Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 59
Filtrar
1.
Radiol Med ; 2024 May 09.
Artigo em Inglês | MEDLINE | ID: mdl-38724697

RESUMO

PURPOSE: To investigate the feasibility of an artificial intelligence (AI)-based semi-automated segmentation for the extraction of ultrasound (US)-derived radiomics features in the characterization of focal breast lesions (FBLs). MATERIAL AND METHODS: Two expert radiologists classified according to US BI-RADS criteria 352 FBLs detected in 352 patients (237 at Center A and 115 at Center B). An AI-based semi-automated segmentation was used to build a machine learning (ML) model on the basis of B-mode US of 237 images (center A) and then validated on an external cohort of B-mode US images of 115 patients (Center B). RESULTS: A total of 202 of 352 (57.4%) FBLs were benign, and 150 of 352 (42.6%) were malignant. The AI-based semi-automated segmentation achieved a success rate of 95.7% for one reviewer and 96% for the other, without significant difference (p = 0.839). A total of 15 (4.3%) and 14 (4%) of 352 semi-automated segmentations were not accepted due to posterior acoustic shadowing at B-Mode US and 13 and 10 of them corresponded to malignant lesions, respectively. In the validation cohort, the characterization made by the expert radiologist yielded values of sensitivity, specificity, PPV and NPV of 0.933, 0.9, 0.857, 0.955, respectively. The ML model obtained values of sensitivity, specificity, PPV and NPV of 0.544, 0.6, 0.416, 0.628, respectively. The combined assessment of radiologists and ML model yielded values of sensitivity, specificity, PPV and NPV of 0.756, 0.928, 0.872, 0.855, respectively. CONCLUSION: AI-based semi-automated segmentation is feasible, allowing an instantaneous and reproducible extraction of US-derived radiomics features of FBLs. The combination of radiomics and US BI-RADS classification led to a potential decrease of unnecessary biopsy but at the expense of a not negligible increase of potentially missed cancers.

2.
Br J Radiol ; 2024 May 07.
Artigo em Inglês | MEDLINE | ID: mdl-38711198

RESUMO

OBJECTIVES: To assess how radiomic features may be combined with plaque morphological and compositional features identified by multi-contrast magnetic resonance imaging (MRI) to improve upon conventional risk assessment models in determining culprit lesions. METHODS: Fifty-five patients (mean age: 62.6; 35 males) with bilateral carotid stenosis who experienced transient ischaemic attack (TIA) or stroke were included from the CARE-II multi-centre carotid imaging trial (ClinicalTrials.gov Identifier: NCT02017756). They underwent MRI within 2 weeks of the event. Classification capability in distinguishing culprit lesions was assessed by machine learning. Repeatability and reproducibility of the results were investigated by assessing the robustness of the radiomic features. RESULTS: Radiomics combined with a relatively conventional plaque morphological and compositional metric-based model provided incremental value over a conventional model alone [area under curve (AUC), 0.819 ± 0.002 vs. 0.689 ± 0.019 respectively, p = 0.014]. The radiomic model alone also provided value over the conventional model [AUC, 0.805 ± 0.003 vs. 0.689 ± 0.019 respectively, p = 0.031]. T2-weighted imaging-based radiomic features had consistently higher robustness and classification capabilities compared with T1-weighted images. Higher-dimensional radiomic features outperformed first-order features. Grey Level Co-occurrence Matrix (GLCM), Grey Level Dependence Matrix (GLDM) and Grey Level Size Zone Matrix (GLSZM) sub-types were particularly useful in identifying textures which could detect vulnerable lesions. CONCLUSIONS: The combination of MRI-based radiomic features and lesion morphological and compositional parameters provided added value to the reference-standard risk assessment for carotid atherosclerosis. This may improve future risk stratification for individuals at risk of major adverse ischemic cerebrovascular events.

3.
Diagnostics (Basel) ; 14(10)2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38786294

RESUMO

Deep learning (DL) networks have shown attractive performance in medical image processing tasks such as brain tumor classification. However, they are often criticized as mysterious "black boxes". The opaqueness of the model and the reasoning process make it difficult for health workers to decide whether to trust the prediction outcomes. In this study, we develop an interpretable multi-part attention network (IMPA-Net) for brain tumor classification to enhance the interpretability and trustworthiness of classification outcomes. The proposed model not only predicts the tumor grade but also provides a global explanation for the model interpretability and a local explanation as justification for the proffered prediction. Global explanation is represented as a group of feature patterns that the model learns to distinguish high-grade glioma (HGG) and low-grade glioma (LGG) classes. Local explanation interprets the reasoning process of an individual prediction by calculating the similarity between the prototypical parts of the image and a group of pre-learned task-related features. Experiments conducted on the BraTS2017 dataset demonstrate that IMPA-Net is a verifiable model for the classification task. A percentage of 86% of feature patterns were assessed by two radiologists to be valid for representing task-relevant medical features. The model shows a classification accuracy of 92.12%, of which 81.17% were evaluated as trustworthy based on local explanations. Our interpretable model is a trustworthy model that can be used for decision aids for glioma classification. Compared with black-box CNNs, it allows health workers and patients to understand the reasoning process and trust the prediction outcomes.

5.
Eur Radiol Exp ; 7(1): 77, 2023 12 07.
Artigo em Inglês | MEDLINE | ID: mdl-38057616

RESUMO

PURPOSE: To determine if pelvic/ovarian and omental lesions of ovarian cancer can be reliably segmented on computed tomography (CT) using fully automated deep learning-based methods. METHODS: A deep learning model for the two most common disease sites of high-grade serous ovarian cancer lesions (pelvis/ovaries and omentum) was developed and compared against the well-established "no-new-Net" framework and unrevised trainee radiologist segmentations. A total of 451 CT scans collected from four different institutions were used for training (n = 276), evaluation (n = 104) and testing (n = 71) of the methods. The performance was evaluated using the Dice similarity coefficient (DSC) and compared using a Wilcoxon test. RESULTS: Our model outperformed no-new-Net for the pelvic/ovarian lesions in cross-validation, on the evaluation and test set by a significant margin (p values being 4 × 10-7, 3 × 10-4, 4 × 10-2, respectively), and for the omental lesions on the evaluation set (p = 1 × 10-3). Our model did not perform significantly differently in segmenting pelvic/ovarian lesions (p = 0.371) compared to a trainee radiologist. On an independent test set, the model achieved a DSC performance of 71 ± 20 (mean ± standard deviation) for pelvic/ovarian and 61 ± 24 for omental lesions. CONCLUSION: Automated ovarian cancer segmentation on CT scans using deep neural networks is feasible and achieves performance close to a trainee-level radiologist for pelvic/ovarian lesions. RELEVANCE STATEMENT: Automated segmentation of ovarian cancer may be used by clinicians for CT-based volumetric assessments and researchers for building complex analysis pipelines. KEY POINTS: • The first automated approach for pelvic/ovarian and omental ovarian cancer lesion segmentation on CT images has been presented. • Automated segmentation of ovarian cancer lesions can be comparable with manual segmentation of trainee radiologists. • Careful hyperparameter tuning can provide models significantly outperforming strong state-of-the-art baselines.


Assuntos
Aprendizado Profundo , Cistos Ovarianos , Neoplasias Ovarianas , Humanos , Feminino , Neoplasias Ovarianas/diagnóstico por imagem , Redes Neurais de Computação , Tomografia Computadorizada por Raios X
6.
Nat Commun ; 14(1): 6756, 2023 10 24.
Artigo em Inglês | MEDLINE | ID: mdl-37875466

RESUMO

High grade serous ovarian carcinoma (HGSOC) is a highly heterogeneous disease that typically presents at an advanced, metastatic state. The multi-scale complexity of HGSOC is a major obstacle to predicting response to neoadjuvant chemotherapy (NACT) and understanding critical determinants of response. Here we present a framework to predict the response of HGSOC patients to NACT integrating baseline clinical, blood-based, and radiomic biomarkers extracted from all primary and metastatic lesions. We use an ensemble machine learning model trained to predict the change in total disease volume using data obtained at diagnosis (n = 72). The model is validated in an internal hold-out cohort (n = 20) and an independent external patient cohort (n = 42). In the external cohort the integrated radiomics model reduces the prediction error by 8% with respect to the clinical model, achieving an AUC of 0.78 for RECIST 1.1 classification compared to 0.47 for the clinical model. Our results emphasize the value of including radiomics data in integrative models of treatment response and provide methods for developing new biomarker-based clinical trials of NACT in HGSOC.


Assuntos
Neoplasias Ovarianas , Humanos , Feminino , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/tratamento farmacológico , Neoplasias Ovarianas/genética , Terapia Neoadjuvante/métodos , Biomarcadores Tumorais/genética
7.
Diagnostics (Basel) ; 13(17)2023 Aug 30.
Artigo em Inglês | MEDLINE | ID: mdl-37685352

RESUMO

Artificial intelligence (AI) methods applied to healthcare problems have shown enormous potential to alleviate the burden of health services worldwide and to improve the accuracy and reproducibility of predictions. In particular, developments in computer vision are creating a paradigm shift in the analysis of radiological images, where AI tools are already capable of automatically detecting and precisely delineating tumours. However, such tools are generally developed in technical departments that continue to be siloed from where the real benefit would be achieved with their usage. Significant effort still needs to be made to make these advancements available, first in academic clinical research and ultimately in the clinical setting. In this paper, we demonstrate a prototype pipeline based entirely on open-source software and free of cost to bridge this gap, simplifying the integration of tools and models developed within the AI community into the clinical research setting, ensuring an accessible platform with visualisation applications that allow end-users such as radiologists to view and interact with the outcome of these AI tools.

8.
Sci Rep ; 13(1): 16239, 2023 09 27.
Artigo em Inglês | MEDLINE | ID: mdl-37758804

RESUMO

Multiple Sclerosis (MS) is an autoimmune demyelinating disease characterised by changes in iron and myelin content. These biomarkers are detectable by Quantitative Susceptibility Mapping (QSM), an advanced Magnetic Resonance Imaging technique detecting magnetic properties. When analysed with radiomic techniques that exploit its intrinsic quantitative nature, QSM may furnish biomarkers to facilitate early diagnosis of MS and timely assessment of progression. In this work, we explore the robustness of QSM radiomic features by varying the number of grey levels (GLs) and echo times (TEs), in a sample of healthy controls and patients with MS. We analysed the white matter in total and within six clinically relevant tracts, including the cortico-spinal tract and the optic radiation. After optimising the number of GLs (n = 64), at least 65% of features were robust for each Volume of Interest (VOI), with no difference (p > .05) between left and right hemispheres. Different outcomes in feature robustness among the VOIs depend on their characteristics, such as volume and variance of susceptibility values. This study validated the processing pipeline for robustness analysis and established the reliability of QSM-based radiomics features against GLs and TEs. Our results provide important insights for future radiomics studies using QSM in clinical applications.


Assuntos
Doenças Autoimunes , Esclerose Múltipla , Humanos , Esclerose Múltipla/diagnóstico por imagem , Reprodutibilidade dos Testes , Pacientes , Imageamento por Ressonância Magnética
9.
Comput Biol Med ; 163: 107096, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37302375

RESUMO

Uncertainty quantification in automated image analysis is highly desired in many applications. Typically, machine learning models in classification or segmentation are only developed to provide binary answers; however, quantifying the uncertainty of the models can play a critical role for example in active learning or machine human interaction. Uncertainty quantification is especially difficult when using deep learning-based models, which are the state-of-the-art in many imaging applications. The current uncertainty quantification approaches do not scale well in high-dimensional real-world problems. Scalable solutions often rely on classical techniques, such as dropout, during inference or training ensembles of identical models with different random seeds to obtain a posterior distribution. In this paper, we present the following contributions. First, we show that the classical approaches fail to approximate the classification probability. Second, we propose a scalable and intuitive framework for uncertainty quantification in medical image segmentation that yields measurements that approximate the classification probability. Third, we suggest the usage of k-fold cross-validation to overcome the need for held out calibration data. Lastly, we motivate the adoption of our method in active learning, creating pseudo-labels to learn from unlabeled images and human-machine collaboration.


Assuntos
Aprendizado Profundo , Humanos , Incerteza , Probabilidade , Calibragem , Processamento de Imagem Assistida por Computador
10.
Radiol Clin North Am ; 61(4): 749-760, 2023 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-37169435

RESUMO

Ovarian cancer, one of the deadliest gynecologic malignancies, is characterized by high intra- and inter-site genomic and phenotypic heterogeneity. The traditional information provided by the conventional interpretation of diagnostic imaging studies cannot adequately represent this heterogeneity. Radiomics analyses can capture the complex patterns related to the microstructure of the tissues and provide quantitative information about them. This review outlines how radiomics and its integration with other quantitative biological information, like genomics and proteomics, can impact the clinical management of ovarian cancer.


Assuntos
Neoplasias Ovarianas , Humanos , Feminino , Neoplasias Ovarianas/diagnóstico por imagem , Neoplasias Ovarianas/genética , Diagnóstico por Imagem , Genômica/métodos
11.
Heliyon ; 9(5): e15984, 2023 May.
Artigo em Inglês | MEDLINE | ID: mdl-37215845

RESUMO

Introduction: The aim of our study was to evaluate the feasibility of texture analysis of epicardial fat (EF) and thoracic subcutaneous fat (TSF) in patients undergoing cardiac CT (CCT). Materials and methods: We compared a consecutive population of 30 patients with BMI ≤25 kg/m2 (Group A, 60.6 ± 13.7 years) with a control population of 30 patients with BMI >25 kg/m2 (Group B, 63.3 ± 11 years). A dedicated computer application for quantification of EF and a texture analysis application for the study of EF and TSF were employed. Results: The volume of EF was higher in group B (mean 116.1 cm3 vs. 86.3 cm3, p = 0.014), despite no differences were found neither in terms of mean density (-69.5 ± 5 HU vs. -68 ± 5 HU, p = 0.28), nor in terms of quartiles distribution (Q1, p = 0.83; Q2, p = 0.22, Q3, p = 0.83, Q4, p = 0.34). The discriminating parameters of the histogram class were mean (p = 0.02), 0,1st (p = 0.001), 10th (p = 0.002), and 50th percentiles (p = 0.02). DifVarnc was the discriminating parameter of the co-occurrence matrix class (p = 0.007).The TSF thickness was 15 ± 6 mm in group A and 19.5 ± 5 mm in group B (p = 0.003). The TSF had a mean density of -97 ± 19 HU in group A and -95.8 ± 19 HU in group B (p = 0.75). The discriminating parameters of texture analysis were 10th (p = 0.03), 50th (p = 0.01), 90th percentiles (p = 0.04), S(0,1)SumAverg (p = 0.02), S(1,-1)SumOfSqs (p = 0.02), S(3,0)Contrast (p = 0.03), S(3,0)SumAverg (p = 0.02), S(4,0)SumAverg (p = 0.04), Horzl_RLNonUni (p = 0.02), and Vertl_LngREmph (p = 0.0005). Conclusions: Texture analysis provides distinctive radiomic parameters of EF and TSF. EF and TSF had different radiomic features as the BMI varies.

12.
Front Oncol ; 13: 1085874, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36860310

RESUMO

Background: High-Grade Serous Ovarian Carcinoma (HGSOC) is the most prevalent and lethal subtype of ovarian cancer, but has a paucity of clinically-actionable biomarkers due to high degrees of multi-level heterogeneity. Radiogenomics markers have the potential to improve prediction of patient outcome and treatment response, but require accurate multimodal spatial registration between radiological imaging and histopathological tissue samples. Previously published co-registration work has not taken into account the anatomical, biological and clinical diversity of ovarian tumours. Methods: In this work, we developed a research pathway and an automated computational pipeline to produce lesion-specific three-dimensional (3D) printed moulds based on preoperative cross-sectional CT or MRI of pelvic lesions. Moulds were designed to allow tumour slicing in the anatomical axial plane to facilitate detailed spatial correlation of imaging and tissue-derived data. Code and design adaptations were made following each pilot case through an iterative refinement process. Results: Five patients with confirmed or suspected HGSOC who underwent debulking surgery between April and December 2021 were included in this prospective study. Tumour moulds were designed and 3D-printed for seven pelvic lesions, covering a range of tumour volumes (7 to 133 cm3) and compositions (cystic and solid proportions). The pilot cases informed innovations to improve specimen and subsequent slice orientation, through the use of 3D-printed tumour replicas and incorporation of a slice orientation slit in the mould design, respectively. The overall research pathway was compatible with implementation within the clinically determined timeframe and treatment pathway for each case, involving multidisciplinary clinical professionals from Radiology, Surgery, Oncology and Histopathology Departments. Conclusions: We developed and refined a computational pipeline that can model lesion-specific 3D-printed moulds from preoperative imaging for a variety of pelvic tumours. This framework can be used to guide comprehensive multi-sampling of tumour resection specimens.

13.
Eur Radiol ; 33(6): 3792-3800, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-36749370

RESUMO

Serial MRI is an essential assessment tool in prostate cancer (PCa) patients enrolled on active surveillance (AS). However, it has only moderate sensitivity for predicting histopathological tumour progression at follow-up, which is in part due to the subjective nature of its clinical reporting and variation among centres and readers. In this study, we used a long short-term memory (LSTM) recurrent neural network (RNN) to develop a time series radiomics (TSR) predictive model that analysed longitudinal changes in tumour-derived radiomic features across 297 scans from 76 AS patients, 28 with histopathological PCa progression and 48 with stable disease. Using leave-one-out cross-validation (LOOCV), we found that an LSTM-based model combining TSR and serial PSA density (AUC 0.86 [95% CI: 0.78-0.94]) significantly outperformed a model combining conventional delta-radiomics and delta-PSA density (0.75 [0.64-0.87]; p = 0.048) and achieved comparable performance to expert-performed serial MRI analysis using the Prostate Cancer Radiologic Estimation of Change in Sequential Evaluation (PRECISE) scoring system (0.84 [0.76-0.93]; p = 0.710). The proposed TSR framework, therefore, offers a feasible quantitative tool for standardising serial MRI assessment in PCa AS. It also presents a novel methodological approach to serial image analysis that can be used to support clinical decision-making in multiple scenarios, from continuous disease monitoring to treatment response evaluation. KEY POINTS: •LSTM RNN can be used to predict the outcome of PCa AS using time series changes in tumour-derived radiomic features and PSA density. •Using all available TSR features and serial PSA density yields a significantly better predictive performance compared to using just two time points within the delta-radiomics framework. •The concept of TSR can be applied to other clinical scenarios involving serial imaging, setting out a new field in AI-driven radiology research.


Assuntos
Antígeno Prostático Específico , Neoplasias da Próstata , Masculino , Humanos , Conduta Expectante , Fatores de Tempo , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Próstata/patologia , Imageamento por Ressonância Magnética/métodos , Estudos Retrospectivos
14.
J Digit Imaging ; 36(2): 739-752, 2023 04.
Artigo em Inglês | MEDLINE | ID: mdl-36474089

RESUMO

The Dice similarity coefficient (DSC) is both a widely used metric and loss function for biomedical image segmentation due to its robustness to class imbalance. However, it is well known that the DSC loss is poorly calibrated, resulting in overconfident predictions that cannot be usefully interpreted in biomedical and clinical practice. Performance is often the only metric used to evaluate segmentations produced by deep neural networks, and calibration is often neglected. However, calibration is important for translation into biomedical and clinical practice, providing crucial contextual information to model predictions for interpretation by scientists and clinicians. In this study, we provide a simple yet effective extension of the DSC loss, named the DSC++ loss, that selectively modulates the penalty associated with overconfident, incorrect predictions. As a standalone loss function, the DSC++ loss achieves significantly improved calibration over the conventional DSC loss across six well-validated open-source biomedical imaging datasets, including both 2D binary and 3D multi-class segmentation tasks. Similarly, we observe significantly improved calibration when integrating the DSC++ loss into four DSC-based loss functions. Finally, we use softmax thresholding to illustrate that well calibrated outputs enable tailoring of recall-precision bias, which is an important post-processing technique to adapt the model predictions to suit the biomedical or clinical task. The DSC++ loss overcomes the major limitation of the DSC loss, providing a suitable loss function for training deep learning segmentation models for use in biomedical and clinical practice. Source code is available at https://github.com/mlyg/DicePlusPlus .


Assuntos
Processamento de Imagem Assistida por Computador , Redes Neurais de Computação , Humanos , Processamento de Imagem Assistida por Computador/métodos
16.
Sci Rep ; 12(1): 15142, 2022 09 07.
Artigo em Inglês | MEDLINE | ID: mdl-36071117

RESUMO

Photoacoustic imaging is an increasingly popular method of exploring the tumour microenvironment, which can provide insight into tumour oxygenation status and potentially treatment response assessment. Currently, the measurements most commonly performed on such images are the mean and median of the pixel values of the tumour volumes of interest. We investigated expanding the set of measurements that can be extracted from these images by adding radiomic features. In particular, we found that Skewness was sensitive to differences between basal and luminal patient derived xenograft cancer models with an [Formula: see text] of 0.86, and that it was robust to variations in confounding factors such as reconstruction type and wavelength. We also built discriminant models with radiomic features that were correlated with the underlying tumour model and were independent from each other. We then ranked features by their importance in the model. Skewness was again found to be an important feature, as were 10th Percentile, Root Mean Squared, and several other texture-based features. In summary, this paper proposes a methodology to select radiomic features extracted from photoacoustic images that are robust to changes in acquisition and reconstruction parameters, and discusses features found to have discriminating power between the underlying tumour models in a pre-clinical dataset.


Assuntos
Neoplasias , Técnicas Fotoacústicas , Animais , Diagnóstico por Imagem , Modelos Animais de Doenças , Xenoenxertos , Humanos , Neoplasias/diagnóstico por imagem , Microambiente Tumoral
17.
Diagnostics (Basel) ; 12(9)2022 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-36140526

RESUMO

Given growing clinical needs, in recent years Artificial Intelligence (AI) techniques have increasingly been used to define the best approaches for survival assessment and prediction in patients with brain tumors. Advances in computational resources, and the collection of (mainly) public databases, have promoted this rapid development. This narrative review of the current state-of-the-art aimed to survey current applications of AI in predicting survival in patients with brain tumors, with a focus on Magnetic Resonance Imaging (MRI). An extensive search was performed on PubMed and Google Scholar using a Boolean research query based on MeSH terms and restricting the search to the period between 2012 and 2022. Fifty studies were selected, mainly based on Machine Learning (ML), Deep Learning (DL), radiomics-based methods, and methods that exploit traditional imaging techniques for survival assessment. In addition, we focused on two distinct tasks related to survival assessment: the first on the classification of subjects into survival classes (short and long-term or eventually short, mid and long-term) to stratify patients in distinct groups. The second focused on quantification, in days or months, of the individual survival interval. Our survey showed excellent state-of-the-art methods for the first, with accuracy up to ∼98%. The latter task appears to be the most challenging, but state-of-the-art techniques showed promising results, albeit with limitations, with C-Index up to ∼0.91. In conclusion, according to the specific task, the available computational methods perform differently, and the choice of the best one to use is non-univocal and dependent on many aspects. Unequivocally, the use of features derived from quantitative imaging has been shown to be advantageous for AI applications, including survival prediction. This evidence from the literature motivates further research in the field of AI-powered methods for survival prediction in patients with brain tumors, in particular, using the wealth of information provided by quantitative MRI techniques.

18.
Diagnostics (Basel) ; 12(8)2022 Jul 31.
Artigo em Inglês | MEDLINE | ID: mdl-36010200

RESUMO

Convolutional neural networks (CNNs) constitute a widely used deep learning approach that has frequently been applied to the problem of brain tumor diagnosis. Such techniques still face some critical challenges in moving towards clinic application. The main objective of this work is to present a comprehensive review of studies using CNN architectures to classify brain tumors using MR images with the aim of identifying useful strategies for and possible impediments in the development of this technology. Relevant articles were identified using a predefined, systematic procedure. For each article, data were extracted regarding training data, target problems, the network architecture, validation methods, and the reported quantitative performance criteria. The clinical relevance of the studies was then evaluated to identify limitations by considering the merits of convolutional neural networks and the remaining challenges that need to be solved to promote the clinical application and development of CNN algorithms. Finally, possible directions for future research are discussed for researchers in the biomedical and machine learning communities. A total of 83 studies were identified and reviewed. They differed in terms of the precise classification problem targeted and the strategies used to construct and train the chosen CNN. Consequently, the reported performance varied widely, with accuracies of 91.63-100% in differentiating meningiomas, gliomas, and pituitary tumors (26 articles) and of 60.0-99.46% in distinguishing low-grade from high-grade gliomas (13 articles). The review provides a survey of the state of the art in CNN-based deep learning methods for brain tumor classification. Many networks demonstrated good performance, and it is not evident that any specific methodological choice greatly outperforms the alternatives, especially given the inconsistencies in the reporting of validation methods, performance metrics, and training data encountered. Few studies have focused on clinical usability.

19.
J Imaging ; 8(8)2022 Jul 22.
Artigo em Inglês | MEDLINE | ID: mdl-35893083

RESUMO

Management of brain tumors is based on clinical and radiological information with presumed grade dictating treatment. Hence, a non-invasive assessment of tumor grade is of paramount importance to choose the best treatment plan. Convolutional Neural Networks (CNNs) represent one of the effective Deep Learning (DL)-based techniques that have been used for brain tumor diagnosis. However, they are unable to handle input modifications effectively. Capsule neural networks (CapsNets) are a novel type of machine learning (ML) architecture that was recently developed to address the drawbacks of CNNs. CapsNets are resistant to rotations and affine translations, which is beneficial when processing medical imaging datasets. Moreover, Vision Transformers (ViT)-based solutions have been very recently proposed to address the issue of long-range dependency in CNNs. This survey provides a comprehensive overview of brain tumor classification and segmentation techniques, with a focus on ML-based, CNN-based, CapsNet-based, and ViT-based techniques. The survey highlights the fundamental contributions of recent studies and the performance of state-of-the-art techniques. Moreover, we present an in-depth discussion of crucial issues and open challenges. We also identify some key limitations and promising future research directions. We envisage that this survey shall serve as a good springboard for further study.

20.
Front Oncol ; 12: 868265, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35785153

RESUMO

Background: Pathological response to neoadjuvant treatment for patients with high-grade serous ovarian carcinoma (HGSOC) is assessed using the chemotherapy response score (CRS) for omental tumor deposits. The main limitation of CRS is that it requires surgical sampling after initial neoadjuvant chemotherapy (NACT) treatment. Earlier and non-invasive response predictors could improve patient stratification. We developed computed tomography (CT) radiomic measures to predict neoadjuvant response before NACT using CRS as a gold standard. Methods: Omental CT-based radiomics models, yielding a simplified fully interpretable radiomic signature, were developed using Elastic Net logistic regression and compared to predictions based on omental tumor volume alone. Models were developed on a single institution cohort of neoadjuvant-treated HGSOC (n = 61; 41% complete response to NCT) and tested on an external test cohort (n = 48; 21% complete response). Results: The performance of the comprehensive radiomics models and the fully interpretable radiomics model was significantly higher than volume-based predictions of response in both the discovery and external test sets when assessed using G-mean (geometric mean of sensitivity and specificity) and NPV, indicating high generalizability and reliability in identifying non-responders when using radiomics. The performance of a fully interpretable model was similar to that of comprehensive radiomics models. Conclusions: CT-based radiomics allows for predicting response to NACT in a timely manner and without the need for abdominal surgery. Adding pre-NACT radiomics to volumetry improved model performance for predictions of response to NACT in HGSOC and was robust to external testing. A radiomic signature based on five robust predictive features provides improved clinical interpretability and may thus facilitate clinical acceptance and application.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...