Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 3 de 3
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Artigo em Inglês | MEDLINE | ID: mdl-39093499

RESUMO

PURPOSE: Automated glioblastoma segmentation from magnetic resonance imaging is generally performed on a four-modality input, including T1, contrast T1, T2 and FLAIR. We hypothesize that information redundancy is present within these image combinations, which can possibly reduce a model's performance. Moreover, for clinical applications, the risk of encountering missing data rises as the number of required input modalities increases. Therefore, this study aimed to explore the relevance and influence of the different modalities used for MRI-based glioblastoma segmentation. METHODS: After the training of multiple segmentation models based on nnU-Net and SwinUNETR architectures, differing only in their amount and combinations of input modalities, each model was evaluated with regard to segmentation accuracy and epistemic uncertainty. RESULTS: Results show that T1CE-based segmentation (for enhanced tumor and tumor core) and T1CE-FLAIR-based segmentation (for whole tumor and overall segmentation) can reach segmentation accuracies comparable to the full-input version. Notably, the highest segmentation accuracy for nnU-Net was found for a three-input configuration of T1CE-FLAIR-T1, suggesting the confounding effect of redundant input modalities. The SwinUNETR architecture appears to suffer less from this, where said three-input and the full-input model yielded statistically equal results. CONCLUSION: The T1CE-FLAIR-based model can therefore be considered as a minimal-input alternative to the full-input configuration. Addition of modalities beyond this does not statistically improve and can even deteriorate accuracy, but does lower the segmentation uncertainty.

2.
J Neurooncol ; 168(1): 159-169, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38502281

RESUMO

PURPOSE: After glioblastoma (GB) recurrence, prognosis is very cumbersome. Therefore, health-related quality of life (HRQoL) and neurocognitive functioning (NCF) have become important endpoints in clinical trials when evaluating novel treatments. We aimed to evaluate the HRQoL and NCF in patients with recurrent glioblastoma (rGB) treated with a combination of surgical intervention (reoperation or biopsy) and intracerebral immune checkpoint inhibition. METHODS: Patients who participated in the trial (N = 23), at a single-center university hospital were included. Data were collected using 3 patient-reported outcome measures (EORTC-QLQ-C30, EORTC-QLQ-BN20, and HADS) and computerized NCF testing. In the responder group, baseline values were compared to results at a 6-month follow-up. Additionally, exploratory analyses compared baseline HRQoL and NCF between responders and non-responders. RESULTS: There were five responders and 18 non-responders. When comparing the mean and individual baseline with follow-up results for the responders, we observed overall a stable to slight clinically relevant improvement of HRQoL in multiple subsets of the questionnaires while maintaining a stable NCF. One patient deteriorated on anxiety and depression symptoms from baseline to follow-up. CONCLUSIONS: In patients that responded to intracerebral immunotherapy in our institutional trial, HRQoL and NCF remained stable over time, suggesting that no detrimental effect on cognitive function or quality of life may be expected with this treatment approach. Furthermore, there seems to be an overall tendency for responders to score better on HRQoL and NCF than non-responders at baseline.


Assuntos
Neoplasias Encefálicas , Glioblastoma , Inibidores de Checkpoint Imunológico , Recidiva Local de Neoplasia , Qualidade de Vida , Humanos , Glioblastoma/psicologia , Glioblastoma/complicações , Glioblastoma/terapia , Masculino , Feminino , Neoplasias Encefálicas/complicações , Neoplasias Encefálicas/psicologia , Pessoa de Meia-Idade , Inibidores de Checkpoint Imunológico/uso terapêutico , Inibidores de Checkpoint Imunológico/efeitos adversos , Recidiva Local de Neoplasia/psicologia , Idoso , Adulto , Seguimentos , Prognóstico
3.
Case Rep Oncol ; 17(1): 10-16, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38179547

RESUMO

Introduction: Pleiomorphic xanthoastrocytoma (PXA) is considered a low-grade glioma with a favorable prognosis following surgical resection. We present a case report of a BRAFV600E mutant malignantly transformed and disseminated PXA that was successfully treated with BRAF-/MEK-targeted therapy (dabrafenib/trametinib). Case Presentation: At the age of 16 years, our patient underwent an initial subtotal resection of a right occipital PXA. Six months later, a reintervention for an asymptomatic tumor recurrence was performed and complete resection was achieved. The patient has been followed up by MRI for 14 years without arguments for recurrence but was lost to follow-up thereafter. At 38 years of age, he presented with a symptomatic local recurrence with extra-cerebral soft tissue extension, for which a third surgical resection was performed. Anatomopathological examination reported a grade 3 anaplastic PXA (aPXA); molecular analysis detected a BRAFV600E mutation. Three months later, before the initiation of radiotherapy, a local tumor recurrence was diagnosed, for which he underwent a fourth surgical resection. Radiotherapy was performed following the surgical debulking. One month after completion of radiotherapy, disease progression was documented including multiple sites of extracranial metastases (skeletal, lung, cervical lymph node, and subcutaneous metastases). Systemic treatment with a combination of BRAF-/MEK-inhibitors (dabrafenib/trametinib) was initiated and resulted in a rapid and deep tumor response (partial response according to RECISTv1.1) and absence of BRAFV600E mutant ctDNA in plasma at 6 weeks after treatment initiation. A near-complete metabolic remission was documented on [18F]FDG-PET/CT 3 months after starting systemic therapy. Conclusion: We present a rare case of malignant transformation and systemic dissemination of a BRAFV600E mutant PXA, occurring 20 years after the initial diagnosis. This case highlights the importance of long-term follow-up of patients diagnosed with these rare central nervous system tumors that initially are considered benign and also illustrates that BRAF/MEK inhibition can be an effective therapy for BRAFV600E mutated PXA, underscoring the importance of performing molecular genetic profiling of these tumors.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA