Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
IEEE Trans Cybern ; PP2024 May 10.
Artigo em Inglês | MEDLINE | ID: mdl-38728131

RESUMO

Radiation therapy treatment planning requires balancing the delivery of the target dose while sparing normal tissues, making it a complex process. To streamline the planning process and enhance its quality, there is a growing demand for knowledge-based planning (KBP). Ensemble learning has shown impressive power in various deep learning tasks, and it has great potential to improve the performance of KBP. However, the effectiveness of ensemble learning heavily depends on the diversity and individual accuracy of the base learners. Moreover, the complexity of model ensembles is a major concern, as it requires maintaining multiple models during inference, leading to increased computational cost and storage overhead. In this study, we propose a novel learning-based ensemble approach named LENAS, which integrates neural architecture search with knowledge distillation for 3-D radiotherapy dose prediction. Our approach starts by exhaustively searching each block from an enormous architecture space to identify multiple architectures that exhibit promising performance and significant diversity. To mitigate the complexity introduced by the model ensemble, we adopt the teacher-student paradigm, leveraging the diverse outputs from multiple learned networks as supervisory signals to guide the training of the student network. Furthermore, to preserve high-level semantic information, we design a hybrid loss to optimize the student network, enabling it to recover the knowledge embedded within the teacher networks. The proposed method has been evaluated on two public datasets: 1) OpenKBP and 2) AIMIS. Extensive experimental results demonstrate the effectiveness of our method and its superior performance to the state-of-the-art methods. Code: github.com/hust-linyi/LENAS.

2.
Artif Intell Med ; 149: 102801, 2024 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-38462290

RESUMO

Since different disease grades require different treatments from physicians, i.e., the low-grade patients may recover with follow-up observations whereas the high-grade may need immediate surgery, the accuracy of disease grading is pivotal in clinical practice. In this paper, we propose a Triplet-Branch Network with ContRastive priOr-knoWledge embeddiNg (TBN-CROWN) for the accurate disease grading, which enables physicians to accordingly take appropriate treatments. Specifically, our TBN-CROWN has three branches, which are implemented for representation learning, classifier learning and grade-related prior-knowledge learning, respectively. The former two branches deal with the issue of class-imbalanced training samples, while the latter one embeds the grade-related prior-knowledge via a novel auxiliary module, termed contrastive embedding module. The proposed auxiliary module takes the features embedded by different branches as input, and accordingly constructs positive and negative embeddings for the model to deploy grade-related prior-knowledge via contrastive learning. Extensive experiments on our private and two publicly available disease grading datasets show that our TBN-CROWN can effectively tackle the class-imbalance problem and yield a satisfactory grading accuracy for various diseases, such as fatigue fracture, ulcerative colitis, and diabetic retinopathy.


Assuntos
Retinopatia Diabética , Médicos , Humanos , Aprendizagem
3.
Med Image Anal ; 93: 103102, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38367598

RESUMO

Rare diseases are characterized by low prevalence and are often chronically debilitating or life-threatening. Imaging phenotype classification of rare diseases is challenging due to the severe shortage of training examples. Few-shot learning (FSL) methods tackle this challenge by extracting generalizable prior knowledge from a large base dataset of common diseases and normal controls and transferring the knowledge to rare diseases. Yet, most existing methods require the base dataset to be labeled and do not make full use of the precious examples of rare diseases. In addition, the extremely small size of the training samples may result in inter-class performance imbalance due to insufficient sampling of the true distributions. To this end, we propose in this work a novel hybrid approach to rare disease imaging phenotype classification, featuring three key novelties targeted at the above drawbacks. First, we adopt the unsupervised representation learning (URL) based on self-supervising contrastive loss, whereby to eliminate the overhead in labeling the base dataset. Second, we integrate the URL with pseudo-label supervised classification for effective self-distillation of the knowledge about the rare diseases, composing a hybrid approach taking advantage of both unsupervised and (pseudo-) supervised learning on the base dataset. Third, we use the feature dispersion to assess the intra-class diversity of training samples, to alleviate the inter-class performance imbalance via dispersion-aware correction. Experimental results of imaging phenotype classification of both simulated (skin lesions and cervical smears) and real clinical rare diseases (retinal diseases) show that our hybrid approach substantially outperforms existing FSL methods (including those using a fully supervised base dataset) via effective integration of the URL, pseudo-label driven self-distillation, and dispersion-aware imbalance correction, thus establishing a new state of the art.


Assuntos
Doenças Raras , Doenças Retinianas , Humanos , Fenótipo , Diagnóstico por Imagem
4.
Med Image Anal ; 93: 103095, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38310678

RESUMO

Segmenting prostate from magnetic resonance imaging (MRI) is a critical procedure in prostate cancer staging and treatment planning. Considering the nature of labeled data scarcity for medical images, semi-supervised learning (SSL) becomes an appealing solution since it can simultaneously exploit limited labeled data and a large amount of unlabeled data. However, SSL relies on the assumption that the unlabeled images are abundant, which may not be satisfied when the local institute has limited image collection capabilities. An intuitive solution is to seek support from other centers to enrich the unlabeled image pool. However, this further introduces data heterogeneity, which can impede SSL that works under identical data distribution with certain model assumptions. Aiming at this under-explored yet valuable scenario, in this work, we propose a separated collaborative learning (SCL) framework for semi-supervised prostate segmentation with multi-site unlabeled MRI data. Specifically, on top of the teacher-student framework, SCL exploits multi-site unlabeled data by: (i) Local learning, which advocates local distribution fitting, including the pseudo label learning that reinforces confirmation of low-entropy easy regions and the cyclic propagated real label learning that leverages class prototypes to regularize the distribution of intra-class features; (ii) External multi-site learning, which aims to robustly mine informative clues from external data, mainly including the local-support category mutual dependence learning, which takes the spirit that mutual information can effectively measure the amount of information shared by two variables even from different domains, and the stability learning under strong adversarial perturbations to enhance robustness to heterogeneity. Extensive experiments on prostate MRI data from six different clinical centers show that our method can effectively generalize SSL on multi-site unlabeled data and significantly outperform other semi-supervised segmentation methods. Besides, we validate the extensibility of our method on the multi-class cardiac MRI segmentation task with data from four different clinical centers.


Assuntos
Práticas Interdisciplinares , Neoplasias da Próstata , Masculino , Humanos , Próstata/diagnóstico por imagem , Neoplasias da Próstata/diagnóstico por imagem , Entropia , Imageamento por Ressonância Magnética
5.
Pattern Recognit ; 138: None, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37781685

RESUMO

Supervised machine learning methods have been widely developed for segmentation tasks in recent years. However, the quality of labels has high impact on the predictive performance of these algorithms. This issue is particularly acute in the medical image domain, where both the cost of annotation and the inter-observer variability are high. Different human experts contribute estimates of the "actual" segmentation labels in a typical label acquisition process, influenced by their personal biases and competency levels. The performance of automatic segmentation algorithms is limited when these noisy labels are used as the expert consensus label. In this work, we use two coupled CNNs to jointly learn, from purely noisy observations alone, the reliability of individual annotators and the expert consensus label distributions. The separation of the two is achieved by maximally describing the annotator's "unreliable behavior" (we call it "maximally unreliable") while achieving high fidelity with the noisy training data. We first create a toy segmentation dataset using MNIST and investigate the properties of the proposed algorithm. We then use three public medical imaging segmentation datasets to demonstrate our method's efficacy, including both simulated (where necessary) and real-world annotations: 1) ISBI2015 (multiple-sclerosis lesions); 2) BraTS (brain tumors); 3) LIDC-IDRI (lung abnormalities). Finally, we create a real-world multiple sclerosis lesion dataset (QSMSC at UCL: Queen Square Multiple Sclerosis Center at UCL, UK) with manual segmentations from 4 different annotators (3 radiologists with different level skills and 1 expert to generate the expert consensus label). In all datasets, our method consistently outperforms competing methods and relevant baselines, especially when the number of annotations is small and the amount of disagreement is large. The studies also reveal that the system is capable of capturing the complicated spatial characteristics of annotators' mistakes.

6.
Med Image Anal ; 88: 102880, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37413792

RESUMO

Semi-supervised learning has greatly advanced medical image segmentation since it effectively alleviates the need of acquiring abundant annotations from experts, wherein the mean-teacher model, known as a milestone of perturbed consistency learning, commonly serves as a standard and simple baseline. Inherently, learning from consistency can be regarded as learning from stability under perturbations. Recent improvement leans toward more complex consistency learning frameworks, yet, little attention is paid to the consistency target selection. Considering that the ambiguous regions from unlabeled data contain more informative complementary clues, in this paper, we improve the mean-teacher model to a novel ambiguity-consensus mean-teacher (AC-MT) model. Particularly, we comprehensively introduce and benchmark a family of plug-and-play strategies for ambiguous target selection from the perspectives of entropy, model uncertainty and label noise self-identification, respectively. Then, the estimated ambiguity map is incorporated into the consistency loss to encourage consensus between the two models' predictions in these informative regions. In essence, our AC-MT aims to find out the most worthwhile voxel-wise targets from the unlabeled data, and the model especially learns from the perturbed stability of these informative regions. The proposed methods are extensively evaluated on left atrium segmentation and brain tumor segmentation. Encouragingly, our strategies bring substantial improvement over recent state-of-the-art methods. The ablation study further demonstrates our hypothesis and shows impressive results under various extreme annotation conditions.


Assuntos
Benchmarking , Neoplasias Encefálicas , Humanos , Neoplasias Encefálicas/diagnóstico por imagem , Consenso , Entropia , Átrios do Coração , Aprendizado de Máquina Supervisionado , Processamento de Imagem Assistida por Computador
7.
IEEE Trans Med Imaging ; 42(10): 3000-3011, 2023 10.
Artigo em Inglês | MEDLINE | ID: mdl-37145949

RESUMO

Pathological primary tumor (pT) stage focuses on the infiltration degree of the primary tumor to surrounding tissues, which relates to the prognosis and treatment choices. The pT staging relies on the field-of-views from multiple magnifications in the gigapixel images, which makes pixel-level annotation difficult. Therefore, this task is usually formulated as a weakly supervised whole slide image (WSI) classification task with the slide-level label. Existing weakly-supervised classification methods mainly follow the multiple instance learning paradigm, which takes the patches from single magnification as the instances and extracts their morphological features independently. However, they cannot progressively represent the contextual information from multiple magnifications, which is critical for pT staging. Therefore, we propose a structure-aware hierarchical graph-based multi-instance learning framework (SGMF) inspired by the diagnostic process of pathologists. Specifically, a novel graph-based instance organization method is proposed, namely structure-aware hierarchical graph (SAHG), to represent the WSI. Based on that, we design a novel hierarchical attention-based graph representation (HAGR) network to capture the critical patterns for pT staging by learning cross-scale spatial features. Finally, the top nodes of SAHG are aggregated by a global attention layer for bag-level representation. Extensive studies on three large-scale multi-center pT staging datasets with two different cancer types demonstrate the effectiveness of SGMF, which outperforms state-of-the-art up to 5.6% in the F1 score.


Assuntos
Aprendizado Profundo , Processamento de Imagem Assistida por Computador
8.
IEEE Trans Med Imaging ; 42(8): 2348-2359, 2023 08.
Artigo em Inglês | MEDLINE | ID: mdl-37027635

RESUMO

Leukemia classification relies on a detailed cytomorphological examination of Bone Marrow (BM) smear. However, applying existing deep-learning methods to it is facing two significant limitations. Firstly, these methods require large-scale datasets with expert annotations at the cell level for good results and typically suffer from poor generalization. Secondly, they simply treat the BM cytomorphological examination as a multi-class cell classification task, thus failing to exploit the correlation among leukemia subtypes over different hierarchies. Therefore, BM cytomorphological estimation as a time-consuming and repetitive process still needs to be done manually by experienced cytologists. Recently, Multi-Instance Learning (MIL) has achieved much progress in data-efficient medical image processing, which only requires patient-level labels (which can be extracted from the clinical reports). In this paper, we propose a hierarchical MIL framework and equip it with Information Bottleneck (IB) to tackle the above limitations. First, to handle the patient-level label, our hierarchical MIL framework uses attention-based learning to identify cells with high diagnostic values for leukemia classification in different hierarchies. Then, following the information bottleneck principle, we propose a hierarchical IB to constrain and refine the representations of different hierarchies for better accuracy and generalization. By applying our framework to a large-scale childhood acute leukemia dataset with corresponding BM smear images and clinical reports, we show that it can identify diagnostic-related cells without the need for cell-level annotations and outperforms other comparison methods. Furthermore, the evaluation conducted on an independent test cohort demonstrates the high generalizability of our framework.


Assuntos
Aprendizado Profundo , Leucemia , Criança , Humanos , Aprendizado de Máquina , Processamento de Imagem Assistida por Computador , Leucemia/diagnóstico por imagem
9.
Med Image Anal ; 83: 102652, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36327654

RESUMO

Cancer region detection (CRD) and subtyping are two fundamental tasks in digital pathology image analysis. The development of data-driven models for CRD and subtyping on whole-slide images (WSIs) would mitigate the burden of pathologists and improve their accuracy in diagnosis. However, the existing models are facing two major limitations. Firstly, they typically require large-scale datasets with precise annotations, which contradicts with the original intention of reducing labor effort. Secondly, for the subtyping task, the non-cancerous regions are treated as the same as cancerous regions within a WSI, which confuses a subtyping model in its training process. To tackle the latter limitation, the previous research proposed to perform CRD first for ruling out the non-cancerous region, then train a subtyping model based on the remaining cancerous patches. However, separately training ignores the interaction of these two tasks, also leads to propagating the error of the CRD task to the subtyping task. To address these issues and concurrently improve the performance on both CRD and subtyping tasks, we propose a semi-supervised multi-task learning (MTL) framework for cancer classification. Our framework consists of a backbone feature extractor, two task-specific classifiers, and a weight control mechanism. The backbone feature extractor is shared by two task-specific classifiers, such that the interaction of CRD and subtyping tasks can be captured. The weight control mechanism preserves the sequential relationship of these two tasks and guarantees the error back-propagation from the subtyping task to the CRD task under the MTL framework. We train the overall framework in a semi-supervised setting, where datasets only involve small quantities of annotations produced by our minimal point-based (min-point) annotation strategy. Extensive experiments on four large datasets with different cancer types demonstrate the effectiveness of the proposed framework in both accuracy and generalization.


Assuntos
Neoplasias , Aprendizado de Máquina Supervisionado , Humanos , Cabeça , Neoplasias/diagnóstico por imagem
10.
JAMA Otolaryngol Head Neck Surg ; 148(7): 612-620, 2022 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-35588049

RESUMO

Importance: Otitis media with effusion (OME) is one of the most common causes of acquired conductive hearing loss (CHL). Persistent hearing loss is associated with poor childhood speech and language development and other adverse consequence. However, to obtain accurate and reliable hearing thresholds largely requires a high degree of cooperation from the patients. Objective: To predict CHL from otoscopic images using deep learning (DL) techniques and a logistic regression model based on tympanic membrane features. Design, Setting, and Participants: A retrospective diagnostic/prognostic study was conducted using 2790 otoscopic images obtained from multiple centers between January 2015 and November 2020. Participants were aged between 4 and 89 years. Of 1239 participants, there were 209 ears from children and adolescents (aged 4-18 years [16.87%]), 804 ears from adults (aged 18-60 years [64.89%]), and 226 ears from older people (aged >60 years, [18.24%]). Overall, 679 ears (54.8%) were from men. The 2790 otoscopic images were randomly assigned into a training set (2232 [80%]), and validation set (558 [20%]). The DL model was developed to predict an average air-bone gap greater than 10 dB. A logistic regression model was also developed based on otoscopic features. Main Outcomes and Measures: The performance of the DL model in predicting CHL was measured using the area under the receiver operating curve (AUC), accuracy, and F1 score (a measure of the quality of a classifier, which is the harmonic mean of precision and recall; a higher F1 score means better performance). In addition, these evaluation parameters were compared to results obtained from the logistic regression model and predictions made by three otologists. Results: The performance of the DL model in predicting CHL showed the AUC of 0.74, accuracy of 81%, and F1 score of 0.89. This was better than the results from the logistic regression model (ie, AUC of 0.60, accuracy of 76%, and F1 score of 0.82), and much improved on the performance of the 3 otologists; accuracy of 16%, 30%, 39%, and F1 scores of 0.09, 0.18, and 0.25, respectively. Furthermore, the DL model took 2.5 seconds to predict from 205 otoscopic images, whereas the 3 otologists spent 633 seconds, 645 seconds, and 692 seconds, respectively. Conclusions and Relevance: The model in this diagnostic/prognostic study provided greater accuracy in prediction of CHL in ears with OME than those obtained from the logistic regression model and otologists. This indicates great potential for the use of artificial intelligence tools to facilitate CHL evaluation when CHL is unable to be measured.


Assuntos
Aprendizado Profundo , Otite Média com Derrame , Otite Média , Adolescente , Adulto , Idoso , Idoso de 80 Anos ou mais , Inteligência Artificial , Criança , Pré-Escolar , Perda Auditiva Condutiva/diagnóstico , Perda Auditiva Condutiva/etiologia , Humanos , Masculino , Pessoa de Meia-Idade , Otite Média/complicações , Otite Média com Derrame/complicações , Otite Média com Derrame/diagnóstico por imagem , Estudos Retrospectivos , Adulto Jovem
11.
IEEE J Biomed Health Inform ; 26(7): 3174-3184, 2022 07.
Artigo em Inglês | MEDLINE | ID: mdl-35324450

RESUMO

Semi-supervised learning has substantially advanced medical image segmentation since it alleviates the heavy burden of acquiring the costly expert-examined annotations. Especially, the consistency-based approaches have attracted more attention for their superior performance, wherein the real labels are only utilized to supervise their paired images via supervised loss while the unlabeled images are exploited by enforcing the perturbation-based "unsupervised" consistency without explicit guidance from those real labels. However, intuitively, the expert-examined real labels contain more reliable supervision signals. Observing this, we ask an unexplored but interesting question: can we exploit the unlabeled data via explicit real label supervision for semi-supervised training? To this end, we discard the previous perturbation-based consistency but absorb the essence of non-parametric prototype learning. Based on the prototypical networks, we then propose a novel cyclic prototype consistency learning (CPCL) framework, which is constructed by a labeled-to-unlabeled (L2U) prototypical forward process and an unlabeled-to-labeled (U2L) backward process. Such two processes synergistically enhance the segmentation network by encouraging morediscriminative and compact features. In this way, our framework turns previous "unsupervised" consistency into new "supervised" consistency, obtaining the "all-around real label supervision" property of our method. Extensive experiments on brain tumor segmentation from MRI and kidney segmentation from CT images show that our CPCL can effectively exploit the unlabeled data and outperform other state-of-the-art semi-supervised medical image segmentation methods.


Assuntos
Neoplasias Encefálicas , Aprendizado de Máquina Supervisionado , Humanos , Rim , Imageamento por Ressonância Magnética
12.
Eur Radiol ; 32(2): 747-758, 2022 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-34417848

RESUMO

OBJECTIVES: The molecular subtyping of diffuse gliomas is important. The aim of this study was to establish predictive models based on preoperative multiparametric MRI. METHODS: A total of 1016 diffuse glioma patients were retrospectively collected from Beijing Tiantan Hospital. Patients were randomly divided into the training (n = 780) and validation (n = 236) sets. According to the 2016 WHO classification, diffuse gliomas can be classified into four binary classification tasks (tasks I-IV). Predictive models based on radiomics and deep convolutional neural network (DCNN) were developed respectively, and their performances were compared with receiver operating characteristic (ROC) curves. Additionally, the radiomics and DCNN features were visualized and compared with the t-distributed stochastic neighbor embedding technique and Spearman's correlation test. RESULTS: In the training set, areas under the curves (AUCs) of the DCNN models (ranging from 0.99 to 1.00) outperformed the radiomics models in all tasks, and the accuracies of the DCNN models (ranging from 0.90 to 0.94) outperformed the radiomics models in tasks I, II, and III. In the independent validation set, the accuracies of the DCNN models outperformed the radiomics models in all tasks (0.74-0.83), and the AUCs of the DCNN models (0.85-0.89) outperformed the radiomics models in tasks I, II, and III. DCNN features demonstrated more superior discriminative capability than the radiomics features in feature visualization analysis, and their general correlations were weak. CONCLUSIONS: Both the radiomics and DCNN models could preoperatively predict the molecular subtypes of diffuse gliomas, and the latter performed better in most circumstances. KEY POINTS: • The molecular subtypes of diffuse gliomas could be predicted with MRI. • Deep learning features tend to outperform radiomics features in large cohorts. • The correlation between the radiomics features and DCNN features was low.


Assuntos
Aprendizado Profundo , Glioma , Glioma/diagnóstico por imagem , Humanos , Imageamento por Ressonância Magnética , Espectroscopia de Ressonância Magnética , Estudos Retrospectivos
13.
IEEE Trans Med Imaging ; 41(3): 595-607, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34606453

RESUMO

Medical images from multicentres often suffer from the domain shift problem, which makes the deep learning models trained on one domain usually fail to generalize well to another. One of the potential solutions for the problem is the generative adversarial network (GAN), which has the capacity to translate images between different domains. Nevertheless, the existing GAN-based approaches are prone to fail at preserving image-objects in image-to-image (I2I) translation, which reduces their practicality on domain adaptation tasks. In this regard, a novel GAN (namely IB-GAN) is proposed to preserve image-objects during cross-domain I2I adaptation. Specifically, we integrate the information bottleneck constraint into the typical cycle-consistency-based GAN to discard the superfluous information (e.g., domain information) and maintain the consistency of disentangled content features for image-object preservation. The proposed IB-GAN is evaluated on three tasks-polyp segmentation using colonoscopic images, the segmentation of optic disc and cup in fundus images and the whole heart segmentation using multi-modal volumes. We show that the proposed IB-GAN can generate realistic translated images and remarkably boost the generalization of widely used segmentation networks (e.g., U-Net).


Assuntos
Processamento de Imagem Assistida por Computador , Disco Óptico , Fundo de Olho , Processamento de Imagem Assistida por Computador/métodos
14.
Med Image Anal ; 70: 102006, 2021 05.
Artigo em Inglês | MEDLINE | ID: mdl-33690025

RESUMO

Cervical cancer causes the fourth most cancer-related deaths of women worldwide. Early detection of cervical intraepithelial neoplasia (CIN) can significantly increase the survival rate of patients. World Health Organization (WHO) divided the CIN into three grades (CIN1, CIN2 and CIN3). In clinical practice, different CIN grades require different treatments. Although existing studies proposed computer aided diagnosis (CAD) systems for cervical cancer diagnosis, most of them are fail to perform accurate separation between CIN1 and CIN2/3, due to the similar appearances under colposcopy. To boost the accuracy of CAD systems, we construct a colposcopic image dataset for GRAding cervical intraepithelial Neoplasia with fine-grained lesion Description (GRAND). The dataset consists of colposcopic images collected from 8,604 patients along with the pathological reports. Additionally, we invite the experienced colposcopist to annotate two main clues, which are usually adopted for clinical diagnosis of CIN grade, i.e., texture of acetowhite epithelium (TAE) and appearance of blood vessel (ABV). A multi-rater model using the annotated clues is benchmarked for our dataset. The proposed framework contains several sub-networks (raters) to exploit the fine-grained lesion features TAE and ABV, respectively, by contrastive learning and a backbone network to extract the global information from colposcopic images. A comprehensive experiment is conducted on our GRAND dataset. The experimental results demonstrate the benefit of using additional lesion descriptions (TAE and ABV), which increases the CIN grading accuracy by over 10%. Furthermore, we conduct a human-machine confrontation to evaluate the potential of the proposed benchmark framework for clinical applications. Particularly, three colposcopists on different professional levels (intern, in-service and professional) are invited to compete with our benchmark framework by investigating a same extra test set-our framework achieves a comparable CIN grading accuracy to that of a professional colposcopist.


Assuntos
Displasia do Colo do Útero , Neoplasias do Colo do Útero , Benchmarking , Colposcopia , Feminino , Humanos , Gravidez , Neoplasias do Colo do Útero/diagnóstico por imagem , Displasia do Colo do Útero/diagnóstico por imagem
15.
J Cancer Res Clin Oncol ; 147(3): 821-833, 2021 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-32852634

RESUMO

PURPOSE: Microvascular invasion (MVI) is a valuable predictor of survival in hepatocellular carcinoma (HCC) patients. This study developed predictive models using eXtreme Gradient Boosting (XGBoost) and deep learning based on CT images to predict MVI preoperatively. METHODS: In total, 405 patients were included. A total of 7302 radiomic features and 17 radiological features were extracted by a radiomics feature extraction package and radiologists, respectively. We developed a XGBoost model based on radiomics features, radiological features and clinical variables and a three-dimensional convolutional neural network (3D-CNN) to predict MVI status. Next, we compared the efficacy of the two models. RESULTS: Of the 405 patients, 220 (54.3%) were MVI positive, and 185 (45.7%) were MVI negative. The areas under the receiver operating characteristic curves (AUROCs) of the Radiomics-Radiological-Clinical (RRC) Model and 3D-CNN Model in the training set were 0.952 (95% confidence interval (CI) 0.923-0.973) and 0.980 (95% CI 0.959-0.993), respectively (p = 0.14). The AUROCs of the RRC Model and 3D-CNN Model in the validation set were 0.887 (95% CI 0.797-0.947) and 0.906 (95% CI 0.821-0.960), respectively (p = 0.83). Based on the MVI status predicted by the RRC and 3D-CNN Models, the mean recurrence-free survival (RFS) was significantly better in the predicted MVI-negative group than that in the predicted MVI-positive group (RRC Model: 69.95 vs. 24.80 months, p < 0.001; 3D-CNN Model: 64.06 vs. 31.05 months, p = 0.027). CONCLUSION: The RRC Model and 3D-CNN models showed considerable efficacy in identifying MVI preoperatively. These machine learning models may facilitate decision-making in HCC treatment but requires further validation.


Assuntos
Carcinoma Hepatocelular/irrigação sanguínea , Aprendizado Profundo , Neoplasias Hepáticas/irrigação sanguínea , Carcinoma Hepatocelular/diagnóstico por imagem , Carcinoma Hepatocelular/patologia , Estudos de Coortes , Intervalo Livre de Doença , Feminino , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Neoplasias Hepáticas/patologia , Masculino , Microcirculação , Pessoa de Meia-Idade , Modelos Estatísticos , Neovascularização Patológica/diagnóstico por imagem , Neovascularização Patológica/patologia , Estudos Retrospectivos
16.
Med Image Anal ; 67: 101876, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33197863

RESUMO

Fully convolutional networks (FCNs) trained with abundant labeled data have been proven to be a powerful and efficient solution for medical image segmentation. However, FCNs often fail to achieve satisfactory results due to the lack of labelled data and significant variability of appearance in medical imaging. To address this challenging issue, this paper proposes a conjugate fully convolutional network (CFCN) where pairwise samples are input for capturing a rich context representation and guide each other with a fusion module. To avoid the overfitting problem introduced by intra-class heterogeneity and boundary ambiguity with a small number of training samples, we propose to explicitly exploit the prior information from the label space, termed as proxy supervision. We further extend the CFCN to a compact conjugate fully convolutional network (C2FCN), which just has one head for fitting the proxy supervision without incurring two additional branches of decoders fitting ground truth of the input pairs compared to CFCN. In the test phase, the segmentation probability is inferred by the learned logical relation implied in the proxy supervision. Quantitative evaluation on the Liver Tumor Segmentation (LiTS) and Combined (CT-MR) Healthy Abdominal Organ Segmentation (CHAOS) datasets shows that the proposed framework achieves a significant performance improvement on both binary segmentation and multi-category segmentation, especially with a limited amount of training data. The source code is available at https://github.com/renzhenwang/pairwise_segmentation.


Assuntos
Processamento de Imagem Assistida por Computador , Neoplasias Hepáticas , Diagnóstico por Imagem , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Probabilidade
17.
BMC Med ; 18(1): 406, 2020 12 22.
Artigo em Inglês | MEDLINE | ID: mdl-33349257

RESUMO

BACKGROUND: Colposcopy diagnosis and directed biopsy are the key components in cervical cancer screening programs. However, their performance is limited by the requirement for experienced colposcopists. This study aimed to develop and validate a Colposcopic Artificial Intelligence Auxiliary Diagnostic System (CAIADS) for grading colposcopic impressions and guiding biopsies. METHODS: Anonymized digital records of 19,435 patients were obtained from six hospitals across China. These records included colposcopic images, clinical information, and pathological results (gold standard). The data were randomly assigned (7:1:2) to a training and a tuning set for developing CAIADS and to a validation set for evaluating performance. RESULTS: The agreement between CAIADS-graded colposcopic impressions and pathology findings was higher than that of colposcopies interpreted by colposcopists (82.2% versus 65.9%, kappa 0.750 versus 0.516, p < 0.001). For detecting pathological high-grade squamous intraepithelial lesion or worse (HSIL+), CAIADS showed higher sensitivity than the use of colposcopies interpreted by colposcopists at either biopsy threshold (low-grade or worse 90.5%, 95% CI 88.9-91.4% versus 83.5%, 81.5-85.3%; high-grade or worse 71.9%, 69.5-74.2% versus 60.4%, 57.9-62.9%; all p < 0.001), whereas the specificities were similar (low-grade or worse 51.8%, 49.8-53.8% versus 52.0%, 50.0-54.1%; high-grade or worse 93.9%, 92.9-94.9% versus 94.9%, 93.9-95.7%; all p > 0.05). The CAIADS also demonstrated a superior ability in predicting biopsy sites, with a median mean-intersection-over-union (mIoU) of 0.758. CONCLUSIONS: The CAIADS has potential in assisting beginners and for improving the diagnostic quality of colposcopy and biopsy in the detection of cervical precancer/cancer.


Assuntos
Inteligência Artificial , Carcinoma de Células Escamosas/diagnóstico , Colposcopia/métodos , Detecção Precoce de Câncer/métodos , Neoplasias do Colo do Útero/diagnóstico , Adulto , Idoso , Biópsia/métodos , Biópsia/estatística & dados numéricos , Carcinoma de Células Escamosas/patologia , Carcinoma de Células Escamosas/prevenção & controle , China/epidemiologia , Colposcopia/estatística & dados numéricos , Confiabilidade dos Dados , Testes Diagnósticos de Rotina/métodos , Detecção Precoce de Câncer/estatística & dados numéricos , Feminino , Humanos , Pessoa de Meia-Idade , Gradação de Tumores/métodos , Valor Preditivo dos Testes , Gravidez , Reprodutibilidade dos Testes , Neoplasias do Colo do Útero/patologia , Neoplasias do Colo do Útero/prevenção & controle , Adulto Jovem
18.
IEEE Trans Med Imaging ; 39(12): 4174-4185, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32755853

RESUMO

Fully convolutional neural networks have made promising progress in joint liver and liver tumor segmentation. Instead of following the debates over 2D versus 3D networks (for example, pursuing the balance between large-scale 2D pretraining and 3D context), in this paper, we novelly identify the wide variation in the ratio between intra- and inter-slice resolutions as a crucial obstacle to the performance. To tackle the mismatch between the intra- and inter-slice information, we propose a slice-aware 2.5D network that emphasizes extracting discriminative features utilizing not only in-plane semantics but also out-of-plane coherence for each separate slice. Specifically, we present a slice-wise multi-input multi-output architecture to instantiate such a design paradigm, which contains a Multi-Branch Decoder (MD) with a Slice-centric Attention Block (SAB) for learning slice-specific features and a Densely Connected Dice (DCD) loss to regularize the inter-slice predictions to be coherent and continuous. Based on the aforementioned innovations, we achieve state-of-the-art results on the MICCAI 2017 Liver Tumor Segmentation (LiTS) dataset. Besides, we also test our model on the ISBI 2019 Segmentation of THoracic Organs at Risk (SegTHOR) dataset, and the result proves the robustness and generalizability of the proposed method in other segmentation tasks.


Assuntos
Processamento de Imagem Assistida por Computador , Neoplasias Hepáticas , Redes Neurais de Computação , Humanos , Neoplasias Hepáticas/diagnóstico por imagem , Órgãos em Risco
19.
Med Image Anal ; 64: 101746, 2020 08.
Artigo em Inglês | MEDLINE | ID: mdl-32544840

RESUMO

Due to the development of deep learning, an increasing number of research works have been proposed to establish automated analysis systems for 3D volumetric medical data to improve the quality of patient care. However, it is challenging to obtain a large number of annotated 3D medical data needed to train a neural network well, as such manual annotation by physicians is time consuming and laborious. Self-supervised learning is one of the potential solutions to mitigate the strong requirement of data annotation by deeply exploiting raw data information. In this paper, we propose a novel self-supervised learning framework for volumetric medical data. Specifically, we propose a pretext task, i.e., Rubik's cube+, to pre-train 3D neural networks. The pretext task involves three operations, namely cube ordering, cube rotating and cube masking, forcing networks to learn translation and rotation invariant features from the original 3D medical data, and tolerate the noise of the data at the same time. Compared to the strategy of training from scratch, fine-tuning from the Rubik's cube+ pre-trained weights can remarkablely boost the accuracy of 3D neural networks on various tasks, such as cerebral hemorrhage classification and brain tumor segmentation, without the use of extra data.


Assuntos
Neoplasias Encefálicas , Imageamento Tridimensional , Humanos , Redes Neurais de Computação
20.
IEEE Trans Med Imaging ; 39(11): 3403-3415, 2020 11.
Artigo em Inglês | MEDLINE | ID: mdl-32406830

RESUMO

Cervical cancer causes the fourth most cancer-related deaths of women worldwide. Early detection of cervical intraepithelial neoplasia (CIN) can significantly increase the survival rate of patients. In this paper, we propose a deep learning framework for the accurate identification of LSIL+ (including CIN and cervical cancer) using time-lapsed colposcopic images. The proposed framework involves two main components, i.e., key-frame feature encoding networks and feature fusion network. The features of the original (pre-acetic-acid) image and the colposcopic images captured at around 60s, 90s, 120s and 150s during the acetic acid test are encoded by the feature encoding networks. Several fusion approaches are compared, all of which outperform the existing automated cervical cancer diagnosis systems using a single time slot. A graph convolutional network with edge features (E-GCN) is found to be the most suitable fusion approach in our study, due to its excellent explainability consistent with the clinical practice. A large-scale dataset, containing time-lapsed colposcopic images from 7,668 patients, is collected from the collaborative hospital to train and validate our deep learning framework. Colposcopists are invited to compete with our computer-aided diagnosis system. The proposed deep learning framework achieves a classification accuracy of 78.33%-comparable to that of an in-service colposcopist-which demonstrates its potential to provide assistance in the realistic clinical scenario.


Assuntos
Displasia do Colo do Útero , Neoplasias do Colo do Útero , Colposcopia , Computadores , Feminino , Humanos , Gravidez , Imagem com Lapso de Tempo , Neoplasias do Colo do Útero/diagnóstico por imagem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA