Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
1.
Cancers (Basel) ; 16(2)2024 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-38254850

RESUMO

Glioblastoma is the most frequent and malignant primary neoplasm of the central nervous system. In a recent breakthrough study on a prospective Discovery cohort, I proposed the first all-inclusive molecular classification of glioblastoma into seven subgroups, G1-G7, based on MAPK pathway activation. New data from a WHO-grade-4 diffuse glioma prospective Validation cohort offers, in this study, an integrated demographic-molecular analysis of a 213-patient Combined cohort. Despite cohort differences in the median age and molecular subgroup distribution, all the prospectively-acquired cases from the Validation cohort mapped into one of the G1-G7 subgroups defined in the Discovery cohort. A younger age of onset, higher tumor mutation burden and expanded G1/EGFR-mutant and G3/NF1 glioblastoma subgroups characterized the glioblastomas from African American/Black relative to Caucasian/White patients. The three largest molecular subgroups were G1/EGFR, G3/NF1 and G7/Other. The fourth largest subgroup, G6/Multi-RTK, was detailed by describing a novel gene fusion ST7-MET, rare PTPRZ1-MET, LMNA-NTRK1 and GOPC-ROS1 fusions and their overexpression mechanisms in glioblastoma. The correlations between the MAPK pathway G1-G7 subgroups and the PI3-kinase/PTEN, TERT, cell cycle G1 phase and p53 pathways defined characteristic subgroup pathway profiles amenable to personalized targeted therapy. This analysis validated the first all-inclusive molecular classification of glioblastoma, showed significant demographic and molecular differences between subgroups, and provided the first ethnic molecular comparison of glioblastoma.

2.
Cell Commun Signal ; 20(1): 123, 2022 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-35978432

RESUMO

Medulloblastoma is the most common pediatric embryonal brain tumor, and may occur in cancer predisposition syndromes. We describe novel associations of medulloblastoma with atypical prolactinoma and dural high-grade sarcoma in Li-Fraumeni syndrome (LFS), and epidural desmoid fibromatosis in familial adenomatous polyposis (FAP)/Turcot syndrome. Genomic analysis showing XRCC3 alterations suggested radiotherapy as contributing factor to the progression of LFS-associated medulloblastoma, and demonstrated different mechanisms of APC inactivation in the FAP-associated tumors. The integrated genomic-transcriptomic analysis uncovered the growth pathways driving tumorigenesis, including the prolactin-prolactin receptor (PRLR) autocrine loop and Shh pathway in the LFS-associated prolactinoma and medulloblastoma, respectively, the Wnt pathway in both FAP-associated neoplasms, and the TGFß and Hippo pathways in the soft tissue tumors, regardless of germline predisposition. In addition, the comparative analysis of paired syndromic neoplasms revealed several growth pathways susceptible to therapeutic intervention by PARP, PRLR, and selective receptor tyrosine kinase (RTK) inhibitors. These could target the defective DNA damage repair in the LFS-associated medulloblastoma, the prolactin autocrine loop in the atypical prolactinoma, the EPHA3/7 and ALK overexpression in the FAP-associated medulloblastoma, and the multi-RTK upregulation in the soft tissue neoplasms. This study presents the spatiotemporal evolution of novel neoplastic associations in syndromic medulloblastoma, and discusses the post-radiotherapy risk for secondary malignancies in syndromic pediatric patients, with important implications for the biology, diagnosis, and therapy of these tumors. Video Abstract.


Assuntos
Polipose Adenomatosa do Colo , Neoplasias Cerebelares , Meduloblastoma , Neoplasias Hipofisárias , Prolactinoma , Polipose Adenomatosa do Colo/genética , Polipose Adenomatosa do Colo/patologia , Neoplasias Cerebelares/genética , Criança , Humanos , Meduloblastoma/genética , Meduloblastoma/patologia , Prolactina
3.
Cancers (Basel) ; 13(18)2021 Sep 09.
Artigo em Inglês | MEDLINE | ID: mdl-34572759

RESUMO

Glioblastoma is the most aggressive and frequent glioma in the adult population. Because current therapy regimens confer only minimal survival benefit, molecular subgrouping to stratify patient prognosis and therapy design is warranted. This study presents a multi-platform classification of glioblastoma by analyzing a large, ethnicity-inclusive 101-adult-patient cohort. It defines seven non-redundant IDH-wild-type glioblastoma molecular subgroups, G1-G7, corresponding to the upstream receptor tyrosine kinase (RTK) and RAS-RAF segment of the ERK/MAPK signal transduction pathway. These glioblastoma molecular subgroups are classified as G1/EGFR, G2/FGFR3, G3/NF1, G4/RAF, G5/PDGFRA, G6/Multi-RTK, and G7/Other. The comprehensive genomic analysis was refined by expression landscaping of all RTK genes, as well as of the major associated growth pathway mediators, and used to hierarchically cluster the subgroups. Parallel demographic, clinical, and histologic pattern analyses were merged with the molecular subgrouping to yield the first inclusive multi-platform classification for IDH-wild-type glioblastoma. This straightforward classification with diagnostic and prognostic significance may be readily used in neuro-oncological practice and lays the foundation for personalized targeted therapy approaches.

4.
Acta Neuropathol Commun ; 9(1): 69, 2021 04 14.
Artigo em Inglês | MEDLINE | ID: mdl-33853673

RESUMO

Prognostic molecular subgrouping of glioblastoma is an ongoing effort and the current classification includes IDH-wild-type and IDH-mutant entities, the latter showing significantly better prognosis. We performed a comparative integrated analysis of the FGFR glioblastoma subgroup consisting of 5 cases from a prospective 101-patient-cohort. FGFR alterations included FGFR2-TACC2 and FGFR2 amplifications arising in a multifocal IDH-mutant glioblastoma with unexpected 2.5-month patient survival, novel FGFR3 carboxy-terminal duplication and FGFR3-TLN1 fusion, and two previously described FGFR3-TACC3 fusions. The FGFR2 tumors showed additional mutations in SERPINE1/PAI-1 and MMP16, as part of extensive extracellular matrix remodeling programs. Whole transcriptomic analysis revealed common proliferation but distinct morphogenetic gene expression programs that correlated with tumor histology. The kinase program revealed EPHA3, LTK and ALK receptor tyrosine kinase overexpression in individual FGFR tumors. Paradoxically, all FGFR-fused glioblastomas shared strong PI3K and MAPK pathway suppression effected by SPRY, DUSP and AKAP12 inhibitors, whereas the FGFR2-TACC2 tumor elicited also EGFR suppression by ERRFI1 upregulation. This integrated analysis outlined the proliferation and morphogenetic expression programs in FGFR glioblastoma, and identified four novel, clinically targetable FGFR2 and FGFR3 alterations that confer aggressive phenotype and trigger canonical pathway feedback inhibition, with important therapeutic implications.


Assuntos
Neoplasias Encefálicas/genética , Glioblastoma/genética , Receptor Tipo 2 de Fator de Crescimento de Fibroblastos/genética , Receptor Tipo 3 de Fator de Crescimento de Fibroblastos/genética , Idoso , Neoplasias Encefálicas/patologia , Feminino , Amplificação de Genes , Glioblastoma/patologia , Humanos , Masculino , Pessoa de Meia-Idade , Proteínas de Fusão Oncogênica , Fenótipo , Prognóstico , Estudos Prospectivos , Transcriptoma
5.
Sci Rep ; 10(1): 10954, 2020 07 02.
Artigo em Inglês | MEDLINE | ID: mdl-32616776

RESUMO

Children diagnosed with brain tumors have the lowest overall survival of all pediatric cancers. Recent molecular studies have resulted in the discovery of recurrent driver mutations in many pediatric brain tumors. However, despite these molecular advances, the clinical outcomes of high grade tumors, including H3K27M diffuse midline glioma (H3K27M DMG), remain poor. To address the paucity of tissue for biological studies, we have established a comprehensive protocol for the coordination and processing of donated specimens at postmortem. Since 2010, 60 postmortem pediatric brain tumor donations from 26 institutions were coordinated and collected. Patient derived xenograft models and cell cultures were successfully created (76% and 44% of attempts respectively), irrespective of postmortem processing time. Histological analysis of mid-sagittal whole brain sections revealed evidence of treatment response, immune cell infiltration and the migratory path of infiltrating H3K27M DMG cells into other midline structures and cerebral lobes. Sequencing of primary and disseminated tumors confirmed the presence of oncogenic driver mutations and their obligate partners. Our findings highlight the importance of postmortem tissue donations as an invaluable resource to accelerate research, potentially leading to improved outcomes for children with aggressive brain tumors.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Glioma/patologia , Histonas/genética , Mutação , Adolescente , Adulto , Animais , Autopsia , Neoplasias Encefálicas/genética , Criança , Pré-Escolar , Feminino , Perfilação da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Glioma/genética , Humanos , Lactente , Masculino , Camundongos Endogâmicos NOD , Camundongos SCID , Células Tumorais Cultivadas , Ensaios Antitumorais Modelo de Xenoenxerto , Adulto Jovem
6.
World Neurosurg ; 138: 115-119, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32147560

RESUMO

BACKGROUND: Nonlethal neural tube defects are developmental malformations with complex pathogenesis usually manifested at birth or in childhood. CASE DESCRIPTION: We report the case of a 61-year-old woman without significant previous clinical history presenting for neck pain and stiffness. An extensive workup detected multiple lytic lesions within the occipital bone and cervical vertebrae, suspicious for multiple myeloma or metastatic disease. Surgical resection of the occipital bone lesions revealed ectopic cerebellar tissue, some containing folia with mature cortical lamination, and no evidence of malignancy. CONCLUSIONS: To our knowledge, this study describes the oldest individual presenting with ectopic cerebellar tissue and the only instance in which oncologic workup for malignancy was carried out prior to resection. It also proposes surgical resection as a diagnostic and curative approach for this complex basicranium and neural developmental defect, and discusses retinoic acid toxicity as a possible cause of its occurrence.


Assuntos
Cerebelo/patologia , Coristoma/patologia , Osso Occipital/patologia , Neoplasias Cranianas/patologia , Neoplasias Cerebelares/patologia , Neoplasias Cerebelares/cirurgia , Cerebelo/cirurgia , Coristoma/diagnóstico , Coristoma/cirurgia , Diagnóstico Diferencial , Feminino , Humanos , Pessoa de Meia-Idade , Cervicalgia/etiologia , Procedimentos Neurocirúrgicos/métodos , Osso Occipital/cirurgia , Neoplasias Cranianas/diagnóstico , Neoplasias Cranianas/cirurgia
7.
Acta Neuropathol Commun ; 8(1): 10, 2020 02 03.
Artigo em Inglês | MEDLINE | ID: mdl-32014051

RESUMO

Glioblastoma is the most frequent and aggressive primary brain tumor, characterized by extensive brain invasion and rarely, systemic metastases. The pathogenesis of metastatic glioblastoma is largely unknown. We present the first integrated clinical/histologic/genetic analysis of 5 distinct brain and lung foci from a unique case of recurrent, multifocal, multicentric and metastatic glioblastoma. The initial right frontotemporal gliosarcoma received standard surgical/chemoradiation therapy and recurred 1.5 years later, co-occurring with three additional masses localized to the ipsilateral temporal lobe, cerebellum and lung. Synchronous metastatic lung carcinoma was suspected in this long-term smoker patient with family history of cancer. However, glioblastoma was confirmed in all tumors, although with different morphologic patterns, including ependymomatous and epithelioid. Genomic profiling revealed a germline FANCD2 variant of unknown significance, and a 4-gene somatic mutation signature shared by all tumors, consisting of TERT promoter and PTEN, RB1 and TP53 tumor suppressor mutations. Additional GRIN2A and ATM heterozygous mutations were selected in the cerebellar and lung foci, but were variably present in the supratentorial foci, indicating reduced post-therapeutic genetic evolution in brain foci despite morphologic variability. Significant genetic drift characterized the lung metastasis, likely explaining the known resistance of circulating glioblastoma cells to systemic seeding. MET overexpression was detected in the initial gliosarcoma and lung metastasis, possibly contributing to invasiveness. This comprehensive analysis sheds light on the temporospatial evolution of glioblastoma and underscores the importance of genetic testing for diagnosis and personalized therapy.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Glioblastoma/genética , Glioblastoma/patologia , Neoplasias Pulmonares/genética , Neoplasias Pulmonares/patologia , Recidiva Local de Neoplasia/genética , Recidiva Local de Neoplasia/patologia , Encéfalo/patologia , Feminino , Humanos , Pulmão/patologia , Neoplasias Pulmonares/secundário , Pessoa de Meia-Idade , Mutação
8.
Cancers (Basel) ; 12(1)2020 Jan 17.
Artigo em Inglês | MEDLINE | ID: mdl-31963394

RESUMO

Chordoid meningioma is a rare WHO grade II histologic variant. Its molecular alterations or their impact on patient risk stratification have not been fully explored. We performed a multicenter, clinical, histological, and genomic analysis of chordoid meningiomas from 30 patients (34 tumors), representing the largest integrated study to date. By NHERF1 microlumen immunohistochemical detection, three epithelial differentiation (ED) groups emerged: #1/fibroblastic-like, #2/epithelial-poorly-differentiated and #3/epithelial-well-differentiated. These ED groups correlated with tumor location and genetic profiling, with NF2 and chromatin remodeling gene mutations clustering in ED group #2, and TRAF7 mutations segregating in ED group #3. Mutations in LRP1B were found in the largest number of cases (36%) across ED groups #2 and #3. Pathogenic ATM and VHL germline mutations occurred in ED group #3 patients, conferring an aggressive or benign course, respectively. The recurrence rate significantly correlated with mutations in NF2, as single gene, and with mutations in chromatin remodeling and DNA damage response genes, as groups. The recurrence rate was very high in ED group #2, moderate in ED group #3, and absent in ED group #1. This study proposes guidelines for tumor recurrence risk stratification and practical considerations for patient management.

9.
Oncotarget ; 10(40): 4038-4052, 2019 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-31258848

RESUMO

Epithelioid glioblastoma is a recognized glioblastoma variant, recently added to the World Health Organization brain tumor classification, with similar prognosis as the classic variant and B-Raf V600E mutations in 50% of the cases. We identified a new subset of epithelioid glioblastoma with periventricular location and subependymal giant cell astrocytoma (SEGA)-like morphology. Genomic profiling of these tumors revealed driver mutations in NF1, subclonal mutations in TSC1, and a novel driver mutation in MTOR, suggesting upregulation of the MAPK/TSC1/mTOR pathway. Strong mTOR activation was confirmed by immunohistochemistry for the mTOR kinase target 4E-BP1. TSC1 and MTOR mutations have been previously described in low-grade glioma, such as SEGA, and focal cortical dysplasia, respectively, that display large cells with abundant cytoplasm, most likely resulting from the biogenetic signaling of mTOR. Unlike these, the mutations in SEGA-like glioblastoma occurred in the context of other genetic aberrations present in high-grade neoplasms, including in the CDKN2A/B, PIK3R1, PIK3CA and EGFR genes. For one patient with two temporally distinct specimens, the subclonal TSC1 pathogenic mutation was detected only in the specimen showing SEGA-like morphology, indicating requirement for mTOR activation as trigger for specific epithelioid/SEGA-like morphology. As FDA-approved kinase inhibitors are available and target many steps of the MAPK/mTOR pathway, recognition of this new subset of periventricular high-grade gliomas with clear phenotypic-genotypic correlates is essential for prompt biomarker testing and appropriate targeted therapeutic management of these patients.

10.
Acta Neuropathol Commun ; 7(1): 42, 2019 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-30876455

RESUMO

Astroblastoma (AB) is a rare CNS tumor demonstrating abundant astroblastomatous pseudorosettes. Its molecular features have not been comprehensively studied and its status as a tumor entity is controversial. We analyzed a cohort of 27 histologically-defined ABs using DNA methylation profiling, copy number analysis, FISH and site-directed sequencing. Most cases demonstrated mutually exclusive MN1 rearrangements (n = 10) or BRAFV600E mutations (n = 7). Two additional cases harbored RELA rearrangements. Other cases lacked these specific genetic alterations (n = 8). By DNA methylation profiling, tumors with MN1 or RELA rearrangement clustered with high-grade neuroepithelial tumor with MN1 alteration (HGNET-MN1) and RELA-fusion ependymoma, respectively. In contrast, BRAFV600E-mutant tumors grouped with pleomorphic xanthoastrocytoma (PXA). Six additional tumors clustered with either supratentorial pilocytic astrocytoma and ganglioglioma (LGG-PA/GG-ST), normal or reactive cerebrum, or with no defined DNA methylation class. While certain histologic features favored one genetic group over another, no group could be reliably distinguished by histopathology alone. Survival analysis between genetic AB subtypes was limited by sample size, but showed that MN1-rearranged AB tumors were characterized by better overall survival compared to other genetic subtypes, in fact, significantly better than BRAFV600E-mutant tumors (P = 0.013). Our data confirm that histologically-defined ABs are molecularly heterogeneous and do not represent a single entity. They rather encompass several low- to higher-grade glial tumors including neuroepithelial tumors with MN1 rearrangement, PXA-like tumors, RELA ependymomas, and possibly yet uncharacterized lesions. Genetic subtyping of tumors exhibiting AB histology, particularly determination of MN1 and BRAFV600E status, is necessary for important prognostic and possible treatment implications.


Assuntos
Neoplasias Encefálicas/genética , Rearranjo Gênico/genética , Genômica/métodos , Neoplasias Neuroepiteliomatosas/genética , Proteínas Proto-Oncogênicas B-raf/genética , Transativadores/genética , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/mortalidade , Neoplasias Encefálicas/patologia , Criança , Pré-Escolar , Estudos de Coortes , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Neoplasias Neuroepiteliomatosas/mortalidade , Neoplasias Neuroepiteliomatosas/patologia , Prognóstico , Taxa de Sobrevida/tendências , Adulto Jovem
11.
Neuro Oncol ; 21(6): 800-808, 2019 06 10.
Artigo em Inglês | MEDLINE | ID: mdl-30726976

RESUMO

BACKGROUND: Dysembryoplastic neuroepithelial tumors (DNETs) are uncommon neural tumors presenting most often in children and young adults and associated with intractable seizures. Rare midline neoplasms with similar histological features to those found in DNETs have been described near the septum pellucidum and termed "DNET-like neoplasms of the septum pellucidum." Due to their rarity, these tumors have been described in just a few reports and their genetic alterations sought only in small series. METHODS: We collected 20 of these tumors for a comprehensive study of their clinical, radiological, and pathological features. RNA sequencing or targeted DNA sequencing was undertaken on 18 tumors, and genome-wide DNA methylation profiling was possible with 11 tumors. Published cases (n = 22) were also reviewed for comparative purposes. RESULTS: The commonest presenting symptoms and signs were related to raised intracranial pressure; 40% of cases required cerebrospinal fluid diversion. Epilepsy was seen in approximately one third of cases. All patients had an indolent disease course, despite metastasis within the neuraxis in a few cases. Radiologically, the septum verum/septal nuclei were involved in all cases and are the proposed site of origin for septal DNET (sDNET). Septal DNET showed a high frequency (~80%) of mutations of platelet derived growth factor receptor A (PDGFRA), and alterations in fibroblast growth factor receptor 1 (FGFR1) and neurofibromatosis type 1 (NF1) were also identified. In a genomic DNA methylation analysis alongside other neural tumors, sDNETs formed a separate molecular group. CONCLUSIONS: Genetic alterations that are different from those of cerebral DNETs and a distinct methylome profile support the proposal that sDNET is a distinct disease entity.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Regulação Neoplásica da Expressão Gênica , Imageamento por Ressonância Magnética/métodos , Mutação , Neoplasias Neuroepiteliomatosas/patologia , Biomarcadores Tumorais/metabolismo , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Criança , Metilação de DNA , Feminino , Humanos , Masculino , Neoplasias Neuroepiteliomatosas/genética , Neoplasias Neuroepiteliomatosas/metabolismo , Prognóstico , Receptor Tipo 1 de Fator de Crescimento de Fibroblastos/genética , Receptor alfa de Fator de Crescimento Derivado de Plaquetas/genética , Taxa de Sobrevida
12.
Oper Neurosurg (Hagerstown) ; 17(2): E58, 2019 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-30715450

RESUMO

Peritrigonal lesions are deeply seated and are surrounded by critical neurovascular structures. Traditional transcortical approaches carry the risk of damage to important surrounding white matter tracts. In this regard, a posterior interhemispheric approach gives a more direct and less invasive route and therefore is a reasonable alternative to transcortical approaches. The 3-dimensional video includes illustrations and animations showing the anatomy of the white matter tracts around the trigone and explains the physiological basis of posterior interhemispheric precuneal approach to this complex region. This also includes a 3-dimensional operative video of the same approach in a 50-yr-old male patient with left periatrial lesion describing surgical techniques and nuances. An informed written consent has been obtained from the patient.

13.
Oper Neurosurg (Hagerstown) ; 16(1): E10-E11, 2019 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-29788437

RESUMO

Rosai-Dorfman disease, originally described by Juan Rosai and Ronald F. Dorfman, is a rare benign histiocytic proliferative disorder, classically presenting with massive lymphadenopathy and a self-limiting clinical course.1 Isolated intracranial skull base involvement is extremely rare and often resembles meningiomas, schwannomas, or other benign skull base lesions.2 The disease is difficult to diagnose radiographically, and tissue diagnosis with open skull base approaches has significant perioperative risks.2,3 We present the case of a 48-yr-old Caucasian male presenting with progressively worsening headaches, giddiness, hearing difficulty, and diplopia. Magnetic resonance imaging of the brain revealed T1-weighted isointense, T2-weighted hypointense, and contrast-enhancing dural-based lesion in the left cerebellopontine angle. The patient underwent maximally safe resection of the lesion through the retromastoid approach with careful preservation of the lower cranial nerve complex. The intraoperative findings of a variegated and lobulated mass adherent to the skull base, the surgical strategy of safe resection, and eventual good outcome in this patient are depicted in this 3-dimensional video presentation. The majority of patients with skull base Rosai-Dorfman disease reported in literature have had stable or regression of disease (78%) after initial conservative surgical treatment and hence aggressive surgical resection is of unproven efficacy.3 The patient has consented to depiction of his surgical video and intraoperative images in this video manuscript.

14.
Oncotarget ; 9(47): 28652-28665, 2018 Jun 19.
Artigo em Inglês | MEDLINE | ID: mdl-29983887

RESUMO

Meningioma is a primary brain tumor arising from the neoplastic transformation of meningothelial cells. Several histological variants of meningioma have been described. Here we show that NHERF1/EBP50, an adaptor protein required for structuring specialized polarized epithelia, can distinguish meningioma variants with epithelial differentiation. NHERF1 decorates the membrane of intracytoplasmic lumens and microlumens in the secretory variant, consistent with a previously described epithelial differentiation of this subtype. NHERF1 also labels microlumens in chordoid meningioma, an epithelial variant not previously known to harbor these structures, and ultrastructural analysis confirmed the presence of microlumens in this variant. NHERF1 associates with the ezrin-radixin-moesin (ERM)-NF2 cytoskeletal proteins, and moesin but not NF2 was detectable in the microlumens. In a meningioma series from 83 patients, NHERF1 revealed microlumens in 87.5% of the chordoid meningioma (n = 25) and meningioma with chordoid component (n = 7) cases, and in 100% of the secretory meningioma cases (n = 12). The most common WHO grade I meningioma variants lacked microlumens. Interestingly, 20% and 66.6% of WHO grades II (n = 20) and III (n = 3) meningiomas, respectively, showed microlumen-like NHERF1 staining of ultrastructural tight microvillar interdigitations, mainly in rhabdoid, papillary-like or sheeting areas, revealing a new subset of high grade meningiomas with epithelial differentiation. NHERF1 failed to detect microlumens in 12 additional cases of chordoid glioma of the 3rd ventricle, chordoma and chondrosarcoma, neoplasms that may mimic the histological appearance of chordoid meningioma. This study uncovers features of epithelial differentiation in meningioma and proposes NHERF1 immunohistochemistry as a method of discriminating chordoid meningioma from neoplasms with similar appearance.

15.
World Neurosurg ; 116: 163-168, 2018 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-29689399

RESUMO

BACKGROUND: Primary focal intracranial leptomeningeal glioma (PFILG) is considered a rare solitary glial tumor arising from the leptomeninges without brain attachment or evidence of primary neoplasm elsewhere within the neuraxis. We report a case of PFILG in a left parietal location and provide a concise review on its clinicoradiologic, histopathologic, and management characteristics. CASE PRESENTATION: A 75-year-old woman presented with focal motor seizures involving the right upper limb with associated occasional headache. Magnetic resonance imaging (MRI) of the brain revealed a small focal lesion attached to the left parietal dura with underlying brain parenchymal signal changes, and contrast-enhanced images showed heterogeneous enhancement of the tumor and meninges. The repeated MRI brain images within a month of primary imaging revealed significant progression of the size of lesion along with invasion of underlying parietal lobe parenchyma. The patient underwent gross total resection of the lesion, and the histopathologic diagnosis was glioblastoma multiforme (GBM), World Health Organization grade 4, isocitrate dehydrogenase wild type. The patient recovered well from surgery without deficits; however, she refused adjuvant treatment. MRI of the brain repeated 3 months after surgery revealed significant progression of the GBM with mass effect. Although adjuvant treatment was then started, she could not tolerate it and died 4 months after surgery. CONCLUSION: The possibility of high-grade PFILG should be considered in any aggressive extra-axial focal lesion. The definitive diagnosis can be made after histologic examination. Although surgery followed by adjuvant treatment is considered the mainstay of treatment, the overall prognostic outcome of high-grade PFILG is dismal.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Glioblastoma/diagnóstico por imagem , Neoplasias Meníngeas/diagnóstico por imagem , Idoso , Neoplasias Encefálicas/cirurgia , Feminino , Glioblastoma/cirurgia , Humanos , Neoplasias Meníngeas/cirurgia
16.
Am J Case Rep ; 19: 99-104, 2018 Jan 26.
Artigo em Inglês | MEDLINE | ID: mdl-29371584

RESUMO

BACKGROUND Invasive mucinous adenocarcinoma (IMA) is a rare variant of adenocarcinoma of the lung. It frequently shows KRAS mutations, while ALK rearrangement is exceedingly rare. We present a case of ALK-rearranged IMA of the lung presenting with an unusual pattern of brain metastases, radiologically mimicking a cavernous angioma. CASE REPORT A 44-year-old non-smoker female was first diagnosed with lung right lower lobe IMA with ALK rearrangement. Five years after surgery followed by chemotherapy, she developed a sudden onset headache. Brain imaging revealed a hemorrhagic left frontal mass, suspicious for a cavernous angioma. However, the pathology of the resected lesion showed an ALK-rearranged brain metastasis from the IMA of the lung. Interestingly, the metastases showed perivascular tumor infiltrates, accompanied by focal mural invasion, vascular disruption, and hemorrhage. CONCLUSIONS To our knowledge, this is the first reported case of brain metastasis from an ALK-rearranged IMA of the lung. Further investigation of the clinical and pathological characteristics of the ALK-rearranged IMA, including awareness of the possibility for development of brain metastases with tumor-associated vasculopathy and hemorrhage, is warranted.


Assuntos
Adenocarcinoma Mucinoso/diagnóstico , Neoplasias Encefálicas/diagnóstico , Rearranjo Gênico , Hemangioma Cavernoso do Sistema Nervoso Central/diagnóstico , Neoplasias Pulmonares/patologia , Receptores Proteína Tirosina Quinases/genética , Adenocarcinoma Mucinoso/genética , Adenocarcinoma Mucinoso/secundário , Adulto , Quinase do Linfoma Anaplásico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/secundário , Diagnóstico Diferencial , Feminino , Humanos
17.
World Neurosurg ; 112: e74-e83, 2018 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-29258946

RESUMO

BACKGROUND: To analyze the differences in tumor progression patterns and histopathologic characteristics between skull base meningiomas (SBMs) and non-skull base meningiomas (NSBMs). METHODS: Retrospective data of 382 patients with SBMs and 200 patients with NSBMs who underwent primary microsurgical resection between January 1995 and December 2016 were analyzed. Data related to clinical history, neuroimaging, surgical technique, and follow-up were reviewed. A separate prospective cohort of 78 meningiomas operated on from June 2016 to July 2017 was analyzed (World Health Organization [WHO] grade and Ki-67 proliferation index) for biologic comparison. RESULTS: Skull base location, WHO grade II tumor, and subtotal resection were independent predictors of unfavorable outcome. The overall tumor progression rate in the SBM group (33%) was higher than in the NSBM group (19.7%) (P = 0.006) (mean follow-up period, 51 months). The 3-year, 5-year, and 10-year progression-free survival (PFS) was 78%, 60%, and 45% in the SBM group, whereas it was 90%, 80%, and 53% in the NSBM group, respectively. Interestingly, there was no difference in the median PFS after 10 years of follow-up (SBMs 210 months vs. NSBM 212 months, P = 0.93). In the prospective cohort of 78 meningiomas, there was no statistically significant difference in the proportion of WHO grade I tumors and in the mean Ki-67 index of WHO grade I meningiomas between the 2 groups. CONCLUSION: There is a divergent tumor progression pattern for surgically treated SBMs vis-à-vis NSBMs. Differences in tumor biology and the proportion of Simpson grade I resections are likely attributable factors.


Assuntos
Neoplasias Meníngeas/patologia , Meningioma/patologia , Neoplasias da Base do Crânio/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Estudos de Coortes , Progressão da Doença , Intervalo Livre de Doença , Feminino , Seguimentos , Humanos , Estimativa de Kaplan-Meier , Masculino , Neoplasias Meníngeas/mortalidade , Meningioma/mortalidade , Pessoa de Meia-Idade , Estudos Retrospectivos , Neoplasias da Base do Crânio/mortalidade , Adulto Jovem
18.
Clin Neuropathol ; 36 (2017)(3): 114-120, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28128725

RESUMO

AIM: We describe a series of three diagnostically challenging, histologically similar fibro-osseous skull masses. METHODS: The cases were identified in our archives among 50,000 neuropathology specimens. A comprehensive review of the histological, immunohistochemical, ultrastructural, and imaging features as well as the clinical outcome was performed. RESULTS: The routine histology was similar in all 3 cases and showed spindle cell proliferations with frequent calcospheres or psammomatoid bodies. There was no evidence of an underlying subdural component. Immunohistochemistry for the meningioma markers EMA and SSTR2A raised the possibility of intraosseous meningioma, as all 3 lesions were convincingly positive for epithelial membrane antigen (EMA) and 1 lesion was convincingly positive for the somatostatin receptor subtype 2A (SSTR2A); weak, questionable positivity for SSTR2 was present in the remaining 2 cases. In addition, electron microscopy was available in 1 case and showed features consistent with meningioma. CONCLUSIONS: Overall, the findings were most consistent with intraosseous meningioma. Primary intraosseous meningiomas are rare lesions that may present a diagnostic challenge. It is important to consider meningiomas in the differential diagnosis, as extradural meningiomas are associated with an increased risk of recurrence and may occasionally undergo malignant transformation.
.


Assuntos
Fibroma Ossificante/diagnóstico , Fibroma Ossificante/patologia , Neoplasias Cranianas/diagnóstico , Neoplasias Cranianas/patologia , Crânio/patologia , Adulto , Proliferação de Células , Diagnóstico Diferencial , Fibroma Ossificante/genética , Humanos , Masculino , Meningioma/diagnóstico , Meningioma/genética , Meningioma/patologia , Microscopia Eletrônica , Pessoa de Meia-Idade , Mucina-1/genética , Receptores de Somatostatina/genética , Neoplasias Cranianas/genética
19.
Neuro Oncol ; 19(1): 31-42, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27416954

RESUMO

BACKGROUND: Astroblastomas (ABs) are rare glial tumors showing overlapping features with astrocytomas, ependymomas, and sometimes other glial neoplasms, and may be challenging to diagnose. METHODS: We examined clinical, histopathological, and molecular features in 28 archival formalin-fixed, paraffin-embedded AB cases and performed survival analyses using Cox proportional hazards and Kaplan-Meier methods. RESULTS: Unlike ependymomas and angiocentric gliomas, ABs demonstrate abundant distinctive astroblastic pseudorosettes and are usually Olig2 immunopositive. They also frequently exhibit rhabdoid cells, multinucleated cells, and eosinophilic granular material. They retain immunoreactivity to alpha thalassemia/mental retardation syndrome X-linked, are immunonegative to isocitrate dehydrogenase-1 R132H mutation, and only occasionally show MGMT promoter hypermethylation differentiating them from many diffuse gliomas. Like pleomorphic xanthoastrocytoma, ganglioglioma, supratentorial pilocytic astrocytoma, and other predominantly cortical-based glial tumors, ABs often harbor the BRAFV600E mutation, present in 38% of cases tested (n = 21), further distinguishing those tumors from ependymomas and angiocentric gliomas. Factors correlating with longer patient survival included age less than 30 years, female gender, absent BRAFV600E , and mitotic index less than 5 mitoses/10 high-power fields; however, only the latter was significant by Cox and Kaplan-Meier analyses (n = 24; P = .024 and .012, respectively). This mitotic cutoff is therefore currently the best criterion to stratify tumors into low-grade ABs and higher-grade anaplastic ABs. CONCLUSIONS: In addition to their own characteristic histological features, ABs share some molecular and histological findings with other, possibly ontologically related, cortical-based gliomas of mostly children and young adults. Importantly, the presence of BRAFV600E mutations in a subset of ABs suggests potential clinical utility of targeted anti-BRAF therapy.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/patologia , Córtex Cerebral/patologia , Mutação/genética , Neoplasias Neuroepiteliomatosas/patologia , Proteínas Proto-Oncogênicas B-raf/genética , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/genética , Córtex Cerebral/metabolismo , Criança , Pré-Escolar , Feminino , Seguimentos , Humanos , Masculino , Pessoa de Meia-Idade , Gradação de Tumores , Neoplasias Neuroepiteliomatosas/genética , Prognóstico , Taxa de Sobrevida , Adulto Jovem
20.
Neoplasia ; 18(8): 512-23, 2016 08.
Artigo em Inglês | MEDLINE | ID: mdl-27566107

RESUMO

NHERF1/EBP50, an adaptor molecule that interacts with ß-catenin, YAP, and PTEN, has been recently implicated in the progression of various human malignancies, including colorectal cancer. We report here that NHERF1 acts as a tumor suppressor in vivo for intestinal adenoma development. NHERF1 is highly expressed at the apical membrane of mucosa intestinal epithelial cells (IECs) and serosa mesothelial cells. NHERF1-deficient mice show overall longer small intestine and colon that most likely could be attributed to a combination of defects, including altered apical brush border of absorbtive IECs and increased number of secretory IECs. NHERF1 deficiency in Apc(Min/+) mice resulted in significantly shorter animal survival due to markedly increased tumor burden. This resulted from a moderate increase of the overall tumor density, more pronounced in females than males, and a massive increase in the number of large adenomas in both genders. The analysis of possible pathways controlling tumor size showed upregulation of Wnt-ß-catenin pathway, higher expression of unphosphorylated YAP, and prominent nuclear expression of cyclin D1 in NHERF1-deficient tumors. Similar YAP changes, with relative decrease of phosphorylated YAP and increase of nuclear YAP expression, were observed as early as the adenoma stages in the progression of human colorectal cancer. This study discusses a complex role of NHERF1 for intestinal morphology and presents indisputable evidence for its in vivo tumor suppressor function upstream of Wnt-ß-catenin and Hippo-YAP pathways.


Assuntos
Transformação Celular Neoplásica/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patologia , Fosfoproteínas/metabolismo , Trocadores de Sódio-Hidrogênio/metabolismo , Via de Sinalização Wnt , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Adenoma/genética , Adenoma/metabolismo , Adenoma/patologia , Animais , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/patologia , Proteínas de Ciclo Celular , Transformação Celular Neoplásica/genética , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Neoplasias Colorretais/patologia , Ciclina D1/metabolismo , Genes APC , Humanos , Camundongos , Camundongos Knockout , Mutação , Fosfoproteínas/genética , Fosforilação , Trocadores de Sódio-Hidrogênio/genética , Carga Tumoral , Proteínas Wnt/metabolismo , Proteínas de Sinalização YAP , beta Catenina/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA