首页 > 最新文献

Computerized Medical Imaging and Graphics最新文献

英文 中文
Unsupervised lung CT image registration via stochastic decomposition of deformation fields 通过随机分解变形场实现无监督肺部 CT 图像配准
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-07 DOI: 10.1016/j.compmedimag.2024.102397
Jing Zou , Youyi Song , Lihao Liu , Angelica I. Aviles-Rivero , Jing Qin

We address the problem of lung CT image registration, which underpins various diagnoses and treatments for lung diseases. The main crux of the problem is the large deformation that the lungs undergo during respiration. This physiological process imposes several challenges from a learning point of view. In this paper, we propose a novel training scheme, called stochastic decomposition, which enables deep networks to effectively learn such a difficult deformation field during lung CT image registration. The key idea is to stochastically decompose the deformation field, and supervise the registration by synthetic data that have the corresponding appearance discrepancy. The stochastic decomposition allows for revealing all possible decompositions of the deformation field. At the learning level, these decompositions can be seen as a prior to reduce the ill-posedness of the registration yielding to boost the performance. We demonstrate the effectiveness of our framework on Lung CT data. We show, through extensive numerical and visual results, that our technique outperforms existing methods.

肺部 CT 图像配准是各种肺部疾病诊断和治疗的基础,我们要解决的问题就是肺部 CT 图像配准。该问题的主要症结在于肺部在呼吸过程中会发生巨大变形。从学习的角度来看,这一生理过程带来了诸多挑战。在本文中,我们提出了一种名为随机分解的新型训练方案,它能让深度网络在肺部 CT 图像配准过程中有效学习这种困难的形变场。其关键思路是随机分解形变场,并通过具有相应外观差异的合成数据来监督配准。随机分解可以揭示形变场的所有可能分解。在学习层面上,这些分解可以被看作是一种先验,可以减少配准的不确定性,从而提高性能。我们在肺部 CT 数据上演示了我们框架的有效性。我们通过大量的数值和视觉结果表明,我们的技术优于现有的方法。
{"title":"Unsupervised lung CT image registration via stochastic decomposition of deformation fields","authors":"Jing Zou ,&nbsp;Youyi Song ,&nbsp;Lihao Liu ,&nbsp;Angelica I. Aviles-Rivero ,&nbsp;Jing Qin","doi":"10.1016/j.compmedimag.2024.102397","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102397","url":null,"abstract":"<div><p>We address the problem of lung CT image registration, which underpins various diagnoses and treatments for lung diseases. The main crux of the problem is the large deformation that the lungs undergo during respiration. This physiological process imposes several challenges from a learning point of view. In this paper, we propose a novel training scheme, called stochastic decomposition, which enables deep networks to effectively learn such a difficult deformation field during lung CT image registration. The key idea is to stochastically decompose the deformation field, and supervise the registration by synthetic data that have the corresponding appearance discrepancy. The stochastic decomposition allows for revealing all possible decompositions of the deformation field. At the learning level, these decompositions can be seen as a prior to reduce the ill-posedness of the registration yielding to boost the performance. We demonstrate the effectiveness of our framework on Lung CT data. We show, through extensive numerical and visual results, that our technique outperforms existing methods.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140910060","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Weakly-supervised preclinical tumor localization associated with survival prediction from lung cancer screening Chest X-ray images 弱监督临床前肿瘤定位与肺癌筛查胸部 X 光图像的生存预测相关联
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-07 DOI: 10.1016/j.compmedimag.2024.102395
Renato Hermoza , Jacinto C. Nascimento , Gustavo Carneiro

In this paper, we hypothesize that it is possible to localize image regions of preclinical tumors in a Chest X-ray (CXR) image by a weakly-supervised training of a survival prediction model using a dataset containing CXR images of healthy patients and their time-to-death label. These visual explanations can empower clinicians in early lung cancer detection and increase patient awareness of their susceptibility to the disease. To test this hypothesis, we train a censor-aware multi-class survival prediction deep learning classifier that is robust to imbalanced training, where classes represent quantized number of days for time-to-death prediction. Such multi-class model allows us to use post-hoc interpretability methods, such as Grad-CAM, to localize image regions of preclinical tumors. For the experiments, we propose a new benchmark based on the National Lung Cancer Screening Trial (NLST) dataset to test weakly-supervised preclinical tumor localization and survival prediction models, and results suggest that our proposed method shows state-of-the-art C-index survival prediction and weakly-supervised preclinical tumor localization results. To our knowledge, this constitutes a pioneer approach in the field that is able to produce visual explanations of preclinical events associated with survival prediction results.

在本文中,我们假设通过使用包含健康患者 CXR 图像及其死亡时间标签的数据集对生存预测模型进行弱监督训练,有可能在胸部 X 光(CXR)图像中定位临床前肿瘤的图像区域。这些可视化解释可以增强临床医生早期检测肺癌的能力,并提高患者对自身易感性的认识。为了验证这一假设,我们训练了一种对不平衡训练具有鲁棒性的审查器感知多类生存预测深度学习分类器,其中类代表了量化的死亡时间预测天数。这种多类模型允许我们使用事后可解释性方法(如 Grad-CAM)来定位临床前肿瘤的图像区域。在实验中,我们提出了一个基于国家肺癌筛查试验(NLST)数据集的新基准,以测试弱监督临床前肿瘤定位和生存预测模型,结果表明我们提出的方法显示了最先进的 C 指数生存预测和弱监督临床前肿瘤定位结果。据我们所知,这是该领域中能够对与生存预测结果相关的临床前事件进行可视化解释的开创性方法。
{"title":"Weakly-supervised preclinical tumor localization associated with survival prediction from lung cancer screening Chest X-ray images","authors":"Renato Hermoza ,&nbsp;Jacinto C. Nascimento ,&nbsp;Gustavo Carneiro","doi":"10.1016/j.compmedimag.2024.102395","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102395","url":null,"abstract":"<div><p>In this paper, we hypothesize that it is possible to localize image regions of preclinical tumors in a Chest X-ray (CXR) image by a weakly-supervised training of a survival prediction model using a dataset containing CXR images of healthy patients and their time-to-death label. These visual explanations can empower clinicians in early lung cancer detection and increase patient awareness of their susceptibility to the disease. To test this hypothesis, we train a censor-aware multi-class survival prediction deep learning classifier that is robust to imbalanced training, where classes represent quantized number of days for time-to-death prediction. Such multi-class model allows us to use post-hoc interpretability methods, such as Grad-CAM, to localize image regions of preclinical tumors. For the experiments, we propose a new benchmark based on the National Lung Cancer Screening Trial (NLST) dataset to test weakly-supervised preclinical tumor localization and survival prediction models, and results suggest that our proposed method shows state-of-the-art C-index survival prediction and weakly-supervised preclinical tumor localization results. To our knowledge, this constitutes a pioneer approach in the field that is able to produce visual explanations of preclinical events associated with survival prediction results.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0895611124000727/pdfft?md5=13bd653784bd57b091f5c80e427ca52e&pid=1-s2.0-S0895611124000727-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140901751","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
GNN-based structural information to improve DNN-based basal ganglia segmentation in children following early brain lesion 基于 GNN 的结构信息改进基于 DNN 的儿童早期脑损伤基底节区段划分
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-07 DOI: 10.1016/j.compmedimag.2024.102396
Patty Coupeau , Jean-Baptiste Fasquel , Lucie Hertz-Pannier , Mickaël Dinomais

Analyzing the basal ganglia following an early brain lesion is crucial due to their noteworthy role in sensory–motor functions. However, the segmentation of these subcortical structures on MRI is challenging in children and is further complicated by the presence of a lesion. Although current deep neural networks (DNN) perform well in segmenting subcortical brain structures in healthy brains, they lack robustness when faced with lesion variability, leading to structural inconsistencies. Given the established spatial organization of the basal ganglia, we propose enhancing the DNN-based segmentation through post-processing with a graph neural network (GNN). The GNN conducts node classification on graphs encoding both class probabilities and spatial information regarding the regions segmented by the DNN. In this study, we focus on neonatal arterial ischemic stroke (NAIS) in children. The approach is evaluated on both healthy children and children after NAIS using three DNN backbones: U-Net, UNETr, and MSGSE-Net. The results show an improvement in segmentation performance, with an increase in the median Dice score by up to 4% and a reduction in the median Hausdorff distance (HD) by up to 93% for healthy children (from 36.45 to 2.57) and up to 91% for children suffering from NAIS (from 40.64 to 3.50). The performance of the method is compared with atlas-based methods. Severe cases of neonatal stroke result in a decline in performance in the injured hemisphere, without negatively affecting the segmentation of the contra-injured hemisphere. Furthermore, the approach demonstrates resilience to small training datasets, a widespread challenge in the medical field, particularly in pediatrics and for rare pathologies.

由于基底节在感觉运动功能中的重要作用,因此在早期脑损伤后分析基底节至关重要。然而,在核磁共振成像上分割这些儿童皮层下结构具有挑战性,而且由于存在病变而变得更加复杂。虽然目前的深度神经网络(DNN)在分割健康大脑皮层下结构时表现良好,但在面对病变变异时却缺乏鲁棒性,导致结构不一致。鉴于基底节的空间组织已经确立,我们建议通过图神经网络(GNN)的后处理来增强基于 DNN 的分割。图神经网络对编码类别概率和 DNN 所分割区域空间信息的图进行节点分类。在本研究中,我们重点关注儿童新生儿动脉缺血性中风(NAIS)。我们使用三种 DNN 主干对健康儿童和新生儿动脉缺血性中风后的儿童进行了评估:U-Net、UNETr 和 MSGSE-Net。结果表明,该方法提高了分割性能,健康儿童的中位 Dice 分数提高了 4%,中位 Hausdorff 距离 (HD) 缩短了 93%(从 36.45 到 2.57),而 NAIS 患儿的中位 Hausdorff 距离缩短了 91%(从 40.64 到 3.50)。该方法的性能与基于地图集的方法进行了比较。严重的新生儿中风会导致受伤半球的性能下降,但不会对受伤半球的分割产生负面影响。此外,该方法还显示出对小规模训练数据集的适应能力,这在医学领域是一个普遍的挑战,尤其是在儿科和罕见病症方面。
{"title":"GNN-based structural information to improve DNN-based basal ganglia segmentation in children following early brain lesion","authors":"Patty Coupeau ,&nbsp;Jean-Baptiste Fasquel ,&nbsp;Lucie Hertz-Pannier ,&nbsp;Mickaël Dinomais","doi":"10.1016/j.compmedimag.2024.102396","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102396","url":null,"abstract":"<div><p>Analyzing the basal ganglia following an early brain lesion is crucial due to their noteworthy role in sensory–motor functions. However, the segmentation of these subcortical structures on MRI is challenging in children and is further complicated by the presence of a lesion. Although current deep neural networks (DNN) perform well in segmenting subcortical brain structures in healthy brains, they lack robustness when faced with lesion variability, leading to structural inconsistencies. Given the established spatial organization of the basal ganglia, we propose enhancing the DNN-based segmentation through post-processing with a graph neural network (GNN). The GNN conducts node classification on graphs encoding both class probabilities and spatial information regarding the regions segmented by the DNN. In this study, we focus on neonatal arterial ischemic stroke (NAIS) in children. The approach is evaluated on both healthy children and children after NAIS using three DNN backbones: U-Net, UNETr, and MSGSE-Net. The results show an improvement in segmentation performance, with an increase in the median Dice score by up to 4% and a reduction in the median Hausdorff distance (HD) by up to 93% for healthy children (from 36.45 to 2.57) and up to 91% for children suffering from NAIS (from 40.64 to 3.50). The performance of the method is compared with atlas-based methods. Severe cases of neonatal stroke result in a decline in performance in the injured hemisphere, without negatively affecting the segmentation of the contra-injured hemisphere. Furthermore, the approach demonstrates resilience to small training datasets, a widespread challenge in the medical field, particularly in pediatrics and for rare pathologies.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-07","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140914459","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Leveraging a realistic synthetic database to learn Shape-from-Shading for estimating the colon depth in colonoscopy images 利用逼真的合成数据库学习阴影形状,以估计结肠镜图像中的结肠深度
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-03 DOI: 10.1016/j.compmedimag.2024.102390
Josué Ruano , Martín Gómez , Eduardo Romero , Antoine Manzanera

Colonoscopy is the choice procedure to diagnose, screening, and treat the colon and rectum cancer, from early detection of small precancerous lesions (polyps), to confirmation of malign masses. However, the high variability of the organ appearance and the complex shape of both the colon wall and structures of interest make this exploration difficult. Learned visuospatial and perceptual abilities mitigate technical limitations in clinical practice by proper estimation of the intestinal depth. This work introduces a novel methodology to estimate colon depth maps in single frames from monocular colonoscopy videos. The generated depth map is inferred from the shading variation of the colon wall with respect to the light source, as learned from a realistic synthetic database. Briefly, a classic convolutional neural network architecture is trained from scratch to estimate the depth map, improving sharp depth estimations in haustral folds and polyps by a custom loss function that minimizes the estimation error in edges and curvatures. The network was trained by a custom synthetic colonoscopy database herein constructed and released, composed of 248 400 frames (47 videos), with depth annotations at the level of pixels. This collection comprehends 5 subsets of videos with progressively higher levels of visual complexity. Evaluation of the depth estimation with the synthetic database reached a threshold accuracy of 95.65%, and a mean-RMSE of 0.451cm, while a qualitative assessment with a real database showed consistent depth estimations, visually evaluated by the expert gastroenterologist coauthoring this paper. Finally, the method achieved competitive performance with respect to another state-of-the-art method using a public synthetic database and comparable results in a set of images with other five state-of-the-art methods. Additionally, three-dimensional reconstructions demonstrated useful approximations of the gastrointestinal tract geometry. Code for reproducing the reported results and the dataset are available at https://github.com/Cimalab-unal/ColonDepthEstimation.

从早期发现小的癌前病变(息肉)到确认恶性肿块,结肠镜检查是诊断、筛查和治疗结肠癌和直肠癌的首选方法。然而,由于器官外观的高度可变性以及结肠壁和相关结构的复杂形状,使得这项探索工作十分困难。学习视觉空间和感知能力可以通过正确估计肠道深度来缓解临床实践中的技术限制。这项工作介绍了一种从单眼结肠镜视频的单帧中估算结肠深度图的新方法。生成的深度图是根据现实合成数据库中结肠壁相对于光源的阴影变化推断出来的。简而言之,从头开始训练经典的卷积神经网络架构来估算深度图,并通过自定义损失函数来改善褶皱和息肉的锐利深度估算,使边缘和曲率的估算误差最小化。该网络由一个定制的合成结肠镜数据库训练而成,该数据库由 248 400 个帧组成(47 个视频),并带有像素级深度注释。该数据库包含 5 个视频子集,其视觉复杂度逐步提高。通过合成数据库对深度估计进行评估,阈值准确率达到 95.65%,平均均方根误差为 0.451 厘米,而通过真实数据库进行的定性评估显示,深度估计的一致性得到了本文合著者之一的胃肠病专家的直观评价。最后,该方法与另一种使用公共合成数据库的先进方法相比,性能具有竞争力,在一组图像中与其他五种先进方法的结果也不相上下。此外,三维重建显示了胃肠道几何形状的近似值。重现报告结果的代码和数据集可在 https://github.com/Cimalab-unal/ColonDepthEstimation 上获取。
{"title":"Leveraging a realistic synthetic database to learn Shape-from-Shading for estimating the colon depth in colonoscopy images","authors":"Josué Ruano ,&nbsp;Martín Gómez ,&nbsp;Eduardo Romero ,&nbsp;Antoine Manzanera","doi":"10.1016/j.compmedimag.2024.102390","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102390","url":null,"abstract":"<div><p>Colonoscopy is the choice procedure to diagnose, screening, and treat the colon and rectum cancer, from early detection of small precancerous lesions (polyps), to confirmation of malign masses. However, the high variability of the organ appearance and the complex shape of both the colon wall and structures of interest make this exploration difficult. Learned visuospatial and perceptual abilities mitigate technical limitations in clinical practice by proper estimation of the intestinal depth. This work introduces a novel methodology to estimate colon depth maps in single frames from monocular colonoscopy videos. The generated depth map is inferred from the shading variation of the colon wall with respect to the light source, as learned from a realistic synthetic database. Briefly, a classic convolutional neural network architecture is trained from scratch to estimate the depth map, improving sharp depth estimations in haustral folds and polyps by a custom loss function that minimizes the estimation error in edges and curvatures. The network was trained by a custom synthetic colonoscopy database herein constructed and released, composed of 248<!--> <!-->400 frames (47 videos), with depth annotations at the level of pixels. This collection comprehends 5 subsets of videos with progressively higher levels of visual complexity. Evaluation of the depth estimation with the synthetic database reached a threshold accuracy of 95.65%, and a mean-RMSE of <span><math><mrow><mn>0</mn><mo>.</mo><mn>451</mn><mi>cm</mi></mrow></math></span>, while a qualitative assessment with a real database showed consistent depth estimations, visually evaluated by the expert gastroenterologist coauthoring this paper. Finally, the method achieved competitive performance with respect to another state-of-the-art method using a public synthetic database and comparable results in a set of images with other five state-of-the-art methods. Additionally, three-dimensional reconstructions demonstrated useful approximations of the gastrointestinal tract geometry. Code for reproducing the reported results and the dataset are available at <span>https://github.com/Cimalab-unal/ColonDepthEstimation</span><svg><path></path></svg>.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0895611124000673/pdfft?md5=bb898a4ca8669cb2b3cd2808af60a2b6&pid=1-s2.0-S0895611124000673-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140842801","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Towards a unified approach for unsupervised brain MRI Motion Artefact Detection with few shot Anomaly Detection 实现无监督脑磁共振成像运动伪影检测与少量异常检测的统一方法
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-03 DOI: 10.1016/j.compmedimag.2024.102391
Niamh Belton , Misgina Tsighe Hagos , Aonghus Lawlor , Kathleen M. Curran

Automated Motion Artefact Detection (MAD) in Magnetic Resonance Imaging (MRI) is a field of study that aims to automatically flag motion artefacts in order to prevent the requirement for a repeat scan. In this paper, we identify and tackle the three current challenges in the field of automated MAD; (1) reliance on fully-supervised training, meaning they require specific examples of Motion Artefacts (MA), (2) inconsistent use of benchmark datasets across different works and use of private datasets for testing and training of newly proposed MAD techniques and (3) a lack of sufficiently large datasets for MRI MAD. To address these challenges, we demonstrate how MAs can be identified by formulating the problem as an unsupervised Anomaly Detection (AD) task. We compare the performance of three State-of-the-Art AD algorithms DeepSVDD, Interpolated Gaussian Descriptor and FewSOME on two open-source Brain MRI datasets on the task of MAD and MA severity classification, with FewSOME achieving a MAD AUC >90% on both datasets and a Spearman Rank Correlation Coefficient of 0.8 on the task of MA severity classification. These models are trained in the few shot setting, meaning large Brain MRI datasets are not required to build robust MAD algorithms. This work also sets a standard protocol for testing MAD algorithms on open-source benchmark datasets. In addition to addressing these challenges, we demonstrate how our proposed ‘anomaly-aware’ scoring function improves FewSOME’s MAD performance in the setting where one and two shots of the anomalous class are available for training. Code available at https://github.com/niamhbelton/Unsupervised-Brain-MRI-Motion-Artefact-Detection/.

磁共振成像(MRI)中的运动伪影自动检测(MAD)是一个研究领域,旨在自动标记运动伪影,以避免重复扫描。在本文中,我们确定并解决了自动运动伪影识别领域当前面临的三大挑战:(1)依赖于完全监督训练,这意味着它们需要运动伪影(MA)的特定示例;(2)不同研究中基准数据集的使用不一致,以及使用私人数据集对新提出的运动伪影识别技术进行测试和训练;(3)缺乏足够大的磁共振成像运动伪影识别数据集。为了应对这些挑战,我们演示了如何通过将问题表述为无监督异常检测(AD)任务来识别 MA。我们在两个开源脑磁共振成像数据集上比较了 DeepSVDD、插值高斯描述符和 FewSOME 这三种最新 AD 算法在 MAD 和 MA 严重程度分类任务上的性能,其中 FewSOME 在两个数据集上的 MAD AUC 均达到 90%,在 MA 严重程度分类任务上的斯皮尔曼等级相关系数达到 0.8。这些模型是在少数几个镜头的设置中训练出来的,这意味着不需要大型脑磁共振成像数据集就能建立稳健的 MAD 算法。这项工作还为在开源基准数据集上测试 MAD 算法制定了标准协议。除了应对这些挑战外,我们还展示了我们提出的 "异常感知 "评分函数如何提高 FewSOME 在有一个和两个异常类镜头可用于训练的情况下的 MAD 性能。代码见 https://github.com/niamhbelton/Unsupervised-Brain-MRI-Motion-Artefact-Detection/。
{"title":"Towards a unified approach for unsupervised brain MRI Motion Artefact Detection with few shot Anomaly Detection","authors":"Niamh Belton ,&nbsp;Misgina Tsighe Hagos ,&nbsp;Aonghus Lawlor ,&nbsp;Kathleen M. Curran","doi":"10.1016/j.compmedimag.2024.102391","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102391","url":null,"abstract":"<div><p>Automated Motion Artefact Detection (MAD) in Magnetic Resonance Imaging (MRI) is a field of study that aims to automatically flag motion artefacts in order to prevent the requirement for a repeat scan. In this paper, we identify and tackle the three current challenges in the field of automated MAD; (1) reliance on fully-supervised training, meaning they require specific examples of Motion Artefacts (MA), (2) inconsistent use of benchmark datasets across different works and use of private datasets for testing and training of newly proposed MAD techniques and (3) a lack of sufficiently large datasets for MRI MAD. To address these challenges, we demonstrate how MAs can be identified by formulating the problem as an unsupervised Anomaly Detection (AD) task. We compare the performance of three State-of-the-Art AD algorithms DeepSVDD, Interpolated Gaussian Descriptor and FewSOME on two open-source Brain MRI datasets on the task of MAD and MA severity classification, with FewSOME achieving a MAD AUC <span><math><mrow><mo>&gt;</mo><mn>90</mn><mtext>%</mtext></mrow></math></span> on both datasets and a Spearman Rank Correlation Coefficient of 0.8 on the task of MA severity classification. These models are trained in the few shot setting, meaning large Brain MRI datasets are not required to build robust MAD algorithms. This work also sets a standard protocol for testing MAD algorithms on open-source benchmark datasets. In addition to addressing these challenges, we demonstrate how our proposed ‘anomaly-aware’ scoring function improves FewSOME’s MAD performance in the setting where one and two shots of the anomalous class are available for training. Code available at <span>https://github.com/niamhbelton/Unsupervised-Brain-MRI-Motion-Artefact-Detection/</span><svg><path></path></svg>.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0895611124000685/pdfft?md5=8275185e5cfc03cae6d8bed048a27239&pid=1-s2.0-S0895611124000685-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140844149","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
3DFRINet: A Framework for the Detection and Diagnosis of Fracture Related Infection in Low Extremities Based on 18F-FDG PET/CT 3D Images 3DFRINet:基于 18F-FDG PET/CT 3D 图像的低位肢体骨折相关感染检测和诊断框架
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-03 DOI: 10.1016/j.compmedimag.2024.102394
Chengfan Li , Liangbing Nie , Zhenkui Sun , Xuehai Ding , Quanyong Luo , Chentian Shen

Fracture related infection (FRI) is one of the most devastating complications after fracture surgery in the lower extremities, which can lead to extremely high morbidity and medical costs. Therefore, early comprehensive evaluation and accurate diagnosis of patients are critical for appropriate treatment, prevention of complications, and good prognosis. 18Fluoro-deoxyglucose positron emission tomography/computed tomography (18F-FDG PET/CT) is one of the most commonly used medical imaging modalities for diagnosing FRI. With the development of deep learning, more neural networks have been proposed and become powerful computer-aided diagnosis tools in medical imaging. Therefore, a fully automated two-stage framework for FRI detection and diagnosis, 3DFRINet (Three Dimension FRI Network), is proposed for 18F-FDG PET/CT 3D imaging. The first stage can effectively extract and fuse the features of both modalities to accurately locate the lesion by the dual-branch design and attention module. The second stage reduces the dimensionality of the image by using the maximum intensity projection, which retains the effective features while reducing the computational effort and achieving excellent diagnostic performance. The diagnostic performance of lesions reached 91.55% accuracy, 0.9331 AUC, and 0.9250 F1 score. 3DFRINet has an advantage over six nuclear medicine experts in each classification metric. The statistical analysis shows that 3DFRINet is equivalent or superior to the primary nuclear medicine physicians and comparable to the senior nuclear medicine physicians. In conclusion, this study first proposed a method based on 18F-FDG PET/CT three-dimensional imaging for FRI location and diagnosis. This method shows superior lesion detection rate and diagnostic efficiency and therefore has good prospects for clinical application.

骨折相关感染(FRI)是下肢骨折手术后最具破坏性的并发症之一,可导致极高的发病率和医疗费用。因此,及早对患者进行全面评估和准确诊断对于适当治疗、预防并发症和良好预后至关重要。18氟脱氧葡萄糖正电子发射断层扫描/计算机断层扫描(18F-FDG PET/CT)是诊断FRI最常用的医学影像模式之一。随着深度学习的发展,越来越多的神经网络被提出并成为医学影像领域强大的计算机辅助诊断工具。因此,针对 18F-FDG PET/CT 三维成像,提出了一种两阶段全自动 FRI 检测和诊断框架--3DFRINet(三维 FRI 网络)。第一阶段通过双分支设计和注意力模块,有效提取和融合两种模式的特征,准确定位病灶。第二阶段利用最大强度投影降低图像维度,在保留有效特征的同时减少了计算量,实现了出色的诊断性能。病变诊断准确率达到 91.55%,AUC 为 0.9331,F1 得分为 0.9250。与六位核医学专家相比,3DFRINet 在各项分类指标上均有优势。统计分析表明,3DFRINet 与初级核医学医师相当或更胜一筹,与高级核医学医师相当。总之,本研究首次提出了一种基于 18F-FDG PET/CT 三维成像的 FRI 定位和诊断方法。该方法病灶检出率高,诊断效率高,具有良好的临床应用前景。
{"title":"3DFRINet: A Framework for the Detection and Diagnosis of Fracture Related Infection in Low Extremities Based on 18F-FDG PET/CT 3D Images","authors":"Chengfan Li ,&nbsp;Liangbing Nie ,&nbsp;Zhenkui Sun ,&nbsp;Xuehai Ding ,&nbsp;Quanyong Luo ,&nbsp;Chentian Shen","doi":"10.1016/j.compmedimag.2024.102394","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102394","url":null,"abstract":"<div><p>Fracture related infection (FRI) is one of the most devastating complications after fracture surgery in the lower extremities, which can lead to extremely high morbidity and medical costs. Therefore, early comprehensive evaluation and accurate diagnosis of patients are critical for appropriate treatment, prevention of complications, and good prognosis. <sup>18</sup>Fluoro-deoxyglucose positron emission tomography/computed tomography (<sup>18</sup>F-FDG PET/CT) is one of the most commonly used medical imaging modalities for diagnosing FRI. With the development of deep learning, more neural networks have been proposed and become powerful computer-aided diagnosis tools in medical imaging. Therefore, a fully automated two-stage framework for FRI detection and diagnosis, 3DFRINet (Three Dimension FRI Network), is proposed for <sup>18</sup>F-FDG PET/CT 3D imaging. The first stage can effectively extract and fuse the features of both modalities to accurately locate the lesion by the dual-branch design and attention module. The second stage reduces the dimensionality of the image by using the maximum intensity projection, which retains the effective features while reducing the computational effort and achieving excellent diagnostic performance. The diagnostic performance of lesions reached 91.55% accuracy, 0.9331 AUC, and 0.9250 F1 score. 3DFRINet has an advantage over six nuclear medicine experts in each classification metric. The statistical analysis shows that 3DFRINet is equivalent or superior to the primary nuclear medicine physicians and comparable to the senior nuclear medicine physicians. In conclusion, this study first proposed a method based on <sup>18</sup>F-FDG PET/CT three-dimensional imaging for FRI location and diagnosis. This method shows superior lesion detection rate and diagnostic efficiency and therefore has good prospects for clinical application.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140844148","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
CAVE: Cerebral artery–vein segmentation in digital subtraction angiography CAVE:数字减影血管造影中的脑动脉-静脉分割
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-01 DOI: 10.1016/j.compmedimag.2024.102392
Ruisheng Su , P. Matthijs van der Sluijs , Yuan Chen , Sandra Cornelissen , Ruben van den Broek , Wim H. van Zwam , Aad van der Lugt , Wiro J. Niessen , Danny Ruijters , Theo van Walsum

Cerebral X-ray digital subtraction angiography (DSA) is a widely used imaging technique in patients with neurovascular disease, allowing for vessel and flow visualization with high spatio-temporal resolution. Automatic artery–vein segmentation in DSA plays a fundamental role in vascular analysis with quantitative biomarker extraction, facilitating a wide range of clinical applications. The widely adopted U-Net applied on static DSA frames often struggles with disentangling vessels from subtraction artifacts. Further, it falls short in effectively separating arteries and veins as it disregards the temporal perspectives inherent in DSA. To address these limitations, we propose to simultaneously leverage spatial vasculature and temporal cerebral flow characteristics to segment arteries and veins in DSA. The proposed network, coined CAVE, encodes a 2D+time DSA series using spatial modules, aggregates all the features using temporal modules, and decodes it into 2D segmentation maps. On a large multi-center clinical dataset, CAVE achieves a vessel segmentation Dice of 0.84 (±0.04) and an artery–vein segmentation Dice of 0.79 (±0.06). CAVE surpasses traditional Frangi-based k-means clustering (P < 0.001) and U-Net (P < 0.001) by a significant margin, demonstrating the advantages of harvesting spatio-temporal features. This study represents the first investigation into automatic artery–vein segmentation in DSA using deep learning. The code is publicly available at https://github.com/RuishengSu/CAVE_DSA.

脑 X 射线数字减影血管造影术(DSA)是一种广泛应用于神经血管疾病患者的成像技术,可实现高时空分辨率的血管和血流可视化。DSA 中的自动动脉血管分割在血管分析和定量生物标记物提取中起着基础性作用,有助于广泛的临床应用。在静态 DSA 帧上广泛采用的 U-Net 通常难以将血管与减影伪影分离。此外,由于它忽略了 DSA 固有的时间视角,因此无法有效分离动脉和静脉。为了解决这些局限性,我们建议同时利用空间血管和时间脑流特征来分割 DSA 中的动脉和静脉。我们提出的网络被称为 CAVE,它使用空间模块对二维+时间 DSA 序列进行编码,使用时间模块对所有特征进行聚合,并将其解码为二维分割图。在一个大型多中心临床数据集上,CAVE 的血管分割 Dice 为 0.84(±0.04),动脉-静脉分割 Dice 为 0.79(±0.06)。CAVE 显著超越了传统的基于 Frangi 的 k-means 聚类(P < 0.001)和 U-Net(P < 0.001),显示了采集时空特征的优势。本研究是首次利用深度学习对 DSA 中的动脉血管进行自动分割的研究。代码可在 https://github.com/RuishengSu/CAVE_DSA 公开获取。
{"title":"CAVE: Cerebral artery–vein segmentation in digital subtraction angiography","authors":"Ruisheng Su ,&nbsp;P. Matthijs van der Sluijs ,&nbsp;Yuan Chen ,&nbsp;Sandra Cornelissen ,&nbsp;Ruben van den Broek ,&nbsp;Wim H. van Zwam ,&nbsp;Aad van der Lugt ,&nbsp;Wiro J. Niessen ,&nbsp;Danny Ruijters ,&nbsp;Theo van Walsum","doi":"10.1016/j.compmedimag.2024.102392","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102392","url":null,"abstract":"<div><p>Cerebral X-ray digital subtraction angiography (DSA) is a widely used imaging technique in patients with neurovascular disease, allowing for vessel and flow visualization with high spatio-temporal resolution. Automatic artery–vein segmentation in DSA plays a fundamental role in vascular analysis with quantitative biomarker extraction, facilitating a wide range of clinical applications. The widely adopted U-Net applied on static DSA frames often struggles with disentangling vessels from subtraction artifacts. Further, it falls short in effectively separating arteries and veins as it disregards the temporal perspectives inherent in DSA. To address these limitations, we propose to simultaneously leverage spatial vasculature and temporal cerebral flow characteristics to segment arteries and veins in DSA. The proposed network, coined CAVE, encodes a 2D+time DSA series using spatial modules, aggregates all the features using temporal modules, and decodes it into 2D segmentation maps. On a large multi-center clinical dataset, CAVE achieves a vessel segmentation Dice of 0.84 (<span><math><mo>±</mo></math></span>0.04) and an artery–vein segmentation Dice of 0.79 (<span><math><mo>±</mo></math></span>0.06). CAVE surpasses traditional Frangi-based <span><math><mi>k</mi></math></span>-means clustering (P <span><math><mo>&lt;</mo></math></span> 0.001) and U-Net (P <span><math><mo>&lt;</mo></math></span> 0.001) by a significant margin, demonstrating the advantages of harvesting spatio-temporal features. This study represents the first investigation into automatic artery–vein segmentation in DSA using deep learning. The code is publicly available at <span>https://github.com/RuishengSu/CAVE_DSA</span><svg><path></path></svg>.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S0895611124000697/pdfft?md5=b8c9ddb6b9334a5a30392653d4a487b2&pid=1-s2.0-S0895611124000697-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140842800","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Cross-modality cerebrovascular segmentation based on pseudo-label generation via paired data 基于配对数据生成伪标签的跨模态脑血管分割
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-05-01 DOI: 10.1016/j.compmedimag.2024.102393
Zhanqiang Guo , Jianjiang Feng , Wangsheng Lu , Yin Yin , Guangming Yang , Jie Zhou

Accurate segmentation of cerebrovascular structures from Computed Tomography Angiography (CTA), Magnetic Resonance Angiography (MRA), and Digital Subtraction Angiography (DSA) is crucial for clinical diagnosis of cranial vascular diseases. Recent advancements in deep Convolution Neural Network (CNN) have significantly improved the segmentation process. However, training segmentation networks for all modalities requires extensive data labeling for each modality, which is often expensive and time-consuming. To circumvent this limitation, we introduce an approach to train cross-modality cerebrovascular segmentation network based on paired data from source and target domains. Our approach involves training a universal vessel segmentation network with manually labeled source domain data, which automatically produces initial labels for target domain training images. We improve the initial labels of target domain training images by fusing paired images, which are then used to refine the target domain segmentation network. A series of experimental arrangements is presented to assess the efficacy of our method in various practical application scenarios. The experiments conducted on an MRA-CTA dataset and a DSA-CTA dataset demonstrate that the proposed method is effective for cross-modality cerebrovascular segmentation and achieves state-of-the-art performance.

从计算机断层扫描血管造影 (CTA)、磁共振血管造影 (MRA) 和数字减影血管造影 (DSA) 中准确分割脑血管结构对于颅脑血管疾病的临床诊断至关重要。深度卷积神经网络(CNN)的最新进展极大地改进了分割过程。然而,训练所有模式的分割网络需要对每种模式进行大量数据标注,这通常既昂贵又耗时。为了规避这一限制,我们引入了一种基于源域和目标域的配对数据来训练跨模态脑血管分割网络的方法。我们的方法包括用人工标注的源域数据训练通用血管分割网络,然后自动生成目标域训练图像的初始标签。我们通过融合配对图像来改进目标域训练图像的初始标签,然后利用这些标签来完善目标域分割网络。我们提出了一系列实验安排,以评估我们的方法在各种实际应用场景中的功效。在 MRA-CTA 数据集和 DSA-CTA 数据集上进行的实验表明,所提出的方法对跨模态脑血管分割非常有效,并达到了最先进的性能。
{"title":"Cross-modality cerebrovascular segmentation based on pseudo-label generation via paired data","authors":"Zhanqiang Guo ,&nbsp;Jianjiang Feng ,&nbsp;Wangsheng Lu ,&nbsp;Yin Yin ,&nbsp;Guangming Yang ,&nbsp;Jie Zhou","doi":"10.1016/j.compmedimag.2024.102393","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102393","url":null,"abstract":"<div><p>Accurate segmentation of cerebrovascular structures from Computed Tomography Angiography (CTA), Magnetic Resonance Angiography (MRA), and Digital Subtraction Angiography (DSA) is crucial for clinical diagnosis of cranial vascular diseases. Recent advancements in deep Convolution Neural Network (CNN) have significantly improved the segmentation process. However, training segmentation networks for all modalities requires extensive data labeling for each modality, which is often expensive and time-consuming. To circumvent this limitation, we introduce an approach to train cross-modality cerebrovascular segmentation network based on paired data from source and target domains. Our approach involves training a universal vessel segmentation network with manually labeled source domain data, which automatically produces initial labels for target domain training images. We improve the initial labels of target domain training images by fusing paired images, which are then used to refine the target domain segmentation network. A series of experimental arrangements is presented to assess the efficacy of our method in various practical application scenarios. The experiments conducted on an MRA-CTA dataset and a DSA-CTA dataset demonstrate that the proposed method is effective for cross-modality cerebrovascular segmentation and achieves state-of-the-art performance.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-05-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140822866","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Motion correction and super-resolution for multi-slice cardiac magnetic resonance imaging via an end-to-end deep learning approach 通过端到端深度学习方法实现多切片心脏磁共振成像的运动校正和超分辨率
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-04-29 DOI: 10.1016/j.compmedimag.2024.102389
Zhennong Chen, Hui Ren, Quanzheng Li, Xiang Li

Accurate reconstruction of a high-resolution 3D volume of the heart is critical for comprehensive cardiac assessments. However, cardiac magnetic resonance (CMR) data is usually acquired as a stack of 2D short-axis (SAX) slices, which suffers from the inter-slice misalignment due to cardiac motion and data sparsity from large gaps between SAX slices. Therefore, we aim to propose an end-to-end deep learning (DL) model to address these two challenges simultaneously, employing specific model components for each challenge. The objective is to reconstruct a high-resolution 3D volume of the heart (VHR) from acquired CMR SAX slices (VLR). We define the transformation from VLR to VHR as a sequential process of motion correction and super-resolution. Accordingly, our DL model incorporates two distinct components. The first component conducts motion correction by predicting displacement vectors to re-position each SAX slice accurately. The second component takes the motion-corrected SAX slices from the first component and performs the super-resolution to fill the data gaps. These two components operate in a sequential way, and the entire model is trained end-to-end. Our model significantly reduced inter-slice misalignment from originally 3.33±0.74 mm to 1.36±0.63 mm and generated accurate high resolution 3D volumes with Dice of 0.974±0.010 for left ventricle (LV) and 0.938±0.017 for myocardium in a simulation dataset. When compared to the LAX contours in a real-world dataset, our model achieved Dice of 0.945±0.023 for LV and 0.786±0.060 for myocardium. In both datasets, our model with specific components for motion correction and super-resolution significantly enhance the performance compared to the model without such design considerations. The codes for our model are available at https://github.com/zhennongchen/CMR_MC_SR_End2End.

准确重建心脏的高分辨率三维容积对于全面的心脏评估至关重要。然而,心脏磁共振(CMR)数据通常是以二维短轴(SAX)切片堆叠的形式获取的,这就会受到心脏运动造成的切片间错位和 SAX 切片间巨大间隙造成的数据稀疏的影响。因此,我们旨在提出一种端到端的深度学习(DL)模型,同时应对这两个挑战,并针对每个挑战采用特定的模型组件。我们的目标是从获取的 CMR SAX 切片(VLR)重建高分辨率的三维心脏容积(VHR)。我们将从 VLR 到 VHR 的转换定义为运动校正和超分辨率的连续过程。因此,我们的 DL 模型包含两个不同的组件。第一个组件通过预测位移向量来进行运动校正,从而准确地重新定位每个 SAX 切片。第二个组件从第一个组件中获取经过运动校正的 SAX 切片,并执行超分辨率以填补数据空白。这两个组件以顺序的方式运行,整个模型是端到端的训练。我们的模型大大减少了切片间的错位,从原来的 3.33±0.74 毫米减少到 1.36±0.63 毫米,并在模拟数据集中生成了精确的高分辨率三维体积,左心室(LV)的 Dice 为 0.974±0.010,心肌的 Dice 为 0.938±0.017。与真实世界数据集中的 LAX 轮廓相比,我们的模型对左心室的 Dice 值为 0.945±0.023,对心肌的 Dice 值为 0.786±0.060。在这两个数据集中,与没有考虑运动校正和超分辨率的模型相比,我们的模型包含了运动校正和超分辨率的特定组件,大大提高了性能。我们的模型代码见 https://github.com/zhennongchen/CMR_MC_SR_End2End。
{"title":"Motion correction and super-resolution for multi-slice cardiac magnetic resonance imaging via an end-to-end deep learning approach","authors":"Zhennong Chen,&nbsp;Hui Ren,&nbsp;Quanzheng Li,&nbsp;Xiang Li","doi":"10.1016/j.compmedimag.2024.102389","DOIUrl":"https://doi.org/10.1016/j.compmedimag.2024.102389","url":null,"abstract":"<div><p>Accurate reconstruction of a high-resolution 3D volume of the heart is critical for comprehensive cardiac assessments. However, cardiac magnetic resonance (CMR) data is usually acquired as a stack of 2D short-axis (SAX) slices, which suffers from the inter-slice misalignment due to cardiac motion and data sparsity from large gaps between SAX slices. Therefore, we aim to propose an end-to-end deep learning (DL) model to address these two challenges simultaneously, employing specific model components for each challenge. The objective is to reconstruct a high-resolution 3D volume of the heart (<span><math><msub><mrow><mi>V</mi></mrow><mrow><mi>HR</mi></mrow></msub></math></span>) from acquired CMR SAX slices (<span><math><msub><mrow><mi>V</mi></mrow><mrow><mi>LR</mi></mrow></msub></math></span>). We define the transformation from <span><math><msub><mrow><mi>V</mi></mrow><mrow><mi>LR</mi></mrow></msub></math></span> to <span><math><msub><mrow><mi>V</mi></mrow><mrow><mi>HR</mi></mrow></msub></math></span> as a sequential process of motion correction and super-resolution. Accordingly, our DL model incorporates two distinct components. The first component conducts motion correction by predicting displacement vectors to re-position each SAX slice accurately. The second component takes the motion-corrected SAX slices from the first component and performs the super-resolution to fill the data gaps. These two components operate in a sequential way, and the entire model is trained end-to-end. Our model significantly reduced inter-slice misalignment from originally 3.33<span><math><mo>±</mo></math></span>0.74 mm to 1.36<span><math><mo>±</mo></math></span>0.63 mm and generated accurate high resolution 3D volumes with Dice of 0.974<span><math><mo>±</mo></math></span>0.010 for left ventricle (LV) and 0.938<span><math><mo>±</mo></math></span>0.017 for myocardium in a simulation dataset. When compared to the LAX contours in a real-world dataset, our model achieved Dice of 0.945<span><math><mo>±</mo></math></span>0.023 for LV and 0.786<span><math><mo>±</mo></math></span>0.060 for myocardium. In both datasets, our model with specific components for motion correction and super-resolution significantly enhance the performance compared to the model without such design considerations. The codes for our model are available at <span>https://github.com/zhennongchen/CMR_MC_SR_End2End</span><svg><path></path></svg>.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140816871","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A deep learning-based pipeline for developing multi-rib shape generative model with populational percentiles or anthropometrics as predictors 基于深度学习的管道,用于开发以人口百分位数或人体测量学为预测指标的多肋形状生成模型
IF 5.7 2区 医学 Q1 Medicine Pub Date : 2024-04-25 DOI: 10.1016/j.compmedimag.2024.102388
Yuan Huang , Sven A. Holcombe , Stewart C. Wang , Jisi Tang

Rib cross-sectional shapes (characterized by the outer contour and cortical bone thickness) affect the rib mechanical response under impact loading, thereby influence the rib injury pattern and risk. A statistical description of the rib shapes or their correlations to anthropometrics is a prerequisite to the development of numerical human body models representing target demographics. Variational autoencoders (VAE) as anatomical shape generators remain to be explored in terms of utilizing the latent vectors to control or interpret the representativeness of the generated results. In this paper, we propose a pipeline for developing a multi-rib cross-sectional shape generative model from CT images, which consists of the achievement of rib cross-sectional shape data from CT images using an anatomical indexing system and regular grids, and a unified framework to fit shape distributions and associate shapes to anthropometrics for different rib categories. Specifically, we collected CT images including 3193 ribs, surface regular grid is generated for each rib based on anatomical coordinates, the rib cross-sectional shapes are characterized by nodal coordinates and cortical bone thickness. The tensor structure of shape data based on regular grids enable the implementation of CNNs in the conditional variational autoencoder (CVAE). The CVAE is trained against an auxiliary classifier to decouple the low-dimensional representations of the inter- and intra- variations and fit each intra-variation by a Gaussian distribution simultaneously. Random tree regressors are further leveraged to associate each continuous intra-class space with the corresponding anthropometrics of the subjects, i.e., age, height and weight. As a result, with the rib class labels and the latent vectors sampled from Gaussian distributions or predicted from anthropometrics as the inputs, the decoder can generate valid rib cross-sectional shapes of given class labels (male/female, 2nd to 11th ribs) for arbitrary populational percentiles or specific age, height and weight, which paves the road for future biomedical and biomechanical studies considering the diversity of rib shapes across the population.

肋骨横截面形状(以外轮廓和皮质骨厚度为特征)会影响肋骨在冲击负荷下的机械响应,从而影响肋骨损伤模式和风险。对肋骨形状或其与人体测量学的相关性进行统计描述,是开发代表目标人群的数字人体模型的先决条件。作为解剖形状生成器的变异自动编码器(VAE)在利用潜在向量控制或解释生成结果的代表性方面仍有待探索。在本文中,我们提出了一种从 CT 图像开发多肋骨横截面形状生成模型的方法,其中包括使用解剖索引系统和规则网格从 CT 图像中获取肋骨横截面形状数据,以及一个统一的框架来拟合不同肋骨类别的形状分布并将形状与人体测量学相关联。具体来说,我们收集了包括 3193 根肋骨在内的 CT 图像,根据解剖坐标为每根肋骨生成表面规则网格,通过节点坐标和皮质骨厚度表征肋骨横截面形状。基于规则网格的形状数据张量结构使 CNN 能够在条件变异自动编码器(CVAE)中实现。CVAE 根据辅助分类器进行训练,以解耦内部和内部变异的低维表示,并同时用高斯分布拟合每个内部变异。进一步利用随机树回归器,将每个连续的类内空间与受试者的相应人体测量数据(即年龄、身高和体重)关联起来。因此,有了肋骨类标签和从高斯分布采样或从人体测量预测的潜向量作为输入,解码器就能为任意人口百分位数或特定年龄、身高和体重生成给定类标签(男性/女性,第 2 至第 11 肋)的有效肋骨横截面形状,这为考虑到整个人群肋骨形状多样性的未来生物医学和生物力学研究铺平了道路。
{"title":"A deep learning-based pipeline for developing multi-rib shape generative model with populational percentiles or anthropometrics as predictors","authors":"Yuan Huang ,&nbsp;Sven A. Holcombe ,&nbsp;Stewart C. Wang ,&nbsp;Jisi Tang","doi":"10.1016/j.compmedimag.2024.102388","DOIUrl":"10.1016/j.compmedimag.2024.102388","url":null,"abstract":"<div><p>Rib cross-sectional shapes (characterized by the outer contour and cortical bone thickness) affect the rib mechanical response under impact loading, thereby influence the rib injury pattern and risk. A statistical description of the rib shapes or their correlations to anthropometrics is a prerequisite to the development of numerical human body models representing target demographics. Variational autoencoders (VAE) as anatomical shape generators remain to be explored in terms of utilizing the latent vectors to control or interpret the representativeness of the generated results. In this paper, we propose a pipeline for developing a multi-rib cross-sectional shape generative model from CT images, which consists of the achievement of rib cross-sectional shape data from CT images using an anatomical indexing system and regular grids, and a unified framework to fit shape distributions and associate shapes to anthropometrics for different rib categories. Specifically, we collected CT images including 3193 ribs, surface regular grid is generated for each rib based on anatomical coordinates, the rib cross-sectional shapes are characterized by nodal coordinates and cortical bone thickness. The tensor structure of shape data based on regular grids enable the implementation of CNNs in the conditional variational autoencoder (CVAE). The CVAE is trained against an auxiliary classifier to decouple the low-dimensional representations of the inter- and intra- variations and fit each intra-variation by a Gaussian distribution simultaneously. Random tree regressors are further leveraged to associate each continuous intra-class space with the corresponding anthropometrics of the subjects, i.e., age, height and weight. As a result, with the rib class labels and the latent vectors sampled from Gaussian distributions or predicted from anthropometrics as the inputs, the decoder can generate valid rib cross-sectional shapes of given class labels (male/female, 2nd to 11th ribs) for arbitrary populational percentiles or specific age, height and weight, which paves the road for future biomedical and biomechanical studies considering the diversity of rib shapes across the population.</p></div>","PeriodicalId":50631,"journal":{"name":"Computerized Medical Imaging and Graphics","volume":null,"pages":null},"PeriodicalIF":5.7,"publicationDate":"2024-04-25","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140791093","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Computerized Medical Imaging and Graphics
全部 Acc. Chem. Res. ACS Applied Bio Materials ACS Appl. Electron. Mater. ACS Appl. Energy Mater. ACS Appl. Mater. Interfaces ACS Appl. Nano Mater. ACS Appl. Polym. Mater. ACS BIOMATER-SCI ENG ACS Catal. ACS Cent. Sci. ACS Chem. Biol. ACS Chemical Health & Safety ACS Chem. Neurosci. ACS Comb. Sci. ACS Earth Space Chem. ACS Energy Lett. ACS Infect. Dis. ACS Macro Lett. ACS Mater. Lett. ACS Med. Chem. Lett. ACS Nano ACS Omega ACS Photonics ACS Sens. ACS Sustainable Chem. Eng. ACS Synth. Biol. Anal. Chem. BIOCHEMISTRY-US Bioconjugate Chem. BIOMACROMOLECULES Chem. Res. Toxicol. Chem. Rev. Chem. Mater. CRYST GROWTH DES ENERG FUEL Environ. Sci. Technol. Environ. Sci. Technol. Lett. Eur. J. Inorg. Chem. IND ENG CHEM RES Inorg. Chem. J. Agric. Food. Chem. J. Chem. Eng. Data J. Chem. Educ. J. Chem. Inf. Model. J. Chem. Theory Comput. J. Med. Chem. J. Nat. Prod. J PROTEOME RES J. Am. Chem. Soc. LANGMUIR MACROMOLECULES Mol. Pharmaceutics Nano Lett. Org. Lett. ORG PROCESS RES DEV ORGANOMETALLICS J. Org. Chem. J. Phys. Chem. J. Phys. Chem. A J. Phys. Chem. B J. Phys. Chem. C J. Phys. Chem. Lett. Analyst Anal. Methods Biomater. Sci. Catal. Sci. Technol. Chem. Commun. Chem. Soc. Rev. CHEM EDUC RES PRACT CRYSTENGCOMM Dalton Trans. Energy Environ. Sci. ENVIRON SCI-NANO ENVIRON SCI-PROC IMP ENVIRON SCI-WAT RES Faraday Discuss. Food Funct. Green Chem. Inorg. Chem. Front. Integr. Biol. J. Anal. At. Spectrom. J. Mater. Chem. A J. Mater. Chem. B J. Mater. Chem. C Lab Chip Mater. Chem. Front. Mater. Horiz. MEDCHEMCOMM Metallomics Mol. Biosyst. Mol. Syst. Des. Eng. Nanoscale Nanoscale Horiz. Nat. Prod. Rep. New J. Chem. Org. Biomol. Chem. Org. Chem. Front. PHOTOCH PHOTOBIO SCI PCCP Polym. Chem.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
0
微信
客服QQ
Book学术公众号 扫码关注我们
反馈
×
意见反馈
请填写您的意见或建议
请填写您的手机或邮箱
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
现在去查看 取消
×
提示
确定
Book学术官方微信
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术
文献互助 智能选刊 最新文献 互助须知 联系我们:info@booksci.cn
Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。
Copyright © 2023 Book学术 All rights reserved.
ghs 京公网安备 11010802042870号 京ICP备2023020795号-1