首页 > 最新文献

Journal of Pathology Informatics最新文献

英文 中文
Publicly available datasets of breast histopathology H&E whole-slide images: A scoping review 可公开获取的乳腺组织病理学 H&E 全切片图像数据集:范围审查
Q2 Medicine Pub Date : 2024-02-01 DOI: 10.1016/j.jpi.2024.100363
Masoud Tafavvoghi , Lars Ailo Bongo , Nikita Shvetsov , Lill-Tove Rasmussen Busund , Kajsa Møllersen

Advancements in digital pathology and computing resources have made a significant impact in the field of computational pathology for breast cancer diagnosis and treatment. However, access to high-quality labeled histopathological images of breast cancer is a big challenge that limits the development of accurate and robust deep learning models. In this scoping review, we identified the publicly available datasets of breast H&E-stained whole-slide images (WSIs) that can be used to develop deep learning algorithms. We systematically searched 9 scientific literature databases and 9 research data repositories and found 17 publicly available datasets containing 10 385 H&E WSIs of breast cancer. Moreover, we reported image metadata and characteristics for each dataset to assist researchers in selecting proper datasets for specific tasks in breast cancer computational pathology. In addition, we compiled 2 lists of breast H&E patches and private datasets as supplementary resources for researchers. Notably, only 28% of the included articles utilized multiple datasets, and only 14% used an external validation set, suggesting that the performance of other developed models may be susceptible to overestimation. The TCGA-BRCA was used in 52% of the selected studies. This dataset has a considerable selection bias that can impact the robustness and generalizability of the trained algorithms. There is also a lack of consistent metadata reporting of breast WSI datasets that can be an issue in developing accurate deep learning models, indicating the necessity of establishing explicit guidelines for documenting breast WSI dataset characteristics and metadata.

数字病理学和计算资源的进步对用于乳腺癌诊断和治疗的计算病理学领域产生了重大影响。然而,获取高质量的乳腺癌标记组织病理学图像是一个巨大的挑战,限制了准确、稳健的深度学习模型的开发。在这篇范围综述中,我们确定了可用于开发深度学习算法的公开可用的乳腺H&E染色全切片图像(WSI)数据集。我们系统地搜索了 9 个科学文献数据库和 9 个研究数据存储库,发现了 17 个公开可用的数据集,包含 10 385 张乳腺癌 H&E WSIs。此外,我们还报告了每个数据集的图像元数据和特征,以帮助研究人员为乳腺癌计算病理学的特定任务选择合适的数据集。此外,我们还编制了两份乳腺 H&E 补丁和私人数据集列表,作为研究人员的补充资源。值得注意的是,只有28%的收录文章使用了多个数据集,只有14%的文章使用了外部验证集,这表明其他已开发模型的性能可能容易被高估。52%的入选研究使用了 TCGA-BRCA。该数据集存在相当大的选择偏差,可能会影响训练算法的稳健性和普适性。此外,乳腺 WSI 数据集缺乏一致的元数据报告,这可能会成为开发精确深度学习模型的一个问题,这表明有必要制定明确的指南来记录乳腺 WSI 数据集的特征和元数据。
{"title":"Publicly available datasets of breast histopathology H&E whole-slide images: A scoping review","authors":"Masoud Tafavvoghi ,&nbsp;Lars Ailo Bongo ,&nbsp;Nikita Shvetsov ,&nbsp;Lill-Tove Rasmussen Busund ,&nbsp;Kajsa Møllersen","doi":"10.1016/j.jpi.2024.100363","DOIUrl":"10.1016/j.jpi.2024.100363","url":null,"abstract":"<div><p>Advancements in digital pathology and computing resources have made a significant impact in the field of computational pathology for breast cancer diagnosis and treatment. However, access to high-quality labeled histopathological images of breast cancer is a big challenge that limits the development of accurate and robust deep learning models. In this scoping review, we identified the publicly available datasets of breast H&amp;E-stained whole-slide images (WSIs) that can be used to develop deep learning algorithms. We systematically searched 9 scientific literature databases and 9 research data repositories and found 17 publicly available datasets containing 10 385 H&amp;E WSIs of breast cancer. Moreover, we reported image metadata and characteristics for each dataset to assist researchers in selecting proper datasets for specific tasks in breast cancer computational pathology. In addition, we compiled 2 lists of breast H&amp;E patches and private datasets as supplementary resources for researchers. Notably, only 28% of the included articles utilized multiple datasets, and only 14% used an external validation set, suggesting that the performance of other developed models may be susceptible to overestimation. The TCGA-BRCA was used in 52% of the selected studies. This dataset has a considerable selection bias that can impact the robustness and generalizability of the trained algorithms. There is also a lack of consistent metadata reporting of breast WSI datasets that can be an issue in developing accurate deep learning models, indicating the necessity of establishing explicit guidelines for documenting breast WSI dataset characteristics and metadata.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-02-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000026/pdfft?md5=e1d6b199f5ede66427075250c84de4c0&pid=1-s2.0-S2153353924000026-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139824478","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
Computational pathology: A survey review and the way forward 计算病理学:调查回顾与前进之路
Q2 Medicine Pub Date : 2024-01-14 DOI: 10.1016/j.jpi.2023.100357
Mahdi S. Hosseini , Babak Ehteshami Bejnordi , Vincent Quoc-Huy Trinh , Lyndon Chan , Danial Hasan , Xingwen Li , Stephen Yang , Taehyo Kim , Haochen Zhang , Theodore Wu , Kajanan Chinniah , Sina Maghsoudlou , Ryan Zhang , Jiadai Zhu , Samir Khaki , Andrei Buin , Fatemeh Chaji , Ala Salehi , Bich Ngoc Nguyen , Dimitris Samaras , Konstantinos N. Plataniotis

Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field’s future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.

计算病理学(CPath)是一门跨学科的科学,它加强了对医学组织病理学图像进行分析和建模的计算方法的开发。CPath 的主要目标是开发数字诊断的基础设施和工作流程,将其作为临床病理学的辅助 CAD 系统,促进癌症诊断和治疗的转型变革,而 CPath 工具主要解决这些问题。随着深度学习和计算机视觉算法的不断发展,以及数字病理学数据流的便捷性,目前 CPath 正在见证一场范式转变。尽管针对癌症图像分析推出了大量工程和科学作品,但在临床实践中采用和整合这些算法方面仍存在相当大的差距。这就提出了一个有关 CPath 发展方向和趋势的重要问题。在本文中,我们对 800 多篇论文进行了全面回顾,从应用和实施的角度探讨了问题设计所面临的挑战。我们将每篇论文编入一个模型卡片,通过研究关键作品和面临的挑战来布局当前的 CPath 领域。我们希望这能帮助社区找到相关作品,并促进对该领域未来发展方向的理解。简而言之,我们将 CPath 的发展划分为多个阶段,这些阶段需要紧密联系在一起,以应对与此类多学科科学相关的挑战。我们从以数据为中心、以模型为中心和以应用为中心的不同角度概述了这一周期。最后,我们概述了剩余的挑战,并为 CPath 的未来技术发展和临床整合提供了方向。有关本调查综述论文的最新信息和访问原始模型卡库,请参阅 GitHub。本草案的更新版本也可在 arXiv 上找到。
{"title":"Computational pathology: A survey review and the way forward","authors":"Mahdi S. Hosseini ,&nbsp;Babak Ehteshami Bejnordi ,&nbsp;Vincent Quoc-Huy Trinh ,&nbsp;Lyndon Chan ,&nbsp;Danial Hasan ,&nbsp;Xingwen Li ,&nbsp;Stephen Yang ,&nbsp;Taehyo Kim ,&nbsp;Haochen Zhang ,&nbsp;Theodore Wu ,&nbsp;Kajanan Chinniah ,&nbsp;Sina Maghsoudlou ,&nbsp;Ryan Zhang ,&nbsp;Jiadai Zhu ,&nbsp;Samir Khaki ,&nbsp;Andrei Buin ,&nbsp;Fatemeh Chaji ,&nbsp;Ala Salehi ,&nbsp;Bich Ngoc Nguyen ,&nbsp;Dimitris Samaras ,&nbsp;Konstantinos N. Plataniotis","doi":"10.1016/j.jpi.2023.100357","DOIUrl":"10.1016/j.jpi.2023.100357","url":null,"abstract":"<div><p>Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field’s future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to <span>GitHub</span><svg><path></path></svg>. Updated version of this draft can also be found from <span>arXiv</span><svg><path></path></svg>.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-14","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001712/pdfft?md5=cc2f8380838ba30f7db624e4fbf72b6e&pid=1-s2.0-S2153353923001712-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139538505","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
Use of n-grams and K-means clustering to classify data from free text bone marrow reports 使用 n-grams 和 K-means 聚类对自由文本骨髓报告中的数据进行分类
Q2 Medicine Pub Date : 2024-01-04 DOI: 10.1016/j.jpi.2023.100358
Richard F. Xiang

Natural language processing (NLP) has been used to extract information from and summarize medical reports. Currently, the most advanced NLP models require large training datasets of accurately labeled medical text. An approach to creating these large datasets is to use low resource intensive classical NLP algorithms. In this manuscript, we examined how an automated classical NLP algorithm was able to classify portions of bone marrow report text into their appropriate sections. A total of 1480 bone marrow reports were extracted from the laboratory information system of a tertiary healthcare network. The free text of these bone marrow reports were preprocessed by separating the reports into text blocks and then removing the section headers. A natural language processing algorithm involving n-grams and K-means clustering was used to classify the text blocks into their appropriate bone marrow sections. The impact of token replacement of numerical values, accession numbers, and clusters of differentiation, varying the number of centroids (1–19) and n-grams (1–5), and utilizing an ensemble algorithm were assessed. The optimal NLP model was found to employ an ensemble algorithm that incorporated token replacement, utilized 1-gram or bag of words, and 10 centroids for K-means clustering. This optimal model was able to classify text blocks with an accuracy of 89%, suggesting that classical NLP models can accurately classify portions of marrow report text.

自然语言处理(NLP)已被用于从医疗报告中提取信息并进行总结。目前,最先进的 NLP 模型需要大量准确标注医学文本的训练数据集。创建这些大型数据集的一种方法是使用低资源密集型经典 NLP 算法。在本手稿中,我们研究了自动经典 NLP 算法如何将骨髓报告文本的部分内容分类到相应的部分。我们从一个三级医疗保健网络的实验室信息系统中提取了总共 1480 份骨髓报告。对这些骨髓报告的自由文本进行了预处理,将报告分成文本块,然后删除章节标题。使用 n-grams 和 K-means 聚类的自然语言处理算法将文本块分类到相应的骨髓部分。评估了标记替换数值、加入号和分化群组、改变中心点数量(1-19)和 n-gram(1-5)以及使用集合算法的影响。结果发现,最佳的 NLP 模型采用了一种包含标记替换、使用 1 个词组或词袋以及 10 个中心点进行 K-means 聚类的集合算法。这个最佳模型能够对文本块进行分类,准确率达到 89%,这表明经典的 NLP 模型能够对骨髓报告文本的部分内容进行准确分类。
{"title":"Use of n-grams and K-means clustering to classify data from free text bone marrow reports","authors":"Richard F. Xiang","doi":"10.1016/j.jpi.2023.100358","DOIUrl":"10.1016/j.jpi.2023.100358","url":null,"abstract":"<div><p>Natural language processing (NLP) has been used to extract information from and summarize medical reports. Currently, the most advanced NLP models require large training datasets of accurately labeled medical text. An approach to creating these large datasets is to use low resource intensive classical NLP algorithms. In this manuscript, we examined how an automated classical NLP algorithm was able to classify portions of bone marrow report text into their appropriate sections. A total of 1480 bone marrow reports were extracted from the laboratory information system of a tertiary healthcare network. The free text of these bone marrow reports were preprocessed by separating the reports into text blocks and then removing the section headers. A natural language processing algorithm involving n-grams and K-means clustering was used to classify the text blocks into their appropriate bone marrow sections. The impact of token replacement of numerical values, accession numbers, and clusters of differentiation, varying the number of centroids (1–19) and n-grams (1–5), and utilizing an ensemble algorithm were assessed. The optimal NLP model was found to employ an ensemble algorithm that incorporated token replacement, utilized 1-gram or bag of words, and 10 centroids for K-means clustering. This optimal model was able to classify text blocks with an accuracy of 89%, suggesting that classical NLP models can accurately classify portions of marrow report text.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-01-04","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001724/pdfft?md5=b7e487105251a8e09617df3f8efc1607&pid=1-s2.0-S2153353923001724-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139395227","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
Artificial intelligence for human gunshot wound classification 用于人体枪伤分类的人工智能
Q2 Medicine Pub Date : 2023-12-30 DOI: 10.1016/j.jpi.2023.100361
Jerome Cheng , Carl Schmidt , Allecia Wilson , Zixi Wang , Wei Hao , Joshua Pantanowitz , Catherine Morris , Randy Tashjian , Liron Pantanowitz

Certain features are helpful in the identification of gunshot entrance and exit wounds, such as the presence of muzzle imprints, peripheral tears, stippling, bone beveling, and wound border irregularity. Some cases are less straightforward and wounds can thus pose challenges to an emergency room doctor or forensic pathologist. In recent years, deep learning has shown promise in various automated medical image classification tasks.

