Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 682
Filter
1.
PLoS One ; 19(9): e0307825, 2024.
Article in English | MEDLINE | ID: mdl-39241003

ABSTRACT

Brain tumors pose significant global health concerns due to their high mortality rates and limited treatment options. These tumors, arising from abnormal cell growth within the brain, exhibits various sizes and shapes, making their manual detection from magnetic resonance imaging (MRI) scans a subjective and challenging task for healthcare professionals, hence necessitating automated solutions. This study investigates the potential of deep learning, specifically the DenseNet architecture, to automate brain tumor classification, aiming to enhance accuracy and generalizability for clinical applications. We utilized the Figshare brain tumor dataset, comprising 3,064 T1-weighted contrast-enhanced MRI images from 233 patients with three prevalent tumor types: meningioma, glioma, and pituitary tumor. Four pre-trained deep learning models-ResNet, EfficientNet, MobileNet, and DenseNet-were evaluated using transfer learning from ImageNet. DenseNet achieved the highest test set accuracy of 96%, outperforming ResNet (91%), EfficientNet (91%), and MobileNet (93%). Therefore, we focused on improving the performance of the DenseNet, while considering it as base model. To enhance the generalizability of the base DenseNet model, we implemented a fine-tuning approach with regularization techniques, including data augmentation, dropout, batch normalization, and global average pooling, coupled with hyperparameter optimization. This enhanced DenseNet model achieved an accuracy of 97.1%. Our findings demonstrate the effectiveness of DenseNet with transfer learning and fine-tuning for brain tumor classification, highlighting its potential to improve diagnostic accuracy and reliability in clinical settings.


Subject(s)
Brain Neoplasms , Deep Learning , Magnetic Resonance Imaging , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/classification , Magnetic Resonance Imaging/methods , Meningioma/diagnostic imaging , Meningioma/pathology , Glioma/diagnostic imaging , Glioma/pathology , Glioma/classification , Male , Female , Pituitary Neoplasms/diagnostic imaging , Pituitary Neoplasms/pathology , Pituitary Neoplasms/classification
2.
Chin Clin Oncol ; 13(Suppl 1): AB093, 2024 Aug.
Article in English | MEDLINE | ID: mdl-39295411

ABSTRACT

BACKGROUND: Central nervous system (CNS) tumours, especially glioma, are a complex disease and many challenges are encountered in their treatment. Artificial intelligence (AI) has made a colossal impact in many walks of life at a low cost. However, this avenue still needs to be explored in healthcare settings, demanding investment of resources towards growth in this area. We aim to develop machine learning (ML) algorithms to facilitate the accurate diagnosis and precise mapping of the brain tumour. METHODS: We queried the data from 2019 to 2022 and brain magnetic resonance imaging (MRI) of glioma patients were extracted. Images that had both T1-contrast and T2-fluid-attenuated inversion recovery (T2-FLAIR) volume sequences available were included. MRI images were annotated by a team supervised by a neuroradiologist. The extracted MRIs thus obtained were then fed to the preprocessing pipeline to extract brains using SynthStrip. They were further fed to the deep learning-based semantic segmentation pipelines using UNet-based architecture with convolutional neural network (CNN) at its backbone. Subsequently, the algorithm was tested to assess the efficacy in the pixel-wise diagnosis of tumours. RESULTS: In total, 69 samples of low-grade glioma (LGG) were used out of which 62 were used for fine-tuning a pre-trained model trained on brain tumor segmentation (BraTS) 2020 and 7 were used for testing. For the evaluation of the model, the Dice coefficient was used as the metric. The average Dice coefficient on the 7 test samples was 0.94. CONCLUSIONS: With the advent of technology, AI continues to modify our lifestyles. It is critical to adapt this technology in healthcare with the aim of improving the provision of patient care. We present our preliminary data for the use of ML algorithms in the diagnosis and segmentation of glioma. The promising result with comparable accuracy highlights the importance of early adaptation of this nascent technology.


Subject(s)
Deep Learning , Glioma , Magnetic Resonance Imaging , Humans , Glioma/classification , Glioma/pathology , Magnetic Resonance Imaging/methods , Male , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/classification , Brain Neoplasms/pathology , Female
3.
Int J Mol Sci ; 25(15)2024 Aug 03.
Article in English | MEDLINE | ID: mdl-39126064

ABSTRACT

Paediatric high-grade gliomas are among the most common malignancies found in children. Despite morphological similarities to their adult counterparts, there are profound biological and molecular differences. Furthermore, and thanks to molecular biology, the diagnostic pathology of paediatric high-grade gliomas has experimented a dramatic shift towards molecular classification, with important prognostic implications, as is appropriately reflected in both the current WHO Classification of Tumours of the Central Nervous System and the WHO Classification of Paediatric Tumours. Emphasis is placed on histone 3, IDH1, and IDH2 alterations, and on Receptor of Tyrosine Kinase fusions. In this review we present the current diagnostic categories from the diagnostic pathology perspective including molecular features.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/genetics , Glioma/pathology , Glioma/classification , Glioma/metabolism , Child , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/classification , Neoplasm Grading , Isocitrate Dehydrogenase/genetics , Histones/metabolism , Histones/genetics , Biomarkers, Tumor/genetics , Prognosis
4.
Bioinformatics ; 40(9)2024 09 02.
Article in English | MEDLINE | ID: mdl-39177104

ABSTRACT

MOTIVATION: Heterogeneity in human diseases presents challenges in diagnosis and treatments due to the broad range of manifestations and symptoms. With the rapid development of labelled multi-omic data, integrative machine learning methods have achieved breakthroughs in treatments by redefining these diseases at a more granular level. These approaches often have limitations in scalability, oversimplification, and handling of missing data. RESULTS: In this study, we introduce Multi-Omic Graph Diagnosis (MOGDx), a flexible command line tool for the integration of multi-omic data to perform classification tasks for heterogeneous diseases. MOGDx has a network taxonomy. It fuses patient similarity networks, augments this integrated network with a reduced vector representation of genomic data and performs classification using a graph convolutional network. MOGDx was evaluated on three datasets from the cancer genome atlas for breast invasive carcinoma, kidney cancer, and low grade glioma. MOGDx demonstrated state-of-the-art performance and an ability to identify relevant multi-omic markers in each task. It integrated more genomic measures with greater patient coverage compared to other network integrative methods. Overall, MOGDx is a promising tool for integrating multi-omic data, classifying heterogeneous diseases, and aiding interpretation of genomic marker data. AVAILABILITY AND IMPLEMENTATION: MOGDx source code is available from https://github.com/biomedicalinformaticsgroup/MOGDx.


Subject(s)
Genomics , Humans , Genomics/methods , Software , Breast Neoplasms , Neoplasms , Kidney Neoplasms/genetics , Kidney Neoplasms/classification , Machine Learning , Computational Biology/methods , Glioma/genetics , Glioma/classification , Multiomics
5.
Medicine (Baltimore) ; 103(34): e39316, 2024 Aug 23.
Article in English | MEDLINE | ID: mdl-39183405

ABSTRACT

This study aimed to investigate the function of disulfidptosis-associated long noncoding RNAs (DAlncRNAs) in low-grade gliomas (LGG) through bioinformatics analysis and construct a signature to predict the classification, prognosis, tumor microenvironment, and selection of immunotherapy and chemotherapy in LGG. Genomic, clinical, and mutational information of 526 patients with LGG was retrieved from The Cancer Genome Atlas repository. A nonnegative matrix factorization algorithm was applied to classify patients with LGG. Univariate, LASSO regression, and multivariate Cox regression analyses were performed to determine prognostic DAlncRNAs. Following the median risk score, we defined the sample as a high-risk (HR) or low-risk group. Finally, survival, receiver operating characteristic curve, risk curve, principal component, independent prognosis, risk difference, functional enrichment, tumor microenvironment, immune cell infiltration, mutation, and drug sensitivity analyses were performed. Patients were classified into C1 and C2 subtypes associated with disulfidptosis. Eight prognostic DAlncRNAs (AC003035.2, AC010157.2, AC010273.3, AC011444.3, AC092667.1, AL450270.1, AL645608.2, and LINC01571) were identified, and a prognostic signature of LGG was developed. The DAlncRNA-based signature was found to be an independent prognostic factor in patients with LGG, thereby constructing a nomogram. In addition, in the HR group, immune function was more active and the tumor mutation burden was higher. The patients were mainly composed of subtype C2, and their prognosis was worse. Immunotherapy and chemotherapy were predicted in the HR and low-risk groups, respectively. Our study, based on DAlncRNAs, highlights 2 disulfidptosis-associated LGG subtypes with different prognostic and immune characteristics and creates a novel disulfidptosis-associated prognostic signature, which may inform the classification, prognosis, molecular pathogenesis, and therapeutic strategies for patients with LGG.


Subject(s)
Brain Neoplasms , Glioma , RNA, Long Noncoding , Tumor Microenvironment , Humans , RNA, Long Noncoding/genetics , Glioma/genetics , Glioma/therapy , Glioma/pathology , Glioma/mortality , Glioma/classification , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology , Prognosis , Male , Brain Neoplasms/genetics , Brain Neoplasms/therapy , Brain Neoplasms/pathology , Female , Middle Aged , Biomarkers, Tumor/genetics , Immunotherapy/methods
6.
Lancet Oncol ; 25(9): e404-e419, 2024 Sep.
Article in English | MEDLINE | ID: mdl-39214112

ABSTRACT

Glioma resection is associated with prolonged survival, but neuro-oncological trials have frequently refrained from quantifying the extent of resection. The Response Assessment in Neuro-Oncology (RANO) resect group is an international, multidisciplinary group that aims to standardise research practice by delineating the oncological role of surgery in diffuse adult-type gliomas as defined per WHO 2021 classification. Favourable survival effects of more extensive resection unfold over months to decades depending on the molecular tumour profile. In tumours with a more aggressive natural history, supramaximal resection might correlate with additional survival benefit. Weighing the expected survival benefits of resection as dictated by molecular tumour profiles against clinical factors, including the introduction of neurological deficits, we propose an algorithm to estimate the oncological effects of surgery for newly diagnosed gliomas. The algorithm serves to select patients who might benefit most from extensive resection and to emphasise the relevance of quantifying the extent of resection in clinical trials.


Subject(s)
Brain Neoplasms , Glioma , World Health Organization , Humans , Glioma/surgery , Glioma/pathology , Glioma/classification , Glioma/mortality , Brain Neoplasms/surgery , Brain Neoplasms/pathology , Brain Neoplasms/classification , Brain Neoplasms/mortality , Algorithms , Adult , Neurosurgical Procedures/adverse effects , Treatment Outcome
7.
BMC Med Imaging ; 24(1): 177, 2024 Jul 19.
Article in English | MEDLINE | ID: mdl-39030508

ABSTRACT

BACKGROUND: Cancer pathology shows disease development and associated molecular features. It provides extensive phenotypic information that is cancer-predictive and has potential implications for planning treatment. Based on the exceptional performance of computational approaches in the field of digital pathogenic, the use of rich phenotypic information in digital pathology images has enabled us to identify low-level gliomas (LGG) from high-grade gliomas (HGG). Because the differences between the textures are so slight, utilizing just one feature or a small number of features produces poor categorization results. METHODS: In this work, multiple feature extraction methods that can extract distinct features from the texture of histopathology image data are used to compare the classification outcomes. The successful feature extraction algorithms GLCM, LBP, multi-LBGLCM, GLRLM, color moment features, and RSHD have been chosen in this paper. LBP and GLCM algorithms are combined to create LBGLCM. The LBGLCM feature extraction approach is extended in this study to multiple scales using an image pyramid, which is defined by sampling the image both in space and scale. The preprocessing stage is first used to enhance the contrast of the images and remove noise and illumination effects. The feature extraction stage is then carried out to extract several important features (texture and color) from histopathology images. Third, the feature fusion and reduction step is put into practice to decrease the number of features that are processed, reducing the computation time of the suggested system. The classification stage is created at the end to categorize various brain cancer grades. We performed our analysis on the 821 whole-slide pathology images from glioma patients in the Cancer Genome Atlas (TCGA) dataset. Two types of brain cancer are included in the dataset: GBM and LGG (grades II and III). 506 GBM images and 315 LGG images are included in our analysis, guaranteeing representation of various tumor grades and histopathological features. RESULTS: The fusion of textural and color characteristics was validated in the glioma patients using the 10-fold cross-validation technique with an accuracy equals to 95.8%, sensitivity equals to 96.4%, DSC equals to 96.7%, and specificity equals to 97.1%. The combination of the color and texture characteristics produced significantly better accuracy, which supported their synergistic significance in the predictive model. The result indicates that the textural characteristics can be an objective, accurate, and comprehensive glioma prediction when paired with conventional imagery. CONCLUSION: The results outperform current approaches for identifying LGG from HGG and provide competitive performance in classifying four categories of glioma in the literature. The proposed model can help stratify patients in clinical studies, choose patients for targeted therapy, and customize specific treatment schedules.


Subject(s)
Algorithms , Brain Neoplasms , Color , Glioma , Neoplasm Grading , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Brain Neoplasms/classification , Glioma/diagnostic imaging , Glioma/pathology , Glioma/classification , Diagnosis, Computer-Assisted/methods , Image Interpretation, Computer-Assisted/methods
8.
Clin Cancer Res ; 30(17): 3824-3836, 2024 Sep 03.
Article in English | MEDLINE | ID: mdl-38976016

ABSTRACT

PURPOSE: Recent artificial intelligence algorithms aided intraoperative decision-making via stimulated Raman histology (SRH) during craniotomy. This study assesses deep learning algorithms for rapid intraoperative diagnosis from SRH images in small stereotactic-guided brain biopsies. It defines a minimum tissue sample size threshold to ensure diagnostic accuracy. EXPERIMENTAL DESIGN: A prospective single-center study examined 121 SRH images from 84 patients with unclear intracranial lesions undergoing stereotactic brain biopsy. Unprocessed, label-free samples were imaged using a portable fiber laser Raman scattering microscope. Three deep learning models were tested to (i) identify tumorous/nontumorous tissue as qualitative biopsy control; (ii) subclassify into high-grade glioma (central nervous system World Health Organization grade 4), diffuse low-grade glioma (central nervous system World Health Organization grades 2-3), metastases, lymphoma, or gliosis; and (iii) molecularly subtype IDH and 1p/19q statuses of adult-type diffuse gliomas. Model predictions were evaluated against frozen section analysis and final neuropathologic diagnoses. RESULTS: The first model identified tumorous/nontumorous tissue with 91.7% accuracy. Sample size on slides impacted accuracy in brain tumor subclassification (81.6%, κ = 0.72 frozen section; 73.9%, κ = 0.61 second model), with SRH images being smaller than hematoxylin and eosin images (4.1 ± 2.5 mm2 vs. 16.7 ± 8.2 mm2, P < 0.001). SRH images with more than 140 high-quality patches and a mean squeezed sample of 5.26 mm2 yielded 89.5% accuracy in subclassification and 93.9% in molecular subtyping of adult-type diffuse gliomas. CONCLUSIONS: Artificial intelligence-based SRH image analysis is non-inferior to frozen section analysis in detecting and subclassifying brain tumors during small stereotactic-guided biopsies once a critical squeezed sample size is reached. Beyond frozen section analysis, it enables valid molecular glioma subtyping, allowing faster treatment decisions in the future; however, refinement is needed for long-term application.


