Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 97
Filter
Add more filters

Country/Region as subject
Publication year range
1.
Cell ; 164(5): 1060-1072, 2016 Feb 25.
Article in English | MEDLINE | ID: mdl-26919435

ABSTRACT

Primitive neuroectodermal tumors of the central nervous system (CNS-PNETs) are highly aggressive, poorly differentiated embryonal tumors occurring predominantly in young children but also affecting adolescents and adults. Herein, we demonstrate that a significant proportion of institutionally diagnosed CNS-PNETs display molecular profiles indistinguishable from those of various other well-defined CNS tumor entities, facilitating diagnosis and appropriate therapy for patients with these tumors. From the remaining fraction of CNS-PNETs, we identify four new CNS tumor entities, each associated with a recurrent genetic alteration and distinct histopathological and clinical features. These new molecular entities, designated "CNS neuroblastoma with FOXR2 activation (CNS NB-FOXR2)," "CNS Ewing sarcoma family tumor with CIC alteration (CNS EFT-CIC)," "CNS high-grade neuroepithelial tumor with MN1 alteration (CNS HGNET-MN1)," and "CNS high-grade neuroepithelial tumor with BCOR alteration (CNS HGNET-BCOR)," will enable meaningful clinical trials and the development of therapeutic strategies for patients affected by poorly differentiated CNS tumors.


Subject(s)
Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/pathology , DNA Methylation , Neuroectodermal Tumors/genetics , Neuroectodermal Tumors/pathology , Amino Acid Sequence , Central Nervous System Neoplasms/classification , Central Nervous System Neoplasms/diagnosis , Child , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Molecular Sequence Data , Neuroectodermal Tumors/classification , Neuroectodermal Tumors/diagnosis , Proto-Oncogene Proteins/chemistry , Proto-Oncogene Proteins/genetics , Repressor Proteins/chemistry , Repressor Proteins/genetics , Signal Transduction , Trans-Activators , Tumor Suppressor Proteins/genetics
2.
Acta Neuropathol ; 145(1): 49-69, 2023 01.
Article in English | MEDLINE | ID: mdl-36437415

ABSTRACT

Pediatric central nervous system (CNS) tumors represent the most common cause of cancer-related death in children aged 0-14 years. They differ from their adult counterparts, showing extensive clinical and molecular heterogeneity as well as a challenging histopathological spectrum that often impairs accurate diagnosis. Here, we use DNA methylation-based CNS tumor classification in combination with copy number, RNA-seq, and ChIP-seq analysis to characterize a newly identified CNS tumor type. In addition, we report histology, patient characteristics, and survival data in this tumor type. We describe a biologically distinct pediatric CNS tumor type (n = 31 cases) that is characterized by focal high-level amplification and resultant overexpression of either PLAGL1 or PLAGL2, and an absence of recurrent genetic alterations characteristic of other pediatric CNS tumor types. Both genes act as transcription factors for a regulatory subset of imprinted genes (IGs), components of the Wnt/ß-Catenin pathway, and the potential drug targets RET and CYP2W1, which are also specifically overexpressed in this tumor type. A derived PLAGL-specific gene expression signature indicates dysregulation of imprinting control and differentiation/development. These tumors occurred throughout the neuroaxis including the cerebral hemispheres, cerebellum, and brainstem, and were predominantly composed of primitive embryonal-like cells lacking robust expression of markers of glial or neuronal differentiation (e.g., GFAP, OLIG2, and synaptophysin). Tumors with PLAGL1 amplification were typically diagnosed during adolescence (median age 10.5 years), whereas those with PLAGL2 amplification were diagnosed during early childhood (median age 2 years). The 10-year overall survival was 66% for PLAGL1-amplified tumors, 25% for PLAGL2-amplified tumors, 18% for male patients, and 82% for female patients. In summary, we describe a new type of biologically distinct CNS tumor characterized by PLAGL1/2 amplification that occurs predominantly in infants and toddlers (PLAGL2) or adolescents (PLAGL1) which we consider best classified as a CNS embryonal tumor and which is associated with intermediate survival. The cell of origin and optimal treatment strategies remain to be defined.


Subject(s)
Central Nervous System Neoplasms , Neuroectodermal Tumors, Primitive , Child , Child, Preschool , Female , Humans , Infant , Male , Cell Cycle Proteins/genetics , Central Nervous System Neoplasms/genetics , DNA Methylation , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Neuroectodermal Tumors, Primitive/genetics , RNA-Binding Proteins/genetics , Transcription Factors/genetics , Transcription Factors/metabolism , Tumor Suppressor Proteins/genetics , Wnt Signaling Pathway/genetics
3.
Acta Neuropathol ; 145(5): 667-680, 2023 05.
Article in English | MEDLINE | ID: mdl-36933012

ABSTRACT

Glioneuronal tumors are a heterogenous group of CNS neoplasms that can be challenging to accurately diagnose. Molecular methods are highly useful in classifying these tumors-distinguishing precise classes from their histological mimics and identifying previously unrecognized types of tumors. Using an unsupervised visualization approach of DNA methylation data, we identified a novel group of tumors (n = 20) that formed a cluster separate from all established CNS tumor types. Molecular analyses revealed ATRX alterations (in 16/16 cases by DNA sequencing and/or immunohistochemistry) as well as potentially targetable gene fusions involving receptor tyrosine-kinases (RTK; mostly NTRK1-3) in all of these tumors (16/16; 100%). In addition, copy number profiling showed homozygous deletions of CDKN2A/B in 55% of cases. Histological and immunohistochemical investigations revealed glioneuronal tumors with isomorphic, round and often condensed nuclei, perinuclear clearing, high mitotic activity and microvascular proliferation. Tumors were mainly located supratentorially (84%) and occurred in patients with a median age of 19 years. Survival data were limited (n = 18) but point towards a more aggressive biology as compared to other glioneuronal tumors (median progression-free survival 12.5 months). Given their molecular characteristics in addition to anaplastic features, we suggest the term glioneuronal tumor with ATRX alteration, kinase fusion and anaplastic features (GTAKA) to describe these tumors. In summary, our findings highlight a novel type of glioneuronal tumor driven by different RTK fusions accompanied by recurrent alterations in ATRX and homozygous deletions of CDKN2A/B. Targeted approaches such as NTRK inhibition might represent a therapeutic option for patients suffering from these tumors.


Subject(s)
Brain Neoplasms , Central Nervous System Neoplasms , Neoplasms, Neuroepithelial , Humans , Young Adult , Biomarkers, Tumor/genetics , Brain/pathology , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Gene Fusion , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Receptor Protein-Tyrosine Kinases/genetics , X-linked Nuclear Protein/genetics
4.
Gastrointest Endosc ; 97(1): 50-58.e4, 2023 01.
Article in English | MEDLINE | ID: mdl-35964683

ABSTRACT

BACKGROUND AND AIMS: Recent advances have introduced molecular subtyping of pancreatic cystic lesions (PCLs) as a possible amendment to the diagnostic algorithm. The study evaluated the feasibility and diagnostic accuracy of molecular analysis and subtyping of PCLs using the recently introduced EUS-guided through-the-needle-biopsy (TTNB) sampling. METHODS: We prospectively included 101 patients in the study who presented with PCLs >15 mm in the largest cross-section. EUS-guided TTNB samples were obtained by a micro-biopsy forceps introduced through a 19-gauge needle. The TTNB samples were analyzed by next-generation sequencing (NGS) for point mutations in tumor suppressors and oncogenes using a 51-gene customized hotspot panel. Sensitivity and specificity were calculated with the histologic diagnosis as reference. RESULTS: After initial microscopic evaluation of the samples, 91 patients had residual TTNB samples available for NGS. Of these, 49 harbored mutations, most frequently in KRAS and GNAS, reflecting an excess frequency of intraductal papillary mucinous neoplasms (IPMNs) in the study population. A sensitivity and specificity of 83.7% (95% confidence interval [CI], 70.3-92.7) and 81.8% (95% CI, 48.2-97.7), respectively, were demonstrated for the diagnosis of a mucinous cyst and 87.2% (95% CI, 74.2-95.2) and 84.6% (95% CI, 54.5-98.1) for the diagnosis of an IPMN. CONCLUSIONS: Thus, molecular analysis of TTNB samples by NGS has high sensitivity and specificity for diagnosing mucinous cysts and IPMNs. Although the procedure comes with a risk of adverse events of 9.9%, TTNB samples are a robust alternative to cyst fluid for a combined histologic and molecular diagnosis of PCLs. (Clinical trial registration number: NCT03578445.).


Subject(s)
Pancreatic Cyst , Pancreatic Neoplasms , Humans , Cyst Fluid , Endoscopic Ultrasound-Guided Fine Needle Aspiration/methods , High-Throughput Nucleotide Sequencing , Pancreas/pathology , Pancreatic Cyst/diagnosis , Pancreatic Cyst/genetics , Pancreatic Cyst/pathology , Pancreatic Neoplasms/diagnosis , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology
5.
PLoS Genet ; 16(12): e1009231, 2020 12.
Article in English | MEDLINE | ID: mdl-33332384

ABSTRACT

PURPOSE: Historically, cancer predisposition syndromes (CPSs) were rarely established for children with cancer. This nationwide, population-based study investigated how frequently children with cancer had or were likely to have a CPS. METHODS: Children (0-17 years) in Denmark with newly diagnosed cancer were invited to participate in whole-genome sequencing of germline DNA. Suspicion of CPS was assessed according to Jongmans'/McGill Interactive Pediatric OncoGenetic Guidelines (MIPOGG) criteria and familial cancer diagnoses were verified using population-based registries. RESULTS: 198 of 235 (84.3%) eligible patients participated, of whom 94/198 (47.5%) carried pathogenic variants (PVs) in a CPS gene or had clinical features indicating CPS. Twenty-nine of 198 (14.6%) patients harbored a CPS, of whom 21/198 (10.6%) harbored a childhood-onset and 9/198 (4.5%) an adult-onset CPS. In addition, 23/198 (11.6%) patients carried a PV associated with biallelic CPS. Seven of the 54 (12.9%) patients carried two or more variants in different CPS genes. Seventy of 198 (35.4%) patients fulfilled the Jongmans' and/or MIPOGG criteria indicating an underlying CPS, including two of the 9 (22.2%) patients with an adult-onset CPS versus 18 of the 21 (85.7%) patients with a childhood-onset CPS (p = 0.0022), eight of the additional 23 (34.8%) patients with a heterozygous PV associated with biallelic CPS, and 42 patients without PVs. Children with a central nervous system (CNS) tumor had family members with CNS tumors more frequently than patients with other cancers (11/44, p = 0.04), but 42 of 44 (95.5%) cases did not have a PV in a CPS gene. CONCLUSION: These results demonstrate the value of systematically screening pediatric cancer patients for CPSs and indicate that a higher proportion of childhood cancers may be linked to predisposing germline variants than previously supposed.


Subject(s)
Genetic Testing/statistics & numerical data , Germ-Line Mutation , Neoplastic Syndromes, Hereditary/epidemiology , Whole Genome Sequencing/statistics & numerical data , Adolescent , Child , Child, Preschool , Female , Humans , Incidence , Infant , Male , Mutation Rate , Neoplastic Syndromes, Hereditary/genetics
6.
Hum Mutat ; 43(1): 85-96, 2022 01.
Article in English | MEDLINE | ID: mdl-34816535

ABSTRACT

Heterozygous POLE or POLD1 germline pathogenic variants (PVs) cause polymerase proofreading associated polyposis (PPAP), a constitutional polymerase proofreading deficiency that typically presents with colorectal adenomas and carcinomas in adulthood. Constitutional mismatch-repair deficiency (CMMRD), caused by germline bi-allelic PVs affecting one of four MMR genes, results in a high propensity for the hematological, brain, intestinal tract, and other malignancies in childhood. Nonmalignant clinical features, such as skin pigmentation alterations, are found in nearly all CMMRD patients and are important diagnostic markers. Here, we excluded CMMRD in three cancer patients with highly suspect clinical phenotypes but identified in each a constitutional heterozygous POLE PV. These, and two additional POLE PVs identified in published CMMRD-like patients, have not previously been reported as germline PVs despite all being well-known somatic mutations in hyper-mutated tumors. Together, these five cases show that specific POLE PVs may have a stronger "mutator" effect than known PPAP-associated POLE PVs and may cause a CMMRD-like phenotype distinct from PPAP. The common underlying mechanism, that is, a constitutional replication error repair defect, and a similar tumor spectrum provide a good rationale for monitoring these patients with a severe constitutional polymerase proofreading deficiency according to protocols proposed for CMMRD.


Subject(s)
Brain Neoplasms , Colorectal Neoplasms , Neoplastic Syndromes, Hereditary , Adult , Brain Neoplasms/genetics , Colorectal Neoplasms/genetics , DNA Mismatch Repair/genetics , Humans , Mutation , Neoplastic Syndromes, Hereditary/diagnosis , Neoplastic Syndromes, Hereditary/genetics , Phenotype
7.
Neuropathol Appl Neurobiol ; 48(3): e12773, 2022 04.
Article in English | MEDLINE | ID: mdl-34799864

ABSTRACT

TERT promoter mutations have been associated with increased risk of recurrence in meningioma cohorts, thus a potential biomarker for aggressive phenotypes. A main purpose of refining tumour classification is better predictions on the patient level. We compiled data from previous published cohorts to investigate patient-level predictions of recurrence based on TERTp-mut status. Implementation of TERTp-mut into the WHO grading led to better patient prognostication by improved prediction of recurrence. Our results support implementation of TERTp-mut into diagnostics and classification of meningiomas.


Subject(s)
Meningeal Neoplasms , Meningioma , Telomerase , Humans , Meningeal Neoplasms/genetics , Meningeal Neoplasms/pathology , Meningioma/genetics , Meningioma/pathology , Mutation , Promoter Regions, Genetic/genetics , Telomerase/genetics , World Health Organization
8.
Acta Neuropathol ; 143(6): 697-711, 2022 06.
Article in English | MEDLINE | ID: mdl-35501487

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.


Subject(s)
Central Nervous System Neoplasms , Neoplasms, Neuroepithelial , Rhabdoid Tumor , Teratoma , Central Nervous System Neoplasms/genetics , DNA Methylation , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Neoplasms, Neuroepithelial/genetics , Prognosis , Rhabdoid Tumor/genetics , SMARCB1 Protein/genetics , SMARCB1 Protein/metabolism , Teratoma/genetics
9.
Eur J Nucl Med Mol Imaging ; 49(13): 4677-4691, 2022 Nov.
Article in English | MEDLINE | ID: mdl-35907033

ABSTRACT

PURPOSE: Both amino acid positron emission tomography (PET) and magnetic resonance imaging (MRI) blood volume (BV) measurements are used in suspected recurrent high-grade gliomas. We compared the separate and combined diagnostic yield of simultaneously acquired dynamic contrast-enhanced (DCE) perfusion MRI and O-(2-[18F]-fluoroethyl)-L-tyrosine ([18F]FET) PET in patients with anaplastic astrocytoma and glioblastoma following standard therapy. METHODS: A total of 76 lesions in 60 hybrid [18F]FET PET/MRI scans with DCE MRI from patients with suspected recurrence of anaplastic astrocytoma and glioblastoma were included retrospectively. BV was measured from DCE MRI employing a 2-compartment exchange model (2CXM). Diagnostic performances of maximal tumour-to-background [18F]FET uptake (TBRmax), maximal BV (BVmax) and normalised BVmax (nBVmax) were determined by ROC analysis using 6-month histopathological (n = 28) or clinical/radiographical follow-up (n = 48) as reference. Sensitivity and specificity at optimal cut-offs were determined separately for enhancing and non-enhancing lesions. RESULTS: In progressive lesions, all BV and [18F]FET metrics were higher than in non-progressive lesions. ROC analyses showed higher overall ROC AUCs for TBRmax than both BVmax and nBVmax in both lesion-wise (all lesions, p = 0.04) and in patient-wise analysis (p < 0.01). Combining TBRmax with BV metrics did not increase ROC AUC. Lesion-wise positive fraction/sensitivity/specificity at optimal cut-offs were 55%/91%/84% for TBRmax, 45%/77%/84% for BVmax and 59%/84%/72% for nBVmax. Combining TBRmax and best-performing BV cut-offs yielded lesion-wise sensitivity/specificity of 75/97%. The fraction of progressive lesions was 11% in concordant negative lesions, 33% in lesions only BV positive, 64% in lesions only [18F]FET positive and 97% in concordant positive lesions. CONCLUSION: The overall diagnostic accuracy of DCE BV imaging is good, but lower than that of [18F]FET PET. Adding DCE BV imaging did not improve the overall diagnostic accuracy of [18F]FET PET, but may improve specificity and allow better lesion-wise risk stratification than [18F]FET PET alone.


Subject(s)
Astrocytoma , Brain Neoplasms , Glioblastoma , Humans , Glioblastoma/diagnostic imaging , Brain Neoplasms/pathology , Retrospective Studies , Positron-Emission Tomography/methods , Astrocytoma/diagnostic imaging , Tyrosine/metabolism , Magnetic Resonance Imaging/methods , Perfusion , Magnetic Resonance Spectroscopy
10.
Neurosurg Rev ; 45(5): 3067-3081, 2022 Oct.
Article in English | MEDLINE | ID: mdl-35984552

ABSTRACT

Treatment-refractory meningiomas have a dismal prognosis and limited treatment options. Meningiomas express high-densities of somatostatin receptors (SSTR), thus potentially susceptible to antitumorigenic effects of somatostatin analogues (SSA). Evidence for SSA in meningiomas is scarce, and it is unclear if published literature would either (1) support wider use of SSA, if (2) more evidence is desirable, or if (3) available evidence is sufficient to discard SSA. We addressed the need for more evidence with a systematic review and meta-analysis. We performed an individual patient data (IPD) meta-analysis. Main outcomes were toxicity, best radiological response, progression-free survival, and overall survival. We applied multivariable logistic regression models to estimate the effect of SSA on the probability of obtaining radiological disease control. The predictive performance was evaluated using area under the curve and Brier scores. We included 16 studies and compiled IPD from 8/9 of all previous cohorts. Quality of evidence was overall ranked "very low." Stable disease was reported in 58% of patients as best radiological response. Per 100 mg increase in total SSA dosage, the odds ratios for obtaining radiological disease control was 1.42 (1.11 to 1.81, P = 0.005) and 1.44 (1.00 to 2.08, P = 0.05) for patients treated with SSA as monodrug therapy vs SSA in combination with everolimus, respectively. Low quality of evidence impeded exact quantification of treatment efficacy, and the association between response and treatment may represent reverse causality. Yet, the SSA treatment was well tolerated, and beneficial effect cannot be disqualified. A prospective trial without bias from inconsistent study designs is warranted to assess SSA therapy for well-defined meningioma subgroups.


Subject(s)
Meningeal Neoplasms , Meningioma , Everolimus/therapeutic use , Humans , Meningeal Neoplasms/drug therapy , Meningioma/drug therapy , Prospective Studies , Receptors, Somatostatin/therapeutic use , Somatostatin/therapeutic use
11.
Acta Neurochir (Wien) ; 164(1): 267-271, 2022 01.
Article in English | MEDLINE | ID: mdl-34748074

ABSTRACT

Meningiomas are benign lesion although anot insignificant number experiences recurrences despite Simpson grade 1 removal. FG001 is a compound with a fluorophore (ICG) that binds to a urokinase-type plasminogen activator receptor (uPAR) and is currently investigated at our institution in a first-in-human trial. The patient presented with a plausible malignant glioma but proved to be a grade 1 meningioma. FG001 could delineated the tumor not only on the surface but supplementary in the cavity to remove safely the dural attachment. We present FG001 as a new promising tool for improved surgical radicality beyond the intended indication, provided that a prospective validation in a consecutive meningioma cohort demonstrates similar results.


Subject(s)
Glioma , Meningeal Neoplasms , Meningioma , Child , Fluorescent Dyes , Humans , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/surgery , Meningioma/diagnostic imaging , Meningioma/surgery , Receptors, Urokinase Plasminogen Activator
12.
Acta Neurochir (Wien) ; 164(2): 343-352, 2022 02.
Article in English | MEDLINE | ID: mdl-34816314

ABSTRACT

INTRODUCTION: Malignant peripheral nerve sheath tumor of the vestibulocochlear nerve (VN-MPNST) is exceedingly rare and carries a poor prognosis. Little is known about its underlying genetics and in particular the process of malignant transformation. There is an ongoing debate on whether the transformation is initiated by ionizing radiation. We present here the analysis and comparison of two post-radiation VN-MPNST and one undergoing spontaneous transformation. METHODS: Four tumors from three patients (radiation-naïve vestibular schwannoma before (VS) and after (VN-MPNST) malignant transformation in addition to two post-radiation VN-MPNST) were subjected to DNA whole-genome microarray and whole-exome sequencing and tumor-specific mutations were called. Mutational signatures were characterized using MuSiCa. RESULTS: The tumor genomes were characterized predominantly by copy-number aberrations with 36-81% of the genome affected. Even the VS genome was grossly aberrated. The spontaneous malignant transformation was characterized by a near-total whole-genome doubling, disappearance of NF2 mutation and new mutations in three cancer-related genes (GNAQ, FOXO4 and PDGFRB). All tumors had homozygous loss of the tumor suppressor CDKN2A. Neither mutational signature nor copy number profile was associated with ionizing radiation. CONCLUSION: The VN-MPNST genome in our cases is characterized by large copy-number aberrations and homozygous deletion of CDKN2A. Our study demonstrates a VS with genetic alterations similar to its malignant counterpart, suggesting the existence of premalignant VS. No consistent mutational signature was associated with ionizing radiation.


Subject(s)
Nerve Sheath Neoplasms , Neuroma, Acoustic , Homozygote , Humans , Mutation/genetics , Neuroma, Acoustic/genetics , Neuroma, Acoustic/pathology , Sequence Deletion , Vestibulocochlear Nerve
13.
Acta Neurochir (Wien) ; 164(1): 229-238, 2022 01.
Article in English | MEDLINE | ID: mdl-34714434

ABSTRACT

INTRODUCTION: The extent of meningioma resection is the most fundamental risk factor for recurrence, and exact knowledge of extent of resection is necessary for prognostication and for planning of adjuvant treatment. Currently used classifications are the EANO-grading and the Simpson grading. The former comprises radiological imaging with contrast-enhanced MRI and differentiation between "gross total removal" and "subtotal removal," while the latter comprises a five-tiered differentiation of the surgeon's impression of the extent of resection. The extent of resection of tumors is usually defined via analyses of resection margins but has until now not been implemented for meningiomas. PET/MRI imaging with 68Ga-DOTATOC allows more sensitive and specific imaging than MRI following surgery of meningiomas. OBJECTIVE: To develop an objective grading system based on microscopic analyses of resection margins and sensitive radiological analyses to improve management of follow-up, adjuvant therapy, and prognostication of meningiomas. Based on the rationale of resection-margin analyses as gold standard and superior imaging performance of 68Ga DOTATOC PET, we propose "Copenhagen Grading" for meningiomas. RESULTS: Copenhagen Grading was described for six pilot patients with examples of positive and negative findings on histopathology and DOTATOC PET scanning. The grading could be traceably implemented and parameters of grading appeared complementary. Copenhagen Grading is prospectively implemented as a clinical standard at Rigshospitalet, Copenhagen. CONCLUSION: Copenhagen Grading provided a comprehensive, logical, and reproducible definition of the extent of resection. It offers promise to be the most sensitive and specific imaging modality available for meningiomas. Clinical and cost-efficacy remain to be established during prospective implementation.


Subject(s)
Meningeal Neoplasms , Meningioma , Humans , Meningeal Neoplasms/diagnostic imaging , Meningeal Neoplasms/surgery , Meningioma/diagnostic imaging , Meningioma/surgery , Neoplasm Grading , Neoplasm Recurrence, Local , Positron-Emission Tomography , Prospective Studies , Retrospective Studies
14.
Acta Neuropathol ; 142(5): 841-857, 2021 11.
Article in English | MEDLINE | ID: mdl-34417833

ABSTRACT

Large-scale molecular profiling studies in recent years have shown that central nervous system (CNS) tumors display a much greater heterogeneity in terms of molecularly distinct entities, cellular origins and genetic drivers than anticipated from histological assessment. DNA methylation profiling has emerged as a useful tool for robust tumor classification, providing new insights into these heterogeneous molecular classes. This is particularly true for rare CNS tumors with a broad morphological spectrum, which are not possible to assign as separate entities based on histological similarity alone. Here, we describe a molecularly distinct subset of predominantly pediatric CNS neoplasms (n = 60) that harbor PATZ1 fusions. The original histological diagnoses of these tumors covered a wide spectrum of tumor types and malignancy grades. While the single most common diagnosis was glioblastoma (GBM), clinical data of the PATZ1-fused tumors showed a better prognosis than typical GBM, despite frequent relapses. RNA sequencing revealed recurrent MN1:PATZ1 or EWSR1:PATZ1 fusions related to (often extensive) copy number variations on chromosome 22, where PATZ1 and the two fusion partners are located. These fusions have individually been reported in a number of glial/glioneuronal tumors, as well as extracranial sarcomas. We show here that they are more common than previously acknowledged, and together define a biologically distinct CNS tumor type with high expression of neural development markers such as PAX2, GATA2 and IGF2. Drug screening performed on the MN1:PATZ1 fusion-bearing KS-1 brain tumor cell line revealed preliminary candidates for further study. In summary, PATZ1 fusions define a molecular class of histologically polyphenotypic neuroepithelial tumors, which show an intermediate prognosis under current treatment regimens.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/pathology , Kruppel-Like Transcription Factors/genetics , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/pathology , Repressor Proteins/genetics , Biomarkers, Tumor/genetics , Child , Child, Preschool , Female , Humans , Male , Oncogene Fusion , Oncogene Proteins, Fusion/genetics
15.
Neurosurg Rev ; 43(2): 749-758, 2020 Apr.
Article in English | MEDLINE | ID: mdl-31183587

ABSTRACT

World Health Organization (WHO) grade I meningiomas are intracranial extracerebral tumors, in which microsurgery as a stand-alone therapy provides high rates of disease control and low recurrence rates. Our aim was to identify prognostic factors of overall survival and time-to-retreat (OS; TTR) in a cohort of patients with surgically managed WHO grade I meningioma. Patients with WHO grade I meningiomas from a retrospectively (1990 to 2002) and prospectively managed (2003 to 2010) databank of Oslo University Hospital, Norway, were included. The mean follow-up was 9.2 ± 5.7 years, with a total of 11,414 patient-years. One thousand three hundred fifty-five patients were included. The mean age was 58 ± 13.2, mean Karnofsky Performance Status (KPS) 92.6 ± 26.1 and female-to-male ratio 2.5:1. The 1-year, 5-year, 10-year, 15-year, and 20-year probabilities were 0.98, 0.91, 0.87, 0.84, and 0.8 for TTR. Patient age (OR 0.92 [0.91, 0.94]), male sex (OR 0.59 [0.45, 0.76]), preoperative KPS ≥ 70 (OR 2.22 [1.59, 3.13]), skull base location (OR 0.77 [0.60, 1]), and the occurrence of a postoperative hematoma (OR 0.44 [0.26, 0.76]) were identified as independent prognostic factors of OS. Patient age (OR 1.02 [1.01, 1.03]) and skull base location (OR 0.30 [0.21, 0.45]) were independent predictors of decreased PFS. Using a recursive partitioning analysis, we suggest a classification tree for the prediction of 5-year PFS based on patient and tumor characteristics. The findings from this cohort of meningioma WHO I patients helps to identify patients at risk of recurrence and tailor the therapeutic management.


Subject(s)
Meningeal Neoplasms/classification , Meningeal Neoplasms/mortality , Meningioma/classification , Meningioma/mortality , Adult , Aged , Aged, 80 and over , Cohort Studies , Disease Progression , Female , Humans , Male , Meningeal Neoplasms/surgery , Meningioma/surgery , Middle Aged , Norway , Prognosis , Retrospective Studies , World Health Organization , Young Adult
16.
Acta Neuropathol ; 138(6): 1075-1089, 2019 12.
Article in English | MEDLINE | ID: mdl-31414211

ABSTRACT

Spinal ependymal tumors form a histologically and molecularly heterogeneous group of tumors with generally good prognosis. However, their treatment can be challenging if infiltration of the spinal cord or dissemination throughout the central nervous system (CNS) occurs and, in these cases, clinical outcome remains poor. Here, we describe a new and relatively rare subgroup of spinal ependymal tumors identified using DNA methylation profiling that is distinct from other molecular subgroups of ependymoma. Copy number variation plots derived from DNA methylation arrays showed MYCN amplification as a characteristic genetic alteration in all cases of our cohort (n = 13), which was subsequently validated using fluorescence in situ hybridization. The histological diagnosis was anaplastic ependymoma (WHO Grade III) in ten cases and classic ependymoma (WHO Grade II) in three cases. Histological re-evaluation in five primary tumors and seven relapses showed characteristic histological features of ependymoma, namely pseudorosettes, GFAP- and EMA positivity. Electron microscopy revealed cilia, complex intercellular junctions and intermediate filaments in a representative sample. Taking these findings into account, we suggest to designate this molecular subgroup spinal ependymoma with MYCN amplification, SP-EPN-MYCN. SP-EPN-MYCN tumors showed distinct growth patterns with intradural, extramedullary localization mostly within the thoracic and cervical spine, diffuse leptomeningeal spread throughout the whole CNS and infiltrative invasion of the spinal cord. Dissemination was observed in 100% of cases. Despite high-intensity treatment, SP-EPN-MYCN showed significantly worse median progression free survival (PFS) (17 months) and median overall survival (OS) (87 months) than all other previously described molecular spinal ependymoma subgroups. OS and PFS were similar to supratentorial ependymoma with RELA-fusion (ST-EPN-RELA) and posterior fossa ependymoma A (PF-EPN-A), further highlighting the aggressiveness of this distinct new subgroup. We, therefore, propose to establish SP-EPN-MYCN as a new molecular subgroup in ependymoma and advocate for testing newly diagnosed spinal ependymal tumors for MYCN amplification.


Subject(s)
Ependymoma/genetics , Ependymoma/pathology , N-Myc Proto-Oncogene Protein/genetics , Spinal Neoplasms/genetics , Spinal Neoplasms/pathology , Adult , Brain Neoplasms/genetics , Brain Neoplasms/pathology , DNA Copy Number Variations/genetics , Female , Gene Expression Regulation, Neoplastic/genetics , Humans , Male , Mutation/genetics
17.
Acta Neuropathol ; 137(5): 837-846, 2019 05.
Article in English | MEDLINE | ID: mdl-30759284

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.


Subject(s)
Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Neoplasms, Neuroepithelial/genetics , Neoplasms, Neuroepithelial/metabolism , Protein Kinase C-alpha/genetics , Protein Kinase C-alpha/metabolism , Adolescent , Adult , Antigens, CD/genetics , Antigens, CD/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Brain/metabolism , Brain/pathology , Brain Neoplasms/pathology , Child , Cohort Studies , Female , Gene Fusion , Humans , Male , Middle Aged , Neoplasms, Neuroepithelial/pathology , Organic Cation Transport Proteins/genetics , Organic Cation Transport Proteins/metabolism , Site-Specific DNA-Methyltransferase (Adenine-Specific)
18.
Neurosurg Rev ; 42(1): 163-173, 2019 Mar.
Article in English | MEDLINE | ID: mdl-29627874

ABSTRACT

To identify differences between skull base meningiomas (SBM) and non-skull base meningiomas (NSBM). All adult patients (18.0-69.9 years) operated for intracranial meningiomas between 1990 and 2010 at our institution were investigated. Al-Mefty's definition was used to dichotomize tumors into SBM and NSBM. Overall, 1148 consecutive patients were identified. Median age at surgery was 54.2 years [18.1-69.9]. Median follow-up was 7.4 years [0.0-20.9]. There were 562 patients (49%) with SBM and 586 (51%) with NSBM. The two groups were similar with respect to patient age, follow-up time, and number of patients. Overall female-to-male ratio was 2.6:1, but 3.2:1 in SBM and 2.2:1 in NSBM (p < 0.005). With respect to presenting symptoms, SBMs had more often neurological deficits (risk ratio (RR) 1.4; p < 0.0001) and less often seizures (RR 0.4; p < 0.0001). Gross total resections were less frequent in SBM than NSBM (62 vs 84%) (RR 1.3; p < 0.0001). SBMs had a lower risk of WHO grades II and III histology (4.5 vs 9.5%) (RR 0.5; p < 0.001). Worsening of neurological function was more frequent in SBM (21 vs 121%) (RR 1.8; p < 0.001). Retreatment-free survival at 5, 10, and 15 years, respectively, was 80, 70, and 62% for SBM versus 90, 82, and 74% for NSBM (p < 0.0001). Overall survival at 5, 10, and 15 years, respectively, was 93, 85, and 78% for SBM and 96, 91, and 79% for NSBM (p = 0.14). Patients with SBMs had more new-onset neurological deficits and significantly shorter retreatment-free survivals, but this did not adversely affect the overall survival.


Subject(s)
Meningeal Neoplasms/surgery , Meningioma/surgery , Skull Base Neoplasms/surgery , Adolescent , Adult , Age Factors , Aged , Female , Follow-Up Studies , Humans , Male , Meningeal Neoplasms/pathology , Meningioma/pathology , Middle Aged , Nervous System Diseases/etiology , Progression-Free Survival , Reoperation/statistics & numerical data , Seizures/etiology , Sex Factors , Skull Base Neoplasms/pathology , Survival Analysis , Treatment Outcome , Young Adult
19.
Neurosurg Rev ; 42(4): 961-972, 2019 Dec.
Article in English | MEDLINE | ID: mdl-29978438

ABSTRACT

To compare outcomes after surgery for skull base meningiomas (SBMs) with non-skull base meningiomas (NSBMs) in the elderly. Overall, 128 consecutive patients ≥ 70 years of age with intracranial SBMs operated between 1990 and 2010 were compared to 193 consecutive patients ≥ 70 years of age with NSBMs operated within the same time period. Median age at surgery was 75.0 years (mean 75.7, range 70.0-92.4). Follow-up was complete with median 4.7 years (mean 5.5, range 0-19). The female-to-male ratio was 2.8 for SBMs and 1.3 for NSBMs (p < 0.005). The groups had similar preoperative KPS (median 80, range 20-100), but SBMs presented significantly more frequently with raised ICP (RR = 2.2, p < 0.005) and less frequently with seizures (RR = 0.6, p < 0.05). WHO I was significantly more frequent in SBMs (p < 0.005). Gross-total resection (GTR) was less frequent in SBMs (63 vs 82%) (RR = 2.1, p < 0.0001). SBMs were similar to NSBMs with respect to neurological outcome at 6-12 months, reoperations for hematomas, postoperative infections, and 30-day mortality. Retreatment rates and time to retreatments were also similar. There were no differences between the two groups with respect to risk of retreatment and overall survival (OS) at 5, 10, 15, and 20 years. In elderly patients with SBMs selected to surgical treatment, the risks of surgery, risk of retreatment, and OS were similar to NBSMs. Therefore, surgery for SBMs may be considered as safe as NSBMs in the elderly population.


Subject(s)
Meningeal Neoplasms/surgery , Meningioma/surgery , Postoperative Complications/epidemiology , Skull Base Neoplasms/surgery , Age Factors , Aged , Aged, 80 and over , Female , Humans , Male , Meningeal Neoplasms/pathology , Meningioma/pathology , Patient Selection , Reoperation , Skull Base , Skull Base Neoplasms/pathology , Treatment Outcome
SELECTION OF CITATIONS
SEARCH DETAIL