This study explores the feasibility of using a deep learning model to classify entry and exit gunshot wounds in digital color images. A collection of 2418 images of entrance and exit gunshot wounds were procured. Of these, 2028 entrance and 1314 exit wounds were cropped, focusing on the area around each gunshot wound. A ConvNext Tiny deep learning model was trained using the Fastai deep learning library, with a train/validation split ratio of 70/30, until a maximum validation accuracy of 92.6% was achieved. An additional 415 entrance and 293 exit wound images were collected for the test (holdout) set. The model achieved an accuracy of 87.99%, precision of 83.99%, recall of 87.71%, and F1-score 85.81% on the holdout set. Correctly classified were 88.19% of entrance wounds and 87.71% of exit wounds. The results are comparable to what a forensic pathologist can achieve without other morphologic cues. This study represents one of the first applications of artificial intelligence to the field of forensic pathology. This work demonstrates that deep learning models can discern entrance and exit gunshot wounds in digital images with high accuracy.

某些特征有助于识别枪弹出入伤口,如枪口印记、周边撕裂、条纹、骨斜面和伤口边缘不规则。有些情况则不那么简单,因此伤口会给急诊室医生或法医病理学家带来挑战。近年来,深度学习在各种自动医学图像分类任务中显示出了良好的前景。本研究探讨了使用深度学习模型对数字彩色图像中的入口和出口枪伤进行分类的可行性。本研究收集了 2418 幅入口和出口枪伤图像。对其中的 2028 个入口伤口和 1314 个出口伤口进行了裁剪,重点是每个枪伤周围的区域。使用 Fastai 深度学习库训练了 ConvNext Tiny 深度学习模型,训练/验证比例为 70/30,直到达到 92.6% 的最高验证准确率。另外还收集了 415 幅入口伤口图像和 293 幅出口伤口图像作为测试(保留)集。在保留集上,该模型的准确率为 87.99%,精确率为 83.99%,召回率为 87.71%,F1 分数为 85.81%。入口伤口的正确分类率为 88.19%,出口伤口的正确分类率为 87.71%。其结果与法医病理学家在没有其他形态线索的情况下所取得的结果相当。这项研究是人工智能在法医病理学领域的首次应用。这项工作表明,深度学习模型可以在数字图像中高精度地辨别入口和出口枪伤。
{"title":"Artificial intelligence for human gunshot wound classification","authors":"Jerome Cheng ,&nbsp;Carl Schmidt ,&nbsp;Allecia Wilson ,&nbsp;Zixi Wang ,&nbsp;Wei Hao ,&nbsp;Joshua Pantanowitz ,&nbsp;Catherine Morris ,&nbsp;Randy Tashjian ,&nbsp;Liron Pantanowitz","doi":"10.1016/j.jpi.2023.100361","DOIUrl":"10.1016/j.jpi.2023.100361","url":null,"abstract":"<div><p>Certain features are helpful in the identification of gunshot entrance and exit wounds, such as the presence of muzzle imprints, peripheral tears, stippling, bone beveling, and wound border irregularity. Some cases are less straightforward and wounds can thus pose challenges to an emergency room doctor or forensic pathologist. In recent years, deep learning has shown promise in various automated medical image classification tasks.</p><p>This study explores the feasibility of using a deep learning model to classify entry and exit gunshot wounds in digital color images. A collection of 2418 images of entrance and exit gunshot wounds were procured. Of these, 2028 entrance and 1314 exit wounds were cropped, focusing on the area around each gunshot wound. A ConvNext Tiny deep learning model was trained using the Fastai deep learning library, with a train/validation split ratio of 70/30, until a maximum validation accuracy of 92.6% was achieved. An additional 415 entrance and 293 exit wound images were collected for the test (holdout) set. The model achieved an accuracy of 87.99%, precision of 83.99%, recall of 87.71%, and F1-score 85.81% on the holdout set. Correctly classified were 88.19% of entrance wounds and 87.71% of exit wounds. The results are comparable to what a forensic pathologist can achieve without other morphologic cues. This study represents one of the first applications of artificial intelligence to the field of forensic pathology. This work demonstrates that deep learning models can discern entrance and exit gunshot wounds in digital images with high accuracy.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S215335392300175X/pdfft?md5=e69bbf6eb449bdb360e3bbcbd84c9a3a&pid=1-s2.0-S215335392300175X-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139190131","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
Multimodal Gated Mixture of Experts Using Whole Slide Image and Flow Cytometry for Multiple Instance Learning Classification of Lymphoma 利用全切片图像和流式细胞仪对淋巴瘤进行多实例学习分类的多模态门控专家混合物
Q2 Medicine Pub Date : 2023-12-29 DOI: 10.1016/j.jpi.2023.100359
Noriaki Hashimoto , Hiroyuki Hanada , Hiroaki Miyoshi , Miharu Nagaishi , Kensaku Sato , Hidekata Hontani , Koichi Ohshima , Ichiro Takeuchi

In this study, we present a deep-learning-based multimodal classification method for lymphoma diagnosis in digital pathology, which utilizes a whole slide image (WSI) as the primary image data and flow cytometry (FCM) data as auxiliary information. In pathological diagnosis of malignant lymphoma, FCM serves as valuable auxiliary information during the diagnosis process, offering useful insights into predicting the major class (superclass) of subtypes. By incorporating both images and FCM data into the classification process, we can develop a method that mimics the diagnostic process of pathologists, enhancing the explainability. In order to incorporate the hierarchical structure between superclasses and their subclasses, the proposed method utilizes a network structure that effectively combines the mixture of experts (MoE) and multiple instance learning (MIL) techniques, where MIL is widely recognized for its effectiveness in handling WSIs in digital pathology. The MoE network in the proposed method consists of a gating network for superclass classification and multiple expert networks for (sub)class classification, specialized for each superclass. To evaluate the effectiveness of our method, we conducted experiments involving a six-class classification task using 600 lymphoma cases. The proposed method achieved a classification accuracy of 72.3%, surpassing the 69.5% obtained through the straightforward combination of FCM and images, as well as the 70.2% achieved by the method using only images. Moreover, the combination of multiple weights in the MoE and MIL allows for the visualization of specific cellular and tumor regions, resulting in a highly explanatory model that cannot be attained with conventional methods. It is anticipated that by targeting a larger number of classes and increasing the number of expert networks, the proposed method could be effectively applied to the real problem of lymphoma diagnosis.

在这项研究中,我们提出了一种基于深度学习的多模态分类方法,该方法利用全切片图像(WSI)作为主要图像数据,流式细胞术(FCM)数据作为辅助信息,用于数字病理学中的淋巴瘤诊断。在恶性淋巴瘤的病理诊断中,FCM 是诊断过程中非常有价值的辅助信息,为预测亚型的主要类别(超类别)提供了有用的见解。通过将图像和 FCM 数据同时纳入分类过程,我们可以开发出一种模仿病理学家诊断过程的方法,从而提高可解释性。为了将超类与子类之间的层次结构结合起来,所提出的方法采用了一种网络结构,该结构有效地结合了专家混合(MoE)和多实例学习(MIL)技术,其中 MIL 因其在数字病理学中处理 WSI 的有效性而得到广泛认可。拟议方法中的混合专家网络由一个用于超类分类的门控网络和多个用于(子)类分类的专家网络组成,每个超类都有专门的专家网络。为了评估我们方法的有效性,我们使用 600 个淋巴瘤病例进行了六类分类任务实验。所提出的方法达到了 72.3% 的分类准确率,超过了直接结合 FCM 和图像所达到的 69.5%,也超过了只使用图像的方法所达到的 70.2%。此外,MoE 和 MIL 中多重权重的组合可实现特定细胞和肿瘤区域的可视化,从而产生传统方法无法实现的高解释性模型。预计通过针对更多的类别和增加专家网络的数量,所提出的方法可以有效地应用于淋巴瘤诊断的实际问题。
{"title":"Multimodal Gated Mixture of Experts Using Whole Slide Image and Flow Cytometry for Multiple Instance Learning Classification of Lymphoma","authors":"Noriaki Hashimoto ,&nbsp;Hiroyuki Hanada ,&nbsp;Hiroaki Miyoshi ,&nbsp;Miharu Nagaishi ,&nbsp;Kensaku Sato ,&nbsp;Hidekata Hontani ,&nbsp;Koichi Ohshima ,&nbsp;Ichiro Takeuchi","doi":"10.1016/j.jpi.2023.100359","DOIUrl":"10.1016/j.jpi.2023.100359","url":null,"abstract":"<div><p>In this study, we present a deep-learning-based multimodal classification method for lymphoma diagnosis in digital pathology, which utilizes a whole slide image (WSI) as the primary image data and flow cytometry (FCM) data as auxiliary information. In pathological diagnosis of malignant lymphoma, FCM serves as valuable auxiliary information during the diagnosis process, offering useful insights into predicting the major class (superclass) of subtypes. By incorporating both images and FCM data into the classification process, we can develop a method that mimics the diagnostic process of pathologists, enhancing the explainability. In order to incorporate the hierarchical structure between superclasses and their subclasses, the proposed method utilizes a network structure that effectively combines the mixture of experts (MoE) and multiple instance learning (MIL) techniques, where MIL is widely recognized for its effectiveness in handling WSIs in digital pathology. The MoE network in the proposed method consists of a gating network for superclass classification and multiple expert networks for (sub)class classification, specialized for each superclass. To evaluate the effectiveness of our method, we conducted experiments involving a six-class classification task using 600 lymphoma cases. The proposed method achieved a classification accuracy of 72.3%, surpassing the 69.5% obtained through the straightforward combination of FCM and images, as well as the 70.2% achieved by the method using only images. Moreover, the combination of multiple weights in the MoE and MIL allows for the visualization of specific cellular and tumor regions, resulting in a highly explanatory model that cannot be attained with conventional methods. It is anticipated that by targeting a larger number of classes and increasing the number of expert networks, the proposed method could be effectively applied to the real problem of lymphoma diagnosis.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001736/pdfft?md5=7da710e168eb41e1143a4b0663efcd4f&pid=1-s2.0-S2153353923001736-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139190722","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
Combining a deep learning model with clinical data better predicts hepatocellular carcinoma behavior following surgery 将深度学习模型与临床数据相结合,可更好地预测肝细胞癌术后行为
Q2 Medicine Pub Date : 2023-12-29 DOI: 10.1016/j.jpi.2023.100360
Benoit Schmauch , Sarah S. Elsoukkary , Amika Moro , Roma Raj , Chase J. Wehrle , Kazunari Sasaki , Julien Calderaro , Patrick Sin-Chan , Federico Aucejo , Daniel E. Roberts

Hepatocellular carcinoma (HCC) is among the most common cancers worldwide, and tumor recurrence following liver resection or transplantation is one of the highest contributors to mortality in HCC patients after surgery. Using artificial intelligence (AI), we developed an interdisciplinary model to predict HCC recurrence and patient survival following surgery. We collected whole-slide H&E images, clinical variables, and follow-up data from 300 patients with HCC who underwent transplant and 169 patients who underwent resection at the Cleveland Clinic. A deep learning model was trained to predict recurrence-free survival (RFS) and disease-specific survival (DSS) from the H&E-stained slides. Repeated cross-validation splits were used to compute robust C-index estimates, and the results were compared to those obtained by fitting a Cox proportional hazard model using only clinical variables. While the deep learning model alone was predictive of recurrence and survival among patients in both cohorts, integrating the clinical and histologic models significantly increased the C-index in each cohort. In every subgroup analyzed, we found that a combined clinical and deep learning model better predicted post-surgical outcome in HCC patients compared to either approach independently.

肝细胞癌(HCC)是全球最常见的癌症之一,肝切除或移植后肿瘤复发是导致 HCC 患者术后死亡率最高的因素之一。我们利用人工智能(AI)开发了一种跨学科模型,用于预测HCC术后复发和患者生存率。我们收集了克利夫兰诊所接受移植手术的 300 名 HCC 患者和接受切除手术的 169 名患者的全切片 H&E 图像、临床变量和随访数据。我们训练了一个深度学习模型,以便从H&E染色切片中预测无复发生存率(RFS)和疾病特异性生存率(DSS)。重复交叉验证用于计算稳健的 C 指数估计值,并将结果与仅使用临床变量拟合 Cox 比例危险模型得出的结果进行比较。虽然单独的深度学习模型可以预测两个队列中患者的复发率和存活率,但整合临床和组织学模型后,每个队列中的C指数都有显著提高。在分析的每个亚组中,我们都发现,与单独使用其中一种方法相比,结合临床和深度学习的模型能更好地预测 HCC 患者的术后预后。
{"title":"Combining a deep learning model with clinical data better predicts hepatocellular carcinoma behavior following surgery","authors":"Benoit Schmauch ,&nbsp;Sarah S. Elsoukkary ,&nbsp;Amika Moro ,&nbsp;Roma Raj ,&nbsp;Chase J. Wehrle ,&nbsp;Kazunari Sasaki ,&nbsp;Julien Calderaro ,&nbsp;Patrick Sin-Chan ,&nbsp;Federico Aucejo ,&nbsp;Daniel E. Roberts","doi":"10.1016/j.jpi.2023.100360","DOIUrl":"10.1016/j.jpi.2023.100360","url":null,"abstract":"<div><p>Hepatocellular carcinoma (HCC) is among the most common cancers worldwide, and tumor recurrence following liver resection or transplantation is one of the highest contributors to mortality in HCC patients after surgery. Using artificial intelligence (AI), we developed an interdisciplinary model to predict HCC recurrence and patient survival following surgery. We collected whole-slide H&amp;E images, clinical variables, and follow-up data from 300 patients with HCC who underwent transplant and 169 patients who underwent resection at the Cleveland Clinic. A deep learning model was trained to predict recurrence-free survival (RFS) and disease-specific survival (DSS) from the H&amp;E-stained slides. Repeated cross-validation splits were used to compute robust C-index estimates, and the results were compared to those obtained by fitting a Cox proportional hazard model using only clinical variables. While the deep learning model alone was predictive of recurrence and survival among patients in both cohorts, integrating the clinical and histologic models significantly increased the C-index in each cohort. In every subgroup analyzed, we found that a combined clinical and deep learning model better predicted post-surgical outcome in HCC patients compared to either approach independently.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-29","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001748/pdfft?md5=765c0e6b2719108fb46126309088e40a&pid=1-s2.0-S2153353923001748-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139191922","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
SlideTiler: A dataset creator software for boosting deep learning on histological whole slide images SlideTiler:一款数据集创建软件,用于在组织学全切片图像上提升深度学习能力
Q2 Medicine Pub Date : 2023-12-08 DOI: 10.1016/j.jpi.2023.100356
Leonardo Barcellona , Lorenzo Nicolè , Rocco Cappellesso , Angelo Paolo Dei Tos , Stefano Ghidoni

The introduction of deep learning caused a significant breakthrough in digital pathology. Thanks to its capability of mining hidden data patterns in digitised histological slides to resolve diagnostic tasks and extract prognostic and predictive information. However, the high performance achieved in classification tasks depends on the availability of large datasets, whose collection and preprocessing are still time-consuming processes. Therefore, strategies to make these steps more efficient are worth investigation. This work introduces SlideTiler, an open-source software with a user-friendly graphical interface. SlideTiler can manage several image preprocessing phases through an intuitive workflow that does not require specific coding skills. The software was designed to provide direct access to virtual slides, allowing custom tiling of specific regions of interest drawn by the user, tile labelling, quality assessment, and direct export to dataset directories. To illustrate the functions and the scalability of SlideTiler, a deep learning-based classifier was implemented to classify 4 different tumour histotypes available in the TCGA repository. The results demonstrate the effectiveness of SlideTiler in facilitating data preprocessing and promoting accessibility to digitised pathology images for research purposes. Considering the increasing interest in deep learning applications of digital pathology, SlideTiler has a positive impact on this field. Moreover, SlideTiler has been conceived as a dynamic tool in constant evolution, and more updated and efficient versions will be released in the future.

深度学习的引入为数字病理学带来了重大突破。由于深度学习能够挖掘数字化组织学切片中隐藏的数据模式,从而解决诊断任务并提取预后和预测信息。然而,分类任务所取得的高性能取决于大型数据集的可用性,而数据集的收集和预处理仍然是耗时的过程。因此,提高这些步骤效率的策略值得研究。本作品介绍了一款图形界面友好的开源软件 SlideTiler。SlideTiler 可通过直观的工作流程管理多个图像预处理阶段,无需特定的编码技能。该软件旨在提供对虚拟幻灯片的直接访问,允许对用户绘制的特定感兴趣区域进行自定义平铺、平铺标记、质量评估以及直接导出到数据集目录。为了说明 SlideTiler 的功能和可扩展性,我们实施了一个基于深度学习的分类器,对 TCGA 存储库中的 4 种不同肿瘤组织型进行分类。结果证明了 SlideTiler 在促进数据预处理和提高数字化病理图像的可访问性方面的有效性。考虑到人们对数字病理学深度学习应用的兴趣与日俱增,SlideTiler 将对这一领域产生积极影响。此外,SlideTiler 被认为是一个不断发展的动态工具,未来还将推出更多更新、更高效的版本。
{"title":"SlideTiler: A dataset creator software for boosting deep learning on histological whole slide images","authors":"Leonardo Barcellona ,&nbsp;Lorenzo Nicolè ,&nbsp;Rocco Cappellesso ,&nbsp;Angelo Paolo Dei Tos ,&nbsp;Stefano Ghidoni","doi":"10.1016/j.jpi.2023.100356","DOIUrl":"10.1016/j.jpi.2023.100356","url":null,"abstract":"<div><p>The introduction of deep learning caused a significant breakthrough in digital pathology. Thanks to its capability of mining hidden data patterns in digitised histological slides to resolve diagnostic tasks and extract prognostic and predictive information. However, the high performance achieved in classification tasks depends on the availability of large datasets, whose collection and preprocessing are still time-consuming processes. Therefore, strategies to make these steps more efficient are worth investigation. This work introduces SlideTiler, an open-source software with a user-friendly graphical interface. SlideTiler can manage several image preprocessing phases through an intuitive workflow that does not require specific coding skills. The software was designed to provide direct access to virtual slides, allowing custom tiling of specific regions of interest drawn by the user, tile labelling, quality assessment, and direct export to dataset directories. To illustrate the functions and the scalability of SlideTiler, a deep learning-based classifier was implemented to classify 4 different tumour histotypes available in the TCGA repository. The results demonstrate the effectiveness of SlideTiler in facilitating data preprocessing and promoting accessibility to digitised pathology images for research purposes. Considering the increasing interest in deep learning applications of digital pathology, SlideTiler has a positive impact on this field. Moreover, SlideTiler has been conceived as a dynamic tool in constant evolution, and more updated and efficient versions will be released in the future.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-12-08","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001700/pdfft?md5=8704f1d3116c95cedb709a6224e1022e&pid=1-s2.0-S2153353923001700-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138624638","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
Artificial intelligence (AI) for tumor microenvironment (TME) and tumor budding (TB) identification in colorectal cancer (CRC) patients: A systematic review 人工智能(AI)用于识别结直肠癌(CRC)患者的肿瘤微环境(TME)和肿瘤萌芽(TB):系统综述
Q2 Medicine Pub Date : 2023-11-22 DOI: 10.1016/j.jpi.2023.100353
Olga Andreevna Lobanova , Anastasia Olegovna Kolesnikova , Valeria Aleksandrovna Ponomareva , Ksenia Andreevna Vekhova , Anaida Lusparonovna Shaginyan , Alisa Borisovna Semenova , Dmitry Petrovich Nekhoroshkov , Svetlana Evgenievna Kochetkova , Natalia Valeryevna Kretova , Alexander Sergeevich Zanozin , Maria Alekseevna Peshkova , Natalia Borisovna Serezhnikova , Nikolay Vladimirovich Zharkov , Evgeniya Altarovna Kogan , Alexander Alekseevich Biryukov , Ekaterina Evgenievna Rudenko , Tatiana Alexandrovna Demura

