Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 149
Filtrar
Mais filtros

Intervalo de ano de publicação
1.
Cell ; 175(6): 1665-1678.e18, 2018 11 29.
Artigo em Inglês | MEDLINE | ID: mdl-30343896

RESUMO

Low-grade gliomas almost invariably progress into secondary glioblastoma (sGBM) with limited therapeutic option and poorly understood mechanism. By studying the mutational landscape of 188 sGBMs, we find significant enrichment of TP53 mutations, somatic hypermutation, MET-exon-14-skipping (METex14), PTPRZ1-MET (ZM) fusions, and MET amplification. Strikingly, METex14 frequently co-occurs with ZM fusion and is present in ∼14% of cases with significantly worse prognosis. Subsequent studies show that METex14 promotes glioma progression by prolonging MET activity. Furthermore, we describe a MET kinase inhibitor, PLB-1001, that demonstrates remarkable potency in selectively inhibiting MET-altered tumor cells in preclinical models. Importantly, this compound also shows blood-brain barrier permeability and is subsequently applied in a phase I clinical trial that enrolls MET-altered chemo-resistant glioma patients. Encouragingly, PLB-1001 achieves partial response in at least two advanced sGBM patients with rarely significant side effects, underscoring the clinical potential for precisely treating gliomas using this therapy.


Assuntos
Neoplasias Encefálicas , Éxons , Glioblastoma , Mutação , Inibidores de Proteínas Quinases , Proteínas Proto-Oncogênicas c-met , Animais , Barreira Hematoencefálica/metabolismo , Barreira Hematoencefálica/patologia , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Sistemas de Liberação de Medicamentos , Feminino , Glioblastoma/tratamento farmacológico , Glioblastoma/genética , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Inibidores de Proteínas Quinases/farmacocinética , Inibidores de Proteínas Quinases/farmacologia , Proteínas Proto-Oncogênicas c-met/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-met/genética , Proteínas Proto-Oncogênicas c-met/metabolismo , Ratos Sprague-Dawley , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo , Ensaios Antitumorais Modelo de Xenoenxerto
2.
Int J Mol Sci ; 25(18)2024 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-39337713

RESUMO

Glial-cell-line-derived neurotrophic factor (GDNF) is highly expressed and is involved in the malignant phenotype in glioblastomas (GBMs). However, uncovering its underlying mechanism for promoting GBM progression is still a challenging work. In this study, we found that serine protease inhibitor family E member 1 (SERPINE1) was a potential downstream gene of GDNF. Further experiments confirmed that SERPINE1 was highly expressed in GBM tissues and cells, and its levels of expression and secretion were enhanced by exogenous GDNF. SERPINE1 knockdown inhibited the migration and invasion of GBM cells promoted by GDNF. Mechanistically, GDNF increased SERPINE1 by promoting the phosphorylation of SMAD2/3. In vivo experiments demonstrated that GDNF facilitated GBM growth and the expressions of proteins related to migration and invasion via SERPINE1. Collectively, our findings revealed that GDNF upregulated SERPINE1 via the SMAD2/3-signaling pathway, thereby accelerating GBM cell migration and invasion. The present work presents a new mechanism of GDNF, supporting GBM development.


Assuntos
Movimento Celular , Fator Neurotrófico Derivado de Linhagem de Célula Glial , Glioblastoma , Invasividade Neoplásica , Inibidor 1 de Ativador de Plasminogênio , Transdução de Sinais , Animais , Humanos , Masculino , Camundongos , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/genética , Linhagem Celular Tumoral , Movimento Celular/genética , Regulação Neoplásica da Expressão Gênica , Fator Neurotrófico Derivado de Linhagem de Célula Glial/metabolismo , Fator Neurotrófico Derivado de Linhagem de Célula Glial/genética , Glioblastoma/metabolismo , Glioblastoma/patologia , Glioblastoma/genética , Camundongos Nus , Inibidor 1 de Ativador de Plasminogênio/metabolismo , Inibidor 1 de Ativador de Plasminogênio/genética , Proteína Smad2/metabolismo , Proteína Smad2/genética , Proteína Smad3/metabolismo , Proteína Smad3/genética
3.
Lab Invest ; 103(5): 100053, 2023 05.
Artigo em Inglês | MEDLINE | ID: mdl-36801645

RESUMO

Gliomas are the most common and lethal primary brain tumors in adults. Glioblastomas, the most frequent and aggressive form of gliomas, represent a therapeutic challenge as no curative treatment exists to date, and the prognosis remains extremely poor. Recently, the transcriptional cofactors Yes-associated protein (YAP) and transcriptional co-activator with PDZ-binding motif (TAZ) belonging to the Hippo pathway have emerged as a major determinant of malignancy in solid tumors, including gliomas. However, the mechanisms involved in its regulation, particularly in brain tumors, remain ill-defined. In glioblastomas, EGFR represents one of the most altered oncogenes affected by chromosomal rearrangements, mutations, amplifications, and overexpression. In this study, we investigated the potential link between epidermal growth factor receptor (EGFR) and the transcriptional cofactors YAP and TAZ by in situ and in vitro approaches. We first studied their activation on tissue microarray, including 137 patients from different glioma molecular subtypes. We observed that YAP and TAZ nuclear location was highly associated with isocitrate dehydrogenase 1/2 (IDH1/2) wild-type glioblastomas and poor patient outcomes. Interestingly, we found an association between EGFR activation and YAP nuclear location in glioblastoma clinical samples, suggesting a link between these 2 markers contrary to its ortholog TAZ. We tested this hypothesis in patient-derived glioblastoma cultures by pharmacologic inhibition of EGFR using gefinitib. We showed an increase of S397-YAP phosphorylation associated with decreased AKT phosphorylation after EGFR inhibition in phosphatase and tensin homolog (PTEN) wild-type cultures, unlike PTEN-mutated cell lines. Finally, we used bpV(HOpic), a potent PTEN inhibitor, to mimic the effect of PTEN mutations. We found that the inhibition of PTEN was sufficient to revert back the effect induced by Gefitinib in PTEN-wild-type cultures. Altogether, to our knowledge, these results show for the first time the regulation of pS397-YAP by the EGFR-AKT axis in a PTEN-dependent manner.


