Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 89
Filtrar
1.
Acta Neuropathol Commun ; 12(1): 143, 2024 Sep 03.
Artículo en Inglés | MEDLINE | ID: mdl-39228008

RESUMEN

Neuroepithelial tumors with fusion of PLAGL1 or amplification of PLAGL1/PLAGL2 have recently been described often with ependymoma-like or embryonal histology respectively. To further evaluate emerging entities with PLAG-family genetic alterations, the histologic, molecular, clinical, and imaging features are described for 8 clinical cases encountered at St. Jude (EWSR1-PLAGL1 fusion n = 6; PLAGL1 amplification n = 1; PLAGL2 amplification n = 1). A histologic feature observed on initial resection in a subset (4/6) of supratentorial neuroepithelial tumors with EWSR1-PLAGL1 rearrangement was the presence of concurrent ependymal and ganglionic differentiation. This ranged from prominent clusters of ganglion cells within ependymoma/subependymoma-like areas, to interspersed ganglion cells of low to moderate frequency among otherwise ependymal-like histology, or focal areas with a ganglion cell component. When present, the combination of ependymal-like and ganglionic features within a supratentorial neuroepithelial tumor may raise consideration for an EWSR1-PLAGL1 fusion, and prompt initiation of appropriate molecular testing such as RNA sequencing and methylation profiling. One of the EWSR1-PLAGL1 fusion cases showed subclonal INI1 loss in a region containing small clusters of rhabdoid/embryonal cells, and developed a prominent ganglion cell component on recurrence. As such, EWSR1-PLAGL1 neuroepithelial tumors are a tumor type in which acquired inactivation of SMARCB1 and development of AT/RT features may occur and lead to clinical progression. In contrast, the PLAGL2 and PLAGL1 amplified cases showed either embryonal histology or contained an embryonal component with a significant degree of desmin staining, which could also serve to raise consideration for a PLAG entity when present. Continued compilation of associated clinical data and histopathologic findings will be critical for understanding emerging entities with PLAG-family genetic alterations.


Asunto(s)
Proteína EWS de Unión a ARN , Neoplasias Supratentoriales , Factores de Transcripción , Humanos , Neoplasias Supratentoriales/genética , Neoplasias Supratentoriales/patología , Femenino , Proteína EWS de Unión a ARN/genética , Masculino , Factores de Transcripción/genética , Niño , Neoplasias Neuroepiteliales/genética , Neoplasias Neuroepiteliales/patología , Preescolar , Adolescente , Adulto , Proteínas de Unión al ADN/genética , Adulto Joven , Diferenciación Celular , Proteínas de Fusión Oncogénica/genética , Epéndimo/patología , Reordenamiento Génico/genética , Proteínas Cromosómicas no Histona/genética
2.
J Neuropathol Exp Neurol ; 83(9): 708-721, 2024 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-38917431

RESUMEN

Mixed pituitary adenoma/PitNET-gangliocytomas (PA/PitNET-GC) have been reported in small series over the past 20 years; some had limited immunohistochemistry (IHC) data. We interrogated our experience over 20 years, focusing on patterns of the GC component and IHC results for anterior pituitary hormones, transcription factors, NFP, and CAM5.2. A search of cases from 2002 to 2023 yielded 20 cases: 7M:13F, ages 20-71 years; 17 macroadenomas, 1 microadenoma, 2 ectopic. GC was co-associated with 4 corticotroph, 2 densely granulated lactotroph, 5 mixed lactotroph-somatotroph, 1 immature PIT1-lineage tumor, and 8 sparsely granulated GH; the latter all had a minor lactotroph component. Patterns were: discrete nodular foci of GC (9/20), extensive GC differentiation often overshadowing the PA/PitNET (7/20), and intimate admixture of smaller bands of neuropil and individual metaplastic ganglion cells within PA/PitNET (4/20). NFP highlighted small cohesive regions of neuropil and identified greater axonal content, including individual axons within "pure" PA/PitNET areas, than appreciated on H&E. CAM5.2 IHC often revealed cells with neuronal morphologies to a greater extent than NFP and in different areas within the same tumor. These data suggest that the combined use of NFP and CAM5.2 IHC best reveals transition from PA to GC phenotype, with CAM5.2 positivity reflecting earlier stages of transformation.


