1
|
Julian DR, Bahramy A, Neal M, Pearce TM, Kofler J. Current Advancements in Digital Neuropathology and Machine Learning for the Study of Neurodegenerative Diseases. THE AMERICAN JOURNAL OF PATHOLOGY 2025:S0002-9440(25)00046-X. [PMID: 39954963 DOI: 10.1016/j.ajpath.2024.12.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/16/2024] [Accepted: 12/30/2024] [Indexed: 02/17/2025]
Abstract
Computational neurodegenerative neuropathology represents a transformative approach in the analysis and understanding of neurodegenerative diseases through utilization of whole slide images (WSIs) and advanced machine learning/artificial intelligence (ML/AI) techniques. This review explores the emerging field of computational neurodegenerative neuropathology, emphasizing its potential to enhance neuropathologic assessment, diagnosis, and research. Recent advancements in ML/AI technologies have significantly affected image-based medical fields, including anatomic pathology, by automating disease staging, identifying novel morphologic biomarkers, and uncovering new clinical insights via multi-modal AI approaches. Despite its promise, the field faces several challenges, including limited expert annotations, slide scanning inaccessibility, inter-institutional variability, and the complexities of sharing large WSI data sets. This review discusses the importance of improving deep learning model accuracy and efficiency for better interpretation of neuropathologic data. It highlights the potential of unsupervised learning to identify patterns in unannotated data. Furthermore, the development of explainable AI models is crucial for experimental neuropathology. By addressing these challenges and leveraging cutting-edge AI techniques, computational neurodegenerative neuropathology has the potential to revolutionize the field and significantly advance our understanding of disease.
Collapse
Affiliation(s)
- Dana R Julian
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Afshin Bahramy
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Clinical and Translational Science Institute, University of Pittsburgh School of Public Health, Pittsburgh, Pennsylvania
| | - Makayla Neal
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Thomas M Pearce
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Julia Kofler
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania; Department of Human Genetics, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania.
| |
Collapse
|
2
|
Zia S, Yildiz-Aktas IZ, Zia F, Parwani AV. An update on applications of digital pathology: primary diagnosis; telepathology, education and research. Diagn Pathol 2025; 20:17. [PMID: 39940046 DOI: 10.1186/s13000-025-01610-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 01/30/2025] [Indexed: 02/14/2025] Open
Abstract
Digital Pathology or whole slide imaging (WSI) is a diagnostic evaluation technique that produces digital images of high quality from tissue fragments. These images are formed on glass slides and evaluated by pathologist with the aid of microscope. As the concept of digital pathology is introduced, these high quality images are digitized and produced on-screen whole slide images in the form of digital files. This has paved the way for pathologists to collaborate with other pathology professionals in case of any additional recommendations and also provides remote working opportunities. The application of digital pathology in clinical practice is glazed with several advantages and adopted by pathologists and researchers for clinical, educational and research purposes. Moreover, digital pathology system integration requires an intensive effort from multiple stakeholders. All pathology departments have different needs, case usage, and blueprints, even though the framework elements and variables for effective clinical integration can be applied to any institution aiming for digital transformation. This article reviews the background and developmental phases of digital pathology and its application in clinical services, educational and research activities.
Collapse
Affiliation(s)
- Shamail Zia
- Department of Pathology, CorePath Laboratories, San Antonio, TX, USA.
| | - Isil Z Yildiz-Aktas
- Department of Pathology, Yale School of Medicine, New Haven, CT, USA
- Department of Pathology, VA CT Healthcare System, West Haven, CT, USA
| | - Fazail Zia
- Department of Pathology, Jinnah Sindh Medical University, Karachi, Sindh, Pakistan
| | - Anil V Parwani
- Department of Pathology, The Ohio State University, Columbus, OH, USA
| |
Collapse
|
3
|
Clunie D, Taylor A, Bisson T, Gutman D, Xiao Y, Schwarz CG, Greve D, Gichoya J, Shih G, Kline A, Kopchick B, Farahani K. Summary of the National Cancer Institute 2023 Virtual Workshop on Medical Image De-identification-Part 2: Pathology Whole Slide Image De-identification, De-facing, the Role of AI in Image De-identification, and the NCI MIDI Datasets and Pipeline. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2025; 38:16-30. [PMID: 38980626 PMCID: PMC11811347 DOI: 10.1007/s10278-024-01183-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/13/2024] [Revised: 06/16/2024] [Accepted: 06/18/2024] [Indexed: 07/10/2024]
Abstract
De-identification of medical images intended for research is a core requirement for data sharing initiatives, particularly as the demand for data for artificial intelligence (AI) applications grows. The Center for Biomedical Informatics and Information Technology (CBIIT) of the United States National Cancer Institute (NCI) convened a two half-day virtual workshop with the intent of summarizing the state of the art in de-identification technology and processes and exploring interesting aspects of the subject. This paper summarizes the highlights of the second day of the workshop, the recordings and presentations of which are publicly available for review. The topics covered included pathology whole slide image de-identification, de-facing, the role of AI in image de-identification, and the NCI Medical Image De-Identification Initiative (MIDI) datasets and pipeline.
Collapse
Affiliation(s)
| | | | - Tom Bisson
- Charité - Universitätsmedizin Berlin, Berlin, Germany
| | | | - Ying Xiao
- Hospital of the University of Pennsylvania, Philadelphia, PA, USA
| | | | | | | | - George Shih
- Weill Cornell Medical College, New York, NY, USA
| | | | | | - Keyvan Farahani
- National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
4
|
Mateos-Aparicio-Ruiz I, Pedraza A, Becker JU, Altini N, Salido J, Bueno G. GNCnn: A QuPath extension for glomerulosclerosis and glomerulonephritis characterization based on deep learning. Comput Struct Biotechnol J 2024; 27:35-47. [PMID: 39802211 PMCID: PMC11719282 DOI: 10.1016/j.csbj.2024.11.049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Revised: 11/29/2024] [Accepted: 11/29/2024] [Indexed: 01/16/2025] Open
Abstract
The digitalization of traditional glass slide microscopy into whole slide images has opened up new opportunities for pathology, such as the application of artificial intelligence techniques. Specialized software is necessary to visualize and analyze these images. One of these applications is QuPath, a popular bioimage analysis tool. This study proposes GNCnn, the first open-source QuPath extension specifically designed for nephropathology. It integrates deep learning models to provide nephropathologists with an accessible, automatic detector and classifier of glomeruli, the basic filtering units of the kidneys. The aim is to offer nephropathologists a freely available application to measure and analyze glomeruli to identify conditions such as glomerulosclerosis and glomerulonephritis. GNCnn offers a user-friendly interface that enables nephropathologists to detect glomeruli with high accuracy (Dice coefficient of 0.807) and categorize them as either sclerotic or non-sclerotic, achieving a balanced accuracy of 98.46%. Furthermore, it facilitates the classification of non-sclerotic glomeruli into 12 commonly diagnosed types of glomerulonephritis, with a top-3 balanced accuracy of 84.41%. GNCnn provides real-time updates of results, which are available at both the glomerulus and slide levels. This allows users to complete a typical analysis task without leaving the main application, QuPath. This tool is the first to integrate the entire workflow for the assessment of glomerulonephritis directly into the nephropathologists' workspace, accelerating and supporting their diagnosis.
Collapse
Affiliation(s)
- Israel Mateos-Aparicio-Ruiz
- VISILAB Group, Universidad de Castilla–La Mancha, Av. Camilo José Cela, Ciudad Real, 13071, Ciudad Real, Spain
| | - Anibal Pedraza
- VISILAB Group, Universidad de Castilla–La Mancha, Av. Camilo José Cela, Ciudad Real, 13071, Ciudad Real, Spain
| | - Jan Ulrich Becker
- Institute of Pathology, University Hospital of Cologne, Cologne, Germany
| | - Nicola Altini
- Department of Electrical and Information Engineering (DEI), Polytechnic University of Bari, Via Edoardo Orabona n.4, Bari, 70126, Italy
| | - Jesus Salido
- VISILAB Group, Universidad de Castilla–La Mancha, Av. Camilo José Cela, Ciudad Real, 13071, Ciudad Real, Spain
| | - Gloria Bueno
- VISILAB Group, Universidad de Castilla–La Mancha, Av. Camilo José Cela, Ciudad Real, 13071, Ciudad Real, Spain
| |
Collapse
|
5
|
Barcellona L, Nicolè L, Cappellesso R, Dei Tos AP, Ghidoni S. SlideTiler: A dataset creator software for boosting deep learning on histological whole slide images. J Pathol Inform 2024; 15:100356. [PMID: 38222323 PMCID: PMC10787253 DOI: 10.1016/j.jpi.2023.100356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 11/15/2023] [Accepted: 12/04/2023] [Indexed: 01/16/2024] Open
Abstract
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.
Collapse
Affiliation(s)
- Leonardo Barcellona
- Department of Information Engineering, University of Padua, Padua, Italy
- Polytechnic University of Turin, Turin, Italy
| | - Lorenzo Nicolè
- Unit of Pathology and Cytopathology, Ospedale dell’Angelo, Mestre, Italy
- Department of Medicine, DIMED, University of Padua, Padua, Italy
| | | | - Angelo Paolo Dei Tos
- Department of Medicine, DIMED, University of Padua, Padua, Italy
- Department of Integrated diagnostics, Azienda Ospedale-Università, Padua, Italy
| | - Stefano Ghidoni
- Department of Information Engineering, University of Padua, Padua, Italy
| |
Collapse
|
6
|
Kim H, Kim J, Yeon SY, You S. Machine learning approaches for spatial omics data analysis in digital pathology: tools and applications in genitourinary oncology. Front Oncol 2024; 14:1465098. [PMID: 39678498 PMCID: PMC11638011 DOI: 10.3389/fonc.2024.1465098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 11/08/2024] [Indexed: 12/17/2024] Open
Abstract
Recent advances in spatial omics technologies have enabled new approaches for analyzing tissue morphology, cell composition, and biomolecule expression patterns in situ. These advances are promoting the development of new computational tools and quantitative techniques in the emerging field of digital pathology. In this review, we survey current trends in the development of computational methods for spatially mapped omics data analysis using digitized histopathology slides and supplementary materials, with an emphasis on tools and applications relevant to genitourinary oncological research. The review contains three sections: 1) an overview of image processing approaches for histopathology slide analysis; 2) machine learning integration with spatially resolved omics data analysis; 3) a discussion of current limitations and future directions for integration of machine learning in the clinical decision-making process.
Collapse
Affiliation(s)
- Hojung Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Jina Kim
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Su Yeon Yeon
- Department of Pathology, University of Illinois at Chicago, Chicago, IL, United States
| | - Sungyong You
- Department of Urology, Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, United States
| |
Collapse
|
7
|
Kauer T, Sehring J, Schmid K, Bartkuhn M, Wiebach B, Crnkovic S, Kwapiszewska G, Acker T, Amsel D. MOTH: Memory-Efficient On-the-Fly Tiling of Histological Image Annotations Using QuPath. J Imaging 2024; 10:292. [PMID: 39590756 PMCID: PMC11595786 DOI: 10.3390/jimaging10110292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/30/2024] [Accepted: 11/12/2024] [Indexed: 11/28/2024] Open
Abstract
The emerging usage of digitalized histopathological images is leading to a novel possibility for data analysis. With the help of artificial intelligence algorithms, it is now possible to detect certain structures and morphological features on whole slide images automatically. This enables algorithms to count, measure, or evaluate those areas when trained properly. To achieve suitable training, datasets must be annotated and curated by users in programs like QuPath. The extraction of this data for artificial intelligence algorithms is still rather tedious and needs to be saved on a local hard drive. We developed a toolkit for integration into existing pipelines and tools, like U-net, for the on-the-fly extraction of annotation tiles from existing QuPath projects. The tiles can be directly used as input for artificial intelligence algorithms, and the results are directly transferred back to QuPath for visual inspection. With the toolkit, we created a convenient way to incorporate QuPath into existing AI workflows.
Collapse
Affiliation(s)
- Thomas Kauer
- Institute of Neuropathology, Justus-Liebig-University Giessen, Arndtstr. 16, 35392 Giessen, Germany; (T.K.); (J.S.); (K.S.); (T.A.)
| | - Jannik Sehring
- Institute of Neuropathology, Justus-Liebig-University Giessen, Arndtstr. 16, 35392 Giessen, Germany; (T.K.); (J.S.); (K.S.); (T.A.)
| | - Kai Schmid
- Institute of Neuropathology, Justus-Liebig-University Giessen, Arndtstr. 16, 35392 Giessen, Germany; (T.K.); (J.S.); (K.S.); (T.A.)
| | - Marek Bartkuhn
- Institute for Lung Health, Justus-Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany; (M.B.); (B.W.); (S.C.); (G.K.)
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Benedikt Wiebach
- Institute for Lung Health, Justus-Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany; (M.B.); (B.W.); (S.C.); (G.K.)
- Biomedical Informatics and Systems Medicine, Justus-Liebig-University Giessen, Aulweg 128, 35392 Giessen, Germany
| | - Slaven Crnkovic
- Institute for Lung Health, Justus-Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany; (M.B.); (B.W.); (S.C.); (G.K.)
- Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Grazyna Kwapiszewska
- Institute for Lung Health, Justus-Liebig University Giessen, Aulweg 128, 35392 Giessen, Germany; (M.B.); (B.W.); (S.C.); (G.K.)
- Medical University of Graz, Neue Stiftingtalstraße 6, 8010 Graz, Austria
| | - Till Acker
- Institute of Neuropathology, Justus-Liebig-University Giessen, Arndtstr. 16, 35392 Giessen, Germany; (T.K.); (J.S.); (K.S.); (T.A.)
| | - Daniel Amsel
- Institute of Neuropathology, Justus-Liebig-University Giessen, Arndtstr. 16, 35392 Giessen, Germany; (T.K.); (J.S.); (K.S.); (T.A.)
| |
Collapse
|
8
|
Ghandian S, Albarghouthi L, Nava K, Sharma SRR, Minaud L, Beckett L, Saito N, DeCarli C, Rissman RA, Teich AF, Jin LW, Dugger BN, Keiser MJ. Learning precise segmentation of neurofibrillary tangles from rapid manual point annotations. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.15.594372. [PMID: 39386601 PMCID: PMC11463656 DOI: 10.1101/2024.05.15.594372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Accumulation of abnormal tau protein into neurofibrillary tangles (NFTs) is a pathologic hallmark of Alzheimer disease (AD). Accurate detection of NFTs in tissue samples can reveal relationships with clinical, demographic, and genetic features through deep phenotyping. However, expert manual analysis is time-consuming, subject to observer variability, and cannot handle the data amounts generated by modern imaging. We present a scalable, open-source, deep-learning approach to quantify NFT burden in digital whole slide images (WSIs) of post-mortem human brain tissue. To achieve this, we developed a method to generate detailed NFT boundaries directly from single-point-per-NFT annotations. We then trained a semantic segmentation model on 45 annotated 2400μm by 1200μm regions of interest (ROIs) selected from 15 unique temporal cortex WSIs of AD cases from three institutions (University of California (UC)-Davis, UC-San Diego, and Columbia University). Segmenting NFTs at the single-pixel level, the model achieved an area under the receiver operating characteristic of 0.832 and an F1 of 0.527 (196-fold over random) on a held-out test set of 664 NFTs from 20 ROIs (7 WSIs). We compared this to deep object detection, which achieved comparable but coarser-grained performance that was 60% faster. The segmentation and object detection models correlated well with expert semi-quantitative scores at the whole-slide level (Spearman's rho ρ=0.654 (p=6.50e-5) and ρ=0.513 (p=3.18e-3), respectively). We openly release this multi-institution deep-learning pipeline to provide detailed NFT spatial distribution and morphology analysis capability at a scale otherwise infeasible by manual assessment.
Collapse
Affiliation(s)
- Sina Ghandian
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, 94158, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Liane Albarghouthi
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, 94158, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Kiana Nava
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Shivam R. Rai Sharma
- Department of Computer Science, University of California, Davis, Davis, CA, 95616, USA
- Robust and Ubiquitous Networking (RUbiNet) Lab, University of California, Davis, Davis, CA, 95616, USA
| | - Lise Minaud
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, 94158, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Laurel Beckett
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Naomi Saito
- Division of Biostatistics, Department of Public Health Sciences, University of California Davis, Davis, CA, USA
| | - Charles DeCarli
- Alzheimer’s Disease Research Center, Department of Neurology, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Robert A. Rissman
- Department of Neurosciences, University of California San Diego, La Jolla, San Diego, CA, USA
| | - Andrew F. Teich
- Taub Institute for Research On Alzheimer’s Disease and Aging Brain, Department of Neurology, Columbia University Medical Center, New York, NY, USA
| | - Lee-Way Jin
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Brittany N. Dugger
- Department of Pathology and Laboratory Medicine, School of Medicine, University of California, Davis, Sacramento, CA, 95817, USA
| | - Michael J. Keiser
- Institute for Neurodegenerative Diseases, University of California, San Francisco, San Francisco, CA, 94158, USA
- Bakar Computational Health Sciences Institute, University of California, San Francisco, CA, 94158, USA
- Department of Pharmaceutical Chemistry, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| |
Collapse
|
9
|
Ingrassia L, Boluda S, Potier MC, Haïk S, Jimenez G, Kar A, Racoceanu D, Delatour B, Stimmer L. Automated deep learning segmentation of neuritic plaques and neurofibrillary tangles in Alzheimer disease brain sections using a proprietary software. J Neuropathol Exp Neurol 2024; 83:752-762. [PMID: 38812098 PMCID: PMC11333827 DOI: 10.1093/jnen/nlae048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024] Open
Abstract
Neuropathological diagnosis of Alzheimer disease (AD) relies on semiquantitative analysis of phosphorylated tau-positive neurofibrillary tangles (NFTs) and neuritic plaques (NPs), without consideration of lesion heterogeneity in individual cases. We developed a deep learning workflow for automated annotation and segmentation of NPs and NFTs from AT8-immunostained whole slide images (WSIs) of AD brain sections. Fifteen WSIs of frontal cortex from 4 biobanks with varying tissue quality, staining intensity, and scanning formats were analyzed. We established an artificial intelligence (AI)-driven iterative procedure to improve the generation of expert-validated annotation datasets for NPs and NFTs thereby increasing annotation quality by >50%. This strategy yielded an expert-validated annotation database with 5013 NPs and 5143 NFTs. We next trained two U-Net convolutional neural networks for detection and segmentation of NPs or NFTs, achieving high accuracy and consistency (mean Dice similarity coefficient: NPs, 0.77; NFTs, 0.81). The workflow showed high generalization performance across different cases. This study serves as a proof-of-concept for the utilization of proprietary image analysis software (Visiopharm) in the automated deep learning segmentation of NPs and NFTs, demonstrating that AI can significantly improve the annotation quality of complex neuropathological features and enable the creation of highly precise models for identifying these markers in AD brain sections.
Collapse
Affiliation(s)
- Lea Ingrassia
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Susana Boluda
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
- Department of Neuropathology Raymond Escourolle, AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Marie-Claude Potier
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Stéphane Haïk
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
- AP-HP, Cellule Nationale de Référence des MCJ, Salpêtrière Hospital, Paris, France
| | - Gabriel Jimenez
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Anuradha Kar
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Daniel Racoceanu
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Benoît Delatour
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| | - Lev Stimmer
- Paris Brain Institute (ICM), Centre National de la Recherche Scientifique (CNRS) UMR 7225, INSERM U1127, Sorbonne Université, Hôpital de la Pitié-Salpêtrière, Paris, France
| |
Collapse
|
10
|
Border S, Ferreira RM, Lucarelli N, Manthey D, Kumar S, Paul A, Mimar S, Naglah A, Cheng YH, Barisoni L, Ray J, Strekalova Y, Rosenberg AZ, Tomaszewski JE, Hodgin JB, El-Achkar TM, Jain S, Eadon MT, Sarder P. FUSION: A web-based application for in-depth exploration of multi-omics data with brightfield histology. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.09.602778. [PMID: 39026885 PMCID: PMC11257503 DOI: 10.1101/2024.07.09.602778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/20/2024]
Abstract
Spatial -OMICS technologies facilitate the interrogation of molecular profiles in the context of the underlying histopathology and tissue microenvironment. Paired analysis of histopathology and molecular data can provide pathologists with otherwise unobtainable insights into biological mechanisms. To connect the disparate molecular and histopathologic features into a single workspace, we developed FUSION (Functional Unit State IdentificatiON in WSIs [Whole Slide Images]), a web-based tool that provides users with a broad array of visualization and analytical tools including deep learning-based algorithms for in-depth interrogation of spatial -OMICS datasets and their associated high-resolution histology images. FUSION enables end-to-end analysis of functional tissue units (FTUs), automatically aggregating underlying molecular data to provide a histopathology-based medium for analyzing healthy and altered cell states and driving new discoveries using "pathomic" features. We demonstrate FUSION using 10x Visium spatial transcriptomics (ST) data from both formalin-fixed paraffin embedded (FFPE) and frozen prepared datasets consisting of healthy and diseased tissue. Through several use-cases, we demonstrate how users can identify spatial linkages between quantitative pathomics, qualitative image characteristics, and spatial --omics.
Collapse
Affiliation(s)
- Samuel Border
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Nicholas Lucarelli
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | | | - Suhas Kumar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Anindya Paul
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Sayat Mimar
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ahmed Naglah
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| | - Ying-Hua Cheng
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Laura Barisoni
- Department of Pathology, Division of AI and Computational Pathology, Duke University, Durham, NC
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC
| | - Jessica Ray
- Department of Health Outcomes & Biomedical Informatics, University of Florida, Gainesville, FL
| | - Yulia Strekalova
- College of Public Health and Health Professions, University of Florida, Gainesville, FL
| | - Avi Z Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine Baltimore, MD
| | - John E Tomaszewski
- Department of Pathology & Anatomical Sciences, Jacobs School of Medicine & Biomedical Sciences, University at Buffalo, State University of New York, Buffalo, NY
| | | | - Tarek M El-Achkar
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
- Indianapolis VA Medical Center, Indianapolis, IN
| | - Sanjay Jain
- Department of Medicine, Division of Nephrology, Washington University School of Medicine, St. Louis, MO
| | - Michael T Eadon
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN
| | - Pinaki Sarder
- Department of Medicine - Section of Quantitative Health, University of Florida, Gainesville, FL
| |
Collapse
|
11
|
Casotti MC, Meira DD, Zetum ASS, Campanharo CV, da Silva DRC, Giacinti GM, da Silva IM, Moura JAD, Barbosa KRM, Altoé LSC, Mauricio LSR, Góes LSBDB, Alves LNR, Linhares SSG, Ventorim VDP, Guaitolini YM, dos Santos EDVW, Errera FIV, Groisman S, de Carvalho EF, de Paula F, de Sousa MVP, Fechine PBA, Louro ID. Integrating frontiers: a holistic, quantum and evolutionary approach to conquering cancer through systems biology and multidisciplinary synergy. Front Oncol 2024; 14:1419599. [PMID: 39224803 PMCID: PMC11367711 DOI: 10.3389/fonc.2024.1419599] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 07/31/2024] [Indexed: 09/04/2024] Open
Abstract
Cancer therapy is facing increasingly significant challenges, marked by a wide range of techniques and research efforts centered around somatic mutations, precision oncology, and the vast amount of big data. Despite this abundance of information, the quest to cure cancer often seems more elusive, with the "war on cancer" yet to deliver a definitive victory. A particularly pressing issue is the development of tumor treatment resistance, highlighting the urgent need for innovative approaches. Evolutionary, Quantum Biology and System Biology offer a promising framework for advancing experimental cancer research. By integrating theoretical studies, translational methods, and flexible multidisciplinary clinical research, there's potential to enhance current treatment strategies and improve outcomes for cancer patients. Establishing stronger links between evolutionary, quantum, entropy and chaos principles and oncology could lead to more effective treatments that leverage an understanding of the tumor's evolutionary dynamics, paving the way for novel methods to control and mitigate cancer. Achieving these objectives necessitates a commitment to multidisciplinary and interprofessional collaboration at the heart of both research and clinical endeavors in oncology. This entails dismantling silos between disciplines, encouraging open communication and data sharing, and integrating diverse viewpoints and expertise from the outset of research projects. Being receptive to new scientific discoveries and responsive to how patients react to treatments is also crucial. Such strategies are key to keeping the field of oncology at the forefront of effective cancer management, ensuring patients receive the most personalized and effective care. Ultimately, this approach aims to push the boundaries of cancer understanding, treating it as a manageable chronic condition, aiming to extend life expectancy and enhance patient quality of life.
Collapse
Affiliation(s)
- Matheus Correia Casotti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Débora Dummer Meira
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | | | - Giulia Maria Giacinti
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Iris Moreira da Silva
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - João Augusto Diniz Moura
- Laboratório de Oncologia Clínica e Experimental, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Karen Ruth Michio Barbosa
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Lorena Souza Castro Altoé
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Lyvia Neves Rebello Alves
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Vinícius do Prado Ventorim
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | - Yasmin Moreto Guaitolini
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | | | - Sonia Groisman
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Elizeu Fagundes de Carvalho
- Instituto de Biologia Roberto Alcântara Gomes (IBRAG), Universidade do Estado do Rio de Janeiro (UERJ), Rio de Janeiro, RJ, Brazil
| | - Flavia de Paula
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| | | | - Pierre Basílio Almeida Fechine
- Group of Chemistry of Advanced Materials (GQMat), Department of Analytical Chemistry and Physical-Chemistry, Federal University of Ceará (UFC), Fortaleza, CE, Brazil
| | - Iuri Drumond Louro
- Núcleo de Genética Humana e Molecular, Universidade Federal do Espírito Santo (UFES), Vitória, ES, Brazil
| |
Collapse
|
12
|
Mi H, Sivagnanam S, Ho WJ, Zhang S, Bergman D, Deshpande A, Baras AS, Jaffee EM, Coussens LM, Fertig EJ, Popel AS. Computational methods and biomarker discovery strategies for spatial proteomics: a review in immuno-oncology. Brief Bioinform 2024; 25:bbae421. [PMID: 39179248 PMCID: PMC11343572 DOI: 10.1093/bib/bbae421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Revised: 07/11/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024] Open
Abstract
Advancements in imaging technologies have revolutionized our ability to deeply profile pathological tissue architectures, generating large volumes of imaging data with unparalleled spatial resolution. This type of data collection, namely, spatial proteomics, offers invaluable insights into various human diseases. Simultaneously, computational algorithms have evolved to manage the increasing dimensionality of spatial proteomics inherent in this progress. Numerous imaging-based computational frameworks, such as computational pathology, have been proposed for research and clinical applications. However, the development of these fields demands diverse domain expertise, creating barriers to their integration and further application. This review seeks to bridge this divide by presenting a comprehensive guideline. We consolidate prevailing computational methods and outline a roadmap from image processing to data-driven, statistics-informed biomarker discovery. Additionally, we explore future perspectives as the field moves toward interfacing with other quantitative domains, holding significant promise for precision care in immuno-oncology.
Collapse
Affiliation(s)
- Haoyang Mi
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Shamilene Sivagnanam
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
| | - Won Jin Ho
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Shuming Zhang
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Daniel Bergman
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
| | - Atul Deshpande
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Alexander S Baras
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Pathology, Johns Hopkins University School of Medicine, MD 21205, United States
- The Sidney Kimmel Comprehensive Cancer Center, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Elizabeth M Jaffee
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
| | - Lisa M Coussens
- The Knight Cancer Institute, Oregon Health and Science University, Portland, OR 97201, United States
- Department of Cell, Development and Cancer Biology, Oregon Health and Science University, Portland, OR 97201, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR 97201, United States
| | - Elana J Fertig
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
- Convergence Institute, Johns Hopkins University, Baltimore, MD 21205, United States
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Applied Mathematics and Statistics, Johns Hopkins University Whiting School of Engineering, Baltimore, MD 21218, United States
| | - Aleksander S Popel
- Department of Biomedical Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, United States
- Department of Oncology, Johns Hopkins University School of Medicine, MD 21205, United States
| |
Collapse
|
13
|
Liu S, Amgad M, More D, Rathore MA, Salgado R, Cooper LAD. A panoptic segmentation dataset and deep-learning approach for explainable scoring of tumor-infiltrating lymphocytes. NPJ Breast Cancer 2024; 10:52. [PMID: 38942745 PMCID: PMC11213912 DOI: 10.1038/s41523-024-00663-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Tumor-Infiltrating Lymphocytes (TILs) have strong prognostic and predictive value in breast cancer, but their visual assessment is subjective. To improve reproducibility, the International Immuno-oncology Working Group recently released recommendations for the computational assessment of TILs that build on visual scoring guidelines. However, existing resources do not adequately address these recommendations due to the lack of annotation datasets that enable joint, panoptic segmentation of tissue regions and cells. Moreover, existing deep-learning methods focus entirely on either tissue segmentation or cell nuclei detection, which complicates the process of TILs assessment by necessitating the use of multiple models and reconciling inconsistent predictions. We introduce PanopTILs, a region and cell-level annotation dataset containing 814,886 nuclei from 151 patients, openly accessible at: sites.google.com/view/panoptils . Using PanopTILs we developed MuTILs, a neural network optimized for assessing TILs in accordance with clinical recommendations. MuTILs is a concept bottleneck model designed to be interpretable and to encourage sensible predictions at multiple resolutions. Using a rigorous internal-external cross-validation procedure, MuTILs achieves an AUROC of 0.93 for lymphocyte detection and a DICE coefficient of 0.81 for tumor-associated stroma segmentation. Our computational score closely matched visual scores from 2 pathologists (Spearman R = 0.58-0.61, p < 0.001). Moreover, computational TILs scores had a higher prognostic value than visual scores, independent of TNM stage and patient age. In conclusion, we introduce a comprehensive open data resource and a modeling approach for detailed mapping of the breast tumor microenvironment.
Collapse
Affiliation(s)
- Shangke Liu
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | - Mohamed Amgad
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| | - Deeptej More
- Department of Pathology, Northwestern University, Chicago, IL, USA
| | | | - Roberto Salgado
- Department of Pathology, GZA-ZNA Ziekenhuizen, Antwerp, Belgium
- Division of Research, Peter MacCallum Cancer Centre, Melbourne, VIC, Australia
| | - Lee A D Cooper
- Department of Pathology, Northwestern University, Chicago, IL, USA.
| |
Collapse
|
14
|
Shephard AJ, Bashir RMS, Mahmood H, Jahanifar M, Minhas F, Raza SEA, McCombe KD, Craig SG, James J, Brooks J, Nankivell P, Mehanna H, Khurram SA, Rajpoot NM. A fully automated and explainable algorithm for predicting malignant transformation in oral epithelial dysplasia. NPJ Precis Oncol 2024; 8:137. [PMID: 38942998 PMCID: PMC11213925 DOI: 10.1038/s41698-024-00624-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Accepted: 05/29/2024] [Indexed: 06/30/2024] Open
Abstract
Oral epithelial dysplasia (OED) is a premalignant histopathological diagnosis given to lesions of the oral cavity. Its grading suffers from significant inter-/intra-observer variability, and does not reliably predict malignancy progression, potentially leading to suboptimal treatment decisions. To address this, we developed an artificial intelligence (AI) algorithm, that assigns an Oral Malignant Transformation (OMT) risk score based on the Haematoxylin and Eosin (H&E) stained whole slide images (WSIs). Our AI pipeline leverages an in-house segmentation model to detect and segment both nuclei and epithelium. Subsequently, a shallow neural network utilises interpretable morphological and spatial features, emulating histological markers, to predict progression. We conducted internal cross-validation on our development cohort (Sheffield; n = 193 cases) and independent validation on two external cohorts (Birmingham and Belfast; n = 89 cases). On external validation, the proposed OMTscore achieved an AUROC = 0.75 (Recall = 0.92) in predicting OED progression, outperforming other grading systems (Binary: AUROC = 0.72, Recall = 0.85). Survival analyses showed the prognostic value of our OMTscore (C-index = 0.60, p = 0.02), compared to WHO (C-index = 0.64, p = 0.003) and binary grades (C-index = 0.65, p < 0.001). Nuclear analyses elucidated the presence of peri-epithelial and intra-epithelial lymphocytes in highly predictive patches of transforming cases (p < 0.001). This is the first study to propose a completely automated, explainable, and externally validated algorithm for predicting OED transformation. Our algorithm shows comparable-to-human-level performance, offering a promising solution to the challenges of grading OED in routine clinical practice.
Collapse
Affiliation(s)
- Adam J Shephard
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | | | - Hanya Mahmood
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Mostafa Jahanifar
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Fayyaz Minhas
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Shan E Ahmed Raza
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK
| | - Kris D McCombe
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Stephanie G Craig
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jacqueline James
- Precision Medicine Centre, Patrick G. Johnston Centre for Cancer Research, Queen's University Belfast, Belfast, UK
| | - Jill Brooks
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Paul Nankivell
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Hisham Mehanna
- Institute of Head and Neck Studies and Education, Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham, UK
| | - Syed Ali Khurram
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Nasir M Rajpoot
- Tissue Image Analytics Centre, Department of Computer Science, University of Warwick, Coventry, UK.
| |
Collapse
|
15
|
Lin S, Zhou M, Cheng L, Shuai Z, Zhao M, Jie R, Wan Q, Peng F, Ding S. Exploring the association of POSTN + cancer-associated fibroblasts with triple-negative breast cancer. Int J Biol Macromol 2024; 268:131560. [PMID: 38631570 DOI: 10.1016/j.ijbiomac.2024.131560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 04/19/2024]
Abstract
Triple-negative breast cancer (TNBC) is a subtype of breast cancer with a poor prognosis. Cancer-associated fibroblasts (CAFs) play a critical role in regulating TNBC tumor development. This study aimed to identify and characterize a specific subtype of CAFs associated with TNBC. Initially, using high-throughput bulk transcriptomic data in two cohorts, we identified three CAF-related subtypes (CS1, CS2, CS3) in TNBC samples. These three CAFs subtypes were closely linked to the tumor microenvironment. The CS1 subtype exhibited a relatively immune-rich microenvironment and a favourable prognosis, whereas the CS3 subtype displayed an immune-deprived tumor microenvironment and an unfavourable prognosis. Through WGCNA analysis, POSTN was identified as a key biomarker for CAFs associated with TNBC. Then, POSTN+CAFs was identified and characterized. Both POSTN and POSTN+CAFs showed significant positive correlations with stromal molecules HGF and MET at both the transcriptional and protein levels. Specifically co-localized with CAFs in the tumor stromal area, POSTN, produced by POSTN+CAFs, could modulate the HGF-MET axis, serving as a bypass activation pathway to regulate tumor cell proliferation in response to EGFR inhibitor and MET inhibitor. This study underscores the significance of POSTN and POSTN+CAFs as crucial targets for the diagnosis and treatment of TNBC.
Collapse
Affiliation(s)
- Shuangyan Lin
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Pathology, Zhejiang Hospital, Zhejiang University School of Medicine, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Miaoni Zhou
- Department of Dermatology, Hangzhou Third People's Hospital, Affiliated Hangzhou Dermatology Hospital, Zhejiang University School of Medicine, 38 Xihu Rd, Hangzhou 310009, Zhejiang, China
| | - Liying Cheng
- Jiaxing University Medical College, 899 Shiguang Rd, Jiaxing 314001, Zhejiang, China
| | - Zhifeng Shuai
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Mingyuan Zhao
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Ruixia Jie
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China
| | - Qun Wan
- Department of Urinary Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, 79 Qingchun Road, Hangzhou 310003, Zhejiang, China
| | - Fang Peng
- Department of Pathology, Zhejiang Hospital, 12 Lingyin Rd, Hangzhou 310013, Zhejiang, China.
| | - Shiping Ding
- Department of Cell Biology and Department of Cardiovascular Surgery, The Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, 57 Zhugan Lane, Hangzhou 310000, Zhejiang, China; Department of Cell Biology, Zhejiang University School of Medicine, 866 Yuhangtang Rd, Hangzhou 310058, Zhejiang, China.
| |
Collapse
|
16
|
Hölscher DL, Goedertier M, Klinkhammer BM, Droste P, Costa IG, Boor P, Bülow RD. tRigon: an R package and Shiny App for integrative (path-)omics data analysis. BMC Bioinformatics 2024; 25:98. [PMID: 38443821 PMCID: PMC10916305 DOI: 10.1186/s12859-024-05721-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2023] [Accepted: 02/26/2024] [Indexed: 03/07/2024] Open
Abstract
BACKGROUND Pathomics facilitates automated, reproducible and precise histopathology analysis and morphological phenotyping. Similar to molecular omics, pathomics datasets are high-dimensional, but also face large outlier variability and inherent data missingness, making quick and comprehensible data analysis challenging. To facilitate pathomics data analysis and interpretation as well as support a broad implementation we developed tRigon (Toolbox foR InteGrative (path-)Omics data aNalysis), a Shiny application for fast, comprehensive and reproducible pathomics analysis. RESULTS tRigon is available via the CRAN repository ( https://cran.r-project.org/web/packages/tRigon ) with its source code available on GitLab ( https://git-ce.rwth-aachen.de/labooratory-ai/trigon ). The tRigon package can be installed locally and its application can be executed from the R console via the command 'tRigon::run_tRigon()'. Alternatively, the application is hosted online and can be accessed at https://labooratory.shinyapps.io/tRigon . We show fast computation of small, medium and large datasets in a low- and high-performance hardware setting, indicating broad applicability of tRigon. CONCLUSIONS tRigon allows researchers without coding abilities to perform exploratory feature analyses of pathomics and non-pathomics datasets on their own using a variety of hardware.
Collapse
Affiliation(s)
- David L Hölscher
- Institute of Pathology, RWTH Aachen University Clinic, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Clinic, Aachen, Germany
| | - Michael Goedertier
- Institute of Pathology, RWTH Aachen University Clinic, Aachen, Germany
- Institute for Computational Genomics, RWTH Aachen University Clinic, Aachen, Germany
| | | | - Patrick Droste
- Institute of Pathology, RWTH Aachen University Clinic, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Clinic, Aachen, Germany
| | - Ivan G Costa
- Institute for Computational Genomics, RWTH Aachen University Clinic, Aachen, Germany
| | - Peter Boor
- Institute of Pathology, RWTH Aachen University Clinic, Aachen, Germany
- Department of Nephrology and Immunology, RWTH Aachen University Clinic, Aachen, Germany
| | - Roman D Bülow
- Institute of Pathology, RWTH Aachen University Clinic, Aachen, Germany.
| |
Collapse
|
17
|
Amgad M, Hodge JM, Elsebaie MAT, Bodelon C, Puvanesarajah S, Gutman DA, Siziopikou KP, Goldstein JA, Gaudet MM, Teras LR, Cooper LAD. A population-level digital histologic biomarker for enhanced prognosis of invasive breast cancer. Nat Med 2024; 30:85-97. [PMID: 38012314 DOI: 10.1038/s41591-023-02643-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 28.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Accepted: 10/13/2023] [Indexed: 11/29/2023]
Abstract
Breast cancer is a heterogeneous disease with variable survival outcomes. Pathologists grade the microscopic appearance of breast tissue using the Nottingham criteria, which are qualitative and do not account for noncancerous elements within the tumor microenvironment. Here we present the Histomic Prognostic Signature (HiPS), a comprehensive, interpretable scoring of the survival risk incurred by breast tumor microenvironment morphology. HiPS uses deep learning to accurately map cellular and tissue structures to measure epithelial, stromal, immune, and spatial interaction features. It was developed using a population-level cohort from the Cancer Prevention Study-II and validated using data from three independent cohorts, including the Prostate, Lung, Colorectal, and Ovarian Cancer trial, Cancer Prevention Study-3, and The Cancer Genome Atlas. HiPS consistently outperformed pathologists in predicting survival outcomes, independent of tumor-node-metastasis stage and pertinent variables. This was largely driven by stromal and immune features. In conclusion, HiPS is a robustly validated biomarker to support pathologists and improve patient prognosis.
Collapse
Affiliation(s)
- Mohamed Amgad
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - James M Hodge
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Maha A T Elsebaie
- Department of Medicine, John H. Stroger, Jr. Hospital of Cook County, Chicago, IL, USA
| | - Clara Bodelon
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | | | - David A Gutman
- Department of Pathology, Emory University School of Medicine, Atlanta, GA, USA
| | - Kalliopi P Siziopikou
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Jeffery A Goldstein
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Mia M Gaudet
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, Bethesda, MD, USA
| | - Lauren R Teras
- Department of Population Science, American Cancer Society, Atlanta, GA, USA
| | - Lee A D Cooper
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
18
|
Lu W, Lashen AG, Wahab N, Miligy IM, Jahanifar M, Toss M, Graham S, Bilal M, Bhalerao A, Atallah NM, Makhlouf S, Ibrahim AY, Snead D, Minhas F, Raza SEA, Rakha E, Rajpoot N. AI-based intra-tumor heterogeneity score of Ki67 expression as a prognostic marker for early-stage ER+/HER2- breast cancer. J Pathol Clin Res 2024; 10:e346. [PMID: 37873865 PMCID: PMC10766021 DOI: 10.1002/cjp2.346] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Revised: 08/11/2023] [Accepted: 09/28/2023] [Indexed: 10/25/2023]
Abstract
Early-stage estrogen receptor positive and human epidermal growth factor receptor negative (ER+/HER2-) luminal breast cancer (BC) is quite heterogeneous and accounts for about 70% of all BCs. Ki67 is a proliferation marker that has a significant prognostic value in luminal BC despite the challenges in its assessment. There is increasing evidence that spatial colocalization, which measures the evenness of different types of cells, is clinically important in several types of cancer. However, reproducible quantification of intra-tumor spatial heterogeneity remains largely unexplored. We propose an automated pipeline for prognostication of luminal BC based on the analysis of spatial distribution of Ki67 expression in tumor cells using a large well-characterized cohort (n = 2,081). The proposed Ki67 colocalization (Ki67CL) score can stratify ER+/HER2- BC patients with high significance in terms of BC-specific survival (p < 0.00001) and distant metastasis-free survival (p = 0.0048). Ki67CL score is shown to be highly significant compared with the standard Ki67 index. In addition, we show that the proposed Ki67CL score can help identify luminal BC patients who can potentially benefit from adjuvant chemotherapy.
Collapse
Affiliation(s)
- Wenqi Lu
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Ayat G Lashen
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
- Department of Pathology, Faculty of MedicineMenoufia UniversityMenoufiaEgypt
| | - Noorul Wahab
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Islam M Miligy
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
- Department of Pathology, Faculty of MedicineMenoufia UniversityMenoufiaEgypt
| | - Mostafa Jahanifar
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Michael Toss
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
| | - Simon Graham
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Mohsin Bilal
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Abhir Bhalerao
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Nehal M Atallah
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
| | - Shorouk Makhlouf
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
| | - Asmaa Y Ibrahim
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
| | - David Snead
- Department of PathologyUniversity Hospitals Coventry and Warwickshire NHS TrustCoventryUK
| | - Fayyaz Minhas
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Shan E Ahmed Raza
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Emad Rakha
- Academic Unit for Translational Medical Sciences, School of MedicineUniversity of NottinghamNottinghamUK
| | - Nasir Rajpoot
- Tissue Image Analytics (TIA) Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| |
Collapse
|
19
|
Vizcarra JC, Pearce TM, Dugger BN, Keiser MJ, Gearing M, Crary JF, Kiely EJ, Morris M, White B, Glass JD, Farrell K, Gutman DA. Toward a generalizable machine learning workflow for neurodegenerative disease staging with focus on neurofibrillary tangles. Acta Neuropathol Commun 2023; 11:202. [PMID: 38110981 PMCID: PMC10726581 DOI: 10.1186/s40478-023-01691-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Accepted: 11/19/2023] [Indexed: 12/20/2023] Open
Abstract
Machine learning (ML) has increasingly been used to assist and expand current practices in neuropathology. However, generating large imaging datasets with quality labels is challenging in fields which demand high levels of expertise. Further complicating matters is the often seen disagreement between experts in neuropathology-related tasks, both at the case level and at a more granular level. Neurofibrillary tangles (NFTs) are a hallmark pathological feature of Alzheimer disease, and are associated with disease progression which warrants further investigation and granular quantification at a scale not currently accessible in routine human assessment. In this work, we first provide a baseline of annotator/rater agreement for the tasks of Braak NFT staging between experts and NFT detection using both experts and novices in neuropathology. We use a whole-slide-image (WSI) cohort of neuropathology cases from Emory University Hospital immunohistochemically stained for Tau. We develop a workflow for gathering annotations of the early stage formation of NFTs (Pre-NFTs) and mature intracellular (iNFTs) and show ML models can be trained to learn annotator nuances for the task of NFT detection in WSIs. We utilize a model-assisted-labeling approach and demonstrate ML models can be used to aid in labeling large datasets efficiently. We also show these models can be used to extract case-level features, which predict Braak NFT stages comparable to expert human raters, and do so at scale. This study provides a generalizable workflow for various pathology and related fields, and also provides a technique for accomplishing a high-level neuropathology task with limited human annotations.
Collapse
Affiliation(s)
- Juan C Vizcarra
- The Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, 313 Ferst Dr NW, Atlanta, GA, 30332, USA
| | - Thomas M Pearce
- Department of Pathology, Division of Neuropathology, University of Pittsburgh Medical Center, Room S701 Scaife Hall 3550 Terrace Street, Pittsburgh, PA, 15261, USA
| | - Brittany N Dugger
- Department of Pathology and Laboratory Medicine, University of California-Davis School of Medicine, 3400A Research Building III Sacramento, Davis, CA, 95817, USA
| | - Michael J Keiser
- Department of Pharmaceutical Chemistry, Department of Bioengineering and Therapeutic Sciences, Institute for Neurodegenerative Diseases, Kavli Institute for Fundamental Neuroscience, and Bakar Computational Health Sciences Institute, University of California, 675 Nelson Rising Ln, Box 0518, San Francisco, CA, 94143, USA
| | - Marla Gearing
- Department of Neurology, Emory University School of Medicine, 12 Executive Park Dr NE, Atlanta, GA, 30322, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - John F Crary
- Departments of Pathology, Neuroscience, and Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank and Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Icahn Building 9th Floor, Room 20A, 1425 Madison Avenue, New York, NY, 10029, USA
| | - Evan J Kiely
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Rd, Atlanta, GA, 30322, USA
| | - Meaghan Morris
- Department of Pathology, Johns Hopkins School of Medicine, Baltimore, MD, 21218, USA
| | - Bartholomew White
- Department of Pathology, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, Boston, MA, 02215, USA
| | - Jonathan D Glass
- Department of Neurology, Emory University School of Medicine, 12 Executive Park Dr NE, Atlanta, GA, 30322, USA
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Rd, Atlanta, GA, 30322, USA
- Center for Neurodegenerative Disease, Emory University School of Medicine, Whitehead Biomedical Research Building, 615 Michael Street, 5th Floor, Suite 500, Atlanta, GA, 30322, USA
| | - Kurt Farrell
- Departments of Pathology, Neuroscience, and Artificial Intelligence and Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank and Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Icahn Building 9th Floor, L9-02C, 1425 Madison, Avenue, New York, NY, USA
| | - David A Gutman
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, 1364 Clifton Rd, Atlanta, GA, 30322, USA.
| |
Collapse
|
20
|
Jesus R, Bastião Silva L, Sousa V, Carvalho L, Garcia Gonzalez D, Carias J, Costa C. Personalizable AI platform for universal access to research and diagnosis in digital pathology. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107787. [PMID: 37717524 DOI: 10.1016/j.cmpb.2023.107787] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND AND MOTIVATION Digital pathology has been evolving over the last years, proposing significant workflow advantages that have fostered its adoption in professional environments. Patient clinical and image data are readily available in remote data banks that can be consumed efficiently over standard communication technologies. The appearance of new imaging techniques and advanced artificial intelligence algorithms has significantly reduced the burden on medical professionals by speeding up the screening process. Despite these advancements, the usage of digital pathology in professional environments has been slowed down by poor interoperability between services resulting from a lack of standard interfaces and integrative solutions. This work addresses this issue by proposing a cloud-based digital pathology platform built on standard and open interfaces. METHODS The work proposes and describes a vendor-neutral platform that provides interfaces for managing digital slides, and medical reports, and integrating digital image analysis services compatible with existing standards. The solution integrates the open-source plugin-based Dicoogle PACS for interoperability and extensibility, which grants the proposed solution great feature customization. RESULTS The solution was developed in collaboration with iPATH research project partners, including the validation by medical pathologists. The result is a pure Web collaborative framework that supports both research and production environments. A total of 566 digital slides from different pathologies were successfully uploaded to the platform. Using the integration interfaces, a mitosis detection algorithm was successfully installed into the platform, and it was trained with 2400 annotations collected from breast carcinoma images. CONCLUSION Interoperability is a key factor when discussing digital pathology solutions, as it facilitates their integration into existing institutions' information systems. Moreover, it improves data sharing and integration of third-party services such as image analysis services, which have become relevant in today's digital pathology workflow. The proposed solution fully embraces the DICOM standard for digital pathology, presenting an interoperable cloud-based solution that provides great feature customization thanks to its extensible architecture.
Collapse
Affiliation(s)
- Rui Jesus
- University of A. Coruña, A Coruña, Spain; BMD Software, Aveiro, Portugal.
| | | | - Vítor Sousa
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lina Carvalho
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - João Carias
- Center for Computer Graphics, Braga, Portugal
| | - Carlos Costa
- IEETA/DETI, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
21
|
Lucarelli N, Ginley B, Zee J, Mimar S, Paul AS, Jain S, Han SS, Rodrigues L, Ozrazgat-Baslanti T, Wong ML, Nadkarni G, Clapp WL, Jen KY, Sarder P. Correlating Deep Learning-Based Automated Reference Kidney Histomorphometry with Patient Demographics and Creatinine. KIDNEY360 2023; 4:1726-1737. [PMID: 37966063 PMCID: PMC10758512 DOI: 10.34067/kid.0000000000000299] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 10/26/2023] [Indexed: 11/16/2023]
Abstract
Key Points The authors leverage the unique benefits of panoptic segmentation to perform the largest ever quantitation of reference kidney morphometry. Kidney features vary with age and sex; and glomeruli size may intricately link to creatinine, defying prior notions. Background Reference histomorphometric data of healthy human kidneys are largely lacking because of laborious quantitation requirements. Correlating histomorphometric features with clinical parameters through machine learning approaches can provide valuable information about natural population variance. To this end, we leveraged deep learning (DL), computational image analysis, and feature analysis to associate the relationship of histomorphometry with patient age, sex, serum creatinine (SCr), and eGFR in a multinational set of reference kidney tissue sections. Methods A panoptic segmentation neural network was developed and used to segment viable and sclerotic glomeruli, cortical and medullary interstitia, tubules, and arteries/arterioles in the digitized images of 79 periodic acid–Schiff-stained human nephrectomy sections showing minimal pathologic changes. Simple morphometrics (e.g. , area, radius, density) were quantified from the segmented classes. Regression analysis aided in determining the association of histomorphometric parameters with age, sex, SCr, and eGFR. Results Our DL model achieved high segmentation performance for all test compartments. The size and density of glomeruli, tubules, and arteries/arterioles varied significantly among healthy humans, with potentially large differences between geographically diverse patients. Glomerular size was significantly correlated with SCr and eGFR. Slight, albeit significant, differences in renal vasculature were observed between sexes. Glomerulosclerosis percentage increased, and cortical density of arteries/arterioles decreased, as a function of increasing age. Conclusions Using DL, we automated precise measurements of kidney histomorphometric features. In the reference kidney tissue, several histomorphometric features demonstrated significant correlation to patient demographics, SCr, and eGFR. DL tools can increase the efficiency and rigor of histomorphometric analysis.
Collapse
Affiliation(s)
- Nicholas Lucarelli
- J. Crayton Pruitt Family, Department of Biomedical Engineering, University of Florida Herbert Wertheim College of Engineering, Gainesville, Florida
| | - Brandon Ginley
- Departments of Pathology and Anatomical Sciences, University at Buffalo Jacobs School of Medicine and Biomedical Sciences – The State University of New York, Buffalo, New York
| | - Jarcy Zee
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania
- Children's Hospital of Philadelphia Research Institute, Philadelphia, Pennsylvania
| | - Sayat Mimar
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, College of Medicine, Gainesville, Florida
| | - Anindya S. Paul
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, College of Medicine, Gainesville, Florida
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, Missouri
- Departments of Pediatrics and Pathology, Washington University School of Medicine, St. Louis, Missouri
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Luis Rodrigues
- University Clinic of Nephrology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Tezcan Ozrazgat-Baslanti
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, College of Medicine, Gainesville, Florida
| | - Michelle L. Wong
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, California
| | - Girish Nadkarni
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, New York
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, New York
| | - William L. Clapp
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida College of Medicine, Gainesville, Florida
| | - Kuang-Yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, California
| | - Pinaki Sarder
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, College of Medicine, Gainesville, Florida
| |
Collapse
|
22
|
Cazzaniga G, Eccher A, Munari E, Marletta S, Bonoldi E, Della Mea V, Cadei M, Sbaraglia M, Guerriero A, Dei Tos AP, Pagni F, L’Imperio V. Natural Language Processing to extract SNOMED-CT codes from pathological reports. Pathologica 2023; 115:318-324. [PMID: 38180139 PMCID: PMC10767798 DOI: 10.32074/1591-951x-952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Accepted: 11/17/2023] [Indexed: 01/06/2024] Open
Abstract
OBJECTIVE The use of standardized structured reports (SSR) and suitable terminologies like SNOMED-CT can enhance data retrieval and analysis, fostering large-scale studies and collaboration. However, the still large prevalence of narrative reports in our laboratories warrants alternative and automated labeling approaches. In this project, natural language processing (NLP) methods were used to associate SNOMED-CT codes to structured and unstructured reports from an Italian Digital Pathology Department. METHODS Two NLP-based automatic coding systems (support vector machine, SVM, and long-short term memory, LSTM) were trained and applied to a series of narrative reports. RESULTS The 1163 cases were tested with both algorithms, showing good performances in terms of accuracy, precision, recall, and F1 score, with SVM showing slightly better performances as compared to LSTM (0.84, 0.87, 0.83, 0.82 vs 0.83, 0.85, 0.83, 0.82, respectively). The integration of an explainability allowed identification of terms and groups of words of importance, enabling fine-tuning, balancing semantic meaning and model performance. CONCLUSIONS AI tools allow the automatic SNOMED-CT labeling of the pathology archives, providing a retrospective fix to the large lack of organization of narrative reports.
Collapse
Affiliation(s)
- Giorgio Cazzaniga
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Italy
| | - Albino Eccher
- Section of Pathology, Department of Medical and Surgical Sciences for Children and Adults, University of Modena and Reggio Emilia, University Hospital of Modena, Modena, Italy
| | - Enrico Munari
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Stefano Marletta
- Department of Diagnostic and Public Health, Section of Pathology, University of Verona, Verona, Italy
| | - Emanuela Bonoldi
- Unit of Surgical Pathology and Cytogenetics, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Vincenzo Della Mea
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Moris Cadei
- Pathology Unit, ASST Spedali Civili di Brescia, Brescia, Italy
| | - Marta Sbaraglia
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Angela Guerriero
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Angelo Paolo Dei Tos
- Surgical Pathology and Cytopathology Unit, Department of Medicine-DIMED, University of Padua School of Medicine, Padua, Italy
| | - Fabio Pagni
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Italy
| | - Vincenzo L’Imperio
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Italy
| |
Collapse
|
23
|
Lutnick B, Ramon AJ, Ginley B, Csiszer C, Kim A, Flament I, Damasceno PF, Cornibe J, Parmar C, Standish K, Carrasco-Zevallos O, Yip SS. Accelerating pharmaceutical R&D with a user-friendly AI system for histopathology image analysis. J Pathol Inform 2023; 14:100337. [PMID: 37860714 PMCID: PMC10582575 DOI: 10.1016/j.jpi.2023.100337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 08/08/2023] [Accepted: 09/19/2023] [Indexed: 10/21/2023] Open
Abstract
A system for analysis of histopathology data within a pharmaceutical R&D environment has been developed with the intention of enabling interdisciplinary collaboration. State-of-the-art AI tools have been deployed as easy-to-use self-service modules within an open-source whole slide image viewing platform, so that non-data scientist users (e.g., clinicians) can utilize and evaluate pre-trained algorithms and retrieve quantitative results. The outputs of analysis are automatically cataloged in the database to track data provenance and can be viewed interactively on the slide as annotations or heatmaps. Commonly used models for analysis of whole slide images including segmentation, extraction of hand-engineered features for segmented regions, and slide-level classification using multi-instance learning are included and new models can be added as needed. The source code that supports running inference with these models internally is backed up by a robust CI/CD pipeline to ensure model versioning, robust testing, and seamless deployment of the latest models. Examples of the use of this system in a pharmaceutical development workflow include glomeruli segmentation, enumeration of podocyte count from WT-1 immuno-histochemistry, measurement of beta-1 integrin target engagement from immunofluorescence, digital glomerular phenotyping from periodic acid-Schiff histology, PD-L1 score prediction using multi-instance learning, and the deployment of the open-source Segment Anything model to speed up annotation.
Collapse
Affiliation(s)
| | | | | | | | - Alex Kim
- Janssen R&D, Data Sciences, Raritan, NJ 08869, USA
| | - Io Flament
- Janssen R&D, Data Sciences, Raritan, NJ 08869, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Sauter D, Lodde G, Nensa F, Schadendorf D, Livingstone E, Kukuk M. Deep learning in computational dermatopathology of melanoma: A technical systematic literature review. Comput Biol Med 2023; 163:107083. [PMID: 37315382 DOI: 10.1016/j.compbiomed.2023.107083] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 05/10/2023] [Accepted: 05/27/2023] [Indexed: 06/16/2023]
Abstract
Deep learning (DL) has become one of the major approaches in computational dermatopathology, evidenced by a significant increase in this topic in the current literature. We aim to provide a structured and comprehensive overview of peer-reviewed publications on DL applied to dermatopathology focused on melanoma. In comparison to well-published DL methods on non-medical images (e.g., classification on ImageNet), this field of application comprises a specific set of challenges, such as staining artifacts, large gigapixel images, and various magnification levels. Thus, we are particularly interested in the pathology-specific technical state-of-the-art. We also aim to summarize the best performances achieved thus far with respect to accuracy, along with an overview of self-reported limitations. Accordingly, we conducted a systematic literature review of peer-reviewed journal and conference articles published between 2012 and 2022 in the databases ACM Digital Library, Embase, IEEE Xplore, PubMed, and Scopus, expanded by forward and backward searches to identify 495 potentially eligible studies. After screening for relevance and quality, a total of 54 studies were included. We qualitatively summarized and analyzed these studies from technical, problem-oriented, and task-oriented perspectives. Our findings suggest that the technical aspects of DL for histopathology in melanoma can be further improved. The DL methodology was adopted later in this field, and still lacks the wider adoption of DL methods already shown to be effective for other applications. We also discuss upcoming trends toward ImageNet-based feature extraction and larger models. While DL has achieved human-competitive accuracy in routine pathological tasks, its performance on advanced tasks is still inferior to wet-lab testing (for example). Finally, we discuss the challenges impeding the translation of DL methods to clinical practice and provide insight into future research directions.
Collapse
Affiliation(s)
- Daniel Sauter
- Department of Computer Science, Fachhochschule Dortmund, 44227 Dortmund, Germany.
| | - Georg Lodde
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | - Felix Nensa
- Institute for AI in Medicine (IKIM), University Hospital Essen, 45131 Essen, Germany; Institute of Diagnostic and Interventional Radiology and Neuroradiology, University Hospital Essen, 45147 Essen, Germany
| | - Dirk Schadendorf
- Department of Dermatology, University Hospital Essen, 45147 Essen, Germany
| | | | - Markus Kukuk
- Department of Computer Science, Fachhochschule Dortmund, 44227 Dortmund, Germany
| |
Collapse
|
25
|
Wicks MN, Glinka M, Hill B, Houghton D, Sharghi M, Ferreira I, Adams D, Din S, Papatheodorou I, Kirkwood K, Cheeseman M, Burger A, Baldock RA, Arends MJ. The Comparative Pathology Workbench: Interactive visual analytics for biomedical data. J Pathol Inform 2023; 14:100328. [PMID: 37693862 PMCID: PMC10491844 DOI: 10.1016/j.jpi.2023.100328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 07/07/2023] [Accepted: 08/04/2023] [Indexed: 09/12/2023] Open
Abstract
Pathologists need to compare histopathological images of normal and diseased tissues between different samples, cases, and species. We have designed an interactive system, termed Comparative Pathology Workbench (CPW), which allows direct and dynamic comparison of images at a variety of magnifications, selected regions of interest, as well as the results of image analysis or other data analyses such as scRNA-seq. This allows pathologists to indicate key diagnostic features, with a mechanism to allow discussion threads amongst expert groups of pathologists and other disciplines. The data and associated discussions can be accessed online from anywhere in the world. The Comparative Pathology Workbench (CPW) is a web-browser-based visual analytics platform providing shared access to an interactive "spreadsheet" style presentation of image and associated analysis data. The CPW provides a grid layout of rows and columns so that images that correspond to matching data can be organised in the form of an image-enabled "spreadsheet". An individual workbench can be shared with other users with read-only or full edit access as required. In addition, each workbench element or the whole bench itself has an associated discussion thread to allow collaborative analysis and consensual interpretation of the data. The CPW is a Django-based web-application that hosts the workbench data, manages users, and user-preferences. All image data are hosted by other resource applications such as OMERO or the Digital Slide Archive. Further resources can be added as required. The discussion threads are managed using WordPress and include additional graphical and image data. The CPW has been developed to allow integration of image analysis outputs from systems such as QuPath or ImageJ. All software is open-source and available from a GitHub repository.
Collapse
Affiliation(s)
- Michael N. Wicks
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Michael Glinka
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Bill Hill
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Derek Houghton
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Mehran Sharghi
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Ingrid Ferreira
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - David Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Shahida Din
- Edinburgh IBD Unit Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Irene Papatheodorou
- European Molecular Biology Laboratory - European Bioinformatics Institute (EMBL-EBI), Hinxton, Cambridge, UK
| | - Kathryn Kirkwood
- Pathology Department, Western General Hospital, NHS Lothian, Edinburgh, UK
| | - Michael Cheeseman
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Albert Burger
- Department of Computer Science, School of Mathematical and Computer Sciences, Heriot-Watt University, Edinburgh, UK
| | - Richard A. Baldock
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| | - Mark J. Arends
- Edinburgh Pathology & Centre for Comparative Pathology, Institute of Genetics & Cancer, University of Edinburgh, Crewe Road, Edinburgh EH4 2XR, UK
| |
Collapse
|
26
|
Berman AG, Orchard WR, Gehrung M, Markowetz F. SliDL: A toolbox for processing whole-slide images in deep learning. PLoS One 2023; 18:e0289499. [PMID: 37549131 PMCID: PMC10406329 DOI: 10.1371/journal.pone.0289499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 07/20/2023] [Indexed: 08/09/2023] Open
Abstract
The inspection of stained tissue slides by pathologists is essential for the early detection, diagnosis and monitoring of disease. Recently, deep learning methods for the analysis of whole-slide images (WSIs) have shown excellent performance on these tasks, and have the potential to substantially reduce the workload of pathologists. However, WSIs present a number of unique challenges for analysis, requiring special consideration of image annotations, slide and image artefacts, and evaluation of WSI-trained model performance. Here we introduce SliDL, a Python library for performing pre- and post-processing of WSIs. SliDL makes WSI data handling easy, allowing users to perform essential processing tasks in a few simple lines of code, bridging the gap between standard image analysis and WSI analysis. We introduce each of the main functionalities within SliDL: from annotation and tile extraction to tissue detection and model evaluation. We also provide 'code snippets' to guide the user in running SliDL. SliDL has been designed to interact with PyTorch, one of the most widely used deep learning libraries, allowing seamless integration into deep learning workflows. By providing a framework in which deep learning methods for WSI analysis can be developed and applied, SliDL aims to increase the accessibility of an important application of deep learning.
Collapse
Affiliation(s)
- Adam G. Berman
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - William R. Orchard
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Marcel Gehrung
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| | - Florian Markowetz
- Cancer Research UK Cambridge Institute, University of Cambridge, Cambridge, United Kingdom
| |
Collapse
|
27
|
Spratt DE, Tang S, Sun Y, Huang HC, Chen E, Mohamad O, Armstrong AJ, Tward JD, Nguyen PL, Lang JM, Zhang J, Mitani A, Simko JP, DeVries S, van der Wal D, Pinckaers H, Monson JM, Campbell HA, Wallace J, Ferguson MJ, Bahary JP, Schaeffer EM, Sandler HM, Tran PT, Rodgers JP, Esteva A, Yamashita R, Feng FY. Artificial Intelligence Predictive Model for Hormone Therapy Use in Prostate Cancer. NEJM EVIDENCE 2023; 2:EVIDoa2300023. [PMID: 38320143 PMCID: PMC11195914 DOI: 10.1056/evidoa2300023] [Citation(s) in RCA: 42] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2024]
Abstract
BACKGROUND: Androgen deprivation therapy (ADT) with radiotherapy can benefit patients with localized prostate cancer. However, ADT can negatively impact quality of life, and there remain no validated predictive models to guide its use. METHODS: We used digital pathology images from pretreatment prostate tissue and clinical data from 5727 patients enrolled in five phase 3 randomized trials, in which treatment was radiotherapy with or without ADT, as our data source to develop and validate an artificial intelligence (AI)–derived predictive patient-specific model that would determine which patients would develop the primary end point of distant metastasis. The model used baseline data to provide a binary output that a given patient will likely benefit from ADT or not. After the model was locked, validation was performed using data from NRG Oncology/Radiation Therapy Oncology Group (RTOG) 9408 (n=1594), a trial that randomly assigned men to radiotherapy plus or minus 4 months of ADT. Fine–Gray regression and restricted mean survival times were used to assess the interaction between treatment and the predictive model and within predictive model–positive, i.e., benefited from ADT, and –negative subgroup treatment effects. RESULTS: Overall, in the NRG/RTOG 9408 validation cohort (14.9 years of median follow-up), ADT significantly improved time to distant metastasis. Of these enrolled patients, 543 (34%) were model positive, and ADT significantly reduced the risk of distant metastasis compared with radiotherapy alone. Of 1051 patients who were model negative, ADT did not provide benefit. CONCLUSIONS: Our AI-based predictive model was able to identify patients with a predominantly intermediate risk for prostate cancer likely to benefit from short-term ADT. (Supported by a grant [U10CA180822] from NRG Oncology Statistical and Data Management Center, a grant [UG1CA189867] from NCI Community Oncology Research Program, a grant [U10CA180868] from NRG Oncology Operations, and a grant [U24CA196067] from NRG Specimen Bank from the National Cancer Institute and by Artera, Inc. ClinicalTrials.gov numbers NCT00767286, NCT00002597, NCT00769548, NCT00005044, and NCT00033631.)
Collapse
Affiliation(s)
- Daniel E Spratt
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland
| | - Siyi Tang
- Department of Electrical Engineering, Stanford University, Stanford, CA
- Artera, Inc., Los Altos, CA
| | - Yilun Sun
- Department of Radiation Oncology, University Hospitals Seidman Cancer Center, Case Western Reserve University, Cleveland
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, Cleveland
| | | | | | - Osama Mohamad
- Department of Radiation Oncology, University of California, San Francisco, San Francisco
| | - Andrew J Armstrong
- Duke Cancer Institute Center for Prostate and Urologic Cancer, Division of Medical Oncology, Department of Medicine, Duke University, Durham, NC
| | - Jonathan D Tward
- Department of Radiation Oncology, Huntsman Cancer Institute, University of Utah, Salt Lake City
| | - Paul L Nguyen
- Department of Radiation Oncology, Dana-Farber/Brigham Cancer Center, Boston
| | - Joshua M Lang
- Division of Hematology/Medical Oncology, University of Wisconsin, Madison, WI
| | | | | | - Jeffry P Simko
- Department of Radiation Oncology, University of California, San Francisco, San Francisco
| | - Sandy DeVries
- NRG Oncology Biospecimen Bank, University of California, San Francisco, San Francisco
| | | | | | - Jedidiah M Monson
- Department of Radiation Oncology, Saint Agnes Medical Center, Fresno, CA
| | - Holly A Campbell
- Department of Radiation Oncology, Saint John Regional Hospital, Saint John, NB, Canada
| | - James Wallace
- University of Chicago Medicine Medical Group, Chicago
| | - Michelle J Ferguson
- Department of Radiation Oncology, Allan Blair Cancer Centre, Regina, SK, Canada
| | - Jean-Paul Bahary
- Department of Radiation Oncology, Centre Hospitalier de l'Universite de Montreal, Montreal
| | - Edward M Schaeffer
- Department of Urology, Northwestern University Feinberg School of Medicine, Chicago
| | - Howard M Sandler
- Department of Radiation Oncology, Cedars-Sinai Medical Center, Los Angeles
| | - Phuoc T Tran
- Department of Radiation Oncology, University of Maryland School of Medicine, Baltimore
| | - Joseph P Rodgers
- Statistics and Data Management Center, NRG Oncology, Philadelphia
- Statistics and Data Management Center, American College of Radiology, Philadelphia
| | | | | | - Felix Y Feng
- Department of Radiation Oncology, University of California, San Francisco, San Francisco
| |
Collapse
|
28
|
McDonnell KJ. Leveraging the Academic Artificial Intelligence Silecosystem to Advance the Community Oncology Enterprise. J Clin Med 2023; 12:4830. [PMID: 37510945 PMCID: PMC10381436 DOI: 10.3390/jcm12144830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/05/2023] [Accepted: 07/07/2023] [Indexed: 07/30/2023] Open
Abstract
Over the last 75 years, artificial intelligence has evolved from a theoretical concept and novel paradigm describing the role that computers might play in our society to a tool with which we daily engage. In this review, we describe AI in terms of its constituent elements, the synthesis of which we refer to as the AI Silecosystem. Herein, we provide an historical perspective of the evolution of the AI Silecosystem, conceptualized and summarized as a Kuhnian paradigm. This manuscript focuses on the role that the AI Silecosystem plays in oncology and its emerging importance in the care of the community oncology patient. We observe that this important role arises out of a unique alliance between the academic oncology enterprise and community oncology practices. We provide evidence of this alliance by illustrating the practical establishment of the AI Silecosystem at the City of Hope Comprehensive Cancer Center and its team utilization by community oncology providers.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
29
|
Liu L, Wang Y, Chang J, Zhang P, Xiong S, Liu H. A correlation graph attention network for classifying chromosomal instabilities from histopathology whole-slide images. iScience 2023; 26:106874. [PMID: 37260749 PMCID: PMC10227422 DOI: 10.1016/j.isci.2023.106874] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 02/23/2023] [Accepted: 05/10/2023] [Indexed: 06/02/2023] Open
Abstract
The chromosome instability (CIN) is one of the hallmarks of cancer and is closely related to tumor metastasis. However, the sheer size and resolution of histopathology whole-slide images (WSIs) already challenges the capabilities of computational pathology. In this study, we propose a correlation graph attention network (MLP-GAT) that can construct graphs for classifying multi-type CINs from the WSIs of breast cancer. We construct a WSIs dataset of breast cancer from the Cancer Genome Atlas Breast Invasive Carcinoma (TCGA-BRCA). Extensive experiments show that MLP-GAT far outperforms accepted state-of-the-art methods and demonstrate the advantages of the constructed graph networks for analyzing WSI data. The visualization shows the difference among the tiles in a WSI. Furthermore, the generalization performance of the proposed method was verified on the stomach cancer. This study provides guidance for studying the relationship between CIN and cancer from the perspective of image phenotype.
Collapse
Affiliation(s)
- Liangliang Liu
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Ying Wang
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Jing Chang
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Pei Zhang
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Shufeng Xiong
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| | - Hebing Liu
- College of Information and Management Science, Henan Agricultural University, Zhengzhou, Henan 450046, P.R. China
| |
Collapse
|
30
|
Bumgardner VKC, Armstrong S, Virodov A, Hickey C. Automated Curation and AI Workflow Management System for Digital Pathology. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE PROCEEDINGS. AMIA JOINT SUMMITS ON TRANSLATIONAL SCIENCE 2023; 2023:71-80. [PMID: 37350884 PMCID: PMC10283146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 06/24/2023]
Abstract
Digital pathology applications present several challenges, including the processing, storage, and distribution of gigapixel images across distributed computational resources and viewing stations. Individual slides must be available for interactive review, and large repositories must be programmatically accessible for dataset and model building. We present a platform to manage and process multi-modal pathology data (images and case information) across multiple locations. Using an agent-based system coupled with open-source automated machine learning and review tools allows not only dynamic load-balancing and cross-network operation but also the development of research and clinical AI models using the data managed by the platform. The platform presented covers end-to-end AI workflow from data acquisition and curation through model training and evaluation allowing for sharing and review. We conclude with a case study of colon and prostate cancer model development utilizing the presented system.
Collapse
|
31
|
Ginley B, Lucarelli N, Zee J, Jain S, Han SS, Rodrigues L, Ozrazgat-Baslanti T, Wong ML, Nadkarni G, Jen KY, Sarder P. Correlating Deep Learning-Based Automated Reference Kidney Histomorphometry with Patient Demographics and Creatinine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.18.541348. [PMID: 37292965 PMCID: PMC10245721 DOI: 10.1101/2023.05.18.541348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Background Reference histomorphometric data of healthy human kidneys are largely lacking due to laborious quantitation requirements. Correlating histomorphometric features with clinical parameters through machine learning approaches can provide valuable information about natural population variance. To this end, we leveraged deep learning, computational image analysis, and feature analysis to investigate the relationship of histomorphometry with patient age, sex, and serum creatinine (SCr) in a multinational set of reference kidney tissue sections. Methods A panoptic segmentation neural network was developed and used to segment viable and sclerotic glomeruli, cortical and medullary interstitia, tubules, and arteries/arterioles in the digitized images of 79 periodic acid-Schiff-stained human nephrectomy sections showing minimal pathologic changes. Simple morphometrics (e.g., area, radius, density) were quantified from the segmented classes. Regression analysis aided in determining the relationship of histomorphometric parameters with age, sex, and SCr. Results Our deep-learning model achieved high segmentation performance for all test compartments. The size and density of nephrons and arteries/arterioles varied significantly among healthy humans, with potentially large differences between geographically diverse patients. Nephron size was significantly dependent on SCr. Slight, albeit significant, differences in renal vasculature were observed between sexes. Glomerulosclerosis percentage increased, and cortical density of arteries/arterioles decreased, as a function of age. Conclusions Using deep learning, we automated precise measurements of kidney histomorphometric features. In the reference kidney tissue, several histomorphometric features demonstrated significant correlation to patient demographics and SCr. Deep learning tools can increase the efficiency and rigor of histomorphometric analysis.
Collapse
Affiliation(s)
- Brandon Ginley
- Departments of Pathology & Anatomical Sciences, University at Buffalo Jacobs School of Medicine and Biomedical Sciences – The State University of New York, Buffalo, NY, USA
| | - Nicholas Lucarelli
- Department of Biomedical Engineering, University of Florida College of Engineering, Gainesville, FL Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Jarcy Zee
- Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- Departments of Pediatrics and Pathology, Washington University School of Medicine, St. Louis, MO, USA
| | - Seung Sook Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Luis Rodrigues
- University Clinic of Nephrology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Nephrology Unit, Centro Hospitalare Universitário de Coimbra, Coimbra, Portugal
| | - Tezcan Ozrazgat-Baslanti
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, FL, USA
| | - Michelle L. Wong
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Girish Nadkarni
- The Mount Sinai Clinical Intelligence Center, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Nephrology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Kuang-Yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Pinaki Sarder
- Quantitative Health Section, Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, FL, USA
- Department of Electrical & Computer Engineering, University of Florida College of Engineering, Gainesville, FL, USA
| |
Collapse
|
32
|
Spratt DE, Tang S, Sun Y, Huang HC, Chen E, Mohamad O, Armstrong AJ, Tward JD, Nguyen PL, Lang JM, Zhang J, Mitani A, Simko JP, DeVries S, van der Wal D, Pinckaers H, Monson JM, Campbell HA, Wallace J, Ferguson MJ, Bahary JP, Schaeffer EM, Sandler HM, Tran PT, Rodgers JP, Esteva A, Yamashita R, Feng FY. Artificial Intelligence Predictive Model for Hormone Therapy Use in Prostate Cancer. RESEARCH SQUARE 2023:rs.3.rs-2790858. [PMID: 37131691 PMCID: PMC10153374 DOI: 10.21203/rs.3.rs-2790858/v1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Background Androgen deprivation therapy (ADT) with radiotherapy can benefit patients with localized prostate cancer. However, ADT can negatively impact quality of life and there remain no validated predictive models to guide its use. Methods Digital pathology image and clinical data from pre-treatment prostate tissue from 5,727 patients enrolled on five phase III randomized trials treated with radiotherapy +/- ADT were used to develop and validate an artificial intelligence (AI)-derived predictive model to assess ADT benefit with the primary endpoint of distant metastasis. After the model was locked, validation was performed on NRG/RTOG 9408 (n = 1,594) that randomized men to radiotherapy +/- 4 months of ADT. Fine-Gray regression and restricted mean survival times were used to assess the interaction between treatment and predictive model and within predictive model positive and negative subgroup treatment effects. Results In the NRG/RTOG 9408 validation cohort (14.9 years of median follow-up), ADT significantly improved time to distant metastasis (subdistribution hazard ratio [sHR] = 0.64, 95%CI [0.45-0.90], p = 0.01). The predictive model-treatment interaction was significant (p-interaction = 0.01). In predictive model positive patients (n = 543, 34%), ADT significantly reduced the risk of distant metastasis compared to radiotherapy alone (sHR = 0.34, 95%CI [0.19-0.63], p < 0.001). There were no significant differences between treatment arms in the predictive model negative subgroup (n = 1,051, 66%; sHR = 0.92, 95%CI [0.59-1.43], p = 0.71). Conclusions Our data, derived and validated from completed randomized phase III trials, show that an AI-based predictive model was able to identify prostate cancer patients, with predominately intermediate-risk disease, who are likely to benefit from short-term ADT.
Collapse
|
33
|
Goldstein JA, Nateghi R, Irmakci I, Cooper LAD. Machine learning classification of placental villous infarction, perivillous fibrin deposition, and intervillous thrombus. Placenta 2023; 135:43-50. [PMID: 36958179 PMCID: PMC10156426 DOI: 10.1016/j.placenta.2023.03.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 03/09/2023] [Accepted: 03/11/2023] [Indexed: 03/17/2023]
Abstract
INTRODUCTION Placental parenchymal lesions are commonly encountered and carry significant clinical associations. However, they are frequently missed or misclassified by general practice pathologists. Interpretation of pathology slides has emerged as one of the most successful applications of machine learning (ML) in medicine with applications ranging from cancer detection and prognostication to transplant medicine. The goal of this study was to use a whole-slide learning model to identify and classify placental parenchymal lesions including villous infarctions, intervillous thrombi (IVT), and perivillous fibrin deposition (PVFD). METHODS We generated whole slide images from placental discs examined at our institution with infarct, IVT, PVFD, or no macroscopic lesion. Slides were analyzed as a set of overlapping patches. We extracted feature vectors from each patch using a pretrained convolutional neural network (EfficientNetV2L). We trained a model to assign attention to each vector and used the attentions as weights to produce a pooled feature vector. The pooled vector was classified as normal or 1 of 3 lesions using a fully connected network. Patch attention was plotted to highlight informative areas of the slide. RESULTS Overall balanced accuracy in a test set of held-out slides was 0.86 with receiver-operator characteristic areas under the curve of 0.917-0.993. Cases of PVFD were frequently miscalled as normal or infarcts, the latter possibly due to the perivillous fibrin found at the periphery of infarctions. We used attention maps to further understand some errors, including one most likely due to poor tissue fixation and processing. DISCUSSION We used a whole-slide learning paradigm to train models to recognize three of the most common placental parenchymal lesions. We used attention maps to gain insight into model function, which differed from intuitive explanations.
Collapse
Affiliation(s)
| | - Ramin Nateghi
- Northwestern University, Department of Pathology, Chicago, IL, USA
| | - Ismail Irmakci
- Northwestern University, Department of Pathology, Chicago, IL, USA
| | - Lee A D Cooper
- Northwestern University, Department of Pathology, Chicago, IL, USA; Northwestern University, McCormick School of Engineering, Evanston, IL, USA
| |
Collapse
|
34
|
Sobral PS, Luz VCC, Almeida JMGCF, Videira PA, Pereira F. Computational Approaches Drive Developments in Immune-Oncology Therapies for PD-1/PD-L1 Immune Checkpoint Inhibitors. Int J Mol Sci 2023; 24:ijms24065908. [PMID: 36982981 PMCID: PMC10054797 DOI: 10.3390/ijms24065908] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2023] [Revised: 03/16/2023] [Accepted: 03/19/2023] [Indexed: 03/30/2023] Open
Abstract
Computational approaches in immune-oncology therapies focus on using data-driven methods to identify potential immune targets and develop novel drug candidates. In particular, the search for PD-1/PD-L1 immune checkpoint inhibitors (ICIs) has enlivened the field, leveraging the use of cheminformatics and bioinformatics tools to analyze large datasets of molecules, gene expression and protein-protein interactions. Up to now, there is still an unmet clinical need for improved ICIs and reliable predictive biomarkers. In this review, we highlight the computational methodologies applied to discovering and developing PD-1/PD-L1 ICIs for improved cancer immunotherapies with a greater focus in the last five years. The use of computer-aided drug design structure- and ligand-based virtual screening processes, molecular docking, homology modeling and molecular dynamics simulations methodologies essential for successful drug discovery campaigns focusing on antibodies, peptides or small-molecule ICIs are addressed. A list of recent databases and web tools used in the context of cancer and immunotherapy has been compilated and made available, namely regarding a general scope, cancer and immunology. In summary, computational approaches have become valuable tools for discovering and developing ICIs. Despite significant progress, there is still a need for improved ICIs and biomarkers, and recent databases and web tools have been compiled to aid in this pursuit.
Collapse
Affiliation(s)
- Patrícia S Sobral
- LAQV and REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Vanessa C C Luz
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - João M G C F Almeida
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Paula A Videira
- UCIBIO, Applied Molecular Biosciences Unit, Department of Life Sciences, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
- Associate Laboratory i4HB-Institute for Health and Bioeconomy, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| | - Florbela Pereira
- LAQV and REQUIMTE, Department of Chemistry, NOVA School of Science and Technology, Universidade NOVA de Lisboa, 2829-516 Caparica, Portugal
| |
Collapse
|
35
|
Huss R, Raffler J, Märkl B. Artificial intelligence and digital biomarker in precision pathology guiding immune therapy selection and precision oncology. Cancer Rep (Hoboken) 2023:e1796. [PMID: 36813293 PMCID: PMC10363837 DOI: 10.1002/cnr2.1796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND The currently available immunotherapies already changed the strategy how many cancers are treated from first to last line. Understanding even the most complex heterogeneity in tumor tissue and mapping the spatial cartography of the tumor immunity allows the best and optimized selection of immune modulating agents to (re-)activate the patient's immune system and direct it against the individual cancer in the most effective way. RECENT FINDINGS Primary cancer and metastases maintain a high degree of plasticity to escape any immune surveillance and continue to evolve depending on many intrinsic and extrinsic factors In the field of immune-oncology (IO) immune modulating agents are recognized as practice changing therapeutic modalities. Recent studies have shown that an optimal and lasting efficacy of IO therapeutics depends on the understanding of the spatial communication network and functional context of immune and cancer cells within the tumor microenvironment. Artificial intelligence (AI) provides an insight into the immune-cancer-network through the visualization of very complex tumor and immune interactions in cancer tissue specimens and allows the computer-assisted development and clinical validation of such digital biomarker. CONCLUSIONS The successful implementation of AI-supported digital biomarker solutions guides the clinical selection of effective immune therapeutics based on the retrieval and visualization of spatial and contextual information from cancer tissue images and standardized data. As such, computational pathology (CP) turns into "precision pathology" delivering individual therapy response prediction. Precision Pathology does not only include digital and computational solutions but also high levels of standardized processes in the routine histopathology workflow and the use of mathematical tools to support clinical and diagnostic decisions as the basic principle of a "precision oncology".
Collapse
Affiliation(s)
- Ralf Huss
- Medical Faculty University Augsburg, Augsburg, Germany
- Institute for Digital Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Johannes Raffler
- Institute for Digital Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Medical Faculty University Augsburg, Augsburg, Germany
| |
Collapse
|
36
|
Razian SA, Jadidi M. Histology Image Viewer and Converter (HIVC): A High-Speed Freeware Software to View and Convert Whole Slide Histology Images. COMPUTER METHODS IN BIOMECHANICS AND BIOMEDICAL ENGINEERING. IMAGING & VISUALIZATION 2023; 11:1652-1660. [PMID: 37994355 PMCID: PMC10662701 DOI: 10.1080/21681163.2023.2174776] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/26/2023] [Indexed: 02/07/2023]
Abstract
Histology images are widely used to assess the microstructure of biological tissues, but scanners often save images in bulky SVS and multi-layered TIFF formats. These formats were designed to archive image blocks and high-resolution textual information and are not compatible with conventional image analysis software. Our goal was to create a freeware Histology Image Viewer and Converter (HIVC) with a graphical user interface that allows viewing and converting whole-slide images in batch. HIVC was developed using C# Language for Windows x64 operating system. HIVC's performance was assessed by converting 20 whole-slide images to a JPG format at 20x and 40x resolution and comparing the results to ImageJ, Cell Profiler, QuPath, Nanoborb, and Aperio ImageScope. HIVC was more than 8-times faster in converting images than other software packages. This software allows high-speed batch conversion of histology images to traditional formats, permitting platform-independent secondary analyses.
Collapse
Affiliation(s)
| | - Majid Jadidi
- Department of Biomechanics, University of Nebraska Omaha, Omaha, NE
| |
Collapse
|
37
|
Li B, Nelson MS, Chacko JV, Cudworth N, Eliceiri KW. Hardware-software co-design of an open-source automatic multimodal whole slide histopathology imaging system. JOURNAL OF BIOMEDICAL OPTICS 2023; 28:026501. [PMID: 36761254 PMCID: PMC9905038 DOI: 10.1117/1.jbo.28.2.026501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Accepted: 01/17/2023] [Indexed: 06/18/2023]
Abstract
Significance Advanced digital control of microscopes and programmable data acquisition workflows have become increasingly important for improving the throughput and reproducibility of optical imaging experiments. Combinations of imaging modalities have enabled a more comprehensive understanding of tissue biology and tumor microenvironments in histopathological studies. However, insufficient imaging throughput and complicated workflows still limit the scalability of multimodal histopathology imaging. Aim We present a hardware-software co-design of a whole slide scanning system for high-throughput multimodal tissue imaging, including brightfield (BF) and laser scanning microscopy. Approach The system can automatically detect regions of interest using deep neural networks in a low-magnification rapid BF scan of the tissue slide and then conduct high-resolution BF scanning and laser scanning imaging on targeted regions with deep learning-based run-time denoising and resolution enhancement. The acquisition workflow is built using Pycro-Manager, a Python package that bridges hardware control libraries of the Java-based open-source microscopy software Micro-Manager in a Python environment. Results The system can achieve optimized imaging settings for both modalities with minimized human intervention and speed up the laser scanning by an order of magnitude with run-time image processing. Conclusions The system integrates the acquisition pipeline and data analysis pipeline into a single workflow that improves the throughput and reproducibility of multimodal histopathological imaging.
Collapse
Affiliation(s)
- Bin Li
- University of Wisconsin–Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
| | - Michael S. Nelson
- University of Wisconsin–Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
| | - Jenu V. Chacko
- University of Wisconsin–Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
| | - Nathan Cudworth
- University of Wisconsin–Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| | - Kevin W. Eliceiri
- University of Wisconsin–Madison, Center for Quantitative Cell Imaging, Madison, Wisconsin, United States
- University of Wisconsin–Madison, Department of Biomedical Engineering, Madison, Wisconsin, United States
- Morgridge Institute for Research, Madison, Wisconsin, United States
- University of Wisconsin–Madison, Department of Medical Physics, Madison, Wisconsin, United States
| |
Collapse
|
38
|
Lewis JE, Shebelut CW, Drumheller BR, Zhang X, Shanmugam N, Attieh M, Horwath MC, Khanna A, Smith GH, Gutman DA, Aljudi A, Cooper LAD, Jaye DL. An Automated Pipeline for Differential Cell Counts on Whole-Slide Bone Marrow Aspirate Smears. Mod Pathol 2023; 36:100003. [PMID: 36853796 PMCID: PMC10310355 DOI: 10.1016/j.modpat.2022.100003] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2022] [Revised: 08/10/2022] [Accepted: 09/18/2022] [Indexed: 01/11/2023]
Abstract
The pathologic diagnosis of bone marrow disorders relies in part on the microscopic analysis of bone marrow aspirate (BMA) smears and the manual counting of marrow nucleated cells to obtain a differential cell count (DCC). This manual process has significant limitations, including the analysis of only a small subset of optimal slide areas and nucleated cells, as well as interobserver variability due to differences in cell selection and classification. To address these shortcomings, we developed an automated machine learning-based pipeline for obtaining 11-component DCCs on whole-slide BMAs. This pipeline uses a sequential process of identifying optimal BMA regions with high proportions of marrow nucleated cells, detecting individual cells within these optimal areas, and classifying these cells into 1 of 11 DCC components. Convolutional neural network models were trained on 396,048 BMA region, 28,914 cell boundary, and 1,510,976 cell class images from manual annotations. The resulting automated pipeline produced 11-component DCCs that demonstrated a high statistical and diagnostic concordance with manual DCCs among a heterogeneous group of testing BMA slides with varying pathologies and cellularities. Additionally, we demonstrated that an automated analysis can reduce the intraslide variance in DCCs by analyzing the whole slide and marrow nucleated cells within all optimal regions. Finally, the pipeline outputs of region classification, cell detection, and cell classification can be visualized using whole-slide image analysis software. This study demonstrates the feasibility of a fully automated pipeline for generating DCCs on scanned whole-slide BMA images, with the potential for improving the current standard of practice for utilizing BMA smears in the laboratory analysis of hematologic disorders.
Collapse
Affiliation(s)
- Joshua E Lewis
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Conrad W Shebelut
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Bradley R Drumheller
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Xuebao Zhang
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Nithya Shanmugam
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Michel Attieh
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Michael C Horwath
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Anurag Khanna
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Geoffrey H Smith
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - David A Gutman
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia
| | - Ahmed Aljudi
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia
| | - Lee A D Cooper
- Department of Pathology, Northwestern University, Chicago, Illinois.
| | - David L Jaye
- Department of Pathology and Laboratory Medicine, Emory University, Atlanta, Georgia; Winship Cancer Institute, Emory University, Atlanta, Georgia.
| |
Collapse
|
39
|
Ginley B, Lucarelli N, Zee J, Jain S, Han SS, Rodrigues L, Wong ML, Jen KY, Sarder P. Automated Reference Kidney Histomorphometry using a Panoptic Segmentation Neural Network Correlates to Patient Demographics and Creatinine. PROCEEDINGS OF SPIE--THE INTERNATIONAL SOCIETY FOR OPTICAL ENGINEERING 2023; 12471:124711R. [PMID: 37818349 PMCID: PMC10563118 DOI: 10.1117/12.2655288] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2023]
Abstract
Reference histomorphometric data of healthy human kidneys are lacking due to laborious quantitation requirements. We leveraged deep learning to investigate the relationship of histomorphometry with patient age, sex, and serum creatinine in a multinational set of reference kidney tissue sections. A panoptic segmentation neural network was developed and used to segment viable and sclerotic glomeruli, cortical and medullary interstitia, tubules, and arteries/arterioles in digitized images of 79 periodic acid-Schiff (PAS)-stained human nephrectomy sections showing minimal pathologic changes. Simple morphometrics (e.g., area, radius, density) were measured from the segmented classes. Regression analysis was used to determine the relationship of histomorphometric parameters with age, sex, and serum creatinine. The model achieved high segmentation performance for all test compartments. We found that the size and density of nephrons, arteries/arterioles, and the baseline level of interstitium vary significantly among healthy humans, with potentially large differences between subjects from different geographic locations. Nephron size in any region of the kidney was significantly dependent on patient creatinine. Slight differences in renal vasculature and interstitium were observed between sexes. Finally, glomerulosclerosis percentage increased and cortical density of arteries/arterioles decreased as a function of age. We show that precise measurements of kidney histomorphometric parameters can be automated. Even in reference kidney tissue sections with minimal pathologic changes, several histomorphometric parameters demonstrated significant correlation to patient demographics and serum creatinine. These robust tools support the feasibility of deep learning to increase efficiency and rigor in histomorphometric analysis and pave the way for future large-scale studies.
Collapse
Affiliation(s)
- Brandon Ginley
- Department of Pathology and Anatomical Sciences, University at Buffalo – The State University of New York, Buffalo, NY, USA
| | - Nicholas Lucarelli
- Department of Biomedical Engineering, University of Florida, Gainesville, FL, USA
| | - Jarcy Zee
- Department of Biostatistics, Epidemiology, and Informatics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- Children’s Hospital of Philadelphia, Philadelphia, PA
| | - Sanjay Jain
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Luis Rodrigues
- University Clinic of Nephrology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
- Nephrology Unit, Centro Hospitalare Universitário de Coimbra, Coimbra, Portugal
| | - Michelle L. Wong
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, California, USA
| | - Kuang-yu Jen
- Department of Pathology and Laboratory Medicine, University of California, Davis School of Medicine, Sacramento, California, USA
| | - Pinaki Sarder
- Division of Nephrology, Hypertension, and Renal Transplantation, Department of Medicine, University of Florida, Gainesville, FL
| |
Collapse
|
40
|
Annotating for Artificial Intelligence Applications in Digital Pathology: A Practical Guide for Pathologists and Researchers. Mod Pathol 2023; 36:100086. [PMID: 36788085 DOI: 10.1016/j.modpat.2022.100086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/24/2022] [Accepted: 12/14/2022] [Indexed: 01/13/2023]
Abstract
Training machine learning models for artificial intelligence (AI) applications in pathology often requires extensive annotation by human experts, but there is little guidance on the subject. In this work, we aimed to describe our experience and provide a simple, useful, and practical guide addressing annotation strategies for AI development in computational pathology. Annotation methodology will vary significantly depending on the specific study's objectives, but common difficulties will be present across different settings. We summarize key aspects and issue guiding principles regarding team interaction, ground-truth quality assessment, different annotation types, and available software and hardware options and address common difficulties while annotating. This guide was specifically designed for pathology annotation, intending to help pathologists, other researchers, and AI developers with this process.
Collapse
|
41
|
Huang W, Tan K, Zhang Z, Hu J, Dong S. A Review of Fusion Methods for Omics and Imaging Data. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:74-93. [PMID: 35044920 DOI: 10.1109/tcbb.2022.3143900] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
The development of omics data and biomedical images has greatly advanced the progress of precision medicine in diagnosis, treatment, and prognosis. The fusion of omics and imaging data, i.e., omics-imaging fusion, offers a new strategy for understanding complex diseases. However, due to a variety of issues such as the limited number of samples, high dimensionality of features, and heterogeneity of different data types, efficiently learning complementary or associated discriminative fusion information from omics and imaging data remains a challenge. Recently, numerous machine learning methods have been proposed to alleviate these problems. In this review, from the perspective of fusion levels and fusion methods, we first provide an overview of preprocessing and feature extraction methods for omics and imaging data, and comprehensively analyze and summarize the basic forms and variations of commonly used and newly emerging fusion methods, along with their advantages, disadvantages and the applicable scope. We then describe public datasets and compare experimental results of various fusion methods on the ADNI and TCGA datasets. Finally, we discuss future prospects and highlight remaining challenges in the field.
Collapse
|
42
|
Sinha S, Sparks HD, Labit E, Robbins HN, Gowing K, Jaffer A, Kutluberk E, Arora R, Raredon MSB, Cao L, Swanson S, Jiang P, Hee O, Pope H, Workentine M, Todkar K, Sharma N, Bharadia S, Chockalingam K, de Almeida LGN, Adam M, Niklason L, Potter SS, Seifert AW, Dufour A, Gabriel V, Rosin NL, Stewart R, Muench G, McCorkell R, Matyas J, Biernaskie J. Fibroblast inflammatory priming determines regenerative versus fibrotic skin repair in reindeer. Cell 2022; 185:4717-4736.e25. [PMID: 36493752 PMCID: PMC9888357 DOI: 10.1016/j.cell.2022.11.004] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 08/24/2022] [Accepted: 11/02/2022] [Indexed: 12/13/2022]
Abstract
Adult mammalian skin wounds heal by forming fibrotic scars. We report that full-thickness injuries of reindeer antler skin (velvet) regenerate, whereas back skin forms fibrotic scar. Single-cell multi-omics reveal that uninjured velvet fibroblasts resemble human fetal fibroblasts, whereas back skin fibroblasts express inflammatory mediators mimicking pro-fibrotic adult human and rodent fibroblasts. Consequently, injury elicits site-specific immune responses: back skin fibroblasts amplify myeloid infiltration and maturation during repair, whereas velvet fibroblasts adopt an immunosuppressive phenotype that restricts leukocyte recruitment and hastens immune resolution. Ectopic transplantation of velvet to scar-forming back skin is initially regenerative, but progressively transitions to a fibrotic phenotype akin to the scarless fetal-to-scar-forming transition reported in humans. Skin regeneration is diminished by intensifying, or enhanced by neutralizing, these pathologic fibroblast-immune interactions. Reindeer represent a powerful comparative model for interrogating divergent wound healing outcomes, and our results nominate decoupling of fibroblast-immune interactions as a promising approach to mitigate scar.
Collapse
Affiliation(s)
- Sarthak Sinha
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Holly D Sparks
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Elodie Labit
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hayley N Robbins
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Kevin Gowing
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Arzina Jaffer
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Eren Kutluberk
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Rohit Arora
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Micha Sam Brickman Raredon
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics, Yale University, New Haven, CT, USA
| | - Leslie Cao
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Peng Jiang
- Morgridge Institute for Research, Madison, WI, USA
| | - Olivia Hee
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Hannah Pope
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Matt Workentine
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Kiran Todkar
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Nilesh Sharma
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Shyla Bharadia
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada
| | | | - Luiz G N de Almeida
- McCaig Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Mike Adam
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Laura Niklason
- Department of Biomedical Engineering, Yale University, New Haven, CT, USA; Vascular Biology and Therapeutics, Yale University, New Haven, CT, USA
| | - S Steven Potter
- Division of Developmental Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, USA
| | - Ashley W Seifert
- Department of Biology, University of Kentucky, Lexington, KY, USA
| | - Antoine Dufour
- McCaig Institute, University of Calgary, Calgary, AB, Canada; Department of Physiology and Pharmacology, University of Calgary, Calgary, AB, Canada
| | - Vincent Gabriel
- Department of Clinical Neurosciences, Cumming School of Medicine, University of Calgary, Calgary, AB, Canada; McCaig Institute, University of Calgary, Calgary, AB, Canada; Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada
| | - Nicole L Rosin
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Ron Stewart
- Morgridge Institute for Research, Madison, WI, USA
| | - Greg Muench
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - Robert McCorkell
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada
| | - John Matyas
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada; McCaig Institute, University of Calgary, Calgary, AB, Canada
| | - Jeff Biernaskie
- Faculty of Veterinary Medicine, University of Calgary, Calgary, AB, Canada; Department of Surgery, Cumming School of Medicine, University of Calgary, Calgary, AB, T2N 4N1, Canada; Hotchkiss Brain Institute, Calgary, AB, Canada; Alberta Children's Hospital Research Institute, Calgary, AB, Canada.
| |
Collapse
|
43
|
Pocock J, Graham S, Vu QD, Jahanifar M, Deshpande S, Hadjigeorghiou G, Shephard A, Bashir RMS, Bilal M, Lu W, Epstein D, Minhas F, Rajpoot NM, Raza SEA. TIAToolbox as an end-to-end library for advanced tissue image analytics. COMMUNICATIONS MEDICINE 2022; 2:120. [PMID: 36168445 PMCID: PMC9509319 DOI: 10.1038/s43856-022-00186-5] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 09/12/2022] [Indexed: 11/12/2022] Open
Abstract
Background Computational pathology has seen rapid growth in recent years, driven by advanced deep-learning algorithms. Due to the sheer size and complexity of multi-gigapixel whole-slide images, to the best of our knowledge, there is no open-source software library providing a generic end-to-end API for pathology image analysis using best practices. Most researchers have designed custom pipelines from the bottom up, restricting the development of advanced algorithms to specialist users. To help overcome this bottleneck, we present TIAToolbox, a Python toolbox designed to make computational pathology accessible to computational, biomedical, and clinical researchers. Methods By creating modular and configurable components, we enable the implementation of computational pathology algorithms in a way that is easy to use, flexible and extensible. We consider common sub-tasks including reading whole slide image data, patch extraction, stain normalization and augmentation, model inference, and visualization. For each of these steps, we provide a user-friendly application programming interface for commonly used methods and models. Results We demonstrate the use of the interface to construct a full computational pathology deep-learning pipeline. We show, with the help of examples, how state-of-the-art deep-learning algorithms can be reimplemented in a streamlined manner using our library with minimal effort. Conclusions We provide a usable and adaptable library with efficient, cutting-edge, and unit-tested tools for data loading, pre-processing, model inference, post-processing, and visualization. This enables a range of users to easily build upon recent deep-learning developments in the computational pathology literature.
Collapse
Affiliation(s)
- Johnathan Pocock
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - Simon Graham
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - Quoc Dang Vu
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | | | - Srijay Deshpande
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | | | - Adam Shephard
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | | | - Mohsin Bilal
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - Wenqi Lu
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - David Epstein
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - Fayyaz Minhas
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | - Nasir M. Rajpoot
- Tissue Image Analytics Centre, University of Warwick, Coventry, UK
| | | |
Collapse
|
44
|
McKenzie AT, Marx GA, Koenigsberg D, Sawyer M, Iida MA, Walker JM, Richardson TE, Campanella G, Attems J, McKee AC, Stein TD, Fuchs TJ, White CL, Farrell K, Crary JF. Interpretable deep learning of myelin histopathology in age-related cognitive impairment. Acta Neuropathol Commun 2022; 10:131. [PMID: 36127723 PMCID: PMC9490907 DOI: 10.1186/s40478-022-01425-5] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Accepted: 08/09/2022] [Indexed: 02/08/2023] Open
Abstract
Age-related cognitive impairment is multifactorial, with numerous underlying and frequently co-morbid pathological correlates. Amyloid beta (Aβ) plays a major role in Alzheimer's type age-related cognitive impairment, in addition to other etiopathologies such as Aβ-independent hyperphosphorylated tau, cerebrovascular disease, and myelin damage, which also warrant further investigation. Classical methods, even in the setting of the gold standard of postmortem brain assessment, involve semi-quantitative ordinal staging systems that often correlate poorly with clinical outcomes, due to imperfect cognitive measurements and preconceived notions regarding the neuropathologic features that should be chosen for study. Improved approaches are needed to identify histopathological changes correlated with cognition in an unbiased way. We used a weakly supervised multiple instance learning algorithm on whole slide images of human brain autopsy tissue sections from a group of elderly donors to predict the presence or absence of cognitive impairment (n = 367 with cognitive impairment, n = 349 without). Attention analysis allowed us to pinpoint the underlying subregional architecture and cellular features that the models used for the prediction in both brain regions studied, the medial temporal lobe and frontal cortex. Despite noisy labels of cognition, our trained models were able to predict the presence of cognitive impairment with a modest accuracy that was significantly greater than chance. Attention-based interpretation studies of the features most associated with cognitive impairment in the top performing models suggest that they identified myelin pallor in the white matter. Our results demonstrate a scalable platform with interpretable deep learning to identify unexpected aspects of pathology in cognitive impairment that can be translated to the study of other neurobiological disorders.
Collapse
Affiliation(s)
- Andrew T McKenzie
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Psychiatry, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gabriel A Marx
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Daniel Koenigsberg
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Mary Sawyer
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Megan A Iida
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jamie M Walker
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Timothy E Richardson
- Department of Pathology, University of Texas Health Science Center, San Antonio, TX, USA
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center, San Antonio, TX, USA
| | - Gabriele Campanella
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Johannes Attems
- Translation and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, NE4 5PL, UK
| | - Ann C McKee
- Department of Pathology, VA Medical Center &, Boston University School of Medicine, Boston, MA, USA
| | - Thor D Stein
- Department of Pathology, VA Medical Center &, Boston University School of Medicine, Boston, MA, USA
| | - Thomas J Fuchs
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Charles L White
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kurt Farrell
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Icahn Building 9th Floor, L9-02C, 1425 Madison Avenue, New York, NY, USA.
| | - John F Crary
- Departments of Pathology, Neuroscience, and Artificial Intelligence & Human Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Neuropathology Brain Bank & Research Core, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Ronald M. Loeb Center for Alzheimer's Disease, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Department of Pathology, Icahn School of Medicine at Mount Sinai, Icahn Building 9th Floor, Room 20A, 1425 Madison Avenue, New York, NY, 10029, USA.
| |
Collapse
|
45
|
Lutnick B, Manthey D, Becker JU, Ginley B, Moos K, Zuckerman JE, Rodrigues L, Gallan AJ, Barisoni L, Alpers CE, Wang XX, Myakala K, Jones BA, Levi M, Kopp JB, Yoshida T, Zee J, Han SS, Jain S, Rosenberg AZ, Jen KY, Sarder P. A user-friendly tool for cloud-based whole slide image segmentation with examples from renal histopathology. COMMUNICATIONS MEDICINE 2022; 2:105. [PMID: 35996627 PMCID: PMC9391340 DOI: 10.1038/s43856-022-00138-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 06/09/2022] [Indexed: 01/21/2023] Open
Abstract
BACKGROUND Image-based machine learning tools hold great promise for clinical applications in pathology research. However, the ideal end-users of these computational tools (e.g., pathologists and biological scientists) often lack the programming experience required for the setup and use of these tools which often rely on the use of command line interfaces. METHODS We have developed Histo-Cloud, a tool for segmentation of whole slide images (WSIs) that has an easy-to-use graphical user interface. This tool runs a state-of-the-art convolutional neural network (CNN) for segmentation of WSIs in the cloud and allows the extraction of features from segmented regions for further analysis. RESULTS By segmenting glomeruli, interstitial fibrosis and tubular atrophy, and vascular structures from renal and non-renal WSIs, we demonstrate the scalability, best practices for transfer learning, and effects of dataset variability. Finally, we demonstrate an application for animal model research, analyzing glomerular features in three murine models. CONCLUSIONS Histo-Cloud is open source, accessible over the internet, and adaptable for segmentation of any histological structure regardless of stain.
Collapse
Affiliation(s)
- Brendon Lutnick
- Department of Pathology and Anatomical Sciences, SUNY Buffalo, Buffalo, USA
| | | | - Jan U. Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Brandon Ginley
- Department of Pathology and Anatomical Sciences, SUNY Buffalo, Buffalo, USA
| | - Katharina Moos
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jonathan E. Zuckerman
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, USA
| | - Luis Rodrigues
- University Clinic of Nephrology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - Laura Barisoni
- Departments of Pathology and Medicine, Duke University, Durham, USA
| | - Charles E. Alpers
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, USA
| | - Xiaoxin X. Wang
- Departments of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC USA
| | - Komuraiah Myakala
- Departments of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC USA
| | - Bryce A. Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, DC USA
| | - Moshe Levi
- Departments of Biochemistry and Molecular & Cellular Biology, Georgetown University, Washington, DC USA
| | | | | | - Jarcy Zee
- Department of Biostatistics, Epidemiology, & Informatics, University of Pennsylvania, Philadelphia, USA
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, South Korea
| | - Sanjay Jain
- Department of Medicine, Nephrology, Washington University School of Medicine, St. Louis, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University, Baltimore, USA
| | - Kuang Yu. Jen
- Department of Pathology and Laboratory Medicine, University of California at Davis, Sacramento, USA
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, SUNY Buffalo, Buffalo, USA
| |
Collapse
|
46
|
Wu Y, Koyuncu CF, Toro P, Corredor G, Feng Q, Buzzy C, Old M, Teknos T, Connelly ST, Jordan RC, Lang Kuhs KA, Lu C, Lewis JS, Madabhushi A. A machine learning model for separating epithelial and stromal regions in oral cavity squamous cell carcinomas using H&E-stained histology images: A multi-center, retrospective study. Oral Oncol 2022; 131:105942. [PMID: 35689952 DOI: 10.1016/j.oraloncology.2022.105942] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 04/12/2022] [Accepted: 05/24/2022] [Indexed: 01/30/2023]
Abstract
OBJECTIVE Tissue slides from Oral cavity squamous cell carcinoma (OC-SCC), particularly the epithelial regions, hold morphologic features that are both diagnostic and prognostic. Yet, previously developed approaches for automated epithelium segmentation in OC-SCC have not been independently tested in a multi-center setting. In this study, we aimed to investigate the effectiveness and applicability of a convolutional neural network (CNN) model to perform epithelial segmentation using digitized H&E-stained diagnostic slides from OC-SCC patients in a multi-center setting. METHODS A CNN model was developed to segment the epithelial regions of digitized slides (n = 810), retrospectively collected from five different centers. Deep learning models were trained and validated using well-annotated tissue microarray (TMA) images (n = 212) at various magnifications. The best performing model was locked down and used for independent testing with a total of 478 whole-slide images (WSIs). Manually annotated epithelial regions were used as the reference standard for evaluation. We also compared the model generated results with IHC-stained epithelium (n = 120) as the reference. RESULTS The locked-down CNN model trained on the TMA image training cohorts with 10x magnification achieved the best segmentation performance. The locked-down model performed consistently and yielded Pixel Accuracy, Recall Rate, Precision Rate, and Dice Coefficient that ranged from 95.8% to 96.6%, 79.1% to 93.8%, 85.7% to 89.3%, and 82.3% to 89.0%, respectively for the three independent testing WSI cohorts. CONCLUSION The automated model achieved a consistently accurate performance for automated epithelial region segmentation compared to manual annotations. This model could be integrated into a computer-aided diagnosis or prognosis system.
Collapse
Affiliation(s)
- Yuxin Wu
- Shandong Junteng Medical Technology Co., Ltd, Jinan, China; College of Computer Science, Shaanxi Normal University, Xian, China
| | - Can F Koyuncu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Paula Toro
- Department of Pathology, Cleveland Clinic, OH, USA
| | - German Corredor
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA
| | - Qianyu Feng
- College of Computer Science, Shaanxi Normal University, Xian, China
| | - Christina Buzzy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Matthew Old
- Department of Otolaryngology, Ohio State University Medical Center, OH, USA
| | - Theodoros Teknos
- Department of Otolaryngology, University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | - Stephen Thaddeus Connelly
- Department of Oral and Maxillofacial Surgery, San Francisco Veterans Affairs Health Care System, University of California, San Francisco, San Francisco, CA, USA
| | - Richard C Jordan
- Departments of Orofacial Sciences, Pathology and Radiation Oncology, University of California San Francisco, CA, USA
| | - Krystle A Lang Kuhs
- Department of Epidemiology, College of Public Health, University of Kentucky, Lexington, KY, USA; Department of Medicine, Vanderbilt University Medical Cancer, Nashville, TN, USA
| | - Cheng Lu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA.
| | - James S Lewis
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA; Department of Otolaryngology - Head and Neck Surgery, Vanderbilt University Medical Center, Nashville, TN, USA.
| | - Anant Madabhushi
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA; Louis Stokes Cleveland VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
47
|
Saednia K, Lagree A, Alera MA, Fleshner L, Shiner A, Law E, Law B, Dodington DW, Lu FI, Tran WT, Sadeghi-Naini A. Quantitative digital histopathology and machine learning to predict pathological complete response to chemotherapy in breast cancer patients using pre-treatment tumor biopsies. Sci Rep 2022; 12:9690. [PMID: 35690630 PMCID: PMC9188550 DOI: 10.1038/s41598-022-13917-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2022] [Accepted: 05/30/2022] [Indexed: 11/17/2022] Open
Abstract
Complete pathological response (pCR) to neoadjuvant chemotherapy (NAC) is a prognostic factor for breast cancer (BC) patients and is correlated with improved survival. However, pCR rates are variable to standard NAC, depending on BC subtype. This study investigates quantitative digital histopathology coupled with machine learning (ML) to predict NAC response a priori. Clinicopathologic data and digitized slides of BC core needle biopsies were collected from 149 patients treated with NAC. The nuclei within the tumor regions were segmented on the histology images of biopsy samples using a weighted U-Net model. Five pathomic feature subsets were extracted from segmented digitized samples, including the morphological, intensity-based, texture, graph-based and wavelet features. Seven ML experiments were conducted with different feature sets to develop a prediction model of therapy response using a gradient boosting machine with decision trees. The models were trained and optimized using a five-fold cross validation on the training data and evaluated using an unseen independent test set. The prediction model developed with the best clinical features (tumor size, tumor grade, age, and ER, PR, HER2 status) demonstrated an area under the ROC curve (AUC) of 0.73. Various pathomic feature subsets resulted in models with AUCs in the range of 0.67 and 0.87, with the best results associated with the graph-based and wavelet features. The selected features among all subsets of the pathomic and clinicopathologic features included four wavelet and three graph-based features and no clinical features. The predictive model developed with these features outperformed the other models, with an AUC of 0.90, a sensitivity of 85% and a specificity of 82% on the independent test set. The results demonstrated the potential of quantitative digital histopathology features integrated with ML methods in predicting BC response to NAC. This study is a step forward towards precision oncology for BC patients to potentially guide future therapies.
Collapse
Affiliation(s)
- Khadijeh Saednia
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Andrew Lagree
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Marie A Alera
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Lauren Fleshner
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Audrey Shiner
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Ethan Law
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - Brianna Law
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
| | - David W Dodington
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Fang-I Lu
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - William T Tran
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada
- Department of Radiation Oncology, University of Toronto, Toronto, ON, Canada
- Temerity Centre for AI Research and Education in Medicine, University of Toronto, Toronto, ON, Canada
| | - Ali Sadeghi-Naini
- Department of Electrical Engineering and Computer Science, Lassonde School of Engineering, York University, Toronto, ON, Canada.
- Department of Radiation Oncology, Sunnybrook Health Sciences Center, Toronto, ON, Canada.
- Temerity Centre for AI Research and Education in Medicine, University of Toronto, Toronto, ON, Canada.
- Physical Sciences Platform, Sunnybrook Research Institute, Toronto, ON, Canada.
| |
Collapse
|
48
|
Escobar Díaz Guerrero R, Carvalho L, Bocklitz T, Popp J, Oliveira JL. Software tools and platforms in Digital Pathology: a review for clinicians and computer scientists. J Pathol Inform 2022; 13:100103. [PMID: 36268075 PMCID: PMC9576980 DOI: 10.1016/j.jpi.2022.100103] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Revised: 05/12/2022] [Accepted: 05/17/2022] [Indexed: 11/20/2022] Open
Abstract
At the end of the twentieth century, a new technology was developed that allowed an entire tissue section to be scanned on an objective slide. Originally called virtual microscopy, this technology is now known as Whole Slide Imaging (WSI). WSI presents new challenges for reading, visualization, storage, and analysis. For this reason, several technologies have been developed to facilitate the handling of these images. In this paper, we analyze the most widely used technologies in the field of digital pathology, ranging from specialized libraries for the reading of these images to complete platforms that allow reading, visualization, and analysis. Our aim is to provide the reader, whether a pathologist or a computational scientist, with the knowledge to choose the technologies to use for new studies, development, or research.
Collapse
Affiliation(s)
- Rodrigo Escobar Díaz Guerrero
- BMD Software, PCI - Creative Science Park, 3830-352 Ilhavo, Portugal
- DETI/IEETA, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Lina Carvalho
- Institute of Anatomical and Molecular Pathology, Faculty of Medicine, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Thomas Bocklitz
- Leibniz Institute of Photonic Technology Jena, Member of Leibniz research alliance ‘Health technologies’, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Friedrich-Schiller-University, Jena, Germany
| | - Juergen Popp
- Leibniz Institute of Photonic Technology Jena, Member of Leibniz research alliance ‘Health technologies’, Albert-Einstein-Straße 9, 07745 Jena, Germany
- Institute of Physical Chemistry and Abbe Center of Photonics (IPC), Friedrich-Schiller-University, Jena, Germany
| | | |
Collapse
|
49
|
Santo BA, Govind D, Daneshpajouhnejad P, Yang X, Wang XX, Myakala K, Jones BA, Levi M, Kopp JB, Yoshida T, Niedernhofer LJ, Manthey D, Moon KC, Han SS, Zee J, Rosenberg AZ, Sarder P. PodoCount: A Robust, Fully Automated, Whole-Slide Podocyte Quantification Tool. Kidney Int Rep 2022; 7:1377-1392. [PMID: 35694561 PMCID: PMC9174049 DOI: 10.1016/j.ekir.2022.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 02/25/2022] [Accepted: 03/04/2022] [Indexed: 12/13/2022] Open
Abstract
Introduction Podocyte depletion is a histomorphologic indicator of glomerular injury and predicts clinical outcomes. Podocyte estimation methods or podometrics are semiquantitative, technically involved, and laborious. Implementation of high-throughput podometrics in experimental and clinical workflows necessitates an automated podometrics pipeline. Recognizing that computational image analysis offers a robust approach to study cell and tissue structure, we developed and validated PodoCount (a computational tool for automated podocyte quantification in immunohistochemically labeled tissues) using a diverse data set. Methods Whole-slide images (WSIs) of tissues immunostained with a podocyte nuclear marker and periodic acid–Schiff counterstain were acquired. The data set consisted of murine whole kidney sections (n = 135) from 6 disease models and human kidney biopsy specimens from patients with diabetic nephropathy (DN) (n = 45). Within segmented glomeruli, podocytes were extracted and image analysis was applied to compute measures of podocyte depletion and nuclear morphometry. Computational performance evaluation and statistical testing were performed to validate podometric and associated image features. PodoCount was disbursed as an open-source, cloud-based computational tool. Results PodoCount produced highly accurate podocyte quantification when benchmarked against existing methods. Podocyte nuclear profiles were identified with 0.98 accuracy and segmented with 0.85 sensitivity and 0.99 specificity. Errors in podocyte count were bounded by 1 podocyte per glomerulus. Podocyte-specific image features were found to be significant predictors of disease state, proteinuria, and clinical outcome. Conclusion PodoCount offers high-performance podocyte quantitation in diverse murine disease models and in human kidney biopsy specimens. Resultant features offer significant correlation with associated metadata and outcome. Our cloud-based tool will provide end users with a standardized approach for automated podometrics from gigapixel-sized WSIs.
Collapse
Affiliation(s)
- Briana A. Santo
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York, USA
| | - Darshana Govind
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York, USA
| | | | - Xiaoping Yang
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
| | - Xiaoxin X. Wang
- Department of Biochemistry, Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| | - Komuraiah Myakala
- Department of Biochemistry, Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| | - Bryce A. Jones
- Department of Pharmacology and Physiology, Georgetown University, Washington, District of Columbia, USA
| | - Moshe Levi
- Department of Biochemistry, Molecular & Cellular Biology, Georgetown University, Washington, District of Columbia, USA
| | - Jeffrey B. Kopp
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Teruhiko Yoshida
- Kidney Disease Section, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, USA
| | - Laura J. Niedernhofer
- Institute on the Biology of Aging and Metabolism, Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, Minnesota, USA
| | | | - Kyung Chul Moon
- Department of Pathology, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Seung Seok Han
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Republic of Korea
| | - Jarcy Zee
- Department of Biostatistics, Epidemiology and Informatics, University of Pennsylvania, Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Avi Z. Rosenberg
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, USA
- Correspondence: Avi Z. Rosenberg, Department of Pathology, Johns Hopkins University School of Medicine, 720 Rutland Avenue, Ross Building, Room 632D, Johns Hopkins Medical Institutions, Baltimore, Maryland 21205, USA.
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, University at Buffalo, Buffalo, New York, USA
- Pinaki Sarder, Department of Pathology and Anatomical Sciences, University at Buffalo, 955 Main Street, Room 4204, Buffalo, New York 14203, USA.
| |
Collapse
|
50
|
Lutnick B, Manthey D, Becker JU, Zuckerman JE, Rodrigues L, Jen KY, Sarder P. A tool for federated training of segmentation models on whole slide images. J Pathol Inform 2022; 13:100101. [PMID: 35910077 PMCID: PMC9326476 DOI: 10.1016/j.jpi.2022.100101] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2022] [Revised: 04/06/2022] [Accepted: 04/09/2022] [Indexed: 11/22/2022] Open
Abstract
The largest bottleneck to the development of convolutional neural network (CNN) models in the computational pathology domain is the collection and curation of diverse training datasets. Training CNNs requires large cohorts of image data, and model generalizability is dependent on training data heterogeneity. Including data from multiple centers enhances the generalizability of CNN-based models, but this is hindered by the logistical challenges of sharing medical data. In this paper, we explore the feasibility of training our recently developed cloud-based segmentation tool (Histo-Cloud) using federated learning. Using a dataset of renal tissue biopsies we show that federated training to segment interstitial fibrosis and tubular atrophy (IFTA) using datasets from three institutions is not found to be different from a training by pooling the data on one server when tested on a fourth (holdout) institution's data. Further, training a model to segment glomeruli for a federated dataset (split by staining) demonstrates similar performance.
Collapse
Affiliation(s)
- Brendon Lutnick
- Department of Pathology and Anatomical Sciences, SUNY Buffalo, Buffalo, NY, USA
| | | | - Jan U. Becker
- Institute of Pathology, University Hospital Cologne, Cologne, Germany
| | - Jonathan E. Zuckerman
- Department of Pathology and Laboratory Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| | - Luis Rodrigues
- University Clinic of Nephrology, Faculty of Medicine, University of Coimbra, Portugal
| | - Kuang-Yu Jen
- University of California, Davis School of Medicine, Sacramento, CA, USA
| | - Pinaki Sarder
- Department of Pathology and Anatomical Sciences, SUNY Buffalo, Buffalo, NY, USA
| |
Collapse
|