首页 > 最新文献

Journal of Pathology Informatics最新文献

英文 中文
Digital mapping of resected cancer specimens: The visual pathology report 对切除的癌症标本进行数字绘图:可视病理报告
Q2 Medicine Pub Date : 2024-09-28 DOI: 10.1016/j.jpi.2024.100399

Background

The current standard-of-care pathology report relies only on lengthy written text descriptions without a visual representation of the resected cancer specimen. This study demonstrates the feasibility of incorporating virtual, three-dimensional (3D) visual pathology reports to improve communication of final pathology reporting.

Materials and methods

Surgical specimens are 3D scanned and virtually mapped alongside the pathology team to replicate grossing. The 3D specimen maps are incorporated into a hybrid visual pathology report which displays the resected specimen and sampled margins alongside gross measurements, tumor characteristics, and microscopic diagnoses.

Results

Visual pathology reports were created for 10 head and neck cancer cases. Each report concisely communicated information from the final pathology report in a single page and contained significantly fewer words (293.4 words) than standard written pathology reports (850.1 words, p < 0.01).

Conclusions

We establish the feasibility of a novel visual pathology report that includes an annotated visual model of the resected cancer specimen in place of lengthy written text of standard of care head and neck cancer pathology reports.
背景目前的标准病理报告仅依赖于冗长的书面文字描述,而没有切除癌症标本的视觉呈现。材料和方法对手术标本进行三维扫描,并与病理团队一起绘制虚拟标本图,以复制大体标本。三维标本图被纳入混合可视病理报告中,该报告显示切除标本和取样边缘,以及大体测量结果、肿瘤特征和显微诊断。每份报告都在一页纸上简明扼要地传达了最终病理报告的信息,与标准的书面病理报告(850.1 字,p <0.01)相比,字数明显减少(293.4 字)。结论我们确定了一种新型可视化病理报告的可行性,这种报告包括切除癌症标本的注释可视化模型,以取代头颈部癌症病理报告中冗长的书面文字。
{"title":"Digital mapping of resected cancer specimens: The visual pathology report","authors":"","doi":"10.1016/j.jpi.2024.100399","DOIUrl":"10.1016/j.jpi.2024.100399","url":null,"abstract":"<div><h3>Background</h3><div>The current standard-of-care pathology report relies only on lengthy written text descriptions without a visual representation of the resected cancer specimen. This study demonstrates the feasibility of incorporating virtual, three-dimensional (3D) visual pathology reports to improve communication of final pathology reporting.</div></div><div><h3>Materials and methods</h3><div>Surgical specimens are 3D scanned and virtually mapped alongside the pathology team to replicate grossing. The 3D specimen maps are incorporated into a hybrid visual pathology report which displays the resected specimen and sampled margins alongside gross measurements, tumor characteristics, and microscopic diagnoses.</div></div><div><h3>Results</h3><div>Visual pathology reports were created for 10 head and neck cancer cases. Each report concisely communicated information from the final pathology report in a single page and contained significantly fewer words (293.4 words) than standard written pathology reports (850.1 words, <em>p</em> &lt; 0.01).</div></div><div><h3>Conclusions</h3><div>We establish the feasibility of a novel visual pathology report that includes an annotated visual model of the resected cancer specimen in place of lengthy written text of standard of care head and neck cancer pathology reports.</div></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-28","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142432514","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 precise machine learning model: Detecting cervical cancer using feature selection and explainable AI 精确的机器学习模型:利用特征选择和可解释人工智能检测宫颈癌
Q2 Medicine Pub Date : 2024-09-26 DOI: 10.1016/j.jpi.2024.100398
Cervical cancer is a cancer that remains a significant global health challenge all over the world. Due to improper screening in the early stages, and healthcare disparities, a large number of women are suffering from this disease, and the mortality rate increases day by day. Hence, in these studies, we presented a precise approach utilizing six different machine learning models (decision tree, logistic regression, naïve bayes, random forest, k nearest neighbors, support vector machine), which can predict the early stage of cervical cancer by analysing 36 risk factor attributes of 858 individuals. In addition, two data balancing techniques—Synthetic Minority Oversampling Technique and Adaptive Synthetic Sampling—were used to mitigate the data imbalance issues. Furthermore, Chi-square and Least Absolute Shrinkage and Selection Operator are two distinct feature selection processes that have been applied to evaluate the feature rank, which are mostly correlated to identify the particular disease, and also integrate an explainable artificial intelligence technique, namely Shapley Additive Explanations, for clarifying the model outcome. The applied machine learning model outcome is evaluated by performance evaluation matrices, namely accuracy, sensitivity, specificity, precision, f1-score, false-positive rate and false-negative rate, and area under the Receiver operating characteristic curve score. The decision tree outperformed in Chi-square feature selection with outstanding accuracy with 97.60%, 98.73% sensitivity, 80% specificity, and 98.73% precision, respectively. During the data imbalance, DT performed 97% accuracy, 99.35% sensitivity, 69.23% specificity, and 97.45% precision. This research is focused on developing diagnostic frameworks with automated tools to improve the detection and management of cervical cancer, as well as on helping healthcare professionals deliver more efficient and personalized care to their patients.
宫颈癌是一种癌症,在全世界仍然是一项重大的全球健康挑战。由于早期筛查不当和医疗保健方面的不平等,大量妇女罹患此病,死亡率与日俱增。因此,在这些研究中,我们提出了一种精确的方法,利用六种不同的机器学习模型(决策树、逻辑回归、奈夫贝叶斯、随机森林、k 近邻、支持向量机),通过分析 858 人的 36 个风险因素属性,预测宫颈癌的早期阶段。此外,还采用了两种数据平衡技术--合成少数群体过度采样技术和自适应合成采样技术,以缓解数据不平衡问题。此外,Chi-square 和 Least Absolute Shrinkage and Selection Operator 是两种不同的特征选择过程,用于评估特征等级,这些特征等级大多与特定疾病的识别相关,同时还集成了一种可解释的人工智能技术,即 Shapley Additive Explanations,用于澄清模型结果。应用机器学习模型的结果由性能评价矩阵进行评价,即准确度、灵敏度、特异性、精确度、f1-分数、假阳性率、假阴性率和接收者工作特征曲线下面积分数。决策树的准确率、灵敏度、特异度和精确度分别为 97.60%、98.73%、80% 和 98.73%,优于 Chi-square 特征选择。在数据不平衡时,决策树的准确率为 97%,灵敏度为 99.35%,特异度为 69.23%,精确度为 97.45%。这项研究的重点是开发带有自动化工具的诊断框架,以改善宫颈癌的检测和管理,并帮助医疗保健专业人员为患者提供更高效、更个性化的护理。
{"title":"A precise machine learning model: Detecting cervical cancer using feature selection and explainable AI","authors":"","doi":"10.1016/j.jpi.2024.100398","DOIUrl":"10.1016/j.jpi.2024.100398","url":null,"abstract":"<div><div>Cervical cancer is a cancer that remains a significant global health challenge all over the world. Due to improper screening in the early stages, and healthcare disparities, a large number of women are suffering from this disease, and the mortality rate increases day by day. Hence, in these studies, we presented a precise approach utilizing six different machine learning models (decision tree, logistic regression, naïve bayes, random forest, k nearest neighbors, support vector machine), which can predict the early stage of cervical cancer by analysing 36 risk factor attributes of 858 individuals. In addition, two data balancing techniques—Synthetic Minority Oversampling Technique and Adaptive Synthetic Sampling—were used to mitigate the data imbalance issues. Furthermore, Chi-square and Least Absolute Shrinkage and Selection Operator are two distinct feature selection processes that have been applied to evaluate the feature rank, which are mostly correlated to identify the particular disease, and also integrate an explainable artificial intelligence technique, namely Shapley Additive Explanations, for clarifying the model outcome. The applied machine learning model outcome is evaluated by performance evaluation matrices, namely accuracy, sensitivity, specificity, precision, f1-score, false-positive rate and false-negative rate, and area under the Receiver operating characteristic curve score. The decision tree outperformed in Chi-square feature selection with outstanding accuracy with 97.60%, 98.73% sensitivity, 80% specificity, and 98.73% precision, respectively. During the data imbalance, DT performed 97% accuracy, 99.35% sensitivity, 69.23% specificity, and 97.45% precision. This research is focused on developing diagnostic frameworks with automated tools to improve the detection and management of cervical cancer, as well as on helping healthcare professionals deliver more efficient and personalized care to their patients.</div></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-26","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142442480","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
ViCE: An automated and quantitative program to assess intestinal tissue morphology ViCE:自动定量评估肠道组织形态的程序
Q2 Medicine Pub Date : 2024-09-13 DOI: 10.1016/j.jpi.2024.100397

Background and objective

The tissue morphology of the intestinal surface is architecturally complex with finger-like projections called villi, and glandular structures called crypts. The ratio of villus height-to-crypt depth ratio (Vh:Cd) is used to quantitatively assess disease severity and response to therapy for intestinal enteropathies, such as celiac disease and is currently quantified manually. Given the time required, manual Vh:Cd measurements have largely been limited to clinical trials and are not used widely in clinical practice. We developed ViCE (Villus Crypt Evaluator), a user-friendly software that automatically quantifies histological parameters in standard hematoxylin and eosin-stained intestinal biopsies.

Methods

ViCE is based on mathematical morphology operations and is scale and staining agnostic. It evaluates tissue orientation, identifies geometrical structure, and outputs key tissue measurements.

Results

The output measurements of Vh:Cd are concordant with manual quantifications across multiple datasets.

Conclusions

The underlying mathematical morphological approach for ViCE is robust, and reproducible and easily adaptable for measurement of morphological features in other tissues.
背景和目的肠表面的组织形态结构复杂,有称为绒毛的指状突起和称为隐窝的腺体结构。绒毛高度与隐窝深度之比(Vh:Cd)用于定量评估疾病的严重程度以及对乳糜泻等肠道疾病的治疗反应,目前采用人工定量的方法。由于需要花费大量时间,手动 Vh:Cd 测量在很大程度上仅限于临床试验,并未广泛应用于临床实践。我们开发了 ViCE(绒毛隐窝评价器),这是一款用户友好型软件,可自动量化标准苏木精和伊红染色肠活检组织学参数。结果在多个数据集中,Vh:Cd 的输出测量结果与人工量化结果一致。结论ViCE 的基本数学形态学方法是稳健的、可重复的,并且很容易适应于其他组织形态特征的测量。
{"title":"ViCE: An automated and quantitative program to assess intestinal tissue morphology","authors":"","doi":"10.1016/j.jpi.2024.100397","DOIUrl":"10.1016/j.jpi.2024.100397","url":null,"abstract":"<div><h3>Background and objective</h3><div>The tissue morphology of the intestinal surface is architecturally complex with finger-like projections called villi, and glandular structures called crypts. The ratio of villus height-to-crypt depth ratio (Vh:Cd) is used to quantitatively assess disease severity and response to therapy for intestinal enteropathies, such as celiac disease and is currently quantified manually. Given the time required, manual Vh:Cd measurements have largely been limited to clinical trials and are not used widely in clinical practice. We developed ViCE (Villus Crypt Evaluator), a user-friendly software that automatically quantifies histological parameters in standard hematoxylin and eosin-stained intestinal biopsies.</div></div><div><h3>Methods</h3><div>ViCE is based on mathematical morphology operations and is scale and staining agnostic. It evaluates tissue orientation, identifies geometrical structure, and outputs key tissue measurements.</div></div><div><h3>Results</h3><div>The output measurements of Vh:Cd are concordant with manual quantifications across multiple datasets.</div></div><div><h3>Conclusions</h3><div>The underlying mathematical morphological approach for ViCE is robust, and reproducible and easily adaptable for measurement of morphological features in other tissues.</div></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-13","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142422215","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 feature batch correction using ComBat for machine learning applications in computational pathology 利用 ComBat 对计算病理学中的机器学习应用进行深度特征批量校正
Q2 Medicine Pub Date : 2024-09-12 DOI: 10.1016/j.jpi.2024.100396

Background

Developing artificial intelligence (AI) models for digital pathology requires large datasets from multiple sources. However, without careful implementation, AI models risk learning confounding site-specific features in datasets instead of clinically relevant information, leading to overestimated performance, poor generalizability to real-world data, and potential misdiagnosis.

Methods

Whole-slide images (WSIs) from The Cancer Genome Atlas (TCGA) colon (COAD), and stomach adenocarcinoma datasets were selected for inclusion in this study. Patch embeddings were obtained using three feature extraction models, followed by ComBat harmonization. Attention-based multiple instance learning models were trained to predict tissue-source site (TSS), as well as clinical and genetic attributes, using raw, Macenko normalized, and Combat-harmonized patch embeddings.

Results

TSS prediction achieved high accuracy (AUROC > 0.95) with all three feature extraction models. ComBat harmonization significantly reduced the AUROC for TSS prediction, with mean AUROCs dropping to approximately 0.5 for most models, indicating successful mitigation of batch effects (e.g., CCL-ResNet50 in TCGA-COAD: Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, p < 0.001). Clinical attributes associated with TSS, such as race and treatment response, showed decreased predictability post-harmonization. Notably, the prediction of genetic features like MSI status remained robust after harmonization (e.g., MSI in TCGA-COAD: Pre-ComBat AUROC = 0.667, Post-ComBat AUROC = 0.669, p=0.952), indicating the preservation of true histological signals.

Conclusion

ComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates. This approach is promising for the integration of large-scale digital pathology datasets.
背景为数字病理学开发人工智能(AI)模型需要来自多个来源的大型数据集。然而,如果不仔细实施,人工智能模型就有可能学习数据集中的混杂部位特异性特征,而不是临床相关信息,从而导致性能被高估、对真实世界数据的普适性差以及潜在的误诊。使用三种特征提取模型获得斑块嵌入,然后进行 ComBat 协调。使用原始、Macenko 归一化和 Combat 协调的斑块嵌入,训练了基于注意力的多实例学习模型,以预测组织来源部位(TSS)以及临床和遗传属性。ComBat 协调大大降低了 TSS 预测的 AUROC,大多数模型的平均 AUROC 降至 0.5 左右,这表明批次效应得到了成功缓解(例如,TCGA-COAD 中的 CCL-ResNet50 模型,其平均 AUROC 降至 0.5 左右):CCL-ResNet50 in TCGA-COAD:Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, p < 0.001)。与 TSS 相关的临床属性(如种族和治疗反应)在协调后的可预测性有所下降。值得注意的是,MSI 状态等遗传特征的预测能力在协调后仍然很强(例如,TCGA-COAD 中的 MSI:ConclusionComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates.这种方法在整合大规模数字病理数据集方面大有可为。
{"title":"Deep feature batch correction using ComBat for machine learning applications in computational pathology","authors":"","doi":"10.1016/j.jpi.2024.100396","DOIUrl":"10.1016/j.jpi.2024.100396","url":null,"abstract":"<div><h3>Background</h3><div>Developing artificial intelligence (AI) models for digital pathology requires large datasets from multiple sources. However, without careful implementation, AI models risk learning confounding site-specific features in datasets instead of clinically relevant information, leading to overestimated performance, poor generalizability to real-world data, and potential misdiagnosis.</div></div><div><h3>Methods</h3><div>Whole-slide images (WSIs) from The Cancer Genome Atlas (TCGA) colon (COAD), and stomach adenocarcinoma datasets were selected for inclusion in this study. Patch embeddings were obtained using three feature extraction models, followed by ComBat harmonization. Attention-based multiple instance learning models were trained to predict tissue-source site (TSS), as well as clinical and genetic attributes, using raw, Macenko normalized, and Combat-harmonized patch embeddings.</div></div><div><h3>Results</h3><div>TSS prediction achieved high accuracy (AUROC &gt; 0.95) with all three feature extraction models. ComBat harmonization significantly reduced the AUROC for TSS prediction, with mean AUROCs dropping to approximately 0.5 for most models, indicating successful mitigation of batch effects (e.g., CCL-ResNet50 in TCGA-COAD: Pre-ComBat AUROC = 0.960, Post-ComBat AUROC = 0.506, <em>p &lt;</em> 0.001). Clinical attributes associated with TSS, such as race and treatment response, showed decreased predictability post-harmonization. Notably, the prediction of genetic features like MSI status remained robust after harmonization (e.g., MSI in TCGA-COAD: Pre-ComBat AUROC = 0.667, Post-ComBat AUROC = 0.669, <em>p</em>=0.952), indicating the preservation of true histological signals.</div></div><div><h3>Conclusion</h3><div>ComBat harmonization of deep learning-derived histology features effectively reduces the risk of AI models learning confounding features in WSIs, ensuring more reliable performance estimates. This approach is promising for the integration of large-scale digital pathology datasets.</div></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-09-12","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142323561","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
LVI-PathNet: Segmentation-classification pipeline for detection of lymphovascular invasion in whole slide images of lung adenocarcinoma LVI-PathNet:用于检测肺腺癌全切片图像中淋巴管侵犯的分割-分类管道
Q2 Medicine Pub Date : 2024-08-30 DOI: 10.1016/j.jpi.2024.100395