Asunto(s)
Adenoma , Ganglioneuroma , Inmunohistoquímica , Neoplasias Hipofisarias , Humanos , Persona de Mediana Edad , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/metabolismo , Femenino , Masculino , Anciano , Adulto , Adenoma/patología , Adenoma/metabolismo , Ganglioneuroma/patología , Ganglioneuroma/metabolismo , Adulto Joven , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/metabolismo
3.
J Neuropathol Exp Neurol ; 83(7): 586-595, 2024 Jun 20.
Artículo en Inglés | MEDLINE | ID: mdl-38777808

RESUMEN

Corticotroph adenomas/pituitary neuroendocrine tumors (PitNETs) are associated with significant morbidity and mortality. Predictors of tumor behavior have not shown high prognostic accuracy. For somatotroph adenomas/PitNETs, E-cadherin expression correlates strongly with prognosis. E-cadherin expression has not been investigated in other PitNETs. A retrospective chart review of adults with corticotroph adenomas/PitNETs was conducted to assess correlation between E-cadherin expression and tumor characteristics. In addition, gene expression microarray was performed in subset of tumors (n = 16). Seventy-seven patients were identified; 71% were female, with median age of cohort 45.2 years. Seventy-five percent had macroadenomas, of which 22% were hormonally active. Ninety-five percent of microadenomas were hormonally active. Adrenocorticotropic hormone granulation pattern by IHC identified 63% as densely granulated (DG) and 34% as sparsely granulated (SG). All microadenomas were DG (p < .001); 50% of macroadenomas were DG associated with increased tumor invasion compared to SG. E-cadherin IHC was positive in 80%, diminished in 17%, and absent in 20% and did not correlate with corticotroph PitNETs subtype, size, or prognosis. In contrast to the distinct transcriptomes of corticotroph PitNETs and normal pituitaries, a comparison of clinically active and silent corticotroph PitNETs demonstrated similar molecular signatures indicating their common origin, but with unique differences related to their secretory status.


Asunto(s)
Adenoma Hipofisario Secretor de ACTH , Cadherinas , Tumores Neuroendocrinos , Adulto , Anciano , Femenino , Humanos , Masculino , Persona de Mediana Edad , Adulto Joven , Adenoma Hipofisario Secretor de ACTH/genética , Adenoma Hipofisario Secretor de ACTH/patología , Adenoma Hipofisario Secretor de ACTH/metabolismo , Cadherinas/genética , Cadherinas/biosíntesis , Cadherinas/metabolismo , Perfilación de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Tumores Neuroendocrinos/genética , Tumores Neuroendocrinos/patología , Tumores Neuroendocrinos/metabolismo , Neoplasias Hipofisarias/genética , Neoplasias Hipofisarias/patología , Neoplasias Hipofisarias/metabolismo , Estudios Retrospectivos , Transcriptoma
4.
Clin Neuropathol ; 43(1): 10-22, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38085092

RESUMEN

Two of the rarest radiation-induced adverse effects are focal neuronal gigantism (FNG) and SMART syndrome (stroke-like migraine attacks after radiation therapy). Both conditions develop years, and sometimes decades, after receipt of therapeutic radiation to the brain. To date, there are only 3 previously reported cases of FNG, all of which describe cortical thickening, enlarged "hypertrophic" neurons, and neuronal cytological changes. No detailed studies exist of histological features of SMART or the comparison between FNG and SMART. In this study, we contrast histological and neuroimaging features of 3 FNG vs. 4 SMART cases, the latter diagnosed by a neuroradiologist, neurooncologist, and/or neurosurgeon. We confirm the cortical thickening, dyslamination, neuronal cytomegaly, and gliosis in FNG vs. cortical architectural preservation and normal neuronal cytology in SMART, although both showed gliosis, scattered neurons with cytoplasmic accumulation of tau and neurofibrillary protein and variable co-existence of other radiation-induced lesions. Both conditions lacked significant inflammation or consistent small vessel hyalinization throughout the entire resection specimen. The absence of pathognomonic histologic alterations in SMART cases suggests underlying vascular dysregulation. Despite differing histology, some overlap may exist in neuroimaging features. Molecular assessment conducted in 2 cases of FNG was negative for significant alterations including in the MAPK pathway.


Asunto(s)
Gigantismo , Humanos , Gigantismo/patología , Gliosis/patología , Encéfalo/patología , Neuroimagen , Neuronas
5.
Clin Neuropathol ; 42(2): 54-65, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36708209

RESUMEN

AIMS: Central nervous system (CNS) and spine are seldom impacted by primary or metastatic sarcomas. We reviewed our 22-year experience with metastatic versus primary mesenchymal sarcomas in adults versus pediatric patients, additionally asking how many might today undergo nomenclature changes using CNS World Health Organization, 5th edition criteria. MATERIALS AND METHODS: Case identification via text word search of pathology databases from our adult and pediatric referral hospitals, 2000 to August 2022, with exclusion of peripheral nervous system and primary chondro-osseous and notochordal tumors. Demographic, immunohistochemical, fluorescence in situ hybridization (FISH), and fusion results performed at the time of original diagnosis were acquired from reports. RESULTS: 57 cases were identified, with a 16 : 15 primary and 19 : 7 metastatic ratio in adult versus pediatric patients. Ewing sarcoma was the most frequent type (n = 18, 7 adult, 11 pediatric), with a rare primary PEComa, 2 alveolar soft part sarcomas, and metastatic angiosarcoma in the cohort. Only 3 cases, an intracranial sarcoma, DICER-1 mutant formerly diagnosed as rhabdomyosarcoma, an intracranial mesenchymal tumor, FET::CREB fusion-positive formerly diagnosed as angiomatoid fibrous histiocytoma, and a CIC-rearranged sarcoma required nomenclature updating by CNS WHO5 criteria. CONCLUSIONS: Few primary or metastatic, adult or pediatric, CNS/spinal sarcomas required nomenclature updates; almost all had been satisfactorily classified at the time of diagnosis, using immunohistochemistry, FISH, or fusion results.


Asunto(s)
Neoplasias Óseas , Neoplasias Encefálicas , Rabdomiosarcoma , Sarcoma de Ewing , Sarcoma , Humanos , Hibridación Fluorescente in Situ , Sarcoma/patología , Sarcoma de Ewing/diagnóstico , Sarcoma de Ewing/patología , Neoplasias Óseas/patología , Neoplasias Encefálicas/genética , Biomarcadores de Tumor
6.
J Neuropathol Exp Neurol ; 82(3): 194-201, 2023 02 21.
Artículo en Inglés | MEDLINE | ID: mdl-36637258

RESUMEN

Leptomeningeal (LM) metastases or dural spread by adult high-grade astrocytomas are problematic; it is unclear which tumor types are predisposed to spread and at what time intervals from original diagnosis. We reviewed our recent experience with these tumor types with LM or dural spread, all of which had assessments that allowed CNS World Health Organization, 5th Edition classification. Following a database search, 2018-present, 10 patients were identified: 4 glioblastomas, IDH-wildtype, WHO grade 4; 4 astrocytomas, IDH-mutant, WHO grade 4; 1 high-grade astrocytoma with piloid features (HGAP) proven by DNA methylation, and 1 high-grade astrocytic tumor that fell closest to the HGAP category by DNA methylation. Most had LM dissemination; 2 had dural spread. Intervals from initial tumor diagnosis to LM spread for 4 astrocytomas, IDH-mutant were 1, 6, 7, and 14 years. Two glioblastomas, IDH-wildtype had dural spread at the time of diagnosis; 1 had a 6-year interval to metastasis; and 1 had a 3-month interval to LM spread. The definite HGAP showed an interval of 7 years to metastasis and the possible HGAP had LM spread recognized at the time of initial diagnosis. All adult high-grade astrocytic tumor types are capable of LM or dural spread, including HGAP.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Carcinomatosis Meníngea , Adulto , Humanos , Neoplasias Encefálicas/patología , Glioblastoma/secundario , Isocitrato Deshidrogenasa/genética , Carcinomatosis Meníngea/secundario , Mutación , Clasificación del Tumor
7.
Pediatr Dev Pathol ; 26(1): 18-29, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36377607

RESUMEN

BACKGROUND: Childhood (c) primary angiitis of the central nervous system (PACNS) is a rare condition that most often affects small vessels (SV), is nearly exclusively lymphocytic, and devoid of vessel necrosis. Diagnosis of cSV-PACNS is challenging. We noted possible histological overlap of cSV-PACNS with myelin oligodendrocyte glycoprotein disease (MOGAD) on biopsy, prompting a 10-year retrospective review of our experience. MATERIALS AND METHODS: Database-search for brain biopsy cases, age <18 years, performed for an acquired neurological deficit with suspicion of vasculitis, with histological evidence of lymphocytic small-vessel inflammation. RESULTS: We identified 7 patients; 2/7 were serum-positive for anti-MOG antibodies and 1/7 for anti-NMDA antibodies. The remaining 4/7 proved to be idiopathic lymphocytic vasculitis/cSV-PACNS. All 7 showed overlapping features of lymphocytes permeating parenchymal SV walls, vessel wall distortion without fibrinoid necrosis, and absence of microglial clusters or intravascular thrombi. Tissue infarction was confined to a single case of idiopathic lymphocytic vasculitis. Although demyelination was diligently sought, only subtle demyelination was identified in the 2 MOGAD cases and absent in the remainder. CONCLUSION: There is considerable histological overlap between cSV-PACNS and at least some cases of MOGAD or anti-NMDA-encephalitis; at diagnosis, the differential should include cSV-PACNS but correct classification requires post-biopsy serological testing.


Asunto(s)
Enfermedades Desmielinizantes , Vasculitis del Sistema Nervioso Central , Humanos , Niño , Encéfalo/patología , Imagen por Resonancia Magnética , Vasculitis del Sistema Nervioso Central/diagnóstico , Vasculitis del Sistema Nervioso Central/complicaciones , Vasculitis del Sistema Nervioso Central/patología , Necrosis/patología , Enfermedades Desmielinizantes/complicaciones , Enfermedades Desmielinizantes/patología
9.
Brain Pathol ; 32(4): e13037, 2022 07.
Artículo en Inglés | MEDLINE | ID: mdl-34821426

RESUMEN

'Intracranial mesenchymal tumor, FET-CREB fusion-positive' occurs primarily in children and young adults and has previously been termed intracranial angiomatoid fibrous histiocytoma (AFH) or intracranial myxoid mesenchymal tumor (IMMT). Here we performed genome-wide DNA methylation array profiling of 20 primary intracranial mesenchymal tumors with FET-CREB fusion to further study their ontology. These tumors resolved into two distinct epigenetic subgroups that were both divergent from all other analyzed intracranial neoplasms and soft tissue sarcomas, including meningioma, clear cell sarcoma of soft tissue (CCS), and AFH of extracranial soft tissue. The first subgroup (Group A, 16 tumors) clustered nearest to but independent of solitary fibrous tumor and AFH of extracranial soft tissue, whereas the second epigenetic subgroup (Group B, 4 tumors) clustered nearest to but independent of CCS and also lacked expression of melanocytic markers (HMB45, Melan A, or MITF) characteristic of CCS. Group A tumors most often occurred in adolescence or early adulthood, arose throughout the neuroaxis, and contained mostly EWSR1-ATF1 and EWSR1-CREB1 fusions. Group B tumors arose most often in early childhood, were located along the cerebral convexities or spinal cord, and demonstrated an enrichment for tumors with CREM as the fusion partner (either EWSR1-CREM or FUS-CREM). Group A tumors more often demonstrated stellate/spindle cell morphology and hemangioma-like vasculature, whereas Group B tumors more often demonstrated round cell or epithelioid/rhabdoid morphology without hemangioma-like vasculature, although robust comparison of these clinical and histologic features requires future study. Patients with Group B tumors had inferior progression-free survival relative to Group A tumors (median 4.5 vs. 49 months, p = 0.001). Together, these findings confirm that intracranial AFH-like neoplasms and IMMT represent histologic variants of a single tumor type ('intracranial mesenchymal tumor, FET-CREB fusion-positive') that is distinct from meningioma and extracranial sarcomas. Additionally, epigenomic evaluation may provide important prognostic subtyping for this unique tumor entity.


Asunto(s)
Neoplasias Encefálicas , Hemangioma , Histiocitoma Fibroso Maligno , Neoplasias Meníngeas , Meningioma , Neoplasias de los Tejidos Blandos , Adolescente , Adulto , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Niño , Preescolar , Epigénesis Genética , Epigenómica , Hemangioma/genética , Histiocitoma Fibroso Maligno/genética , Humanos , Neoplasias Meníngeas/genética , Meningioma/genética , Proteínas de Fusión Oncogénica/genética , Proteína EWS de Unión a ARN/genética , Neoplasias de los Tejidos Blandos/genética , Neoplasias de los Tejidos Blandos/patología , Adulto Joven
10.
Clin Neuropathol ; 41(2): 46-52, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-34958303

RESUMEN

AIMS: Cerebral amyloidomas (CAs) are mass-producing congophilic lesions most commonly due to λ light chain deposits, contrasting them with light chain deposition disease (LCDD) which has non-polarizable, often κ light chain deposition. MATERIALS AND METHODS: Although usual histological features are well known, we detail 3 recent CAs with unusual morphological findings and review the literature specifically for these features. RESULTS: Two women, aged 56 and 58 years, had right cerebral white matter CAs. The biopsy of case 1 disclosed congophilic polarizable deposits with prominent dystrophic mineralization as well as scant plasma cells. Case 2 had a CA with significant multinucleated giant cell reaction to the amyloid and additionally contained an area suspicious for marginal zone B-cell lymphoma. Case 3 was a clinically unsuspected CA identified at autopsy in a 75-year-old woman that manifested as several contiguous left frontal lobe white matter erythematous, hyperemic lesions; microscopy showed nodular and concentric amyloid deposits and thick perivascular cuffs of plasma cells. Mass spectrometry proved λ light and α heavy chain amyloid deposits in all 3 cases. CONCLUSION: These 3 CA cases illustrate several unusual gross and microscopic features that are discussed in context with the literature.


Asunto(s)
Amiloidosis , Linfoma de Células B , Neoplasias de los Tejidos Blandos , Anciano , Amiloide/análisis , Amiloidosis/diagnóstico , Amiloidosis/patología , Femenino , Humanos , Linfoma de Células B/patología , Persona de Mediana Edad , Células Plasmáticas/patología , Neoplasias de los Tejidos Blandos/patología
11.
Acta Neuropathol ; 142(6): 1025-1043, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34661724

RESUMEN

Pituicytoma (PITUI), granular cell tumor (GCT), and spindle cell oncocytoma (SCO) are rare tumors of the posterior pituitary. Histologically, they may be challenging to distinguish and have been proposed to represent a histological spectrum of a single entity. We performed targeted next-generation sequencing, DNA methylation profiling, and copy number analysis on 47 tumors (14 PITUI; 12 GCT; 21 SCO) to investigate molecular features and explore possibilities of clinically meaningful tumor subclassification. We detected two main epigenomic subgroups by unsupervised clustering of DNA methylation data, though the overall methylation differences were subtle. The largest group (n = 23) contained most PITUIs and a subset of SCOs and was enriched for pathogenic mutations within genes in the MAPK/PI3K pathways (12/17 [71%] of sequenced tumors: FGFR1 (3), HRAS (3), BRAF (2), NF1 (2), CBL (1), MAP2K2 (1), PTEN (1)) and two with accompanying TERT promoter mutation. The second group (n = 16) contained most GCTs and a subset of SCOs, all of which mostly lacked identifiable genetic drivers. Outcome analysis demonstrated that the presence of chromosomal imbalances was significantly associated with reduced progression-free survival especially within the combined PITUI and SCO group (p = 0.031). In summary, we observed only subtle DNA methylation differences between posterior pituitary tumors, indicating that these tumors may be best classified as subtypes of a single entity. Nevertheless, our data indicate differences in mutation patterns and clinical outcome. For a clinically meaningful subclassification, we propose a combined histo-molecular approach into three subtypes: one subtype is defined by granular cell histology, scarcity of identifiable oncogenic mutations, and favorable outcome. The other two subtypes have either SCO or PITUI histology but are segregated by chromosomal copy number profile into a favorable group (no copy number changes) and a less favorable group (copy number imbalances present). Both of the latter groups have recurrent MAPK/PI3K genetic alterations that represent potential therapeutic targets.


Asunto(s)
Adenoma Oxifílico/genética , Tumor de Células Granulares/genética , Neoplasias Hipofisarias/genética , Epigénesis Genética , Humanos
15.
Clin Neuropathol ; 40(1): 4-10, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-32631482

RESUMEN

Although increasing numbers of central nervous system (CNS) tumors with stereotypic morphological, molecular, and/or site-specific features have been recently reported, morphological diversity is often recognized within a tumor category as more cases are encountered. Such was the case with diffuse midline gliomas, H3K27M-mutant. Therefore, it is not surprising that two cases of H3G34-mutant. CNS tumors with advanced ganglionic differentiation were recently published, especially given the posited role of this mutation in neuronal differentiation. We have encountered, in a 17-year-old female, a third example with advanced neoplastic ganglion cell differentiation that mimicked anaplastic ganglioglioma, with the ganglionic elements further confirmed as neoplastic by H3G34 immunohistochemistry (IHC). We therefore sought to review our experience with H3G34-mutant tumors, assessing for morphological diversity, supplemented by IHC. Six cases (ages 17 - 33 years), all confirmed on mutational analyses, were identified that were further negative for BRAFV600E or other major oncogenic mutations/fusions. The index anaplastic ganglioglioma-like case manifested multifocal large dysmorphic ganglion cells IHC+ for synaptophysin, chromogranin, and neurofilament, but no CD34 immunopositivity. A tumor from a 33-year-old male contained rare neuronal-like cells, with subtler enlargement, that were synaptophysin and neurofilament protein IHC+ and exceeded the size expected in "primitive neuroectodermal like (PNET)" tumors. A third example with morphological diversity was a glioblastoma with prominent epithelioid cells. We conclude that a spectrum of morphological differentiation does occur beyond the well-known glioblastoma or PNET-like morphology in H3G34-mutant tumors, adding to the literature one more example with advanced ganglionic differentiation and one with epithelioid features.


Asunto(s)
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Glioma/genética , Glioma/patología , Histonas/genética , Mutación/genética , Adolescente , Adulto , Neoplasias Encefálicas/metabolismo , Estudios de Cohortes , Femenino , Glioma/metabolismo , Humanos , Masculino , Adulto Joven
16.
Brain Pathol ; 31(4): e12918, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33141488

RESUMEN

Intracranial mesenchymal tumors with FET-CREB fusions are a recently described group of neoplasms in children and young adults characterized by fusion of a FET family gene (usually EWSR1, but rarely FUS) to a CREB family transcription factor (ATF1, CREB1, or CREM), and have been variously termed intracranial angiomatoid fibrous histiocytoma or intracranial myxoid mesenchymal tumor. The clinical outcomes, histologic features, and genomic landscape are not well defined. Here, we studied 20 patients with intracranial mesenchymal tumors proven to harbor FET-CREB fusion by next-generation sequencing (NGS). The 16 female and four male patients had a median age of 14 years (range 4-70). Tumors were uniformly extra-axial or intraventricular and located at the cerebral convexities (n = 7), falx (2), lateral ventricles (4), tentorium (2), cerebellopontine angle (4), and spinal cord (1). NGS demonstrated that eight tumors harbored EWSR1-ATF1 fusion, seven had EWSR1-CREB1, four had EWSR1-CREM, and one had FUS-CREM. Tumors were uniformly well circumscribed and typically contrast enhancing with solid and cystic growth. Tumors with EWSR1-CREB1 fusions more often featured stellate/spindle cell morphology, mucin-rich stroma, and hemangioma-like vasculature compared to tumors with EWSR1-ATF1 fusions that most often featured sheets of epithelioid cells with mucin-poor collagenous stroma. These tumors demonstrated polyphenotypic immunoprofiles with frequent positivity for desmin, EMA, CD99, MUC4, and synaptophysin, but absence of SSTR2A, myogenin, and HMB45 expression. There was a propensity for local recurrence with a median progression-free survival of 12 months and a median overall survival of greater than 60 months, with three patients succumbing to disease (all with EWSR1-ATF1 fusions). In combination with prior case series, this study provides further insight into intracranial mesenchymal tumors with FET-CREB fusion, which represent a distinct group of CNS tumors encompassing both intracranial myxoid mesenchymal tumor and angiomatoid fibrous histiocytoma-like neoplasms.


Asunto(s)
Neoplasias Encefálicas/patología , Histiocitoma Fibroso Benigno/patología , Histiocitoma Fibroso Maligno/patología , Proteínas de Fusión Oncogénica/metabolismo , Adolescente , Adulto , Anciano , Biomarcadores de Tumor/genética , Neoplasias Encefálicas/diagnóstico , Neoplasias Encefálicas/genética , Niño , Preescolar , Femenino , Fusión Génica/genética , Histiocitoma Fibroso Benigno/diagnóstico , Histiocitoma Fibroso Benigno/metabolismo , Histiocitoma Fibroso Maligno/diagnóstico , Histiocitoma Fibroso Maligno/genética , Humanos , Masculino , Persona de Mediana Edad , Proteínas de Fusión Oncogénica/genética , Adulto Joven
17.
Acta Neuropathol Commun ; 8(1): 151, 2020 08 28.
Artículo en Inglés | MEDLINE | ID: mdl-32859279

RESUMEN

The FGFR1 gene encoding fibroblast growth factor receptor 1 has emerged as a frequently altered oncogene in the pathogenesis of multiple low-grade neuroepithelial tumor (LGNET) subtypes including pilocytic astrocytoma, dysembryoplastic neuroepithelial tumor (DNT), rosette-forming glioneuronal tumor (RGNT), and extraventricular neurocytoma (EVN). These activating FGFR1 alterations in LGNET can include tandem duplication of the exons encoding the intracellular tyrosine kinase domain, in-frame gene fusions most often with TACC1 as the partner, or hotspot missense mutations within the tyrosine kinase domain (either at p.N546 or p.K656). However, the specificity of these different FGFR1 events for the various LGNET subtypes and accompanying genetic alterations are not well defined. Here we performed comprehensive genomic and epigenomic characterization on a diverse cohort of 30 LGNET with FGFR1 alterations. We identified that RGNT harbors a distinct epigenetic signature compared to other LGNET with FGFR1 alterations, and is uniquely characterized by FGFR1 kinase domain hotspot missense mutations in combination with either PIK3CA or PIK3R1 mutation, often with accompanying NF1 or PTPN11 mutation. In contrast, EVN harbors its own distinct epigenetic signature and is characterized by FGFR1-TACC1 fusion as the solitary pathogenic alteration. Additionally, DNT and pilocytic astrocytoma are characterized by either kinase domain tandem duplication or hotspot missense mutations, occasionally with accompanying NF1 or PTPN11 mutation, but lacking the accompanying PIK3CA or PIK3R1 mutation that characterizes RGNT. The glial component of LGNET with FGFR1 alterations typically has a predominantly oligodendroglial morphology, and many of the pilocytic astrocytomas with FGFR1 alterations lack the biphasic pattern, piloid processes, and Rosenthal fibers that characterize pilocytic astrocytomas with BRAF mutation or fusion. Together, this analysis improves the classification and histopathologic stratification of LGNET with FGFR1 alterations.


Asunto(s)
Neoplasias Neuroepiteliales/clasificación , Neoplasias Neuroepiteliales/genética , Neoplasias Neuroepiteliales/patología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/genética , Adolescente , Adulto , Anciano , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patología , Niño , Femenino , Humanos , Masculino , Persona de Mediana Edad , Mutación , Neoplasias de la Médula Espinal/clasificación , Neoplasias de la Médula Espinal/genética , Neoplasias de la Médula Espinal/patología , Adulto Joven
18.
Acta Neurochir (Wien) ; 162(8): 1941-1947, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32291590

RESUMEN

BACKGROUND: The purpose of this study was to assess the reliability of fluorescein sodium in predicting conclusive tissue diagnosis in stereotactic brain biopsies and to characterize features of contrast-enhancing and non-enhancing MRI lesions associated with fluorescence. METHODS: A total of 19 patients were studied, 14 of which had contrast-enhancing and 5 of which had non-enhancing lesions on preoperative T1 post-gadolinium MRI scan. All patients received 3 mg/kg fluorescein sodium during anesthesia induction. Biopsy specimens were photographed under the operating microscope, using the Yellow560 module, prior to histopathological analysis. Two observers blinded to the MRI scans and histopathological results categorized the photographs retrospectively as "fluorescent" or "not fluorescent." Inter-rater agreement was assessed using Cohen's kappa coefficient. Sensitivity, specificity, and positive predictive value of fluorescence reliability were calculated for MRI contrast-enhancing lesions and confirmed location-concordance of tumor pathology based on rater's fluorescence status assessment. Results were correlated finally with final results on permanent sections. RESULTS: Strength of inter-rater fluorescence status agreement was found to be "substantial" (kappa = 0.771). Sensitivity, specificity, and positive predictive value for "fluorescent" and "not fluorescent" specimen in comparison with MRI contrast-enhancing lesions were 97%, 40%, and 82%, respectively. Sensitivity, specificity, and positive predictive value for confirmed tumor pathology were 100%, 63%, and 91%, respectively. Permanent pathology revealed high-grade glioma n = 5, low-grade glioma n = 3, lymphoma n = 5, pineal tumor n = 2, hamartoma n = 1, and nonspecific hypercellularity n = 3. CONCLUSIONS: Fluorescein-assisted stereotactic brain biopsies demonstrated a high likelihood to manifest fluorescence in contrast-enhancing MRI lesions, while adequately predicting conclusive tumor pathology.


Asunto(s)
Neoplasias Encefálicas/patología , Fluoresceína/normas , Glioma/patología , Técnicas Estereotáxicas/normas , Adulto , Neoplasias Encefálicas/diagnóstico por imagen , Neoplasias Encefálicas/cirugía , Femenino , Glioma/diagnóstico por imagen , Glioma/cirugía , Humanos , Imagen por Resonancia Magnética , Masculino , Persona de Mediana Edad , Reproducibilidad de los Resultados
19.
Pediatr Blood Cancer ; 67(1): e28028, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31595628

RESUMEN

BACKGROUND: The use of next-generation sequencing for fusion identification is being increasingly applied and aids our understanding of tumor biology. Some fusions are responsive to approved targeted agents, while others have future potential for therapeutic targeting. Although some pediatric central nervous system tumors may be cured with surgery alone, many require adjuvant therapy associated with acute and long-term toxicities. Identification of targetable fusions can shift the treatment paradigm toward earlier integration of molecularly targeted agents. METHODS: Patients diagnosed with glial, glioneuronal, and ependymal tumors between 2002 and 2019 were retrospectively reviewed for fusion testing. Testing was done primarily using the ArcherDx FusionPlex Solid Tumor panel, which assesses fusions in 53 genes. In contrast to many previously published series chronicling fusions in pediatric patients, we compared histological features and the tumor classification subtype with the specific fusion identified. RESULTS: We report 24 cases of glial, glioneuronal, or ependymal tumors from pediatric patients with identified fusions. With the exception of BRAF:KIAA1549 and pilocytic/pilomyxoid astrocytoma morphology, and possibly QKI-MYB and angiocentric glioma, there was not a strong correlation between histological features/tumor subtype and the specific fusion. We report the unusual fusions of PPP1CB-ALK, CIC-LEUTX, FGFR2-KIAA159, and MN1-CXXC5 and detail their morphological features. CONCLUSIONS: Fusion testing proved to be informative in a high percentage of cases. A large majority of fusion events in pediatric glial, glioneuronal, and ependymal tumors can be identified by relatively small gene panels.


Asunto(s)
Biomarcadores de Tumor/genética , Neoplasias Encefálicas/patología , Ependimoma/patología , Glioma/patología , Neoplasias Neuroepiteliales/patología , Proteínas de Fusión Oncogénica/genética , Neoplasias Encefálicas/clasificación , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/terapia , Niño , Preescolar , Terapia Combinada , Ependimoma/clasificación , Ependimoma/genética , Ependimoma/terapia , Femenino , Estudios de Seguimiento , Glioma/clasificación , Glioma/genética , Glioma/terapia , Humanos , Lactante , Masculino , Neoplasias Neuroepiteliales/clasificación , Neoplasias Neuroepiteliales/genética , Neoplasias Neuroepiteliales/terapia , Pronóstico , Estudios Retrospectivos
20.
J Neuropathol Exp Neurol ; 79(1): 62-66, 2020 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-31769844

RESUMEN

Transgender individuals may receive long-term hormonal treatment as part of their sexual transition; limited literature has suggested that they consequently may be predisposed to development of prolactinomas. We questioned whether we had encountered such cases. Pathology databases were searched for the years 2000-2019 for tissue specimens from transgender individuals; Sixty surgical specimens from 58 individuals and 8 cytology specimens were identified. Two of these 60 were pituitary adenomas, neither of which were lactotroph adenomas (prolactinomas).The first occurred in a 71-year-old transgender male-to-female who had undergone high-dose hormone therapy, followed by orchiectomy 30 years prior. Chronic hypertension, dizziness, and vertigo prompted an endocrine workup which revealed elevated IGF-1 and prolactin; The pituitary mass proved to be a mixed somatotroph/lactotroph adenoma. The second occurred in a 53-year-old transgender male-to-female who was being evaluated by an endocrinologist prior to initiating hormone therapy for transition when a slightly elevated prolactin level was discovered. This pituitary macroadenoma proved to be a gonadotroph adenoma. The most common surgical specimens were 33 bilateral mastectomies, 13 hysterectomies, and 4 orchiectomies, almost all for gender transition purposes rather than medical conditions. Pathologists may wish to be aware of the occurrence of pituitary adenomas in transgender individuals, although the incidence is quite low.


Asunto(s)
Adenoma/etiología , Hormonas/efectos adversos , Neoplasias Hipofisarias/etiología , Procedimientos de Reasignación de Sexo/efectos adversos , Personas Transgénero , Adenoma/cirugía , Anciano , Bases de Datos Factuales , Femenino , Hormonas/uso terapéutico , Humanos , Hipofisectomía , Masculino , Persona de Mediana Edad , Orquiectomía , Neoplasias Hipofisarias/cirugía , Prolactinoma/etiología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA