42 results on '"Annika K. Wefers"'
Search Results
2. Atypical neurofibromas reveal distinct epigenetic features with proximity to benign peripheral nerve sheath tumor entities
- Author
-
Catena Kresbach, Matthias Dottermusch, Alicia Eckhardt, Inka Ristow, Petros Paplomatas, Lea Altendorf, Annika K Wefers, Michael Bockmayr, Sarra Belakhoua, Ivy Tran, Lara Pohl, Sina Neyazi, Helena Bode, Said Farschtschi, Lennart Well, Reinhard E Friedrich, David Reuss, Matija Snuderl, Christian Hagel, Victor-Felix Mautner, and Ulrich Schüller
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Background Plexiform neurofibromas can transform into atypical neurofibromas (ANF) and then further progress to aggressive malignant peripheral nerve sheath tumors (MPNST). ANF have been described to harbor distinct histological features and frequent loss of CDKN2A/B. However, histological evaluation may be rater-dependent, and detailed knowledge about the molecular mechanisms of malignant transformation is scarce. In general, malignant transformation can be accompanied by significant epigenetic changes, and global DNA methylation profiling is able to differentiate relevant tumor subgroups. Therefore, epigenetic profiling might provide a valuable tool to distinguish and characterize ANF with differing extent of histopathological atypia from neurofibromas and MPNST. Methods We investigated 40 tumors histologically diagnosed as ANF and compared their global methylation profile to other peripheral nerve sheath tumors. Results Unsupervised class discovery and t-SNE analysis indicated that 36/40 ANF cluster with benign peripheral nerve sheath tumors with clear separation from MPNST. 21 ANF formed a molecularly distinct cluster in proximity to schwannomas. Tumors in this cluster had a frequent heterozygous or homozygous loss of CDKN2A/B and significantly more lymphocyte infiltration than MPNST, schwannomas, and NF. Few ANF clustered closely with neurofibromas, schwannomas, or MPNST, raising the question, whether diagnosis based on histological features alone might pose a risk to both over- and underestimate the aggressiveness of these lesions. Conclusions Our data suggest that ANF with varying histological morphology show distinct epigenetic similarities and cluster in proximity to benign peripheral nerve sheath tumor entities. Future investigations should pay special respect to correlating this methylation pattern to clinical outcomes.
- Published
- 2023
3. Pleomorphic xanthoastrocytoma is a heterogeneous entity with pTERT mutations prognosticating shorter survival
- Author
-
Azadeh Ebrahimi, Andrey Korshunov, Guido Reifenberger, David Capper, Joerg Felsberg, Elena Trisolini, Bianca Pollo, Chiara Calatozzolo, Marco Prinz, Ori Staszewski, Leonille Schweizer, Jens Schittenhelm, Patrick N. Harter, Werner Paulus, Christian Thomas, Patricia Kohlhof-Meinecke, Marcel Seiz-Rosenhagen, Till Milde, Belén M. Casalini, Abigail Suwala, Annika K. Wefers, Annekathrin Reinhardt, Philipp Sievers, Christof M. Kramm, Nima Etminam, Andreas Unterberg, Wolfgang Wick, Christel Herold-Mende, Dominik Sturm, Stefan M. Pfister, Martin Sill, David T. W. Jones, Daniel Schrimpf, David E. Reuss, Ken Aldape, Zied Abdullaev, Felix Sahm, Andreas von Deimling, and Damian Stichel
- Subjects
Brain Neoplasms ,Epithelioid glioblastoma ,BRAF V600E ,Research ,Astrocytoma ,DNA Methylation ,Prognosis ,Pathology and Forensic Medicine ,Survival Rate ,Cellular and Molecular Neuroscience ,pTERT mutation ,Mutation ,Humans ,Neurology (clinical) ,Neurology. Diseases of the nervous system ,Pleomorphic xanthoastrocytoma ,DNA methylation array profiling ,RC346-429 ,Telomerase ,Ganglioglioma - Abstract
Pleomorphic xanthoastrocytoma (PXA) in its classic manifestation exhibits distinct morphological features and is assigned to CNS WHO grade 2 or grade 3. Distinction from glioblastoma variants and lower grade glial and glioneuronal tumors is a common diagnostic challenge. We compared a morphologically defined set of PXA (histPXA) with an independent set, defined by DNA methylation analysis (mcPXA). HistPXA encompassed 144 tumors all subjected to DNA methylation array analysis. Sixty-two histPXA matched to the methylation class mcPXA. These were combined with the cases that showed the mcPXA signature but had received a histopathological diagnosis other than PXA. This cohort constituted a set of 220 mcPXA. Molecular and clinical parameters were analyzed in these groups. Morphological parameters were analyzed in a subset of tumors with FFPE tissue available. HistPXA revealed considerable heterogeneity in regard to methylation classes, with methylation classes glioblastoma and ganglioglioma being the most frequent mismatches. Similarly, the mcPXA cohort contained tumors of diverse histological diagnoses, with glioblastoma constituting the most frequent mismatch. Subsequent analyses demonstrated the presence of canonical pTERT mutations to be associated with unfavorable prognosis among mcPXA. Based on these data, we consider the tumor type PXA to be histologically more varied than previously assumed. Histological approach to diagnosis will predominantly identify cases with the established archetypical morphology. DNA methylation analysis includes additional tumors in the tumor class PXA that share similar DNA methylation profile but lack the typical morphology of a PXA. DNA methylation analysis also assist in separating other tumor types with morphologic overlap to PXA. Our data suggest the presence of canonical pTERT mutations as a robust indicator for poor prognosis in methylation class PXA. Supplementary Information The online version contains supplementary material available at 10.1186/s40478-021-01308-1.
- Published
- 2022
4. Risk prediction in early childhood sonic hedgehog medulloblastoma treated with radiation-avoiding chemotherapy: Evidence for more than 2 subgroups
- Author
-
Svenja Tonn, Andrey Korshunov, Denise Obrecht, Martin Sill, Michael Spohn, Katja von Hoff, Till Milde, Torsten Pietsch, Tobias Goschzik, Brigitte Bison, Björn-Ole Juhnke, Nina Struve, Dominik Sturm, Felix Sahm, Michael Bockmayr, Carsten Friedrich, André O von Bueren, Nicolas U Gerber, Martin Benesch, David T W Jones, Marcel Kool, Annika K Wefers, Ulrich Schüller, Stefan M Pfister, Stefan Rutkowski, and Martin Mynarek
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Background The prognostic impact of clinical risk factors and DNA methylation patterns in sonic hedgehog (SHH)-activated early childhood desmoplastic/nodular medulloblastoma (DMB) or medulloblastoma with extensive nodularity (MBEN) were evaluated to better identify patients at risk for relapse. Methods One hundred and forty-four patients with DMB (n = 99) or MBEN (n = 45) aged Results Patients with DMB had less favorable 5-year progression-free survival than MBEN (5y-PFS, 71% [DMB] vs. 93% [MBEN]). Patients aged >3 years were associated with more unfavorable 5y-PFS (47% [>3 years] vs. 85% [ Conclusions These data suggest further heterogeneity within early childhood SHH-DMB/MBEN: SHH-2 splits into a very low-risk group SHH-2a enriched for MBEN histology and SMO mutations, and SHH-2b comprising older DMB patients with a higher risk of relapse.
- Published
- 2023
5. Analysing Cerebrospinal Fluid with Explainable Deep Learning: from Diagnostics to Insights
- Author
-
Leonille Schweizer, Philipp Seegerer, Hee‐yeong Kim, René Saitenmacher, Amos Muench, Liane Barnick, Anja Osterloh, Carsten Dittmayer, Ruben Jödicke, Debora Pehl, Annekathrin Reinhardt, Klemens Ruprecht, Werner Stenzel, Annika K. Wefers, Patrick N. Harter, Ulrich Schüller, Frank L. Heppner, Maximilian Alber, Klaus‐Robert Müller, and Frederick Klauschen
- Subjects
Histology ,deep learning ,000 Informatik, Informationswissenschaft, allgemeine Werke::000 Informatik, Wissen, Systeme::005 Computerprogrammierung, Programme, Daten ,heatmaps ,cerebrospinal fluid ,explainable AI ,Pathology and Forensic Medicine ,Deep Learning ,Neurology ,Artificial Intelligence ,Physiology (medical) ,methods [Image Processing, Computer-Assisted] ,Humans ,Neurology (clinical) ,ddc:610 ,Neural Networks, Computer ,cell detection - Abstract
Aim: Analysis of cerebrospinal fluid (CSF) is essential for diagnostic workup of patients with neurological diseases and includes differential cell typing. The current gold standard is based on microscopic examination by specialised technicians and neuropathologists, which is time‐consuming, labour‐intensive and subjective. Methods: We, therefore, developed an image analysis approach based on expert annotations of 123,181 digitised CSF objects from 78 patients corresponding to 15 clinically relevant categories and trained a multiclass convolutional neural network (CNN). Results: The CNN classified the 15 categories with high accuracy (mean AUC 97.3%). By using explainable artificial intelligence (XAI), we demonstrate that the CNN identified meaningful cellular substructures in CSF cells recapitulating human pattern recognition. Based on the evaluation of 511 cells selected from 12 different CSF samples, we validated the CNN by comparing it with seven board‐certified neuropathologists blinded for clinical information. Inter‐rater agreement between the CNN and the ground truth was non‐inferior (Krippendorff's alpha 0.79) compared with the agreement of seven human raters and the ground truth (mean Krippendorff's alpha 0.72, range 0.56–0.81). The CNN assigned the correct diagnostic label (inflammatory, haemorrhagic or neoplastic) in 10 out of 11 clinical samples, compared with 7–11 out of 11 by human raters. Conclusions: Our approach provides the basis to overcome current limitations in automated cell classification for routine diagnostics and demonstrates how a visual explanation framework can connect machine decision‐making with cell properties and thus provide a novel versatile and quantitative method for investigating CSF manifestations of various neurological diseases.
- Published
- 2023
6. Anaplastic ganglioglioma-A diagnosis comprising several distinct tumour types
- Author
-
Annekathrin Reinhardt, Kristin Pfister, Daniel Schrimpf, Damian Stichel, Felix Sahm, David E. Reuss, David Capper, Annika K. Wefers, Azadeh Ebrahimi, Martin Sill, Joerg Felsberg, Guido Reifenberger, Albert Becker, Marco Prinz, Ori Staszewski, Christian Hartmann, Jens Schittenhelm, Dorothee Gramatzki, Michael Weller, Adriana Olar, Elisabeth Jane Rushing, Markus Bergmann, Michael A. Farrell, Ingmar Blümcke, Roland Coras, Jan Beckervordersandforth, Se Hoon Kim, Fabio Rogerio, Petia S. Dimova, Pitt Niehusmann, Andreas Unterberg, Michael Platten, Stefan M. Pfister, Wolfgang Wick, Christel Herold‐Mende, Andreas von Deimling, MUMC+: DA Pat Pathologie (9), and RS: GROW - R2 - Basic and Translational Cancer Biology
- Subjects
Histology ,Brain Neoplasms ,Glioma ,Astrocytoma ,Isocitrate Dehydrogenase ,Pathology and Forensic Medicine ,Central Nervous System Neoplasms ,Neurology ,Physiology (medical) ,Humans ,Neurology (clinical) ,Child ,Ganglioglioma ,Retrospective Studies - Abstract
Anaplastic ganglioglioma is a rare tumour, and diagnosis has been based on histological criteria. The 5th edition of the World Health Organization Classification of Tumours of the Central Nervous System (CNS WHO) does not list anaplastic ganglioglioma as a distinct diagnosis due to lack of molecular data in previous publications. We retrospectively compiled a cohort of 54 histologically diagnosed anaplastic gangliogliomas to explore whether the molecular profiles of these tumours represent a separate type or resolve into other entities.Samples were subjected to histological review, desoxyribonucleic acid (DNA) methylation profiling and next-generation sequencing. Morphological and molecular data were summarised to an integrated diagnosis.The majority of tumours designated as anaplastic gangliogliomas resolved into other CNS WHO diagnoses, most commonly pleomorphic xanthoastrocytoma (16/54), glioblastoma, isocitrate dehydrogenase protein (IDH) wild type and diffuse paediatric-type high-grade glioma, H3 wild type and IDH wild type (11 and 2/54), followed by low-grade glial or glioneuronal tumours including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumour and diffuse leptomeningeal glioneuronal tumour (5/54), IDH mutant astrocytoma (4/54) and others (6/54). A subset of tumours (10/54) was not assignable to a CNS WHO diagnosis, and common molecular profiles pointing to a separate entity were not evident.In summary, we show that tumours histologically diagnosed as anaplastic ganglioglioma comprise a wide spectrum of CNS WHO tumour types with different prognostic and therapeutic implications. We therefore suggest assigning this designation with caution and recommend comprehensive molecular workup.
