首页 > 最新文献

Radiology-Artificial Intelligence最新文献

英文 中文
Radiomics Beyond the Hype: A Critical Evaluation Toward Oncologic Clinical Use. eRadiomics 超越炒作:面向肿瘤临床应用的严格评估。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230437
Natally Horvat, Nikolaos Papanikolaou, Dow-Mu Koh

Radiomics is a promising and fast-developing field within oncology that involves the mining of quantitative high-dimensional data from medical images. Radiomics has the potential to transform cancer management, whereby radiomics data can be used to aid early tumor characterization, prognosis, risk stratification, treatment planning, treatment response assessment, and surveillance. Nevertheless, certain challenges have delayed the clinical adoption and acceptability of radiomics in routine clinical practice. The objectives of this report are to (a) provide a perspective on the translational potential and potential impact of radiomics in oncology; (b) explore frequent challenges and mistakes in its derivation, encompassing study design, technical requirements, standardization, model reproducibility, transparency, data sharing, privacy concerns, quality control, as well as the complexity of multistep processes resulting in less radiologist-friendly interfaces; (c) discuss strategies to overcome these challenges and mistakes; and (d) propose measures to increase the clinical use and acceptability of radiomics, taking into account the different perspectives of patients, health care workers, and health care systems. Keywords: Radiomics, Oncology, Cancer Management, Artificial Intelligence © RSNA, 2024.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现影响内容的错误。放射组学是肿瘤学中一个前景广阔、发展迅速的领域,涉及从医学影像中挖掘定量高维数据。放射组学具有改变癌症管理的潜力,放射组学数据可用于辅助早期肿瘤特征描述、预后判断、风险分层、治疗计划、治疗反应评估和监测等。然而,放射组学在常规临床实践中的临床应用和可接受性还面临着一些挑战。本报告的目的是(a) 展望放射组学在肿瘤学领域的转化潜力和潜在影响;(b) 探讨放射组学发展过程中经常遇到的挑战和失误,包括研究设计、技术要求、标准化、模型可重复性、透明度、数据共享、隐私问题、质量控制,以及多步骤流程的复杂性导致放射科医生界面不够友好;(c) 讨论克服这些挑战和错误的策略;以及 (d) 考虑到患者、医护人员和医疗系统的不同观点,提出提高放射组学临床应用和可接受性的措施。©RSNA,2024 年。
{"title":"Radiomics Beyond the Hype: A Critical Evaluation Toward Oncologic Clinical Use.","authors":"Natally Horvat, Nikolaos Papanikolaou, Dow-Mu Koh","doi":"10.1148/ryai.230437","DOIUrl":"10.1148/ryai.230437","url":null,"abstract":"<p><p>Radiomics is a promising and fast-developing field within oncology that involves the mining of quantitative high-dimensional data from medical images. Radiomics has the potential to transform cancer management, whereby radiomics data can be used to aid early tumor characterization, prognosis, risk stratification, treatment planning, treatment response assessment, and surveillance. Nevertheless, certain challenges have delayed the clinical adoption and acceptability of radiomics in routine clinical practice. The objectives of this report are to (<i>a</i>) provide a perspective on the translational potential and potential impact of radiomics in oncology; (<i>b</i>) explore frequent challenges and mistakes in its derivation, encompassing study design, technical requirements, standardization, model reproducibility, transparency, data sharing, privacy concerns, quality control, as well as the complexity of multistep processes resulting in less radiologist-friendly interfaces; (<i>c</i>) discuss strategies to overcome these challenges and mistakes; and (<i>d</i>) propose measures to increase the clinical use and acceptability of radiomics, taking into account the different perspectives of patients, health care workers, and health care systems. <b>Keywords:</b> Radiomics, Oncology, Cancer Management, Artificial Intelligence © RSNA, 2024.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294952/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140877467","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Two-Stage Training Framework Using Multicontrast MRI Radiomics for IDH Mutation Status Prediction in Glioma. 利用多对比核磁共振成像放射组学预测胶质瘤中 IDH 突变状态的两阶段训练框架
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230218
Nghi C D Truong, Chandan Ganesh Bangalore Yogananda, Benjamin C Wagner, James M Holcomb, Divya Reddy, Niloufar Saadat, Kimmo J Hatanpaa, Toral R Patel, Baowei Fei, Matthew D Lee, Rajan Jain, Richard J Bruce, Marco C Pinho, Ananth J Madhuranthakam, Joseph A Maldjian

Purpose To develop a radiomics framework for preoperative MRI-based prediction of isocitrate dehydrogenase (IDH) mutation status, a crucial glioma prognostic indicator. Materials and Methods Radiomics features (shape, first-order statistics, and texture) were extracted from the whole tumor or the combination of nonenhancing, necrosis, and edema regions. Segmentation masks were obtained via the federated tumor segmentation tool or the original data source. Boruta, a wrapper-based feature selection algorithm, identified relevant features. Addressing the imbalance between mutated and wild-type cases, multiple prediction models were trained on balanced data subsets using random forest or XGBoost and assembled to build the final classifier. The framework was evaluated using retrospective MRI scans from three public datasets (The Cancer Imaging Archive [TCIA, 227 patients], the University of California San Francisco Preoperative Diffuse Glioma MRI dataset [UCSF, 495 patients], and the Erasmus Glioma Database [EGD, 456 patients]) and internal datasets collected from the University of Texas Southwestern Medical Center (UTSW, 356 patients), New York University (NYU, 136 patients), and University of Wisconsin-Madison (UWM, 174 patients). TCIA and UTSW served as separate training sets, while the remaining data constituted the test set (1617 or 1488 testing cases, respectively). Results The best performing models trained on the TCIA dataset achieved area under the receiver operating characteristic curve (AUC) values of 0.89 for UTSW, 0.86 for NYU, 0.93 for UWM, 0.94 for UCSF, and 0.88 for EGD test sets. The best performing models trained on the UTSW dataset achieved slightly higher AUCs: 0.92 for TCIA, 0.88 for NYU, 0.96 for UWM, 0.93 for UCSF, and 0.90 for EGD. Conclusion This MRI radiomics-based framework shows promise for accurate preoperative prediction of IDH mutation status in patients with glioma. Keywords: Glioma, Isocitrate Dehydrogenase Mutation, IDH Mutation, Radiomics, MRI Supplemental material is available for this article. Published under a CC BY 4.0 license. See also commentary by Moassefi and Erickson in this issue.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现影响内容的错误。目的 建立一个放射组学框架,用于术前基于 MRI 预测 IDH 突变状态,这是胶质瘤预后的一个重要指标。材料与方法 从整个肿瘤或非增强、坏死和水肿区域的组合中提取放射组学特征(形状、一阶统计和纹理)。分割掩膜通过联合肿瘤分割工具或原始数据源获得。Boruta是一种基于包装的特征选择算法,可识别相关特征。为了解决突变型病例和野生型病例之间的不平衡问题,使用随机森林或 XGBoost 在平衡数据子集上训练了多个预测模型,并将其组合起来建立最终分类器。利用三个公共数据集(癌症成像档案(TCIA,227 名患者)、加州大学旧金山分校术前弥漫性胶质瘤 MRI 数据集(UCSF,495 名患者))的回顾性 MRI 扫描对该框架进行了评估、和伊拉斯谟胶质瘤数据库(EGD,456 名患者))以及UT 西南医学中心(UTSW,356 名患者)、纽约大学(NYU,136 名患者)和威斯康星大学麦迪逊分校(UWM,174 名患者)收集的内部数据集。TCIA和UTSW作为单独的训练集,其余数据构成测试集(分别有1617个或1488个测试病例)。结果 在 TCIA 数据集上训练的表现最好的模型,其接收者操作特征曲线下面积(AUC)值分别为:UTSW 0.89、NYU 0.86、UWM 0.93、UCSF 0.94、EGD 测试集 0.88。在UTSW数据集上训练的表现最好的模型的AUC略高:TCIA为0.92,NYU为0.88,UWM为0.96,UCSF为0.93,EGD为0.90。结论 这种基于磁共振成像放射组学的框架有望准确预测胶质瘤患者的术前IDH突变状态。以 CC BY 4.0 许可发布。
{"title":"Two-Stage Training Framework Using Multicontrast MRI Radiomics for <i>IDH</i> Mutation Status Prediction in Glioma.","authors":"Nghi C D Truong, Chandan Ganesh Bangalore Yogananda, Benjamin C Wagner, James M Holcomb, Divya Reddy, Niloufar Saadat, Kimmo J Hatanpaa, Toral R Patel, Baowei Fei, Matthew D Lee, Rajan Jain, Richard J Bruce, Marco C Pinho, Ananth J Madhuranthakam, Joseph A Maldjian","doi":"10.1148/ryai.230218","DOIUrl":"10.1148/ryai.230218","url":null,"abstract":"<p><p>Purpose To develop a radiomics framework for preoperative MRI-based prediction of isocitrate dehydrogenase (<i>IDH</i>) mutation status, a crucial glioma prognostic indicator. Materials and Methods Radiomics features (shape, first-order statistics, and texture) were extracted from the whole tumor or the combination of nonenhancing, necrosis, and edema regions. Segmentation masks were obtained via the federated tumor segmentation tool or the original data source. Boruta, a wrapper-based feature selection algorithm, identified relevant features. Addressing the imbalance between mutated and wild-type cases, multiple prediction models were trained on balanced data subsets using random forest or XGBoost and assembled to build the final classifier. The framework was evaluated using retrospective MRI scans from three public datasets (The Cancer Imaging Archive [TCIA, 227 patients], the University of California San Francisco Preoperative Diffuse Glioma MRI dataset [UCSF, 495 patients], and the Erasmus Glioma Database [EGD, 456 patients]) and internal datasets collected from the University of Texas Southwestern Medical Center (UTSW, 356 patients), New York University (NYU, 136 patients), and University of Wisconsin-Madison (UWM, 174 patients). TCIA and UTSW served as separate training sets, while the remaining data constituted the test set (1617 or 1488 testing cases, respectively). Results The best performing models trained on the TCIA dataset achieved area under the receiver operating characteristic curve (AUC) values of 0.89 for UTSW, 0.86 for NYU, 0.93 for UWM, 0.94 for UCSF, and 0.88 for EGD test sets. The best performing models trained on the UTSW dataset achieved slightly higher AUCs: 0.92 for TCIA, 0.88 for NYU, 0.96 for UWM, 0.93 for UCSF, and 0.90 for EGD. Conclusion This MRI radiomics-based framework shows promise for accurate preoperative prediction of <i>IDH</i> mutation status in patients with glioma. <b>Keywords:</b> Glioma, Isocitrate Dehydrogenase Mutation, <i>IDH</i> Mutation, Radiomics, MRI <i>Supplemental material is available for this article.</i> Published under a CC BY 4.0 license. See also commentary by Moassefi and Erickson in this issue.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294953/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141074538","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Evaluating Sex-specific Differences in Abdominal Fat Volume and Proton Density Fat Fraction at MRI Using Automated nnU-Net-based Segmentation. 利用基于 nnU-Net 的自动分割技术评估磁共振成像扫描中腹部脂肪量和质子密度脂肪率的性别差异
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230471
Arun Somasundaram, Mingming Wu, Anna Reik, Selina Rupp, Jessie Han, Stella Naebauer, Daniela Junker, Lisa Patzelt, Meike Wiechert, Yu Zhao, Daniel Rueckert, Hans Hauner, Christina Holzapfel, Dimitrios C Karampinos

Sex-specific abdominal organ volume and proton density fat fraction (PDFF) in people with obesity during a weight loss intervention was assessed with automated multiorgan segmentation of quantitative water-fat MRI. An nnU-Net architecture was employed for automatic segmentation of abdominal organs, including visceral and subcutaneous adipose tissue, liver, and psoas and erector spinae muscle, based on quantitative chemical shift-encoded MRI and using ground truth labels generated from participants of the Lifestyle Intervention (LION) study. Each organ's volume and fat content were examined in 127 participants (73 female and 54 male participants; body mass index, 30-39.9 kg/m2) and in 81 (54 female and 32 male participants) of these participants after an 8-week formula-based low-calorie diet. Dice scores ranging from 0.91 to 0.97 were achieved for the automatic segmentation. PDFF was found to be lower in visceral adipose tissue compared with subcutaneous adipose tissue in both male and female participants. Before intervention, female participants exhibited higher PDFF in subcutaneous adipose tissue (90.6% vs 89.7%; P < .001) and lower PDFF in liver (8.6% vs 13.3%; P < .001) and visceral adipose tissue (76.4% vs 81.3%; P < .001) compared with male participants. This relation persisted after intervention. As a response to caloric restriction, male participants lost significantly more visceral adipose tissue volume (1.76 L vs 0.91 L; P < .001) and showed a higher decrease in subcutaneous adipose tissue PDFF (2.7% vs 1.5%; P < .001) than female participants. Automated body composition analysis on quantitative water-fat MRI data provides new insights for understanding sex-specific metabolic response to caloric restriction and weight loss in people with obesity. Keywords: Obesity, Chemical Shift-encoded MRI, Abdominal Fat Volume, Proton Density Fat Fraction, nnU-Net ClinicalTrials.gov registration no. NCT04023942 Supplemental material is available for this article. Published under a CC BY 4.0 license.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校样审核。请注意,在制作最终校对稿的过程中,可能会发现影响内容的错误。利用定量水-脂肪核磁共振成像的自动多器官分割技术评估了减肥干预期间肥胖症患者腹部器官体积和质子密度脂肪分数(PDFF)的性别特异性。根据定量化学位移编码核磁共振成像,并使用生活方式干预(LION)研究参与者生成的地面实况标签,采用 nnU-Net 架构自动分割腹部器官,包括内脏脂肪组织(VAT)和皮下脂肪组织(SAT)、肝脏、腰肌和竖脊肌。研究人员对 127 名参与者(73 名女性,54 名男性;体重指数为 30-39.9 kg/m2)以及其中 81 名参与者(54 名女性,32 名男性)进行了为期 8 周的配方低热量饮食后,对每个器官的体积和脂肪含量进行了检测。自动分段的骰子得分从 0.91 到 0.97 不等。研究发现,在男性和女性参与者中,VAT 的 PDFF 均低于 SAT。干预前,与男性相比,女性在 SAT(90.6% 对 89.7%,P < .001)和肝脏(8.6% 对 13.3%,P < .001)以及 VAT(76.4% 对 81.3%,P < .001)中表现出更高的 PDFF。这种关系在干预后仍然存在。作为对热量限制的反应,与女性参与者相比,男性参与者的增值脂肪体积明显减少(1.76 升比 0.91 升,P < .001),SAT PDFF 的下降幅度也更高(2.7% 比 1.5%,P < .001)。对定量水-脂肪核磁共振成像数据进行自动身体成分分析为了解肥胖症患者对热量限制和体重减轻的性别特异性代谢反应提供了新的视角。以 CC BY 4.0 许可发布。
{"title":"Evaluating Sex-specific Differences in Abdominal Fat Volume and Proton Density Fat Fraction at MRI Using Automated nnU-Net-based Segmentation.","authors":"Arun Somasundaram, Mingming Wu, Anna Reik, Selina Rupp, Jessie Han, Stella Naebauer, Daniela Junker, Lisa Patzelt, Meike Wiechert, Yu Zhao, Daniel Rueckert, Hans Hauner, Christina Holzapfel, Dimitrios C Karampinos","doi":"10.1148/ryai.230471","DOIUrl":"10.1148/ryai.230471","url":null,"abstract":"<p><p>Sex-specific abdominal organ volume and proton density fat fraction (PDFF) in people with obesity during a weight loss intervention was assessed with automated multiorgan segmentation of quantitative water-fat MRI. An nnU-Net architecture was employed for automatic segmentation of abdominal organs, including visceral and subcutaneous adipose tissue, liver, and psoas and erector spinae muscle, based on quantitative chemical shift-encoded MRI and using ground truth labels generated from participants of the Lifestyle Intervention (LION) study. Each organ's volume and fat content were examined in 127 participants (73 female and 54 male participants; body mass index, 30-39.9 kg/m<sup>2</sup>) and in 81 (54 female and 32 male participants) of these participants after an 8-week formula-based low-calorie diet. Dice scores ranging from 0.91 to 0.97 were achieved for the automatic segmentation. PDFF was found to be lower in visceral adipose tissue compared with subcutaneous adipose tissue in both male and female participants. Before intervention, female participants exhibited higher PDFF in subcutaneous adipose tissue (90.6% vs 89.7%; <i>P</i> < .001) and lower PDFF in liver (8.6% vs 13.3%; <i>P</i> < .001) and visceral adipose tissue (76.4% vs 81.3%; <i>P</i> < .001) compared with male participants. This relation persisted after intervention. As a response to caloric restriction, male participants lost significantly more visceral adipose tissue volume (1.76 L vs 0.91 L; <i>P</i> < .001) and showed a higher decrease in subcutaneous adipose tissue PDFF (2.7% vs 1.5%; <i>P</i> < .001) than female participants. Automated body composition analysis on quantitative water-fat MRI data provides new insights for understanding sex-specific metabolic response to caloric restriction and weight loss in people with obesity. <b>Keywords:</b> Obesity, Chemical Shift-encoded MRI, Abdominal Fat Volume, Proton Density Fat Fraction, nnU-Net ClinicalTrials.gov registration no. NCT04023942 <i>Supplemental material is available for this article.</i> Published under a CC BY 4.0 license.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294970/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141162496","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
The University of California San Francisco Adult Longitudinal Post-Treatment Diffuse Glioma MRI Dataset. 加州大学旧金山分校成人纵向弥漫性胶质瘤治疗后(UCSF-ALPTDG)磁共振成像数据集。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230182
Brandon K K Fields, Evan Calabrese, John Mongan, Soonmee Cha, Christopher P Hess, Leo P Sugrue, Susan M Chang, Tracy L Luks, Javier E Villanueva-Meyer, Andreas M Rauschecker, Jeffrey D Rudie

Supplemental material is available for this article.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现一些可能影响内容的错误。加利福尼亚大学旧金山分校成人弥漫性胶质瘤纵向治疗后 MRI 数据集(UCSF-ALPTDG)是一个公开的注释数据集,包含 298 名弥漫性胶质瘤患者在两次连续随访(共 596 次扫描)时拍摄的多模态脑 MRI 图像,以及相应的临床病史和专家体素注释。©RSNA,2024。
{"title":"The University of California San Francisco Adult Longitudinal Post-Treatment Diffuse Glioma MRI Dataset.","authors":"Brandon K K Fields, Evan Calabrese, John Mongan, Soonmee Cha, Christopher P Hess, Leo P Sugrue, Susan M Chang, Tracy L Luks, Javier E Villanueva-Meyer, Andreas M Rauschecker, Jeffrey D Rudie","doi":"10.1148/ryai.230182","DOIUrl":"10.1148/ryai.230182","url":null,"abstract":"<p><p>\u0000 <i>Supplemental material is available for this article.</i>\u0000 </p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294954/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141307008","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Impact of Transfer Learning Using Local Data on Performance of a Deep Learning Model for Screening Mammography. 使用本地数据进行迁移学习对乳腺筛查深度学习模型性能的影响。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230383
James J J Condon, Vincent Trinh, Kelly A Hall, Michelle Reintals, Andrew S Holmes, Lauren Oakden-Rayner, Lyle J Palmer

Purpose To investigate the issues of generalizability and replication of deep learning models by assessing performance of a screening mammography deep learning system developed at New York University (NYU) on a local Australian dataset. Materials and Methods In this retrospective study, all individuals with biopsy or surgical pathology-proven lesions and age-matched controls were identified from a South Australian public mammography screening program (January 2010 to December 2016). The primary outcome was deep learning system performance-measured with area under the receiver operating characteristic curve (AUC)-in classifying invasive breast cancer or ductal carcinoma in situ (n = 425) versus no malignancy (n = 490) or benign lesions (n = 44). The NYU system, including models without (NYU1) and with (NYU2) heatmaps, was tested in its original form, after training from scratch (without transfer learning), and after retraining with transfer learning. Results The local test set comprised 959 individuals (mean age, 62.5 years ± 8.5 [SD]; all female). The original AUCs for the NYU1 and NYU2 models were 0.83 (95% CI: 0.82, 0.84) and 0.89 (95% CI: 0.88, 0.89), respectively. When NYU1 and NYU2 were applied in their original form to the local test set, the AUCs were 0.76 (95% CI: 0.73, 0.79) and 0.84 (95% CI: 0.82, 0.87), respectively. After local training without transfer learning, the AUCs were 0.66 (95% CI: 0.62, 0.69) and 0.86 (95% CI: 0.84, 0.88). After retraining with transfer learning, the AUCs were 0.82 (95% CI: 0.80, 0.85) and 0.86 (95% CI: 0.84, 0.88). Conclusion A deep learning system developed using a U.S. dataset showed reduced performance when applied "out of the box" to an Australian dataset. Local retraining with transfer learning using available model weights improved model performance. Keywords: Screening Mammography, Convolutional Neural Network (CNN), Deep Learning Algorithms, Breast Cancer Supplemental material is available for this article. © RSNA, 2024 See also commentary by Cadrin-Chênevert in this issue.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现影响文章内容的错误。目的 通过评估纽约大学(NYU)在澳大利亚本地数据集上开发的乳腺 X 射线筛查 DL 系统的性能,研究深度学习(DL)模型的可推广性和可复制性问题。材料与方法 在这项回顾性研究中,我们从南澳大利亚公共乳腺放射摄影筛查项目(2010 年 1 月至 2016 年 12 月)中确定了所有活检和手术病理证实病变的个体以及年龄匹配的对照组。主要结果是DL系统在将浸润性乳腺癌或导管原位癌(n = 425)从无恶性病变(n = 490)或良性病变(n = 44)的年龄匹配对照中进行分类时的性能,用接收器操作特征曲线下面积(AUC)来衡量。对 NYU 系统(包括无热图(NYU1)和有热图(NYU2)的模型)进行了原始测试、从头开始训练(无迁移学习;TL)和用迁移学习重新训练后的测试。结果 本地测试集包括 959 人(平均年龄 62.5 岁 [SD, 8.5];均为女性)。NYU1 和 NYU2 模型的原始 AUC 分别为 0.83(95%CI = 0.82-0.84)和 0.89(95%CI = 0.88-0.89)。当以原始形式应用于本地测试集时,AUC 分别为 0.76 (95%CI = 0.73-0.79) 和 0.84 (95%CI = 0.82-0.87)。在不使用 TL 进行局部训练后,AUC 分别为 0.66(95%CI = 0.62-0.69)和 0.86(95%CI = 0.84-0.88)。使用 TL 重新训练后,AUC 分别为 0.82(95%CI = 0.80-0.85)和 0.86(95%CI = 0.84-0.88)。结论 使用美国数据集开发的深度学习系统在 "开箱即用 "澳大利亚数据集时,性能有所下降。利用现有模型权重进行迁移学习的局部再训练提高了模型性能。©RSNA,2024。
{"title":"Impact of Transfer Learning Using Local Data on Performance of a Deep Learning Model for Screening Mammography.","authors":"James J J Condon, Vincent Trinh, Kelly A Hall, Michelle Reintals, Andrew S Holmes, Lauren Oakden-Rayner, Lyle J Palmer","doi":"10.1148/ryai.230383","DOIUrl":"10.1148/ryai.230383","url":null,"abstract":"<p><p>Purpose To investigate the issues of generalizability and replication of deep learning models by assessing performance of a screening mammography deep learning system developed at New York University (NYU) on a local Australian dataset. Materials and Methods In this retrospective study, all individuals with biopsy or surgical pathology-proven lesions and age-matched controls were identified from a South Australian public mammography screening program (January 2010 to December 2016). The primary outcome was deep learning system performance-measured with area under the receiver operating characteristic curve (AUC)-in classifying invasive breast cancer or ductal carcinoma in situ (<i>n</i> = 425) versus no malignancy (<i>n</i> = 490) or benign lesions (<i>n</i> = 44). The NYU system, including models without (NYU1) and with (NYU2) heatmaps, was tested in its original form, after training from scratch (without transfer learning), and after retraining with transfer learning. Results The local test set comprised 959 individuals (mean age, 62.5 years ± 8.5 [SD]; all female). The original AUCs for the NYU1 and NYU2 models were 0.83 (95% CI: 0.82, 0.84) and 0.89 (95% CI: 0.88, 0.89), respectively. When NYU1 and NYU2 were applied in their original form to the local test set, the AUCs were 0.76 (95% CI: 0.73, 0.79) and 0.84 (95% CI: 0.82, 0.87), respectively. After local training without transfer learning, the AUCs were 0.66 (95% CI: 0.62, 0.69) and 0.86 (95% CI: 0.84, 0.88). After retraining with transfer learning, the AUCs were 0.82 (95% CI: 0.80, 0.85) and 0.86 (95% CI: 0.84, 0.88). Conclusion A deep learning system developed using a U.S. dataset showed reduced performance when applied \"out of the box\" to an Australian dataset. Local retraining with transfer learning using available model weights improved model performance. <b>Keywords:</b> Screening Mammography, Convolutional Neural Network (CNN), Deep Learning Algorithms, Breast Cancer <i>Supplemental material is available for this article.</i> © RSNA, 2024 See also commentary by Cadrin-Chênevert in this issue.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294949/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140877468","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Stepwise Transfer Learning for Expert-level Pediatric Brain Tumor MRI Segmentation in a Limited Data Scenario. 在数据有限的情况下,针对专家级小儿脑肿瘤磁共振成像分割的逐步迁移学习。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230254
Aidan Boyd, Zezhong Ye, Sanjay P Prabhu, Michael C Tjong, Yining Zha, Anna Zapaishchykova, Sridhar Vajapeyam, Paul J Catalano, Hasaan Hayat, Rishi Chopra, Kevin X Liu, Ali Nabavizadeh, Adam C Resnick, Sabine Mueller, Daphne A Haas-Kogan, Hugo J W L Aerts, Tina Y Poussaint, Benjamin H Kann

Purpose To develop, externally test, and evaluate clinical acceptability of a deep learning pediatric brain tumor segmentation model using stepwise transfer learning. Materials and Methods In this retrospective study, the authors leveraged two T2-weighted MRI datasets (May 2001 through December 2015) from a national brain tumor consortium (n = 184; median age, 7 years [range, 1-23 years]; 94 male patients) and a pediatric cancer center (n = 100; median age, 8 years [range, 1-19 years]; 47 male patients) to develop and evaluate deep learning neural networks for pediatric low-grade glioma segmentation using a stepwise transfer learning approach to maximize performance in a limited data scenario. The best model was externally tested on an independent test set and subjected to randomized blinded evaluation by three clinicians, wherein they assessed clinical acceptability of expert- and artificial intelligence (AI)-generated segmentations via 10-point Likert scales and Turing tests. Results The best AI model used in-domain stepwise transfer learning (median Dice score coefficient, 0.88 [IQR, 0.72-0.91] vs 0.812 [IQR, 0.56-0.89] for baseline model; P = .049). With external testing, the AI model yielded excellent accuracy using reference standards from three clinical experts (median Dice similarity coefficients: expert 1, 0.83 [IQR, 0.75-0.90]; expert 2, 0.81 [IQR, 0.70-0.89]; expert 3, 0.81 [IQR, 0.68-0.88]; mean accuracy, 0.82). For clinical benchmarking (n = 100 scans), experts rated AI-based segmentations higher on average compared with other experts (median Likert score, 9 [IQR, 7-9] vs 7 [IQR 7-9]) and rated more AI segmentations as clinically acceptable (80.2% vs 65.4%). Experts correctly predicted the origin of AI segmentations in an average of 26.0% of cases. Conclusion Stepwise transfer learning enabled expert-level automated pediatric brain tumor autosegmentation and volumetric measurement with a high level of clinical acceptability. Keywords: Stepwise Transfer Learning, Pediatric Brain Tumors, MRI Segmentation, Deep Learning Supplemental material is available for this article. © RSNA, 2024.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现影响内容的错误。目的 开发、外部测试和评估使用逐步转移学习的深度学习(DL)儿科脑肿瘤分割模型的临床可接受性。材料与方法 在这项回顾性研究中,作者利用两个 T2 加权磁共振成像数据集(2001 年 5 月至 2015 年 12 月),分别来自一个国家脑肿瘤联盟(n = 184;中位年龄 7 岁(范围:1-23 岁);94 名男性)和一个儿科癌症中心(n = 100;中位年龄 8 岁(范围:1-19 岁);47 名男性),开发并评估了用于儿科低级别胶质瘤分割的 DL 神经网络,采用了一种新颖的逐步转移学习方法,以在有限的数据场景中实现性能最大化。最佳模型在独立测试集上进行了外部测试,并由三位临床医生进行了随机、盲测评估,他们通过 10 分李克特量表和图灵测试评估了专家和人工智能(AI)生成的分割结果的临床可接受性。结果 最佳人工智能模型采用了域内逐步转移学习(DSC 中位数:0.88 [IQR 0.72-0.91] 而基线模型为 0.812 [0.56-0.89];P = .049)。在外部测试中,人工智能模型使用三位临床专家提供的参考标准(专家-1:0.83 [0.75-0.90];专家-2:0.81 [0.70-0.89];专家-3:0.81 [0.68-0.88];平均准确度:0.82))获得了极高的准确度。在临床基准测试(n = 100 次扫描)中,专家对基于人工智能的分割的平均评分高于其他专家(Likert 评分中位数:中位数 9 [IQR 7-9]) 对 7 [IQR 7-9]),并将更多人工智能分割评为临床可接受(80.2% 对 65.4%)。专家平均在 26.0% 的病例中正确预测了人工智能分割的起源。结论 逐步迁移学习实现了专家级的自动化小儿脑肿瘤自动分割和体积测量,并具有较高的临床可接受性。©RSNA, 2024.
{"title":"Stepwise Transfer Learning for Expert-level Pediatric Brain Tumor MRI Segmentation in a Limited Data Scenario.","authors":"Aidan Boyd, Zezhong Ye, Sanjay P Prabhu, Michael C Tjong, Yining Zha, Anna Zapaishchykova, Sridhar Vajapeyam, Paul J Catalano, Hasaan Hayat, Rishi Chopra, Kevin X Liu, Ali Nabavizadeh, Adam C Resnick, Sabine Mueller, Daphne A Haas-Kogan, Hugo J W L Aerts, Tina Y Poussaint, Benjamin H Kann","doi":"10.1148/ryai.230254","DOIUrl":"10.1148/ryai.230254","url":null,"abstract":"<p><p>Purpose To develop, externally test, and evaluate clinical acceptability of a deep learning pediatric brain tumor segmentation model using stepwise transfer learning. Materials and Methods In this retrospective study, the authors leveraged two T2-weighted MRI datasets (May 2001 through December 2015) from a national brain tumor consortium (<i>n</i> = 184; median age, 7 years [range, 1-23 years]; 94 male patients) and a pediatric cancer center (<i>n</i> = 100; median age, 8 years [range, 1-19 years]; 47 male patients) to develop and evaluate deep learning neural networks for pediatric low-grade glioma segmentation using a stepwise transfer learning approach to maximize performance in a limited data scenario. The best model was externally tested on an independent test set and subjected to randomized blinded evaluation by three clinicians, wherein they assessed clinical acceptability of expert- and artificial intelligence (AI)-generated segmentations via 10-point Likert scales and Turing tests. Results The best AI model used in-domain stepwise transfer learning (median Dice score coefficient, 0.88 [IQR, 0.72-0.91] vs 0.812 [IQR, 0.56-0.89] for baseline model; <i>P</i> = .049). With external testing, the AI model yielded excellent accuracy using reference standards from three clinical experts (median Dice similarity coefficients: expert 1, 0.83 [IQR, 0.75-0.90]; expert 2, 0.81 [IQR, 0.70-0.89]; expert 3, 0.81 [IQR, 0.68-0.88]; mean accuracy, 0.82). For clinical benchmarking (<i>n</i> = 100 scans), experts rated AI-based segmentations higher on average compared with other experts (median Likert score, 9 [IQR, 7-9] vs 7 [IQR 7-9]) and rated more AI segmentations as clinically acceptable (80.2% vs 65.4%). Experts correctly predicted the origin of AI segmentations in an average of 26.0% of cases. Conclusion Stepwise transfer learning enabled expert-level automated pediatric brain tumor autosegmentation and volumetric measurement with a high level of clinical acceptability. <b>Keywords:</b> Stepwise Transfer Learning, Pediatric Brain Tumors, MRI Segmentation, Deep Learning <i>Supplemental material is available for this article</i>. © RSNA, 2024.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294948/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141564667","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Navigating Clinical Variability: Transfer Learning's Impact on Imaging Model Performance. 驾驭临床变异性:迁移学习对成像模型性能的影响。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.240263
Alexandre Cadrin-Chênevert
{"title":"Navigating Clinical Variability: Transfer Learning's Impact on Imaging Model Performance.","authors":"Alexandre Cadrin-Chênevert","doi":"10.1148/ryai.240263","DOIUrl":"10.1148/ryai.240263","url":null,"abstract":"","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294946/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141427772","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
A New Era of Text Mining in Radiology with Privacy-Preserving LLMs. 用保护隐私的 LLMs 开启放射学文本挖掘新纪元
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.240261
Tugba Akinci D'Antonoli, Christian Bluethgen
{"title":"A New Era of Text Mining in Radiology with Privacy-Preserving LLMs.","authors":"Tugba Akinci D'Antonoli, Christian Bluethgen","doi":"10.1148/ryai.240261","DOIUrl":"10.1148/ryai.240261","url":null,"abstract":"","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294969/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141427770","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Reproducibility Analysis of Radiomic Features on T2-weighted MR Images after Processing and Segmentation Alterations in Neuroblastoma Tumors. 神经母细胞瘤 T2 加权核磁共振成像处理和分割交替后放射学特征的再现性分析
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230208
Diana Veiga-Canuto, Matías Fernández-Patón, Leonor Cerdà Alberich, Ana Jiménez Pastor, Armando Gomis Maya, Jose Miguel Carot Sierra, Cinta Sangüesa Nebot, Blanca Martínez de Las Heras, Ulrike Pötschger, Sabine Taschner-Mandl, Emanuele Neri, Adela Cañete, Ruth Ladenstein, Barbara Hero, Ángel Alberich-Bayarri, Luis Martí-Bonmatí

Purpose To evaluate the reproducibility of radiomics features extracted from T2-weighted MR images in patients with neuroblastoma. Materials and Methods A retrospective study included 419 patients (mean age, 29 months ± 34 [SD]; 220 male, 199 female) with neuroblastic tumors diagnosed between 2002 and 2023, within the scope of the PRedictive In-silico Multiscale Analytics to support cancer personalized diaGnosis and prognosis, Empowered by imaging biomarkers (ie, PRIMAGE) project, involving 746 T2/T2*-weighted MRI sequences at diagnosis and/or after initial chemotherapy. Images underwent processing steps (denoising, inhomogeneity bias field correction, normalization, and resampling). Tumors were automatically segmented, and 107 shape, first-order, and second-order radiomics features were extracted, considered as the reference standard. Subsequently, the previous image processing settings were modified, and volumetric masks were applied. New radiomics features were extracted and compared with the reference standard. Reproducibility was assessed using the concordance correlation coefficient (CCC); intrasubject repeatability was measured using the coefficient of variation (CoV). Results When normalization was omitted, only 5% of the radiomics features demonstrated high reproducibility. Statistical analysis revealed significant changes in the normalization and resampling processes (P < .001). Inhomogeneities removal had the least impact on radiomics (83% of parameters remained stable). Shape features remained stable after mask modifications, with a CCC greater than 0.90. Mask modifications were the most favorable changes for achieving high CCC values, with a radiomics features stability of 70%. Only 7% of second-order radiomics features showed an excellent CoV of less than 0.10. Conclusion Modifications in the T2-weighted MRI preparation process in patients with neuroblastoma resulted in changes in radiomics features, with normalization identified as the most influential factor for reproducibility. Inhomogeneities removal had the least impact on radiomics features. Keywords: Pediatrics, MR Imaging, Oncology, Radiomics, Reproducibility, Repeatability, Neuroblastic Tumors Supplemental material is available for this article. © RSNA, 2024 See also the commentary by Safdar and Galaria in this issue.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现影响内容的错误。目的 评估神经母细胞瘤患者从 T2 加权磁共振成像中提取的放射组学特征的可重复性。材料与方法 一项回顾性研究纳入了 419 例神经母细胞瘤患者(平均(标清)年龄 29(34)岁;男性 220 例,女性 199 例),这些患者在 2002-2023 年间被诊断出患有神经母细胞瘤,属于 PRIMAGE 项目的研究范围,涉及诊断时和/或初始化疗后的 746 个 MRI T2/T2* 加权序列。图像经过了处理步骤(去噪、不均匀偏倚场校正、归一化和重采样)。自动分割肿瘤并提取 107 个形状、一阶和二阶放射学特征,作为参考标准。随后,修改了之前的图像处理设置,并应用了容积掩膜。提取新的放射组学特征并与参考标准进行比较。使用一致性相关系数(CCC)评估再现性,使用变异系数(CoV)测量受试者内的可重复性。结果 省略归一化后,只有 5%的放射组学特征显示出较高的可重复性。统计分析表明,归一化和重新取样过程发生了重大变化(P < .001)。去除不均匀性对放射组学的影响最小(83%的参数保持稳定)。掩膜修改后,形状特征保持稳定,CCC > 0.90。掩膜修改是获得高 CCC 值最有利的修改,70% 的放射组学特征保持稳定。只有 7% 的二阶放射组学特征显示出小于 0.10 的出色 CoV。结论 神经母细胞瘤患者T2加权磁共振成像制备过程的改变会导致放射组学特征的变化,而正常化被认为是对可重复性影响最大的因素。去除不均匀性对放射组学特征的影响最小。©RSNA,2024。
{"title":"Reproducibility Analysis of Radiomic Features on T2-weighted MR Images after Processing and Segmentation Alterations in Neuroblastoma Tumors.","authors":"Diana Veiga-Canuto, Matías Fernández-Patón, Leonor Cerdà Alberich, Ana Jiménez Pastor, Armando Gomis Maya, Jose Miguel Carot Sierra, Cinta Sangüesa Nebot, Blanca Martínez de Las Heras, Ulrike Pötschger, Sabine Taschner-Mandl, Emanuele Neri, Adela Cañete, Ruth Ladenstein, Barbara Hero, Ángel Alberich-Bayarri, Luis Martí-Bonmatí","doi":"10.1148/ryai.230208","DOIUrl":"10.1148/ryai.230208","url":null,"abstract":"<p><p>Purpose To evaluate the reproducibility of radiomics features extracted from T2-weighted MR images in patients with neuroblastoma. Materials and Methods A retrospective study included 419 patients (mean age, 29 months ± 34 [SD]; 220 male, 199 female) with neuroblastic tumors diagnosed between 2002 and 2023, within the scope of the PRedictive In-silico Multiscale Analytics to support cancer personalized diaGnosis and prognosis, Empowered by imaging biomarkers (ie, PRIMAGE) project, involving 746 T2/T2*-weighted MRI sequences at diagnosis and/or after initial chemotherapy. Images underwent processing steps (denoising, inhomogeneity bias field correction, normalization, and resampling). Tumors were automatically segmented, and 107 shape, first-order, and second-order radiomics features were extracted, considered as the reference standard. Subsequently, the previous image processing settings were modified, and volumetric masks were applied. New radiomics features were extracted and compared with the reference standard. Reproducibility was assessed using the concordance correlation coefficient (CCC); intrasubject repeatability was measured using the coefficient of variation (CoV). Results When normalization was omitted, only 5% of the radiomics features demonstrated high reproducibility. Statistical analysis revealed significant changes in the normalization and resampling processes (<i>P</i> < .001). Inhomogeneities removal had the least impact on radiomics (83% of parameters remained stable). Shape features remained stable after mask modifications, with a CCC greater than 0.90. Mask modifications were the most favorable changes for achieving high CCC values, with a radiomics features stability of 70%. Only 7% of second-order radiomics features showed an excellent CoV of less than 0.10. Conclusion Modifications in the T2-weighted MRI preparation process in patients with neuroblastoma resulted in changes in radiomics features, with normalization identified as the most influential factor for reproducibility. Inhomogeneities removal had the least impact on radiomics features. <b>Keywords:</b> Pediatrics, MR Imaging, Oncology, Radiomics, Reproducibility, Repeatability, Neuroblastic Tumors <i>Supplemental material is available for this article.</i> © RSNA, 2024 See also the commentary by Safdar and Galaria in this issue.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294951/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141307007","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
Deep Learning Prostate MRI Segmentation Accuracy and Robustness: A Systematic Review. 深度学习前列腺 MRI 分段准确性和鲁棒性:系统性综述。
IF 8.1 Q1 COMPUTER SCIENCE, ARTIFICIAL INTELLIGENCE Pub Date : 2024-07-01 DOI: 10.1148/ryai.230138
Mohammad-Kasim Fassia, Adithya Balasubramanian, Sungmin Woo, Hebert Alberto Vargas, Hedvig Hricak, Ender Konukoglu, Anton S Becker

Purpose To investigate the accuracy and robustness of prostate segmentation using deep learning across various training data sizes, MRI vendors, prostate zones, and testing methods relative to fellowship-trained diagnostic radiologists. Materials and Methods In this systematic review, Embase, PubMed, Scopus, and Web of Science databases were queried for English-language articles using keywords and related terms for prostate MRI segmentation and deep learning algorithms dated to July 31, 2022. A total of 691 articles from the search query were collected and subsequently filtered to 48 on the basis of predefined inclusion and exclusion criteria. Multiple characteristics were extracted from selected studies, such as deep learning algorithm performance, MRI vendor, and training dataset features. The primary outcome was comparison of mean Dice similarity coefficient (DSC) for prostate segmentation for deep learning algorithms versus diagnostic radiologists. Results Forty-eight studies were included. Most published deep learning algorithms for whole prostate gland segmentation (39 of 42 [93%]) had a DSC at or above expert level (DSC ≥ 0.86). The mean DSC was 0.79 ± 0.06 (SD) for peripheral zone, 0.87 ± 0.05 for transition zone, and 0.90 ± 0.04 for whole prostate gland segmentation. For selected studies that used one major MRI vendor, the mean DSCs of each were as follows: General Electric (three of 48 studies), 0.92 ± 0.03; Philips (four of 48 studies), 0.92 ± 0.02; and Siemens (six of 48 studies), 0.91 ± 0.03. Conclusion Deep learning algorithms for prostate MRI segmentation demonstrated accuracy similar to that of expert radiologists despite varying parameters; therefore, future research should shift toward evaluating segmentation robustness and patient outcomes across diverse clinical settings. Keywords: MRI, Genital/Reproductive, Prostate Segmentation, Deep Learning Systematic review registration link: osf.io/nxaev © RSNA, 2024.

"刚刚接受 "的论文经过同行评审,已被接受在《放射学》上发表:人工智能》上发表。这篇文章在以最终版本发表之前,还将经过校对、排版和校对审核。请注意,在制作最终校对稿的过程中,可能会发现一些错误,从而影响文章内容。目的 研究相对于接受过研究员培训的放射诊断医师,使用深度学习对各种训练数据大小、核磁共振成像供应商、前列腺区域和测试方法进行前列腺分割的准确性和鲁棒性。材料与方法 在这篇系统性综述中,我们使用关键词和相关术语在 EMBASE、PubMed、Scopus 和 Web of Science 数据库中查询了截至 2022 年 7 月 31 日有关前列腺 MRI 分割和深度学习算法的英文文章。搜索结果共收集到 691 篇文章,随后根据预定义的纳入和排除标准筛选出 48 篇文章。从所选研究中提取了多种特征,如深度学习算法性能、核磁共振成像供应商和训练数据集特征。主要结果是比较深度学习算法与放射诊断医师在前列腺分割方面的平均狄斯相似系数(DSC)。结果 共纳入 48 项研究。绝大多数已发表的全前列腺分割深度学习算法(39/42 或 93%)的 DSC 达到或超过专家水平(DSC ≥ 0.86)。外周区的平均 DSC 为 0.79 ± 0.06,过渡区为 0.87 ± 0.05,整个前列腺的平均 DSC 为 0.90 ± 0.04。对于使用一家主要核磁共振成像供应商的选定研究,每项研究的平均 DSCs 如下:通用电气(3/48 项研究)0.92 ± 0.03,飞利浦(4/48 项研究)0.92 ± 0.02,西门子(6/48 项研究)0.91 ± 0.03。结论 用于前列腺 MRI 分段的深度学习算法尽管参数不同,但其准确性与放射科专家相当,因此未来的研究应转向评估不同临床环境下的分段稳健性和患者预后。©RSNA,2024。
{"title":"Deep Learning Prostate MRI Segmentation Accuracy and Robustness: A Systematic Review.","authors":"Mohammad-Kasim Fassia, Adithya Balasubramanian, Sungmin Woo, Hebert Alberto Vargas, Hedvig Hricak, Ender Konukoglu, Anton S Becker","doi":"10.1148/ryai.230138","DOIUrl":"10.1148/ryai.230138","url":null,"abstract":"<p><p>Purpose To investigate the accuracy and robustness of prostate segmentation using deep learning across various training data sizes, MRI vendors, prostate zones, and testing methods relative to fellowship-trained diagnostic radiologists. Materials and Methods In this systematic review, Embase, PubMed, Scopus, and Web of Science databases were queried for English-language articles using keywords and related terms for prostate MRI segmentation and deep learning algorithms dated to July 31, 2022. A total of 691 articles from the search query were collected and subsequently filtered to 48 on the basis of predefined inclusion and exclusion criteria. Multiple characteristics were extracted from selected studies, such as deep learning algorithm performance, MRI vendor, and training dataset features. The primary outcome was comparison of mean Dice similarity coefficient (DSC) for prostate segmentation for deep learning algorithms versus diagnostic radiologists. Results Forty-eight studies were included. Most published deep learning algorithms for whole prostate gland segmentation (39 of 42 [93%]) had a DSC at or above expert level (DSC ≥ 0.86). The mean DSC was 0.79 ± 0.06 (SD) for peripheral zone, 0.87 ± 0.05 for transition zone, and 0.90 ± 0.04 for whole prostate gland segmentation. For selected studies that used one major MRI vendor, the mean DSCs of each were as follows: General Electric (three of 48 studies), 0.92 ± 0.03; Philips (four of 48 studies), 0.92 ± 0.02; and Siemens (six of 48 studies), 0.91 ± 0.03. Conclusion Deep learning algorithms for prostate MRI segmentation demonstrated accuracy similar to that of expert radiologists despite varying parameters; therefore, future research should shift toward evaluating segmentation robustness and patient outcomes across diverse clinical settings. <b>Keywords:</b> MRI, Genital/Reproductive, Prostate Segmentation, Deep Learning Systematic review registration link: osf.io/nxaev © RSNA, 2024.</p>","PeriodicalId":29787,"journal":{"name":"Radiology-Artificial Intelligence","volume":null,"pages":null},"PeriodicalIF":8.1,"publicationDate":"2024-07-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.ncbi.nlm.nih.gov/pmc/articles/PMC11294957/pdf/","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"140874810","PeriodicalName":null,"FirstCategoryId":null,"ListUrlMain":null,"RegionNum":0,"RegionCategory":"","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":"OA","EPubDate":null,"PubModel":null,"JCR":null,"JCRName":null,"Score":null,"Total":0}
引用次数: 0
期刊
Radiology-Artificial Intelligence
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