Lymphovascular invasion (LVI) in lung cancer is a significant prognostic factor that influences treatment and outcomes, yet its reliable detection is challenging due to interobserver variability. This study aims to develop a deep learning model for LVI detection using whole slide images (WSIs) and evaluate its effectiveness within a pathologist's information system. Experienced pathologists annotated blood vessels and invading tumor cells in 162 WSIs of non-mucinous lung adenocarcinoma sourced from two external and one internal datasets. Two models were trained to segment vessels and identify images with LVI features. DeepLabV3+ model achieved an Intersection-over-Union of 0.8840 and an area under the receiver operating characteristic curve (AUC-ROC) of 0.9869 in vessel segmentation. For LVI classification, the ensemble model achieved a F1-score of 0.9683 and an AUC-ROC of 0.9987. The model demonstrated robustness and was unaffected by variations in staining and image quality. The pilot study showed that pathologists' evaluation time for LVI detecting decreased by an average of 16.95%, and by 21.5% in “hard cases”. The model facilitated consistent diagnostic assessments, suggesting potential for broader applications in detecting pathological changes in blood vessels and other lung pathologies.

肺癌中的淋巴管侵犯(LVI)是影响治疗和预后的重要预后因素,但由于观察者之间的差异,其可靠检测具有挑战性。本研究旨在开发一种利用全切片图像(WSI)进行淋巴管侵犯检测的深度学习模型,并评估其在病理学家信息系统中的有效性。经验丰富的病理学家对来自两个外部数据集和一个内部数据集的 162 张非粘液性肺腺癌 WSI 图像中的血管和入侵肿瘤细胞进行了标注。对两个模型进行了训练,以利用 LVI 特征分割血管和识别图像。DeepLabV3+ 模型在血管分割方面取得了 0.8840 的 "联合交叉"(Intersection-over-Union)和 0.9869 的接收者操作特征曲线下面积(AUC-ROC)。在 LVI 分类中,集合模型的 F1 分数为 0.9683,AUC-ROC 为 0.9987。该模型具有鲁棒性,不受染色和图像质量变化的影响。试点研究表明,病理学家检测 LVI 的评估时间平均减少了 16.95%,在 "疑难病例 "中减少了 21.5%。该模型有助于进行一致的诊断评估,表明它在检测血管病理变化和其他肺部病变方面具有更广泛的应用潜力。
{"title":"LVI-PathNet: Segmentation-classification pipeline for detection of lymphovascular invasion in whole slide images of lung adenocarcinoma","authors":"","doi":"10.1016/j.jpi.2024.100395","DOIUrl":"10.1016/j.jpi.2024.100395","url":null,"abstract":"<div><p>Lymphovascular invasion (LVI) in lung cancer is a significant prognostic factor that influences treatment and outcomes, yet its reliable detection is challenging due to interobserver variability. This study aims to develop a deep learning model for LVI detection using whole slide images (WSIs) and evaluate its effectiveness within a pathologist's information system. Experienced pathologists annotated blood vessels and invading tumor cells in 162 WSIs of non-mucinous lung adenocarcinoma sourced from two external and one internal datasets. Two models were trained to segment vessels and identify images with LVI features. DeepLabV3+ model achieved an Intersection-over-Union of 0.8840 and an area under the receiver operating characteristic curve (AUC-ROC) of 0.9869 in vessel segmentation. For LVI classification, the ensemble model achieved a F1-score of 0.9683 and an AUC-ROC of 0.9987. The model demonstrated robustness and was unaffected by variations in staining and image quality. The pilot study showed that pathologists' evaluation time for LVI detecting decreased by an average of 16.95%, and by 21.5% in “hard cases”. The model facilitated consistent diagnostic assessments, suggesting potential for broader applications in detecting pathological changes in blood vessels and other lung pathologies.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-30","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000348/pdfft?md5=9a1e9217891b1539c144069b2cb2703f&pid=1-s2.0-S2153353924000348-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142238092","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
Globalization of a telepathology network with artificial intelligence applications in Colombia: The GLORIA program study protocol 哥伦比亚应用人工智能的远程病理网络全球化:GLORIA 计划研究协议
Q2 Medicine Pub Date : 2024-08-15 DOI: 10.1016/j.jpi.2024.100394

In Colombia, cancer is recognized as a high-cost pathology by the national government and the Colombian High-Cost Disease Fund. As of 2020, the situation is most critical for adult cancer patients, particularly those under public healthcare and residing in remote regions of the country. The highest lag time for a diagnosis was observed for cervical cancer (79.13 days), followed by prostate (77.30 days), and breast cancer (70.25 days). Timely and accurate histopathological reporting plays a vital role in the diagnosis of cancer. In recent years, digital pathology has been globally implemented as a technological tool in two main areas: telepathology (TP) and computational pathology. TP has been shown to improve rapid and timely diagnosis in anatomic pathology by facilitating interaction between general laboratories and specialized pathologists worldwide through information and telecommunication technologies. Computational pathology provides diagnostic and prognostic assistance based on histopathological patterns, molecular, and clinical information, aiding pathologists in making more accurate diagnoses. We present the study protocol of the GLORIA digital pathology network, a pioneering initiative, and national grant-approved program aiming to design and pilot a Colombian digital pathology transformation focused on TP and computational pathology, in response to the general needs of pathology laboratories for diagnosing complex malignant tumors. The study protocol describes the design of a TP network to expand oncopathology services across all Colombian regions. It also describes an artificial intelligence proposal for lung cancer, one of Colombia's most prevalent cancers, and a freely accessible national histopathological image database to facilitate image analysis studies.

在哥伦比亚,癌症被国家政府和哥伦比亚高成本疾病基金认定为高成本病症。截至 2020 年,成年癌症患者的情况最为严峻,尤其是那些享受公共医疗服务和居住在偏远地区的患者。宫颈癌的诊断滞后时间最长(79.13 天),其次是前列腺癌(77.30 天)和乳腺癌(70.25 天)。及时准确的组织病理学报告在癌症诊断中起着至关重要的作用。近年来,数字病理学作为一种技术工具已在全球范围内广泛应用,主要涉及两个领域:远程病理学(TP)和计算病理学。远程病理学通过信息和电信技术促进了全球普通实验室和专业病理学家之间的互动,从而提高了解剖病理学诊断的快速性和及时性。计算病理学根据组织病理学模式、分子和临床信息提供诊断和预后帮助,帮助病理学家做出更准确的诊断。我们介绍了 GLORIA 数字病理学网络的研究方案,该网络是一项开创性计划,也是国家拨款批准的计划,旨在设计和试点哥伦比亚数字病理学转型,重点关注 TP 和计算病理学,以满足病理实验室诊断复杂恶性肿瘤的普遍需求。该研究计划介绍了如何设计一个 TP 网络,以在哥伦比亚所有地区扩展肿瘤病理学服务。它还介绍了针对肺癌(哥伦比亚最常见的癌症之一)的人工智能提案,以及可免费访问的国家组织病理学图像数据库,以促进图像分析研究。
{"title":"Globalization of a telepathology network with artificial intelligence applications in Colombia: The GLORIA program study protocol","authors":"","doi":"10.1016/j.jpi.2024.100394","DOIUrl":"10.1016/j.jpi.2024.100394","url":null,"abstract":"<div><p>In Colombia, cancer is recognized as a high-cost pathology by the national government and the Colombian High-Cost Disease Fund. As of 2020, the situation is most critical for adult cancer patients, particularly those under public healthcare and residing in remote regions of the country. The highest lag time for a diagnosis was observed for cervical cancer (79.13 days), followed by prostate (77.30 days), and breast cancer (70.25 days). Timely and accurate histopathological reporting plays a vital role in the diagnosis of cancer. In recent years, digital pathology has been globally implemented as a technological tool in two main areas: telepathology (TP) and computational pathology. TP has been shown to improve rapid and timely diagnosis in anatomic pathology by facilitating interaction between general laboratories and specialized pathologists worldwide through information and telecommunication technologies. Computational pathology provides diagnostic and prognostic assistance based on histopathological patterns, molecular, and clinical information, aiding pathologists in making more accurate diagnoses. We present the study protocol of the GLORIA digital pathology network, a pioneering initiative, and national grant-approved program aiming to design and pilot a Colombian digital pathology transformation focused on TP and computational pathology, in response to the general needs of pathology laboratories for diagnosing complex malignant tumors. The study protocol describes the design of a TP network to expand oncopathology services across all Colombian regions. It also describes an artificial intelligence proposal for lung cancer, one of Colombia's most prevalent cancers, and a freely accessible national histopathological image database to facilitate image analysis studies.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-15","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000336/pdfft?md5=861e86fc08dee64d7bef49370be8286b&pid=1-s2.0-S2153353924000336-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"142075800","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
Non-diagnostic time in digital pathology: An empirical study over 10 years 数字病理学的非诊断时间:十年实证研究
Q2 Medicine Pub Date : 2024-08-05 DOI: 10.1016/j.jpi.2024.100393
{"title":"Non-diagnostic time in digital pathology: An empirical study over 10 years","authors":"","doi":"10.1016/j.jpi.2024.100393","DOIUrl":"10.1016/j.jpi.2024.100393","url":null,"abstract":"","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-08-05","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000324/pdfft?md5=215132a8d517d7691de823ffcf6bf232&pid=1-s2.0-S2153353924000324-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141963948","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
Engineered feature embeddings meet deep learning: A novel strategy to improve bone marrow cell classification and model transparency 工程特征嵌入与深度学习的结合:改善骨髓细胞分类和模型透明度的新策略
Q2 Medicine Pub Date : 2024-07-03 DOI: 10.1016/j.jpi.2024.100390

Cytomorphology evaluation of bone marrow cell is the initial step to diagnose different hematological diseases. This assessment is still manually performed by trained specialists, who may be a bottleneck within the clinical process. Deep learning algorithms are a promising approach to automate this bone marrow cell evaluation. These artificial intelligence models have focused on limited cell subtypes, mainly associated to a particular disease, and are frequently presented as black boxes. The herein introduced strategy presents an engineered feature representation, the region-attention embedding, which improves the deep learning classification performance of a cytomorphology with 21 bone marrow cell subtypes. This embedding is built upon a specific organization of cytology features within a squared matrix by distributing them after pre-segmented cell regions, i.e., cytoplasm, nucleus, and whole-cell. This novel cell image representation, aimed to preserve spatial/regional relations, is used as input of the network. Combination of region-attention embedding and deep learning networks (Xception and ResNet50) provides local relevance associated to image regions, adding up interpretable information to the prediction. Additionally, this approach is evaluated in a public database with the largest number of cell subtypes (21) by a thorough evaluation scheme with three iterations of a 3-fold cross-validation, performed in 80% of the images (n = 89,484), and a testing process in an unseen set of images composed by the remaining 20% of the images (n = 22,371). This evaluation process demonstrates the introduced strategy outperforms previously published approaches in an equivalent validation set, with a f1-score of 0.82, and presented competitive results on the unseen data partition with a f1-score of 0.56.

骨髓细胞的细胞形态学评估是诊断各种血液病的第一步。这种评估仍由训练有素的专家手工完成,这可能是临床过程中的一个瓶颈。深度学习算法是一种有望实现骨髓细胞评估自动化的方法。这些人工智能模型侧重于有限的细胞亚型,主要与特定疾病相关,通常以黑盒形式呈现。本文介绍的策略提出了一种工程特征表征--区域注意嵌入,它提高了 21 种骨髓细胞亚型的细胞形态学深度学习分类性能。这种嵌入建立在方形矩阵中细胞学特征的特定组织之上,将它们分布在预先分割的细胞区域(即细胞质、细胞核和全细胞)之后。这种旨在保留空间/区域关系的新型细胞图像表示法被用作网络的输入。区域注意嵌入和深度学习网络(Xception 和 ResNet50)的结合提供了与图像区域相关的局部相关性,为预测增加了可解释的信息。此外,我们还在一个拥有最多细胞亚型的公共数据库(21)中对该方法进行了全面评估,评估方案包括对 80% 的图像(n = 89,484 张)进行三次迭代的 3 倍交叉验证,以及对由剩余 20% 的图像(n = 22,371 张)组成的未见图像集进行测试。评估结果表明,在等效验证集上,引入的策略优于之前发布的方法,f1 分数为 0.82,而在未见数据分区上,引入的策略也取得了具有竞争力的结果,f1 分数为 0.56。
{"title":"Engineered feature embeddings meet deep learning: A novel strategy to improve bone marrow cell classification and model transparency","authors":"","doi":"10.1016/j.jpi.2024.100390","DOIUrl":"10.1016/j.jpi.2024.100390","url":null,"abstract":"<div><p>Cytomorphology evaluation of bone marrow cell is the initial step to diagnose different hematological diseases. This assessment is still manually performed by trained specialists, who may be a bottleneck within the clinical process. Deep learning algorithms are a promising approach to automate this bone marrow cell evaluation. These artificial intelligence models have focused on limited cell subtypes, mainly associated to a particular disease, and are frequently presented as black boxes. The herein introduced strategy presents an engineered feature representation, the region-attention embedding, which improves the deep learning classification performance of a cytomorphology with 21 bone marrow cell subtypes. This embedding is built upon a specific organization of cytology features within a squared matrix by distributing them after pre-segmented cell regions, i.e., cytoplasm, nucleus, and whole-cell. This novel cell image representation, aimed to preserve spatial/regional relations, is used as input of the network. Combination of region-attention embedding and deep learning networks (Xception and ResNet50) provides local relevance associated to image regions, adding up interpretable information to the prediction. Additionally, this approach is evaluated in a public database with the largest number of cell subtypes (21) by a thorough evaluation scheme with three iterations of a 3-fold cross-validation, performed in 80% of the images (<em>n</em> = 89,484), and a testing process in an unseen set of images composed by the remaining 20% of the images (<em>n</em> = 22,371). This evaluation process demonstrates the introduced strategy outperforms previously published approaches in an equivalent validation set, with a f1-score of 0.82, and presented competitive results on the unseen data partition with a f1-score of 0.56.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-03","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000294/pdfft?md5=87a5b2e97447248282a9f8d40bb281e3&pid=1-s2.0-S2153353924000294-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141629985","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
Validation of AI-assisted ThinPrep® Pap test screening using the GeniusTM Digital Diagnostics System 使用 GeniusTM 数字诊断系统对人工智能辅助 ThinPrep® Pap 测试筛查进行验证
Q2 Medicine Pub Date : 2024-07-02 DOI: 10.1016/j.jpi.2024.100391

Advances in whole-slide imaging and artificial intelligence present opportunities for improvement in Pap test screening. To date, there have been limited studies published regarding how best to validate newer AI-based digital systems for screening Pap tests in clinical practice. In this study, we validated the Genius™ Digital Diagnostics System (Hologic) by comparing the performance to traditional manual light microscopic diagnosis of ThinPrep® Pap test slides. A total of 319 ThinPrep® Pap test cases were prospectively assessed by six cytologists and three cytopathologists by light microscopy and digital evaluation and the results compared to the original ground truth Pap test diagnosis. Concordance with the original diagnosis was significantly different by digital and manual light microscopy review when comparing across: (i) exact Bethesda System diagnostic categories (62.1% vs 55.8%, respectively, p = 0.014), (ii) condensed diagnostic categories (76.8% vs 71.5%, respectively, p = 0.027), and (iii) condensed diagnoses based on clinical management (71.5% vs 65.2%, respectively, p = 0.017). Time to evaluate cases was shorter for digital (M = 3.2 min, SD = 2.2) compared to manual (M = 5.9 min, SD = 3.1) review (t(352) = 19.44, p < 0.001, Cohen's d = 1.035, 95% CI [0.905, 1.164]). Not only did our validation study demonstrate that AI-based digital Pap test evaluation had improved diagnostic accuracy and reduced screening time compared to light microscopy, but that participants reported a positive experience using this system.

全切片成像和人工智能的进步为改进巴氏试验筛查带来了机遇。迄今为止,关于如何在临床实践中验证较新的人工智能巴氏试验筛查数字系统的研究还很有限。在本研究中,我们将 Genius™ 数字诊断系统(Hologic)的性能与 ThinPrep® 巴氏试验玻片的传统人工光学显微镜诊断进行了比较,从而对其进行了验证。六位细胞学专家和三位细胞病理学专家通过光学显微镜和数字评估对总共 319 例 ThinPrep® Pap 测试病例进行了前瞻性评估,并将评估结果与原始的地面真实 Pap 测试诊断结果进行了比较。数字光镜检查和人工光镜检查与原始诊断的一致性在以下方面有显著差异:(i) 贝塞斯达系统精确诊断类别(分别为 62.1% 对 55.8%,p = 0.014);(ii) 简化诊断类别(分别为 76.8% 对 71.5%,p = 0.027);(iii) 基于临床管理的简化诊断(分别为 71.5% 对 65.2%,p = 0.017)。与人工复查(M = 5.9 min, SD = 3.1)相比,数字复查(M = 3.2 min, SD = 2.2)的病例评估时间更短(t(352) = 19.44, p < 0.001, Cohen's d = 1.035, 95% CI [0.905, 1.164])。我们的验证研究表明,与光学显微镜检查相比,基于人工智能的数字巴氏试验评估不仅提高了诊断准确性,缩短了筛查时间,而且参与者对该系统的使用体验表示肯定。
{"title":"Validation of AI-assisted ThinPrep® Pap test screening using the GeniusTM Digital Diagnostics System","authors":"","doi":"10.1016/j.jpi.2024.100391","DOIUrl":"10.1016/j.jpi.2024.100391","url":null,"abstract":"<div><p>Advances in whole-slide imaging and artificial intelligence present opportunities for improvement in Pap test screening. To date, there have been limited studies published regarding how best to validate newer AI-based digital systems for screening Pap tests in clinical practice. In this study, we validated the Genius™ Digital Diagnostics System (Hologic) by comparing the performance to traditional manual light microscopic diagnosis of ThinPrep<strong>®</strong> Pap test slides. A total of 319 ThinPrep<strong>®</strong> Pap test cases were prospectively assessed by six cytologists and three cytopathologists by light microscopy and digital evaluation and the results compared to the original ground truth Pap test diagnosis. Concordance with the original diagnosis was significantly different by digital and manual light microscopy review when comparing across: (i) exact Bethesda System diagnostic categories (62.1% vs 55.8%, respectively, <em>p</em> = 0.014), (ii) condensed diagnostic categories (76.8% vs 71.5%, respectively, <em>p</em> = 0.027), and (iii) condensed diagnoses based on clinical management (71.5% vs 65.2%, respectively, <em>p</em> = 0.017). Time to evaluate cases was shorter for digital (M = 3.2 min, SD = 2.2) compared to manual (M = 5.9 min, SD = 3.1) review (t(352) = 19.44, <em>p</em> &lt; 0.001, Cohen's d = 1.035, 95% CI [0.905, 1.164]). Not only did our validation study demonstrate that AI-based digital Pap test evaluation had improved diagnostic accuracy and reduced screening time compared to light microscopy, but that participants reported a positive experience using this system.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000300/pdfft?md5=f678b76ba4ddf0bb5fbfba56b65df94c&pid=1-s2.0-S2153353924000300-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141639228","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
An explainable AI-based blood cell classification using optimized convolutional neural network 利用优化的卷积神经网络实现基于人工智能的可解释血细胞分类
Q2 Medicine Pub Date : 2024-07-02 DOI: 10.1016/j.jpi.2024.100389

White blood cells (WBCs) are a vital component of the immune system. The efficient and precise classification of WBCs is crucial for medical professionals to diagnose diseases accurately. This study presents an enhanced convolutional neural network (CNN) for detecting blood cells with the help of various image pre-processing techniques. Various image pre-processing techniques, such as padding, thresholding, erosion, dilation, and masking, are utilized to minimize noise and improve feature enhancement. Additionally, performance is further enhanced by experimenting with various architectural structures and hyperparameters to optimize the proposed model. A comparative evaluation is conducted to compare the performance of the proposed model with three transfer learning models, including Inception V3, MobileNetV2, and DenseNet201.The results indicate that the proposed model outperforms existing models, achieving a testing accuracy of 99.12%, precision of 99%, and F1-score of 99%. In addition, We utilized SHAP (Shapley Additive explanations) and LIME (Local Interpretable Model-agnostic Explanations) techniques in our study to improve the interpretability of the proposed model, providing valuable insights into how the model makes decisions. Furthermore, the proposed model has been further explained using the Grad-CAM and Grad-CAM++ techniques, which is a class-discriminative localization approach, to improve trust and transparency. Grad-CAM++ performed slightly better than Grad-CAM in identifying the predicted area's location. Finally, the most efficient model has been integrated into an end-to-end (E2E) system, accessible through both web and Android platforms for medical professionals to classify blood cell.

白细胞(WBC)是免疫系统的重要组成部分。对白细胞进行高效、精确的分类对于医学专家准确诊断疾病至关重要。本研究提出了一种增强型卷积神经网络(CNN),可借助各种图像预处理技术检测血细胞。利用各种图像预处理技术,如填充、阈值处理、侵蚀、扩张和遮蔽,可以最大限度地减少噪音,提高特征增强效果。此外,还通过试验各种架构结构和超参数来优化所提出的模型,从而进一步提高性能。结果表明,拟议模型的性能优于现有模型,测试准确率达到 99.12%,精确率达到 99%,F1 分数达到 99%。此外,我们还在研究中使用了 SHAP(夏普利相加解释)和 LIME(局部可解释模型-不可知解释)技术,以提高所提模型的可解释性,为了解模型如何做出决策提供了宝贵的见解。此外,我们还使用 Grad-CAM 和 Grad-CAM++ 技术进一步解释了所提出的模型。在识别预测区域位置方面,Grad-CAM++ 的表现略好于 Grad-CAM。最后,最有效的模型被集成到一个端到端(E2E)系统中,通过网络和安卓平台供医疗专业人员对血细胞进行分类。
{"title":"An explainable AI-based blood cell classification using optimized convolutional neural network","authors":"","doi":"10.1016/j.jpi.2024.100389","DOIUrl":"10.1016/j.jpi.2024.100389","url":null,"abstract":"<div><p>White blood cells (WBCs) are a vital component of the immune system. The efficient and precise classification of WBCs is crucial for medical professionals to diagnose diseases accurately. This study presents an enhanced convolutional neural network (CNN) for detecting blood cells with the help of various image pre-processing techniques. Various image pre-processing techniques, such as padding, thresholding, erosion, dilation, and masking, are utilized to minimize noise and improve feature enhancement. Additionally, performance is further enhanced by experimenting with various architectural structures and hyperparameters to optimize the proposed model. A comparative evaluation is conducted to compare the performance of the proposed model with three transfer learning models, including Inception V3, MobileNetV2, and DenseNet201.The results indicate that the proposed model outperforms existing models, achieving a testing accuracy of 99.12%, precision of 99%, and F1-score of 99%. In addition, We utilized SHAP (Shapley Additive explanations) and LIME (Local Interpretable Model-agnostic Explanations) techniques in our study to improve the interpretability of the proposed model, providing valuable insights into how the model makes decisions. Furthermore, the proposed model has been further explained using the Grad-CAM and Grad-CAM++ techniques, which is a class-discriminative localization approach, to improve trust and transparency. Grad-CAM++ performed slightly better than Grad-CAM in identifying the predicted area's location. Finally, the most efficient model has been integrated into an end-to-end (E2E) system, accessible through both web and Android platforms for medical professionals to classify blood cell.</p></div>","PeriodicalId":37769,"journal":{"name":"Journal of Pathology Informatics","volume":null,"pages":null},"PeriodicalIF":0.0,"publicationDate":"2024-07-02","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://www.sciencedirect.com/science/article/pii/S2153353924000282/pdfft?md5=357d6d2314681f04709e94998615c5a1&pid=1-s2.0-S2153353924000282-main.pdf","citationCount":null,"resultStr":null,"platform":"Semanticscholar","paperid":"141708134","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