Subject(s)
Brain Neoplasms , Deep Learning , Glioma , Spectrum Analysis, Raman , Humans , Brain Neoplasms/pathology , Brain Neoplasms/classification , Brain Neoplasms/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/surgery , Spectrum Analysis, Raman/methods , Male , Female , Middle Aged , Glioma/pathology , Glioma/classification , Glioma/genetics , Glioma/surgery , Glioma/diagnosis , Aged , Adult , Prospective Studies , Stereotaxic Techniques , Biopsy , Neoplasm Grading , Algorithms
9.
J Neurooncol ; 169(2): 287-297, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38874844

ABSTRACT

PURPOSE: To evaluate the performance of multi-pool Chemical exchange saturation transfer (CEST) MRI in prediction of glioma grade, isocitrate dehydrogenase (IDH) mutation, alpha-thalassemia/mental retardation syndrome X-linked (ATRX) loss and Ki-67 labeling index (LI), based on the fifth edition of the World Health Organization classification of central nervous system tumors (WHO CNS5). METHODS: 95 patients with adult-type diffuse gliomas were analyzed. The amide, direct water saturation (DS), nuclear Overhauser enhancement (NOE), semi-solid magnetization transfer (MT) and amine signals were derived using Lorentzian fitting, and asymmetry-based amide proton transfer-weighted (APTwasym) signal was calculated. The mean value of tumor region was measured and intergroup differences were estimated using student-t test. The receiver operating curve (ROC) and area under the curve (AUC) analysis were used to evaluate the diagnostic performance of signals and their combinations. Spearman correlation analysis was performed to evaluate tumor proliferation. RESULTS: The amide and DS signals were significantly higher in high-grade gliomas compared to low-grade gliomas, as well as in IDH-wildtype gliomas compared to IDH-mutant gliomas (all p < 0.001). The DS, MT and amine signals showed significantly differences between ATRX loss and retention in grade 2/3 IDH-mutant gliomas (all p < 0.05). The combination of signals showed the highest AUC in prediction of grade (0.857), IDH mutation (0.814) and ATRX loss (0.769). Additionally, the amide and DS signals were positively correlated with Ki-67 LI (both p < 0.001). CONCLUSION: Multi-pool CEST MRI demonstrated good potential to predict glioma grade, IDH mutation, ATRX loss and Ki-67 LI.


Subject(s)
Brain Neoplasms , Glioma , Isocitrate Dehydrogenase , Magnetic Resonance Imaging , Mutation , Neoplasm Grading , Humans , Glioma/genetics , Glioma/diagnostic imaging , Glioma/metabolism , Glioma/pathology , Glioma/classification , Male , Female , Middle Aged , Adult , Brain Neoplasms/genetics , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , Magnetic Resonance Imaging/methods , Isocitrate Dehydrogenase/genetics , Aged , Young Adult , Cell Proliferation , X-linked Nuclear Protein/genetics , X-linked Nuclear Protein/metabolism , Ki-67 Antigen/metabolism
10.
Cancer Med ; 13(11): e7377, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38850123

ABSTRACT

OBJECTIVE: The study aimed to identify if clinical features and survival outcomes of insular glioma patients are associated with our classification based on the tumor spread. METHODS: Our study included 283 consecutive patients diagnosed with histological grade 2 and 3 insular gliomas. A new classification was proposed, and tumors restricted to the paralimbic system were defined as type 1. When tumors invaded the limbic system (referred to as the hippocampus and its surrounding structures in this study) simultaneously, they were defined as type 2. Tumors with additional internal capsule involvement were defined as type 3. RESULTS: Tumors defined as type 3 had a higher age at diagnosis (p = 0.002) and a higher preoperative volume (p < 0.001). Furthermore, type 3 was more likely to be diagnosed as IDH wild type (p < 0.001), with a higher rate of Ki-67 index (p = 0.015) and a lower rate of gross total resection (p < 0.001). Type 1 had a slower tumor growth rate than type 2 (mean 3.3%/month vs. 19.8%/month; p < 0.001). Multivariate Cox regression analysis revealed the extent of resection (HR 0.259, p = 0.004), IDH status (HR 3.694, p = 0.012), and tumor spread type (HR = 1.874, p = 0.012) as independent predictors of overall survival (OS). Tumor grade (HR 2.609, p = 0.008), the extent of resection (HR 0.488, p = 0.038), IDH status (HR 2.225, p = 0.025), and tumor spread type (HR 1.531, p = 0.038) were significant in predicting progression-free survival (PFS). CONCLUSION: The current study proposes a classification of the insular glioma according to the tumor spread. It indicates that the tumors defined as type 1 have a relatively better nature and biological characteristics, and those defined as type 3 can be more aggressive and refractory. Besides its predictive value for prognosis, the classification has potential value in formulating surgical strategies for patients with insular gliomas.


Subject(s)
Brain Neoplasms , Glioma , Neoplasm Grading , Humans , Glioma/pathology , Glioma/mortality , Glioma/classification , Glioma/surgery , Male , Female , Middle Aged , Brain Neoplasms/pathology , Brain Neoplasms/mortality , Brain Neoplasms/classification , Adult , Aged , Prognosis , Isocitrate Dehydrogenase/genetics , Retrospective Studies , Young Adult , World Health Organization
11.
Sensors (Basel) ; 24(12)2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38931588

ABSTRACT

This study describes a novel method for grading pathological sections of gliomas. Our own integrated hyperspectral imaging system was employed to characterize 270 bands of cancerous tissue samples from microarray slides of gliomas. These samples were then classified according to the guidelines developed by the World Health Organization, which define the subtypes and grades of diffuse gliomas. We explored a hyperspectral feature extraction model called SMLMER-ResNet using microscopic hyperspectral images of brain gliomas of different malignancy grades. The model combines the channel attention mechanism and multi-scale image features to automatically learn the pathological organization of gliomas and obtain hierarchical feature representations, effectively removing the interference of redundant information. It also completes multi-modal, multi-scale spatial-spectral feature extraction to improve the automatic classification of glioma subtypes. The proposed classification method demonstrated high average classification accuracy (>97.3%) and a Kappa coefficient (0.954), indicating its effectiveness in improving the automatic classification of hyperspectral gliomas. The method is readily applicable in a wide range of clinical settings, offering valuable assistance in alleviating the workload of clinical pathologists. Furthermore, the study contributes to the development of more personalized and refined treatment plans, as well as subsequent follow-up and treatment adjustment, by providing physicians with insights into the underlying pathological organization of gliomas.


Subject(s)
Brain Neoplasms , Glioma , Neoplasm Grading , Glioma/pathology , Glioma/classification , Humans , Brain Neoplasms/pathology , Brain Neoplasms/classification , Neoplasm Grading/methods , Hyperspectral Imaging/methods , Algorithms , Image Processing, Computer-Assisted/methods
12.
Math Biosci Eng ; 21(4): 5250-5282, 2024 Mar 06.
Article in English | MEDLINE | ID: mdl-38872535

ABSTRACT

The increasing global incidence of glioma tumors has raised significant healthcare concerns due to their high mortality rates. Traditionally, tumor diagnosis relies on visual analysis of medical imaging and invasive biopsies for precise grading. As an alternative, computer-assisted methods, particularly deep convolutional neural networks (DCNNs), have gained traction. This research paper explores the recent advancements in DCNNs for glioma grading using brain magnetic resonance images (MRIs) from 2015 to 2023. The study evaluated various DCNN architectures and their performance, revealing remarkable results with models such as hybrid and ensemble based DCNNs achieving accuracy levels of up to 98.91%. However, challenges persisted in the form of limited datasets, lack of external validation, and variations in grading formulations across diverse literature sources. Addressing these challenges through expanding datasets, conducting external validation, and standardizing grading formulations can enhance the performance and reliability of DCNNs in glioma grading, thereby advancing brain tumor classification and extending its applications to other neurological disorders.


Subject(s)
Brain Neoplasms , Deep Learning , Glioma , Magnetic Resonance Imaging , Neoplasm Grading , Neural Networks, Computer , Humans , Glioma/diagnostic imaging , Glioma/pathology , Glioma/classification , Magnetic Resonance Imaging/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Reproducibility of Results , Algorithms , Brain/diagnostic imaging , Brain/pathology , Image Processing, Computer-Assisted/methods , Image Interpretation, Computer-Assisted/methods
13.
J Neurol Sci ; 461: 123058, 2024 Jun 15.
Article in English | MEDLINE | ID: mdl-38781807

ABSTRACT

The World Health Organization (WHO) published the 5th edition classification of tumors of central nervous system in 2021, commonly abbreviated as WHO CNS5, which became the new standard for brain tumor diagnosis and therapy. This edition dramatically impacted tumor diagnostics. In short it introduced new tumors, changed the names of previously recognized tumors, and modified the working definition of previously known tumors. The new system appears complex due to the integration of morphological and multiple molecular criteria. The most radical changes occurred in the field of glial and glioneuronal tumors, which constitutes the lengthy first chapter of this new edition. Herein we present an illustrative outline of the evolving concepts of glial and glioneuronal tumors. We also attempt to explain the rationales behind this substantial change in tumor classification and the challenges to update and integrate it into clinical practice. We aim to present a concise and precise roadmap to aid navigation through the intricate conceptual framework of glial and glioneuronal tumors in the context of WHO CNS5.


Subject(s)
Brain Neoplasms , Glioma , Humans , Glioma/classification , Glioma/pathology , Glioma/diagnostic imaging , Glioma/diagnosis , Brain Neoplasms/classification , Brain Neoplasms/pathology , Brain Neoplasms/diagnostic imaging , World Health Organization
14.
Sci Rep ; 14(1): 11977, 2024 05 25.
Article in English | MEDLINE | ID: mdl-38796531

ABSTRACT

The preoperative diagnosis of brain tumors is important for therapeutic planning as it contributes to the tumors' prognosis. In the last few years, the development in the field of artificial intelligence and machine learning has contributed greatly to the medical area, especially the diagnosis of the grades of brain tumors through radiological images and magnetic resonance images. Due to the complexity of tumor descriptors in medical images, assessing the accurate grade of glioma is a major challenge for physicians. We have proposed a new classification system for glioma grading by integrating novel MRI features with an ensemble learning method, called Ensemble Learning based on Adaptive Power Mean Combiner (EL-APMC). We evaluate and compare the performance of the EL-APMC algorithm with twenty-one classifier models that represent state-of-the-art machine learning algorithms. Results show that the EL-APMC algorithm achieved the best performance in terms of classification accuracy (88.73%) and F1-score (93.12%) over the MRI Brain Tumor dataset called BRATS2015. In addition, we showed that the differences in classification results among twenty-two classifier models have statistical significance. We believe that the EL-APMC algorithm is an effective method for the classification in case of small-size datasets, which are common cases in medical fields. The proposed method provides an effective system for the classification of glioma with high reliability and accurate clinical findings.


Subject(s)
Algorithms , Brain Neoplasms , Glioma , Machine Learning , Magnetic Resonance Imaging , Neoplasm Grading , Humans , Glioma/diagnostic imaging , Glioma/classification , Glioma/pathology , Magnetic Resonance Imaging/methods , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/classification , Brain Neoplasms/pathology
15.
Interdiscip Sci ; 16(3): 727-740, 2024 Sep.
Article in English | MEDLINE | ID: mdl-38637440

ABSTRACT

Gliomas are highly heterogeneous in molecular, histology, and microenvironment. However, a classification of gliomas by integrating different tumor microenvironment (TME) components remains unexplored. Based on the enrichment scores of 17 pathways involved in immune, stromal, DNA repair, and nervous system signatures in diffuse gliomas, we performed consensus clustering to uncover novel subtypes of gliomas. Consistently in three glioma datasets (TCGA-glioma, CGGA325, and CGGA301), we identified three subtypes: Stromal-enriched (Str-G), Nerve-enriched (Ner-G), and mixed (Mix-G). Ner-G was charactered by low immune infiltration levels, stromal contents, tumor mutation burden, copy number alterations, DNA repair activity, cell proliferation, epithelial-mesenchymal transformation, stemness, intratumor heterogeneity, androgen receptor expression and EGFR, PTEN, NF1 and MUC16 mutation rates, while high enrichment of neurons and nervous system pathways, and high tumor purity, estrogen receptor expression, IDH1 and CIC mutation rates, temozolomide response rate and overall and disease-free survival rates. In contrast, Str-G displayed contrastive characteristics to Ner-G. Our analysis indicates that the heterogeneity between glioma cells and neurons is lower than that between glioma cells and immune and stromal cells. Furthermore, the abundance of neurons is positively associated with clinical outcomes in gliomas, while the enrichment of immune and stromal cells has a negative association with them. Our classification method provides new insights into the tumor biology of gliomas, as well as clinical implications for the precise management of this disease.


Subject(s)
Glioma , Tumor Microenvironment , Glioma/genetics , Glioma/pathology , Glioma/metabolism , Glioma/classification , Humans , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/metabolism , Mutation
16.
Clin Cancer Res ; 30(14): 2860-2861, 2024 Jul 15.
Article in English | MEDLINE | ID: mdl-38652677

ABSTRACT

The diagnosis and classification of glioma by liquid biopsy represent a critical unmet need in neuro-oncology. A recent study demonstrates targeted next-generation sequencing of cell-free DNA from cerebrospinal fluid as an evolving option for liquid biopsy in patients with glioma. See related article by Iser et al., p. 2974.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Cell-Free Nucleic Acids , Glioma , High-Throughput Nucleotide Sequencing , Humans , Liquid Biopsy/methods , Glioma/genetics , Glioma/diagnosis , Glioma/pathology , Glioma/classification , Biomarkers, Tumor/genetics , High-Throughput Nucleotide Sequencing/methods , Brain Neoplasms/genetics , Brain Neoplasms/diagnosis , Brain Neoplasms/pathology , Brain Neoplasms/classification , Circulating Tumor DNA/genetics
17.
J Cancer Res Clin Oncol ; 150(4): 220, 2024 Apr 29.
Article in English | MEDLINE | ID: mdl-38684578

ABSTRACT

PURPOSE: The purpose of this study is to develop accurate and automated detection and segmentation methods for brain tumors, given their significant fatality rates, with aggressive malignant tumors like Glioblastoma Multiforme (GBM) having a five-year survival rate as low as 5 to 10%. This underscores the urgent need to improve diagnosis and treatment outcomes through innovative approaches in medical imaging and deep learning techniques. METHODS: In this work, we propose a novel approach utilizing the two-headed UNetEfficientNets model for simultaneous segmentation and classification of brain tumors from Magnetic Resonance Imaging (MRI) images. The model combines the strengths of EfficientNets and a modified two-headed Unet model. We utilized a publicly available dataset consisting of 3064 brain MR images classified into three tumor classes: Meningioma, Glioma, and Pituitary. To enhance the training process, we performed 12 types of data augmentation on the training dataset. We evaluated the methodology using six deep learning models, ranging from UNetEfficientNet-B0 to UNetEfficientNet-B5, optimizing the segmentation and classification heads using binary cross entropy (BCE) loss with Dice and BCE with focal loss, respectively. Post-processing techniques such as connected component labeling (CCL) and ensemble models were applied to improve segmentation outcomes. RESULTS: The proposed UNetEfficientNet-B4 model achieved outstanding results, with an accuracy of 99.4% after postprocessing. Additionally, it obtained high scores for DICE (94.03%), precision (98.67%), and recall (99.00%) after post-processing. The ensemble technique further improved segmentation performance, with a global DICE score of 95.70% and Jaccard index of 91.20%. CONCLUSION: Our study demonstrates the high efficiency and accuracy of the proposed UNetEfficientNet-B4 model in the automatic and parallel detection and segmentation of brain tumors from MRI images. This approach holds promise for improving diagnosis and treatment planning for patients with brain tumors, potentially leading to better outcomes and prognosis.


Subject(s)
Brain Neoplasms , Deep Learning , Magnetic Resonance Imaging , Humans , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/classification , Brain Neoplasms/pathology , Magnetic Resonance Imaging/methods , Image Processing, Computer-Assisted/methods , Glioblastoma/diagnostic imaging , Glioblastoma/classification , Glioblastoma/pathology , Glioma/diagnostic imaging , Glioma/classification , Glioma/pathology
18.
Comput Biol Med ; 174: 108404, 2024 May.
Article in English | MEDLINE | ID: mdl-38582000

ABSTRACT

BACKGROUND: Glioma is a common and aggressive primary malignant cancer known for its high morbidity, mortality, and recurrence rates. Despite this, treatment options for glioma are currently restricted. The dysregulation of RBPs has been linked to the advancement of several types of cancer, but their precise role in glioma evolution is still not fully understood. This study sought to investigate how RBPs may impact the development and prognosis of glioma, with potential implications for prognosis and therapy. METHODS: RNA-seq profiles of glioma and corresponding clinical data from the CGGA database were initially collected for analysis. Unsupervised clustering was utilized to identify crucial tumor subtypes in glioma development. Subsequent time-series analysis and MS model were employed to track the progression of these identified subtypes. RBPs playing a significant role in glioma progression were then pinpointed using WGCNA and Lasso Cox regression models. Functional analysis of these key RBP-related genes was conducted through GSEA. Additionally, the CIBERSORT algorithm was utilized to estimate immune infiltrating cells, while the STRING database was consulted to uncover potential mechanisms of the identified biomarkers. RESULTS: Six tumor subgroups were identified and found to be highly homogeneous within each subgroup. The progression stages of these tumor subgroups were determined using time-series analysis and a MS model. Through WGCNA, Lasso Cox, and multivariate Cox regression analysis, it was confirmed that BCLAF1 is correlated with survival in glioma patients and is closely linked to glioma progression. Functional annotation suggests that BCLAF1 may impact glioma progression by influencing RNA splicing, which in turn affects the cell cycle, Wnt signaling pathway, and other cancer development pathways. CONCLUSIONS: The study initially identified six subtypes of glioma progression and assessed their malignancy ranking. Furthermore, it was determined that BCLAF1 could serve as an RBP-related prognostic marker, offering significant implications for the clinical diagnosis and personalized treatment of glioma.


Subject(s)
Biomarkers, Tumor , Brain Neoplasms , Glioma , RNA-Binding Proteins , Glioma/genetics , Glioma/classification , Glioma/metabolism , Humans , Brain Neoplasms/genetics , Brain Neoplasms/classification , Brain Neoplasms/metabolism , RNA-Binding Proteins/genetics , RNA-Binding Proteins/metabolism , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Databases, Genetic , Gene Expression Regulation, Neoplastic
19.
World Neurosurg ; 186: 204-218.e2, 2024 06.
Article in English | MEDLINE | ID: mdl-38580093

ABSTRACT

BACKGROUND: Classifying brain tumors accurately is crucial for treatment and prognosis. Machine learning (ML) shows great promise in improving tumor classification accuracy. This study evaluates ML algorithms for differentiating various brain tumor types. METHODS: A systematic review and meta-analysis were conducted, searching PubMed, Embase, and Web of Science up to March 14, 2023. Studies that only investigated image segmentation accuracy or brain tumor detection instead of classification were excluded. We extracted binary diagnostic accuracy data, constructing contingency tables to derive sensitivity and specificity. RESULTS: Fifty-one studies were included. The pooled area under the curve for glioblastoma versus lymphoma and low-grade versus high-grade gliomas were 0.99 (95% confidence interval [CI]: 0.98-1.00) and 0.89, respectively. The pooled sensitivity and specificity for benign versus malignant tumors were 0.90 (95% CI: 0.85-0.93) and 0.93 (95% CI: 0.90-0.95), respectively. The pooled sensitivity and specificity for low-grade versus high-grade gliomas were 0.99 (95% CI: 0.97-1.00) and 0.94, (95% CI: 0.79-0.99), respectively. Primary versus metastatic tumor identification yields sensitivity and specificity of 0.89, (95% CI: 0.83-0.93) and 0.87 (95% CI: 0.82-0.91), correspondingly. The differentiation of gliomas from pituitary tumors yielded the highest results among primary brain tumor classifications: sensitivity of 0.99 (95% CI: 0.99-1.00) and specificity of 0.99 (95% CI: 0.98-1.00). CONCLUSIONS: ML demonstrated excellent performance in classifying brain tumor images, with near-maximum area under the curves, sensitivity, and specificity.


Subject(s)
Brain Neoplasms , Machine Learning , Humans , Brain Neoplasms/classification , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/pathology , Glioblastoma/classification , Glioblastoma/diagnostic imaging , Glioblastoma/pathology , Glioma/classification , Glioma/diagnostic imaging , Glioma/pathology , Sensitivity and Specificity
20.
Eur Radiol ; 34(10): 6751-6762, 2024 Oct.
Article in English | MEDLINE | ID: mdl-38528135

ABSTRACT

OBJECTIVES: To distinguish isocitrate dehydrogenase (IDH) genotypes and tumor subtypes of adult-type diffuse gliomas based on the fifth edition of the World Health Organization classification of central nervous system tumors (WHO CNS5) in 2021 using standard, high, and ultra-high b-value diffusion-weighted imaging (DWI). MATERIALS AND METHODS: This prospective study enrolled 70 patients with adult-type diffuse gliomas who underwent multiple b-value DWI. Apparent diffusion coefficient (ADC) values including ADCb500/b1000, ADCb500/b2000, ADCb500/b3000, ADCb500/b4000, ADCb500/b6000, ADCb500/b8000, and ADCb500/b10000 in tumor parenchyma (TP) and contralateral normal-appearing white matter (NAWM) were calculated. The ADC ratios of TP/NAWM were assessed for correlations with IDH genotypes, tumor subtypes, and Ki-67 status; diagnostic performances were compared. RESULTS: All ADCs were significantly higher in IDH mutant gliomas than in IDH wild-type gliomas (p < 0.01 for all); ADCb500/b8000 had the highest area under the curve (AUC) of 0.866. All ADCs were significantly lower in glioblastoma than in astrocytoma (p < 0.01 for all). ADCs other than ADCb500/b1000 were significantly lower in glioblastoma than in oligodendroglioma (p < 0.05 for all). ADCb500/b8000 and ADCb500/b10000 were significantly higher in oligodendroglioma than in astrocytoma (p = 0.034 and 0.023). The highest AUCs were 0.818 for ADCb500/b6000 when distinguishing glioblastoma from astrocytoma, 0.979 for ADCb500/b8000 and ADCb500/b10000 when distinguishing glioblastoma from oligodendroglioma, and 0.773 for ADCb500/b10000 when distinguishing astrocytoma from oligodendroglioma. Additionally, all ADCs were negatively correlated with Ki-67 status (p < 0.05 for all). CONCLUSION: Ultra-high b-value DWI can reliably separate IDH genotypes and tumor subtypes of adult-type diffuse gliomas using WHO CNS5 criteria. CLINICAL RELEVANCE STATEMENT: Ultra-high b-value diffusion-weighted imaging can accurately distinguish isocitrate dehydrogenase genotypes and tumor subtypes of adult-type diffuse gliomas, which may facilitate personalized treatment and prognostic assessment for patients with glioma. KEY POINTS: • Ultra-high b-value diffusion-weighted imaging can accurately distinguish subtle differences in water diffusion among biological tissues. • Ultra-high b-value diffusion-weighted imaging can reliably separate isocitrate dehydrogenase genotypes and tumor subtypes of adult-type diffuse gliomas. • Compared with standard b-value diffusion-weighted imaging, high and ultra-high b-value diffusion-weighted imaging demonstrate better diagnostic performances.


Subject(s)
Brain Neoplasms , Diffusion Magnetic Resonance Imaging , Glioma , Adult , Aged , Female , Humans , Male , Middle Aged , Young Adult , Brain Neoplasms/diagnostic imaging , Brain Neoplasms/genetics , Diffusion Magnetic Resonance Imaging/methods , Genotype , Glioma/diagnostic imaging , Glioma/genetics , Glioma/classification , Isocitrate Dehydrogenase/genetics , Prospective Studies
SELECTION OF CITATIONS
SEARCH DETAIL