首页 | 本学科首页   官方微博 | 高级检索  
相似文献
 共查询到20条相似文献,搜索用时 62 毫秒
1.
Deep convolutional neural networks have been highly effective in segmentation tasks. However, segmentation becomes more difficult when training images include many complex instances to segment, such as the task of nuclei segmentation in histopathology images. Weakly supervised learning can reduce the need for large-scale, high-quality ground truth annotations by involving non-expert annotators or algorithms to generate supervision information for segmentation. However, there is still a significant performance gap between weakly supervised learning and fully supervised learning approaches. In this work, we propose a weakly-supervised nuclei segmentation method in a two-stage training manner that only requires annotation of the nuclear centroids. First, we generate boundary and superpixel-based masks as pseudo ground truth labels to train our SAC-Net, which is a segmentation network enhanced by a constraint network and an attention network to effectively address the problems caused by noisy labels. Then, we refine the pseudo labels at the pixel level based on Confident Learning to train the network again. Our method shows highly competitive performance of cell nuclei segmentation in histopathology images on three public datasets. Code will be available at: https://github.com/RuoyuGuo/MaskGA_Net.  相似文献   

2.
Supervised deep learning-based methods yield accurate results for medical image segmentation. However, they require large labeled datasets for this, and obtaining them is a laborious task that requires clinical expertise. Semi/self-supervised learning-based approaches address this limitation by exploiting unlabeled data along with limited annotated data. Recent self-supervised learning methods use contrastive loss to learn good global level representations from unlabeled images and achieve high performance in classification tasks on popular natural image datasets like ImageNet. In pixel-level prediction tasks such as segmentation, it is crucial to also learn good local level representations along with global representations to achieve better accuracy. However, the impact of the existing local contrastive loss-based methods remains limited for learning good local representations because similar and dissimilar local regions are defined based on random augmentations and spatial proximity; not based on the semantic label of local regions due to lack of large-scale expert annotations in the semi/self-supervised setting. In this paper, we propose a local contrastive loss to learn good pixel level features useful for segmentation by exploiting semantic label information obtained from pseudo-labels of unlabeled images alongside limited annotated images with ground truth (GT) labels. In particular, we define the proposed contrastive loss to encourage similar representations for the pixels that have the same pseudo-label/GT label while being dissimilar to the representation of pixels with different pseudo-label/GT label in the dataset. We perform pseudo-label based self-training and train the network by jointly optimizing the proposed contrastive loss on both labeled and unlabeled sets and segmentation loss on only the limited labeled set. We evaluated the proposed approach on three public medical datasets of cardiac and prostate anatomies, and obtain high segmentation performance with a limited labeled set of one or two 3D volumes. Extensive comparisons with the state-of-the-art semi-supervised and data augmentation methods and concurrent contrastive learning methods demonstrate the substantial improvement achieved by the proposed method. The code is made publicly available at https://github.com/krishnabits001/pseudo_label_contrastive_training.  相似文献   

3.
Mitosis counting of biopsies is an important biomarker for breast cancer patients, which supports disease prognostication and treatment planning. Developing a robust mitotic cell detection model is highly challenging due to its complex growth pattern and high similarities with non-mitotic cells. Most mitosis detection algorithms have poor generalizability across image domains and lack reproducibility and validation in multicenter settings. To overcome these issues, we propose a generalizable and robust mitosis detection algorithm (called FMDet), which is independently tested on multicenter breast histopathological images. To capture more refined morphological features of cells, we convert the object detection task as a semantic segmentation problem. The pixel-level annotations for mitotic nuclei are obtained by taking the intersection of the masks generated from a well-trained nuclear segmentation model and the bounding boxes provided by the MIDOG 2021 challenge. In our segmentation framework, a robust feature extractor is developed to capture the appearance variations of mitotic cells, which is constructed by integrating a channel-wise multi-scale attention mechanism into a fully convolutional network structure. Benefiting from the fact that the changes in the low-level spectrum do not affect the high-level semantic perception, we employ a Fourier-based data augmentation method to reduce domain discrepancies by exchanging the low-frequency spectrum between two domains. Our FMDet algorithm has been tested in the MIDOG 2021 challenge and ranked first place. Further, our algorithm is also externally validated on four independent datasets for mitosis detection, which exhibits state-of-the-art performance in comparison with previously published results. These results demonstrate that our algorithm has the potential to be deployed as an assistant decision support tool in clinical practice. Our code has been released at https://github.com/Xiyue-Wang/1st-in-MICCAI-MIDOG-2021-challenge.  相似文献   

4.
Post-prostatectomy radiotherapy requires accurate annotation of the prostate bed (PB), i.e., the residual tissue after the operative removal of the prostate gland, to minimize side effects on surrounding organs-at-risk (OARs). However, PB segmentation in computed tomography (CT) images is a challenging task, even for experienced physicians. This is because PB is almost a “virtual” target with non-contrast boundaries and highly variable shapes depending on neighboring OARs. In this work, we propose an asymmetric multi-task attention network (AMTA-Net) for the concurrent segmentation of PB and surrounding OARs. Our AMTA-Net mimics experts in delineating the non-contrast PB by explicitly leveraging its critical dependency on the neighboring OARs (i.e., the bladder and rectum), which are relatively easy to distinguish in CT images. Specifically, we first adopt a U-Net as the backbone network for the low-level (or prerequisite) task of the OAR segmentation. Then, we build an attention sub-network upon the backbone U-Net with a series of cascaded attention modules, which can hierarchically transfer the OAR features and adaptively learn discriminative representations for the high-level (or primary) task of the PB segmentation. We comprehensively evaluate the proposed AMTA-Net on a clinical dataset composed of 186 CT images. According to the experimental results, our AMTA-Net significantly outperforms current clinical state-of-the-arts (i.e., atlas-based segmentation methods), indicating the value of our method in reducing time and labor in the clinical workflow. Our AMTA-Net also presents better performance than the technical state-of-the-arts (i.e., the deep learning-based segmentation methods), especially for the most indistinguishable and clinically critical part of the PB boundaries. Source code is released at https://github.com/superxuang/amta-net.  相似文献   

5.
Deep convolutional neural networks (CNNs) have been widely used for medical image segmentation. In most studies, only the output layer is exploited to compute the final segmentation results and the hidden representations of the deep learned features have not been well understood. In this paper, we propose a prototype segmentation (ProtoSeg) method to compute a binary segmentation map based on deep features. We measure the segmentation abilities of the features by computing the Dice between the feature segmentation map and ground-truth, named as the segmentation ability score (SA score for short). The corresponding SA score can quantify the segmentation abilities of deep features in different layers and units to understand the deep neural networks for segmentation. In addition, our method can provide a mean SA score which can give a performance estimation of the output on the test images without ground-truth. Finally, we use the proposed ProtoSeg method to compute the segmentation map directly on input images to further understand the segmentation ability of each input image. Results are presented on segmenting tumors in brain MRI, lesions in skin images, COVID-related abnormality in CT images, prostate segmentation in abdominal MRI, and pancreatic mass segmentation in CT images. Our method can provide new insights for interpreting and explainable AI systems for medical image segmentation. Our code is available on: https://github.com/shengfly/ProtoSeg.  相似文献   

6.
Automatic segmentation of coronary arteries provides vital assistance to enable accurate and efficient diagnosis and evaluation of coronary artery disease (CAD). However, the task of coronary artery segmentation (CAS) remains highly challenging due to the large-scale variations exhibited by coronary arteries, their complicated anatomical structures and morphologies, as well as the low contrast between vessels and their background. To comprehensively tackle these challenges, we propose a novel multi-attention, multi-scale 3D deep network for CAS, which we call CAS-Net. Specifically, we first propose an attention-guided feature fusion (AGFF) module to efficiently fuse adjacent hierarchical features in the encoding and decoding stages to capture more effectively latent semantic information. Then, we propose a scale-aware feature enhancement (SAFE) module, aiming to dynamically adjust the receptive fields to extract more expressive features effectively, thereby enhancing the feature representation capability of the network. Furthermore, we employ the multi-scale feature aggregation (MSFA) module to learn a more distinctive semantic representation for refining the vessel maps. In addition, considering that the limited training data annotated with a quality golden standard are also a significant factor restricting the development of CAS, we construct a new dataset containing 119 cases consisting of coronary computed tomographic angiography (CCTA) volumes and annotated coronary arteries. Extensive experiments on our self-collected dataset and three publicly available datasets demonstrate that the proposed method has good segmentation performance and generalization ability, outperforming multiple state-of-the-art algorithms on various metrics. Compared with U-Net3D, the proposed method significantly improves the Dice similarity coefficient (DSC) by at least 4% on each dataset, due to the synergistic effect among the three core modules, AGFF, SAFE, and MSFA. Our implementation is released at https://github.com/Cassie-CV/CAS-Net.  相似文献   

7.
Cells/nuclei deliver massive information of microenvironment. An automatic nuclei segmentation approach can reduce pathologists’ workload and allow precise of the microenvironment for biological and clinical researches. Existing deep learning models have achieved outstanding performance under the supervision of a large amount of labeled data. However, when data from the unseen domain comes, we still have to prepare a certain degree of manual annotations for training for each domain. Unfortunately, obtaining histopathological annotations is extremely difficult. It is high expertise-dependent and time-consuming. In this paper, we attempt to build a generalized nuclei segmentation model with less data dependency and more generalizability. To this end, we propose a meta multi-task learning (Meta-MTL) model for nuclei segmentation which requires fewer training samples. A model-agnostic meta-learning is applied as the outer optimization algorithm for the segmentation model. We introduce a contour-aware multi-task learning model as the inner model. A feature fusion and interaction block (FFIB) is proposed to allow feature communication across both tasks. Extensive experiments prove that our proposed Meta-MTL model can improve the model generalization and obtain a comparable performance with state-of-the-art models with fewer training samples. Our model can also perform fast adaptation on the unseen domain with only a few manual annotations. Code is available at https://github.com/ChuHan89/Meta-MTL4NucleiSegmentation  相似文献   

8.
Video feedback provides a wealth of information about surgical procedures and is the main sensory cue for surgeons. Scene understanding is crucial to computer assisted interventions (CAI) and to post-operative analysis of the surgical procedure. A fundamental building block of such capabilities is the identification and localization of surgical instruments and anatomical structures through semantic segmentation. Deep learning has advanced semantic segmentation techniques in the recent years but is inherently reliant on the availability of labelled datasets for model training. This paper introduces a dataset for semantic segmentation of cataract surgery videos complementing the publicly available CATARACTS challenge dataset. In addition, we benchmark the performance of several state-of-the-art deep learning models for semantic segmentation on the presented dataset. The dataset is publicly available at https://cataracts-semantic-segmentation2020.grand-challenge.org/.  相似文献   

9.
Tissue-level semantic segmentation is a vital step in computational pathology. Fully-supervised models have already achieved outstanding performance with dense pixel-level annotations. However, drawing such labels on the giga-pixel whole slide images is extremely expensive and time-consuming. In this paper, we use only patch-level classification labels to achieve tissue semantic segmentation on histopathology images, finally reducing the annotation efforts. We propose a two-step model including a classification and a segmentation phases. In the classification phase, we propose a CAM-based model to generate pseudo masks by patch-level labels. In the segmentation phase, we achieve tissue semantic segmentation by our propose Multi-Layer Pseudo-Supervision. Several technical novelties have been proposed to reduce the information gap between pixel-level and patch-level annotations. As a part of this paper, we introduce a new weakly-supervised semantic segmentation (WSSS) dataset for lung adenocarcinoma (LUAD-HistoSeg). We conduct several experiments to evaluate our proposed model on two datasets. Our proposed model outperforms five state-of-the-art WSSS approaches. Note that we can achieve comparable quantitative and qualitative results with the fully-supervised model, with only around a 2% gap for MIoU and FwIoU. By comparing with manual labeling on a randomly sampled 100 patches dataset, patch-level labeling can greatly reduce the annotation time from hours to minutes. The source code and the released datasets are available at: https://github.com/ChuHan89/WSSS-Tissue.  相似文献   

10.
Semantic image segmentation is an important prerequisite for context-awareness and autonomous robotics in surgery. The state of the art has focused on conventional RGB video data acquired during minimally invasive surgery, but full-scene semantic segmentation based on spectral imaging data and obtained during open surgery has received almost no attention to date. To address this gap in the literature, we are investigating the following research questions based on hyperspectral imaging (HSI) data of pigs acquired in an open surgery setting: (1) What is an adequate representation of HSI data for neural network-based fully automated organ segmentation, especially with respect to the spatial granularity of the data (pixels vs. superpixels vs. patches vs. full images)? (2) Is there a benefit of using HSI data compared to other modalities, namely RGB data and processed HSI data (e.g. tissue parameters like oxygenation), when performing semantic organ segmentation? According to a comprehensive validation study based on 506 HSI images from 20 pigs, annotated with a total of 19 classes, deep learning-based segmentation performance increases — consistently across modalities — with the spatial context of the input data. Unprocessed HSI data offers an advantage over RGB data or processed data from the camera provider, with the advantage increasing with decreasing size of the input to the neural network. Maximum performance (HSI applied to whole images) yielded a mean DSC of 0.90 ((standard deviation (SD)) 0.04), which is in the range of the inter-rater variability (DSC of 0.89 ((standard deviation (SD)) 0.07)). We conclude that HSI could become a powerful image modality for fully-automatic surgical scene understanding with many advantages over traditional imaging, including the ability to recover additional functional tissue information. Our code and pre-trained models are available at https://github.com/IMSY-DKFZ/htc.  相似文献   

11.
Paediatric echocardiography is a standard method for screening congenital heart disease (CHD). The segmentation of paediatric echocardiography is essential for subsequent extraction of clinical parameters and interventional planning. However, it remains a challenging task due to (1) the considerable variation of key anatomic structures, (2) the poor lateral resolution affecting accurate boundary definition, (3) the existence of speckle noise and artefacts in echocardiographic images. In this paper, we propose a novel deep network to address these challenges comprehensively. We first present a dual-path feature extraction module (DP-FEM) to extract rich features via a channel attention mechanism. A high- and low-level feature fusion module (HL-FFM) is devised based on spatial attention, which selectively fuses rich semantic information from high-level features with spatial cues from low-level features. In addition, a hybrid loss is designed to deal with pixel-level misalignment and boundary ambiguities. Based on the segmentation results, we derive key clinical parameters for diagnosis and treatment planning. We extensively evaluate the proposed method on 4,485 two-dimensional (2D) paediatric echocardiograms from 127 echocardiographic videos. The proposed method consistently achieves better segmentation performance than other state-of-the-art methods, whichdemonstratesfeasibility for automatic segmentation and quantitative analysis of paediatric echocardiography. Our code is publicly available at https://github.com/end-of-the-century/Cardiac.  相似文献   

12.
Glaucoma is one of the leading causes of irreversible but preventable blindness in working age populations. Color fundus photography (CFP) is the most cost-effective imaging modality to screen for retinal disorders. However, its application to glaucoma has been limited to the computation of a few related biomarkers such as the vertical cup-to-disc ratio. Deep learning approaches, although widely applied for medical image analysis, have not been extensively used for glaucoma assessment due to the limited size of the available data sets. Furthermore, the lack of a standardize benchmark strategy makes difficult to compare existing methods in a uniform way. In order to overcome these issues we set up the Retinal Fundus Glaucoma Challenge, REFUGE (https://refuge.grand-challenge.org), held in conjunction with MICCAI 2018. The challenge consisted of two primary tasks, namely optic disc/cup segmentation and glaucoma classification. As part of REFUGE, we have publicly released a data set of 1200 fundus images with ground truth segmentations and clinical glaucoma labels, currently the largest existing one. We have also built an evaluation framework to ease and ensure fairness in the comparison of different models, encouraging the development of novel techniques in the field. 12 teams qualified and participated in the online challenge. This paper summarizes their methods and analyzes their corresponding results. In particular, we observed that two of the top-ranked teams outperformed two human experts in the glaucoma classification task. Furthermore, the segmentation results were in general consistent with the ground truth annotations, with complementary outcomes that can be further exploited by ensembling the results.  相似文献   

13.
Breast cancer is one of the most common causes of death among women worldwide. Early signs of breast cancer can be an abnormality depicted on breast images (e.g., mammography or breast ultrasonography). However, reliable interpretation of breast images requires intensive labor and physicians with extensive experience. Deep learning is evolving breast imaging diagnosis by introducing a second opinion to physicians. However, most deep learning-based breast cancer analysis algorithms lack interpretability because of their black box nature, which means that domain experts cannot understand why the algorithms predict a label. In addition, most deep learning algorithms are formulated as a single-task-based model that ignores correlations between different tasks (e.g., tumor classification and segmentation). In this paper, we propose an interpretable multitask information bottleneck network (MIB-Net) to accomplish simultaneous breast tumor classification and segmentation. MIB-Net maximizes the mutual information between the latent representations and class labels while minimizing information shared by the latent representations and inputs. In contrast from existing models, our MIB-Net generates a contribution score map that offers an interpretable aid for physicians to understand the model’s decision-making process. In addition, MIB-Net implements multitask learning and further proposes a dual prior knowledge guidance strategy to enhance deep task correlation. Our evaluations are carried out on three breast image datasets in different modalities. Our results show that the proposed framework is not only able to help physicians better understand the model’s decisions but also improve breast tumor classification and segmentation accuracy over representative state-of-the-art models. Our code is available at https://github.com/jxw0810/MIB-Net.  相似文献   

14.
Automatic lesion segmentation on thoracic CT enables rapid quantitative analysis of lung involvement in COVID-19 infections. However, obtaining a large amount of voxel-level annotations for training segmentation networks is prohibitively expensive. Therefore, we propose a weakly-supervised segmentation method based on dense regression activation maps (dRAMs). Most weakly-supervised segmentation approaches exploit class activation maps (CAMs) to localize objects. However, because CAMs were trained for classification, they do not align precisely with the object segmentations. Instead, we produce high-resolution activation maps using dense features from a segmentation network that was trained to estimate a per-lobe lesion percentage. In this way, the network can exploit knowledge regarding the required lesion volume. In addition, we propose an attention neural network module to refine dRAMs, optimized together with the main regression task. We evaluated our algorithm on 90 subjects. Results show our method achieved 70.2% Dice coefficient, substantially outperforming the CAM-based baseline at 48.6%. We published our source code at https://github.com/DIAGNijmegen/bodyct-dram.  相似文献   

15.
In this work, we report the set-up and results of the Liver Tumor Segmentation Benchmark (LiTS), which was organized in conjunction with the IEEE International Symposium on Biomedical Imaging (ISBI) 2017 and the International Conferences on Medical Image Computing and Computer-Assisted Intervention (MICCAI) 2017 and 2018. The image dataset is diverse and contains primary and secondary tumors with varied sizes and appearances with various lesion-to-background levels (hyper-/hypo-dense), created in collaboration with seven hospitals and research institutions. Seventy-five submitted liver and liver tumor segmentation algorithms were trained on a set of 131 computed tomography (CT) volumes and were tested on 70 unseen test images acquired from different patients. We found that not a single algorithm performed best for both liver and liver tumors in the three events. The best liver segmentation algorithm achieved a Dice score of 0.963, whereas, for tumor segmentation, the best algorithms achieved Dices scores of 0.674 (ISBI 2017), 0.702 (MICCAI 2017), and 0.739 (MICCAI 2018). Retrospectively, we performed additional analysis on liver tumor detection and revealed that not all top-performing segmentation algorithms worked well for tumor detection. The best liver tumor detection method achieved a lesion-wise recall of 0.458 (ISBI 2017), 0.515 (MICCAI 2017), and 0.554 (MICCAI 2018), indicating the need for further research. LiTS remains an active benchmark and resource for research, e.g., contributing the liver-related segmentation tasks in http://medicaldecathlon.com/. In addition, both data and online evaluation are accessible via https://competitions.codalab.org/competitions/17094.  相似文献   

16.
Most image segmentation algorithms are trained on binary masks formulated as a classification task per pixel. However, in applications such as medical imaging, this “black-and-white” approach is too constraining because the contrast between two tissues is often ill-defined, i.e., the voxels located on objects’ edges contain a mixture of tissues (a partial volume effect). Consequently, assigning a single “hard” label can result in a detrimental approximation. Instead, a soft prediction containing non-binary values would overcome that limitation. In this study, we introduce SoftSeg, a deep learning training approach that takes advantage of soft ground truth labels, and is not bound to binary predictions. SoftSeg aims at solving a regression instead of a classification problem. This is achieved by using (i) no binarization after preprocessing and data augmentation, (ii) a normalized ReLU final activation layer (instead of sigmoid), and (iii) a regression loss function (instead of the traditional Dice loss). We assess the impact of these three features on three open-source MRI segmentation datasets from the spinal cord gray matter, the multiple sclerosis brain lesion, and the multimodal brain tumor segmentation challenges. Across multiple random dataset splittings, SoftSeg outperformed the conventional approach, leading to an increase in Dice score of 2.0% on the gray matter dataset (p=0.001), 3.3% for the brain lesions, and 6.5% for the brain tumors. SoftSeg produces consistent soft predictions at tissues’ interfaces and shows an increased sensitivity for small objects (e.g., multiple sclerosis lesions). The richness of soft labels could represent the inter-expert variability, the partial volume effect, and complement the model uncertainty estimation, which is typically unclear with binary predictions. The developed training pipeline can easily be incorporated into most of the existing deep learning architectures. SoftSeg is implemented in the freely-available deep learning toolbox ivadomed (https://ivadomed.org).  相似文献   

17.
Magnetic resonance imaging (MRI) is an increasingly important tool for the diagnosis and treatment of prostate cancer. However, interpretation of MRI suffers from high inter-observer variability across radiologists, thereby contributing to missed clinically significant cancers, overdiagnosed low-risk cancers, and frequent false positives. Interpretation of MRI could be greatly improved by providing radiologists with an answer key that clearly shows cancer locations on MRI. Registration of histopathology images from patients who had radical prostatectomy to pre-operative MRI allows such mapping of ground truth cancer labels onto MRI. However, traditional MRI-histopathology registration approaches are computationally expensive and require careful choices of the cost function and registration hyperparameters. This paper presents ProsRegNet, a deep learning-based pipeline to accelerate and simplify MRI-histopathology image registration in prostate cancer. Our pipeline consists of image preprocessing, estimation of affine and deformable transformations by deep neural networks, and mapping cancer labels from histopathology images onto MRI using estimated transformations. We trained our neural network using MR and histopathology images of 99 patients from our internal cohort (Cohort 1) and evaluated its performance using 53 patients from three different cohorts (an additional 12 from Cohort 1 and 41 from two public cohorts). Results show that our deep learning pipeline has achieved more accurate registration results and is at least 20 times faster than a state-of-the-art registration algorithm. This important advance will provide radiologists with highly accurate prostate MRI answer keys, thereby facilitating improvements in the detection of prostate cancer on MRI. Our code is freely available at https://github.com/pimed//ProsRegNet.  相似文献   

18.
High throughput nuclear segmentation and classification of whole slide images (WSIs) is crucial to biological analysis, clinical diagnosis and precision medicine. With the advances of CNN algorithms and the continuously growing datasets, considerable progress has been made in nuclear segmentation and classification. However, few works consider how to reasonably deal with nuclear heterogeneity in the following two aspects: imbalanced data distribution and diversified morphology characteristics. The minority classes might be dominated by the majority classes due to the imbalanced data distribution and the diversified morphology characteristics may lead to fragile segmentation results. In this study, a cost-Sensitive MultI-task LEarning (SMILE) framework is conducted to tackle the data heterogeneity problem. Based on the most popular multi-task learning backbone in nuclei segmentation and classification, we propose a multi-task correlation attention (MTCA) to perform feature interaction of multiple high relevant tasks to learn better feature representation. A cost-sensitive learning strategy is proposed to solve the imbalanced data distribution by increasing the penalization for the error classification of the minority classes. Furthermore, we propose a novel post-processing step based on the coarse-to-fine marker-controlled watershed scheme to alleviate fragile segmentation when nuclei are with large size and unclear contour. Extensive experiments show that the proposed method achieves state-of-the-art performances on CoNSeP and MoNuSAC 2020 datasets. The code is available at: https://github.com/panxipeng/nuclear_segandcls.  相似文献   

19.
In digital pathology, segmentation is a fundamental task for the diagnosis and treatment of diseases. Existing fully supervised methods often require accurate pixel-level annotations that are both time-consuming and laborious to generate. Typical approaches first pre-process histology images into patches to meet memory constraints and later perform stitching for segmentation; at times leading to lower performance given the lack of global context. Since image level labels are cheaper to acquire, weakly supervised learning is a more practical alternative for training segmentation algorithms. In this work, we present a weakly supervised framework for histopathology segmentation using only image-level labels by refining class activation maps (CAM) with self-supervision. First, we compress gigapixel histology images with an unsupervised contrastive learning technique to retain high-level spatial context. Second, a network is trained on the compressed images to jointly predict image-labels and refine the initial CAMs via self-supervised losses. In particular, we achieve refinement via a pixel correlation module (PCM) that leverages self-attention between the initial CAM and the input to encourage fine-grained activations. Also, we introduce a feature masking technique that performs spatial dropout on the compressed input to suppress low confidence predictions. To effectively train our model, we propose a loss function that includes a classification objective with image-labels, self-supervised regularization and entropy minimization between the CAM predictions. Experimental results on two curated datasets show that our approach is comparable to fully-supervised methods and can outperform existing state-of-the-art patch-based methods. https://github.com/PhilipChicco/wsshisto  相似文献   

20.
In this paper, we propose a novel mutual consistency network (MC-Net+) to effectively exploit the unlabeled data for semi-supervised medical image segmentation. The MC-Net+ model is motivated by the observation that deep models trained with limited annotations are prone to output highly uncertain and easily mis-classified predictions in the ambiguous regions (e.g., adhesive edges or thin branches) for medical image segmentation. Leveraging these challenging samples can make the semi-supervised segmentation model training more effective. Therefore, our proposed MC-Net+ model consists of two new designs. First, the model contains one shared encoder and multiple slightly different decoders (i.e., using different up-sampling strategies). The statistical discrepancy of multiple decoders’ outputs is computed to denote the model’s uncertainty, which indicates the unlabeled hard regions. Second, we apply a novel mutual consistency constraint between one decoder’s probability output and other decoders’ soft pseudo labels. In this way, we minimize the discrepancy of multiple outputs (i.e., the model uncertainty) during training and force the model to generate invariant results in such challenging regions, aiming at regularizing the model training. We compared the segmentation results of our MC-Net+ model with five state-of-the-art semi-supervised approaches on three public medical datasets. Extension experiments with two standard semi-supervised settings demonstrate the superior performance of our model over other methods, which sets a new state of the art for semi-supervised medical image segmentation. Our code is released publicly at https://github.com/ycwu1997/MC-Net.  相似文献   

设为首页 | 免责声明 | 关于勤云 | 加入收藏

Copyright©北京勤云科技发展有限公司  京ICP备09084417号