Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 48.236
Filtrar
1.
Skin Res Technol ; 30(5): e13607, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38742379

RESUMO

BACKGROUND: Timely diagnosis plays a critical role in determining melanoma prognosis, prompting the development of deep learning models to aid clinicians. Questions persist regarding the efficacy of clinical images alone or in conjunction with dermoscopy images for model training. This study aims to compare the classification performance for melanoma of three types of CNN models: those trained on clinical images, dermoscopy images, and a combination of paired clinical and dermoscopy images from the same lesion. MATERIALS AND METHODS: We divided 914 image pairs into training, validation, and test sets. Models were built using pre-trained Inception-ResNetV2 convolutional layers for feature extraction, followed by binary classification. Training comprised 20 models per CNN type using sets of random hyperparameters. Best models were chosen based on validation AUC-ROC. RESULTS: Significant AUC-ROC differences were found between clinical versus dermoscopy models (0.661 vs. 0.869, p < 0.001) and clinical versus clinical + dermoscopy models (0.661 vs. 0.822, p = 0.001). Significant sensitivity differences were found between clinical and dermoscopy models (0.513 vs. 0.799, p = 0.01), dermoscopy versus clinical + dermoscopy models (0.799 vs. 1.000, p = 0.02), and clinical versus clinical + dermoscopy models (0.513 vs. 1.000, p < 0.001). Significant specificity differences were found between dermoscopy versus clinical + dermoscopy models (0.800 vs. 0.288, p < 0.001) and clinical versus clinical + dermoscopy models (0.650 vs. 0.288, p < 0.001). CONCLUSION: CNN models trained on dermoscopy images outperformed those relying solely on clinical images under our study conditions. The potential advantages of incorporating paired clinical and dermoscopy images for CNN-based melanoma classification appear less clear based on our findings.


Assuntos
Dermoscopia , Melanoma , Redes Neurais de Computação , Neoplasias Cutâneas , Humanos , Melanoma/diagnóstico por imagem , Melanoma/patologia , Melanoma/classificação , Dermoscopia/métodos , Neoplasias Cutâneas/diagnóstico por imagem , Neoplasias Cutâneas/patologia , Neoplasias Cutâneas/classificação , Aprendizado Profundo , Sensibilidade e Especificidade , Feminino , Curva ROC , Interpretação de Imagem Assistida por Computador/métodos , Masculino
2.
BMC Med Imaging ; 24(1): 107, 2024 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-38734629

RESUMO

This study addresses the critical challenge of detecting brain tumors using MRI images, a pivotal task in medical diagnostics that demands high accuracy and interpretability. While deep learning has shown remarkable success in medical image analysis, there remains a substantial need for models that are not only accurate but also interpretable to healthcare professionals. The existing methodologies, predominantly deep learning-based, often act as black boxes, providing little insight into their decision-making process. This research introduces an integrated approach using ResNet50, a deep learning model, combined with Gradient-weighted Class Activation Mapping (Grad-CAM) to offer a transparent and explainable framework for brain tumor detection. We employed a dataset of MRI images, enhanced through data augmentation, to train and validate our model. The results demonstrate a significant improvement in model performance, with a testing accuracy of 98.52% and precision-recall metrics exceeding 98%, showcasing the model's effectiveness in distinguishing tumor presence. The application of Grad-CAM provides insightful visual explanations, illustrating the model's focus areas in making predictions. This fusion of high accuracy and explainability holds profound implications for medical diagnostics, offering a pathway towards more reliable and interpretable brain tumor detection tools.


Assuntos
Neoplasias Encefálicas , Aprendizado Profundo , Imageamento por Ressonância Magnética , Humanos , Neoplasias Encefálicas/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Interpretação de Imagem Assistida por Computador/métodos
3.
Radiology ; 311(2): e230750, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38713024

RESUMO

Background Multiparametric MRI (mpMRI) improves prostate cancer (PCa) detection compared with systematic biopsy, but its interpretation is prone to interreader variation, which results in performance inconsistency. Artificial intelligence (AI) models can assist in mpMRI interpretation, but large training data sets and extensive model testing are required. Purpose To evaluate a biparametric MRI AI algorithm for intraprostatic lesion detection and segmentation and to compare its performance with radiologist readings and biopsy results. Materials and Methods This secondary analysis of a prospective registry included consecutive patients with suspected or known PCa who underwent mpMRI, US-guided systematic biopsy, or combined systematic and MRI/US fusion-guided biopsy between April 2019 and September 2022. All lesions were prospectively evaluated using Prostate Imaging Reporting and Data System version 2.1. The lesion- and participant-level performance of a previously developed cascaded deep learning algorithm was compared with histopathologic outcomes and radiologist readings using sensitivity, positive predictive value (PPV), and Dice similarity coefficient (DSC). Results A total of 658 male participants (median age, 67 years [IQR, 61-71 years]) with 1029 MRI-visible lesions were included. At histopathologic analysis, 45% (294 of 658) of participants had lesions of International Society of Urological Pathology (ISUP) grade group (GG) 2 or higher. The algorithm identified 96% (282 of 294; 95% CI: 94%, 98%) of all participants with clinically significant PCa, whereas the radiologist identified 98% (287 of 294; 95% CI: 96%, 99%; P = .23). The algorithm identified 84% (103 of 122), 96% (152 of 159), 96% (47 of 49), 95% (38 of 40), and 98% (45 of 46) of participants with ISUP GG 1, 2, 3, 4, and 5 lesions, respectively. In the lesion-level analysis using radiologist ground truth, the detection sensitivity was 55% (569 of 1029; 95% CI: 52%, 58%), and the PPV was 57% (535 of 934; 95% CI: 54%, 61%). The mean number of false-positive lesions per participant was 0.61 (range, 0-3). The lesion segmentation DSC was 0.29. Conclusion The AI algorithm detected cancer-suspicious lesions on biparametric MRI scans with a performance comparable to that of an experienced radiologist. Moreover, the algorithm reliably predicted clinically significant lesions at histopathologic examination. ClinicalTrials.gov Identifier: NCT03354416 © RSNA, 2024 Supplemental material is available for this article.


Assuntos
Aprendizado Profundo , Imageamento por Ressonância Magnética Multiparamétrica , Neoplasias da Próstata , Masculino , Humanos , Neoplasias da Próstata/diagnóstico por imagem , Neoplasias da Próstata/patologia , Idoso , Estudos Prospectivos , Imageamento por Ressonância Magnética Multiparamétrica/métodos , Pessoa de Meia-Idade , Algoritmos , Próstata/diagnóstico por imagem , Próstata/patologia , Biópsia Guiada por Imagem/métodos , Interpretação de Imagem Assistida por Computador/métodos , Imageamento por Ressonância Magnética/métodos
4.
Radiol Cardiothorac Imaging ; 6(3): e230177, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38722232

RESUMO

Purpose To develop a deep learning model for increasing cardiac cine frame rate while maintaining spatial resolution and scan time. Materials and Methods A transformer-based model was trained and tested on a retrospective sample of cine images from 5840 patients (mean age, 55 years ± 19 [SD]; 3527 male patients) referred for clinical cardiac MRI from 2003 to 2021 at nine centers; images were acquired using 1.5- and 3-T scanners from three vendors. Data from three centers were used for training and testing (4:1 ratio). The remaining data were used for external testing. Cines with downsampled frame rates were restored using linear, bicubic, and model-based interpolation. The root mean square error between interpolated and original cine images was modeled using ordinary least squares regression. In a prospective study of 49 participants referred for clinical cardiac MRI (mean age, 56 years ± 13; 25 male participants) and 12 healthy participants (mean age, 51 years ± 16; eight male participants), the model was applied to cines acquired at 25 frames per second (fps), thereby doubling the frame rate, and these interpolated cines were compared with actual 50-fps cines. The preference of two readers based on perceived temporal smoothness and image quality was evaluated using a noninferiority margin of 10%. Results The model generated artifact-free interpolated images. Ordinary least squares regression analysis accounting for vendor and field strength showed lower error (P < .001) with model-based interpolation compared with linear and bicubic interpolation in internal and external test sets. The highest proportion of reader choices was "no preference" (84 of 122) between actual and interpolated 50-fps cines. The 90% CI for the difference between reader proportions favoring collected (15 of 122) and interpolated (23 of 122) high-frame-rate cines was -0.01 to 0.14, indicating noninferiority. Conclusion A transformer-based deep learning model increased cardiac cine frame rates while preserving both spatial resolution and scan time, resulting in images with quality comparable to that of images obtained at actual high frame rates. Keywords: Functional MRI, Heart, Cardiac, Deep Learning, High Frame Rate Supplemental material is available for this article. © RSNA, 2024.


Assuntos
Aprendizado Profundo , Imagem Cinética por Ressonância Magnética , Humanos , Masculino , Imagem Cinética por Ressonância Magnética/métodos , Pessoa de Meia-Idade , Feminino , Estudos Prospectivos , Estudos Retrospectivos , Coração/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos
6.
Comput Biol Med ; 175: 108412, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38691914

RESUMO

Brain tumor segmentation and classification play a crucial role in the diagnosis and treatment planning of brain tumors. Accurate and efficient methods for identifying tumor regions and classifying different tumor types are essential for guiding medical interventions. This study comprehensively reviews brain tumor segmentation and classification techniques, exploring various approaches based on image processing, machine learning, and deep learning. Furthermore, our study aims to review existing methodologies, discuss their advantages and limitations, and highlight recent advancements in this field. The impact of existing segmentation and classification techniques for automated brain tumor detection is also critically examined using various open-source datasets of Magnetic Resonance Images (MRI) of different modalities. Moreover, our proposed study highlights the challenges related to segmentation and classification techniques and datasets having various MRI modalities to enable researchers to develop innovative and robust solutions for automated brain tumor detection. The results of this study contribute to the development of automated and robust solutions for analyzing brain tumors, ultimately aiding medical professionals in making informed decisions and providing better patient care.


Assuntos
Neoplasias Encefálicas , Imageamento por Ressonância Magnética , Humanos , Neoplasias Encefálicas/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Aprendizado Profundo , Interpretação de Imagem Assistida por Computador/métodos , Encéfalo/diagnóstico por imagem , Aprendizado de Máquina , Processamento de Imagem Assistida por Computador/métodos , Neuroimagem/métodos
7.
Comput Biol Med ; 175: 108459, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38701588

RESUMO

Diabetic retinopathy (DR) is the most common diabetic complication, which usually leads to retinal damage, vision loss, and even blindness. A computer-aided DR grading system has a significant impact on helping ophthalmologists with rapid screening and diagnosis. Recent advances in fundus photography have precipitated the development of novel retinal imaging cameras and their subsequent implementation in clinical practice. However, most deep learning-based algorithms for DR grading demonstrate limited generalization across domains. This inferior performance stems from variance in imaging protocols and devices inducing domain shifts. We posit that declining model performance between domains arises from learning spurious correlations in the data. Incorporating do-operations from causality analysis into model architectures may mitigate this issue and improve generalizability. Specifically, a novel universal structural causal model (SCM) was proposed to analyze spurious correlations in fundus imaging. Building on this, a causality-inspired diabetic retinopathy grading framework named CauDR was developed to eliminate spurious correlations and achieve more generalizable DR diagnostics. Furthermore, existing datasets were reorganized into 4DR benchmark for DG scenario. Results demonstrate the effectiveness and the state-of-the-art (SOTA) performance of CauDR. Diabetic retinopathy (DR) is the most common diabetic complication, which usually leads to retinal damage, vision loss, and even blindness. A computer-aided DR grading system has a significant impact on helping ophthalmologists with rapid screening and diagnosis. Recent advances in fundus photography have precipitated the development of novel retinal imaging cameras and their subsequent implementation in clinical practice. However, most deep learning-based algorithms for DR grading demonstrate limited generalization across domains. This inferior performance stems from variance in imaging protocols and devices inducing domain shifts. We posit that declining model performance between domains arises from learning spurious correlations in the data. Incorporating do-operations from causality analysis into model architectures may mitigate this issue and improve generalizability. Specifically, a novel universal structural causal model (SCM) was proposed to analyze spurious correlations in fundus imaging. Building on this, a causality-inspired diabetic retinopathy grading framework named CauDR was developed to eliminate spurious correlations and achieve more generalizable DR diagnostics. Furthermore, existing datasets were reorganized into 4DR benchmark for DG scenario. Results demonstrate the effectiveness and the state-of-the-art (SOTA) performance of CauDR.


Assuntos
Retinopatia Diabética , Retinopatia Diabética/diagnóstico por imagem , Retinopatia Diabética/diagnóstico , Humanos , Fundo de Olho , Algoritmos , Aprendizado Profundo , Interpretação de Imagem Assistida por Computador/métodos
8.
Comput Biol Med ; 175: 108523, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38701591

RESUMO

Diabetic retinopathy is considered one of the most common diseases that can lead to blindness in the working age, and the chance of developing it increases as long as a person suffers from diabetes. Protecting the sight of the patient or decelerating the evolution of this disease depends on its early detection as well as identifying the exact levels of this pathology, which is done manually by ophthalmologists. This manual process is very consuming in terms of the time and experience of an expert ophthalmologist, which makes developing an automated method to aid in the diagnosis of diabetic retinopathy an essential and urgent need. In this paper, we aim to propose a new hybrid deep learning method based on a fine-tuning vision transformer and a modified capsule network for automatic diabetic retinopathy severity level prediction. The proposed approach consists of a new range of computer vision operations, including the power law transformation technique and the contrast-limiting adaptive histogram equalization technique in the preprocessing step. While the classification step builds up on a fine-tuning vision transformer, a modified capsule network, and a classification model combined with a classification model, The effectiveness of our approach was evaluated using four datasets, including the APTOS, Messidor-2, DDR, and EyePACS datasets, for the task of severity levels of diabetic retinopathy. We have attained excellent test accuracy scores on the four datasets, respectively: 88.18%, 87.78%, 80.36%, and 78.64%. Comparing our results with the state-of-the-art, we reached a better performance.


Assuntos
Aprendizado Profundo , Retinopatia Diabética , Retinopatia Diabética/diagnóstico por imagem , Humanos , Redes Neurais de Computação , Bases de Dados Factuais , Interpretação de Imagem Assistida por Computador/métodos , Algoritmos
9.
Artif Intell Med ; 152: 102872, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38701636

RESUMO

Accurately measuring the evolution of Multiple Sclerosis (MS) with magnetic resonance imaging (MRI) critically informs understanding of disease progression and helps to direct therapeutic strategy. Deep learning models have shown promise for automatically segmenting MS lesions, but the scarcity of accurately annotated data hinders progress in this area. Obtaining sufficient data from a single clinical site is challenging and does not address the heterogeneous need for model robustness. Conversely, the collection of data from multiple sites introduces data privacy concerns and potential label noise due to varying annotation standards. To address this dilemma, we explore the use of the federated learning framework while considering label noise. Our approach enables collaboration among multiple clinical sites without compromising data privacy under a federated learning paradigm that incorporates a noise-robust training strategy based on label correction. Specifically, we introduce a Decoupled Hard Label Correction (DHLC) strategy that considers the imbalanced distribution and fuzzy boundaries of MS lesions, enabling the correction of false annotations based on prediction confidence. We also introduce a Centrally Enhanced Label Correction (CELC) strategy, which leverages the aggregated central model as a correction teacher for all sites, enhancing the reliability of the correction process. Extensive experiments conducted on two multi-site datasets demonstrate the effectiveness and robustness of our proposed methods, indicating their potential for clinical applications in multi-site collaborations to train better deep learning models with lower cost in data collection and annotation.


Assuntos
Aprendizado Profundo , Imageamento por Ressonância Magnética , Esclerose Múltipla , Esclerose Múltipla/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética/métodos , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos
10.
Comput Methods Programs Biomed ; 250: 108205, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38703435

RESUMO

The pancreas is a vital organ in digestive system which has significant health implications. It is imperative to evaluate and identify malignant pancreatic lesions promptly in light of the high mortality rate linked to such malignancies. Endoscopic Ultrasound (EUS) is a non-invasive precise technique to detect pancreas disorders, but it is highly operator dependent. Artificial intelligence (AI), including traditional machine learning (ML) and deep learning (DL) techniques can play a pivotal role to enhancing the performance of EUS regardless of operator. AI performs a critical function in the detection, classification, and segmentation of medical images. The utilization of AI-assisted systems has improved the accuracy and productivity of pancreatic analysis, including the detection of diverse pancreatic disorders (e.g., pancreatitis, masses, and cysts) as well as landmarks and parenchyma. This systematic review examines the rapidly developing domain of AI-assisted system in EUS of the pancreas. Its objective is to present a thorough study of the present research status and developments in this area. This paper explores the significant challenges of AI-assisted system in pancreas EUS imaging, highlights the potential of AI techniques in addressing these challenges, and suggests the scope for future research in domain of AI-assisted EUS systems.


Assuntos
Inteligência Artificial , Endossonografia , Pâncreas , Humanos , Endossonografia/métodos , Pâncreas/diagnóstico por imagem , Aprendizado de Máquina , Aprendizado Profundo , Neoplasias Pancreáticas/diagnóstico por imagem , Pancreatopatias/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos
11.
Comput Biol Med ; 175: 108549, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38704901

RESUMO

In this paper, we propose a multi-task learning (MTL) network based on the label-level fusion of metadata and hand-crafted features by unsupervised clustering to generate new clustering labels as an optimization goal. We propose a MTL module (MTLM) that incorporates an attention mechanism to enable the model to learn more integrated, variable information. We propose a dynamic strategy to adjust the loss weights of different tasks, and trade off the contributions of multiple branches. Instead of feature-level fusion, we propose label-level fusion and combine the results of our proposed MTLM with the results of the image classification network to achieve better lesion prediction on multiple dermatological datasets. We verify the effectiveness of the proposed model by quantitative and qualitative measures. The MTL network using multi-modal clues and label-level fusion can yield the significant performance improvement for skin lesion classification.


Assuntos
Pele , Humanos , Pele/diagnóstico por imagem , Pele/patologia , Interpretação de Imagem Assistida por Computador/métodos , Aprendizado de Máquina , Neoplasias Cutâneas/diagnóstico por imagem , Neoplasias Cutâneas/patologia , Redes Neurais de Computação , Algoritmos , Dermatopatias/diagnóstico por imagem
12.
Sci Rep ; 14(1): 9336, 2024 04 23.
Artigo em Inglês | MEDLINE | ID: mdl-38653997

RESUMO

Skin cancer is the most prevalent kind of cancer in people. It is estimated that more than 1 million people get skin cancer every year in the world. The effectiveness of the disease's therapy is significantly impacted by early identification of this illness. Preprocessing is the initial detecting stage in enhancing the quality of skin images by removing undesired background noise and objects. This study aims is to compile preprocessing techniques for skin cancer imaging that are currently accessible. Researchers looking into automated skin cancer diagnosis might use this article as an excellent place to start. The fully convolutional encoder-decoder network and Sparrow search algorithm (FCEDN-SpaSA) are proposed in this study for the segmentation of dermoscopic images. The individual wolf method and the ensemble ghosting technique are integrated to generate a neighbour-based search strategy in SpaSA for stressing the correct balance between navigation and exploitation. The classification procedure is accomplished by using an adaptive CNN technique to discriminate between normal skin and malignant skin lesions suggestive of disease. Our method provides classification accuracies comparable to commonly used incremental learning techniques while using less energy, storage space, memory access, and training time (only network updates with new training samples, no network sharing). In a simulation, the segmentation performance of the proposed technique on the ISBI 2017, ISIC 2018, and PH2 datasets reached accuracies of 95.28%, 95.89%, 92.70%, and 98.78%, respectively, on the same dataset and assessed the classification performance. It is accurate 91.67% of the time. The efficiency of the suggested strategy is demonstrated through comparisons with cutting-edge methodologies.


Assuntos
Algoritmos , Dermoscopia , Redes Neurais de Computação , Neoplasias Cutâneas , Humanos , Neoplasias Cutâneas/diagnóstico , Neoplasias Cutâneas/diagnóstico por imagem , Neoplasias Cutâneas/classificação , Neoplasias Cutâneas/patologia , Dermoscopia/métodos , Processamento de Imagem Assistida por Computador/métodos , Interpretação de Imagem Assistida por Computador/métodos , Pele/patologia , Pele/diagnóstico por imagem
13.
Tomography ; 10(4): 459-470, 2024 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-38668393

RESUMO

BACKGROUND: Left atrial (LA) assessment is an important marker of adverse cardiovascular outcomes. Cardiovascular magnetic resonance (CMR) accurately quantifies LA volume and function based on biplane long-axis imaging. We aimed to validate single-plane-derived LA indices against the biplane method to simplify the post-processing of cine CMR. METHODS: In this study, 100 patients from Leeds Teaching Hospitals were used as the derivation cohort. Bias correction for the single plane method was applied and subsequently validated in 79 subjects. RESULTS: There were significant differences between the biplane and single plane mean LA maximum and minimum volumes and LA ejection fraction (EF) (all p < 0.01). After correcting for biases in the validation cohort, significant correlations in all LA indices were observed (0.89 to 0.98). The area under the curve (AUC) for the single plane to predict biplane cutoffs of LA maximum volume ≥ 112 mL was 0.97, LA minimum volume ≥ 44 mL was 0.99, LA stroke volume (SV) ≤ 21 mL was 1, and LA EF ≤ 46% was 1, (all p < 0.001). CONCLUSIONS: LA volumetric and functional assessment by the single plane method has a systematic bias compared to the biplane method. After bias correction, single plane LA volume and function are comparable to the biplane method.


Assuntos
Átrios do Coração , Imagem Cinética por Ressonância Magnética , Humanos , Imagem Cinética por Ressonância Magnética/métodos , Feminino , Masculino , Átrios do Coração/diagnóstico por imagem , Pessoa de Meia-Idade , Idoso , Volume Sistólico/fisiologia , Reprodutibilidade dos Testes , Adulto , Interpretação de Imagem Assistida por Computador/métodos
14.
Artif Intell Med ; 151: 102866, 2024 May.
Artigo em Inglês | MEDLINE | ID: mdl-38593684

RESUMO

An echocardiogram is a sophisticated ultrasound imaging technique employed to diagnose heart conditions. The transthoracic echocardiogram, one of the most prevalent types, is instrumental in evaluating significant cardiac diseases. However, interpreting its results heavily relies on the clinician's expertise. In this context, artificial intelligence has emerged as a vital tool for helping clinicians. This study critically analyzes key state-of-the-art research that uses deep learning techniques to automate transthoracic echocardiogram analysis and support clinical judgments. We have systematically organized and categorized articles that proffer solutions for view classification, enhancement of image quality and dataset, segmentation and identification of cardiac structures, detection of cardiac function abnormalities, and quantification of cardiac functions. We compared the performance of various deep learning approaches within each category, identifying the most promising methods. Additionally, we highlight limitations in current research and explore promising avenues for future exploration. These include addressing generalizability issues, incorporating novel AI approaches, and tackling the analysis of rare cardiac diseases.


Assuntos
Aprendizado Profundo , Ecocardiografia , Humanos , Ecocardiografia/métodos , Cardiopatias/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Inteligência Artificial
15.
Comput Biol Med ; 175: 108410, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38678938

RESUMO

Latent diffusion models (LDMs) have emerged as a state-of-the-art image generation method, outperforming previous Generative Adversarial Networks (GANs) in terms of training stability and image quality. In computational pathology, generative models are valuable for data sharing and data augmentation. However, the impact of LDM-generated images on histopathology tasks compared to traditional GANs has not been systematically studied. We trained three LDMs and a styleGAN2 model on histology tiles from nine colorectal cancer (CRC) tissue classes. The LDMs include 1) a fine-tuned version of stable diffusion v1.4, 2) a Kullback-Leibler (KL)-autoencoder (KLF8-DM), and 3) a vector quantized (VQ)-autoencoder deploying LDM (VQF8-DM). We assessed image quality through expert ratings, dimensional reduction methods, distribution similarity measures, and their impact on training a multiclass tissue classifier. Additionally, we investigated image memorization in the KLF8-DM and styleGAN2 models. All models provided a high image quality, with the KLF8-DM achieving the best Frechet Inception Distance (FID) and expert rating scores for complex tissue classes. For simpler classes, the VQF8-DM and styleGAN2 models performed better. Image memorization was negligible for both styleGAN2 and KLF8-DM models. Classifiers trained on a mix of KLF8-DM generated and real images achieved a 4% improvement in overall classification accuracy, highlighting the usefulness of these images for dataset augmentation. Our systematic study of generative methods showed that KLF8-DM produces the highest quality images with negligible image memorization. The higher classifier performance in the generatively augmented dataset suggests that this augmentation technique can be employed to enhance histopathology classifiers for various tasks.


Assuntos
Neoplasias Colorretais , Humanos , Neoplasias Colorretais/patologia , Neoplasias Colorretais/diagnóstico por imagem , Interpretação de Imagem Assistida por Computador/métodos , Processamento de Imagem Assistida por Computador/métodos , Algoritmos
16.
Comput Biol Med ; 175: 108502, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38678943

RESUMO

OBJECTIVES: Musculoskeletal (MSK) tumors, given their high mortality rate and heterogeneity, necessitate precise examination and diagnosis to guide clinical treatment effectively. Magnetic resonance imaging (MRI) is pivotal in detecting MSK tumors, as it offers exceptional image contrast between bone and soft tissue. This study aims to enhance the speed of detection and the diagnostic accuracy of MSK tumors through automated segmentation and grading utilizing MRI. MATERIALS AND METHODS: The research included 170 patients (mean age, 58 years ±12 (standard deviation), 84 men) with MSK lesions, who underwent MRI scans from April 2021 to May 2023. We proposed a deep learning (DL) segmentation model MSAPN based on multi-scale attention and pixel-level reconstruction, and compared it with existing algorithms. Using MSAPN-segmented lesions to extract their radiomic features for the benign and malignant classification of tumors. RESULTS: Compared to the most advanced segmentation algorithms, MSAPN demonstrates better performance. The Dice similarity coefficients (DSC) are 0.871 and 0.815 in the testing set and independent validation set, respectively. The radiomics model for classifying benign and malignant lesions achieves an accuracy of 0.890. Moreover, there is no statistically significant difference between the radiomics model based on manual segmentation and MSAPN segmentation. CONCLUSION: This research contributes to the advancement of MSK tumor diagnosis through automated segmentation and predictive classification. The integration of DL algorithms and radiomics shows promising results, and the visualization analysis of feature maps enhances clinical interpretability.


Assuntos
Neoplasias Ósseas , Aprendizado Profundo , Imageamento por Ressonância Magnética , Humanos , Masculino , Pessoa de Meia-Idade , Feminino , Imageamento por Ressonância Magnética/métodos , Idoso , Neoplasias Ósseas/diagnóstico por imagem , Neoplasias Ósseas/classificação , Algoritmos , Adulto , Interpretação de Imagem Assistida por Computador/métodos , Neoplasias Musculares/diagnóstico por imagem , Radiômica
17.
Artif Intell Med ; 152: 102871, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38685169

RESUMO

For the diagnosis and outcome prediction of gastric cancer (GC), machine learning methods based on whole slide pathological images (WSIs) have shown promising performance and reduced the cost of manual analysis. Nevertheless, accurate prediction of GC outcome may rely on multiple modalities with complementary information, particularly gene expression data. Thus, there is a need to develop multimodal learning methods to enhance prediction performance. In this paper, we collect a dataset from Ruijin Hospital and propose a multimodal learning method for GC diagnosis and outcome prediction, called GaCaMML, which is featured by a cross-modal attention mechanism and Per-Slide training scheme. Additionally, we perform feature attribution analysis via integrated gradient (IG) to identify important input features. The proposed method improves prediction accuracy over the single-modal learning method on three tasks, i.e., survival prediction (by 4.9% on C-index), pathological stage classification (by 11.6% on accuracy), and lymph node classification (by 12.0% on accuracy). Especially, the Per-Slide strategy addresses the issue of a high WSI-to-patient ratio and leads to much better results compared with the Per-Person training scheme. For the interpretable analysis, we find that although WSIs dominate the prediction for most samples, there is still a substantial portion of samples whose prediction highly relies on gene expression information. This study demonstrates the great potential of multimodal learning in GC-related prediction tasks and investigates the contribution of WSIs and gene expression, respectively, which not only shows how the model makes a decision but also provides insights into the association between macroscopic pathological phenotypes and microscopic molecular features.


Assuntos
Aprendizado de Máquina , Neoplasias Gástricas , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Humanos , Interpretação de Imagem Assistida por Computador/métodos , Prognóstico , Perfilação da Expressão Gênica/métodos
18.
Comput Biol Med ; 175: 108519, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38688128

RESUMO

Lung cancer has seriously threatened human health due to its high lethality and morbidity. Lung adenocarcinoma, in particular, is one of the most common subtypes of lung cancer. Pathological diagnosis is regarded as the gold standard for cancer diagnosis. However, the traditional manual screening of lung cancer pathology images is time consuming and error prone. Computer-aided diagnostic systems have emerged to solve this problem. Current research methods are unable to fully exploit the beneficial features inherent within patches, and they are characterized by high model complexity and significant computational effort. In this study, a deep learning framework called Multi-Scale Network (MSNet) is proposed for the automatic detection of lung adenocarcinoma pathology images. MSNet is designed to efficiently harness the valuable features within data patches, while simultaneously reducing model complexity, computational demands, and storage space requirements. The MSNet framework employs a dual data stream input method. In this input method, MSNet combines Swin Transformer and MLP-Mixer models to address global information between patches and the local information within each patch. Subsequently, MSNet uses the Multilayer Perceptron (MLP) module to fuse local and global features and perform classification to output the final detection results. In addition, a dataset of lung adenocarcinoma pathology images containing three categories is created for training and testing the MSNet framework. Experimental results show that the diagnostic accuracy of MSNet for lung adenocarcinoma pathology images is 96.55 %. In summary, MSNet has high classification performance and shows effectiveness and potential in the classification of lung adenocarcinoma pathology images.


Assuntos
Adenocarcinoma de Pulmão , Neoplasias Pulmonares , Redes Neurais de Computação , Humanos , Adenocarcinoma de Pulmão/diagnóstico por imagem , Adenocarcinoma de Pulmão/patologia , Adenocarcinoma de Pulmão/classificação , Neoplasias Pulmonares/diagnóstico por imagem , Neoplasias Pulmonares/patologia , Neoplasias Pulmonares/classificação , Aprendizado Profundo , Interpretação de Imagem Assistida por Computador/métodos , Diagnóstico por Computador/métodos
19.
J Comput Assist Tomogr ; 48(3): 343-353, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38595087

RESUMO

PURPOSE: Accurate quantification of liver iron concentration (LIC) can be achieved via magnetic resonance imaging (MRI). Maps of liver T2*/R2* are provided by commercially available, vendor-provided, 3-dimensional (3D) multiecho Dixon sequences and allow automated, inline postprocessing, which removes the need for manual curve fitting associated with conventional 2-dimensional (2D) gradient echo (GRE)-based postprocessing. The main goal of our study was to investigate the relationship among LIC estimates generated by 3D multiecho Dixon sequence to values generated by 2D GRE-based R2* relaxometry as the reference standard. METHODS: A retrospective review of patients who had undergone MRI scans for estimation of LIC with conventional T2* relaxometry and 3D multiecho Dixon sequences was performed. A 1.5 T scanner was used to acquire the magnetic resonance studies. Acquisition of standard multislice multiecho T2*-based sequences was performed, and R2* values with corresponding LIC were estimated. The comparison between R2* and corresponding LIC estimates obtained by the 2 methods was analyzed via the correlation coefficients and Bland-Altman difference plots. RESULTS: This study included 104 patients (51 male and 53 female patients) with 158 MRI scans. The mean age of the patients at the time of scan was 15.2 (SD, 8.8) years. There was a very strong correlation between the 2 LIC estimation methods for LIC values up to 3.2 mg/g (LIC quantitative multiecho Dixon [qDixon; from region of interest R2*] vs LIC GRE [in-house]: r = 0.83, P < 0.01; LIC qDixon [from segmentation volume R2*] vs LIC GRE [in-house]: r = 0.92, P < 0.01); and very weak correlation between the 2 methods at liver iron levels >7 mg/g. CONCLUSION: Three-dimensional-based multiecho Dixon technique can accurately measure LIC up to 7 mg/g and has the potential to replace 2D GRE-based relaxometry methods.


Assuntos
Imageamento Tridimensional , Sobrecarga de Ferro , Fígado , Imageamento por Ressonância Magnética , Humanos , Feminino , Masculino , Sobrecarga de Ferro/diagnóstico por imagem , Imageamento por Ressonância Magnética/métodos , Estudos Retrospectivos , Adulto , Imageamento Tridimensional/métodos , Fígado/diagnóstico por imagem , Pessoa de Meia-Idade , Adulto Jovem , Idoso , Interpretação de Imagem Assistida por Computador/métodos , Adolescente , Reprodutibilidade dos Testes , Ferro
20.
Comput Biol Med ; 175: 108368, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38663351

RESUMO

BACKGROUND: The issue of using deep learning to obtain accurate gross tumor volume (GTV) and metastatic lymph nodes (MLN) segmentation for nasopharyngeal carcinoma (NPC) on heterogeneous magnetic resonance imaging (MRI) images with limited labeling remains unsolved. METHOD: We collected 918 patients with MRI images from three hospitals to develop and validate models and proposed a semi-supervised framework for the fine delineation of multi-center NPC boundaries by integrating uncertainty-based implicit neural representations named SIMN. The framework utilizes the deep mutual learning approach with CNN and Transformer, incorporating dynamic thresholds. Additionally, domain adaptive algorithms are employed to enhance the performance. RESULTS: SIMN predictions have a high overlap ratio with the ground truth. Under the 20 % labeled cases, for the internal test cohorts, the average DSC in GTV and MLN are 0.7981 and 0.7804, respectively; for external test cohort Wu Zhou Red Cross Hospital, the average DSC in GTV and MLN are 0.7217 and 0.7581, respectively; for external test cohorts First People Hospital of Foshan, the average DSC in GTV and MLN are 0.7004 and 0.7692, respectively. No significant differences are found in DSC, HD95, ASD, and Recall for patients with different clinical categories. Moreover, SIMN outperformed existing classical semi-supervised methods. CONCLUSIONS: SIMN showed a highly accurate GTV and MLN segmentation for NPC on multi-center MRI images under Semi-Supervised Learning (SSL), which can easily transfer to other centers without fine-tuning. It suggests that it has the potential to act as a generalized delineation solution for heterogeneous MRI images with limited labels in clinical deployment.


Assuntos
Imageamento por Ressonância Magnética , Carcinoma Nasofaríngeo , Neoplasias Nasofaríngeas , Humanos , Imageamento por Ressonância Magnética/métodos , Carcinoma Nasofaríngeo/diagnóstico por imagem , Neoplasias Nasofaríngeas/diagnóstico por imagem , Masculino , Feminino , Pessoa de Meia-Idade , Adulto , Aprendizado Profundo , Algoritmos , Interpretação de Imagem Assistida por Computador/métodos , Redes Neurais de Computação
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA