Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
1.
Cell Death Dis ; 13(7): 588, 2022 07 08.
Artigo em Inglês | MEDLINE | ID: mdl-35803912

RESUMO

Due to the negligence of the complex tumor immune microenvironment, traditional treatment for glioblastoma has reached its limitation and cannot achieve a satisfying outcome in the past decade. The emergence of immunotherapy based on the theory of cancer-immunity cycle has brought a new dawn to glioblastoma patients. However, the results of most phase II and phase III clinical trials are not optimistic due to the simple focus on T cells activation rather than other immune cells involved in anti-tumor immunity. NK cells play a critical role in both innate and adaptive immunity, having the ability to coordinate immune response in inflammation, autoimmune disease and cancer. They are expected to cooperate with T cells to maximize the anti-tumor immune effect and have great potential in treating glioblastoma. Here, we describe the traditional treatment methods and current immunotherapy strategies for glioblastoma. Then, we list a microenvironment map and discuss the reasons for glioblastoma inhibitory immunity from multiple perspectives. More importantly, we focus on the advantages of NK cells as potential immune regulatory cells and the ways to maximize their anti-tumor immune effect. Finally, our outlook on the directions and potential applications of NK cell-based therapy combining with the advance technologies is presented. This review depicts NK cell awakening as the precondition to unleash the cancer-immunity cycle against glioblastoma and elaborate this idea from biology to clinical treatment.


Assuntos
Glioblastoma , Neoplasias , Imunidade Adaptativa , Glioblastoma/patologia , Humanos , Imunoterapia/métodos , Imunoterapia Adotiva/métodos , Células Matadoras Naturais/metabolismo , Neoplasias/metabolismo , Microambiente Tumoral
3.
Front Oncol ; 11: 650153, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34055619

RESUMO

BACKGROUND: Glioma is the most common malignant brain tumor in adults, with its tumor-promoting immune microenvironment always being intricate to handle with. Amounts of evidence has accumulated to suggest that alternative splicing (AS) is related to tumor immune microenvironment. However, comprehensive analysis of immune-related AS events and their clinical significance are still lacking in glioma. METHODS: AS events and transcriptome data of 653 glioma patients were downloaded online. ssGSEA was performed on transcriptome data of 653 patients to divided them into low, medium and high immune cell infiltration groups. Immune-related AS events were filtrated based on this grouping. Then lasso Cox regression analysis and multivariate Cox regression analysis were done to achieve an immune-related AS events prognostic signature for glioma. Kaplan-Meier analysis, ROC analyses, univariate Cox regression and multivariate Cox regression were performed to reveal the independent prognostic role of this signature. Meanwhile, a nomogram was constructed to achieved better prognostic value for glioma patients. Besides, functional enrichment analyses and correlation analyses with immune cells infiltration were used to validated the immune-related characteristic of this signature. RESULTS: 36 immune-related AS events were achieved based on the grouping mentioned above. A nine-immune-related alternative splicing event signature was built for glioma patients. This signature showed an independent prognostic value and a nomogram containing gender, age, Karnofsky performance score, grade, IDH status, MGMT promoter status and risk score derived from the signature was constructed with a higher predictive ability for overall survival. Association with the infiltration of immune cell subtypes was validated and functional enrichment analysis found that the signature was mainly enriched in immune-related and pro-tumor functions. CONCLUSION: Our research presented all immune-related AS events in glioma, identified an immune-related prognostic AS events risk model and a nomogram was constructed to predict the prognosis individually and more precisely. Tight connection was verified between this signature and clinical characteristics. Also, immune cells infiltration and immune checkpoints expression level were proved to link to risk scores, which enhanced the understanding of relationship between AS events and glioma immune microenvironment, firstly revealing the potential role of AS in immunotherapy of glioma.

4.
Am J Cancer Res ; 11(4): 1069-1086, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33948346

RESUMO

Glioblastoma is one of the most common malignant tumors in the central nervous system. Due to the high plasticity, heterogeneity and complexity of the tumor microenvironment, these tumors are resistant to almost all therapeutic strategies when they reach an advanced stage. Along with being a unique and effective way to kill cancer cells, tumor-treating fields (TTFields) has emerged as a breakthrough among glioblastoma therapies since the advent of temozolomide (TMZ), and the combination of these treatments has gradually been promoted and applied in the clinic. The combination of TTFields with other therapies is particularly suitable for this type of "cold" tumors and has attracted a large amount of attention from clinicians and researchers in the era of cancer cocktail therapy. Here, we introduced the current treatment regimen for glioblastoma, highlighting the unique advantages of TTFields in the treatment of glioblastoma. Then, we summarized current glioblastoma clinical trials that combine TTFields and other therapies. In addition, the main and potential mechanisms of TTFields were introduced to further understand the rationale for each combination therapy. Finally, we focused on the most advanced technologies applied in glioblastoma research and treatment and the prospect of their combination with TTFields. This review provides a unique overview of glioblastoma treatment.

5.
Cancer Sci ; 112(7): 2835-2844, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33932065

RESUMO

This study aims to build a radiological model based on standard MR sequences for detecting methylguanine methyltransferase (MGMT) methylation in gliomas using texture analysis. A retrospective cross-sectional study was undertaken in a cohort of 53 glioma patients who underwent standard preoperative magnetic resonance (MR) imaging. Conventional visual radiographic features and clinical factors were compared between MGMT promoter methylated and unmethylated groups. Texture analysis extracted the top five most powerful texture features of MR images in each sequence quantitatively for detecting the MGMT promoter methylation status. The radiomic signature (Radscore) was generated by a linear combination of the five features and estimates in each sequence. The combined model based on each Radscore was established using multivariate logistic regression analysis. A receiver operating characteristic (ROC) curve, nomogram, calibration, and decision curve analysis (DCA) were used to evaluate the performance of the model. No significant differences were observed in any of the visual radiographic features or clinical factors between different MGMT methylated statuses. The top five most powerful features were selected from a total of 396 texture features of T1, contrast-enhanced T1, T2, and T2 FLAIR. Each sequence's Radscore can distinguish MGMT methylated status. A combined model based on Radscores showed differentiation between methylated MGMT and unmethylated MGMT both in the glioblastoma (GBM) dataset as well as the dataset for all other gliomas. The area under the ROC curve values for the combined model was 0.818, with 90.5% sensitivity and 72.7% specificity, in the GBM dataset, and 0.833, with 70.2% sensitivity and 90.6% specificity, in the overall gliomas dataset. Nomogram, calibration, and DCA also validated the performance of the combined model. The combined model based on texture features could be considered as a noninvasive imaging marker for detecting MGMT methylation status in glioma.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Neoplasias Encefálicas/enzimologia , Metilases de Modificação do DNA/metabolismo , Enzimas Reparadoras do DNA/metabolismo , Glioma/diagnóstico por imagem , Glioma/enzimologia , Proteínas Supressoras de Tumor/metabolismo , Adulto , Idoso , Neoplasias Encefálicas/patologia , Meios de Contraste , Estudos Transversais , Metilação de DNA , Reparo do DNA , Técnicas de Apoio para a Decisão , Feminino , Glioblastoma/diagnóstico por imagem , Glioblastoma/enzimologia , Glioblastoma/patologia , Glioma/patologia , Humanos , Imageamento por Ressonância Magnética , Masculino , Pessoa de Meia-Idade , Nomogramas , Curva ROC , Estudos Retrospectivos , Sensibilidade e Especificidade , Adulto Jovem
6.
Biomed Res Int ; 2021: 8872977, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33553434

RESUMO

INTRODUCTION: Glioblastoma (GBM) is one of the most frequent primary intracranial malignancies, with limited treatment options and poor overall survival rates. Alternated glucose metabolism is a key metabolic feature of tumour cells, including GBM cells. However, due to high cellular heterogeneity, accurately predicting the prognosis of GBM patients using a single biomarker is difficult. Therefore, identifying a novel glucose metabolism-related biomarker signature is important and may contribute to accurate prognosis prediction for GBM patients. METHODS: In this research, we performed gene set enrichment analysis and profiled four glucose metabolism-related gene sets containing 327 genes related to biological processes. Univariate and multivariate Cox regression analyses were specifically completed to identify genes to build a specific risk signature, and we identified ten mRNAs (B4GALT7, CHST12, G6PC2, GALE, IL13RA1, LDHB, SPAG4, STC1, TGFBI, and TPBG) within the Cox proportional hazards regression model for GBM. RESULTS: Depending on this glucose metabolism-related gene signature, we divided patients into high-risk (with poor outcomes) and low-risk (with satisfactory outcomes) subgroups. The results of the multivariate Cox regression analysis demonstrated that the prognostic potential of this ten-gene signature is independent of clinical variables. Furthermore, we used two other GBM databases (Chinese Glioma Genome Atlas (CGGA) and REMBRANDT) to validate this model. In the functional analysis results, the risk signature was associated with almost every step of cancer progression, such as adhesion, proliferation, angiogenesis, drug resistance, and even an immune-suppressed microenvironment. Moreover, we found that IL31RA expression was significantly different between the high-risk and low-risk subgroups. CONCLUSION: The 10 glucose metabolism-related gene risk signatures could serve as an independent prognostic factor for GBM patients and might be valuable for the clinical management of GBM patients. The differential gene IL31RA may be a potential treatment target in GBM.


Assuntos
Neoplasias Encefálicas/genética , Neoplasias Encefálicas/mortalidade , Glioblastoma/genética , Glioblastoma/mortalidade , Glucose/metabolismo , Idoso , Biomarcadores Tumorais/genética , Neoplasias Encefálicas/metabolismo , Feminino , Regulação Neoplásica da Expressão Gênica , Glioblastoma/metabolismo , Glioma/genética , Gluconeogênese/genética , Glucose/genética , Glicólise/genética , Humanos , Masculino , Pessoa de Meia-Idade , Prognóstico , Modelos de Riscos Proporcionais , RNA Mensageiro , Reprodutibilidade dos Testes
7.
J Comput Assist Tomogr ; 45(1): 110-120, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33475317

RESUMO

OBJECTIVE: To investigate the value of radiomics analyses based on different magnetic resonance (MR) sequences in the noninvasive evaluation of glioma characteristics for the differentiation of low-grade glioma versus high-grade glioma, isocitrate dehydrogenase (IDH)1 mutation versus IDH1 wild-type, and mutation status and 6-methylguanine-DNA methyltransferase (MGMT) promoter methylation (+) versus MGMT promoter methylation (-) glioma. METHODS: Fifty-nine patients with untreated glioma who underwent a standard 3T-MR tumor protocol were included in the study. A total of 396 radiomics features were extracted from the MR images, with the manually delineated tumor as the volume of interest. Clinical imaging diagnostic features (tumor location, necrosis/cyst change, crossing midline, and the degree of enhancement or peritumoral edema) were analyzed by univariate logistic regression to select independent clinical factors. Radiomics and combined clinical-radiomics models were established for grading and molecular genomic typing of glioma by multiple logistic regression and cross-validation. The performance of the models based on different sequences was evaluated by using receiver operating characteristic curves, nomograms, and decision curves. RESULTS: The radiomics model based on T1-CE performed better than models based on other sequences in predicting the tumor grade and the IDH1 status of the glioma. The radiomics model based on T2 performed better than models based on other sequences in predicting the MGMT methylation status of glioma. Only the T1 combined clinical-radiomics model showed improved prediction performance in predicting tumor grade and the IDH1 status. CONCLUSIONS: The results demonstrate that state-of-the-art radiomics analysis methods based on multiparametric MR image data and radiomics features can significantly contribute to pretreatment glioma grading and molecular subtype classification.


Assuntos
Neoplasias Encefálicas/diagnóstico por imagem , Metilases de Modificação do DNA/genética , Enzimas Reparadoras do DNA/genética , Glioma/diagnóstico por imagem , Isocitrato Desidrogenase/genética , Imageamento por Ressonância Magnética/métodos , Interpretação de Imagem Radiográfica Assistida por Computador/métodos , Proteínas Supressoras de Tumor/genética , Adolescente , Adulto , Idoso , Neoplasias Encefálicas/genética , Neoplasias Encefálicas/patologia , Criança , Metilação de DNA , Feminino , Glioma/genética , Glioma/patologia , Humanos , Modelos Logísticos , Masculino , Pessoa de Meia-Idade , Mutação , Estadiamento de Neoplasias , Regiões Promotoras Genéticas , Adulto Jovem
8.
Clin Neurol Neurosurg ; 201: 106421, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33370626

RESUMO

OBJECTIVE: To study the expression of FAM46A in glioblastoma (GBM) and analyze its significance in predicting the prognosis of patients. MATERIALS AND METHODS: mRNA expression and clinical data of patients with GBM were retrieved from ONCOMINE databases and The Cancer Genome Atlas (TCGA) database. Immunohistochemistry was performed in a tissue microarray including 110 GBM cases and 12 normal controls to determine the expression of FAM46A protein. Then, Kaplan-Meier curve and Cox regression model were used to investigate the relationship between FAM46A expression and clinical outcome. Coexpressed genes of FAM46A were analyzed by Linked Omics, Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG). RESULTS: Upregulated expression of FAM46A was found in both TCGA and our cohort. High FAM46A expression was associated with the poor prognosis of patients with GBM and could be identified as an independent risk factor for overall survival (HR = 1.652, p = 0.022). Further bioinformatics analysis revealed that FAM46A might be involved in cell motility and endoplasmic reticulum proteostasis and stress to promote GBM progression. CONCLUSION: Our findings suggest that increased expression of FAM46A in GBM is a novel biomarker for predicting poor outcome of patients and that targeting FAM46A may serve as a potential therapy for this disease.


Assuntos
Biomarcadores Tumorais/genética , Neoplasias Encefálicas/metabolismo , Regulação Neoplásica da Expressão Gênica/genética , Glioblastoma/metabolismo , Polinucleotídeo Adenililtransferase/metabolismo , Neoplasias Encefálicas/mortalidade , Estudos de Coortes , Biologia Computacional/métodos , Feminino , Glioblastoma/mortalidade , Humanos , Masculino , Pessoa de Meia-Idade , Polinucleotídeo Adenililtransferase/genética , Prognóstico
9.
Cancer Manag Res ; 12: 12011-12020, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-33262651

RESUMO

PURPOSE: To explore the regulatory mechanism of long non-coding RNA small nucleolar RNA host gene 1 (SNHG1) in glioma. MATERIALS AND METHODS: The expression of SNHG1 and miR-140-5p in glioma tissues and glioma cell lines (LN-18, KNS-81, and KALS-1) was determined, and the effect of the two on cell proliferation, invasion, and PI3K/AKT pathway was analyzed. RESULTS: SNHG1 was overexpressed in glioma tissues, while miR-140-5p was underexpressed in them, and there was a significant negative correlation between SNHG1 and miR-140-5p. In addition, both down-regulation of SNHG1 and up-regulation of miR-140-5p significantly inhibited the malignant proliferation and invasion of glioma, intensified the apoptosis, and also significantly suppressed the activation of the PI3K/AKT pathway. The dual-luciferase reporter assay, RNA pull-down assay, and RIP determination all confirmed that there was a targeting relationship between SNHG1 and miR-140-5p, and there was no difference between KNS-81 and KALS-1 cells transfected with SNHG1+mimics and si-SNHG1+inhibitor and those in the si-NC group with unrelated sequences in terms of cell malignant progression. CONCLUSION: SNHG1/miR-140-5p axis and its regulation on PI3K/AKT pathway might be a novel therapeutic direction to curb the malignant progression of glioma.

10.
Am J Cancer Res ; 10(8): 2242-2257, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32905502

RESUMO

The high mortality and poor clinical prognosis of glioblastoma multiforme (GBM) are concerns for many GBM patients as well as clinicians and researchers. The lack of a preclinical model that can easily be established and accurately recapitulate tumour biology and the tumour microenvironment further complicates GBM research and its clinical translation. GBM organoids (GBOs) are promising high-fidelity models that can be applied to model the disease, develop drugs, establish a living biobank, mimic therapeutic responses and explore personalized therapy. However, GBO models face some challenges, including deficient immune responses, absent vascular system and controversial reliability. In recent years, considerable progress has been achieved in the improvement of brain tumour organoid models and research based on such models. In addition to the traditional cultivation method, these models can be cultivated via genetic engineering and co-culture of cerebral organoids and GBM. In this review, we summarize the applications of GBM organoids and related advances and provide our opinions on associated limitations and challenges.

11.
Endokrynol Pol ; 71(5): 425-431, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32797477

RESUMO

INTRODUCTION: Pituitary adenomas constitute one of the most common intracranial tumours. The mouse double minute 2 homologue (MDM2) is considered as an important oncogene in many tumours, but it has been little studied in pituitary adenomas and the mechanism is not well understood. The purpose of this study was to investigate the function of MDM2 and its primary mechanism of action in pituitary adenoma cells. MATERIAL AND METHODS: The expression of MDM2 in pituitary adenoma cell lines and normal cells was determined by real-time polymerase chain reaction (RT-PCR). The proliferation and apoptosis of pituitary adenoma cells after inhibition of MDM2 expression were detected by MTS and flow cytometry, respectively. The protein expressions of MDM2 and p53 were detected by western blot. Co-IP was used to detect the direct binding between MDM2 and p53. RESULTS: The results of RT-PCR showed that MDM2 was significantly up-regulated in pituitary adenoma cell lines. Inhibition of MDM2 suppressed the proliferation and promoted apoptosis of pituitary adenoma cells. However, inhibiting the expression of MDM2 can promotethe protein expression of p53. The results of co-IP showed that MDM2 interacted with p53 by direct combination. Then, we inhibited the expressions of p53 and MDM2 simultaneously in the pituitary adenoma cells by co-transfecting siRNAs, and the results showed that, compared with the group that inhibited MDM2 alone, cell proliferation of the co-transfected group increased and apoptosis of the cotransfected group decreased, which was similar to the NC group. CONCLUSIONS: Taken together, these results suggest that MDM2 promoted the proliferation and inhibited the apoptosis of pituitary adenoma cells by directly interacting with p53 in pituitary adenoma cells. Therefore, MDM2-p53 may serve as a novel marker and therapeutic target for pituitary adenomas.


Assuntos
Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Apoptose/fisiologia , Proliferação de Células , Humanos , Neoplasias Hipofisárias/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Células Tumorais Cultivadas
12.
Biosci Biotechnol Biochem ; 84(5): 911-918, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31959058

RESUMO

Pituitary adenomas constitute one of the most common intracranial tumors. MicroRNAs play an important role in development and progression of pituitary adenomas. In this study, we showed that miR-219a-2-3p was significantly down-regulated in pituitary adenomas cells. Overexpression of miR-219a-2-3p suppressed the proliferation and promoted apoptosis of pituitary adenomas cells. After bioinformatics analysis, we found that MDM2 was one of the downstream targets of miR-219a-2-3p. Further researches showed that miR-219a-2-3p could reduce the protein level of MDM2 by binding to the 3'-UTR of MDM2 and promoted p53 expression. Then, we overexpressed both miR-219a-2-3p and MDM2 in the same group and found that it could counteract the effect of overexpressing miR-219a-2-3p alone on proliferation and apoptosis of pituitary adenoma cells. Taken together, these results suggested that miR-219a-2-3p regulated the proliferation and apoptosis by targeting MDM2/p53 in pituitary adenomas. Therefore, miR-219a-2-3p may serve as a novel marker and therapeutic target for pituitary adenomas.


Assuntos
Adenoma/metabolismo , Apoptose/genética , Proliferação de Células/genética , MicroRNAs/metabolismo , Neoplasias Hipofisárias/metabolismo , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Transdução de Sinais/genética , Proteína Supressora de Tumor p53/metabolismo , Regiões 3' não Traduzidas/genética , Adenoma/patologia , Animais , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Linhagem Celular Tumoral , Camundongos , MicroRNAs/genética , Neoplasias Hipofisárias/patologia , Transfecção
13.
FEBS Open Bio ; 9(3): 527-537, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30868061

RESUMO

Cancer stem cells contribute to cancer progression, but the mechanisms underlying neuroblastoma stem cell development are unclear. Here, we examined the roles of the transcription factor SLC34A2 in regulating the stemness of neuroblastoma cells. We found that SLC34A2 expression was negatively correlated with the overall survival and relapse-free survival probability of neuroblastoma patients. Additionally, SLC34A2 expression was observed to be remarkably increased in spheroids derived from neuroblastoma cells. Knockdown of SLC34A2 attenuated the expression of stemness markers and spheroid formation capacity of neuroblastoma cell-derived spheroids, and overexpression of SLC34A2 exerted the opposite effects in neuroblastoma cells. Mechanistically, SLC34A2 was found to directly bind to the promoter of MIR25, which targets glycogen synthesis kinase 3ß (Gsk3ß), an antagonist of Wnt signaling. Transfection of miR-25 inhibitor or a Gsk3ß overexpression plasmid attenuated the effects of SLC34A2 overexpression on the stemness of neuroblastoma cells. Our results demonstrate that miR-25/Gsk3ß-mediated activation of Wnt signaling is responsible for SLC34A2-induced enhancement of neuroblastoma cell stemness.


Assuntos
Glicogênio Sintase Quinase 3 beta/metabolismo , MicroRNAs/metabolismo , Neuroblastoma/metabolismo , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/metabolismo , beta Catenina/metabolismo , Humanos , Neuroblastoma/patologia , Proteínas Cotransportadoras de Sódio-Fosfato Tipo IIb/genética , Análise de Sobrevida , Células Tumorais Cultivadas , Via de Sinalização Wnt
14.
J Neurooncol ; 140(1): 15-26, 2018 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29916101

RESUMO

INTRODUCTION: Runt-related transcription factor 3 (RUNX3) exerts a tumor suppressor gene associated with gastric and other cancers, including glioma. However, how its anti-tumor mechanism in functions glioma is unclear. METHODS: We assayed expression of RUNX3 with a tissue microarray (TMA), frozen cancer tissues and malignant glioma cell lines using immunohistochemistry, qRT-PCR and Western bolt analysis. Cell proliferation, invasion, cell cycle distribution and apoptosis were also examined to confirm the effect of RUNX3 medicated malignant phenotype. TOP/FOP experiment was used to detect the ß-catenin/Tcf-4 transcription activity by RUNX3. RESULTS: Enforced RUNX3 expression inhibited proliferation and invasion, induced cell cycle arrest and promoted apoptosis in vitro and in vivo, Bim siRNA partically reversed the effect of RUNX3-induced apoptosis in LN229 and U87 cells, suggesting a dependent role of Bim-caspase pathway. Moreover, Mechanism investigations revealed that restoration of RUNX3 suppressed ß-catenin/Tcf-4 transcription activity. CONCLUSIONS: RUNX3 plays a pivotal role in glioma initiation and progression as a tumor suppressor via attenuation of Wnt signaling, highlighting it as a potential therapeutic target for glioma.


Assuntos
Neoplasias Encefálicas , Subunidade alfa 3 de Fator de Ligação ao Core/metabolismo , Glioma , Transdução de Sinais/fisiologia , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/patologia , Neoplasias Encefálicas/fisiopatologia , Regulação Neoplásica da Expressão Gênica , Glioma/metabolismo , Glioma/patologia , Glioma/fisiopatologia , Humanos , Invasividade Neoplásica/fisiopatologia , Análise Serial de Tecidos , Fator de Transcrição 4/metabolismo , beta Catenina/metabolismo
15.
World Neurosurg ; 95: 77-84, 2016 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-27485530

RESUMO

BACKGROUND: As the population ages, the proportion of elderly patients with glioblastomas has increased. Recently, many researchers have focused on the treatments available to and prognoses in elderly patients with glioblastomas. METHODS: We conducted a retrospective study of glioblastoma patients aged 60 years old or older who were treated at the Neurosurgery Center at Beijing Tiantan Hospital from 2012 to 2014. Their clinical features, immunohistochemical characteristics, treatments, and outcomes were evaluated to determine treatment tolerance and identify prognostic factors. RESULTS: Among the 70 included patients, the median survival time was 15 months. In the univariate analysis, patients who underwent a gross total resection had longer overall survival times than patients who had a subtotal resection (P < 0.05), and patients who received postoperative adjuvant therapy had longer overall survival times than those with no postoperative adjuvant therapy (P < 0.05). The expression of the p53 protein significantly affected overall survival. Patients with low p53 protein expression had a median survival of 17 months, whereas those who had high p53 protein expression had a median survival of 11.50 months (P < 0.05). Undergoing a gross total resection, receiving postoperative adjuvant therapy and having low p53 protein expression were factors that independently contributed to longer overall survival times in multivariate analysis. CONCLUSIONS: Maximal safe surgical resection followed by radiotherapy with concurrent and adjuvant temozolomide significantly prolonged overall survival times and was well tolerated in elderly patients with glioblastomas. In addition, low p53 protein expression was a significant favorable prognostic indicator in this population.


Assuntos
Antineoplásicos Alquilantes/uso terapêutico , Neoplasias Encefálicas/terapia , Quimiorradioterapia Adjuvante , Dacarbazina/análogos & derivados , Glioblastoma/terapia , Procedimentos Neurocirúrgicos , Idoso , Idoso de 80 Anos ou mais , Neoplasias Encefálicas/metabolismo , Neoplasias Encefálicas/mortalidade , Quimioterapia Adjuvante , Terapia Combinada , Dacarbazina/uso terapêutico , Feminino , Glioblastoma/metabolismo , Glioblastoma/mortalidade , Humanos , Avaliação de Estado de Karnofsky , Masculino , Pessoa de Meia-Idade , Análise Multivariada , Prognóstico , Modelos de Riscos Proporcionais , Radioterapia Adjuvante , Estudos Retrospectivos , Taxa de Sobrevida , Temozolomida , Proteína Supressora de Tumor p53/metabolismo
16.
World Neurosurg ; 94: 111-119, 2016 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-27392897

RESUMO

OBJECTIVE: To present our experience with microsurgical technique for patients with giant meningiomas (maximum diameter ≥7 cm) that obstruct the superior sagittal sinus (SSS). METHODS: All patients who were preoperatively diagnosed (between 2010 and 2014) with giant meningiomas involving the SSS in Ward 10 at the Neurosurgery Department of Beijing Tiantan Hospital were enrolled in this study. Patient charts, imaging findings, and outcomes were examined. RESULTS: The study included 6 male and 4 female patients with a mean age of 46.8 ± 10.7 years. The tumor sizes varied from 7 to 12 cm (mean, 8.8 ± 2.0 cm). All patients underwent customized craniotomies, and aggressive surgery for resection of the invaded SSS was performed. Simpson grade I removals were achieved in all cases. No cases of perioperative mortality occurred. Three patients required cranioplasty as a result of a decompressive craniectomy that was performed during the primary surgery. Histologic examinations showed 1 malignant and 9 benign meningiomas. During the follow-up period (mean, 29.0 ± 9.7 months), recurrence/progression occurred in 1 patient, and 1 patient was lost to follow-up. The recent Karnofsky Performance Score was 80 ± 32.3 and was improved in 5 patients and stabilized in 3 patients. In addition, 7 patients lived independently. CONCLUSIONS: The rigorous preservation of cortical veins, draining veins, and eloquent areas should be implemented during the resection of large tumors that obstruct the SSS. Suitable individualized approaches associated with full exposure and low cerebral perfusion pressure levels after surgery are critical for favorable results, and the reconstruction of the SSS may not be necessary.


Assuntos
Neoplasias Meníngeas/cirurgia , Meningioma/cirurgia , Procedimentos Neurocirúrgicos/métodos , Seio Sagital Superior/cirurgia , Adulto , Craniectomia Descompressiva , Epilepsia/epidemiologia , Estudos de Viabilidade , Feminino , Hematoma/epidemiologia , Humanos , Avaliação de Estado de Karnofsky , Masculino , Neoplasias Meníngeas/diagnóstico por imagem , Neoplasias Meníngeas/patologia , Meningioma/diagnóstico por imagem , Meningioma/patologia , Pessoa de Meia-Idade , Recidiva Local de Neoplasia/epidemiologia , Paresia/epidemiologia , Complicações Pós-Operatórias/epidemiologia , Procedimentos de Cirurgia Plástica , Estudos Retrospectivos , Crânio/cirurgia , Seio Sagital Superior/diagnóstico por imagem , Seio Sagital Superior/patologia , Resultado do Tratamento , Carga Tumoral
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA