40 results on '"M Kool"'
Search Results
2. SWI/SNF-deficient tumors of the central nervous system: An update.
- Author
-
Hasselblatt M, Kool M, and Frühwald MC
- Subjects
- Humans, SMARCB1 Protein genetics, Central Nervous System pathology, DNA Helicases genetics, Nuclear Proteins genetics, Transcription Factors genetics, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Neoplasms, Neuroepithelial
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant tumor of the central nervous system characterized by biallelic inactivation of SWI/SNF chromatin remodeling complex members SMARCB1/INI1 or (rarely) SMARCA4/BRG1. Most high-grade central nervous system lesions showing loss of nuclear SMARCB1 or SMARCA4 protein expression can indeed be categorized as AT/RT. However, some high-grade lesions have been identified, whose clinical and/or molecular features justify separation from AT/RT. Furthermore, other recently described tumor types such as desmoplastic myxoid tumor, SMARCB1-mutant, and low-grade diffusely infiltrative tumor, SMARCB1-mutant, may even manifest as low-grade lesions. Here, we review recent developments in the definition of the molecular landscape of AT/RT and give an update on other rare high- and low-grade SWI/SNF-deficient central nervous system tumors.
- Published
- 2024
- Full Text
- View/download PDF
3. Recurrent atypical teratoid/rhabdoid tumors (AT/RT) reveal discrete features of progression on histology, epigenetics, copy number profiling, and transcriptomics.
- Author
-
Johann PD, Altendorf L, Efremova EM, Holsten T, Steinbügl M, Nemes K, Eckhardt A, Kresbach C, Bockmayr M, Koch A, Haberler C, Antonelli M, DeSisto J, Schuhmann MU, Hauser P, Siebert R, Bens S, Kool M, Green AL, Hasselblatt M, Frühwald MC, and Schüller U
- Subjects
- Child, Child, Preschool, Female, Humans, Infant, Male, Chromosomes, Human, Pair 1 genetics, Chromosomes, Human, Pair 10 genetics, Cohort Studies, Dendritic Cells, DNA Methylation, Histology, Mitosis, Sequence Analysis, RNA, Transcription Factors genetics, Gene Expression Regulation, Neoplastic genetics, Disease Progression, DNA Copy Number Variations genetics, Epigenesis, Genetic, Gene Expression Profiling, Recurrence, Rhabdoid Tumor classification, Rhabdoid Tumor genetics, Rhabdoid Tumor immunology, Rhabdoid Tumor pathology, Teratoma classification, Teratoma genetics, Teratoma immunology, Teratoma pathology
- Abstract
Atypical teratoid/rhabdoid tumors (AT/RT) are the most common malignant brain tumors manifesting in infancy. They split into four molecular types. The major three (AT/RT-SHH, AT/RT-TYR, and AT/RT-MYC) all carry mutations in SMARCB1, the fourth quantitatively smaller type is characterized by SMARCA4 mutations (AT/RT-SMARCA4). Molecular characteristics of disease recurrence or metastatic spread, which go along with a particularly dismal outcome, are currently unclear. Here, we investigated tumor tissue from 26 patients affected by AT/RT to identify signatures of recurrences in comparison with matched primary tumor samples. Microscopically, AT/RT recurrences demonstrated a loss of architecture and significantly enhanced mitotic activity as compared to their related primary tumors. Based on DNA methylation profiling, primary tumor and related recurrence were grossly similar, but three out of 26 tumors belonged to a different molecular type or subtype after second surgery compared to related primary lesions. Copy number variations (CNVs) differed in six cases, showing novel gains on chromosome 1q or losses of chromosome 10 in recurrences as the most frequent alterations. To consolidate these observations, our cohort was combined with a data set of unmatched primary and recurrent AT/RT, which demonstrated chromosome 1q gain and 10 loss in 18% (n = 7) and 11% (n = 4) of the recurrences (n = 38) as compared to 7% (n = 3) and 0% (n = 0) in the primary tumors (n = 44), respectively. Similar to the observations made by DNA methylation profiling, RNA sequencing of our cohort revealed AT/RT primary tumors and matched recurrences clustering closely together. However, a number of genes showed significantly altered expression in AT/RT-SHH recurrences. Many of them are known tumor driving growth factors, involved in embryonal development and tumorigenesis, or are cell-cycle-associated. Overall, our work identifies subtle molecular changes that occur in the course of the disease and that may help define novel therapeutic targets for AT/RT recurrences., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
4. Atypical teratoid/rhabdoid tumoroids reveal subgroup-specific drug vulnerabilities.
- Author
-
Paassen I, Williams J, Ríos Arceo C, Ringnalda F, Mercer KS, Buhl JL, Moreno N, Federico A, Franke NE, Kranendonk M, Upadhyaya SA, Kerl K, van de Wetering M, Clevers H, Kool M, Hoving EW, Roussel MF, and Drost J
- Subjects
- Child, Humans, SMARCB1 Protein genetics, Receptors, Notch, Epigenomics, DNA Helicases, Nuclear Proteins, Transcription Factors genetics, Teratoma drug therapy, Teratoma genetics, Brain Neoplasms drug therapy, Brain Neoplasms genetics, Rhabdoid Tumor drug therapy, Rhabdoid Tumor genetics, Rhabdoid Tumor metabolism
- Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) represent a rare, but aggressive pediatric brain tumor entity. They are genetically defined by alterations in the SWI/SNF chromatin remodeling complex members SMARCB1 or SMARCA4. ATRTs can be further classified in different molecular subgroups based on their epigenetic profiles. Although recent studies suggest that the different subgroups have distinct clinical features, subgroup-specific treatment regimens have not been developed thus far. This is hampered by the lack of pre-clinical in vitro models representative of the different molecular subgroups. Here, we describe the establishment of ATRT tumoroid models from the ATRT-MYC and ATRT-SHH subgroups. We demonstrate that ATRT tumoroids retain subgroup-specific epigenetic and gene expression profiles. High throughput drug screens on our ATRT tumoroids revealed distinct drug sensitivities between and within ATRT-MYC and ATRT-SHH subgroups. Whereas ATRT-MYC universally displayed high sensitivity to multi-targeted tyrosine kinase inhibitors, ATRT-SHH showed a more heterogeneous response with a subset showing high sensitivity to NOTCH inhibitors, which corresponded to high expression of NOTCH receptors. Our ATRT tumoroids represent the first pediatric brain tumor organoid model, providing a representative pre-clinical model which enables the development of subgroup-specific therapies., (© 2023. The Author(s).)
- Published
- 2023
- Full Text
- View/download PDF
5. Primary cilia contribute to the aggressiveness of atypical teratoid/rhabdoid tumors.
- Author
-
Blümel L, Qin N, Berlandi J, Paisana E, Cascão R, Custódia C, Pauck D, Picard D, Langini M, Stühler K, Meyer FD, Göbbels S, Malzkorn B, Liebau MC, Barata JT, Jeibmann A, Kerl K, Erkek S, Kool M, Pfister SM, Johann PD, Frühwald MC, Borkhardt A, Reifenberger G, Faria CC, Fischer U, Hasselblatt M, Bartl J, and Remke M
- Subjects
- Animals, Cilia metabolism, DNA Helicases metabolism, Humans, Mice, Nuclear Proteins metabolism, Signal Transduction, Transcription Factors genetics, Transcription Factors therapeutic use, Brain Neoplasms genetics, Rhabdoid Tumor genetics, Rhabdoid Tumor metabolism, Rhabdoid Tumor pathology, Teratoma genetics, Teratoma pathology
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant brain tumor in infants that is characterized by loss of nuclear expression of SMARCB1 or SMARCA4 proteins. Recent studies show that AT/RTs comprise three molecular subgroups, namely AT/RT-TYR, AT/RT-MYC and AT/RT-SHH. The subgroups show distinct expression patterns of genes involved in ciliogenesis, however, little is known about the functional roles of primary cilia in the biology of AT/RT. Here, we show that primary cilia are present across all AT/RT subgroups with specific enrichment in AT/RT-TYR patient samples. Furthermore, we demonstrate that primary ciliogenesis contributes to AT/RT biology in vitro and in vivo. Specifically, we observed a significant decrease in proliferation and clonogenicity following disruption of primary ciliogenesis in AT/RT cell line models. Additionally, apoptosis was significantly increased via the induction of STAT1 and DR5 signaling, as detected by proteogenomic profiling. In a Drosophila model of SMARCB1 deficiency, concomitant knockdown of several cilia-associated genes resulted in a substantial shift of the lethal phenotype with more than 20% of flies reaching adulthood. We also found significantly extended survival in an orthotopic xenograft mouse model of AT/RT upon disruption of primary ciliogenesis. Taken together, our findings indicate that primary ciliogenesis or its downstream signaling contributes to the aggressiveness of AT/RT and, therefore, may constitute a novel therapeutic target., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
6. SMARCB1-deficient and SMARCA4-deficient Malignant Brain Tumors With Complex Copy Number Alterations and TP53 Mutations May Represent the First Clinical Manifestation of Li-Fraumeni Syndrome.
- Author
-
Hasselblatt M, Thomas C, Federico A, Nemes K, Johann PD, Bison B, Bens S, Dahlum S, Kordes U, Redlich A, Lessel L, Pajtler KW, Mawrin C, Schüller U, Nolte K, Kramm CM, Hinz F, Sahm F, Giannini C, Penkert J, Kratz CP, Pfister SM, Siebert R, Paulus W, Kool M, and Frühwald MC
- Subjects
- Child, DNA Copy Number Variations, DNA Helicases genetics, DNA Helicases metabolism, Humans, Mutation, Nuclear Proteins genetics, Nuclear Proteins metabolism, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Transcription Factors genetics, Transcription Factors metabolism, Tumor Suppressor Protein p53 genetics, Brain Neoplasms complications, Brain Neoplasms genetics, Li-Fraumeni Syndrome complications, Li-Fraumeni Syndrome genetics, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a malignant central nervous system tumor predominantly affecting infants. Mutations of SMARCB1 or (rarely) SMARCA4 causing loss of nuclear SMARCB1 or SMARCA4 protein expression are characteristic features, but further recurrent genetic alterations are lacking. Most AT/RTs occur de novo, but secondary AT/RTs arising from other central nervous system tumors have been reported. Malignant gliomas, IDH wild-type, arising in patients with Li-Fraumeni syndrome typically show somatic mutations of TP53 as well as complex copy number alterations, but little is known about the loss of SMARCB1 or SMARCA4 protein expression in this context. Here, we report 2 children in whom malignant supratentorial brain tumors with SMARCB1 deficiency, complex copy number alterations, and somatic TP53 mutations lead to the discovery of pathogenic/likely pathogenic TP53 variants in the germline. Screening of the molecularneuropathology.org dataset for cases with similar genetic and epigenetic alterations yielded another case with SMARCA4 deficiency in a young adult with Li-Fraumeni syndrome. In conclusion, SMARCB1-deficient or SMARCA4-deficient malignant brain tumors with complex copy number alterations and somatic TP53 mutations in children and young adults may represent the first clinical manifestation of Li-Fraumeni syndrome and should prompt genetic counseling and investigation for TP53 germline status., Competing Interests: Conflicts of Interest and Source of Funding: Supported by IZKF Münster (Ha3/017/20). C.P.K. has been supported by the Deutsche Kinderkrebsstiftung (DKS2019.13) and BMBF ADDRess (01GM1909A). B.B. is supported by the Deutsche Kinderkrebsstiftung (DKS 2020.05). M.C.F. and R.S. are supported Deutsche Krebshilfe (DKH 70113981, 70114040). The authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article., (Copyright © 2022 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2022
- Full Text
- View/download PDF
7. ATRT-SHH comprises three molecular subgroups with characteristic clinical and histopathological features and prognostic significance.
- Author
-
Federico A, Thomas C, Miskiewicz K, Woltering N, Zin F, Nemes K, Bison B, Johann PD, Hawes D, Bens S, Kordes U, Albrecht S, Dohmen H, Hauser P, Keyvani K, van Landeghem FKH, Lund EL, Scheie D, Mawrin C, Monoranu CM, Parm Ulhøi B, Pietsch T, Reinhard H, Riemenschneider MJ, Sehested A, Sumerauer D, Siebert R, Paulus W, Frühwald MC, Kool M, and Hasselblatt M
- Subjects
- DNA Methylation, Hedgehog Proteins genetics, Hedgehog Proteins metabolism, Humans, Prognosis, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Central Nervous System Neoplasms genetics, Neoplasms, Neuroepithelial genetics, Rhabdoid Tumor genetics, Teratoma genetics
- Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is an aggressive central nervous system tumor characterized by loss of SMARCB1/INI1 protein expression and comprises three distinct molecular groups, ATRT-TYR, ATRT-MYC and ATRT-SHH. ATRT-SHH represents the largest molecular group and is heterogeneous with regard to age, tumor location and epigenetic profile. We, therefore, aimed to investigate if heterogeneity within ATRT-SHH might also have biological and clinical importance. Consensus clustering of DNA methylation profiles and confirmatory t-SNE analysis of 65 ATRT-SHH yielded three robust molecular subgroups, i.e., SHH-1A, SHH-1B and SHH-2. These subgroups differed by median age of onset (SHH-1A: 18 months, SHH-1B: 107 months, SHH-2: 13 months) and tumor location (SHH-1A: 88% supratentorial; SHH-1B: 85% supratentorial; SHH-2: 93% infratentorial, often extending to the pineal region). Subgroups showed comparable SMARCB1 mutational profiles, but pathogenic/likely pathogenic SMARCB1 germline variants were over-represented in SHH-2 (63%) as compared to SHH-1A (20%) and SHH-1B (0%). Protein expression of proneural marker ASCL1 (enriched in SHH-1B) and glial markers OLIG2 and GFAP (absent in SHH-2) as well as global mRNA expression patterns differed, but all subgroups were characterized by overexpression of SHH as well as Notch pathway members. In a Drosophila model, knockdown of Snr1 (the fly homologue of SMARCB1) in hedgehog activated cells not only altered hedgehog signaling, but also caused aberrant Notch signaling and formation of tumor-like structures. Finally, on survival analysis, molecular subgroup and age of onset (but not ASCL1 staining status) were independently associated with overall survival, older patients (> 3 years) harboring SHH-1B experiencing relatively favorable outcome. In conclusion, ATRT-SHH comprises three subgroups characterized by SHH and Notch pathway activation, but divergent molecular and clinical features. Our data suggest that molecular subgrouping of ATRT-SHH has prognostic relevance and might aid to stratify patients within future clinical trials., (© 2022. The Author(s).)
- Published
- 2022
- Full Text
- View/download PDF
8. Low-grade diffusely infiltrative tumour (LGDIT), SMARCB1-mutant: A clinical and histopathological distinct entity showing epigenetic similarity with ATRT-MYC.
- Author
-
Hasselblatt M, Thomas C, Federico A, Bens S, Hellström M, Casar-Borota O, Kordes U, Neumann JE, Dottermusch M, Rodriguez FJ, Lo AC, Cheng S, Hendson G, Hukin J, Hartmann C, Koch A, Capper D, Siebert R, Paulus W, Nemes K, Johann PD, Frühwald MC, and Kool M
- Subjects
- Child, Epigenesis, Genetic, Humans, SMARCB1 Protein genetics, Young Adult, Rhabdoid Tumor pathology, Teratoma
- Abstract
Low-grade diffusely infiltrative tumour (LGDIT), SMARCB1-mutant, is a histopathological distinct low-grade lesion encountered in older children and young adults that shows epigenetic similarity with ATRT-MYC and has the potential for malignant progression., (© 2022 The Authors. Neuropathology and Applied Neurobiology published by John Wiley & Sons Ltd on behalf of British Neuropathological Society.)
- Published
- 2022
- Full Text
- View/download PDF
9. Histopathological patterns in atypical teratoid/rhabdoid tumors are related to molecular subgroup.
- Author
-
Zin F, Cotter JA, Haberler C, Dottermusch M, Neumann J, Schüller U, Schweizer L, Thomas C, Nemes K, Johann PD, Kool M, Frühwald MC, Paulus W, Judkins A, and Hasselblatt M
- Subjects
- Adolescent, Adult, Central Nervous System Neoplasms genetics, Child, DNA Methylation genetics, Female, Humans, Male, Neoplasms, Neuroepithelial genetics, Neoplasms, Neuroepithelial pathology, SMARCB1 Protein genetics, Young Adult, Biomarkers, Tumor genetics, Central Nervous System Neoplasms pathology, Rhabdoid Tumor pathology, Teratoma pathology
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant tumor that may not only contain rhabdoid tumor cells but also poorly differentiated small-round-blue cells as well as areas with mesenchymal or epithelial differentiation. Little is known on factors associated with histopathological diversity. Recent studies demonstrated three molecular subgroups of AT/RT, namely ATRT-TYR, ATRT-SHH, and ATRT-MYC. We thus aimed to investigate if morphological patterns might be related to molecular subgroup status. Hematoxylin-eosin stained sections of 114 AT/RT with known molecular subgroup status were digitalized and independently categorized by nine blinded observers into four morphological categories, that is, "rhabdoid," "small-round-blue," "epithelial," and "mesenchymal." The series comprised 48 ATRT-SHH, 40 ATRT-TYR, and 26 ATRT-MYC tumors. Inter-observer agreement was moderate but significant (Fleiss' kappa = 0.47; 95% C.I. 0.41-0.53; p < 0.001) and there was a highly significant overall association between morphological categories and molecular subgroups for each of the nine observers (p < 0.0001). Specifically, the category "epithelial" was found to be over-represented in ATRT-TYR (p < 0.000001) and the category "small-round-blue" to be over-represented in ATRT-SHH (p < 0.01). The majority of ATRT-MYC was categorized as "mesenchymal" or "rhabdoid," but this association was less compelling. The specificity of the category "epithelial" for ATRT-TYR was highest and accounted for 97% (range: 88-99%) whereas sensitivity was low [49% (range: 35%-63%)]. In line with these findings, cytokeratin-positivity was highly overrepresented in ATRT-TYR. In conclusion, morphological features of AT/RT might reflect molecular alterations and may also provide a first hint on molecular subgroup status, which will need to be confirmed by DNA methylation profiling., (© 2021 The Authors. Brain Pathology published by John Wiley & Sons Ltd on behalf of International Society of Neuropathology.)
- Published
- 2021
- Full Text
- View/download PDF
10. Atypical Teratoid/Rhabdoid Tumor (AT/RT) With Molecular Features of Pleomorphic Xanthoastrocytoma.
- Author
-
Thomas C, Federico A, Sill M, Bens S, Oyen F, Nemes K, Johann PD, Hartmann C, Hartmann W, Sumerauer D, Paterno V, Samii A, Kordes U, Siebert R, Frühwald MC, Paulus W, Kool M, and Hasselblatt M
- Subjects
- Adolescent, Child, Female, Humans, Mutation, Proto-Oncogene Proteins B-raf genetics, SMARCB1 Protein genetics, Astrocytoma genetics, Brain Neoplasms genetics, Rhabdoid Tumor genetics, Teratoma genetics
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a highly malignant central nervous system tumor predominantly occurring in infants that may also arise in older children and adults. Rare secondary AT/RT developing from other tumors such as pleomorphic xanthoastrocytoma (PXA) are on record, but AT/RT presenting with molecular features of PXA have not been described. Here, we report 3 malignant central nervous system tumors in children (10, 13, and 18 y old). All tumors were located in the temporal lobe. In 2 cases, there was no history of a low-grade precursor lesion; in 1 case anaplastic PXA had been diagnosed 3 months earlier. Histopathologically, all tumors were composed of RT cells and showed frank signs of malignancy as well as loss of nuclear SMARCB1/INI1 protein expression. Two cases displayed homozygous deletions of the SMARCB1 region while the third case showed an exon 7 mutation (c.849_850delGT; p.Met283Ilefs*77). Of note, DNA methylation profiles did not group with AT/RT or other tumor entities using the Heidelberg Brain Tumor Classifier (version v11b4). By unsupervised t-distributed stochastic neighbor embedding analysis and hierarchical clustering analysis, however, all tumors clearly grouped with PXA. Genome-wide copy number analysis revealed homozygous CDNK2A/B deletions and gains of whole chromosome 7. BRAF V600E mutations could be demonstrated in all cases. In conclusion, the possibility of AT/RT with molecular features of PXA needs to be taken into account and warrants molecular characterization of AT/RT especially in older children. Since treatments targeting mutated BRAF are available, identification of such cases may also have therapeutic consequences., Competing Interests: Conflicts of Interest and Source of Funding: M.H. and C.T. are supported by Deutsche Forschungsgemeinschaft (HA 3060/8-1 and TH 2345/1-1). The authors have disclosed that they have no significant relationships with, or financial interest in, any commercial companies pertaining to this article., (Copyright © 2021 Wolters Kluwer Health, Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
11. Organoid-based drug screening reveals neddylation as therapeutic target for malignant rhabdoid tumors.
- Author
-
Calandrini C, van Hooff SR, Paassen I, Ayyildiz D, Derakhshan S, Dolman MEM, Langenberg KPS, van de Ven M, de Heus C, Liv N, Kool M, de Krijger RR, Tytgat GAM, van den Heuvel-Eibrink MM, Molenaar JJ, and Drost J
- Subjects
- Animals, Cell Line, Tumor, Female, Humans, Male, Mice, Mice, Inbred NOD, Mice, SCID, Xenograft Model Antitumor Assays, Cyclopentanes pharmacology, Gene Expression Regulation, Neoplastic drug effects, Organoids metabolism, Pyrimidines pharmacology, Rhabdoid Tumor drug therapy, Rhabdoid Tumor metabolism, Unfolded Protein Response drug effects
- Abstract
Malignant rhabdoid tumors (MRTs) represent one of the most aggressive childhood malignancies. No effective treatment options are available, and prognosis is, therefore, dismal. Previous studies have demonstrated that tumor organoids capture the heterogeneity of patient tumors and can be used to predict patient response to therapy. Here, we perform drug screening on patient-derived normal and tumor organoids to identify MRT-specific therapeutic vulnerabilities. We identify neddylation inhibitor MLN4924 as a potential therapeutic agent. Mechanistically, we find increased neddylation in MRT organoids and tissues and show that MLN4924 induces a cytotoxic response via upregulation of the unfolded protein response. Lastly, we demonstrate in vivo efficacy in an MRT PDX mouse model, in which single-agent MLN4924 treatment significantly extends survival. Our study demonstrates that organoids can be used to find drugs selectively targeting tumor cells while leaving healthy cells unharmed and proposes neddylation inhibition as a therapeutic strategy in MRT., Competing Interests: Declaration of interests The authors declare no competing interests., (Copyright © 2021 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2021
- Full Text
- View/download PDF
12. Transposable element insertion as a mechanism of SMARCB1 inactivation in atypical teratoid/rhabdoid tumor.
- Author
-
Thomas C, Oehl-Huber K, Bens S, Soschinski P, Koch A, Nemes K, Oyen F, Kordes U, Kool M, Frühwald MC, Hasselblatt M, and Siebert R
- Subjects
- Brain Neoplasms pathology, DNA Transposable Elements, Female, Humans, Infant, Mutagenesis, Insertional, Rhabdoid Tumor pathology, Teratoma pathology, Brain Neoplasms genetics, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Teratoma genetics
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT) is a malignant brain tumor predominantly occurring in infants. Biallelic SMARCB1 mutations causing loss of nuclear SMARCB1/INI1 protein expression represent the characteristic genetic lesion. Pathogenic SMARCB1 mutations comprise single nucleotide variants, small insertions/deletions, large deletions, which may be also present in the germline (rhabdoid tumor predisposition syndrome 1), as well as somatic copy-number neutral loss of heterozygosity (LOH). In some SMARCB1-deficient AT/RT underlying biallelic mutations cannot be identified. Here we report the case of a 24-months-old girl diagnosed with a large brain tumor. The malignant rhabdoid tumor showed loss of nuclear SMARCB1/INI1 protein expression and the diagnosis of AT/RT was confirmed by DNA methylation profiling. While FISH, MLPA, Sanger sequencing and DNA methylation data-based imbalance analysis did not disclose alterations affecting SMARCB1, OncoScan array analysis revealed a 28.29 Mb sized region of copy-number neutral LOH on chromosome 22q involving the SMARCB1 locus. Targeted next-generation sequencing did also not detect a single nucleotide variant but instead revealed insertion of an AluY element into exon 2 of SMARCB1. Specific PCR-based Sanger sequencing verified the Alu insertion (SMARCB1 c.199_200 Alu ins) resulting in a frame-shift truncation not present in the patient's germline. In conclusion, transposable element insertion represents a hitherto not widely recognized mechanism of SMARCB1 disruption in AT/RT, which might not be detected by several widely applied conventional diagnostics assays. This finding has particular clinical implications, if rhabdoid predisposition syndrome 1 is suspected, but germline SMARCB1 alterations cannot be identified., (© 2021 The Authors. Genes, Chromosomes and Cancer published by Wiley Periodicals LLC.)
- Published
- 2021
- Full Text
- View/download PDF
13. Inhibition of nuclear export restores nuclear localization and residual tumor suppressor function of truncated SMARCB1/INI1 protein in a molecular subset of atypical teratoid/rhabdoid tumors.
- Author
-
Pathak R, Zin F, Thomas C, Bens S, Gayden T, Karamchandani J, Dudley RW, Nemes K, Johann PD, Oyen F, Kordes U, Jabado N, Siebert R, Paulus W, Kool M, Frühwald MC, Albrecht S, Kalpana GV, and Hasselblatt M
- Subjects
- Central Nervous System Neoplasms genetics, Central Nervous System Neoplasms metabolism, Child, Preschool, Female, Genes, Tumor Suppressor physiology, Humans, Infant, Male, Mutation genetics, Neoplasm, Residual metabolism, Neoplasms, Neuroepithelial genetics, Neoplasms, Neuroepithelial metabolism, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Teratoma genetics, Active Transport, Cell Nucleus physiology, Neoplasm, Residual genetics, Rhabdoid Tumor metabolism, SMARCB1 Protein metabolism
- Abstract
Loss of nuclear SMARCB1 (INI1/hSNF5/BAF47) protein expression due to biallelic mutations of the SMARCB1 tumor suppressor gene is a hallmark of atypical teratoid/rhabdoid tumors (ATRT), but the presence of cytoplasmic SMARCB1 protein in these tumors has not yet been described. In a series of 102 primary ATRT, distinct cytoplasmic SMARCB1 staining on immunohistochemistry was encountered in 19 cases (19%) and was highly over-represented in cases showing pathogenic sequence variants leading to truncation or mutation of the C-terminal part of SMARCB1 (15/19 vs. 4/83; Chi-square: 56.04, p = 1.0E-10) and, related to this, in tumors of the molecular subgroup ATRT-TYR (16/36 vs. 3/66; Chi-square: 24.47, p = 7.6E-7). Previous reports have indicated that while SMARCB1 lacks a bona fide nuclear localization signal, it harbors a masked nuclear export signal (NES) and that truncation of the C-terminal region results in unmasking of this NES leading to cytoplasmic localization. To determine if cytoplasmic localization found in ATRT is due to unmasking of NES, we generated GFP fusions of one of the SMARCB1 truncating mutations (p.Q318X) found in the tumors along with a p.L266A mutation, which was shown to disrupt the interaction of SMARCB1-NES with exportin-1. We found that while the GFP-SMARCB1(Q318X) mutant localized to the cytoplasm, the double mutant GFP-SMARCB1(Q318X;L266A) localized to the nucleus, confirming NES requirement for cytoplasmic localization. Furthermore, cytoplasmic SMARCB1(Q318X) was unable to cause senescence as determined by morphological observations and by senescence-associated β-galactosidase assay, while nuclear SMARCB1(Q318X;L266A) mutant regained this function. Selinexor, a selective exportin-1 inhibitor, was effective in inhibiting the nuclear export of SMARCB1(Q318X) and caused rapid cell death in rhabdoid tumor cells. In conclusion, inhibition of nuclear export restores nuclear localization and residual tumor suppressor function of truncated SMARCB1. Therapies aimed at preventing nuclear export of mutant SMARCB1 protein may represent a promising targeted therapy in ATRT harboring truncating C-terminal SMARCB1 mutations., (© 2021. The Author(s).)
- Published
- 2021
- Full Text
- View/download PDF
14. Relevance of Molecular Groups in Children with Newly Diagnosed Atypical Teratoid Rhabdoid Tumor: Results from Prospective St. Jude Multi-institutional Trials.
- Author
-
Upadhyaya SA, Robinson GW, Onar-Thomas A, Orr BA, Johann P, Wu G, Billups CA, Tatevossian RG, Dhanda SK, Srinivasan A, Broniscer A, Qaddoumi I, Vinitsky A, Armstrong GT, Bendel AE, Hassall T, Partap S, Fisher PG, Crawford JR, Chintagumpala M, Bouffet E, Gururangan S, Mostafavi R, Sanders RP, Klimo P Jr, Patay Z, Indelicato DJ, Nichols KE, Boop FA, Merchant TE, Kool M, Ellison DW, and Gajjar A
- Subjects
- Antineoplastic Combined Chemotherapy Protocols adverse effects, Antineoplastic Combined Chemotherapy Protocols therapeutic use, Child, Child, Preschool, DNA Copy Number Variations, DNA Methylation, Diagnosis, Differential, Disease Management, Disease Susceptibility, Female, Germ-Line Mutation, Humans, Infant, Male, Mutation, Prognosis, Rhabdoid Tumor mortality, Rhabdoid Tumor therapy, SMARCB1 Protein genetics, Teratoma mortality, Teratoma therapy, Treatment Outcome, Biomarkers, Tumor, Rhabdoid Tumor diagnosis, Rhabdoid Tumor etiology, Teratoma diagnosis, Teratoma etiology
- Abstract
Purpose: Report relevance of molecular groups to clinicopathologic features, germline SMARCB1/SMARCA4 alterations (GLA), and survival of children with atypical teratoid rhabdoid tumor (ATRT) treated in two multi-institutional clinical trials., Materials and Methods: Seventy-four participants with newly diagnosed ATRT were treated in two trials: infants (SJYC07: age < 3 years; n = 52) and children (SJMB03: age 3-21 years; n = 22), using surgery, conventional chemotherapy (infants), or dose-dense chemotherapy with autologous stem cell rescue (children), and age- and risk-adapted radiotherapy [focal (infants) and craniospinal (CSI; children)]. Molecular groups ATRT-MYC (MYC), ATRT-SHH (SHH), and ATRT-TYR (TYR) were determined from tumor DNA methylation profiles., Results: Twenty-four participants (32%) were alive at time of analysis at a median follow-up of 8.4 years (range, 3.1-14.1 years). Methylation profiling classified 64 ATRTs as TYR ( n = 21), SHH ( n = 30), and MYC ( n = 13), SHH group being associated with metastatic disease. Among infants, TYR group had the best overall survival (OS; P = 0.02). However, outcomes did not differ by molecular groups among infants with nonmetastatic (M0) disease. Children with M0 disease and <1.5 cm
2 residual tumor had a 5-year progression-free survival (PFS) of 72.7 ± 12.7% and OS of 81.8 ± 11%. Infants with M0 disease had a 5-year PFS of 39.1 ± 11.5% and OS of 51.8 ± 12%. Those with metastases fared poorly [5-year OS 25 ± 12.5% (children) and 0% (infants)]. SMARCB1 GLAs were not associated with PFS., Conclusions: Among infants, those with ATRT-TYR had the best OS. ATRT-SHH was associated with metastases and consequently with inferior outcomes. Children with nonmetastatic ATRT benefit from postoperative CSI and adjuvant chemotherapy., (©2021 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
15. Delta-24-RGD, an Oncolytic Adenovirus, Increases Survival and Promotes Proinflammatory Immune Landscape Remodeling in Models of AT/RT and CNS-PNET.
- Author
-
Garcia-Moure M, Gonzalez-Huarriz M, Labiano S, Guruceaga E, Bandres E, Zalacain M, Marrodan L, de Andrea C, Villalba M, Martinez-Velez N, Laspidea V, Puigdelloses M, Gallego Perez-Larraya J, Iñigo-Marco I, Stripecke R, Chan JA, Raabe EH, Kool M, Gomez-Manzano C, Fueyo J, Patiño-García A, and Alonso MM
- Subjects
- Animals, Apoptosis, Cell Proliferation, Central Nervous System Neoplasms immunology, Central Nervous System Neoplasms mortality, Central Nervous System Neoplasms pathology, Female, Humans, Immunity, Cellular, Mice, Mice, Inbred C57BL, Mice, Nude, Neuroectodermal Tumors, Primitive immunology, Neuroectodermal Tumors, Primitive mortality, Neuroectodermal Tumors, Primitive pathology, Rhabdoid Tumor immunology, Rhabdoid Tumor mortality, Rhabdoid Tumor pathology, Teratoma immunology, Teratoma mortality, Teratoma pathology, Tumor Cells, Cultured, Xenograft Model Antitumor Assays, Central Nervous System Neoplasms therapy, Neuroectodermal Tumors, Primitive therapy, Oligopeptides genetics, Oncolytic Virotherapy methods, Oncolytic Viruses genetics, Rhabdoid Tumor therapy, Teratoma therapy
- Abstract
Purpose: Atypical teratoid/rhabdoid tumors (AT/RT) and central nervous system primitive neuroectodermal tumors (CNS-PNET) are pediatric brain tumors with poor survival and life-long negative side effects. Here, the aim was to characterize the efficacy and safety of the oncolytic adenovirus, Delta-24-RGD, which selectively replicates in and kills tumor cells., Experimental Design: Delta-24-RGD determinants for infection and replication were evaluated in patient expression datasets. Viral replication and cytotoxicity were assessed in vitro in a battery of CNS-PNET and AT/RT cell lines. In vivo , efficacy was determined in different orthotopic mouse models, including early and established tumor models, a disseminated AT/RT lesion model, and immunocompetent humanized mouse models (hCD34
+ -NSG-SGM3)., Results: Delta-24-RGD infected and replicated efficiently in all the cell lines tested. In addition, the virus induced dose-dependent cytotoxicity [IC50 value below 1 plaque-forming unit (PFU)/cell] and the release of immunogenic markers. In vivo , a single intratumoral Delta-24-RGD injection (107 or 108 PFU) significantly increased survival and led to long-term survival in AT/RT and PNET models. Delta-24-RGD hindered the dissemination of AT/RTs and increased survival, leading to 70% of long-term survivors. Of relevance, viral administration to established tumor masses (30 days after engraftment) showed therapeutic benefit. In humanized immunocompetent models, Delta-24-RGD significantly extended the survival of mice bearing AT/RTs or PNETs (ranging from 11 to 27 days) and did not display any toxicity associated with inflammation. Immunophenotyping of Delta-24-RGD-treated tumors revealed increased CD8+ T-cell infiltration., Conclusions: Delta-24-RGD is a feasible therapeutic option for AT/RTs and CNS-PNETs. This work constitutes the basis for potential translation to the clinical setting., (©2020 American Association for Cancer Research.)- Published
- 2021
- Full Text
- View/download PDF
16. Somatic mutations and single-cell transcriptomes reveal the root of malignant rhabdoid tumours.
- Author
-
Custers L, Khabirova E, Coorens THH, Oliver TRW, Calandrini C, Young MD, Vieira Braga FA, Ellis P, Mamanova L, Segers H, Maat A, Kool M, Hoving EW, van den Heuvel-Eibrink MM, Nicholson J, Straathof K, Hook L, de Krijger RR, Trayers C, Allinson K, Behjati S, and Drost J
- Subjects
- Cell Differentiation genetics, DNA Methylation, Drug Screening Assays, Antitumor, Gene Expression Profiling, Gene Expression Regulation, Neoplastic drug effects, Histone Deacetylase Inhibitors pharmacology, Humans, Neural Crest pathology, Phylogeny, Rhabdoid Tumor drug therapy, SMARCB1 Protein genetics, Single-Cell Analysis, TOR Serine-Threonine Kinases antagonists & inhibitors, Tissue Culture Techniques methods, Mutation, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology
- Abstract
Malignant rhabdoid tumour (MRT) is an often lethal childhood cancer that, like many paediatric tumours, is thought to arise from aberrant fetal development. The embryonic root and differentiation pathways underpinning MRT are not firmly established. Here, we study the origin of MRT by combining phylogenetic analyses and single-cell mRNA studies in patient-derived organoids. Comparison of somatic mutations shared between cancer and surrounding normal tissues places MRT in a lineage with neural crest-derived Schwann cells. Single-cell mRNA readouts of MRT differentiation, which we examine by reverting the genetic driver mutation underpinning MRT, SMARCB1 loss, suggest that cells are blocked en route to differentiating into mesenchyme. Quantitative transcriptional predictions indicate that combined HDAC and mTOR inhibition mimic MRT differentiation, which we confirm experimentally. Our study defines the developmental block of MRT and reveals potential differentiation therapies.
- Published
- 2021
- Full Text
- View/download PDF
17. Atypical teratoid/rhabdoid tumors (ATRTs) with SMARCA4 mutation are molecularly distinct from SMARCB1-deficient cases.
- Author
-
Holdhof D, Johann PD, Spohn M, Bockmayr M, Safaei S, Joshi P, Masliah-Planchon J, Ho B, Andrianteranagna M, Bourdeaut F, Huang A, Kool M, Upadhyaya SA, Bendel AE, Indenbirken D, Foulkes WD, Bush JW, Creytens D, Kordes U, Frühwald MC, Hasselblatt M, and Schüller U
- Subjects
- Adolescent, Adult, Age of Onset, Central Nervous System Neoplasms pathology, Child, Child, Preschool, Computational Biology, DNA Methylation, Gene Expression Profiling, Humans, Middle Aged, Mutation genetics, Rhabdoid Tumor pathology, Survival Analysis, Teratoma pathology, Young Adult, Central Nervous System Neoplasms genetics, DNA Helicases genetics, Nuclear Proteins genetics, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Teratoma genetics, Transcription Factors genetics
- Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) are very aggressive childhood malignancies of the central nervous system. The underlying genetic cause are inactivating bi-allelic mutations in SMARCB1 or (rarely) in SMARCA4. ATRT-SMARCA4 have been associated with a higher frequency of germline mutations, younger age, and an inferior prognosis in comparison to SMARCB1 mutated cases. Based on their DNA methylation profiles and transcriptomics, SMARCB1 mutated ATRTs have been divided into three distinct molecular subgroups: ATRT-TYR, ATRT-SHH, and ATRT-MYC. These subgroups differ in terms of age at diagnosis, tumor location, type of SMARCB1 alterations, and overall survival. ATRT-SMARCA4 are, however, less well understood, and it remains unknown, whether they belong to one of the described ATRT subgroups. Here, we examined 14 ATRT-SMARCA4 by global DNA methylation analyses. We show that they form a separate group segregating from SMARCB1 mutated ATRTs and from other SMARCA4-deficient tumors like small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) or SMARCA4 mutated extra-cranial malignant rhabdoid tumors. In contrast, medulloblastoma (MB) samples with heterozygous SMARCA4 mutations do not group separately, but with established MB subgroups. RNA sequencing of ATRT-SMARCA4 confirmed the clustering results based on DNA methylation profiling and displayed an absence of typical signature genes upregulated in SMARCB1 deleted ATRT. In summary, our results suggest that, in line with previous clinical observations, ATRT-SMARCA4 should be regarded as a distinct molecular subgroup.
- Published
- 2021
- Full Text
- View/download PDF
18. SMARCB1 loss interacts with neuronal differentiation state to block maturation and impact cell stability.
- Author
-
Parisian AD, Koga T, Miki S, Johann PD, Kool M, Crawford JR, and Furnari FB
- Subjects
- Gene Deletion, Gene Expression Profiling, Gene Expression Regulation, Neoplastic genetics, Gene Knockdown Techniques, Humans, Induced Pluripotent Stem Cells, Organoids cytology, Organoids physiopathology, Brain Neoplasms genetics, Carcinogenesis genetics, Cell Differentiation genetics, Neurons cytology, Rhabdoid Tumor genetics, SMARCB1 Protein genetics
- Abstract
Atypical teratoid rhabdoid tumors (ATRTs) are challenging pediatric brain cancers that are predominantly associated with inactivation of the gene SMARCB1 , a conserved subunit of the chromatin remodeling BAF complex, which has known contributions to developmental processes. To identify potential interactions between SMARCB1 loss and the process of neural development, we introduced an inducible SMARCB1 loss-of-function system into human induced pluripotent stem cells (iPSCs) that were subjected to either directed neuronal differentiation or differentiation into cerebral organoids. Using this system, we identified substantial differences in the downstream effects of SMARCB1 loss depending on differentiation state and identified an interaction between SMARCB1 loss and neural differentiation pressure that causes a resistance to terminal differentiation and a defect in maintenance of a normal cell state. Our results provide insight into how SMARCB1 loss might interact with neural development in the process of ATRT tumorigenesis., (© 2020 Parisian et al.; Published by Cold Spring Harbor Laboratory Press.)
- Published
- 2020
- Full Text
- View/download PDF
19. Age and DNA methylation subgroup as potential independent risk factors for treatment stratification in children with atypical teratoid/rhabdoid tumors.
- Author
-
Frühwald MC, Hasselblatt M, Nemes K, Bens S, Steinbügl M, Johann PD, Kerl K, Hauser P, Quiroga E, Solano-Paez P, Biassoni V, Gil-da-Costa MJ, Perek-Polnik M, van de Wetering M, Sumerauer D, Pears J, Stabell N, Holm S, Hengartner H, Gerber NU, Grotzer M, Boos J, Ebinger M, Tippelt S, Paulus W, Furtwängler R, Hernáiz-Driever P, Reinhard H, Rutkowski S, Schlegel PG, Schmid I, Kortmann RD, Timmermann B, Warmuth-Metz M, Kordes U, Gerss J, Nysom K, Schneppenheim R, Siebert R, Kool M, and Graf N
- Subjects
- Adolescent, Adult, Age Distribution, Child, DNA Methylation, Europe, Female, Humans, In Situ Hybridization, Fluorescence, Male, Risk Factors, Young Adult, Rhabdoid Tumor genetics, Rhabdoid Tumor therapy, Teratoma genetics, Teratoma therapy
- Abstract
Background: Controversy exists as to what may be defined as standard of care (including markers for stratification) for patients with atypical teratoid/rhabdoid tumors (ATRTs). The European Rhabdoid Registry (EU-RHAB) recruits uniformly treated patients and offers standardized genetic and DNA methylation analyses., Methods: Clinical, genetic, and treatment data of 143 patients from 13 European countries were analyzed (2009-2017). Therapy consisted of surgery, anthracycline-based induction, and either radiotherapy or high dose chemotherapy following a consensus among European experts. Fluorescence in situ hybridization, multiplex ligation-dependent probe amplification, and sequencing were employed for assessment of somatic and germline mutations in SWItch/sucrose nonfermentable related, matrix associated, actin dependent regulator of chromatin, subfamily B (SMARCB1). Molecular subgroups (ATRT-SHH, ATRT-TYR, and ATRT-MYC) were determined using DNA methylation arrays, resulting in profiles of 84 tumors., Results: Median age at diagnosis of 67 girls and 76 boys was 29.5 months. Five-year overall survival (OS) and event-free survival (EFS) were 34.7 ± 4.5% and 30.5 ± 4.2%, respectively. Tumors displayed allelic partial/whole gene deletions (66%; 122/186 alleles) or single nucleotide variants (34%; 64/186 alleles) of SMARCB1. Germline mutations were detected in 26% of ATRTs (30/117). The patient cohort consisted of 47% ATRT-SHH (39/84), 33% ATRT-TYR (28/84), and 20% ATRT-MYC (17/84). Age <1 year, non-TYR signature (ATRT-SHH or -MYC), metastatic or synchronous tumors, germline mutation, incomplete remission, and omission of radiotherapy were negative prognostic factors in univariate analyses (P < 0.05). An adjusted multivariate model identified age <1 year and a non-TYR signature as independent negative predictors of OS: high risk (<1 y + non-TYR; 5-y OS = 0%), intermediate risk (<1 y + ATRT-TYR or ≥1 y + non-TYR; 5-y OS = 32.5 ± 8.7%), and standard risk (≥1 y + ATRT-TYR, 5-y OS = 71.5 ± 12.2%)., Conclusions: Age and molecular subgroup status are independent risk factors for survival in children with ATRT. Our model warrants validation within future clinical trials., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology. All rights reserved. For permissions, please e-mail: journals.permissions@oup.com.)
- Published
- 2020
- Full Text
- View/download PDF
20. Molecular subgrouping of atypical teratoid/rhabdoid tumors-a reinvestigation and current consensus.
- Author
-
Ho B, Johann PD, Grabovska Y, De Dieu Andrianteranagna MJ, Yao F, Frühwald M, Hasselblatt M, Bourdeaut F, Williamson D, Huang A, and Kool M
- Subjects
- Consensus, DNA Methylation, Humans, Neoplasms, Neuroepithelial, Rhabdoid Tumor genetics, Teratoma genetics
- Abstract
Background: Atypical teratoid/rhabdoid tumors (ATRTs) are known to exhibit molecular and clinical heterogeneity even though SMARCB1 inactivation is the sole recurrent genetic event present in nearly all cases. Indeed, recent studies demonstrated 3 molecular subgroups of ATRTs that are genetically, epigenetically, and clinically distinct. As these studies included different numbers of tumors, various subgrouping techniques, and naming, an international working group sought to align previous findings and to reach a consensus on nomenclature and clinicopathological significance of ATRT subgroups., Methods: We integrated various methods to perform a meta-analysis on published and unpublished DNA methylation and gene expression datasets of ATRTs and associated clinicopathological data., Results: In concordance with previous studies, the analyses identified 3 main molecular subgroups of ATRTs, for which a consensus was reached to name them ATRT-TYR, ATRT-SHH, and ATRT-MYC. The ATRT-SHH subgroup exhibited further heterogeneity, segregating further into 2 subtypes associated with a predominant supratentorial (ATRT-SHH-1) or infratentorial (ATRT-SHH-2) location. For each ATRT subgroup we provide an overview of its main molecular and clinical characteristics, including SMARCB1 alterations and pathway activation., Conclusions: The introduction of a common classification, characterization, and nomenclature of ATRT subgroups will facilitate future research and serve as a common ground for subgrouping patient samples and ATRT models, which will aid in refining subgroup-based therapies for ATRT patients., (© The Author(s) 2019. Published by Oxford University Press on behalf of the Society for Neuro-Oncology.)
- Published
- 2020
- Full Text
- View/download PDF
21. Locoregionally administered B7-H3-targeted CAR T cells for treatment of atypical teratoid/rhabdoid tumors.
- Author
-
Theruvath J, Sotillo E, Mount CW, Graef CM, Delaidelli A, Heitzeneder S, Labanieh L, Dhingra S, Leruste A, Majzner RG, Xu P, Mueller S, Yecies DW, Finetti MA, Williamson D, Johann PD, Kool M, Pfister S, Hasselblatt M, Frühwald MC, Delattre O, Surdez D, Bourdeaut F, Puget S, Zaidi S, Mitra SS, Cheshier S, Sorensen PH, Monje M, and Mackall CL
- Subjects
- Adult, Animals, Brain drug effects, Brain immunology, Brain pathology, Brain Neoplasms immunology, Brain Neoplasms pathology, Cells, Cultured, Child, Preschool, Female, Fetus pathology, Humans, Infant, Injections, Intraventricular, Mice, Mice, Inbred NOD, Mice, SCID, Receptors, Chimeric Antigen administration & dosage, Receptors, Chimeric Antigen genetics, Receptors, Chimeric Antigen immunology, Rhabdoid Tumor immunology, Rhabdoid Tumor pathology, T-Lymphocytes immunology, T-Lymphocytes metabolism, T-Lymphocytes transplantation, Teratoma immunology, Teratoma pathology, Xenograft Model Antitumor Assays, B7 Antigens immunology, Brain Neoplasms therapy, Cancer Vaccines administration & dosage, Immunotherapy, Adoptive methods, Rhabdoid Tumor therapy, Teratoma therapy
- Abstract
Atypical teratoid/rhabdoid tumors (ATRTs) typically arise in the central nervous system (CNS) of children under 3 years of age. Despite intensive multimodal therapy (surgery, chemotherapy and, if age permits, radiotherapy), median survival is 17 months
1,2 . We show that ATRTs robustly express B7-H3/CD276 that does not result from the inactivating mutations in SMARCB1 (refs.3,4 ), which drive oncogenesis in ATRT, but requires residual SWItch/Sucrose Non-Fermentable (SWI/SNF) activity mediated by BRG1/SMARCA4. Consistent with the embryonic origin of ATRT5,6 , B7-H3 is highly expressed on the prenatal, but not postnatal, brain. B7-H3.BB.z-chimeric antigen receptor (CAR) T cells administered intracerebroventricularly or intratumorally mediate potent antitumor effects against cerebral ATRT xenografts in mice, with faster kinetics, greater potency and reduced systemic levels of inflammatory cytokines compared to CAR T cells administered intravenously. CAR T cells administered ICV also traffic from the CNS into the periphery; following clearance of ATRT xenografts, B7-H3.BB.z-CAR T cells administered intracerebroventricularly or intravenously mediate antigen-specific protection from tumor rechallenge, both in the brain and periphery. These results identify B7-H3 as a compelling therapeutic target for this largely incurable pediatric tumor and demonstrate important advantages of locoregional compared to systemic delivery of CAR T cells for the treatment of CNS malignancies.- Published
- 2020
- Full Text
- View/download PDF
22. Tyrosinase immunohistochemistry can be employed for the diagnosis of atypical teratoid/rhabdoid tumours of the tyrosinase subgroup (ATRT-TYR).
- Author
-
Hasselblatt M, Thomas C, Nemes K, Monoranu CM, Riemenschneider MJ, Koch A, Sumerauer D, Hauser P, Paulus W, Johann PD, Kool M, and Frühwald MC
- Subjects
- Child, Preschool, DNA Methylation, Female, Humans, Immunohistochemistry, Infant, Male, Monophenol Monooxygenase metabolism, Rhabdoid Tumor metabolism, Sensitivity and Specificity, Teratoma metabolism, Monophenol Monooxygenase analysis, Rhabdoid Tumor diagnosis, Teratoma diagnosis
- Published
- 2020
- Full Text
- View/download PDF
23. Desmoplastic myxoid tumor, SMARCB1-mutant: clinical, histopathological and molecular characterization of a pineal region tumor encountered in adolescents and adults.
- Author
-
Thomas C, Wefers A, Bens S, Nemes K, Agaimy A, Oyen F, Vogelgesang S, Rodriguez FJ, Brett FM, McLendon R, Bodi I, Burel-Vandenbos F, Keyvani K, Tippelt S, Poulsen FR, Lipp ES, Giannini C, Reifenberger G, Kuchelmeister K, Pietsch T, Kordes U, Siebert R, Frühwald MC, Johann PD, Sill M, Kool M, von Deimling A, Paulus W, and Hasselblatt M
- Subjects
- Adolescent, Adult, Age Factors, Brain Neoplasms mortality, Brain Neoplasms pathology, Cohort Studies, Female, Humans, Male, Middle Aged, Rhabdoid Tumor mortality, Survival Rate, Young Adult, Brain Neoplasms genetics, Mutation genetics, Pineal Gland, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, SMARCB1 Protein genetics
- 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 40 years (range 15-61 years). 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 2 bp 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 48 months, 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
- Full Text
- View/download PDF
24. Spinal cord atypical teratoid/rhabdoid tumors in children: Clinical, genetic, and outcome characteristics in a representative European cohort.
- Author
-
Benesch M, Nemes K, Neumayer P, Hasselblatt M, Timmermann B, Bison B, Ebetsberger-Dachs G, Bourdeaut F, Dufour C, Biassoni V, Morales La Madrid A, Entz-Werle N, Laithier V, Quehenberger F, Weis S, Sumerauer D, Siebert R, Bens S, Schneppenheim R, Kool M, Modena P, Fouyssac F, and C Frühwald M
- Subjects
- Child, Child, Preschool, Combined Modality Therapy, DNA Helicases genetics, Female, Follow-Up Studies, Humans, Infant, Male, Nuclear Proteins genetics, Prognosis, Retrospective Studies, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Rhabdoid Tumor therapy, SMARCB1 Protein genetics, Spinal Cord Neoplasms genetics, Spinal Cord Neoplasms pathology, Spinal Cord Neoplasms therapy, Survival Rate, Teratoma genetics, Teratoma pathology, Teratoma therapy, Transcription Factors genetics, Biomarkers, Tumor genetics, Rhabdoid Tumor mortality, Spinal Cord Neoplasms mortality, Teratoma mortality
- Abstract
Background: Case reports have portrayed spinal cord atypical teratoid/rhabdoid tumor (spATRT) as an aggressive form of ATRT. We conducted a retrospective European survey to collect data on clinical characteristics, molecular biology, treatment, and outcome of children with intramedullary spATRT., Methods: Scrutinizing a French national series and the European Rhabdoid Registry database, we identified 13 patients (median age 32 months; metastatic disease at diagnosis, n = 6). Systemic postoperative chemotherapy was administered to all patients; three received intrathecal therapy and six were irradiated (craniospinal, n = 3; local, n = 3)., Results: Median observation time was 8 (range, 1-93) months. Progression-free and overall survival rates at 1 and (2 years) were 35.2% ± 13.9% (26.4% ± 12.9%) and 38.5% ± 13.5% (23.1% ± 11.7%). Four patients (ATRT-SHH, n = 2; ATRT-MYC, n = 1; DNA methylation subgroup not available, n = 1) achieved complete remission (CR); two of them are alive in CR 69 and 72 months from diagnosis. One patient relapsed after CR and is alive with progressive disease (PD) and one died of the disease. Three patients (ATRT-MYC, n = 2; subgroup not available, n = 1) died after 7 to 22 months due to PD after having achieved a partial remission (n = 1) or stabilization (n = 2). Five patients (ATRT-MYC, n = 2; subgroup not available, n = 3) developed early PD and died. One patient (ATRT-MYC) died of intracerebral hemorrhage prior to response evaluation., Conclusions: Long-term survival is achievable in selected patients with spATRT using aggressive multimodality treatment. Larger case series and detailed molecular analyses are needed to understand differences between spATRT and their inracranial counterparts and the group of extradural malignant rhabdoid tumors., (© 2019 Wiley Periodicals, Inc.)
- Published
- 2020
- Full Text
- View/download PDF
25. Identification and Analyses of Extra-Cranial and Cranial Rhabdoid Tumor Molecular Subgroups Reveal Tumors with Cytotoxic T Cell Infiltration.
- Author
-
Chun HE, Johann PD, Milne K, Zapatka M, Buellesbach A, Ishaque N, Iskar M, Erkek S, Wei L, Tessier-Cloutier B, Lever J, Titmuss E, Topham JT, Bowlby R, Chuah E, Mungall KL, Ma Y, Mungall AJ, Moore RA, Taylor MD, Gerhard DS, Jones SJM, Korshunov A, Gessler M, Kerl K, Hasselblatt M, Frühwald MC, Perlman EJ, Nelson BH, Pfister SM, Marra MA, and Kool M
- Subjects
- Child, DNA Methylation genetics, DNA Methylation physiology, Female, Humans, Lymphocytes, Tumor-Infiltrating metabolism, Lymphocytes, Tumor-Infiltrating pathology, Male, Mutation genetics, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Skull Base Neoplasms metabolism, Skull Base Neoplasms pathology, T-Lymphocytes metabolism, T-Lymphocytes pathology, Teratoma metabolism, Teratoma pathology, Rhabdoid Tumor metabolism, Rhabdoid Tumor pathology, T-Lymphocytes, Cytotoxic metabolism, T-Lymphocytes, Cytotoxic pathology
- Abstract
Extra-cranial malignant rhabdoid tumors (MRTs) and cranial atypical teratoid RTs (ATRTs) are heterogeneous pediatric cancers driven primarily by SMARCB1 loss. To understand the genome-wide molecular relationships between MRTs and ATRTs, we analyze multi-omics data from 140 MRTs and 161 ATRTs. We detect similarities between the MYC subgroup of ATRTs (ATRT-MYC) and extra-cranial MRTs, including global DNA hypomethylation and overexpression of HOX genes and genes involved in mesenchymal development, distinguishing them from other ATRT subgroups that express neural-like features. We identify five DNA methylation subgroups associated with anatomical sites and SMARCB1 mutation patterns. Groups 1, 3, and 4 exhibit cytotoxic T cell infiltration and expression of immune checkpoint regulators, consistent with a potential role for immunotherapy in rhabdoid tumor patients., (Copyright © 2019 The Author(s). Published by Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
26. Two molecularly distinct atypical teratoid/rhabdoid tumors (or tumor components) occurring in an infant with rhabdoid tumor predisposition syndrome 1.
- Author
-
Thomas C, Knerlich-Lukoschus F, Reinhard H, Johann PD, Sturm D, Sahm F, Bens S, Vogt J, Nemes K, Oyen F, Kordes U, Siebert R, Schneppenheim R, Messing-Jünger M, Pietsch T, von Deimling A, Paulus W, Pfister SM, Kool M, Frühwald MC, and Hasselblatt M
- Subjects
- Brain Neoplasms diagnostic imaging, Brain Neoplasms genetics, Brain Neoplasms pathology, Fatal Outcome, Humans, Infant, Kidney Neoplasms diagnostic imaging, Kidney Neoplasms genetics, Kidney Neoplasms pathology, Male, Mutation, Neoplasms, Multiple Primary diagnostic imaging, Neoplasms, Multiple Primary genetics, Neoplasms, Multiple Primary metabolism, Neoplasms, Multiple Primary pathology, Rhabdoid Tumor diagnostic imaging, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, SMARCB1 Protein genetics, SMARCB1 Protein metabolism, Teratoma diagnostic imaging, Teratoma genetics, Teratoma pathology, Brain Neoplasms metabolism, Kidney Neoplasms metabolism, Rhabdoid Tumor metabolism, Teratoma metabolism
- Published
- 2019
- Full Text
- View/download PDF
27. Comprehensive Analysis of Chromatin States in Atypical Teratoid/Rhabdoid Tumor Identifies Diverging Roles for SWI/SNF and Polycomb in Gene Regulation.
- Author
-
Erkek S, Johann PD, Finetti MA, Drosos Y, Chou HC, Zapatka M, Sturm D, Jones DTW, Korshunov A, Rhyzova M, Wolf S, Mallm JP, Beck K, Witt O, Kulozik AE, Frühwald MC, Northcott PA, Korbel JO, Lichter P, Eils R, Gajjar A, Roberts CWM, Williamson D, Hasselblatt M, Chavez L, Pfister SM, and Kool M
- Subjects
- Binding Sites, Brain metabolism, Cell Line, Tumor, Chromatin Immunoprecipitation, Epigenomics methods, Gene Expression Regulation, Neoplastic, Histones metabolism, Humans, SMARCB1 Protein chemistry, Sequence Analysis, DNA, Survival Analysis, Chromatin metabolism, Polycomb-Group Proteins metabolism, Repressor Proteins metabolism, Rhabdoid Tumor metabolism, SMARCB1 Protein metabolism, Teratoma metabolism
- Abstract
Biallelic inactivation of SMARCB1, encoding a member of the SWI/SNF chromatin remodeling complex, is the hallmark genetic aberration of atypical teratoid rhabdoid tumors (ATRT). Here, we report how loss of SMARCB1 affects the epigenome in these tumors. Using chromatin immunoprecipitation sequencing (ChIP-seq) on primary tumors for a series of active and repressive histone marks, we identified the chromatin states differentially represented in ATRTs compared with other brain tumors and non-neoplastic brain. Re-expression of SMARCB1 in ATRT cell lines enabled confirmation of our genome-wide findings for the chromatin states. Additional generation of ChIP-seq data for SWI/SNF and Polycomb group proteins and the transcriptional repressor protein REST determined differential dependencies of SWI/SNF and Polycomb complexes in regulation of diverse gene sets in ATRTs., (Copyright © 2018 Elsevier Inc. All rights reserved.)
- Published
- 2019
- Full Text
- View/download PDF
28. Functional relevance of genes predicted to be affected by epigenetic alterations in atypical teratoid/rhabdoid tumors.
- Author
-
Tegeder I, Thiel K, Erkek S, Johann PD, Berlandi J, Thatikonda V, Frühwald MC, Kool M, Jeibmann A, and Hasselblatt M
- Subjects
- Animals, Cell Line, Tumor, Drosophila melanogaster, Histones metabolism, Humans, Brain Neoplasms genetics, DNA-Binding Proteins genetics, Drosophila Proteins genetics, Epigenesis, Genetic, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Teratoma genetics, Transcription Factors genetics
- Abstract
Purpose: Atypical teratoid/rhabdoid tumor (ATRT) is a highly malignant brain tumor predominantly arising in infants. Mutations of SWI/SNF chromatin remodeling complex members SMARCB1/INI1 or (rarely) SMARCA4/Brg1 are the sole recurrent genetic lesions. Epigenetic studies revealed a large number of genes predicted to be affected by differential histone modifications in ATRT, but the role of these genes in the biology of ATRT remains uncertain. We therefore aimed at exploring the role of these genes in the detrimental effects of SMARCB1-deficiency., Methods: The functional relevance of 1083 genes predicted to be affected by epigenetic alterations in ATRT was examined in vivo using a Drosophila melanogaster model of SMARCB1-deficiency. Human orthologues of genes whose knockdown modified the phenotype in the Gal4-UAS fly model were further examined in ATRT samples and SMARCB1-deficient rhabdoid tumor cells., Results: Knockdown of Snr1, the fly orthologue of SMARCB1, resulted in a lethal phenotype and epigenetic alterations in the fly model. The lethal phenotype was shifted to later stages of development upon additional siRNA knockdown of 89 of 1083 genes screened in vivo. These included TGF-beta receptor signaling pathway related genes, e.g. CG10348, the fly orthologue of transcriptional regulator PRDM16. Subsequently, PRDM16 was found to be over-expressed in ATRT samples and knockdown of PRDM16 in SMARCB1-deficient rhabdoid tumor cells reduced proliferation., Conclusions: These results suggest that a subset of genes affected by differential histone modification in ATRT is involved in the detrimental effects of SMARCB1-deficiency and also relevant in the biology of ATRT.
- Published
- 2019
- Full Text
- View/download PDF
29. Magnetic resonance imaging surrogates of molecular subgroups in atypical teratoid/rhabdoid tumor.
- Author
-
Nowak J, Nemes K, Hohm A, Vandergrift LA, Hasselblatt M, Johann PD, Kool M, Frühwald MC, and Warmuth-Metz M
- Subjects
- Biomarkers, Tumor analysis, Child, Preschool, Diagnosis, Differential, Female, Follow-Up Studies, Humans, Male, Prognosis, Retrospective Studies, Rhabdoid Tumor genetics, Rhabdoid Tumor metabolism, Teratoma genetics, Teratoma metabolism, Hedgehog Proteins genetics, Magnetic Resonance Imaging methods, Proto-Oncogene Proteins c-myc genetics, Rhabdoid Tumor diagnosis, Teratoma diagnosis, Tyrosine metabolism
- Abstract
Background: Recently, 3 molecular subgroups of atypical teratoid/rhabdoid tumor (ATRT) were identified, but little is known of their clinical and magnetic resonance imaging (MRI) characteristics., Methods: A total of 43 patients with known molecular subgroup status (ATRT-sonic hedgehog [SHH], n = 17; ATRT-tyrosine [TYR], n = 16; ATRT-myelocytomatosis oncogene [MYC], n = 10) were retrieved from the EU-RHAB Registry and analyzed for clinical and MRI features., Results: On MRI review, differences in preferential tumor location were confirmed, with ATRT-TYR being predominantly located infratentorially (P < 0.05). Peritumoral edema was more pronounced in ATRT-MYC compared with ATRT-SHH (P < 0.05) and ATRT-TYR (P < 0.05). Conversely, peripheral tumor cysts were found more frequently in ATRT-SHH (71%) and ATRT-TYR (94%) compared with ATRT-MYC (40%, P < 0.05). Contrast enhancement was absent in 29% of ATRT-SHH (0% of ATRT-TYR; 10% of ATRT-MYC; P < 0.05), and there was a trend toward strong contrast enhancement in ATRT-TYR and ATRT-MYC. We found the characteristic (bandlike) enhancement in 28% of ATRT as well as restricted diffusion in the majority of tumors. A midline/off-midline location in the posterior fossa was also not subgroup specific. Visible meningeal spread (M2) at diagnosis was rare throughout all subgroups., Conclusion: These exploratory findings suggest that MRI features vary across the 3 molecular subgroups of ATRT. Within future prospective trials, MRI may aid diagnosis and treatment stratification.
- Published
- 2018
- Full Text
- View/download PDF
30. Sellar Region Atypical Teratoid/Rhabdoid Tumors (ATRT) in Adults Display DNA Methylation Profiles of the ATRT-MYC Subgroup.
- Author
-
Johann PD, Bens S, Oyen F, Wagener R, Giannini C, Perry A, Raisanen JM, Reis GF, Nobusawa S, Arita K, Felsberg J, Reifenberger G, Agaimy A, Buslei R, Capper D, Pfister SM, Schneppenheim R, Siebert R, Frühwald MC, Paulus W, Kool M, and Hasselblatt M
- Subjects
- Adult, Age Factors, Aged, Female, Gene Deletion, Genetic Predisposition to Disease, Humans, Male, Middle Aged, Phenotype, Pituitary Neoplasms pathology, Point Mutation, Rhabdoid Tumor pathology, Teratoma pathology, Young Adult, Biomarkers, Tumor genetics, DNA Methylation, Epigenesis, Genetic, Pituitary Neoplasms genetics, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Teratoma genetics
- Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is a highly malignant brain tumor predominantly encountered in infants. Mutations of the SMARCB1 gene are the characteristic genetic lesion. A small group of ATRT stands out clinically, because these tumors are located in the sellar region of adults. To investigate if sellar region ATRT in adults represents a molecular distinct entity, we characterized molecular alterations in 7 sellar region ATRTs in adults as compared with 150 pediatric ATRTs and 47 pituitary adenomas using SMARCB1 sequencing, multiplex ligation-dependent probe amplification and fluorescence in situ hybridization as well as DNA methylation profiling. The median age of the 6 female and 1 male patients was 56 years. On histopathologic examination, all tumors were malignant rhabdoid tumors showing loss of SMARCB1/INI1 protein expression. Two cases displayed compound heterozygous SMARCB1 point mutations, 3 cases showed heterozygous SMARCB1 deletions with point mutations of the other allele and 1 case a homozygous SMARCB1 deletion; in 1 case, underlying SMARCB1 alterations could not be identified. On unsupervised hierarchical cluster analysis of DNA methylation profiles, sellar region ATRTs did not form a distinct group, but clustered with ATRT-MYC, 1 of 3 recently described molecular subgroups of ATRT. On analysis of DNA methylation array intensity data, only 1 sellar region ATRT showed characteristic features of pediatric ATRT-MYC, that is, major copy number losses affecting the SMARCB1 region. In conclusion, these results suggest that sellar region ATRTs in adults form a clinically distinct entity with a different mutational spectrum, but epigenetic similarities with pediatric ATRTs of the ATRT-MYC subgroup.
- Published
- 2018
- Full Text
- View/download PDF
31. Reduced histone H3 K27 trimethylation is encountered in about 50% of atypical teratoid/rhabdoid tumors (AT/RT) but is not associated with molecular subgroup status and outcome.
- Author
-
Hasselblatt M, Johann PD, Kool M, and Frühwald MC
- Subjects
- Brain Neoplasms genetics, Brain Neoplasms pathology, Humans, Prognosis, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Teratoma genetics, Teratoma pathology, Brain Neoplasms metabolism, Histones metabolism, Rhabdoid Tumor metabolism, Teratoma metabolism
- Published
- 2017
- Full Text
- View/download PDF
32. Cribriform neuroepithelial tumor: molecular characterization of a SMARCB1-deficient non-rhabdoid tumor with favorable long-term outcome.
- Author
-
Johann PD, Hovestadt V, Thomas C, Jeibmann A, Heß K, Bens S, Oyen F, Hawkins C, Pierson CR, Aldape K, Kim SP, Widing E, Sumerauer D, Hauser P, van Landeghem F, Ryzhova M, Korshunov A, Capper D, Jones DTW, Pfister SM, Schneppenheim R, Siebert R, Paulus W, Frühwald MC, Kool M, and Hasselblatt M
- Subjects
- Child, Child, Preschool, DNA Methylation genetics, Female, Humans, Infant, Kaplan-Meier Estimate, Male, Neoplasms, Neuroepithelial pathology, Rhabdoid Tumor pathology, Statistics, Nonparametric, Brain Neoplasms genetics, Mutation genetics, Neoplasms, Neuroepithelial genetics, Rhabdoid Tumor genetics, SMARCB1 Protein deficiency, SMARCB1 Protein genetics
- Abstract
Rhabdoid phenotype and loss of SMARCB1 expression in a brain tumor are characteristic features of atypical teratoid/rhabdoid tumors (ATRT). Rare non-rhabdoid brain tumors showing cribriform growth pattern and SMARCB1 loss have been designated cribriform neuroepithelial tumor (CRINET). Small case series suggest that CRINETs may have a relatively favorable prognosis. However, the long-term outcome is unclear and it remains uncertain whether CRINET represents a distinct entity or a variant of ATRT. Therefore, 10 CRINETs were clinically and molecularly characterized and compared with 10 ATRTs of each of three recently described molecular subgroups (i.e. ATRT-TYR, ATRT-SHH and ATRT-MYC) using Illumina Infinium HumanMethylation450 arrays, FISH, MLPA, and sequencing. Furthermore, outcome was compared to a larger cohort of 27 children with ATRT-TYR. Median age of the 6 boys and 4 girls harboring a CRINET was 20 months. On histopathological examination, all CRINETs demonstrated a cribriform growth pattern and distinct tyrosinase staining. On unsupervised cluster analysis of methylation data, all CRINETs examined exclusively clustered within the ATRT-TYR molecular subgroup. As ATRT-TYR, CRINETs mainly showed large heterozygous 22q deletions (9/10) and SMARCB1 mutations of the other allele. In two patients, SMARCB1 mutations were also present in the germline. Estimated mean overall survival in patients with CRINETs was 125 months (95% confidence interval 100-151 months) as compared to only 53 (33-74) months in patients with ATRTs of the ATRT-TYR subgroup (Log-Rank P < 0.05). In conclusion, CRINET represents a SMARCB1-deficient non-rhabdoid tumor, which shares molecular similarities with the ATRT-TYR subgroup but has distinct histopathological features and favorable long-term outcome., (© 2016 International Society of Neuropathology.)
- Published
- 2017
- Full Text
- View/download PDF
33. SMAD dependent signaling plays a detrimental role in a fly model of SMARCB1-deficiency and the biology of atypical teratoid/rhabdoid tumors.
- Author
-
Jeibmann A, Schulz J, Eikmeier K, Johann PD, Thiel K, Tegeder I, Ambrée O, Frühwald MC, Pfister SM, Kool M, Paulus W, and Hasselblatt M
- Subjects
- Animals, Benzamides administration & dosage, Dioxoles administration & dosage, Disease Models, Animal, Drosophila Proteins genetics, Drosophila melanogaster, Female, Humans, Male, RNA, Messenger metabolism, Rhabdoid Tumor genetics, SMARCB1 Protein genetics, Smad3 Protein metabolism, Smad6 Protein metabolism, Teratoma genetics, Transcription Factors genetics, Transforming Growth Factor beta antagonists & inhibitors, Transforming Growth Factor beta metabolism, Drosophila Proteins metabolism, Rhabdoid Tumor metabolism, SMARCB1 Protein metabolism, Signal Transduction, Smad Proteins metabolism, Teratoma metabolism, Transcription Factors metabolism
- Abstract
Atypical teratoid/rhabdoid tumors (ATRT) are highly malignant brain tumors arising in young children. The majority of ATRT is characterized by inactivation of the chromatin remodeling complex member SMARCB1 (INI1/hSNF5). Little is known, however, on downstream pathways involved in the detrimental effects of SMARCB1 deficiency which might also represent targets for treatment. Using Drosophila melanogaster and the Gal4-UAS system, modifier screens were performed in order to identify the role of SMAD dependent signaling in the lethal phenotype associated with knockdown of snr1, the fly homolog of SMARCB1. Expression and functional role of human homologs was next investigated in ATRT tumor samples and SMARCB1-deficient rhabdoid tumor cells. The lethal phenotype associated with snr1 knockdown in Drosophila melanogaster could be shifted to later stages of development upon additional knockdown of several decapentaplegic pathway members including Smox, and Med. Similarly, the transforming growth factor beta (TGFbeta) receptor type I kinase inhibitor SB431542 ameliorated the detrimental effect of snr1 knockdown in the fruit fly. Examination of homologs of candidate decapentaplegic pathway members in human SMARCB1-deficent ATRT samples revealed SMAD3 and SMAD6 to be over-expressed. In SMARCB1-deficent rhabdoid tumor cells, siRNA-mediated silencing of SMAD3 or SMAD6 expression reduced TGFbeta signaling activity and resulted in decreased proliferation. Similar results were obtained upon pharmacological inhibition of TGFbeta signaling using SB431542. Our data suggest that SMAD dependent signaling is involved in the detrimental effects of SMARCB1-deficiency and provide a rationale for the investigation of TGFbeta targeted treatments in ATRT.
- Published
- 2017
- Full Text
- View/download PDF
34. Atypical teratoid/rhabdoid tumor arising in a malignant glioma.
- Author
-
Bozzai B, Hasselblatt M, Turányi E, Frühwald MC, Siebert R, Bens S, Schneppenheim R, Kool M, Stelczer G, Hortobágyi T, and Hauser P
- Subjects
- Child, Preschool, Combined Modality Therapy, Female, Glioma pathology, Humans, Neoplasm Recurrence, Local etiology, Neoplasm Staging, Neoplasms, Second Primary etiology, Prognosis, Rhabdoid Tumor etiology, Teratoma etiology, Glioma therapy, Neoplasm Recurrence, Local diagnosis, Neoplasms, Second Primary diagnosis, Rhabdoid Tumor diagnosis, Teratoma diagnosis
- Abstract
Atypical teratoid/rhabdoid tumor (AT/RT), a highly malignant brain tumor in young children, usually arises de novo and has only rarely been described as a secondary malignancy. Here, we present a case of a child with glioblastoma, who was treated postoperatively by a combination of temozolomide, irradiation, and bevacizumab. AT/RT was diagnosed as a secondary tumor, 2.5 years following primary diagnosis. The child died 13 months after the diagnosis of AT/RT. This case demonstrates that malignant gliomas may give rise to AT/RT. It also emphasizes the diagnostic value of a repeated tumor biopsy in the recurrence setting., (© 2016 Wiley Periodicals, Inc.)
- Published
- 2017
- Full Text
- View/download PDF
35. Atypical Teratoid/Rhabdoid Tumors Are Comprised of Three Epigenetic Subgroups with Distinct Enhancer Landscapes.
- Author
-
Johann PD, Erkek S, Zapatka M, Kerl K, Buchhalter I, Hovestadt V, Jones DTW, Sturm D, Hermann C, Segura Wang M, Korshunov A, Rhyzova M, Gröbner S, Brabetz S, Chavez L, Bens S, Gröschel S, Kratochwil F, Wittmann A, Sieber L, Geörg C, Wolf S, Beck K, Oyen F, Capper D, van Sluis P, Volckmann R, Koster J, Versteeg R, von Deimling A, Milde T, Witt O, Kulozik AE, Ebinger M, Shalaby T, Grotzer M, Sumerauer D, Zamecnik J, Mora J, Jabado N, Taylor MD, Huang A, Aronica E, Bertoni A, Radlwimmer B, Pietsch T, Schüller U, Schneppenheim R, Northcott PA, Korbel JO, Siebert R, Frühwald MC, Lichter P, Eils R, Gajjar A, Hasselblatt M, Pfister SM, and Kool M
- Subjects
- Brain Neoplasms genetics, Chromosomal Proteins, Non-Histone genetics, DNA-Binding Proteins genetics, Humans, Mutation genetics, SMARCB1 Protein, Transcription Factors genetics, Epigenesis, Genetic genetics, Rhabdoid Tumor genetics, Teratoma genetics
- Abstract
Atypical teratoid/rhabdoid tumor (ATRT) is one of the most common brain tumors in infants. Although the prognosis of ATRT patients is poor, some patients respond favorably to current treatments, suggesting molecular inter-tumor heterogeneity. To investigate this further, we genetically and epigenetically analyzed 192 ATRTs. Three distinct molecular subgroups of ATRTs, associated with differences in demographics, tumor location, and type of SMARCB1 alterations, were identified. Whole-genome DNA and RNA sequencing found no recurrent mutations in addition to SMARCB1 that would explain the differences between subgroups. Whole-genome bisulfite sequencing and H3K27Ac chromatin-immunoprecipitation sequencing of primary tumors, however, revealed clear differences, leading to the identification of subgroup-specific regulatory networks and potential therapeutic targets., (Copyright © 2016 Elsevier Inc. All rights reserved.)
- Published
- 2016
- Full Text
- View/download PDF
36. Molecular analyses reveal close similarities between small cell carcinoma of the ovary, hypercalcemic type and atypical teratoid/rhabdoid tumor.
- Author
-
Fahiminiya S, Witkowski L, Nadaf J, Carrot-Zhang J, Goudie C, Hasselblatt M, Johann P, Kool M, Lee RS, Gayden T, Roberts CW, Biegel JA, Jabado N, Majewski J, and Foulkes WD
- Subjects
- Cystadenocarcinoma, Serous genetics, DNA Helicases genetics, DNA Methylation, Epigenomics methods, Exome genetics, Female, Genome-Wide Association Study methods, Humans, Mutation, Nuclear Proteins genetics, Sequence Analysis, DNA methods, Transcription Factors genetics, Carcinoma, Small Cell genetics, Genetic Predisposition to Disease genetics, Hypercalcemia genetics, Ovarian Neoplasms genetics, Rhabdoid Tumor genetics, Teratoma genetics
- Abstract
Small cell carcinoma of the ovary, hypercalcemic type (SCCOHT) is the most common undifferentiated ovarian malignancy diagnosed in women under age 40. We and others recently determined that germline and/or somatic deleterious mutations in SMARCA4 characterize SCCOHT. Alterations in this gene, or the related SWI/SNF chromatin remodeling gene SMARCB1, have been previously reported in atypical teratoid/rhabdoid tumors (ATRTs) and malignant rhabdoid tumors (MRTs). To further describe the somatic landscape of SCCOHT, we performed whole exome sequencing on 14 tumors and their matched normal tissues and compared their genomic alterations with those in ATRT and ovarian high grade serous carcinoma (HGSC). We confirmed that SMARCA4 is the only recurrently mutated gene in SCCOHT, and show that recurrent allelic imbalance is observed exclusively on chromosome 19p, where SMARCA4 resides. By comparing genomic alterations between SCCOHT, ATRT and HGSC, we demonstrate that SCCOHTs, like ATRTs, have a remarkably simple genome and harbor significantly fewer somatic protein-coding mutations and chromosomal alterations than HGSC. Furthermore, a comparison of global DNA methylation profiles of 45 SCCOHTs, 65 ATRTs, and 92 HGSCs demonstrates a strong epigenetic correlation between SCCOHT and ATRT. Our results further confirm that the genomic and epigenomic signatures of SCCOHT are more similar to those of ATRT than HGSC, supporting our previous hypothesis that SCCOHT is a rhabdoid tumor and should be renamed MRT of the ovary. Furthermore, we conclude that SMARCA4 inactivation is the main cause of SCCOHT, and that new distinct therapeutic approaches should be developed to specifically target this devastating tumor.
- Published
- 2016
- Full Text
- View/download PDF
37. Analysis of the antiproliferative effects of 3-deazaneoplanocin A in combination with standard anticancer agents in rhabdoid tumor cell lines.
- Author
-
Unland R, Borchardt C, Clemens D, Kool M, Dirksen U, and Frühwald MC
- Subjects
- Adenosine administration & dosage, Adenosine pharmacology, Apoptosis drug effects, Apoptosis genetics, Azacitidine analogs & derivatives, Azacitidine pharmacology, Cell Line, Tumor drug effects, Cell Proliferation drug effects, Decitabine, Doxorubicin administration & dosage, Enhancer of Zeste Homolog 2 Protein, Epigenesis, Genetic, Etoposide administration & dosage, Gene Expression Regulation, Neoplastic drug effects, Humans, Hydroxamic Acids administration & dosage, Molecular Targeted Therapy methods, Polycomb Repressive Complex 2 antagonists & inhibitors, Rhabdoid Tumor genetics, Rhabdoid Tumor pathology, Vorinostat, Adenosine analogs & derivatives, Antineoplastic Combined Chemotherapy Protocols pharmacology, Rhabdoid Tumor drug therapy
- Abstract
Rhabdoid tumors (RTs) are highly aggressive pediatric malignancies with a rather poor prognosis. New therapeutic approaches and optimization of already established treatment protocols are urgently needed. The histone methyltransferase enhancer of zeste homolog 2 (EZH2) is highly overexpressed in RTs and associated strongly with epigenetic silencing in cancer. EZH2 is involved in aggressive cell growth and stem cell maintenance. Thus, EZH2 is an attractive therapeutic target in RTs. The aim of the study presented here was to analyze the effects of a pharmacological inhibition of EZH2 alone and in combination with other anticancer drugs on RTs cells in vitro. The antitumor activity of the S-adenosyl-homocysteine-hydrolase inhibitor 3-deazaneplanocin A (DZNep) alone and in combination with conventional cytostatic drugs (doxorubicin, etoposide) or epigenetic active compounds [5-Aza-CdR, suberoylanilide hydroxamic acid (SAHA)] was assessed by MTT cell proliferation assays on three RT cell lines (A204, BT16, G401). Combinatorial treatment with DZNep synergistically and significantly enhanced the antiproliferative activity of etoposide, 5-Aza-CdR, and SAHA. In functional analyses, pretreatment with DZNep significantly increased the effects of 5-Aza-CdR and SAHA on apoptosis, cell cycle progression, and clonogenicity. Microarray analyses following sequential treatment with DZNep and 5-Aza-CdR or SAHA showed changes in global gene expression affecting apoptosis, neuronal development, and metabolic processes. In-vitro analyses presented here show that pharmacological inhibition of EZH2 synergistically affects the antitumor activity of the epigenetic active compounds 5-Aza-CdR and SAHA. Sequential treatment with these drugs combined with DZNep may represent a new therapeutic approach in RTs.
- Published
- 2015
- Full Text
- View/download PDF
38. Arsenic trioxide inhibits tumor cell growth in malignant rhabdoid tumors in vitro and in vivo by targeting overexpressed Gli1.
- Author
-
Kerl K, Moreno N, Holsten T, Ahlfeld J, Mertins J, Hotfilder M, Kool M, Bartelheim K, Schleicher S, Handgretinger R, Schüller U, Meisterernst M, and Frühwald MC
- Subjects
- Animals, Apoptosis, Arsenic Trioxide, Cell Cycle, Cell Proliferation, Computational Biology, Gene Expression Profiling, Gene Expression Regulation, Hedgehog Proteins metabolism, Humans, Mice, Mice, SCID, Neoplasm Transplantation, Prognosis, Signal Transduction, Zinc Finger Protein GLI1, Antineoplastic Agents pharmacology, Arsenicals pharmacology, Gene Expression Regulation, Neoplastic, Kruppel-Like Transcription Factors metabolism, Oxides pharmacology, Rhabdoid Tumor drug therapy, Transcription Factors metabolism
- Abstract
Rhabdoid tumors are highly aggressive tumors occurring in infants and very young children. Despite multimodal and intensive therapy prognosis remains poor. Molecular analyses have uncovered several deregulated pathways, among them the CDK4/6-Rb-, the WNT- and the Sonic hedgehog (SHH) pathways. The SHH pathway is activated in rhabdoid tumors by GLI1 overexpression. Here, we demonstrate that arsenic trioxide (ATO) inhibits tumor cell growth of malignant rhabdoid tumors in vitro and in a mouse xenograft model by suppressing Gli1. Our data uncover ATO as a promising therapeutic approach to improve prognosis for rhabdoid tumor patients., (© 2014 UICC.)
- Published
- 2014
- Full Text
- View/download PDF
39. Identification of genes involved in the biology of atypical teratoid/rhabdoid tumours using Drosophila melanogaster.
- Author
-
Jeibmann A, Eikmeier K, Linge A, Kool M, Koos B, Schulz J, Albrecht S, Bartelheim K, Frühwald MC, Pfister SM, Paulus W, and Hasselblatt M
- Subjects
- Animals, Chromosomal Proteins, Non-Histone genetics, DNA-Binding Proteins genetics, Drosophila melanogaster, Gene Knockdown Techniques, Humans, Intracellular Signaling Peptides and Proteins genetics, Membrane Proteins genetics, Neurofibromin 2 genetics, Protein Serine-Threonine Kinases genetics, SMARCB1 Protein, Signal Transduction, Tumor Suppressor Proteins genetics, Brain Neoplasms genetics, Drosophila Proteins genetics, Rhabdoid Tumor genetics, Teratoma genetics, Transcription Factors genetics
- Abstract
Atypical teratoid/rhabdoid tumours (AT/RT) are malignant brain tumours. Unlike most other human brain tumours, AT/RT are characterized by inactivation of one single gene, SMARCB1. SMARCB1 is a member of the evolutionarily conserved SWI/SNF chromatin remodelling complex, which has an important role in the control of cell differentiation and proliferation. Little is known, however, about the pathways involved in the oncogenic effects of SMARCB1 inactivation, which might also represent targets for treatment. Here we report a comprehensive genetic screen in the fruit fly that revealed several genes not yet associated with loss of snr1, the Drosophila homologue of SMARCB1. We confirm the functional role of identified genes (including merlin, kibra and expanded, known to regulate hippo signalling pathway activity) in human rhabdoid tumour cell lines and AT/RT tumour samples. These results demonstrate that fly models can be employed for the identification of clinically relevant pathways in human cancer.
- Published
- 2014
- Full Text
- View/download PDF
40. The histone deacetylase inhibitor SAHA acts in synergism with fenretinide and doxorubicin to control growth of rhabdoid tumor cells.
- Author
-
Kerl K, Ries D, Unland R, Borchert C, Moreno N, Hasselblatt M, Jürgens H, Kool M, Görlich D, Eveslage M, Jung M, Meisterernst M, and Frühwald M
- Subjects
- Apoptosis drug effects, Blotting, Western, Cell Line, Tumor, Doxorubicin administration & dosage, Drug Synergism, Fenretinide administration & dosage, Humans, Hydroxamic Acids administration & dosage, Oligonucleotide Array Sequence Analysis, Real-Time Polymerase Chain Reaction, Rhabdoid Tumor pathology, Vorinostat, Antineoplastic Combined Chemotherapy Protocols pharmacology, Histone Deacetylase Inhibitors administration & dosage, Histone Deacetylases metabolism, Rhabdoid Tumor metabolism
- Abstract
Background: Rhabdoid tumors are highly aggressive malignancies affecting infants and very young children. In many instances these tumors are resistant to conventional type chemotherapy necessitating alternative approaches., Methods: Proliferation assays (MTT), apoptosis (propidium iodide/annexin V) and cell cycle analysis (DAPI), RNA expression microarrays and western blots were used to identify synergism of the HDAC (histone deacetylase) inhibitor SAHA with fenretinide, tamoxifen and doxorubicin in rhabdoidtumor cell lines., Results: HDAC1 and HDAC2 are overexpressed in primary rhabdoid tumors and rhabdoid tumor cell lines. Targeting HDACs in rhabdoid tumors induces cell cycle arrest and apoptosis. On the other hand HDAC inhibition induces deregulated gene programs (MYCC-, RB program and the stem cell program) in rhabdoid tumors. These programs are in general associated with cell cycle progression. Targeting these activated pro-proliferative genes by combined approaches of HDAC-inhibitors plus fenretinide, which inhibits cyclinD1, exhibit strong synergistic effects on induction of apoptosis. Furthermore, HDAC inhibition sensitizes rhabdoid tumor cell lines to cell death induced by chemotherapy., Conclusion: Our data demonstrate that HDAC inhibitor treatment in combination with fenretinide or conventional chemotherapy is a promising tool for the treatment of chemoresistant rhabdoid tumors.
- Published
- 2013
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.