- Published
- 2022
7. Neurofibromatosis type 2 predisposes to ependymomas of various localization, histology, and molecular subtype
- Author
-
Abigail K. Suwala, Leonille Schweizer, Brigitte Bison, Annika K. Wefers, Christian Hagel, Andreas von Deimling, Katja Kloth-Stachnau, Lara Engertsberger, Martin Mynarek, Michael Spohn, Catena Kresbach, Martin Benesch, Stefan Rutkowski, Ulrich Schüller, Viktor-F Mautner, and Mario M. Dorostkar
- Subjects
Adult ,Male ,genetics [Ependymoma] ,Pathology ,medicine.medical_specialty ,Neurofibromatosis 2 ,genetics [Central Nervous System Neoplasms] ,Adolescent ,MEDLINE ,genetics [DNA Methylation] ,Pathology and Forensic Medicine ,Central Nervous System Neoplasms ,Cellular and Molecular Neuroscience ,Text mining ,Genes, Neurofibromatosis 2 ,Correspondence ,medicine ,Humans ,pathology [Neurofibromatosis 2] ,Neurosciences ,ddc:610 ,Neurofibromatosis type 2 ,Child ,genetics [Neurofibromatosis 2] ,business.industry ,pathology [Ependymoma] ,pathology [Central Nervous System Neoplasms] ,Histology ,DNA Methylation ,medicine.disease ,Magnetic Resonance Imaging ,diagnostic imaging [Central Nervous System Neoplasms] ,complications [Neurofibromatosis 2] ,Ependymoma ,Child, Preschool ,Mutation ,Female ,Neurology (clinical) ,business ,diagnostic imaging [Ependymoma] - Published
- 2021
8. Molecular characterization of CNS paragangliomas identifies cauda equina paragangliomas as a distinct tumor entity
- Author
-
Alexander Younsi, David E. Reuss, Johannes Kahn, David Capper, Anne Schöler, Georgios Ntoulias, Daniel Teichmann, Hee-Yeong Kim, Dag Moskopp, Diaa Al Safatli, Abigail K. Suwala, Jan Walter, Sven-Axel May, Peter Vajkoczy, David Kaul, Bettina Knie, Frank L. Heppner, Wolfgang Hartmann, Andreas Unterberg, Martin Hasselblatt, Leonille Schweizer, Damian Stichel, Felix Thierfelder, Christian Hartmann, Andreas Jödicke, Arend Koch, Martin Misch, Ruben Jödicke, Annika K. Wefers, Patrick Soschinski, Andreas von Deimling, Michael Bockmayr, Lars Wessels, and Christian Thomas
- Subjects
Male ,musculoskeletal diseases ,Pathology ,medicine.medical_specialty ,genetics [Central Nervous System Neoplasms] ,genetics [Paraganglioma] ,SDHB ,diagnosis [Neoplasm Recurrence, Local] ,Neuroendocrine tumors ,Biology ,Cauda equina ,Pathology and Forensic Medicine ,Central Nervous System Neoplasms ,Diagnosis, Differential ,Paraganglioma ,Head and neck ,Cellular and Molecular Neuroscience ,Cytokeratin ,genetics [Neuroendocrine Tumors] ,Germline mutation ,pathology [Cauda Equina] ,pathology [Neuroendocrine Tumors] ,GATA3 ,medicine ,Humans ,ddc:610 ,pathology [Neoplasm Recurrence, Local] ,diagnosis [Neuroendocrine Tumors] ,Germ-Line Mutation ,Exome sequencing ,genetics [Germ-Line Mutation] ,Original Paper ,DNA methylation ,Adrenal gland ,pathology [Central Nervous System Neoplasms] ,Prognosis ,medicine.disease ,genetics [Neoplasm Recurrence, Local] ,pathology [Paraganglioma] ,Neuroendocrine Tumors ,medicine.anatomical_structure ,Female ,Neurology (clinical) ,Neoplasm Recurrence, Local ,600 Technik, Medizin, angewandte Wissenschaften::610 Medizin und Gesundheit::610 Medizin und Gesundheit - Abstract
Paragangliomas/pheochromocytomas are rare neuroendocrine tumors that arise from the adrenal gland or ganglia at various sites throughout the body. They display a remarkable diversity of driver alterations and are associated with germline mutations in up to 40% of the cases. Comprehensive molecular profiling of abdomino-thoracic paragangliomas revealed four molecularly defined and clinically relevant subtypes. Paragangliomas of the cauda equina region are considered to belong to one of the defined molecular subtypes, but a systematic molecular analysis has not yet been performed. In this study, we analyzed genome-wide DNA methylation profiles of 57 cauda equina paragangliomas and show that these tumors are epigenetically distinct from non-spinal paragangliomas and other tumors. In contrast to paragangliomas of other sites, chromosomal imbalances are widely lacking in cauda equina paragangliomas. Furthermore, RNA and DNA exome sequencing revealed that frequent genetic alterations found in non-spinal paragangliomas—including the prognostically relevant SDH mutations—are absent in cauda equina paragangliomas. Histologically, cauda equina paragangliomas show frequently gangliocytic differentiation and strong immunoreactivity to pan-cytokeratin and cytokeratin 18, which is not common in paragangliomas of other sites. None of our cases had a familial paraganglioma syndrome. Tumors rarely recurred (9%) or presented with multiple lesions within the spinal compartment (7%), but did not metastasize outside the CNS. In summary, we show that cauda equina paragangliomas represent a distinct, sporadic tumor entity defined by a unique clinical and morpho-molecular profile. Electronic supplementary material The online version of this article (10.1007/s00401-020-02218-7) contains supplementary material, which is available to authorized users.
- Published
- 2020
9. Comprehensive profiling of myxopapillary ependymomas identifies a distinct molecular subtype with relapsing disease
- Author
-
Michael Bockmayr, Kim Harnisch, Lara C Pohl, Leonille Schweizer, Theresa Mohme, Meik Körner, Malik Alawi, Abigail K Suwala, Mario M Dorostkar, Camelia M Monoranu, Martin Hasselblatt, Annika K Wefers, David Capper, Jürgen Hench, Stephan Frank, Timothy E Richardson, Ivy Tran, Elisa Liu, Matija Snuderl, Lara Engertsberger, Martin Benesch, Andreas von Deimling, Denise Obrecht, Martin Mynarek, Stefan Rutkowski, Markus Glatzel, Julia E Neumann, and Ulrich Schüller
- Subjects
Adult ,Cancer Research ,DNA methylation ,pathology [Spinal Cord Neoplasms] ,pathology [Ependymoma] ,Myxopapillary ependymoma ,RNA sequencing ,DNA Methylation ,Middle Aged ,Cohort Studies ,histology ,Oncology ,Ependymoma ,Recurrence ,Basic and Translational Investigations ,outcome ,Humans ,Neurology (clinical) ,Spinal Cord Neoplasms ,ddc:610 - Abstract
Background Myxopapillary ependymoma (MPE) is a heterogeneous disease regarding histopathology and outcome. The underlying molecular biology is poorly understood, and markers that reliably predict the patients’ clinical course are unknown. Methods We assembled a cohort of 185 tumors classified as MPE based on DNA methylation. Methylation patterns, copy number profiles, and MGMT promoter methylation were analyzed for all tumors, 106 tumors were evaluated histomorphologically, and RNA sequencing was performed for 37 cases. Based on methylation profiling, we defined two subtypes MPE-A and MPE-B, and explored associations with epidemiological, clinical, pathological, and molecular characteristics of these tumors. Results MPE-A occurred at a median age of 27 years and were enriched with tumors demonstrating papillary morphology and MGMT promoter hypermethylation. Half of these tumors could not be totally resected, and 85% relapsed within 10 years. Copy number alterations were more common in MPE-A. RNA sequencing revealed an enrichment for extracellular matrix and immune system-related signatures in MPE-A. MPE-B occurred at a median age of 45 years and included many tumors with a histological diagnosis of WHO grade II and tanycytic morphology. Patients within this subtype had a significantly better outcome with a relapse rate of 33% in 10 years (P = 3.4e-06). Conclusions We unraveled the morphological and clinical heterogeneity of MPE by identifying two molecularly distinct subtypes. These subtypes significantly differed in progression-free survival and will likely need different protocols for surveillance and treatment.
- Published
- 2022
10. An H3F3A K27M‐mutation in a sonic hedgehog medulloblastoma
- Author
-
Nesrin Uksul, Matthias Dottermusch, Annika K. Wefers, Bernhard Erdlenbruch, and Ulrich J. Knappe
- Subjects
K27m mutation ,Mutant ,Pathology and Forensic Medicine ,Histones ,medicine ,Humans ,Hedgehog Proteins ,Sonic hedgehog ,Cerebellar Neoplasms ,neoplasms ,Gene ,Medulloblastoma ,biology ,Brain Neoplasms ,General Neuroscience ,Glioma ,medicine.disease ,nervous system diseases ,Histone ,Mutation ,Mutation (genetic algorithm) ,biology.protein ,Cancer research ,Neurology (clinical) ,Antibody - Abstract
Medulloblastomas are malignant embryonal brain tumours that may harbour mutations in histone-modifying genes, while mutations in histone genes have not been detected to date. We here describe the first SHH medulloblastoma with H3 K27M mutation. This may have diagnostic implications as H3 K27M mutations are the hallmark of diffuse midline gliomas, H3 K27M mutant, WHO grade IV. Medulloblastomas arise in midline structures and thus must not be mistaken for DMG when using an antibody detecting the H3 K27M mutation.
- Published
- 2021
11. An optimized workflow to improve reliability of detection of KIAA1549:BRAF fusions from RNA sequencing data
- Author
-
Felix Sahm, Pengbo Sun, Andrey Korshunov, David T.W. Jones, Stefan M. Pfister, Andreas von Deimling, Sebastian Uhrig, Daniel Sinnett, Damian Stichel, Annika K. Wefers, Alexander C Sommerkamp, Pascal St-Onge, Natalie Jäger, and Nada Jabado
- Subjects
Oncogene Proteins ,Oncogene Proteins, Fusion ,Sequence analysis ,Computer science ,business.industry ,Sequence Analysis, RNA ,Sequencing data ,RNA ,Computational biology ,Astrocytoma ,Pathology and Forensic Medicine ,Workflow ,Cellular and Molecular Neuroscience ,Text mining ,Correspondence ,Humans ,Neurology (clinical) ,business ,Reliability (statistics) - Published
- 2020
12. Molecular characterization of histopathological ependymoma variants
- Author
-
Marcel Kool, Mario M. Dorostkar, Markus Glatzel, Christian Thomas, Martin Mynarek, Hendrik Witt, Annika K. Wefers, Camelia-Maria Monoranu, Arend Koch, Stephan Frank, Denise Obrecht, Michael Spohn, Julia E Neumann, Martin Hasselblatt, Stefan Rutkowski, Ulrich Schüller, and Kristian W. Pajtler
- Subjects
Adult ,Male ,0301 basic medicine ,Ependymoma ,genetics [Ependymoma] ,Pathology ,medicine.medical_specialty ,pathology [Brain Neoplasms] ,mortality [Ependymoma] ,Adolescent ,Papillary Ependymoma ,Brain tumor ,Pathology and Forensic Medicine ,Cohort Studies ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Glioma ,medicine ,Humans ,ddc:610 ,Child ,Aged ,Aged, 80 and over ,Brain Neoplasms ,business.industry ,pathology [Ependymoma] ,Papillary tumor ,DNA Methylation ,Middle Aged ,medicine.disease ,Progression-Free Survival ,genetics [Brain Neoplasms] ,mortality [Brain Neoplasms] ,Survival Rate ,030104 developmental biology ,DNA methylation ,Female ,Neurology (clinical) ,Neoplasm Grading ,Neurocytoma ,business ,030217 neurology & neurosurgery ,Clear cell - Abstract
According to the WHO classification, ependymal tumors are classified as subependymomas, myxopapillary ependymomas, classic ependymomas, anaplastic ependymomas, and RELA-fusion-positive ependymomas (RELA-EPN). Among classic ependymomas, the WHO defines rare histological variants, i.e., the clear cell, papillary, and tanycytic ependymoma. In parallel, global DNA methylation patterns distinguish nine molecular groups, some of which tightly overlap with histopathological subgroups. However, the match of the aforementioned histological variants to DNA methylation classes remains unclear. We analyzed histomorphology, clinical parameters, and global DNA methylation of tumors with the initial histological diagnoses of tanycytic (n = 12), clear cell (n = 14), or papillary ependymoma (n = 19). Forty percent of these tumors did not match to the epigenetic profile of ependymomas, using a previously published DNA methylation-based classifier for brain tumors. Instead, they were classified as low-grade glioma (n = 3), plexus tumor (n = 2), CNS high-grade neuroepithelial tumor with MN1 alteration (n = 2), papillary tumor of the pineal region (n = 2), neurocytoma (n = 1), or did not match to any known brain tumor methylation class (n = 8). Overall, integrated diagnosis had to be changed in 35.6% of cases as compared to the initial diagnosis. Among the tumors molecularly classified as ependymoma (27/45 cases), tanycytic ependymomas were mostly located in the spine (5/7 cases) and matched to spinal or myxopapillary ependymoma. 6/8 clear cell ependymomas were found supratentorially and fell into the methylation class of RELA-EPN. Papillary ependymomas with a positive ependymoma match (12/19 cases) showed either a 'papillary' (n = 5), a 'trabecular' (n = 1), or a 'pseudo-papillary' (n = 6) growth pattern. The papillary growth pattern was strongly associated with the methylation class B of posterior fossa ependymoma (PFB, 5/5 cases) and tumors displayed DNA methylation sites that were significantly different when compared to PFB ependymomas without papillary growth. Tumors with pseudo-papillary histology matched to the methylation class of myxopapillary ependymoma (4/6 cases), whereas the trabecular case was anatomically and molecularly a spinal ependymoma. Our results show that the diagnosis of histological ependymoma variants is challenging and epigenetic profiles may improve diagnostic accuracy of these cases. Whereas clear cell and papillary ependymomas display correlations between localization, histology, and methylation, tanycytic ependymoma does not represent a molecularly distinct subgroup.
- Published
- 2019
13. Rosette-forming glioneuronal tumors share a distinct DNA methylation profile and mutations in FGFR1, with recurrent co-mutation of PIK3CA and NF1
- Author
-
Karin de Stricker, Wolfgang Brück, Eleonora Aronica, Felice Giangaspero, Benedicte Parm Ulhøi, Simone Schmid, Annika K. Wefers, Viktoria Ruf, David T.W. Jones, Christel Herold-Mende, Jeanette Krogh Petersen, Daniel Schrimpf, Andreas von Deimling, Henning B. Boldt, Maria Gardberg, David Capper, Karima Mokhtari, Felix Sahm, Philipp Sievers, Romain Appay, Stefan M. Pfister, Dominique Figarella-Branger, Sebastian Brandner, Bjarne Winther Kristensen, Martin Hasselblatt, Elisabeth J. Rushing, Wolfgang Wick, Daniel Hänggi, David E. Reuss, Pieter Wesseling, Annekathrin Reinhardt, Damian Stichel, Benoit Lhermitte, Franck Bielle, Roland Coras, Pathology, APH - Aging & Later Life, APH - Mental Health, and CCA - Cancer biology and immunology
- Subjects
0301 basic medicine ,MAPK/ERK pathway ,Male ,PI3K ,DNA methylation profile ,0302 clinical medicine ,Rosette-forming glioneuronal tumor ,Child ,Neurons ,Neurofibromin 1 ,molecular classification ,Brain Neoplasms ,Glioma ,Middle Aged ,Molecular classification ,DNA methylation ,Female ,Signal transduction ,RGNT ,brain tumor ,Adult ,Tumor suppressor gene ,Adolescent ,Class I Phosphatidylinositol 3-Kinases ,Brain tumor ,Biology ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,medicine ,FGFR1 ,MAPK ,NF1 ,PIK3CA ,Humans ,Epigenetics ,Receptor, Fibroblast Growth Factor, Type 1 ,PI3K/AKT/mTOR pathway ,Aged ,Retrospective Studies ,Fibroblast growth factor receptor 1 ,DNA Methylation ,medicine.disease ,030104 developmental biology ,Mutation ,Cancer research ,Neurology (clinical) ,030217 neurology & neurosurgery - Abstract
Rosette-forming glioneuronal tumor (RGNT) is a rare brain neoplasm that primarily affects young adults. Although alterations affecting the mitogen-activated protein kinase (MAPK) and phosphoinositide 3-kinase (PI3K) signaling pathway have been associated with this low-grade entity, comprehensive molecular investigations of RGNT in larger series have not been performed to date, and an integrated view of their genetic and epigenetic profiles is still lacking. Here we describe a genome-wide DNA methylation and targeted sequencing-based characterization of a molecularly distinct class of tumors (n = 30), initially identified through genome-wide DNA methylation screening among a cohort of > 30,000 tumors, of which most were diagnosed histologically as RGNT. FGFR1 hotspot mutations were observed in all tumors analyzed, with co-occurrence of PIK3CA mutations in about two-thirds of the cases (63%). Additional loss-of-function mutations in the tumor suppressor gene NF1 were detected in a subset of cases (33%). Notably, in contrast to most other low-grade gliomas, these tumors often displayed co-occurrence of two or even all three of these mutations. Our data highlight that molecularly defined RGNTs are characterized by highly recurrent combined genetic alterations affecting both MAPK and PI3K signaling pathways. Thus, these two pathways appear to synergistically interact in the formation of RGNT, and offer potential therapeutic targets for this disease.
- Published
- 2019
14. Routine RNA sequencing of formalin-fixed paraffin-embedded specimens in neuropathology diagnostics identifies diagnostically and therapeutically relevant gene fusions
- Author
-
David T.W. Jones, Christian Koelsche, Tobias Kessler, Till Milde, Jochen Meyer, Dominik Sturm, Andrey Korshunov, Felix Sahm, Annika K. Wefers, Daniel Schrimpf, Christel Herold-Mende, Annekathrin Reinhardt, Olaf Witt, Belen Casalini, Stefan M. Pfister, Damian Stichel, Andreas von Deimling, Francisco Fernández-Klett, Wolfgang Wick, David E. Reuss, Azadeh Ebrahimi, Jonas Ecker, and Philipp Sievers
- Subjects
0301 basic medicine ,DNA Mutational Analysis ,Brain tumor ,Druggability ,Neuropathology ,Computational biology ,Pathology and Forensic Medicine ,Fusion gene ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Humans ,Medicine ,Gene ,Paraffin Embedding ,Base Sequence ,Brain Neoplasms ,Sequence Analysis, RNA ,business.industry ,High-Throughput Nucleotide Sequencing ,medicine.disease ,Molecular diagnostics ,030104 developmental biology ,MRNA Sequencing ,Mutation ,DNA methylation ,Neurology (clinical) ,Gene Fusion ,business ,030217 neurology & neurosurgery - Abstract
Molecular markers have become pivotal in brain tumor diagnostics. Mutational analyses by targeted next-generation sequencing of DNA and array-based DNA methylation assessment with copy number analyses are increasingly being used in routine diagnostics. However, the broad variety of gene fusions occurring in brain tumors is marginally covered by these technologies and often only assessed by targeted assays. Here, we assessed the feasibility and clinical value of investigating gene fusions in formalin-fixed paraffin-embedded (FFPE) tumor tissues by next-generation mRNA sequencing in a routine diagnostic setting. After establishment and optimization of a workflow applicable in a routine setting, prospective diagnostic application in a neuropathology department for 26 months yielded relevant fusions in 66 out of 101 (65%) analyzed cases. In 43 (43%) cases, the fusions were of decisive diagnostic relevance and in 40 (40%) cases the fusion genes rendered a druggable target. A major strength of this approach was its ability to detect fusions beyond the canonical alterations for a given entity, and the unbiased search for any fusion event in cases with uncertain diagnosis and, thus, uncertain spectrum of expected fusions. This included both rare variants of established fusions which had evaded prior targeted analyses as well as the detection of previously unreported fusion events. While the impact of fusion detection on diagnostics is highly relevant, it is especially the detection of "druggable" fusions which will most likely provide direct benefit to the patients. The wider application of this approach for unbiased fusion identification therefore promises to be a major advance in identifying alterations with immediate impact on patient care.
- Published
- 2019
15. OTHR-41. Amplification of the PLAG family genes – PLAGL1 and PLAGL2 – is a key feature of a novel embryonal CNS tumor type
- Author
-
Michaela-Kristina Keck, Martin Sill, Andrea Wittmann, Piyush Joshi Kumar, Damian Stichel, Philipp Sievers, Annika K Wefers, Federico Roncaroli, James Hayden, Martin G McCabe, Mariëtte E G Kranendonk, Michal Zapotocky, Alexandre Vasiljevic, Ulrich Schüller, Dominik Sturm, Mirjam Blattner-Johnson, Andreas von Deimling, Andrey Korshunov, Felix Sahm, Arie Perry, David Solomon, Stefan Pfister, and David T W Jones
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Pediatric central nervous system (CNS) tumors differ substantially from their adult counterparts, are marked by considerable molecular and clinical heterogeneity, and diagnosis through histopathology alone can be challenging. Using DNA methylation-based CNS tumor classification in combination with copy number and RNAseq analysis, we identify a rare, novel pediatric CNS tumor type (n=32) which is characterized by focal high-level amplification and consecutive overexpression of one of the PLAG family genes – PLAGL1 or PLAGL2. It is epigenetically divergent from other known tumor types such as high-grade gliomas, medulloblastomas, embryonal tumors, or CNS sarcomas. The wide range of original histopathologic diagnosis rendered attests to their polyphenotypic nature in terms of morphology. We suggest that these tumors may arise from early to intermediate neural progenitor cells with some neuronal commitment. Using ChIPseq data, we show that both PLAGL1 and PLAGL2 act as transcription factors for: i) the oncogenic kinase RET, a potential drug target, that was overexpressed in our cohort; ii) components of the Wnt/β-Catenin pathway; iii) a set of imprinted genes, reported to regulate the imprinted gene network in mouse models, that was deregulated in the PLAGL-amplified tumors. Consequently, a 250-gene expression PLAGL-signature indicated dysregulation of imprinting control and differentiation/development as a prominent feature. We report differences regarding age and sex distribution between PLAGL1- and PLAGL2-amplified tumors and shed light on differences in clinical behavior and outcomes between these subtypes in male and female patients. PLAGL1-amplified tumors were more prevalent in school-age children and teenagers, while PLAGL2-amplified cases occurred in very young patients. Kaplan-Meier analysis showed a trend towards a more favorable outcome in patients with PLAGL1-amplified tumors and in female patients. Survival rates remained constant after 5 years with a five-/ten-year overall survival of 75% for PLAGL1, 24% for PLAGL2, 18% for male patients, and 88% for female patients.
- Published
- 2022
16. HGG-45. Characterization of spinal diffuse midline gliomas, H3 K28M-mutant
- Author
-
Lotte Stegat, Christian Thomas, Leonille Schweizer, Sina Neyazi, Lara Pohl, Stephan Frank, Mario M Dorostkar, Ulrich Schüller, and Annika K Wefers
- Subjects
Cancer Research ,Oncology ,Neurology (clinical) - Abstract
Diffuse midline gliomas (DMGs) are malignant gliomas that arise in the midline structures of the central nervous system. Due to their aggressive and diffuse growth and a two-year survival rate of less than 10%, DMGs are assigned to CNS WHO grade 4. Depending on the localization, median age of patients is about 11‒20 years. Genetically, most tumors are defined by a K28M-mutation in one of the highly homologous genes encoding histone protein H3. Since DMGs most frequently occur in pons and thalamus, comparatively little is known about spinal DMGs. Therefore, we histologically, molecularly, and clinically characterized spinal DMGs and analyzed, in which aspects they differ from DMGs of other localizations. Our cohort currently consists of 25 spinal DMGs and 40 pontine/thalamic reference cases. Histological, immunohistochemical and molecular analyses (DNA methylation, DNA panel sequencing) were done from FFPE tissue. Spinal DMGs were histologically very heterogeneous, both regarding different areas of single tumors as well as in comparison to other spinal and reference cases. First cluster analyses of DNA methylation data indicated a separation into three main clusters enriched for pontine, thalamic or spinal cases. The cluster enriched for spinal cases contained many tumors from elderly patients. Overall, mean age of patients with spinal DMGs was 28 years. Patients were significantly older than those with pontine DMGs. 19/20 spinal DMGs were H3-3A K28M-mutant, while one tumor had an H3-2B mutation. 4/19 (21%) spinal DMGs had mutations in FGFR1, and 6/10 (60%) in NF1. Three tumors had KRAS or BRAF mutations. In summary, first analyses suggest slight histological differences of spinal DMGs compared to DMGs of other localizations. Preliminary cluster analyses of DNA methylation data showed an enrichment of clusters for different localizations. About one third of spinal DMGs had mutations in a gene associated with the MAPK-signaling pathway.
- Published
- 2022
17. Primary mismatch repair deficient IDH-mutant astrocytoma (PMMRDIA) is a distinct type with a poor prognosis
- Author
-
Christian F. Freyschlag, Annika K. Wefers, Abigail K. Suwala, Stefan M. Pfister, Arnulf Pekrun, Matija Snuderl, Christian Hartmann, Andrey Korshunov, Christian P. Kratz, Philipp Sievers, Camelia M. Monoranu, Eleonora Aronica, David T.W. Jones, Daniel Schrimpf, Sybren L. N. Maas, Malak Abedalthagafi, Dominik Sturm, Andreas Unterberg, Matthias Kloor, Damian Stichel, Andrew Dodgshun, Zied Abdullaev, Leonille Schweizer, Christof M. Kramm, Felix Sahm, Felix Hinz, Wolfgang Wick, David E. Reuss, Uri Tabori, Pieter Wesseling, Martin Hasselblatt, Andreas von Deimling, Annekathrin Reinhardt, Marcel Kool, Markus Bergmann, Pathology, CCA - Cancer biology and immunology, APH - Aging & Later Life, APH - Mental Health, and ANS - Cellular & Molecular Mechanisms
- Subjects
Adult ,Male ,IDH ,X-linked Nuclear Protein ,Adolescent ,Gene Dosage ,Lynch ,Subtype ,Kaplan-Meier Estimate ,Astrocytoma ,MLH1 ,DNA Mismatch Repair ,Pathology and Forensic Medicine ,Mismatch repair ,Cellular and Molecular Neuroscience ,Young Adult ,Germline mutation ,medicine ,Humans ,Child ,neoplasms ,ATRX ,Original Paper ,DNA methylation ,business.industry ,Brain Neoplasms ,Microsatellite instability ,medicine.disease ,Prognosis ,Immunohistochemistry ,Survival Analysis ,Isocitrate Dehydrogenase ,CMMRD ,MSH6 ,Gene Expression Regulation, Neoplastic ,MSH2 ,Mutation ,Cancer research ,DNA mismatch repair ,Female ,Microsatellite Instability ,Neurology (clinical) ,Neoplasm Recurrence, Local ,business ,Glioblastoma ,Signal Transduction - Abstract
Diffuse IDH-mutant astrocytoma mostly occurs in adults and carries a favorable prognosis compared to IDH-wildtype malignant gliomas. Acquired mismatch repair deficiency is known to occur in recurrent IDH-mutant gliomas as resistance mechanism towards alkylating chemotherapy. In this multi-institutional study, we report a novel epigenetic group of 32 IDH-mutant gliomas with proven or suspected hereditary mismatch repair deficiency. None of the tumors exhibited a combined 1p/19q deletion. These primary mismatch repair-deficient IDH-mutant astrocytomas (PMMRDIA) were histologically high-grade and were mainly found in children, adolescents and young adults (median age 14 years). Mismatch repair deficiency syndromes (Lynch or Constitutional Mismatch Repair Deficiency Syndrom (CMMRD)) were clinically diagnosed and/or germline mutations in DNA mismatch repair genes (MLH1, MSH6, MSH2) were found in all cases, except one case with a family and personal history of colon cancer and another case with MSH6-deficiency available only as recurrent tumor. Loss of at least one of the mismatch repair proteins was detected via immunohistochemistry in all, but one case analyzed. Tumors displayed a hypermutant genotype and microsatellite instability was present in more than half of the sequenced cases. Integrated somatic mutational and chromosomal copy number analyses showed frequent inactivation of TP53, RB1 and activation of RTK/PI3K/AKT pathways. In contrast to the majority of IDH-mutant gliomas, more than 60% of the samples in our cohort presented with an unmethylated MGMT promoter. While the rate of immuno-histochemical ATRX loss was reduced, variants of unknown significance were more frequently detected possibly indicating a higher frequency of ATRX inactivation by protein malfunction. Compared to reference cohorts of other IDH-mutant gliomas, primary mismatch repair-deficient IDH-mutant astrocytomas have by far the worst clinical outcome with a median survival of only 15 months irrespective of histological or molecular features. The findings reveal a so far unknown entity of IDH-mutant astrocytoma with high prognostic relevance. Diagnosis can be established by aligning with the characteristic DNA methylation profile, by DNA-sequencing-based proof of mismatch repair deficiency or immunohistochemically demonstrating loss-of-mismatch repair proteins.
- Published
- 2021
18. Accurate calling of KIAA1549-BRAF fusions from DNA of human brain tumours using methylation array-based copy number and gene panel sequencing data
- Author
-
Martin Sill, Andrey Korshunov, Pablo Hernáiz Driever, Michèle Simon, Dominik Sturm, Alexander C Sommerkamp, David E. Reuss, Till Milde, Arend Koch, Ulrich Schüller, Olaf Witt, Florian Selt, Felix Sahm, David T.W. Jones, Belen Casalini, Abigail K. Suwala, David Capper, Annika K. Wefers, Annekathrin Reinhardt, Jonas Ecker, Cornelis M. van Tilburg, Damian Stichel, Andreas von Deimling, Daniel Schrimpf, Philipp Sievers, Stefan M. Pfister, and Astrid Gnekow
- Subjects
0301 basic medicine ,Histology ,Oncogene Proteins, Fusion ,Gene Dosage ,Computational biology ,Biology ,Pathology and Forensic Medicine ,Fusion gene ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Physiology (medical) ,medicine ,Biomarkers, Tumor ,Humans ,ddc:610 ,Pilocytic astrocytoma ,Brain Neoplasms ,Gene Expression Profiling ,Chromosome ,Methylation ,Sequence Analysis, DNA ,DNA Methylation ,medicine.disease ,030104 developmental biology ,Neurology ,chemistry ,DNA methylation ,Neurology (clinical) ,Tandem exon duplication ,Sarcoma ,030217 neurology & neurosurgery ,DNA - Abstract
Aims KIAA1549-BRAF fusions occur in certain brain tumours and provide druggable targets due to a constitutive activation of the MAP-kinase pathway. We introduce workflows for calling the KIAA1549-BRAF fusion from DNA methylation-array-derived copy number as well as DNA panel sequencing data. Methods Copy number profiles were analysed by automated screening and visual verification of a tandem duplication on chromosome 7q34, indicative of the KIAA1549-BRAF fusion. Pilocytic astrocytomas of the ICGC cohort with known fusion status were used for validation. KIAA1549-BRAF fusions were called from DNA panel sequencing data using the fusion callers Manta, Arriba with modified filtering criteria and deFuse. We screened DNA methylation and panel sequencing data of 7790 specimens from brain tumour and sarcoma entities. Results We identified the fusion in 337 brain tumours with both DNA methylation and panel sequencing data. Among these, we detected the fusion from copy number data in 84% and from DNA panel sequencing data in more than 90% using Arriba with modified filters. While in 74% the KIAA1549-BRAF fusion was detected from both methylation-array-derived copy number and panel sequencing data, in 9% it was detected from copy number data only and in 16% from panel data only. The fusion was almost exclusively found in pilocytic astrocytomas, diffuse leptomeningeal glioneuronal tumours and high-grade astrocytomas with piloid features. Conclusions The KIAA1549-BRAF fusion can be reliably detected from either DNA methylation array or DNA panel data. The use of both methods is recommended for the most sensitive detection of this diagnostically and therapeutically important marker.
- Published
- 2020
19. Desmoplastic myxoid tumor, SMARCB1-mutant: clinical, histopathological and molecular characterization of a pineal region tumor encountered in adolescents and adults
- Author
-
Werner Paulus, Martin Sill, Annika K. Wefers, Susanne Bens, Fanny Burel-Vandenbos, Christian Thomas, Michael C. Frühwald, Torsten Pietsch, Francesca Brett, Florian Oyen, Abbas Agaimy, Marcel Kool, Karolina Nemes, Silke Vogelgesang, Guido Reifenberger, Frantz Rom Poulsen, Reiner Siebert, Fausto J. Rodriguez, Roger E. McLendon, Caterina Giannini, Pascal Johann, Eric S. Lipp, Stefan Tippelt, Kathy Keyvani, Klaus Kuchelmeister, Martin Hasselblatt, Andreas von Deimling, Uwe Kordes, Istvan Bodi, Thomas C., Wefers A., Bens S., Nemes K., Agaimy A., Oyen F., Vogelgesang S., Rodriguez F.J., Brett F.M., McLendon R., Bodi I., Burel-Vandenbos F., Keyvani K., Tippelt S., Poulsen F.R., Lipp E.S., Giannini C., Reifenberger G., Kuchelmeister K., Pietsch T., Kordes U., Siebert R., Fruhwald M.C., Johann P.D., Sill M., Kool M., von Deimling A., Paulus W., and Hasselblatt M.
- Subjects
0301 basic medicine ,Adult ,Male ,Pathology ,medicine.medical_specialty ,Proliferation index ,Adolescent ,Medizin ,Brain tumor ,Biology ,Pineal Gland ,Pathology and Forensic Medicine ,Frameshift mutation ,Cohort Studies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,0302 clinical medicine ,Atypical teratoid/rhabdoid tumor (ATRT), myxoid stroma, pineal region ,medicine ,Humans ,SMARCB1 ,Rhabdoid Tumor ,Brain Neoplasms ,Myxoid tumor ,Age Factors ,SMARCB1 Protein ,Middle Aged ,medicine.disease ,Survival Rate ,030104 developmental biology ,Tumor progression ,Atypical teratoid rhabdoid tumor ,Mutation ,Atypical teratoid/rhabdoid tumor, SMARCB1 gene, pineal ,Female ,Neurology (clinical) ,Epithelioid cell ,030217 neurology & neurosurgery - Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is a highly malignant brain tumor predominantly occurring in infants. Mutations of the SMARCB1 gene are the characteristic genetic lesion. SMARCB1-mutant tumors in adolescents and adults are rare and may show uncommon histopathological and clinical features. Here we report seven SMARCB1-deficient intracranial tumors sharing distinct clinical, histopathological and molecular features. Median age of the four females and three males was 40years (range 15–61years). All tumors were located in the pineal region. Histopathologically, these tumors displayed spindled and epithelioid cells embedded in a desmoplastic stroma alternating with a variable extent of a loose myxoid matrix. All cases showed loss of nuclear SMARCB1/INI1 protein expression, expression of EMA and CD34 was frequent and the Ki67/MIB1 proliferation index was low in the majority of cases (median 3%). Three cases displayed heterozygous SMARCB1 deletions and two cases a homozygous SMARCB1 deletion. On sequencing, one tumor showed a 2bp deletion in exon 4 (c.369_370del) and one a short duplication in exon 3 (c.237_276dup) both resulting in frameshift mutations. Most DNA methylation profiles were not classifiable using the Heidelberg Brain Tumor Classifier (version v11b4). By unsupervised t-SNE analysis and hierarchical clustering analysis, however, all tumors grouped closely together and showed similarities with ATRT-MYC. After a median observation period of 48months, three patients were alive with stable disease, whereas one patient experienced tumor progression and three patients had succumbed to disease. In conclusion, our series represents an entity with distinct clinical, histopathological and molecular features showing epigenetic similarities with ATRT-MYC. We propose the designation desmoplastic myxoid tumor (DMT), SMARCB1-mutant, for these tumors.
- Published
- 2020
20. Practical implementation of DNA methylation and copy-number-based CNS tumor diagnostics: the Heidelberg experience
- Author
-
Anne Schöler, Andreas Unterberg, Kristin Huang, Daniel Hänggi, Stefan M. Pfister, Till Milde, David Capper, David T.W. Jones, Daniel Schrimpf, Daniel Teichmann, Michael Platten, Andrey Korshunov, Martin Sill, Wolfgang Wick, David E. Reuss, Azadeh Ebrahimi, Philipp Sievers, Simone Schmid, Damian Stichel, Annekathrin Reinhardt, Andreas von Deimling, Annika K. Wefers, Felix Sahm, Arend Koch, Volker Hovestadt, Christian Koelsche, and Olaf Witt
- Subjects
Male ,0301 basic medicine ,DNA Copy Number Variations ,Computational biology ,EPIC array ,Pathology and Forensic Medicine ,Central Nervous System Neoplasms ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,chemistry.chemical_compound ,0302 clinical medicine ,Humans ,Medicine ,Copy-number variation ,CNS TUMORS ,DNA Modification Methylases ,Retrospective Studies ,Original Paper ,DNA methylation ,business.industry ,Tumor classification ,Tumor Suppressor Proteins ,Receptor Protein-Tyrosine Kinases ,Isocitrate Dehydrogenase ,Neoplasm Proteins ,Dna methylation profiling ,DNA Repair Enzymes ,030104 developmental biology ,chemistry ,Female ,Neurology (clinical) ,business ,030217 neurology & neurosurgery ,DNA - Abstract
Recently, we described a machine learning approach for classification of central nervous system tumors based on the analysis of genome-wide DNA methylation patterns [6]. Here, we report on DNA methylation-based central nervous system (CNS) tumor diagnostics conducted in our institution between the years 2015 and 2018. In this period, more than 1000 tumors from the neurosurgical departments in Heidelberg and Mannheim and more than 1000 tumors referred from external institutions were subjected to DNA methylation analysis for diagnostic purposes. We describe our current approach to the integrated diagnosis of CNS tumors with a focus on constellations with conflicts between morphological and molecular genetic findings. We further describe the benefit of integrating DNA copy-number alterations into diagnostic considerations and provide a catalog of copy-number changes for individual DNA methylation classes. We also point to several pitfalls accompanying the diagnostic implementation of DNA methylation profiling and give practical suggestions for recurring diagnostic scenarios. Electronic supplementary material The online version of this article (10.1007/s00401-018-1879-y) contains supplementary material, which is available to authorized users.
- Published
- 2018
21. Rapid detection of 2-hydroxyglutarate in frozen sections of IDH mutant tumors by MALDI-TOF mass spectrometry
- Author
-
Mark Kriegsmann, Aubry K. Miller, Peter Schirmacher, Andreas von Deimling, Stefan Pusch, Rémi Longuespée, Annika K. Wefers, Christel Herold-Mende, Elena De Vita, Wolfgang Wick, and David E. Reuss
- Subjects
0301 basic medicine ,Time Factors ,Mutant ,0601 Biochemistry and Cell Biology ,METABOLITES ,LIPIDOMICS ,lcsh:RC346-429 ,0302 clinical medicine ,610 Medical sciences Medicine ,Chemistry ,Brain Neoplasms ,Methodology Article ,Assay ,Brain ,Glioma ,GLIOMAS ,Immunohistochemistry ,Isocitrate Dehydrogenase ,Isocitrate dehydrogenase ,Diffuse glioma ,Life Sciences & Biomedicine ,MALDI-TOF ,IDH1 ,BRAIN-TUMORS ,2-hydroxyglutarate ,Mass spectrometry ,Biochemical detection ,IDH2 ,Pathology and Forensic Medicine ,Glutarates ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Biomarkers, Tumor ,Humans ,lcsh:Neurology. Diseases of the nervous system ,Brain Chemistry ,Cryopreservation ,Science & Technology ,MUTATIONS ,Neurosciences ,1103 Clinical Sciences ,Molecular biology ,IDH mutation ,030104 developmental biology ,Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization ,PROTON SPONGE ,Neurology (clinical) ,Neurosciences & Neurology ,Time-of-flight mass spectrometry ,1109 Neurosciences ,MATRIX ,030217 neurology & neurosurgery - Abstract
All isocitrate dehydrogenase (IDH) mutant solid neoplasms exhibit highly elevated levels of D-2-hydroxyglutarate (D-2HG). Detection of 2HG in tumor tissues currently is performed by gas or liquid chromatography-mass spectrometry (GC- or LC-MS) or biochemical detection. While these methods are highly accurate, a considerable amount of time for tissue preparation and a relatively high amount of tissue is required for testing. We here present a rapid approach to detect 2HG in brain tumor tissue based on matrix-assisted laser desorption ionization - time of flight mass spectrometry (MALDI-TOF). We analyzed 26 brain tumor samples with known IDH1 or IDH2 mutation and compared readouts to those from 28 brain tumor samples of wildtype IDH status. IDH mutant samples exhibited a clear positive signal for 2HG which was not observed in any of the IDH wildtype tumors. Our analytical pipeline allowed for 2HG detection in less than 5 min. Data were validated by determining 2HG levels in all tissues with a biochemical assay. In conclusion, we developed a protocol for rapid detection of 2HG levels and illustrate the possibility to use MALDI-TOF for the detection of metabolites on frozen tissue sections in a diagnostic setting. Electronic supplementary material The online version of this article (10.1186/s40478-018-0523-3) contains supplementary material, which is available to authorized users.
- Published
- 2018
22. PATH-34. MOLECULAR AND CLINICAL HETEROGENEITY WITHIN SPINAL EPENDYMOMAS
- Author
-
Martin Mynarek, Lan Kluwe, Lara Pohl, Catena Kresbach, Katrin Lamszus, Mario M. Dorostkar, Abigail K. Suwala, Christian Hagel, Theresa Mohme, V. F. Mautner, Leonille Schweizer, Martin Benesch, Stefan Rutkowski, Ulrich Schüller, Andreas von Deimling, Annika K. Wefers, Sina Al-Kershi, and Lara Engertsberger
- Subjects
Cancer Research ,Oncology ,Computer science ,Clinical heterogeneity ,Path (graph theory) ,Neurology (clinical) ,Topology - Abstract
Ependymomas encompass multiple clinically relevant tumor types based on localization, genetic alterations, as well as epigenetic and transcriptomic profiles. Distinct global DNA methylation signatures serve as the most powerful diagnostic tool to distinguish these types. The methylation class of spinal ependymomas (SP-EPN) comprises mostly WHO°II tumors with slow progression and incomplete surgical resection rate. Molecular data of SP-EPN are scarce and clear treatment recommendations are lacking although these neoplasms represent the most common intramedullary tumors in children and adults. The only known recurrent genetic events in SP-EPN are the loss of chromosome 22q and mutations of the NF2 gene. However, data on the frequency of NF2 mutations range from 16 % to 71 % and originate from small series that lack epigenetic or transcriptomic characterization. Furthermore, it remains unclear whether SP-EPN with germline or sporadic NF2 mutation or with NF2 wild type status display clinical and other molecular differences. Finally, the underlying genomic and transcriptomic changes of SP-EPN without NF2 mutations are fully unclear. To provide a comprehensive molecular profile of SP-EPN, we integrated genomic and epigenetic analyses and clinical data of 170 cases. Unsupervised hierarchical clustering and t-SNE analyses of methylation data revealed three distinct molecular SP-EPN subtypes. Of the three subtypes, only subtype 1 and subtype 2 contained tumors with NF2 mutations, either as previously known germline mutations or as sporadic mutations without evidence for a syndromic disease (p< 0.0001). Besides the lack of NF2 mutations, subtype 3 tumors showed a higher frequency of MGMT promoter methylation (p= 0.0015) and occurred in significantly older patients compared to tumors of subtypes 1 and 2 (p= 0.0038). Further investigations such as whole-exome sequencing, copy number variation profiling, gene expression analysis, and histological evaluation are ongoing and will add to the picture of molecular and clinical heterogeneity within SP-EPN.
- Published
- 2021
23. LGG-33. ISOMORPHIC DIFFUSE GLIOMA HAS RECURRENT GENE FUSIONS OF MYBL1 OR MYB AND CAN BE DISTINGUISHED FROM OTHER MYB/MYBL1 ALTERED GLIOMAS BASED ON A DISTINCT MORPHOLOGY AND DNA METHYLATION PROFILE
- Author
-
David T.W. Jones, Ingmar Blümcke, David Capper, Annika K. Wefers, Andreas von Deimling, Felix Sahm, Daniel Schrimpf, and Damian Stichel
- Subjects
Cancer Research ,Diffuse Glioma ,Oncology ,Low Grade Glioma ,AcademicSubjects/MED00300 ,Morphology (biology) ,MYB ,AcademicSubjects/MED00310 ,Neurology (clinical) ,DNA Methylation Profile ,Biology ,Gene ,Molecular biology - Abstract
Isomorphic diffuse glioma (IDG) was first described in 2004 as an epilepsy-associated supratentorial diffuse glioma with low cellularity, low proliferation and very monomorphic tumour cells. Most patients had seizures since childhood but were operated on as adults. To study the position of these lesions among brain tumours we histologically, molecularly and clinically analysed 26 histologically typical IDGs. Tumour cells were GFAP-positive, MAP2-, OLIG2- and CD34-negative and the nuclear ATRX-expression was retained. Proliferation was very low. Sequencing of 24 cases revealed an IDH-wildtype status. Cluster analyses of DNA methylation data showed that IDG has a DNA methylation profile distinct from those of different glial/glio-neuronal brain tumours and normal hemispheric tissue. About half of IDGs had copy number alterations of MYBL1 or MYB (13/25) and half of the cases analysed by RNA-sequencing had gene fusions of MYBL1 or MYB with various gene partners (11/22), often associated with an increased RNA-expression of the respective MYB-family gene. Integrating all data available, 77% of IDGs had either MYBL1 (54%) or MYB (23%) alterations. All patients had a good outcome and most were seizure-free after surgery. In summary, we show that isomorphic diffuse glioma is a distinct benign tumour in the family of MYB/MYBL1-altered gliomas. DNA methylation analysis is very helpful for their identification. More recent analyses of a large cohort of MYB/MYBL1-altered brain tumours suggest the presence of a third methylation group that primarily contains paediatric cases and seems to be distinct from IDG and angiocentric gliomas. Further histological, molecular and clinical analyses are ongoing.
- Published
- 2020
24. PATH-26. RNA SEQUENCING OF FORMALIN-FIXED PARAFFIN-EMBEDDED SPECIMENS IN DIAGNOSTIC ROUTINE IDENTIFIES CLINICALLY RELEVANT GENE FUSIONS
- Author
-
Damian Stichel, Daniel Schrimpf, Wolfgang Wick, Felix Sahm, Annika K. Wefers, Andreas von Deimling, Jochen Meyer, Stefan M. Pfister, Philipp Sievers, and David T.W. Jones
- Subjects
Cancer Research ,Oncology ,Formalin fixed paraffin embedded ,Path (graph theory) ,AcademicSubjects/MED00300 ,RNA ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Computational biology ,Biology ,Gene ,Pathology and Molecular Diagnosis - Abstract
Pediatric brain tumor entities harbor a variety of gene fusions. Whilst other molecular parameters like somatic mutations and copy number alterations have become pivotal for brain tumor diagnostics, gene fusions are only less well covered by routinely applied methylation arrays or targeted next-generation sequencing of DNA. In a routine diagnostic setting we established and optimized a workflow for investigation of gene fusions in formalin-fixed paraffin-embedded (FFPE) tumor tissues by using RNA sequencing. Assessing different tools for calling fusions from raw data, we found relevant fusions in 66 out of 101 (65%) analyzed cases in a prospective cohort collected over 26 months. In 43 (43%) cases the fusions were of decisive diagnostic relevance and in 40 (40%) cases the fusion genes rendered a druggable target. Besides the relevance of pathognomonic fusions for diagnostics, especially the detection of druggable gene fusions yields direct benefit to the patients. This approach allows for an unbiased search for fusion events in the tested samples. Besides rare variants of established fusions which were not detected by prior targeted analyses, we identified previously unreported fusion events. Exemplified on KIAA1549:BRAF fusion, we in addition provide an overview of the detection accuracy of different methods, including breakpoint detection in DNA methylation array data and fusion gene detection in DNA panel sequencing data. Our data show that RNA sequencing has great diagnostic as well as therapeutic value by clinically detecting relevant alterations.
- Published
- 2020
25. Migration of Interneuron Precursors in the Nascent Cerebellar Cortex
- Author
-
Ronald Jabs, Lachezar Surchev, Karl Schilling, Annika K. Wefers, Christian Haberlandt, and Christian Steinhäuser
- Subjects
0301 basic medicine ,Cerebellum ,Interneuron ,Green Fluorescent Proteins ,Mice, Transgenic ,Histogenesis ,Biology ,Inhibitory postsynaptic potential ,Cerebellar Cortex ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Neural Stem Cells ,Cell Movement ,Interneurons ,medicine ,Animals ,PAX2 Transcription Factor ,Gene Expression Regulation, Developmental ,Cell migration ,Granule cell ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Animals, Newborn ,Neurology ,Cerebellar cortex ,Hepatic stellate cell ,Neurology (clinical) ,Neuroscience ,030217 neurology & neurosurgery - Abstract
The cerebellum arguably constitutes one of the best characterized central nervous circuits, and its structure, cellular function, and histogenesis have been described in exceptional quantitative detail. A notable exception to this is the development of its inhibitory interneurons, and in particular the extensive migrations of future basket and stellate cells. Here, we used acute slices from 8-day-old mice to assess the migration of Pax2-EGFP-tagged precursors of these cells en route to the molecular layer during their transit through the nascent cerebellar cortex. We document that movement of these cells is highly directed. Their speed and directional persistence are larger in the nascent granule cell layer than in the molecular layer. And they migrate periodically, with periods of effective, directed translocation separated by bouts of rather local movement. Finally, we document that the arrangement of these cells in the adult molecular layer is characterized by clustering. These data are discussed with a focus on potential generative mechanisms for the developmental pattern observed.
- Published
- 2017
26. PATH-16. HISTOPATHOLOGICAL EPENDYMOMA VARIANTS ARE ASSOCIATED WITH DISTINCT CLINICAL PARAMETERS AND DNA METHYLATION PATTERNS
- Author
-
Annika K. Wefers, Markus Glatzel, Christian Thomas, Camelia-Maria Monoranu, Marcel Kool, Martin Mynarek, Hendrik Witt, Arend Koch, Martin Hasselblatt, Julia E Neumann, Denise Obrecht, Michael Spohn, Stephan Frank, Stefan Rutkowski, Ulrich Schüller, and Mario M. Dorostkar
- Subjects
Ependymoma ,Genetics ,Cancer Research ,Oncology ,Path (graph theory) ,DNA methylation ,medicine ,Neurology (clinical) ,Methylation ,Biology ,medicine.disease ,Molecular Pathology and Classification - Adult and Pediatric - Abstract
According to the current WHO classification, ependymal tumors are classified as subependymomas, myxopapillary ependymomas, classic ependymomas, anaplastic ependymomas and RELA-fusion positive ependymoma (RELA-EPN). Among classic ependymomas, the WHO defines rare histological variants, i.e. the clear-cell, papillary, and tanycytic ependymoma. In parallel to this WHO classification scheme, DNA methylation patterns can distinguish nine distinct molecular ependymoma subgroups, some of which tightly overlap with certain histopathological subgroups, e.g. subependyomas or myxopapillary ependymomas. Since very little is known about the molecular background of histological classic ependymoma variants, we analyzed histomorphology, clinical parameters and global DNA methylation patterns of diagnosed tanycytic ependymomas (n=12), clear-cell ependymomas (n=14) and papillary ependymomas (n=19). Surprisingly, up to 42% of these variants did not match to ependymomas using a previously published DNA methylation-based classifier for brain tumors. Among the tumors with a match to one of the nine known ependymoma methylation classes, tanycytic ependymomas were predominantly located in the spine, but showed diverse molecular methylation patterns. Most clear-cell ependymomas showed a common histomorphology, were found supratentorially and fell into the methylation class of RELA-EPN. Papillary ependymomas showed a “papillary”, “trabecular” or “pseudo-papillary” growth pattern. Interestingly, a true papillary growth pattern was strongly associated with the molecular class B of posterior fossa ependymoma (PFB), but tumors displayed DNA methylation sites that were significantly different when compared to PFB ependymomas without papillary growth. Our results show that the diagnosis of classic histological ependymoma variants can be challenging. While clear-cell and papillary ependymomas harbor common molecular features, tanycytic ependymoma may not represent a molecularly distinct subgroup.
- Published
- 2019
27. Papillary glioneuronal tumor (PGNT) exhibits a characteristic methylation profile and fusions involving PRKCA
- Author
-
Marco Prinz, Andrey Korshunov, Stefan Pusch, Daniel Schrimpf, Karin de Stricker, Leonille Schweizer, Sebastian Brandner, Werner Paulus, Mélanie Pagès, David T.W. Jones, Ingmar Blümcke, Guido Reifenberger, Andreas von Deimling, Stefan M. Pfister, Jürgen Hench, Damian Stichel, Bianca Pollo, Azadeh Ebrahimi, Pascale Varlet, Francisco Fernández-Klett, Felix Sahm, Jorge Pinheiro, David Scheie, David Capper, Karl H. Plate, Belen Casalini, Ulrich Schüller, Jochen Meier, Luca Bertero, Annekathrin Reinhardt, Yanghao Hou, Torsten Pietsch, Wolfgang Wick, David E. Reuss, Philipp Sievers, Annika K. Wefers, Christian Hartmann, Christian Koelsche, Stephan Frank, and Andreas Unterberg
- Subjects
Adult ,Male ,0301 basic medicine ,Site-Specific DNA-Methyltransferase (Adenine-Specific) ,SLC44A1 ,Protein Kinase C-alpha ,Adolescent ,Organic Cation Transport Proteins ,PRKCA ,Papillary glioneuronal tumor ,Brain tumor ,Biology ,Pathology and Forensic Medicine ,Cohort Studies ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,NOTCH1 ,Antigens, CD ,Methylation analysis ,Biomarkers, Tumor ,medicine ,Humans ,CNS TUMORS ,Child ,DNA methylation ,RNA sequencing ,Pilocytic astrocytoma ,Brain Neoplasms ,Dysembryoplastic Neuroepithelial Tumor ,Brain ,Methylation ,Middle Aged ,medicine.disease ,Neoplasms, Neuroepithelial ,030104 developmental biology ,Cancer research ,Female ,Neurology (clinical) ,Gene Fusion ,030217 neurology & neurosurgery - Abstract
Papillary glioneuronal tumor (PGNT) is a WHO-defined brain tumor entity that poses a major diagnostic challenge. Recently, SLC44A1-PRKCA fusions have been described in PGNT. We subjected 28 brain tumors from different institutions histologically diagnosed as PGNT to molecular and morphological analysis. Array-based methylation analysis revealed that 17/28 tumors exhibited methylation profiles typical for other tumor entities, mostly dysembryoplastic neuroepithelial tumor and hemispheric pilocytic astrocytoma. Conversely, 11/28 tumors exhibited a unique profile, thus constituting a distinct methylation class PGNT. By screening the extended Heidelberg cohort containing over 25,000 CNS tumors, we identified three additional tumors belonging to this methylation cluster but originally histologically diagnosed otherwise. RNA sequencing for the detection of SLC44A1-PRKCA fusions could be performed on 19 of the tumors, 10 of them belonging to the methylation class PGNT. In two additional cases, SLC44A1-PRKCA fusions were confirmed by FISH. We detected fusions involving PRKCA in all cases of this methylation class with material available for analyses: the canonical SLC44A1-PRKCA fusion was observed in 11/12 tumors, while the remaining case exhibited a NOTCH1-PRKCA fusion. Neither of the fusions was found in the tumors belonging to other methylation classes. Our results point towards a high misclassification rate of the morphological diagnosis PGNT and clearly demonstrate the necessity of molecular analyses. PRKCA fusions are highly diagnostic for PGNT, and detection by RNA sequencing enables the identification of rare fusion partners. Methylation analysis recognizes a unique methylation class PGNT irrespective of the nature of the PRKCA fusion.
- Published
- 2019
28. Isomorphic diffuse glioma is a morphologically and molecularly distinct tumour entity with recurrent gene fusions of MYBL1 or MYB and a benign disease course
- Author
-
Ingmar Blümcke, Ute Pohl, Olinda Rebelo, Andrey Korshunov, Ori Staszewski, Jochen Meyer, Thomas S. Jacques, Zane Jaunmuktane, Stefan M. Pfister, Eleonora Aronica, Anna Sophia Japp, Dina Marnoto, Daniel Schrimpf, David T.W. Jones, Daniel Schwarz, Arnault Tauziède-Espariat, Jens Schittenhelm, Ruth G. Tatevossian, Kristin Huang, José Pimentel, David Capper, Edmund Cheesman, Annekathrin Reinhardt, Martin Hasselblatt, Azadeh Ebrahimi, Damian Stichel, Abhijit Joshi, Keith L. Ligon, Maria Thom, David E. Reuss, Roland Coras, Sebastian Brandner, Elvis Terci Valera, Christian Koelsche, Pascale Varlet, M. Belén Casalini, Volkmar Hans, Philipp Sievers, Rosane G.P. Queiroz, Adriana Olar, Felix Sahm, Hu Liang Low, Michel Mittelbronn, Ekkehard Hewer, Annika K. Wefers, Mrinalini Honavar, Andreas von Deimling, Albert J. Becker, David W. Ellison, Figen Soylemezoglu, Ricardo Santos de Oliveira, Mélanie Pagès, Repositório da Universidade de Lisboa, Pathology, ANS - Cellular & Molecular Mechanisms, APH - Aging & Later Life, and APH - Mental Health
- Subjects
Adult ,Male ,0301 basic medicine ,Pathology ,medicine.medical_specialty ,DNA Copy Number Variations ,MYBL1 ,MYB ,PROTEÍNAS PROTO-ONCOGÊNICAS ,Biology ,Article ,Pathology and Forensic Medicine ,OLIG2 ,Fusion gene ,Proto-Oncogene Proteins c-myb ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Diffuse Glioma ,0302 clinical medicine ,Diffuse Astrocytoma ,Proto-Oncogene Proteins ,Glioma ,medicine ,Humans ,Oncogene Fusion ,610 Medicine & health ,Child ,Gene ,Epilepsy ,Brain Neoplasms ,DNA Methylation ,Middle Aged ,Isomorphic diffuse glioma ,medicine.disease ,030104 developmental biology ,Child, Preschool ,DNA methylation ,Trans-Activators ,570 Life sciences ,biology ,Female ,Neurology (clinical) ,030217 neurology & neurosurgery ,Gene fusion - Abstract
© Springer-Verlag GmbH Germany, part of Springer Nature 2019., The "isomorphic subtype of diffuse astrocytoma" was identified histologically in 2004 as a supratentorial, highly differentiated glioma with low cellularity, low proliferation and focal diffuse brain infiltration. Patients typically had seizures since childhood and all were operated on as adults. To define the position of these lesions among brain tumours, we histologically, molecularly and clinically analysed 26 histologically prototypical isomorphic diffuse gliomas. Immunohistochemically, they were GFAP-positive, MAP2-, OLIG2- and CD34-negative, nuclear ATRX-expression was retained and proliferation was low. All 24 cases sequenced were IDH-wildtype. In cluster analyses of DNA methylation data, isomorphic diffuse gliomas formed a group clearly distinct from other glial/glio-neuronal brain tumours and normal hemispheric tissue, most closely related to paediatric MYB/MYBL1-altered diffuse astrocytomas and angiocentric gliomas. Half of the isomorphic diffuse gliomas had copy number alterations of MYBL1 or MYB (13/25, 52%). Gene fusions of MYBL1 or MYB with various gene partners were identified in 11/22 (50%) and were associated with an increased RNA-expression of the respective MYB-family gene. Integrating copy number alterations and available RNA sequencing data, 20/26 (77%) of isomorphic diffuse gliomas demonstrated MYBL1 (54%) or MYB (23%) alterations. Clinically, 89% of patients were seizure-free after surgery and all had a good outcome. In summary, we here define a distinct benign tumour class belonging to the family of MYB/MYBL1-altered gliomas. Isomorphic diffuse glioma occurs both in children and adults, has a concise morphology, frequent MYBL1 and MYB alterations and a specific DNA methylation profile. As an exclusively histological diagnosis may be very challenging and as paediatric MYB/MYBL1-altered diffuse astrocytomas may have the same gene fusions, we consider DNA methylation profiling very helpful for their identification., This study was supported by an International League against Epilepsy (ILAE) Grant to D. Capper. I. Blumcke was supported by the European Union (FP6 DESIRE Grant Agreement #602531). M. Mittelbronn would like to thank the Luxembourg National Research Fund (FNR) for the support (FNR PEARL P16/BM/11192868 grant). K. Ligon is funded by the Paediatric Low Grade Astrocytoma Foundation and NCI R01CA215489. T. Jacques is funded by The Brain Tumour Charity, Children with Cancer UK, Great Ormond Street Hospital Children’s Charity, Cancer Research UK, the Olivia Hodson Cancer Fund and the National Institute of Health Research. T. Jacques’s work is partly funded by the NIHR GOSH BRC. The views expressed are those of the authors and not necessarily those of the NHS, the NIHR or the Department of Health. We thank the biomedical scientists in the Division of Neuropathology, the National Hospital for Neurology and Neurosurgery (NHNN) for excellent technical assistance. We also thank clinicians and neuropathologists for referring cases for molecular analysis. Part of the study was funded by the National Institute for Health Research to UCLH Biomedical research centre (BRC399/NS/RB/101410). S. Brandner and Z. Jaunmuktane are also supported by the Department of Health’s NIHR Biomedical Research Centre’s funding scheme. Additional funding was provided by the Brain Tumour Charity (UK) for the Everest Centre for Paediatric Low-Grade Brain Tumour Research
- Published
- 2019
29. LGG-35. FUNCTIONAL GENOMIC APPROACHES TO IDENTIFY THERAPEUTIC TARGETS IN MYB AND MYBL1 EXPRESSING PEDIATRIC LOW-GRADE GLIOMAS
- Author
-
David E. Root, Clauda L Kleinman, Selin Jessa, Amy Goodale, Jessica W Tsai, Federica Piccioni, Cecile Rouleau, Pratiti Bandopadhayay, Jeyna Doshi, Alexandra-Larisa Condurat, Annika K. Wefers, Prasidda Khadka, Graham Buchan, David T.W. Jones, Nicole S. Persky, Nada Jabado, Tanaz Abid, Elizabeth Gonzalez, Rameen Beroukhim, and Keith L. Ligon
- Subjects
Cancer Research ,Multicatalytic endopeptidase complex ,business.industry ,MTOR Serine-Threonine Kinases ,Astrocytoma ,Low Grade Glioma ,Cell cycle ,medicine.disease ,Genome ,Oncology ,Glioma ,Cancer research ,Medicine ,CRISPR ,AcademicSubjects/MED00300 ,MYB ,AcademicSubjects/MED00310 ,Neurology (clinical) ,business - Abstract
AIM Recurrent structural variants involving MYB and MYBL1 transcription factors were recently identified in pediatric low-grade gliomas (pLGGs), such as the MYB-QKI rearrangement in Angiocentric Gliomas and truncations of MYBL1 (MYBL1tr) in Diffuse Astrocytomas. However, therapeutic dependencies induced by these alterations remain unexplored. METHODOLOGY We have generated in vitro pLGG mouse neural stem cell (NSCs) models engineered to harbor distinct MYB/MYBL1 genomic alterations. We used single cell RNA sequencing approaches to determine the transcriptional profile and dissect the central regulatory networks of our in vitro pLGG models over time. To identify specific genetic dependencies associated with MYB/MYBL1 mutations, we employed the Brie genome-wide mouse CRISPR lentiviral knockout pooled library, consisting of 78,637 single guide RNAs (sgRNAs) targeting 19,674 mouse genes. RESULTS MYB/MYBL1 expression in neural stem cells induced activation of cell-cycle related, glioma-related and senescence-related pathways that are involved in normal development, including activation of MAPK and mTOR signaling which are also activated in human pLGG samples. Genome-scale CRISPR-cas9 screens in isogenic NSCs expressing MYB-QKI or MYBL1tr identified differential genetic dependencies relative to GFP controls. These included regulators of cell-cycle progression and several modulators of the ubiquitin-proteasome degradation pathway. Analysis of RNA-sequencing data from human tumors revealed several of these dependencies identified in the cell line model to be differentially expressed in MYB-altered pLGG tumors relative to normal brain. CONCLUSION Expression of MYB family alterations induces expression of key developmental and oncogenic pathways and genetic dependencies that represent potential therapeutic targets for MYB or MYBL1 rearranged pLGGs.
- Published
- 2020
30. MODL-11. COMPARISON OF HUMAN & MURINE PA/PXA CHARACTERISTICS
- Author
-
Andrey Korshunov, Jan Gronych, Alexander C Sommerkamp, Andrea Wittmann, David T.W. Jones, Britta Ismer, Pengbo Sun, Kathrin Schramm, Natalie Jäger, Stefan M. Pfister, Andreas von Deimling, and Annika K. Wefers
- Subjects
Cancer Research ,Oncology ,AcademicSubjects/MED00300 ,AcademicSubjects/MED00310 ,Neurology (clinical) ,Preclinical Models/Experimental Therapy/Drug Discovery - Abstract
Pediatric low-grade gliomas (pLGGs) are the most common brain tumors in children. Despite recent advances in the molecular characterization of this heterogeneous set of tumors, the separation of specific tumor types is still not fully established. Pilocytic astrocytoma (PA; WHO grade I) and pleomorphic xanthoastrocytoma (PXA; WHO grade II) are two pLGG types that can be difficult to distinguish based on histology alone. Even though their clinical course is different, they are often grouped as ‘pLGG’ in clinical trials (and therefore treated similarly). Based on a cohort of 89 human pediatric tumor samples, we show that PAs and PXAs have clearly distinct methylation and transcriptome profiles. The difference in gene expression is mainly caused by cell cycle- and development-associated genes, suggesting a key difference in the regulatory circuits involved in tumor growth. In addition to BRAF V600E, we found NTRK fusions and a previously unknown EGFR:BRAF fusion as mutually exclusive driving events in PXAs. Both tumor types show marked signs of immune cell infiltration, but with significant qualitative differences, which might represent therapeutic vulnerabilities. To pave the way for further research on PA and PXA, we developed corresponding mouse models using the virus-based RCAS system, which allows introduction of an oncogenic driver into immunocompetent mice for molecular and preclinical research. The murine tumors do not only histologically resemble their human counterparts but also show a similar growth behavior. Expression analysis revealed that the murine PXAs have a stronger gene signature of proliferation and immune cell infiltration compared to PAs.
- Published
- 2020
31. Overexpression of Lin28b in Neural Stem Cells is Insufficient for Brain Tumor Formation, but Induces Pathological Lobulation of the Developing Cerebellum
- Author
-
Annika K. Wefers, Sven Lindner, Johannes H. Schulte, and Ulrich Schüller
- Subjects
0301 basic medicine ,Cerebellum ,Mice, 129 Strain ,PAX6 Transcription Factor ,Carcinogenesis ,Medizin ,Mice, Transgenic ,Nerve Tissue Proteins ,Biology ,Nestin ,03 medical and health sciences ,Neural Stem Cells ,medicine ,Animals ,Neurons ,Medulloblastoma ,Brain Neoplasms ,PAX2 Transcription Factor ,Nuclear Proteins ,RNA-Binding Proteins ,medicine.disease ,Granule cell ,Immunohistochemistry ,Neural stem cell ,Cell biology ,DNA-Binding Proteins ,Mice, Inbred C57BL ,Phenotype ,030104 developmental biology ,CXCL3 ,medicine.anatomical_structure ,nervous system ,Neurology ,Cerebellar vermis ,Neurology (clinical) ,PAX6 ,Neuroscience - Abstract
LIN28B is a homologue of the RNA-binding protein LIN28A and regulates gene expression during development and carcinogenesis. It is strongly upregulated in a variety of brain tumors, such as medulloblastoma, embryonal tumor with multilayered rosettes (ETMR), atypical teratoid/rhabdoid tumor (AT/RT), or glioblastoma, but the effect of an in vivo overexpression of LIN28B on the developing central nervous system is unknown. We generated transgenic mice that either overexpressed Lin28b in Math1-positive cerebellar granule neuron precursors or in a broad range of Nestin-positive neural precursors. Sections of the cerebellar vermis from adult Math1-Cre::lsl-Lin28b mice had an additional subfissure in lobule IV. Vermes from p0 and p7 Nestin-Cre::lsl-Lin28b mice appeared normal, but we found a pronounced vermal hypersublobulation at p15 and p21 in these mice. Also, the external granule cell layer (EGL) was thicker at p15 than in controls, contained more proliferating cells, and persisted up to p21. Consistently, some Pax6- and NeuN-positive cells were present in the EGL of Nestin-Cre::lsl-Lin28b mice even at p21, and we detected more NeuN-positive granule neuron precursors in the molecular layer (ML) as compared to control. Finally, we found some residual Pax2-positive precursors of inhibitory interneurons in the ML of Nestin-Cre::lsl-Lin28b mice at p21, which have already disappeared in controls. We conclude that while overexpression of LIN28B in Nestin-positive cells does not lead to tumor formation, it results in a protracted development of granule cells and inhibitory interneurons and leads to a hypersublobulation of the cerebellar vermis.
- Published
- 2016
32. Mutational patterns and regulatory networks in epigenetic subgroups of meningioma
- Author
-
Hans-Georg Wirsching, Rachel Grossmann, Johannes Werner, Philipp Sievers, Ralf Ketter, Stephanie Brehmer, Matthias Schlesner, Andreas von Deimling, Umut H. Toprak, Nagarajan Paramasivam, Stefan M. Pfister, David T.W. Jones, Daniel Schrimpf, Marian Christoph Neidert, Felix Sahm, Annekathrin Reinhardt, Annika K. Wefers, Zvi Ram, Wolfgang Wick, Sebastian Uhrig, David E. Reuss, Matthias Schick, Daniel Hänggi, Roland Eils, Daniel Hübschmann, Michael Weller, Katja Beck, Melanie Bewerunge-Hudler, Damian Stichel, Marcel Seiz-Rosenhagen, Naveed Ishaque, Zuguang Gu, Christel Herold-Mende, and Steffi Urbschat
- Subjects
0301 basic medicine ,Genome instability ,Chromatin Immunoprecipitation ,Neoplasms, Radiation-Induced ,Gene Dosage ,Biology ,Polymorphism, Single Nucleotide ,Genomic Instability ,Pathology and Forensic Medicine ,Meningioma ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,otorhinolaryngologic diseases ,medicine ,Meningeal Neoplasms ,Humans ,ddc:610 ,Epigenetics ,RNA, Messenger ,RNA, Neoplasm ,Hox gene ,Gene ,Genetics ,Whole genome sequencing ,Regulation of gene expression ,Whole Genome Sequencing ,Recombinational DNA Repair ,DNA, Neoplasm ,DNA Methylation ,medicine.disease ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030104 developmental biology ,DNA methylation ,Mutation ,Neurology (clinical) ,Transcriptome ,Sequence Alignment ,030217 neurology & neurosurgery ,Transcription Factors - Abstract
DNA methylation patterns delineate clinically relevant subgroups of meningioma. We previously established the six meningioma methylation classes (MC) benign 1-3, intermediate A and B, and malignant. Here, we set out to identify subgroup-specific mutational patterns and gene regulation. Whole genome sequencing was performed on 62 samples across all MCs and WHO grades from 62 patients with matched blood control, including 40 sporadic meningiomas and 22 meningiomas arising after radiation (Mrad). RNA sequencing was added for 18 of these cases and chromatin-immunoprecipitation for histone H3 lysine 27 acetylation (H3K27ac) followed by sequencing (ChIP-seq) for 16 samples. Besides the known mutations in meningioma, structural variants were found as the mechanism of NF2 inactivation in a small subset (5%) of sporadic meningiomas, similar to previous reports for Mrad. Aberrations of DMD were found to be enriched in MCs with NF2 mutations, and DMD was among the most differentially upregulated genes in NF2 mutant compared to NF2 wild-type cases. The mutational signature AC3, which has been associated with defects in homologous recombination repair (HRR), was detected in both sporadic meningioma and Mrad, but widely distributed across the genome in sporadic cases and enriched near genomic breakpoints in Mrad. Compared to the other MCs, the number of single nucleotide variants matching the AC3 pattern was significantly higher in the malignant MC, which also exhibited higher genomic instability, determined by the numbers of both large segments affected by copy number alterations and breakpoints between large segments. ChIP-seq analysis for H3K27ac revealed a specific activation of genes regulated by the transcription factor FOXM1 in the malignant MC. This analysis also revealed a super enhancer near the HOXD gene cluster in this MC, which, together with general upregulation of HOX genes in the malignant MC, indicates a role of HOX genes in meningioma aggressiveness. This data elucidates the biological mechanisms rendering different epigenetic subgroups of meningiomas, and suggests leveraging HRR as a novel therapeutic target.
- Published
- 2018
33. Review: Challenges in the histopathological classification of ganglioglioma and DNT: microscopic agreement studies and a preliminary genotype-phenotype analysis
- Author
-
Ingmar Blümcke, Hajime Miyata, Roland Coras, Angelika Mühlebner, Albert J. Becker, Thomas J Stone, David Capper, Eleonora Aronica, Thomas S. Jacques, Maria Thom, José Pimentel, Mrinalini Honavar, Figen Soylemezoglu, Annika K. Wefers, Pathology, ANS - Cellular & Molecular Mechanisms, APH - Aging & Later Life, and APH - Mental Health
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,Histology ,Leat ,Neuropathology ,Pathology and Forensic Medicine ,Genotype phenotype ,Ganglioglioma ,03 medical and health sciences ,Epilepsy ,0302 clinical medicine ,Physiology (medical) ,medicine ,Humans ,Epilepsy surgery ,business.industry ,Brain Neoplasms ,Treatment options ,Glioma ,medicine.disease ,Oligodendroglia ,030104 developmental biology ,Phenotype ,Neurology ,Histopathology ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Abstract
Low-grade epilepsy-associated brain tumours (LEAT) are the second most common cause for drug-resistant, focal epilepsy, that is ganglioglioma (GG) and dysembryoplastic neuroepithelial tumours (DNT). However, molecular pathogenesis, risk factors for malignant progression and their frequent association with drug-resistant focal seizures remain poorly understood. This contrasts recent progress in understanding the molecular-genetic basis and targeted treatment options in diffuse gliomas. The Neuropathology Task Force of the International League Against Epilepsy examined available literature to identify common obstacles in diagnosis and research of LEAT. Analysis of 10 published tumour series from epilepsy surgery pointed to poor inter-rater agreement for the histopathology diagnosis. The Task Force tested this hypothesis using a web-based microscopy agreement study. In a series of 30 LEAT, 25 raters from 18 countries agreed in only 40% of cases. Highest discordance in microscopic diagnosis occurred between GG and DNT variants, when oligodendroglial-like cell patterns prevail, or ganglion cells were difficult to discriminate from pre-existing neurons. Suggesting new terminology or major histopathological criteria did not satisfactorily increase the yield of histopathology agreement in four consecutive trials. To this end, the Task Force applied the WHO 2016 strategy of integrating phenotype analysis with molecular-genetic data obtained from panel sequencing and 450k methylation arrays. This strategy was helpful to distinguish DNT from GG variants in all cases. The Task Force recommends, therefore, to further develop diagnostic panels for the integration of phenotype-genotype analysis in order to reliably classify the spectrum of LEAT, carefully characterize clinically meaningful entities and make better use of published literature.
- Published
- 2018
34. Novel, improved grading system(s) for IDH-mutant astrocytic gliomas
- Author
-
Andreas Unterberg, Andrey Korshunov, Christine Jungk, David N. Louis, Hiroshi Nanjo, Annekathrin Reinhardt, Yusuke Kobayashi, Pim J. French, Damian Stichel, Ryo Nishikawa, Kazuhiko Mishima, Kristin Huang, Felix Sahm, Roger Stupp, David Capper, Atsushi Sasaki, Melanie Bewerunge-Hudler, Daniel Hänggi, Christian Koelsche, Brigitta G. Baumert, Peter Sinn, Andreas von Deimling, Martin J. van den Bent, Christel Herold-Mende, Michael Platten, Jun-ichi Adachi, Antje Wick, Stefan M. Pfister, Tomonari Suzuki, Hiroaki Shimizu, Michael Weller, Annika K. Wefers, Mitsuaki Shirahata, Oksana Absalyamova, Takahiro Ono, Andrey Golanov, Wolfgang Wick, David E. Reuss, Monika E. Hegi, David T.W. Jones, Philipp Sievers, Frank Winkler, Daniel Schrimpf, Yohei Miyake, Marina Ryzhova, Thierry Gorlia, RS: GROW - R3 - Innovative Cancer Diagnostics & Therapy, and Neurology
- Subjects
Oncology ,Male ,IDH ,GLIOBLASTOMA-MULTIFORME ,0302 clinical medicine ,Diffuse Astrocytoma ,CDKN2A ,Grading (education) ,Brain Neoplasms ,Astrocytoma ,CDKN2A/B ,Middle Aged ,Isocitrate Dehydrogenase ,Neoplasm Proteins ,Gene Expression Regulation, Neoplastic ,030220 oncology & carcinogenesis ,SURVIVAL ,Immunohistochemistry ,Female ,Algorithms ,Adult ,medicine.medical_specialty ,BRAIN-TUMORS ,Adolescent ,III GLIOMAS ,World Health Organization ,Models, Biological ,CLASSIFICATION ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,MITOTIC INDEX ,Internal medicine ,medicine ,Humans ,IMMUNOHISTOCHEMISTRY ,Cyclin-Dependent Kinase Inhibitor p16 ,Aged ,Neoplasm Grading ,Astrocytic Tumor ,business.industry ,MUTATIONS ,CENTRAL-NERVOUS-SYSTEM ,medicine.disease ,GENOMIC ANALYSIS ,Grading ,Ki-67 Antigen ,Mutation ,Neurology (clinical) ,business ,Glioblastoma ,030217 neurology & neurosurgery ,Anaplastic astrocytoma - Abstract
According to the 2016 World Health Organization Classification of Tumors of the Central Nervous System (2016 CNS WHO), IDH-mutant astrocytic gliomas comprised WHO grade II diffuse astrocytoma, IDH-mutant (AII(IDHmut)), WHO grade III anaplastic astrocytoma, IDH-mutant (AAIII(IDHmut)), and WHO grade IV glioblastoma, IDH-mutant (GBM(IDHmut)). Notably, IDH gene status has been made the major criterion for classification while the manner of grading has remained unchanged: it is based on histological criteria that arose from studies which antedated knowledge of the importance of IDH status in diffuse astrocytic tumor prognostic assessment. Several studies have now demonstrated that the anticipated differences in survival between the newly defined AII(IDHmut) and AAIII(IDHmut) have lost their significance. In contrast, GBM(IDHmut) still exhibits a significantly worse outcome than its lower grade IDH-mutant counterparts. To address the problem of establishing prognostically significant grading for IDH-mutant astrocytic gliomas in the IDH era, we undertook a comprehensive study that included assessment of histological and genetic approaches to prognosis in these tumors. A discovery cohort of 211 IDH-mutant astrocytic gliomas with an extended observation was subjected to histological review, image analysis, and DNA methylation studies. Tumor group-specific methylation profiles and copy number variation (CNV) profiles were established for all gliomas. Algorithms for automated CNV analysis were developed. All tumors exhibiting 1p/19q codeletion were excluded from the series. We developed algorithms for grading, based on molecular, morphological and clinical data. Performance of these algorithms was compared with that of WHO grading. Three independent cohorts of 108, 154 and 224 IDH-mutant astrocytic gliomas were used to validate this approach. In the discovery cohort several molecular and clinical parameters were of prognostic relevance. Most relevant for overall survival (OS) was CDKN2A/B homozygous deletion. Other parameters with major influence were necrosis and the total number of CNV. Proliferation as assessed by mitotic count, which is a key parameter in 2016 CNS WHO grading, was of only minor influence. Employing the parameters most relevant for OS in our discovery set, we developed two models for grading these tumors. These models performed significantly better than WHO grading in both the discovery and the validation sets. Our novel algorithms for grading IDH-mutant astrocytic gliomas overcome the challenges caused by introduction of IDH status into the WHO classification of diffuse astrocytic tumors. We propose that these revised approaches be used for grading of these tumors and incorporated into future WHO criteria.
- Published
- 2018
35. FGFR1:TACC1 fusion is a frequent event in molecularly defined extraventricular neurocytoma
- Author
-
Roland Coras, Elisabeth J. Rushing, Rudi Beschorner, Kristian W. Pajtler, David E. Reuss, Walter Stummer, Daniel Schrimpf, Andreas von Deimling, Stefan M. Pfister, Caterina Giannini, Christian Hagel, Felice Giangaspero, Philipp Sievers, Uta Schick, Christel Herold-Mende, Annika K. Wefers, Azadeh Ebrahimi, Patricia Kohlhof, Kristin Huang, Andrey Korshunov, Ori Staszewski, Francesca Diomedi-Camassei, David T.W. Jones, Christian Koelsche, Guido Reifenberger, Felix Sahm, Yanghao Hou, Damian Stichel, Annekathrin Reinhardt, Christian Hartmann, Martin Hasselblatt, Kathy Keyvani, Sievers P., Stichel D., Schrimpf D., Sahm F., Koelsche C., Reuss D.E., Wefers A.K., Reinhardt A., Huang K., Ebrahimi A., Hou Y., Pajtler K.W., Pfister S.M., Hasselblatt M., Stummer W., Schick U., Hartmann C., Hagel C., Staszewski O., Reifenberger G., Beschorner R., Coras R., Keyvani K., Kohlhof P., Diomedi-Camassei F., Herold-Mende C., Giangaspero F., Rushing E., Giannini C., Korshunov A., Jones D.T.W., von Deimling A., University of Zurich, and von Deimling, Andreas
- Subjects
Fetal Proteins ,Male ,0301 basic medicine ,Pathology ,Oncogene Proteins, Fusion ,2804 Cellular and Molecular Neuroscience ,Medizin ,Kaplan-Meier Estimate ,DNA methylation profile ,Histones ,0302 clinical medicine ,Retrospective Studie ,Neurocytoma ,Nuclear Protein ,Brain Neoplasms ,FGFR ,Nuclear Proteins ,Methylation ,Isocitrate Dehydrogenase ,Histone ,2728 Neurology (clinical) ,Extraventricular neurocytoma ,Molecular classification ,DNA methylation ,Female ,Microtubule-Associated Proteins ,Human ,medicine.medical_specialty ,10208 Institute of Neuropathology ,Clinical Neurology ,Brain tumor ,Copy number analysis ,610 Medicine & health ,Biology ,Pathology and Forensic Medicine ,Brain Neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Text mining ,Fetal Protein ,Parenchyma ,medicine ,Central neurocytoma ,Humans ,Receptor, Fibroblast Growth Factor, Type 1 ,Epigenetics ,Fusion ,Retrospective Studies ,business.industry ,Microtubule-Associated Protein ,DNA Methylation ,medicine.disease ,FGFR1–TACC1 ,2734 Pathology and Forensic Medicine ,Ki-67 Antigen ,030104 developmental biology ,570 Life sciences ,biology ,brain tumor ,extraventricular neurocytoma ,fusion ,molecular classification ,Neurology (clinical) ,Transcriptome ,business ,030217 neurology & neurosurgery - Abstract
Extraventricular neurocytoma (EVN) is a rare primary brain tumor occurring in brain parenchyma outside the ventricular system. Histopathological characteristics resemble those of central neurocytoma but exhibit a wider morphologic spectrum. Accurate diagnosis of these histologically heterogeneous tumors is often challenging because of the overlapping morphological features and the lack of defining molecular markers. Here, we explored the molecular landscape of 40 tumors diagnosed histologically as EVN by investigating copy number profiles and DNA methylation array data. DNA methylation profiles were compared with those of relevant differential diagnoses of EVN and with a broader spectrum of diverse brain tumor entities. Based on this, our tumor cohort segregated into different groups. While a large fraction (n = 22) formed a separate epigenetic group clearly distinct from established DNA methylation profiles of other entities, a subset (n = 14) of histologically diagnosed EVN grouped with clusters of other defined entities. Three cases formed a small group close to but separated from the epigenetically distinct EVN cases, and one sample clustered with non-neoplastic brain tissue. Four additional samples originally diagnosed otherwise were found to molecularly resemble EVN. Thus, our results highlight a distinct DNA methylation pattern for the majority of tumors diagnosed as EVN, but also indicate that approximately onethird of morphological diagnoses of EVN epigenetically correspond to other brain tumor entities. Copy number analysis and confirmation through RNA sequencing revealed FGFR1–TACC1 fusion as a distinctive, recurrent feature within the EVN methylation group (60%), in addition to a small number of other FGFR rearrangements (13%). In conclusion, our data demonstrate a specific epigenetic signature of EVN suitable for characterization of these tumors as a molecularly distinct entity, and reveal a high frequency of potentially druggable FGFR pathway activation in this tumor group.
- Published
- 2018
36. DNA methylation-based reclassification of olfactory neuroblastoma
- Author
-
Stefanie Glöss, Martin Forster, Werner Paulus, Miguel Sáinz-Jaspeado, Rolf Buslei, Camelia M. Monoranu, Jörg Felsberg, Simone Schmid, Matt Lechner, Javier Alonso, Martin Sill, David T.W. Jones, Adriana Olar, Xavier Garcia del Muro, Daniel Schrimpf, Judith Niesen, Markus Glatzel, Manel Esteller, Volker Gudziol, Guido Reifenberger, Arend Koch, Keith L. Ligon, Sebastian Moran, Ulrich Schüller, Oscar M. Tirado, Matthias Meinhardt, Nils W. Engel, Jaume Mora, Sven Perner, Stefan M. Pfister, David Capper, Oliver Weigert, Christian Koelsche, Valerie J. Lund, Damian Stichel, Stephan Frank, Andreas von Deimling, Julika Ribbat-Idel, and Annika K. Wefers
- Subjects
Adult ,Male ,0301 basic medicine ,Adolescent ,Copy number analysis ,Biology ,medicine.disease_cause ,Pathology and Forensic Medicine ,Diagnosis, Differential ,Neuroblastoma ,Olfaction Disorders ,Young Adult ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Cytokeratin ,0302 clinical medicine ,Esthesioneuroblastoma ,Biomarkers, Tumor ,medicine ,Humans ,Child ,Aged ,Aged, 80 and over ,Mutation ,CpG Island Methylator Phenotype ,Olfactory Neuroblastoma ,Chromosome ,DNA Methylation ,Middle Aged ,medicine.disease ,Isocitrate Dehydrogenase ,030104 developmental biology ,030220 oncology & carcinogenesis ,DNA methylation ,Cancer research ,Female ,Neurology (clinical) ,Transcriptome - Abstract
Olfactory neuroblastoma/esthesioneuroblastoma (ONB) is an uncommon neuroectodermal neoplasm thought to arise from the olfactory epithelium. Little is known about its molecular pathogenesis. For this study, a retrospective cohort of n = 66 tumor samples with the institutional diagnosis of ONB was analyzed by immunohistochemistry, genome-wide DNA methylation profiling, copy number analysis, and in a subset, next-generation panel sequencing of 560 tumor-associated genes. DNA methylation profiles were compared to those of relevant differential diagnoses of ONB. Unsupervised hierarchical clustering analysis of DNA methylation data revealed four subgroups among institutionally diagnosed ONB. The largest group (n = 42, 64%, Core ONB) presented with classical ONB histology and no overlap with other classes upon methylation profiling-based t-distributed stochastic neighbor embedding (t-SNE) analysis. A second DNA methylation group (n = 7, 11%) with CpG island methylator phenotype (CIMP) consisted of cases with strong expression of cytokeratin, no or scarce chromogranin A expression and IDH2 hotspot mutation in all cases. T-SNE analysis clustered these cases together with sinonasal carcinoma with IDH2 mutation. Four cases (6%) formed a small group characterized by an overall high level of DNA methylation, but without CIMP. The fourth group consisted of 13 cases that had heterogeneous DNA methylation profiles and strong cytokeratin expression in most cases. In t-SNE analysis, these cases mostly grouped among sinonasal adenocarcinoma, squamous cell carcinoma, and undifferentiated carcinoma. Copy number analysis indicated highly recurrent chromosomal changes among Core ONB with a high frequency of combined loss of chromosome 1-4, 8-10, and 12. NGS sequencing did not reveal highly recurrent mutations in ONB, with the only recurrently mutated genes being TP53 and DNMT3A. In conclusion, we demonstrate that institutionally diagnosed ONB are a heterogeneous group of tumors. Expression of cytokeratin, chromogranin A, the mutational status of IDH2 as well as DNA methylation patterns may greatly aid in the precise classification of ONB.
- Published
- 2018
37. Correction to: DNA methylation-based reclassification of olfactory neuroblastoma
- Author
-
Matt Lechner, Rolf Buslei, Adriana Olar, Volker Gudziol, Valerie J. Lund, Sven Perner, Sebastian Moran, Stephan Frank, Judith Niesen, Martin Forster, Simone Schmid, Jaume Mora, Christian Kölsche, Javier Alonso, Markus Glatzel, David T.W. Jones, Manel Esteller, Damian Stichel, Keith L. Ligon, Xavier Garcia del Muro, Werner Paulus, Oliver Weigert, Miguel Sáinz-Jaspeado, Camelia M. Monoranu, Daniel Schrimpf, Martin Sill, Matthias Meinhardt, David Capper, Andreas von Deimling, Jörg Felsberg, Stefanie Glöss, Nils W. Engel, Stefan M. Pfister, Guido Reifenberger, Arend Koch, Julika Ribbat-Idel, Annika K. Wefers, Oscar M. Tirado, and Ulrich Schüller
- Subjects
Cellular and Molecular Neuroscience ,Olfactory Neuroblastoma ,business.industry ,Published Erratum ,DNA methylation ,MEDLINE ,Medicine ,Neurology (clinical) ,Bioinformatics ,business ,Pathology and Forensic Medicine - Abstract
In the original publication, the second name of the twentieth author was incorrect. It should read as 'Miguel Sáinz-Jaspeado'. The original publication of the article has been updated to reflect the change. This correction was authored by Ulrich Schüller on behalf of all authors of the original publication.
- Published
- 2018
38. Anaplastic astrocytoma with piloid features, a novel molecular class of IDH wildtype glioma with recurrent MAPK pathway, CDKN2A/B and ATRX alterations
- Author
-
Camelia M. Monoranu, Andreas von Deimling, Albert J. Becker, Joerg Felsberg, Jens Schittenhelm, Martina Deckert, Marco Prinz, Rolf Buslei, Till Acker, Katharina Heß, Ute Pohl, Volker Hovestadt, Wolf Mueller, Patricia Kohlhof, Dorothee Gramatzki, Muin S. A. Tuffaha, Amulya NageswaraRao, Andrey Korshunov, Benjamin Brokinkel, Daniel Schrimpf, David T.W. Jones, Annekathrin Reinhardt, Ulrich W. Thomale, Werner Paulus, Ekkehard Hewer, Christian Koelsche, Christian Mawrin, Damian Stichel, Ori Staszewski, Wolfgang Wick, David E. Reuss, Almuth F. Kessler, Caterina Giannini, Annika K. Wefers, Michael Platten, Martin Sill, Daniel Hänggi, Kristin Huang, Christian Hartmann, Adriana Olar, David Capper, Volkmar Hans, Andreas Unterberg, Zane Jaunmuktane, Sebastian Brandner, Nuno Miguel Nunes, Christel Herold-Mende, Felix Sahm, Uri Tabori, Guido Reifenberger, Arend Koch, Mario Loehr, Michael Weller, Hildegard Dohmen, Stefan M. Pfister, Fausto J. Rodriguez, Reinhardt A., Stichel D., Schrimpf D., Sahm F., Korshunov A., Reuss D.E., Koelsche C., Huang K., Wefers A.K., Hovestadt V., Sill M., Gramatzki D., Felsberg J., Reifenberger G., Koch A., Thomale U.-W., Becker A., Hans V.H., Prinz M., Staszewski O., Acker T., Dohmen H., Hartmann C., Mueller W., Tuffaha M.S.A., Paulus W., Hess K., Brokinkel B., Schittenhelm J., Monoranu C.-M., Kessler A.F., Loehr M., Buslei R., Deckert M., Mawrin C., Kohlhof P., Hewer E., Olar A., Rodriguez F.J., Giannini C., NageswaraRao A.A., Tabori U., Nunes N.M., Weller M., Pohl U., Jaunmuktane Z., Brandner S., Unterberg A., Hanggi D., Platten M., Pfister S.M., Wick W., Herold-Mende C., Jones D.T.W., von Deimling A., and Capper D.
- Subjects
0301 basic medicine ,Male ,Medizin ,Kaplan-Meier Estimate ,Mitogen-Activated Protein Kinase Kinase ,Histones ,0302 clinical medicine ,CDKN2A ,Retrospective Studie ,Age Factor ,610 Medicine & health ,Child ,DNA Modification Methylases ,Aged, 80 and over ,Pilocytic astrocytoma ,Brain Neoplasms ,DNA Repair Enzyme ,Age Factors ,CDKN2A/B ,Middle Aged ,Molecular characterization ,Isocitrate Dehydrogenase ,Histone ,ATRX ,Child, Preschool ,DNA methylation ,Female ,MGMT ,Human ,Signal Transduction ,Adult ,X-linked Nuclear Protein ,IDH1 ,Adolescent ,Panel sequencing ,Biology ,Astrocytoma ,Pathology and Forensic Medicine ,BRAF ,DNA copy number alteration ,Brain Neoplasm ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Young Adult ,Glioma ,DNA Modification Methylase ,medicine ,Humans ,Gene ,neoplasms ,Cyclin-Dependent Kinase Inhibitor p16 ,Retrospective Studies ,Pilocytic astrocytoma with anaplasia ,Aged ,Mitogen-Activated Protein Kinase Kinases ,Tumor Suppressor Protein ,Methylation profile based classification ,Tumor Suppressor Proteins ,Infant ,DNA Methylation ,medicine.disease ,Anaplastic pilocytic astrocytoma ,030104 developmental biology ,DNA Repair Enzymes ,FGFR1 ,NF1 ,Mutation ,Cancer research ,570 Life sciences ,biology ,Neurology (clinical) ,030217 neurology & neurosurgery ,Anaplastic astrocytoma - Abstract
Tumors with histological features of pilocytic astrocytoma (PA), but with increased mitotic activity and additional high-grade features (particularly microvascular proliferation and palisading necrosis) have often been designated anaplastic pilocytic astrocytomas. The status of these tumors as a separate entity has not yet been conclusively demonstrated and molecular features have only been partially characterized. We performed DNA methylation profiling of 102 histologically defined anaplastic pilocytic astrocytomas. T-distributed stochastic neighbor-embedding(t-SNE) and hierarchical clustering analysis of these 102 cases against 158 reference cases from 12 glioma reference classes revealed that a subset of 83 of these tumors share a common DNA methylation profile that is distinct from the reference classes. These 83 tumors were thus denominated DNA methylation class anaplastic astrocytoma with piloid features (MC AAP). The 19 remaining tumors were distributed amongst the reference classes, with additional testing confirming the molecular diagnosis in most cases. Median age of patients with MC AAP was 41.5years. The most frequent localization was the posterior fossa (74%). Deletions of CDKN2A/B (66/83, 80%), MAPK pathway gene alterations (49/65, 75%, most frequently affecting NF1, followed by BRAF and FGFR1) and mutations of ATRX or loss of ATRX expression (33/74, 45%) were the most common molecular alterations. All tumors were IDH1/2 wildtype. The MGMT promoter was methylated in 38/83 tumors (45%). Outcome analysis confirmed an unfavorable clinical course in comparison to PA, but better than IDH wildtype glioblastoma. In conclusion, we show that a subset of histologically defined anaplastic pilocytic astrocytomas forms a separate DNA methylation cluster, harbors recurrent alterations in MAPK pathway genes in combination with alterations of CDKN2A/B and ATRX, affects patients who are on average older than those diagnosed with PA and has an intermediate clinical outcome.
- Published
- 2018
39. Gain of 12p encompassing CCND2 is associated with gemistocytic histology in IDH mutant astrocytomas
- Author
-
Damian Stichel, Matthias Schick, Daniel Schrimpf, Michel Mittelbronn, Andrey Korshunov, Felix Sahm, Andreas von Deimling, Kristin Huang, Annika K. Wefers, David Capper, Melanie Bewerunge-Hudler, Andreas Unterberg, David T.W. Jones, Astrid Jeibmann, Christel Herold-Mende, Christian Koelsche, Annekathrin Kratz, Daniel Hänggi, Michael Platten, Sebastian Brandner, Stefan M. Pfister, Wolfgang Wick, and David E. Reuss
- Subjects
0301 basic medicine ,Pathology ,medicine.medical_specialty ,DNA Copy Number Variations ,Mutant ,Astrocytoma ,Tp53 mutation ,Pathology and Forensic Medicine ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,Text mining ,medicine ,Cyclin D2 ,Humans ,Fibrillary astrocytoma ,business.industry ,Brain Neoplasms ,Histology ,medicine.disease ,Isocitrate Dehydrogenase ,Clinical neurology ,030104 developmental biology ,Mutation ,Neurology (clinical) ,business ,030217 neurology & neurosurgery - Published
- 2016
40. Poorly differentiated chordoma with SMARCB1/INI1 loss: a distinct molecular entity with dismal prognosis
- Author
-
Pascal Johann, Yvonne Crede, Nasir Ud Din, Frank van Landeghem, Volker Hovestadt, David Capper, Annika K. Wefers, Susanne Peetz-Dienhart, Felice Giangaspero, Marcel Kool, Arie Perry, Daniel Schrimpf, Manila Antonelli, Reiner Siebert, Markus J. Riemenschneider, Stefan M. Pfister, David Sumerauer, Hannes Vogel, Christian Thomas, Michael C. Frühwald, Florian Oyen, Caterina Giannini, Susanne Bens, Andrey Korshunov, Peter Hauser, Reinhard Schneppenheim, David T.W. Jones, Marie Christine Bernardo, Kathy Keyvani, Martin Hasselblatt, Hasselblatt M., Thomas C., Hovestadt V., Schrimpf D., Johann P., Bens S., Oyen F., Peetz-Dienhart S., Crede Y., Wefers A., Vogel H., Riemenschneider M.J., Antonelli M., Giangaspero F., Bernardo M.C., Giannini C., Ud Din N., Perry A., Keyvani K., van Landeghem F., Sumerauer D., Hauser P., Capper D., Korshunov A., Jones D.T.W., Pfister S.M., Schneppenheim R., Siebert R., Fruhwald M.C., and Kool M.
- Subjects
tumors ,Male ,DNA Copy Number Variations ,Prognosi ,neurology (clinical) ,SMARCB1 ,cellular and molecular neuroscience ,chordomas ,skull base and spine ,Medizin ,Pathology and Forensic Medicine ,Brain Neoplasm ,Diagnosis, Differential ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,0302 clinical medicine ,medicine ,Biomarkers, Tumor ,Chordoma ,Cluster Analysis ,Humans ,Child ,Rhabdoid Tumor ,Molecular entity ,DNA Copy Number Variation ,Cluster Analysi ,business.industry ,Brain Neoplasms ,Poorly differentiated ,SMARCB1 Protein ,Teratoma ,Infant ,DNA Methylation ,medicine.disease ,Prognosis ,Smarcb1 ini1 ,Survival Analysis ,030220 oncology & carcinogenesis ,Child, Preschool ,Cancer research ,Female ,Neurology (clinical) ,Survival Analysi ,Differential diagnosis ,business ,030217 neurology & neurosurgery ,Human - Abstract
Lettera no abstract
- Published
- 2016
41. Subgroup-specific localization of human medulloblastoma based on pre-operative MRI
- Author
-
Torsten Pietsch, André O. von Bueren, Camelia-Maria Monoranu, Aurelia Peraud, Christian Mawrin, Arend Koch, Klaus Seelos, Antoinette Y. N. Schouten-van Meeteren, Stefan Rutkowski, Dannis G. van Vuurden, Ulrich Schüller, Annika K. Wefers, Jörg-Christian Tonn, Monika Warmuth-Metz, Julia Pöschl, Christel Herold-Mende, Marcel Kool, Stefan M. Pfister, Katja von Hoff, Pediatric surgery, CCA - Innovative therapy, Cancer Center Amsterdam, Amsterdam Public Health, Paediatric Oncology, and Other departments
- Subjects
Adult ,Male ,Oncology ,medicine.medical_specialty ,Adolescent ,MEDLINE ,Pathology and Forensic Medicine ,Cohort Studies ,Young Adult ,Cellular and Molecular Neuroscience ,Text mining ,Internal medicine ,medicine ,Humans ,Young adult ,Child ,Medulloblastoma ,Intraoperative Care ,Brain Neoplasms ,business.industry ,Brain ,Infant ,medicine.disease ,Magnetic Resonance Imaging ,Pre operative ,Child, Preschool ,Female ,Neurology (clinical) ,business ,Cohort study - Published
- 2014
42. HG-68COMBINED ALTERATIONS IN MAPK PATHWAY GENES, CDKN2A/B AND ATRX CHARACTERIZE ANAPLASTIC PILOCYTIC ASTROCYTOMA
- Author
-
Martina Deckert, Hildegard Dohmen-Scheufler, Andreas von Deimling, Albert J. Becker, Ulrich W. Thomale, Werner Paulus, Jens Schittenhelm, Caterina Giannini, Almuth F. Keßler, Wolf Mueller, Michael Weller, Sebastian Brandner, David T.W. Jones, Patricia Kohlhof, Christian Mawrin, Amulya NageswaraRao, Camelia M. Monoranu, Christian Hartmann, Muin S. A. Tuffaha, Stefan M. Pfister, Ute Pohl, Dorothee Gramatzki, Kristin Huang, Adriana Olar, David Reuß, Ekkehard Hewer, Fausto J. Rodriguez, Christian Kölsche, David Capper, Rolf Buslei, Marco Prinz, Jörg Felsberg, Annika K. Wefers, Annekathrin Kratz, Katharina Heß, Ori Staszewski, Till Acker, Volkmar Hans, Guido Reifenberger, Arend Koch, Volker Hovestadt, Daniel Schrimpf, Benjamin Brokinkel, and Felix Sahm
- Subjects
MAPK/ERK pathway ,Cancer Research ,Pathology ,medicine.medical_specialty ,Necrosis ,Pilocytic astrocytoma ,Biology ,Cdkn2a b ,medicine.disease ,nervous system diseases ,Abstracts ,nervous system ,Oncology ,medicine ,Cancer research ,Microvascular Proliferation ,Neurology (clinical) ,medicine.symptom ,neoplasms ,Gene ,Mitosis ,ATRX - Abstract
Introduction: Tumors with histological features of pilocytic astrocytoma but with increased mitotic activity and additional high grade features (i.e. microvascular proliferation, necrosis) have been designated anaplastic pilocytic astrocytomas (APA). Patients with such tumors are thought to have an [for full text, please go to the a.m. URL]
- Published
- 2016
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.