Assuntos
Glioblastoma , Adulto , Humanos , Glioblastoma/genética , Proteínas de Sinalização YAP , Proteínas Proto-Oncogênicas c-akt/metabolismo , Tensinas/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Receptores ErbB/metabolismo
4.
Curr Issues Mol Biol ; 45(7): 5276-5292, 2023 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-37504251

RESUMO

Genomic alterations of CDKN2A and CDKN2B in astrocytomas have been an evolving area of study for decades. Most recently, there has been considerable interest in the effect of CDKN2A and/or CDKN2B (CDKN2A/B) homozygous deletions (HD) on the prognosis of isocitrate dehydrogenase (IDH)-mutant astrocytomas. This is highlighted by the adoption of CDKN2A/B HD as an essential criterion for astrocytoma and IDH-mutant central nervous system (CNS) WHO grade 4 in the fifth edition of the World Health Organisation (WHO) Classification of Central Nervous System Tumours (2021). The CDKN2A and CDKN2B genes are located on the short arm of chromosome 9. CDKN2A encodes for two proteins, p14 and p16, and CDKN2B encodes for p15. These proteins regulate cell growth and angiogenesis. Interpreting the impact of CDKN2A/B alterations on astrocytoma prognosis is complicated by recent changes in tumour classification and a lack of uniform standards for testing CDKN2A/B. While the prognostic impact of CDKN2A/B HD is established, the role of different CDKN2A/B alterations-heterozygous deletions (HeD), point mutations, and promoter methylation-is less clear. Consequently, how these alternations should be incorporated into patient management remains controversial. To this end, we reviewed the literature on different CDKN2A/B alterations in IDH-mutant astrocytomas and their impact on diagnosis and management. We also provided a historical review of the changing impact of CDKN2A/B alterations as glioma classification has evolved over time. Through this historical context, we demonstrate that CDKN2A/B HD is an important negative prognostic marker in IDH-mutant astrocytomas; however, the historical data is challenging to interpret given changes in tumour classification over time, variation in the quality of evidence, and variations in the techniques used to identify CDKN2A/B deletions. Therefore, future prospective studies using uniform classification and detection techniques are required to improve the clinical interpretation of this molecular marker.

5.
Environ Toxicol ; 38(7): 1690-1701, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-36988300

RESUMO

Glioblastoma (GBM) is classified as a stage-IV glioma. Unfortunately, there are currently no curative treatments for GBM. Poly(rC)-binding protein 1 (PCBP1) is a cytosolic iron chaperone with diverse functions. PCBP1 is also known to regulate autophagy, but the role of PCBP1 in ferroptosis, iron-dependent cell death pathway, remains unrevealed in GBM cells. Here, we investigated the effects of borax, a boron compound, on the ferroptosis signaling pathway mediated by PCBP1 and autophagy. The study analyzed cell viability, proliferation, and cell cycle on U87-MG and HMC3 cells to investigate the effects of borax. After determining the cytotoxic concentrations of borax, morphological analyzes and measurement of PCBP1, Beclin1, malondialdehyde (MDA), glutathione (GSH), glutathione peroxidase 4 (GPx4) and acyl-CoA synthetase long chain family member 4 (ACSL4) levels were performed. Finally, expression levels of PCBP1, Beclin1, GPx4 and ACSL4, and caspase-3/7 activity were determined. We found that borax reduced U87-MG cell viability in a concentration- and time-dependent manner. Additionally, borax altered cell proliferation and remarkably reduced S phase in the U87-MG cells and exhibited selectivity by having an opposite effect on normal cells (HMC3). According to DAPI staining, borax caused nuclear deficits in U87-MG cells. The result showed that borax in U87-MG cells induced reduction of the PCBP1, GSH, and GPx4 and enhancement of Beclin1, MDA, and ACSL4. Furthermore, borax triggered apoptosis by activating caspase 3/7 in U87-MG cells. Our study indicated that the borax has potential as an anticancer treatment for GBM via regulating PCBP1/Beclin1/GPx4/ACSL4 signaling pathways.