Evaluation of the parameters such as tumor microenvironment (TME) and tumor budding (TB) is one of the most important steps in colorectal cancer (CRC) diagnosis and cancer development prognosis. In recent years, artificial intelligence (AI) has been successfully used to solve such problems. In this paper, we summarize the latest data on the use of artificial intelligence to predict tumor microenvironment and tumor budding in histological scans of patients with colorectal cancer. We performed a systematic literature search using 2 databases (Medline and Scopus) with the following search terms: ("tumor microenvironment" OR "tumor budding") AND ("colorectal cancer" OR CRC) AND ("artificial intelligence" OR "machine learning " OR "deep learning"). During the analysis, we gathered from the articles performance scores such as sensitivity, specificity, and accuracy of identifying TME and TB using artificial intelligence. The systematic review showed that machine learning and deep learning successfully cope with the prediction of these parameters. The highest accuracy values in TB and TME prediction were 97.7% and 97.3%, respectively. This review led us to the conclusion that AI platforms can already be used as diagnostic aids, which will greatly facilitate the work of pathologists in detection and estimation of TB and TME as instruments and second-opinion services. A key limitation in writing this systematic review was the heterogeneous use of performance metrics for machine learning models by different authors, as well as relatively small datasets used in some studies.

肿瘤微环境(TME)和肿瘤萌芽(TB)等参数的评估是结直肠癌(CRC)诊断和癌症发展预后中最重要的步骤之一。近年来,人工智能(AI)已被成功用于解决此类问题。在本文中,我们总结了利用人工智能预测结直肠癌患者组织学扫描中肿瘤微环境和肿瘤萌芽的最新数据。我们使用两个数据库(Medline 和 Scopus)进行了系统的文献检索,检索词如下:("肿瘤微环境 "或 "肿瘤萌芽")和("结直肠癌 "或 CRC)和("人工智能 "或 "机器学习 "或 "深度学习")。在分析过程中,我们从文章中收集了使用人工智能识别 TME 和 TB 的敏感性、特异性和准确性等性能评分。系统综述显示,机器学习和深度学习成功地应对了这些参数的预测。结核病和TME预测的最高准确率分别为97.7%和97.3%。这一综述使我们得出结论:人工智能平台已经可以用作诊断辅助工具,这将极大地促进病理学家在结核病和TME的检测和估算方面的工作,并将其作为仪器和第二意见服务。撰写本系统综述的一个主要限制因素是不同作者对机器学习模型性能指标的使用不尽相同,以及一些研究中使用的数据集相对较小。
{"title":"Artificial intelligence (AI) for tumor microenvironment (TME) and tumor budding (TB) identification in colorectal cancer (CRC) patients: A systematic review","authors":"Olga Andreevna Lobanova ,&nbsp;Anastasia Olegovna Kolesnikova ,&nbsp;Valeria Aleksandrovna Ponomareva ,&nbsp;Ksenia Andreevna Vekhova ,&nbsp;Anaida Lusparonovna Shaginyan ,&nbsp;Alisa Borisovna Semenova ,&nbsp;Dmitry Petrovich Nekhoroshkov ,&nbsp;Svetlana Evgenievna Kochetkova ,&nbsp;Natalia Valeryevna Kretova ,&nbsp;Alexander Sergeevich Zanozin ,&nbsp;Maria Alekseevna Peshkova ,&nbsp;Natalia Borisovna Serezhnikova ,&nbsp;Nikolay Vladimirovich Zharkov ,&nbsp;Evgeniya Altarovna Kogan ,&nbsp;Alexander Alekseevich Biryukov ,&nbsp;Ekaterina Evgenievna Rudenko ,&nbsp;Tatiana Alexandrovna Demura","doi":"10.1016/j.jpi.2023.100353","DOIUrl":"https://doi.org/10.1016/j.jpi.2023.100353","url":null,"abstract":"<div><p>Evaluation of the parameters such as tumor microenvironment (TME) and tumor budding (TB) is one of the most important steps in colorectal cancer (CRC) diagnosis and cancer development prognosis. In recent years, artificial intelligence (AI) has been successfully used to solve such problems. In this paper, we summarize the latest data on the use of artificial intelligence to predict tumor microenvironment and tumor budding in histological scans of patients with colorectal cancer. We performed a systematic literature search using 2 databases (Medline and Scopus) with the following search terms: (\"tumor microenvironment\" OR \"tumor budding\") AND (\"colorectal cancer\" OR CRC) AND (\"artificial intelligence\" OR \"machine learning \" OR \"deep learning\"). During the analysis, we gathered from the articles performance scores such as sensitivity, specificity, and accuracy of identifying TME and TB using artificial intelligence. The systematic review showed that machine learning and deep learning successfully cope with the prediction of these parameters. The highest accuracy values in TB and TME prediction were 97.7% and 97.3%, respectively. This review led us to the conclusion that AI platforms can already be used as diagnostic aids, which will greatly facilitate the work of pathologists in detection and estimation of TB and TME as instruments and second-opinion services. A key limitation in writing this systematic review was the heterogeneous use of performance metrics for machine learning models by different authors, as well as relatively small datasets used in some studies.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001670/pdfft?md5=02bdc3221d561cd21e9de3f4dfe5954d&pid=1-s2.0-S2153353923001670-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"139038548","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 deep learning model to predict Ki-67 positivity in oral squamous cell carcinoma 预测口腔鳞状细胞癌 Ki-67 阳性的深度学习模型
Q2 Medicine Pub Date : 2023-11-22 DOI: 10.1016/j.jpi.2023.100354
Francesco Martino , Gennaro Ilardi , Silvia Varricchio , Daniela Russo , Rosa Maria Di Crescenzo , Stefania Staibano , Francesco Merolla

Anatomical pathology is undergoing its third revolution, transitioning from analogical to digital pathology and incorporating new artificial intelligence technologies into clinical practice. Aside from classification, detection, and segmentation models, predictive models are gaining traction since they can impact diagnostic processes and laboratory activity, lowering consumable usage and turnaround time. Our research aimed to create a deep-learning model to generate synthetic Ki-67 immunohistochemistry from Haematoxylin and Eosin (H&E) stained images. We used 175 oral squamous cell carcinoma (OSCC) from the University Federico II’s Pathology Unit’s archives to train our model to generate 4 Tissue Micro Arrays (TMAs). We sectioned one slide from each TMA, first stained with H&E and then re-stained with anti-Ki-67 immunohistochemistry (IHC). In digitised slides, cores were disarrayed, and the matching cores of the 2 stained were aligned to construct a dataset to train a Pix2Pix algorithm to convert H&E images to IHC. Pathologists could recognise the synthetic images in only half of the cases in a specially designed likelihood test. Hence, our model produced realistic synthetic images. We next used QuPath to quantify IHC positivity, achieving remarkable levels of agreement between genuine and synthetic IHC.

Furthermore, a categorical analysis employing 3 Ki-67 positivity cut-offs (5%, 10%, and 15%) revealed high positive-predictive values. Our model is a promising tool for collecting Ki-67 positivity information directly on H&E slides, reducing laboratory demand and improving patient management. It is also a valuable option for smaller laboratories to easily and quickly screen bioptic samples and prioritise them in a digital pathology workflow.

解剖病理学正在经历第三次革命,从模拟病理学过渡到数字病理学,并将新的人工智能技术纳入临床实践。除了分类、检测和分割模型外,预测模型也越来越受到重视,因为它们可以影响诊断流程和实验室活动,降低耗材使用率,缩短周转时间。我们的研究旨在创建一个深度学习模型,从血红素和伊红(H&E)染色图像中生成合成的 Ki-67 免疫组化结果。我们使用费德里科二世大学病理科档案中的 175 例口腔鳞状细胞癌(OSCC)来训练模型,生成 4 个组织微阵列(TMA)。我们从每个 TMA 中切下一张切片,先用 H&E 染色,然后用抗-Ki-67 免疫组化 (IHC) 重新染色。在数字化的切片中,我们对核心部分进行了排列,并将两种染色的匹配核心部分对齐,以构建一个数据集来训练 Pix2Pix 算法,从而将 H&E 图像转换为 IHC 图像。在专门设计的似然性测试中,病理学家仅能识别半数病例的合成图像。因此,我们的模型生成了真实的合成图像。接下来,我们使用 QuPath 对 IHC 阳性进行量化,结果发现真实 IHC 与合成 IHC 之间的一致性非常高。我们的模型是一种很有前途的工具,可直接在 H&E 切片上收集 Ki-67 阳性信息,减少实验室需求并改善患者管理。对于规模较小的实验室来说,它也是一种有价值的选择,可以方便快捷地筛选生物样本,并在数字病理工作流程中对其进行优先排序。
{"title":"A deep learning model to predict Ki-67 positivity in oral squamous cell carcinoma","authors":"Francesco Martino ,&nbsp;Gennaro Ilardi ,&nbsp;Silvia Varricchio ,&nbsp;Daniela Russo ,&nbsp;Rosa Maria Di Crescenzo ,&nbsp;Stefania Staibano ,&nbsp;Francesco Merolla","doi":"10.1016/j.jpi.2023.100354","DOIUrl":"https://doi.org/10.1016/j.jpi.2023.100354","url":null,"abstract":"<div><p>Anatomical pathology is undergoing its third revolution, transitioning from analogical to digital pathology and incorporating new artificial intelligence technologies into clinical practice. Aside from classification, detection, and segmentation models, predictive models are gaining traction since they can impact diagnostic processes and laboratory activity, lowering consumable usage and turnaround time. Our research aimed to create a deep-learning model to generate synthetic Ki-67 immunohistochemistry from Haematoxylin and Eosin (H&amp;E) stained images. We used 175 oral squamous cell carcinoma (OSCC) from the University Federico II’s Pathology Unit’s archives to train our model to generate 4 Tissue Micro Arrays (TMAs). We sectioned one slide from each TMA, first stained with H&amp;E and then re-stained with anti-Ki-67 immunohistochemistry (IHC). In digitised slides, cores were disarrayed, and the matching cores of the 2 stained were aligned to construct a dataset to train a Pix2Pix algorithm to convert H&amp;E images to IHC. Pathologists could recognise the synthetic images in only half of the cases in a specially designed likelihood test. Hence, our model produced realistic synthetic images. We next used QuPath to quantify IHC positivity, achieving remarkable levels of agreement between genuine and synthetic IHC.</p><p>Furthermore, a categorical analysis employing 3 Ki-67 positivity cut-offs (5%, 10%, and 15%) revealed high positive-predictive values. Our model is a promising tool for collecting Ki-67 positivity information directly on H&amp;E slides, reducing laboratory demand and improving patient management. It is also a valuable option for smaller laboratories to easily and quickly screen bioptic samples and prioritise them in a digital pathology workflow.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-22","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001682/pdfft?md5=1e50c2def78451a443dc9e3c3d022cbc&pid=1-s2.0-S2153353923001682-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138558826","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
Digital analysis of the prostate tumor microenvironment with high-order chromogenic multiplexing 利用高阶色原复用技术对前列腺肿瘤微环境进行数字化分析
Q2 Medicine Pub Date : 2023-11-21 DOI: 10.1016/j.jpi.2023.100352
Rahul Rajendran, Rachel C. Beck, Morteza M. Waskasi, Brian D. Kelly, Daniel R. Bauer

As our understanding of the tumor microenvironment grows, the pathology field is increasingly utilizing multianalyte diagnostic assays to understand important characteristics of tumor growth. In clinical settings, brightfield chromogenic assays represent the gold-standard and have developed significant trust as the first-line diagnostic method. However, conventional brightfield tests have been limited to low-order assays that are visually interrogated. We have developed a hybrid method of brightfield chromogenic multiplexing that overcomes these limitations and enables high-order multiplex assays. However, how compatible high-order brightfield multiplexed images are with advanced analytical algorithms has not been extensively evaluated. In the present study, we address this gap by developing a novel 6-marker prostate cancer assay that targets diverse aspects of the tumor microenvironment such as prostate-specific biomarkers (PSMA and p504s), immune biomarkers (CD8 and PD-L1), a prognostic biomarker (Ki-67), as well as an adjunctive diagnostic biomarker (basal cell cocktail) and apply the assay to 143 differentially graded adenocarcinoma prostate tissues. The tissues were then imaged on our spectroscopic multiplexing imaging platform and mined for proteomic and spatial features that were correlated with cancer presence and disease grade. Extracted features were used to train a UMAP model that differentiated healthy from cancerous tissue with an accuracy of 89% and identified clusters of cells based on cancer grade. For spatial analysis, cell-to-cell distances were calculated for all biomarkers and differences between healthy and adenocarcinoma tissues were studied. We report that p504s positive cells were at least 2× closer to cells expressing PD-L1, CD8, Ki-67, and basal cell in adenocarcinoma tissues relative to the healthy control tissues. These findings offer a powerful insight to understand the fingerprint of the prostate tumor microenvironment and indicate that high-order chromogenic multiplexing is compatible with digital analysis. Thus, the presented chromogenic multiplexing system combines the clinical applicability of brightfield assays with the emerging diagnostic power of high-order multiplexing in a digital pathology friendly format that is well-suited for translational studies to better understand mechanisms of tumor development and growth.

随着我们对肿瘤微环境了解的加深,病理领域越来越多地利用多分析物诊断分析来了解肿瘤生长的重要特征。在临床环境中,明场显色法代表了金标准,并作为一线诊断方法发展了显著的信任。然而,传统的明场测试仅限于低阶分析,是视觉询问。我们开发了一种混合的明场显色多路复用方法,克服了这些限制,实现了高阶多路复用分析。然而,如何兼容高阶明场复用图像与先进的分析算法还没有得到广泛的评估。在本研究中,我们通过开发一种新的6标记前列腺癌检测来解决这一空白,该检测针对肿瘤微环境的各个方面,如前列腺特异性生物标志物(PSMA和p504s)、免疫生物标志物(CD8和PD-L1)、预后生物标志物(Ki-67)以及辅助诊断生物标志物(基底细胞混合物),并将该检测应用于143种不同分级的腺癌前列腺组织。然后在我们的光谱多路复用成像平台上对组织进行成像,并挖掘与癌症存在和疾病等级相关的蛋白质组学和空间特征。提取的特征用于训练UMAP模型,该模型区分健康组织和癌组织的准确率为89%,并根据癌症等级识别细胞簇。为了进行空间分析,计算了所有生物标志物的细胞间距离,并研究了健康组织和腺癌组织之间的差异。我们报道p504s阳性细胞与腺癌组织中表达PD-L1、CD8、Ki-67和基底细胞的细胞至少接近健康对照组织的2倍。这些发现为理解前列腺肿瘤微环境的指纹图谱提供了有力的见解,并表明高阶显色复用与数字分析是兼容的。因此,提出的显色多路复用系统结合了明场分析的临床适用性和高阶多路复用的新兴诊断能力,以数字病理友好的形式,非常适合用于转化研究,以更好地了解肿瘤的发展和生长机制。
{"title":"Digital analysis of the prostate tumor microenvironment with high-order chromogenic multiplexing","authors":"Rahul Rajendran,&nbsp;Rachel C. Beck,&nbsp;Morteza M. Waskasi,&nbsp;Brian D. Kelly,&nbsp;Daniel R. Bauer","doi":"10.1016/j.jpi.2023.100352","DOIUrl":"https://doi.org/10.1016/j.jpi.2023.100352","url":null,"abstract":"<div><p>As our understanding of the tumor microenvironment grows, the pathology field is increasingly utilizing multianalyte diagnostic assays to understand important characteristics of tumor growth. In clinical settings, brightfield chromogenic assays represent the gold-standard and have developed significant trust as the first-line diagnostic method. However, conventional brightfield tests have been limited to low-order assays that are visually interrogated. We have developed a hybrid method of brightfield chromogenic multiplexing that overcomes these limitations and enables high-order multiplex assays. However, how compatible high-order brightfield multiplexed images are with advanced analytical algorithms has not been extensively evaluated. In the present study, we address this gap by developing a novel 6-marker prostate cancer assay that targets diverse aspects of the tumor microenvironment such as prostate-specific biomarkers (PSMA and p504s), immune biomarkers (CD8 and PD-L1), a prognostic biomarker (Ki-67), as well as an adjunctive diagnostic biomarker (basal cell cocktail) and apply the assay to 143 differentially graded adenocarcinoma prostate tissues. The tissues were then imaged on our spectroscopic multiplexing imaging platform and mined for proteomic and spatial features that were correlated with cancer presence and disease grade. Extracted features were used to train a UMAP model that differentiated healthy from cancerous tissue with an accuracy of 89% and identified clusters of cells based on cancer grade. For spatial analysis, cell-to-cell distances were calculated for all biomarkers and differences between healthy and adenocarcinoma tissues were studied. We report that p504s positive cells were at least 2× closer to cells expressing PD-L1, CD8, Ki-67, and basal cell in adenocarcinoma tissues relative to the healthy control tissues. These findings offer a powerful insight to understand the fingerprint of the prostate tumor microenvironment and indicate that high-order chromogenic multiplexing is compatible with digital analysis. Thus, the presented chromogenic multiplexing system combines the clinical applicability of brightfield assays with the emerging diagnostic power of high-order multiplexing in a digital pathology friendly format that is well-suited for translational studies to better understand mechanisms of tumor development and growth.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2023-11-21","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353923001669/pdfft?md5=0e916a6f16983a6fd85779bc435e915c&pid=1-s2.0-S2153353923001669-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"138633481","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
Journal of Pathology Informatics
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