Assuntos
Ferroptose , Glioblastoma , Humanos , Glioblastoma/metabolismo , Ferro , Proteína Beclina-1 , Autofagia
6.
Neuropathol Appl Neurobiol ; 48(4): e12802, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35191072

RESUMO

OBJECTIVE: We aimed to characterise glioblastomas of adolescents and young adults (AYAs) that were isocitrate dehydrogenase (IDH) wild type (wt) and H3wt. MATERIALS AND METHODS: Fifty such patients (aged 16-32) were studied by methylation profiling, targeted sequencing and targeted RNA-seq. RESULTS: Tumours predominantly clustered into three methylation classes according to the terminology of Capper et al. (2018): (anaplastic) pleomorphic xanthoastrocytoma (PXA) (21 cases), GBM_midline (15 cases) and glioblastoma RTK/mesenchymal (seven cases). Two cases clustered with ANA_PA, four cases with LGG classes and one with GBM_MYCN. Only fifteen cases reached a calibrated score >0.84 when the cases were uploaded to DKFZ Classifier. GBM_midline-clustered tumours had a poorer overall survival (OS) compared with the PXA-clustered tumours (p = 0.030). LGG-clustered cases had a significantly better survival than GBM_midline-clustered tumours and glioblastoma RTK/mesenchymal-clustered tumours. Only 13/21 (62%) of PXA-clustered cases were BRAF V600E mutated. Most GBM_midline-clustered cases were not located in the midline. GBM_midline-clustered cases were characterised by PDGFRA amplification/mutation (73.3%), mutations of mismatch repair genes (40.0%), and all showed H3K27me3 and EZH1P loss, and an unmethylated MGMT promoter. Across the whole cohort, MGMT promoter methylation and wt TERT promoter were favourable prognosticators. Mismatch repair gene mutations were poor prognosticators and together with methylation class and MGMT methylation, maintained their significance in multivariate analyses. BRAF mutation was a good prognosticator in the PXA-clustered tumours. CONCLUSION: Methylation profiling is a useful tool in the diagnosis and prognostication of AYA glioblastomas, and the methylation classes have distinct molecular characteristics. The usual molecular diagnostic criteria for adult IDHwt glioblastoma should be applied with caution within the AYA age group.


Assuntos
Astrocitoma , Neoplasias Encefálicas , Glioblastoma , Adolescente , Astrocitoma/patologia , Neoplasias Encefálicas/patologia , Metilação de DNA , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Glioblastoma/patologia , Humanos , Isocitrato Desidrogenase/genética , Mutação , Proteínas Proto-Oncogênicas B-raf/genética , Adulto Jovem
7.
Microbiol Immunol ; 66(10): 465-476, 2022 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-35718749

RESUMO

The antitumoral contribution of γδT cells depends on their activation and differentiation into effectors. This depends on different molecules and membrane receptors, which conditions their physiology. This study aimed to determine the phenotypic characteristics of γδT cells in glioblastoma (GBM) according to five layers of membrane receptors. Among ten GBM cases initially enrolled, five of them who had been confirmed by pathological examination and ten healthy controls underwent phenotyping of peripheral γδT cells by flow cytometry, using the following staining: αßTCR, γδTCR, CD3, CD4, CD8, CD16, CD25, CD27, CD28, CD45, CD45RA, CD56, NKG2D, CD272(BTLA), and CD279(PD-1). Compared with the controls, the results showed no significant change in the number of γδT cells. However, there was a decrease of double-negative (CD4- CD8- ) Tγδ cells and an increase of naive γδT cells, a lack of CD25 expression, a decrease of the expression of CD279, and a remarkable, but not significant, increase in the expression of the CD27 and CD28 costimulation markers. Among the γδT cell subsets, the number of Vδ2 decreased in glioblastoma and showed no significant difference in the expression of CD16, CD56, and NKG2D. In contrast, the number of Vδ1 increased in glioblastoma with overexpression of CD16, CD56, and NKG2D. Our results showed that γδT cells are prone to adopt a pro-inflammatory profile in the glioblastoma context, which suggests that they might be a potential tool to consider in T cell-based immunotherapy in glioblastoma. However, this requires additional investigation on a larger sample size.


Assuntos
Glioblastoma , Subpopulações de Linfócitos T , Antígenos CD28/metabolismo , Glioblastoma/metabolismo , Humanos , Subfamília K de Receptores Semelhantes a Lectina de Células NK/metabolismo , Receptor de Morte Celular Programada 1/metabolismo , Subpopulações de Linfócitos T/metabolismo
8.
Int J Mol Sci ; 23(5)2022 Mar 02.
Artigo em Inglês | MEDLINE | ID: mdl-35269915

RESUMO

Glioblastoma is the most aggressive brain tumour with short survival, partly due to resistance to conventional therapy. Glioma stem cells (GSC) are likely to be involved in treatment resistance, by releasing extracellular vesicles (EVs) containing specific molecular cargoes. Here, we studied the EVs secreted by glioma stem cells (GSC-EVs) and their effects on radiation resistance and glioma progression. EVs were isolated from 3 GSCs by serial centrifugation. NanoSight measurement, cryo-electron microscopy and live imaging were used to study the EVs size, morphology and uptake, respectively. The non-GSC glioma cell lines LN229 and U118 were utilised as a recipient cell model. Wound healing assays were performed to detect cell migration. Colony formation, cell viability and invadopodium assays were conducted to detect cell survival of irradiated recipient cells and cell invasion post GSC-EV treatment. NanoString miRNA global profiling was used to select for the GSC-EVs' specific miRNAs. All three GSC cell lines secreted different amounts of EVs, and all expressed consistent levels of CD9 but different level of Alix, TSG101 and CD81. EVs were taken up by both LN229 and U118 recipient cells. In the presence of GSC-EVs, these recipient cells survived radiation exposure and initiated colony formation. After GSC-EVs exposure, LN229 and U118 cells exhibited an invasive phenotype, as indicated by an increase in cell migration. We also identified 25 highly expressed miRNAs in the GSC-EVs examined, and 8 of these miRNAs can target PTEN. It is likely that GSC-EVs and their specific miRNAs induced the phenotypic changes in the recipient cells due to the activation of the PTEN/Akt pathway. This study demonstrated that GSC-EVs have the potential to induce radiation resistance and modulate the tumour microenvironment to promote glioma progression. Future therapeutic studies should be designed to interfere with these GSC-EVs and their specific miRNAs.


Assuntos
Vesículas Extracelulares , Glioma , MicroRNAs , Microscopia Crioeletrônica , Vesículas Extracelulares/metabolismo , Glioma/genética , Glioma/metabolismo , Glioma/radioterapia , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Células-Tronco Neoplásicas/metabolismo , Microambiente Tumoral
9.
Neurosurg Rev ; 44(3): 1747-1754, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-32845414

RESUMO

Pediatric cerebellar glioblastomas (pcGBMs) are rare and their characteristics remain ill-defined. We conducted a retrospective analysis of pediatric cerebellar glioblastomas who underwent surgery from 2008 to 2019 in our department. Besides, we performed a literature review of the literature data on pcGBMs. Ten children with mean age of 9.4 years were included. During the follow-up, six patients died with mean survival time of 11.7 months, four patients survived with mean follow-up of 28 months. Seven patients underwent molecular analysis, no patients detected IDH1 mutations, four patients (57.1%) had H3K27M mutations, and two patients (28.6%) had MGMT promoter methylation. The literature review identified 38 pcGBMs cases (including ours), with mean age of 8.84 ± 4.20 years (range, 1-16 years). Increased ICP was the commonest sign. Eighteen (47.4%) patients underwent GTR and fifteen (45.5%) patients received STR. Postoperative radiation (RT) was conducted in 28 patients (75.7%) and 23 patients (65.7%) received chemotherapy. During the follow-up, 25 patients died with mean survival time of 12.21 months and 11 patients survived with average follow-up of 29.3 months. Kaplan-Meier survival depicted chemotherapy (P < 0.001) or radiation (P < 0.001) had positive impact on overall survival. Multivariate analysis revealed chemotherapy was a significant predictor of survival with a hazard ratio of 3.264 (P = 0.038). Our study found mean overall survival time for pcGBMs patients was 12.21 months. PcGBMs may have distinct molecular features, with higher incidence of H3K27M mutation and were always IDH1 wild-type. We recommend the routine postoperative radiotherapy and chemotherapy in pcGBMs.


Assuntos
Neoplasias Cerebelares/diagnóstico por imagem , Neoplasias Cerebelares/terapia , Gerenciamento Clínico , Glioblastoma/diagnóstico por imagem , Glioblastoma/terapia , Adolescente , Antineoplásicos/administração & dosagem , Neoplasias Cerebelares/mortalidade , Criança , Pré-Escolar , Feminino , Seguimentos , Glioblastoma/mortalidade , Humanos , Masculino , Radiocirurgia/métodos , Radiocirurgia/mortalidade , Estudos Retrospectivos , Taxa de Sobrevida/tendências
10.
Artigo em Russo | MEDLINE | ID: mdl-34951763

RESUMO

BACKGROUND: Despite the combined treatment in accordance with modern standards, recurrent glioblastoma usually occurs within several months after resection and causes low relapse-free and overall survival. One of the most effective methods for malignant glioma progression is repeated radiotherapy. Indications for this approach have expanded after introduction of stereotactic irradiation into routine clinical practice. OBJECTIVE: To evaluate the results of radiosurgery in patients with recurrent glioblastoma and to identify the factors determining its effectiveness. MATERIAL AND METHODS: Radiosurgery has been carried out in 168 patients with relapses of glioblastoma between 2005 and 2021. This study enrolled 88 patients with 180 foci of local and distant progression. Mean age of patients was 42.8±2.1 years (range 4-73). Mean period between diagnosis and repeated irradiation was 12.7 months. Mean volume of focus was 2.4 cm3, mean dose - 20 Gy. Median follow-up period after radiosurgery was 11.2 months. RESULTS: Repeated irradiation with correction of systemic therapy improved progression-free survival and overall survival with insignificant radiation-induced toxicity. Annual overall survival was 62.2%, median of overall survival after radiosurgery - 15.1 months. Significant factors of local control were marginal dose of at least 18 Gy and distant relapse. Median of progression-free survival in the group of distant progression of glioblastoma was only 3.6 months vs. 9.1 months in patients with local recurrence. CONCLUSION: Repeated irradiation in radiosurgery mode with a dose of 18 Gy and higher is an effective option for local treatment increasing progression-free and overall survival in patients with progression of glioblastoma.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Radiocirurgia , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/radioterapia , Neoplasias Encefálicas/cirurgia , Criança , Pré-Escolar , Glioblastoma/radioterapia , Glioblastoma/cirurgia , Humanos , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/cirurgia , Recidiva , Resultado do Tratamento , Adulto Jovem
11.
Molecules ; 25(6)2020 Mar 23.
Artigo em Inglês | MEDLINE | ID: mdl-32210117

RESUMO

Temozolomide (TMZ)-induced chemoresistance to human glioblastomas is a critical challenge now. Our previous studies showed that honokiol, a major bioactive constituent of Magnolia officinalis (Houpo), can kill human glioblastoma cells and suppresses glioblastoma growth. This study was further aimed to evaluate the effects of honokiol on human drug-resistant glioblastoma cells and the possible mechanisms. The results by data mining in the cancer genome atlas (TCGA) database and immunohistochemistry displayed that expression of caspase-9 mRNA and protein in human glioblastomas was induced. Human TMZ-resistant U87-MG-R9 glioblastoma cells were selected and prepared from human drug-sensitive U87-MG cells. Compared to human drug-sensitive U87-MG cells, TMZ did not affect viability of U87-MG-R9 glioblastoma cells. Interestingly, treatment with honokiol suppressed proliferation and survival of human drug-resistant glioblastoma cells in concentration- and time-dependent manners. Compared to caspase-8 activation, honokiol chiefly increased activity of caspase-9 in U87-MG-R9 cells. Successively, levels of cleaved caspase-3 and activities of caspase-3 and caspase-6 in human TMZ-tolerant glioblastoma cells were augmented following honokiol administration. In parallel, honokiol triggered DNA fragmentation of U87-MG-R9 cells. Accordingly, treatment of human TMZ-resistant glioblastoma cells with honokiol induced cell apoptosis but did not affect cell necrosis. Fascinatingly, suppressing caspase-9 activity using its specific inhibitors repressed honokiol-induced caspase-6 activation, DNA fragmentation, and cell apoptosis. Taken together, this study has shown the major roles of caspase-9 in transducing honokiol-induced mitochondria-dependent apoptosis in human drug-resistant glioblastoma cells. Thus, honokiol may be clinically applied as a drug candidate for treatment of glioblastoma patients with chemoresistance.


Assuntos
Apoptose/efeitos dos fármacos , Compostos de Bifenilo/farmacologia , Caspase 9/metabolismo , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Glioblastoma , Lignanas/farmacologia , Proteínas de Neoplasias/metabolismo , Linhagem Celular Tumoral , Ativação Enzimática/efeitos dos fármacos , Glioblastoma/tratamento farmacológico , Glioblastoma/enzimologia , Glioblastoma/patologia , Humanos , Mitocôndrias/metabolismo , Mitocôndrias/patologia , Temozolomida/farmacologia
12.
Rev Neurol (Paris) ; 176(9): 724-732, 2020 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-32307112

RESUMO

Glioblastoma multiforme (GBM) is the most common primary brain tumor in adults. The incidence of malignant gliomas is growing in the elderly population. Unfortunately, increasing age is one of the most important negative prognostic factors for this tumor. For a long time, the treatment of elderly patients with GBM was controversial. Currently, more active strategies are the rule. Indeed, as in the younger population, prospective randomized studies have recently established the benefit of radiotherapy associated with concomitant and adjuvant chemotherapy by temozolomide in older patients suffering from malignant gliomas with good functional status. The application of chemotherapy alone may be especially useful in patients with poor functional status and O-6-methylguanine-DNA methyltransferase (MGMT) promotor methylation. For the portion of the elderly population identified as frail, treatment decisions should be made in the context of a comprehensive geriatric evaluation while also taking into account quality of life and concomitant pathologies. The willingness of the patient and his or her caregivers will also be key to the therapeutic decision. Symptomatic treatments such as corticosteroids and antiepileptic drugs may be less tolerated in this population compared to younger patients and should be used only if requested. In the future, it will be necessary to continue to develop specific schedules of treatment in the frail population. For this reason, prospective randomized clinical trials are still needed to pursue improvements in the pattern of care of malignant glioma in elderly individuals.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Idoso , Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/tratamento farmacológico , Glioblastoma/tratamento farmacológico , Humanos , Estudos Prospectivos , Qualidade de Vida , Temozolomida
13.
Glia ; 67(8): 1558-1570, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31058365

RESUMO

GPR124 is involved in embryonic development and remains expressed by select organs. The importance of GPR124 during development suggests that its aberrant expression might participate in tumor growth. Here we show that both increases and decreases in GPR124 expression in glioblastoma cells reduce cell proliferation by differentially altering the duration mitotic progression. Using mass spectrometry-based proteomics, we discovered that GPR124 interacts with ch-TOG, a known regulator of both microtubule (MT)-plus-end assembly and mitotic progression. Accordingly, changes in GPR124 expression and ch-TOG similarly affect MT assembly measured by real-time microscopy in cells. Our study describes a novel molecular interaction involving GPR124 and ch-TOG at the plasma membrane that controls glioblastoma cell proliferation by modifying MT assembly rates and controlling the progression of distinct phases of mitosis.


Assuntos
Neoplasias Encefálicas/metabolismo , Proliferação de Células/fisiologia , Glioblastoma/metabolismo , Microtúbulos/metabolismo , Mitose/fisiologia , Receptores Acoplados a Proteínas G/metabolismo , Adulto , Encéfalo/metabolismo , Membrana Celular/metabolismo , Células Cultivadas , Feminino , Expressão Gênica , Humanos , Masculino , Proteínas Associadas aos Microtúbulos/metabolismo , Pessoa de Meia-Idade
14.
J Magn Reson Imaging ; 49(1): 184-194, 2019 01.
Artigo em Inglês | MEDLINE | ID: mdl-29676844

RESUMO

BACKGROUND: Accurate differentiation of brain infections from necrotic glioblastomas (GBMs) may not always be possible on morphologic MRI or on diffusion tensor imaging (DTI) and dynamic susceptibility contrast perfusion-weighted imaging (DSC-PWI) if these techniques are used independently. PURPOSE: To investigate the combined analysis of DTI and DSC-PWI in distinguishing brain injections from necrotic GBMs. STUDY TYPE: Retrospective. POPULATION: Fourteen patients with brain infections and 21 patients with necrotic GBMs. FIELD STRENGTH/SEQUENCE: 3T MRI, DTI, and DSC-PWI. ASSESSMENT: Parametric maps of mean diffusivity (MD), fractional anisotropy (FA), coefficient of linear (CL), and planar anisotropy (CP) and leakage corrected cerebral blood volume (CBV) were computed and coregistered with postcontrast T1 -weighted and FLAIR images. All lesions were segmented into the central core and enhancing region. For each region, median values of MD, FA, CL, CP, relative CBV (rCBV), and top 90th percentile of rCBV (rCBVmax ) were measured. STATISTICAL TESTS: All parameters from both regions were compared between brain infections and necrotic GBMs using Mann-Whitney tests. Logistic regression analyses were performed to obtain the best model in distinguishing these two conditions. RESULTS: From the central core, significantly lower MD (0.90 × 10-3 ± 0.44 × 10-3 mm2 /s vs. 1.66 × 10-3 ± 0.62 × 10-3 mm2 /s, P = 0.001), significantly higher FA (0.15 ± 0.06 vs. 0.09 ± 0.03, P < 0.001), and CP (0.07 ± 0.03 vs. 0.04 ± 0.02, P = 0.009) were observed in brain infections compared to those in necrotic GBMs. Additionally, from the contrast-enhancing region, significantly lower rCBV (1.91 ± 0.95 vs. 2.76 ± 1.24, P = 0.031) and rCBVmax (3.46 ± 1.41 vs. 5.89 ± 2.06, P = 0.001) were observed from infective lesions compared to necrotic GBMs. FA from the central core and rCBVmax from enhancing region provided the best classification model in distinguishing brain infections from necrotic GBMs, with a sensitivity of 91% and a specificity of 93%. DATA CONCLUSION: Combined analysis of DTI and DSC-PWI may provide better performance in differentiating brain infections from necrotic GBMs. LEVEL OF EVIDENCE: 1 Technical Efficacy: Stage 2 J. Magn. Reson. Imaging 2019;49:184-194.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Encéfalo/diagnóstico por imagem , Imagem de Difusão por Ressonância Magnética , Glioblastoma/diagnóstico por imagem , Infecções/diagnóstico por imagem , Angiografia por Ressonância Magnética , Necrose/diagnóstico por imagem , Adulto , Idoso , Anisotropia , Encéfalo/microbiologia , Meios de Contraste/administração & dosagem , Diagnóstico Diferencial , Feminino , Humanos , Masculino , Pessoa de Meia-Idade , Reprodutibilidade dos Testes , Estudos Retrospectivos
15.
J Neurooncol ; 142(1): 149-159, 2019 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-30535596

RESUMO

PURPOSE: There are no widely accepted MRI markers that predict treatment outcomes of bevacizumab among patients with recurrent glioblastoma (GB). We aimed to determine if conventional MRI features of recurrent GB predict survival of patients receiving bevacizumab. METHODS: Patients with recurrent GB were retrospectively included if they received bevacizumab monotherapy between 2008 and 2017 after failure of standard treatment. Their MRI studies obtained at baseline and tumor recurrence, prior to bevacizumab treatment, were evaluated for multiple MRI features including measurable tumor, baseline multicentric tumors, distant recurrence, non-contrast-enhancing tumor, deep white matter invasion, multiple parenchymal tumors, bilateral cerebral involvement, ependymal extension and leptomeningeal dissemination. Predictive values of MRI features and patient characteristics on patient survival were statistically analyzed. RESULTS: A total of 103 patients were included. Baseline multicentric tumors (OR = 4.07; P = 0.042) and distant recurrence (OR = 28.5; P < 0.001) were two significant predictors of 3-month progression-free survival (PFS) rate. Distant recurrence (HR = 3.94; P < 0.001) was the only independent predictor of PFS. Baseline multicentric tumors (HR = 1.97; P = 0.028), distant recurrence (HR = 4.73; P < 0.001) and leptomeningeal dissemination (HR = 2.28; P = 0.044) were three independent predictors of overall survival. CONCLUSIONS: Baseline multicentric tumors, distant recurrence and leptomeningeal dissemination predicted poor survival among patients receiving bevacizumab for recurrent GB. Conventional MRI may help selecting patients with recurrent GB for bevacizumab treatment.


Assuntos
Antineoplásicos Imunológicos/uso terapêutico , Bevacizumab/uso terapêutico , Neoplasias Encefálicas/patologia , Glioblastoma/patologia , Recidiva Local de Neoplasia/patologia , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/tratamento farmacológico , Neoplasias Encefálicas/mortalidade , Progressão da Doença , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/tratamento farmacológico , Glioblastoma/mortalidade , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/diagnóstico por imagem , Recidiva Local de Neoplasia/tratamento farmacológico , Recidiva Local de Neoplasia/mortalidade , Prognóstico , Estudos Retrospectivos , Taxa de Sobrevida , Resultado do Tratamento , Adulto Jovem
16.
Acta Radiol ; 60(12): 1663-1672, 2019 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-31084193

RESUMO

BACKGROUND: Isocitrate dehydrogenase (IDH) mutation has diagnostic and prognostic values in glioblastomas. Peritumoral invasion of glioma cells is a cardinal feature of glioblastomas. PURPOSE: To evaluate the contribution of DWI and DSC-PWI in the enhancing and peri-enhancing region for discriminating glioblastomas IDH genotypes, and the diagnostic values of combining two techniques in the peri-enhancing region compared with those in the enhancing region. MATERIAL AND METHODS: We retrospectively reviewed the conventional MRI (cMRI), DWI and DSC-PWI obtained from 10 patients with IDH-mutated (IDH-m) glioblastomas and 65 patients with IDH wild-type (IDH-w) glioblastomas. Features of cMRI, relative minimum ADC in the enhancing region (rADCmin-t) and peri-enhancing area (rADCmin-p), and relative maximum CBV values in the enhancing region (rCBVmax-t) and peri-enhancing region (rCBVmax-p) were compared between two groups. Receiver operating characteristic curves and logistic regression analysis were used to assess diagnostic performance. RESULTS: IDH-m glioblastomas tended to present in frontal lobes and younger patients. The rADCmin-t (P = 0.042) were significantly lower in IDH-w than IDH-m. Both rCBVmax-t and rCBVmax-p showed significant differences between two subgroups (all P < 0.001). The optimal cutoff values in prediction of IDH-m were >0.98 for rADCmin-t, <7.27 for rCBVmax-t, and < 0.97 for rCBVmax-p. Multivariate logistic regression revealed that the combination of rADCmin-t and rCBVmax-t yielded the highest sensitivity and specificity. CONCLUSION: The rCBVmax-t or rCBVmax-p may serve as preferable and comparable imaging biomarkers for evaluation of glioblastomas IDH status. The combination of rADCmin-t and rCBVmax-t may yield the maximum predictive power for differentiating IDH status.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/genética , Glioblastoma/diagnóstico por imagem , Glioblastoma/genética , Isocitrato Desidrogenase/genética , Imageamento por Ressonância Magnética/métodos , Adulto , Feminino , Genótipo , Humanos , Masculino , Pessoa de Meia-Idade , Mutação , Invasividade Neoplásica , Estudos Retrospectivos
17.
Acta Neurochir (Wien) ; 161(6): 1109-1124, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-30980242

RESUMO

There is a distinct need for new and second-line therapies to delay or prevent local tumor regrowth after current standard of care therapy. Intracavitary radioimmunotherapy, in combination with radiotherapy, is discussed in the present review as a therapeutic strategy of high potential. We performed a systematic literature search following the Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA). The available body of literature on intracavitary radioimmunotherapy (iRIT) in glioblastoma and anaplastic astrocytomas is presented. Several past and current phase I and II clinical trials, using mostly an anti-tenascin monoclonal antibody labeled with I-131, have shown median overall survival of 19-25 months in glioblastoma, while adverse events remain low. Tenascin, followed by EGFR and variants, or smaller peptides have been used as targets, and most clinical studies were performed with I-131 or Y-90 as radionuclides while only recently Re-188, I-125, and Bi-213 were applied. The pharmacokinetics of iRIT, as well as the challenges encountered with this therapy, is comprehensively discussed. This promising approach deserves further exploration in future studies by incorporating several innovative modifications.


Assuntos
Neoplasias Encefálicas/radioterapia , Glioma/radioterapia , Radioimunoterapia/métodos , Anticorpos Monoclonais/uso terapêutico , Neoplasias Encefálicas/patologia , Glioma/patologia , Humanos , Radioisótopos do Iodo/uso terapêutico , Radioimunoterapia/tendências , Radioisótopos de Ítrio/uso terapêutico
18.
Pharmacol Res ; 132: 211-219, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29274786

RESUMO

Blood perfusion was always lower in tumor tissues as compared with that in surrounding normal tissues which lead to inadequate nanomedicine delivery to tumors. Inspired by the upregulation of both endothelin-1 (ET1) and its ETA receptor in tumor tissues and the crucial contribution of ET1-ETA receptor signaling to maintain myogenic tone of tumor vessels, we supposed that inhibition of ET1-ETA receptor signaling might selectively improve tumor perfusion and help deliver nanomedicine to tumors. Using human U87 MG glioblastomas with abundant vessels as the tumor model, immunofluorescence staining demonstrated that ETA receptor was overexpressed by in glioblastomas tissues compared with normal brain tissues. A single administration of ETA receptor antagonist BQ123 at the dose of 0.5 mg/kg could effectively improve tumor perfusion which was evidenced by in vivo photoacoustic imaging. Additionally, a single treatment of BQ123 could significantly improve the accumulation of nanoparticles (NPs) around 115 nm in tumors with a more homogeneous distribution pattern by in vivo imaging, ex vivo imaging as well as in vivo distribution experiments. Furthermore, BQ123 successfully increased the therapeutic benefits of paclitaxel-loaded NPs and significantly elongated the survival time of orthotropic glioblastomas-bearing animal models. In summary, the present study provided a new strategy to selectively improve tumor perfusion and therefore benefit nanomedicine delivery for tumor therapy. As ET1-ETA receptor signaling was upregulated in a variety of tumors, this strategy might open a new avenue for tumor treatment.


Assuntos
Antineoplásicos Fitogênicos/administração & dosagem , Antagonistas dos Receptores de Endotelina/administração & dosagem , Glioblastoma/tratamento farmacológico , Nanopartículas/administração & dosagem , Paclitaxel/administração & dosagem , Peptídeos Cíclicos/administração & dosagem , Animais , Antineoplásicos Fitogênicos/farmacocinética , Linhagem Celular Tumoral , Sistemas de Liberação de Medicamentos , Antagonistas dos Receptores de Endotelina/farmacocinética , Glioblastoma/metabolismo , Humanos , Masculino , Camundongos Endogâmicos BALB C , Camundongos Nus , Paclitaxel/farmacocinética
19.
Ann Diagn Pathol ; 37: 62-66, 2018 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-30286327

RESUMO

Astrocytomas represent the majority of cerebral gliomas. Studies show that the anti-inflammatory protein Annexin-A1 (ANXA1) is associated with the tumor invasion process and that its actions can be mediated by the receptor for formylated peptides (FPR). Therefore, we evaluated the expression of ANXA1, the receptor FPR2 and matrix metalloproteinases 2 and 9 (MMP-2 and MMP-9) in brain astrocytomas. Detection of proteins was performed in sections of diffuse astrocytomas (grade II), anaplastic astrocytomas (grade III) and glioblastomas (GBM, grade IV) and quantifications were made by densitometry. Our analyses showed increased expression of ANXA1 in astrocytomas of all grades, but especially in GBM. The expression of FPR2 is similar to that found for ANXA1, being higher in GBM. Immunostaining for MMPs is also stronger as the degree of malignancy increases, especially with respect to MMP-9. The positive correlation between ANXA1/FPR2 and ANXA1/MMP-9 was observed in all tumors studied. The data indicate the possible action of ANXA1 and FPR2 on the development and progression of astrocytomas, related to increased expression of MMP-9. Thereby, ANXA1 and FPR2 are involved in the biology and malignancy of diffuse astrocytic tumors.


Assuntos
Anexina A1/biossíntese , Astrocitoma/patologia , Biomarcadores Tumorais/biossíntese , Neoplasias Encefálicas/patologia , Receptores de Formil Peptídeo/biossíntese , Receptores de Lipoxinas/biossíntese , Adulto , Idoso , Feminino , Humanos , Masculino , Metaloproteinase 2 da Matriz/biossíntese , Metaloproteinase 9 da Matriz/biossíntese , Pessoa de Meia-Idade
20.
Br J Neurosurg ; 32(5): 528-535, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-30073866

RESUMO

OBJECT: It is becoming well-established that increasing extent of resection with decreasing residual volume is associated with delayed recurrence and prolonged survival for patients with glioblastoma (GBM). These prior studies are based on evaluating the contrast-enhancing (CE) tumour and not the surrounding fluid attenuated inversion recovery (FLAIR) volume. It therefore remains unclear if the resection beyond the CE portion of the tumour if it translates into improved outcomes for patients with GBM. METHODS: Adult patients who underwent resection of a primary glioblastoma at a tertiary care institution between January 1, 2007 and December 31, 2012 and underwent radiation and temozolomide chemotherapy were retrospectively reviewed. Pre and postoperative MRI images were measured for CE tumour and FLAIR volumes. Multivariate proportional hazards were used to assess associations with both time to recurrence and death. Values with p < 0.05 were considered statistically significant. RESULTS: 245 patients met the inclusion criteria. The median [IQR] preoperative CE and FLAIR tumour volumes were 31.9 [13.9-56.1] cm3 and 78.3 [44.7-115.6] cm3, respectively. Following surgery, the median [IQR] postoperative CE and FLAIR tumour volumes were 1.9 [0-7.1] cm3 and 59.7 [29.7-94.2] cm3, respectively. In multivariate analyses, the postoperative FLAIR volume was not associated with recurrence and/or survival (p > 0.05). However, the postoperative CE tumour volume was significantly associated with both recurrence [HR (95%CI); 1.026 (1.005-1.048), p = 0.01] and survival [HR (95%CI); 1.027 (1.007-1.032), p = 0.001]. The postoperative FLAIR volume was also not associated with recurrence and/or survival among patients who underwent gross total resection of the CE portion of the tumour as well as those who underwent supratotal resection. CONCLUSIONS: In this study, the volume of CE tumour remaining after resection is more important than FLAIR volume in regards to recurrence and survival for patients with GBM.


Assuntos
Neoplasias Encefálicas/cirurgia , Glioblastoma/cirurgia , Adulto , Idoso , Neoplasias Encefálicas/patologia , Estudos de Viabilidade , Feminino , Glioblastoma/patologia , Humanos , Imageamento por Ressonância Magnética/métodos , Masculino , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/cirurgia , Neoplasia Residual/patologia , Cuidados Pós-Operatórios , Estudos Retrospectivos , Resultado do Tratamento , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA