1
|
Zhang H, Wang P, Liu J, Qin J. pFedBCC: Personalizing Federated multi-target domain adaptive segmentation via Bi-pole Collaborative Calibration. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2025; 263:108635. [PMID: 39956050 DOI: 10.1016/j.cmpb.2025.108635] [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: 10/22/2024] [Revised: 01/19/2025] [Accepted: 02/01/2025] [Indexed: 02/18/2025]
Abstract
BACKGROUND AND OBJECTIVE Multi-target domain adaptation (MTDA) is a well-established technology for unsupervised segmentation. It can significantly reduce the workload of large-scale data annotations, but assumes that each domain data can be freely accessed. However, data privacy limit its deployment in real-world medical scenes. Aiming at this problem, federated learning (FL) commits a paradigm to handle private cross-institution data. METHODS This paper makes the first attempt to apply FedMTDA to medical image segmentation by proposing a personalized Federated Bi-pole Collaborative Calibration (pFedBCC) framework, which leverages unannotated private client data and a public source-domain model to learn a global model at the central server for unsupervised multi-type immunohistochemically (IHC) image segmentation. Concretely, pFedBCC tackles two significant challenges in FedMTDA including client-side prediction drift and server-side aggregation drift via Semantic-affinity-driven Personalized Label Calibration (SPLC) and Source-knowledge-oriented Consistent Gradient Calibration (SCGC). To alleviate local prediction drift, SPLC personalizes a cross-domain graph reasoning module for each client, which achieves semantic-affinity alignment between high-level source- and target-domain features to produce pseudo labels that are semantically consistent with source-domain labels to guide client training. To further alleviate global aggregation drift, SCGC develops a new conflict-gradient clipping scheme, which takes the source-domain gradient as a guidance to ensure that all clients update with similar gradient directions and magnitudes, thereby improving the generalization of the global model. RESULTS pFedBCC is evaluated on private and public IHC benchmarks, including the proposed MT-IHC dataset, and the panCK, BCData, DLBC-Morph and LYON19 datasets. Overall, pFedBCC achieves the best performance of 88.8% PA on MT-IHC, as well as 88.4% PA on the LYON19 dataset, respectively. CONCLUSIONS The proposed pFedBCC performs better than all comparison methods. The ablation study also confirms the contribution of SPLC and SCGC for unsupervised multi-type IHC image segmentation. This paper constructs a MT-IHC dataset containing more than 19,000 IHC images of 10 types (CgA, CK, Syn, CD, Ki67, P40, P53, EMA, TdT and BCL). Extensive experiments on the MT-IHC and public IHC datasets confirm that pFedBCC outperforms existing FL and DA methods.
Collapse
Affiliation(s)
- Huaqi Zhang
- Department of Information Management, The National Police University for Criminal Justice, Baoding Hebei, China
| | - Pengyu Wang
- Sports Artificial Intelligence Institute, Capital University of Physical Education and Sports, Beijing, China.
| | - Jie Liu
- School of Computer and Information Technology, Beijing Jiaotong University, Beijing, China
| | - Jing Qin
- School of Nursing, The Hong Kong Polytechnic University, Hong Kong Special Administrative Region of China
| |
Collapse
|
2
|
Beltzung F, Le VL, Molnar I, Boutault E, Darcha C, Le Loarer F, Kossai M, Saut O, Biau J, Penault-Llorca F, Chautard E. Leveraging Deep Learning for Immune Cell Quantification and Prognostic Evaluation in Radiotherapy-Treated Oropharyngeal Squamous Cell Carcinomas. J Transl Med 2025; 105:104094. [PMID: 39826685 DOI: 10.1016/j.labinv.2025.104094] [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: 07/27/2024] [Revised: 12/24/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025] Open
Abstract
The tumor microenvironment plays a critical role in cancer progression and therapeutic responsiveness, with the tumor immune microenvironment (TIME) being a key modulator. In head and neck squamous cell carcinomas (HNSCCs), immune cell infiltration significantly influences the response to radiotherapy (RT). A better understanding of the TIME in HNSCCs could help identify patients most likely to benefit from combining RT with immunotherapy. Standardized, cost-effective methods for studying TIME in HNSCCs are currently lacking. This study aims to leverage deep learning (DL) to quantify immune cell densities using immunohistochemistry in untreated oropharyngeal squamous cell carcinoma (OPSCC) biopsies of patients scheduled for curative RT and assess their prognostic value. We analyzed 84 pretreatment formalin-fixed paraffin-embedded tumor biopsies from OPSCC patients. Immunohistochemistry was performed for CD3, CD8, CD20, CD163, and FOXP3, and whole slide images were digitized for analysis using a U-Net-based DL model. Two quantification approaches were applied: a cell-counting method and an area-based method. These methods were applied to stained regions. The DL model achieved high accuracy in detecting stained cells across all biomarkers. Strong correlations were found between our DL pipeline, the HALO Image Analysis Platform, and the open-source QuPath software for estimating immune cell densities. Our DL pipeline provided an accurate and reproducible approach for quantifying immune cells in OPSCC. The area-based method demonstrated superior prognostic value for recurrence-free survival, when compared with the cell-counting method. Elevated densities of CD3, CD8, CD20, and FOXP3 were associated with improved recurrence-free survival, whereas CD163 showed no significant prognostic association. These results highlight the potential of DL in digital pathology for assessing TIME and predicting patient outcomes.
Collapse
Affiliation(s)
- Fanny Beltzung
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Hôpital Haut-Lévêque, CHU de Bordeaux, Pessac, France.
| | - Van-Linh Le
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France; Department of Data and Digital Health, Bergonié Institute, Bordeaux, France
| | - Ioana Molnar
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Clinical Research Division, Clinical Research & Innovation Division, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Erwan Boutault
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France
| | - Claude Darcha
- Department of Pathology, CHU Clermont-Ferrand, Clermont-Ferrand, France
| | - François Le Loarer
- Department of Pathology, Bergonié Institute, Bordeaux, France; Bordeaux Institute of Oncology (BRIC U1312), INSERM, Université de Bordeaux, Institut Bergonié, Bordeaux, France
| | - Myriam Kossai
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Olivier Saut
- MONC team, Center INRIA at University of Bordeaux, Talence, France; Bordeaux Mathematics Institute (IMB), UMR CNRS 5251, University of Bordeaux, Talence, France
| | - Julian Biau
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Radiation Therapy, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Frédérique Penault-Llorca
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| | - Emmanuel Chautard
- Department of Molecular Imaging & Theragnostic Strategies (IMOST), University Clermont Auvergne, INSERM U1240, Clermont-Ferrand, France; Department of Pathology, Centre Jean PERRIN, Clermont-Ferrand, France
| |
Collapse
|
3
|
Hussain I, Boza J, Lukande R, Ayanga R, Semeere A, Cesarman E, Martin J, Maurer T, Erickson D. Automated Detection of Kaposi Sarcoma-Associated Herpesvirus-Infected Cells in Immunohistochemical Images of Skin Biopsies. JCO Glob Oncol 2025; 11:e2400536. [PMID: 40239145 DOI: 10.1200/go-24-00536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 01/11/2025] [Accepted: 02/18/2025] [Indexed: 04/18/2025] Open
Abstract
PURPOSE Immunohistochemical staining for the antigen of Kaposi sarcoma (KS)-associated herpesvirus, latency-associated nuclear antigen (LANA), is helpful in diagnosing KS. A challenge lies in distinguishing anti-LANA-positive cells from morphologically similar brown counterparts. This work aims to develop an automated framework for localization and quantification of LANA positivity in whole-slide images (WSI) of skin biopsies. METHODS The proposed framework leverages weakly supervised multiple-instance learning (MIL) to reduce false-positive predictions. A novel morphology-based slide aggregation method is introduced to improve accuracy. The framework generates interpretable heatmaps for cell localization and provides quantitative values for the percentage of positive tiles. The framework was trained and tested with a KS pathology data set prepared from skin biopsies of KS-suspected patients in Uganda. RESULTS The developed MIL framework achieved an area under the receiver operating characteristic curve of 0.99, with a sensitivity of 98.15% and specificity of 96.00% in predicting anti-LANA-positive WSIs in a test data set. CONCLUSION The framework shows promise for the automated detection of LANA in skin biopsies, offering a reliable and accurate tool for identifying anti-LANA-positive cells. This method may be especially impactful in resource-limited areas that lack trained pathologists, potentially improving diagnostic capabilities in settings with limited access to expert analysis.
Collapse
Affiliation(s)
- Iftak Hussain
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
| | - Juan Boza
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| | - Robert Lukande
- Department of Pathology, Makerere University College of Health Sciences, Kampala, Uganda
| | - Racheal Ayanga
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Aggrey Semeere
- Infectious Diseases Institute, Makerere University College of Health Sciences, Kampala, Uganda
| | - Ethel Cesarman
- Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, NY
| | - Jeffrey Martin
- Department of Epidemiology and Biostatistics, University of California, San Francisco, CA
| | - Toby Maurer
- Department of Dermatology, Indiana University School of Medicine, Indianapolis, IN
| | - David Erickson
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, NY
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY
| |
Collapse
|
4
|
Zhang H, Chen L, Li L, Liu Y, Das B, Zhai S, Tan J, Jiang Y, Turco S, Yao Y, Frishman D. Prediction and analysis of tumor infiltrating lymphocytes across 28 cancers by TILScout using deep learning. NPJ Precis Oncol 2025; 9:76. [PMID: 40108446 PMCID: PMC11923303 DOI: 10.1038/s41698-025-00866-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 03/06/2025] [Indexed: 03/22/2025] Open
Abstract
The density of tumor-infiltrating lymphocytes (TILs) serves as a valuable indicator for predicting anti-tumor responses, but its broad impact across various types of cancers remains underexplored. We introduce TILScout, a pan-cancer deep-learning approach to compute patch-level TIL scores from whole slide images (WSIs). TILScout achieved accuracies of 0.9787 and 0.9628, and AUCs of 0.9988 and 0.9934 in classifying WSI patches into three categories-TIL-positive, TIL-negative, and other/necrotic-on validation and independent test sets, respectively, surpassing previous studies. The biological significance of TILScout-derived TIL scores across 28 cancers was validated through comprehensive functional and correlational analyses. A consistent decrease in TIL scores with an increase in cancer stage provides direct evidence that the lower TIL content may stimulate cancer progression. Additionally, TIL scores correlated with immune checkpoint gene expression and genomic variation in common cancer driver genes. Our comprehensive pan-cancer survey highlights the critical prognostic significance of TILs within the tumor microenvironment.
Collapse
Affiliation(s)
- Huibo Zhang
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lulu Chen
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Lan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yang Liu
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Barnali Das
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Shuang Zhai
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yan Jiang
- Department of Pathology, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Simona Turco
- Electrical Engineering, Eindhoven University of Technology, Den Dolech 12, Eindhoven, 5612AZ, the Netherlands
| | - Yi Yao
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, China.
| | - Dmitrij Frishman
- Department of Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
5
|
García-Barberán V, Gómez Del Pulgar ME, Guamán HM, Benito-Martin A. The times they are AI-changing: AI-powered advances in the application of extracellular vesicles to liquid biopsy in breast cancer. EXTRACELLULAR VESICLES AND CIRCULATING NUCLEIC ACIDS 2025; 6:128-140. [PMID: 40206803 PMCID: PMC11977355 DOI: 10.20517/evcna.2024.51] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 01/03/2025] [Accepted: 01/25/2025] [Indexed: 04/11/2025]
Abstract
Artificial intelligence (AI) is revolutionizing scientific research by facilitating a paradigm shift in data analysis and discovery. This transformation is characterized by a fundamental change in scientific methods and concepts due to AI's ability to process vast datasets with unprecedented speed and accuracy. In breast cancer research, AI aids in early detection, prognosis, and personalized treatment strategies. Liquid biopsy, a noninvasive tool for detecting circulating tumor traits, could ideally benefit from AI's analytical capabilities, enhancing the detection of minimal residual disease and improving treatment monitoring. Extracellular vesicles (EVs), which are key elements in cell communication and cancer progression, could be analyzed with AI to identify disease-specific biomarkers. AI combined with EV analysis promises an enhancement in diagnosis precision, aiding in early detection and treatment monitoring. Studies show that AI can differentiate cancer types and predict drug efficacy, exemplifying its potential in personalized medicine. Overall, the integration of AI in biomedical research and clinical practice promises significant changes and advancements in diagnostics, personalized medicine-based approaches, and our understanding of complex diseases like cancer.
Collapse
Affiliation(s)
- Vanesa García-Barberán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - María Elena Gómez Del Pulgar
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Heidy M. Guamán
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
| | - Alberto Benito-Martin
- Molecular Oncology Laboratory, Medical Oncology Department, Hospital Clínico Universitario San Carlos, Instituto de Investigación Sanitaria San Carlos (IdISSC), Madrid 28040, Spain
- Facultad de Medicina, Universidad Alfonso X el Sabio, Madrid 28691, Spain
| |
Collapse
|
6
|
Chen Z, Xie T, Chen S, Li Z, Yao S, Lu X, He W, Tang C, Yang D, Li S, Shi F, Lin H, Li Z, Madabhushi A, Zhao X, Liu Z, Lu C. AI-based tumor-infiltrating lymphocyte scoring system for assessing HCC prognosis in patients undergoing liver resection. JHEP Rep 2025; 7:101270. [PMID: 39927235 PMCID: PMC11803844 DOI: 10.1016/j.jhepr.2024.101270] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Revised: 10/22/2024] [Accepted: 11/04/2024] [Indexed: 02/11/2025] Open
Abstract
Background & Aims Tumor-infiltrating lymphocytes (TILs), particularly CD8+ TILs, are key prognostic markers in many cancers. However, their prognostic value in hepatocellular carcinoma (HCC) remains controversial, with different evidence. Given the heterogeneous outcomes in patients with HCC undergoing liver resection, this study aims to develop an AI-based system to quantify CD8+ TILs and assess their prognostic value for patients with HCC. Methods We conducted a retrospective multicenter study on patients undergoing liver resection across three cohorts (N = 514). We trained a deep neural network and a random forest model to segment tumor regions and locate CD8+ TILs in H&E and CD8-stained whole-slide images. We quantified CD8+ TIL density and established an Automated CD8+ Tumor-infiltrating Lymphocyte Scoring (ATLS-8) system to assess its prognostic value. Results In the discovery cohort, the 5-year overall survival (OS) rates were 34.05% for ATLS-8 low-score and 65.03% for ATLS-8 high-score groups (hazard ratio [HR] 2.40; 95% CI, 1.37-4.19; p = 0.015). These findings were confirmed in validation cohort 1, which had 5-year OS rates of 28.57% and 68.73% (HR 3.38; 95% CI, 1.27-9.02; p = 0.0098), and validation cohort 2, which had 59.26% and 81.48% (HR 2.74; 95% CI, 1.05-7.15; p = 0.031). ATLS-8 improved the prognostic model based on clinical variables (C-index 0.770 vs. 0.757; 0.769 vs. 0.727; 0.712 vs. 0.642 in three cohorts). Conclusions We developed an automated system using CD8-stained whole-slide images to assess immune infiltration (ATLS-8). In patients with HCC undergoing resection, higher CD8+ TIL density correlates with better OS, as per ATLS-8 assessment. This system is a promising tool for advancing clinical immune microenvironment assessment and outcome prediction. Impact and implications CD8+ tumor-infiltrating lymphocytes (TILs) have been identified as a prognostic factor associated with many cancers. In this study, CD8+ TILs were identified as an independent prognostic factor for overall survival in patients with hepatocellular carcinoma who undergoing liver resection. Therefore, ATLS-8, a novel digital biomarker based on whole-slide image-level CD8+ TILs, could play an important role in the prognostic assessment of patients with HCC and could be integrated into clinicopathological models to participate in the decision-making and prognostic assessment of patients. The scoring system combined with artificial intelligence is essential for automated, quantitative, whole-slide image-level assessment of TILs, which can be widely applied to quantify the immune profile of multi-cancer disease types with the discussion of subsequent immunotherapy.
Collapse
Affiliation(s)
- Zhiyang Chen
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- School of Clinical Dentistry, University of Sheffield, Sheffield, UK
| | - Tingting Xie
- Medical Imaging Center, Peking University Shenzhen Hospital, Shenzhen, Guangdong, China
| | - Shuting Chen
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, Yunnan, China
| | - Su Yao
- Department of Pathology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Xuanjun Lu
- School of Electronics Engineering, Xi’an Shiyou University, Xi’an, China
| | - Wenfeng He
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Chao Tang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- College of Information Engineering, Northwest A&F University, Yangling, Shaanxi, China
| | - Dacheng Yang
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou China
| | - Shaohua Li
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Guangzhou, China
| | - Feng Shi
- Department of Interventional Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, China
| | - Huan Lin
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zipei Li
- School of Computer Science, University of St Andrews, Fife, UK
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Radiology and Imaging Sciences, Biomedical Informatics (BMI) and Pathology, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Atlanta Veterans Administration Medical Center, Atlanta, GA, USA
| | - Xiangtian Zhao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
| | - Cheng Lu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, China
- Medical Research Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou China
| |
Collapse
|
7
|
Caputo A, Maffei E, Gupta N, Cima L, Merolla F, Cazzaniga G, Pepe P, Verze P, Fraggetta F. Computer-assisted diagnosis to improve diagnostic pathology: A review. INDIAN J PATHOL MICR 2025; 68:3-10. [PMID: 40162930 DOI: 10.4103/ijpm.ijpm_339_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 02/17/2025] [Indexed: 04/02/2025] Open
Abstract
ABSTRACT With an increasing demand for accuracy and efficiency in diagnostic pathology, computer-assisted diagnosis (CAD) emerges as a prominent and transformative solution. This review aims to explore the practical applications, implications, strengths, and weaknesses of CAD applied to diagnostic pathology. A comprehensive literature search was conducted to include English-language studies focusing on CAD tools, digital pathology, and Artificial intelligence (AI) applications in pathology. The review underscores the transformative potential of CAD tools in pathology, particularly in streamlining diagnostic processes, reducing turnaround times, and augmenting diagnostic accuracy. It emphasizes the strides made in digital pathology, the integration of AI, and the promising prospects for prognostic biomarker discovery using computational methods. Additionally, ethical considerations regarding data privacy, equity, and trust in AI deployment are examined. CAD has the potential to revolutionize diagnostic pathology. The insights gleaned from this review offer a panoramic view of recent advancements. Ultimately, this review aims to guide future research, influence clinical practice, and inform policy-making by elucidating the promising horizons and potential pitfalls of integrating CAD tools in pathology.
Collapse
Affiliation(s)
- Alessandro Caputo
- Department of Pathology, University Hospital "San Giovanni di Dio e Ruggi D'Aragona", Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Elisabetta Maffei
- Department of Pathology, University Hospital "San Giovanni di Dio e Ruggi D'Aragona", Salerno, Italy
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
| | - Nalini Gupta
- Department of Cytology and Gynecological Pathology, Postgraduate Institute of Medical Education and Research (PGIMER), Chandigarh, India
| | - Luca Cima
- Department of Diagnostic and Public Health, Section of Pathology, University and Hospital Trust of Verona, Campobasso, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| | - Giorgio Cazzaniga
- Department of Medicine and Surgery, Pathology, IRCCS Fondazione San Gerardo dei Tintori, University of Milano-Bicocca, Catania, Italy
| | - Pietro Pepe
- Department of Urology, Cannizzaro Hospital, Catania, Italy
| | - Paolo Verze
- Department of Medicine and Surgery, University of Salerno, Baronissi, Italy
- Department of Urology, University Hospital "San Giovanni di Dio e Ruggi D'Aragona", Salerno, Italy
| | - Filippo Fraggetta
- Department of Pathology, Pathology Unit, Gravina Hospital, Caltagirone, Italy
| |
Collapse
|
8
|
Sultan S, Gorris MAJ, Martynova E, van der Woude LL, Buytenhuijs F, van Wilpe S, Verrijp K, Figdor CG, de Vries IJM, Textor J. ImmuNet: a segmentation-free machine learning pipeline for immune landscape phenotyping in tumors by multiplex imaging. Biol Methods Protoc 2024; 10:bpae094. [PMID: 39866377 PMCID: PMC11769680 DOI: 10.1093/biomethods/bpae094] [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: 11/14/2024] [Accepted: 12/16/2024] [Indexed: 01/28/2025] Open
Abstract
Tissue specimens taken from primary tumors or metastases contain important information for diagnosis and treatment of cancer patients. Multiplex imaging allows in situ visualization of heterogeneous cell populations, such as immune cells, in tissue samples. Most image processing pipelines first segment cell boundaries and then measure marker expression to assign cell phenotypes. In dense tissue environments, this segmentation-first approach can be inaccurate due to segmentation errors or overlapping cells. Here, we introduce the machine-learning pipeline "ImmuNet", which identifies positions and phenotypes of cells without segmenting them. ImmuNet is easy to train: human annotators only need to click on an immune cell and score its expression of each marker-drawing a full cell outline is not required. We trained and evaluated ImmuNet on multiplex images from human tonsil, lung cancer, prostate cancer, melanoma, and bladder cancer tissue samples and found it to consistently achieve error rates below 5%-10% across tissue types, cell types, and tissue densities, outperforming a segmentation-based baseline method. Furthermore, we externally validate ImmuNet results by comparing them to flow cytometric cell count measurements from the same tissue. In summary, ImmuNet is an effective, simpler alternative to segmentation-based approaches when only cell positions and phenotypes, but not their shapes, are required for downstream analyses. Thus, ImmuNet helps researchers to analyze cell positions in multiplex tissue images more easily and accurately.
Collapse
Affiliation(s)
- Shabaz Sultan
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, The Netherlands
| | - Mark A J Gorris
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Oncode Institute, Radboudumc, Nijmegen 6525 GA, The Netherlands
| | - Evgenia Martynova
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, The Netherlands
| | - Lieke L van der Woude
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Oncode Institute, Radboudumc, Nijmegen 6525 GA, The Netherlands
- Department of Pathology, Radboudumc, Nijmegen 6525 GA, The Netherlands
| | - Franka Buytenhuijs
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, The Netherlands
| | - Sandra van Wilpe
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Department of Medical Oncology, Radboudumc, Nijmegen 6525 GA, The Netherlands
| | - Kiek Verrijp
- Oncode Institute, Radboudumc, Nijmegen 6525 GA, The Netherlands
- Department of Pathology, Radboudumc, Nijmegen 6525 GA, The Netherlands
| | - Carl G Figdor
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Oncode Institute, Radboudumc, Nijmegen 6525 GA, The Netherlands
| | | | - Johannes Textor
- Medical BioSciences, Radboudumc, Nijmegen 6562 GA, The Netherlands
- Data Science Group, Institute for Computing and Information Sciences, Radboud University, Nijmegen 6525 EC, The Netherlands
| |
Collapse
|
9
|
Brattoli B, Mostafavi M, Lee T, Jung W, Ryu J, Park S, Park J, Pereira S, Shin S, Choi S, Kim H, Yoo D, Ali SM, Paeng K, Ock CY, Cho SI, Kim S. A universal immunohistochemistry analyzer for generalizing AI-driven assessment of immunohistochemistry across immunostains and cancer types. NPJ Precis Oncol 2024; 8:277. [PMID: 39627299 PMCID: PMC11615360 DOI: 10.1038/s41698-024-00770-z] [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: 06/27/2024] [Accepted: 11/22/2024] [Indexed: 12/06/2024] Open
Abstract
Immunohistochemistry (IHC) is the common companion diagnostics in targeted therapies. However, quantifying protein expressions in IHC images present a significant challenge, due to variability in manual scoring and inherent subjective interpretation. Deep learning (DL) offers a promising approach to address these issues, though current models require extensive training for each cancer and IHC type, limiting the practical application. We developed a Universal IHC (UIHC) analyzer, a DL-based tool that quantifies protein expression across different cancers and IHC types. This multi-cohort trained model outperformed conventional single-cohort models in analyzing unseen IHC images (Kappa score 0.578 vs. up to 0.509) and demonstrated consistent performance across varying positive staining cutoff values. In a discovery application, the UIHC model assigned higher tumor proportion scores to MET amplification cases, but not MET exon 14 splicing or other non-small cell lung cancer cases. This UIHC model represents a novel role for DL that further advances quantitative analysis of IHC.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Sangjoon Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Hyojin Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | | | | | | | | | | | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea.
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea.
| |
Collapse
|
10
|
Crispino A, Varricchio S, Ilardi G, Russo D, Di Crescenzo RM, Staibano S, Merolla F. A Digital Workflow for Automated Assessment of Tumor-Infiltrating Lymphocytes in Oral Squamous Cell Carcinoma Using QuPath and a StarDist-Based Model. Pathologica 2024; 116:390-403. [PMID: 39748724 DOI: 10.32074/1591-951x-1069] [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: 08/12/2024] [Accepted: 12/10/2024] [Indexed: 01/04/2025] Open
Abstract
The search for reliable prognostic markers in oral squamous cell carcinoma (OSCC) remains a critical need. Tumor-infiltrating lymphocytes (TILs), particularly T lymphocytes, play a pivotal role in the immune response against tumors and are strongly correlated with favorable prognoses. Computational pathology has proven highly effective for histopathological image analysis, automating tasks such as cell detection, classification, and segmentation. In the present study, we developed a StarDist-based model to automatically detect T lymphocytes in hematoxylin and eosin (H&E)-stained whole-slide images (WSIs) of OSCC, bypassing the need for traditional immunohistochemistry (IHC). Using QuPath, we generated training datasets from annotated slides, employing IHC as the ground truth. Our model was validated on Cancer Genome Atlas-derived OSCC images, and survival analyses demonstrated that higher TIL densities correlated with improved patient outcomes. This work introduces an efficient, AI-powered workflow for automated immune profiling in OSCC, offering a reproducible and scalable approach for diagnostic and prognostic applications.
Collapse
Affiliation(s)
- Angela Crispino
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Silvia Varricchio
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Gennaro Ilardi
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Daniela Russo
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Rosa Maria Di Crescenzo
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Stefania Staibano
- Department of Advanced Biomedical Sciences, Pathology Unit, University of Naples "Federico II", Naples, Italy
| | - Francesco Merolla
- Department of Medicine and Health Sciences "V. Tiberio", University of Molise, Campobasso, Italy
| |
Collapse
|
11
|
Hosseini MS, Bejnordi BE, Trinh VQH, Chan L, Hasan D, Li X, Yang S, Kim T, Zhang H, Wu T, Chinniah K, Maghsoudlou S, Zhang R, Zhu J, Khaki S, Buin A, Chaji F, Salehi A, Nguyen BN, Samaras D, Plataniotis KN. Computational pathology: A survey review and the way forward. J Pathol Inform 2024; 15:100357. [PMID: 38420608 PMCID: PMC10900832 DOI: 10.1016/j.jpi.2023.100357] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2023] [Revised: 12/21/2023] [Accepted: 12/23/2023] [Indexed: 03/02/2024] Open
Abstract
Computational Pathology (CPath) is an interdisciplinary science that augments developments of computational approaches to analyze and model medical histopathology images. The main objective for CPath is to develop infrastructure and workflows of digital diagnostics as an assistive CAD system for clinical pathology, facilitating transformational changes in the diagnosis and treatment of cancer that are mainly address by CPath tools. With evergrowing developments in deep learning and computer vision algorithms, and the ease of the data flow from digital pathology, currently CPath is witnessing a paradigm shift. Despite the sheer volume of engineering and scientific works being introduced for cancer image analysis, there is still a considerable gap of adopting and integrating these algorithms in clinical practice. This raises a significant question regarding the direction and trends that are undertaken in CPath. In this article we provide a comprehensive review of more than 800 papers to address the challenges faced in problem design all-the-way to the application and implementation viewpoints. We have catalogued each paper into a model-card by examining the key works and challenges faced to layout the current landscape in CPath. We hope this helps the community to locate relevant works and facilitate understanding of the field's future directions. In a nutshell, we oversee the CPath developments in cycle of stages which are required to be cohesively linked together to address the challenges associated with such multidisciplinary science. We overview this cycle from different perspectives of data-centric, model-centric, and application-centric problems. We finally sketch remaining challenges and provide directions for future technical developments and clinical integration of CPath. For updated information on this survey review paper and accessing to the original model cards repository, please refer to GitHub. Updated version of this draft can also be found from arXiv.
Collapse
Affiliation(s)
- Mahdi S. Hosseini
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | | | - Vincent Quoc-Huy Trinh
- Institute for Research in Immunology and Cancer of the University of Montreal, Montreal, QC H3T 1J4, Canada
| | - Lyndon Chan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Danial Hasan
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Xingwen Li
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Stephen Yang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Taehyo Kim
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Haochen Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Theodore Wu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Kajanan Chinniah
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Sina Maghsoudlou
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ryan Zhang
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Jiadai Zhu
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Samir Khaki
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| | - Andrei Buin
- Huron Digitial Pathology, St. Jacobs, ON N0B 2N0, Canada
| | - Fatemeh Chaji
- Department of Computer Science and Software Engineering (CSSE), Concordia Univeristy, Montreal, QC H3H 2R9, Canada
| | - Ala Salehi
- Department of Electrical and Computer Engineering, University of New Brunswick, Fredericton, NB E3B 5A3, Canada
| | - Bich Ngoc Nguyen
- University of Montreal Hospital Center, Montreal, QC H2X 0C2, Canada
| | - Dimitris Samaras
- Department of Computer Science, Stony Brook University, Stony Brook, NY 11794, United States
| | - Konstantinos N. Plataniotis
- The Edward S. Rogers Sr. Department of Electrical & Computer Engineering (ECE), University of Toronto, Toronto, ON M5S 3G4, Canada
| |
Collapse
|
12
|
Fiorin A, López Pablo C, Lejeune M, Hamza Siraj A, Della Mea V. Enhancing AI Research for Breast Cancer: A Comprehensive Review of Tumor-Infiltrating Lymphocyte Datasets. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2996-3008. [PMID: 38806950 PMCID: PMC11612116 DOI: 10.1007/s10278-024-01043-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/19/2024] [Accepted: 02/07/2024] [Indexed: 05/30/2024]
Abstract
The field of immunology is fundamental to our understanding of the intricate dynamics of the tumor microenvironment. In particular, tumor-infiltrating lymphocyte (TIL) assessment emerges as essential aspect in breast cancer cases. To gain comprehensive insights, the quantification of TILs through computer-assisted pathology (CAP) tools has become a prominent approach, employing advanced artificial intelligence models based on deep learning techniques. The successful recognition of TILs requires the models to be trained, a process that demands access to annotated datasets. Unfortunately, this task is hampered not only by the scarcity of such datasets, but also by the time-consuming nature of the annotation phase required to create them. Our review endeavors to examine publicly accessible datasets pertaining to the TIL domain and thereby become a valuable resource for the TIL community. The overall aim of the present review is thus to make it easier to train and validate current and upcoming CAP tools for TIL assessment by inspecting and evaluating existing publicly available online datasets.
Collapse
Affiliation(s)
- Alessio Fiorin
- Oncological Pathology and Bioinformatics Research Group, Institut d'Investigació Sanitària Pere Virgili (IISPV), C/Esplanetes no 14, 43500, Tortosa, Spain.
- Department of Pathology, Hospital de Tortosa Verge de la Cinta (HTVC), Institut Català de la Salut (ICS), C/Esplanetes no 14, 43500, Tortosa, Spain.
- Department of Computer Engineering and Mathematics, Universitat Rovira i Virgili (URV), Tarragona, Spain.
| | - Carlos López Pablo
- Oncological Pathology and Bioinformatics Research Group, Institut d'Investigació Sanitària Pere Virgili (IISPV), C/Esplanetes no 14, 43500, Tortosa, Spain.
- Department of Pathology, Hospital de Tortosa Verge de la Cinta (HTVC), Institut Català de la Salut (ICS), C/Esplanetes no 14, 43500, Tortosa, Spain.
- Department of Computer Engineering and Mathematics, Universitat Rovira i Virgili (URV), Tarragona, Spain.
| | - Marylène Lejeune
- Oncological Pathology and Bioinformatics Research Group, Institut d'Investigació Sanitària Pere Virgili (IISPV), C/Esplanetes no 14, 43500, Tortosa, Spain
- Department of Pathology, Hospital de Tortosa Verge de la Cinta (HTVC), Institut Català de la Salut (ICS), C/Esplanetes no 14, 43500, Tortosa, Spain
- Department of Computer Engineering and Mathematics, Universitat Rovira i Virgili (URV), Tarragona, Spain
| | - Ameer Hamza Siraj
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| | - Vincenzo Della Mea
- Department of Mathematics, Computer Science and Physics, University of Udine, Udine, Italy
| |
Collapse
|
13
|
Ercan C, Renne SL, Di Tommaso L, Ng CKY, Piscuoglio S, Terracciano LM. Hepatocellular Carcinoma Immune Microenvironment Analysis: A Comprehensive Assessment with Computational and Classical Pathology. Clin Cancer Res 2024; 30:5105-5115. [PMID: 39264292 DOI: 10.1158/1078-0432.ccr-24-0960] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 07/16/2024] [Accepted: 09/10/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE The spatial variability and clinical relevance of the tumor immune microenvironment (TIME) are still poorly understood for hepatocellular carcinoma (HCC). In this study, we aim to develop a deep learning (DL)-based image analysis model for the spatial analysis of immune cell biomarkers and microscopically evaluate the distribution of immune infiltration. EXPERIMENTAL DESIGN Ninety-two HCC surgical liver resections and 51 matched needle biopsies were histologically classified according to their immunophenotypes: inflamed, immune-excluded, and immune-desert. To characterize the TIME on immunohistochemistry (IHC)-stained slides, we designed a multistage DL algorithm, IHC-TIME, to automatically detect immune cells and their localization in the TIME in tumor-stroma and center-border segments. RESULTS Two models were trained to detect and localize the immune cells on IHC-stained slides. The framework models (i.e., immune cell detection models and tumor-stroma segmentation) reached 98% and 91% accuracy, respectively. Patients with inflamed tumors showed better recurrence-free survival than those with immune-excluded or immune-desert tumors. Needle biopsies were found to be 75% accurate in representing the immunophenotypes of the main tumor. Finally, we developed an algorithm that defines immunophenotypes automatically based on the IHC-TIME analysis, achieving an accuracy of 80%. CONCLUSIONS Our DL-based tool can accurately analyze and quantify immune cells on IHC-stained slides of HCC. Microscopic classification of the TIME can stratify HCC according to the patient prognosis. Needle biopsies can provide valuable insights for TIME-related prognostic prediction, albeit with specific constraints. The computational pathology tool provides a new way to study the HCC TIME.
Collapse
Affiliation(s)
- Caner Ercan
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
| | - Salvatore Lorenzo Renne
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Luca Di Tommaso
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| | - Charlotte K Y Ng
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Salvatore Piscuoglio
- Institute of Medical Genetics and Pathology, University Hospital Basel, University of Basel, Basel, Switzerland
- IRCCS Humanitas Research Hospital, Milan, Italy
| | - Luigi M Terracciano
- IRCCS Humanitas Research Hospital, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Milan, Italy
| |
Collapse
|
14
|
Li X, Heirman CC, Rickard AG, Sotolongo G, Castillo R, Adanlawo T, Everitt JI, Hodgin JB, Watts TL, Janowczyk A, Mowery YM, Barisoni L, Lafata KJ. Computational staining of CD3/CD20 positive lymphocytes in human tissues with experimental confirmation in a genetically engineered mouse model. Front Immunol 2024; 15:1451261. [PMID: 39530103 PMCID: PMC11550988 DOI: 10.3389/fimmu.2024.1451261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 09/18/2024] [Indexed: 11/16/2024] Open
Abstract
Introduction Immune dysregulation plays a major role in cancer progression. The quantification of lymphocytic spatial inflammation may enable spatial system biology, improve understanding of therapeutic resistance, and contribute to prognostic imaging biomarkers. Methods In this paper, we propose a knowledge-guided deep learning framework to measure the lymphocytic spatial architecture on human H&E tissue, where the fidelity of training labels is maximized through single-cell resolution image registration of H&E to IHC. We demonstrate that such an approach enables pixel-perfect ground-truth labeling of lymphocytes on H&E as measured by IHC. We then experimentally validate our technique in a genetically engineered, immune-compromised Rag2 mouse model, where Rag2 knockout mice lacking mature lymphocytes are used as a negative experimental control. Such experimental validation moves beyond the classical statistical testing of deep learning models and demonstrates feasibility of more rigorous validation strategies that integrate computational science and basic science. Results Using our developed approach, we automatically annotated more than 111,000 human nuclei (45,611 CD3/CD20 positive lymphocytes) on H&E images to develop our model, which achieved an AUC of 0.78 and 0.71 on internal hold-out testing data and external testing on an independent dataset, respectively. As a measure of the global spatial architecture of the lymphocytic microenvironment, the average structural similarity between predicted lymphocytic density maps and ground truth lymphocytic density maps was 0.86 ± 0.06 on testing data. On experimental mouse model validation, we measured a lymphocytic density of 96.5 ± %1% in a Rag2+/- control mouse, compared to an average of 16.2 ± %5% in Rag2-/- immune knockout mice (p<0.0001, ANOVA-test). Discussion These results demonstrate that CD3/CD20 positive lymphocytes can be accurately detected and characterized on H&E by deep learning and generalized across species. Collectively, these data suggest that our understanding of complex biological systems may benefit from computationally-derived spatial analysis, as well as integration of computational science and basic science.
Collapse
Affiliation(s)
- Xiang Li
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States
| | - Casey C. Heirman
- Medical Physics Graduate Program, Duke University, Durham, NC, United States
| | - Ashlyn G. Rickard
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | - Gina Sotolongo
- Department of Pathology, Duke University, Durham, NC, United States
| | - Rico Castillo
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | - Temitayo Adanlawo
- Department of Radiation Oncology, Duke University, Durham, NC, United States
| | | | - Jeffery B. Hodgin
- Department of Pathology, University of Michigan, Ann Arbor, MI, United States
| | - Tammara L. Watts
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham, NC, United States
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, United States
- Department of Oncology, Division of Precision Oncology, Geneva University Hospitals, Geneva, Switzerland
- Department of Diagnostics, Division of Clinical Pathology, Geneva University Hospitals, Geneva, Switzerland
| | - Yvonne M. Mowery
- Department of Radiation Oncology, Duke University, Durham, NC, United States
- Department of Head and Neck Surgery & Communication Sciences, Duke University, Durham, NC, United States
- Department of Radiation Oncology, UPMC Hillman Cancer Center/University of Pittsburgh, Pittsburgh, PA, United States
| | - Laura Barisoni
- Department of Pathology, Duke University, Durham, NC, United States
- Department of Medicine, Division of Nephrology, Duke University, Durham, NC, United States
| | - Kyle J. Lafata
- Department of Electrical and Computer Engineering, Duke University, Durham, NC, United States
- Medical Physics Graduate Program, Duke University, Durham, NC, United States
- Department of Radiation Oncology, Duke University, Durham, NC, United States
- Department of Pathology, Duke University, Durham, NC, United States
- Department of Radiology, Duke University, Durham, NC, United States
| |
Collapse
|
15
|
Baharun NB, Adam A, Zailani MAH, Rajpoot NM, Xu Q, Zin RRM. Automated scoring methods for quantitative interpretation of Tumour infiltrating lymphocytes (TILs) in breast cancer: a systematic review. BMC Cancer 2024; 24:1202. [PMID: 39350098 PMCID: PMC11440723 DOI: 10.1186/s12885-024-12962-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Accepted: 09/18/2024] [Indexed: 10/04/2024] Open
Abstract
Tumour microenvironment (TME) of breast cancer mainly comprises malignant, stromal, immune, and tumour infiltrating lymphocyte (TILs). Assessment of TILs is crucial for determining the disease's prognosis. Manual TIL assessments are hampered by multiple limitations, including low precision, poor inter-observer reproducibility, and time consumption. In response to these challenges, automated scoring emerges as a promising approach. The aim of this systematic review is to assess the evidence on the approaches and performance of automated scoring methods for TILs assessment in breast cancer. This review presents a comprehensive compilation of studies related to automated scoring of TILs, sourced from four databases (Web of Science, Scopus, Science Direct, and PubMed), employing three primary keywords (artificial intelligence, breast cancer, and tumor-infiltrating lymphocytes). The PICOS framework was employed for study eligibility, and reporting adhered to the PRISMA guidelines. The initial search yielded a total of 1910 articles. Following screening and examination, 27 studies met the inclusion criteria and data were extracted for the review. The findings indicate a concentration of studies on automated TILs assessment in developed countries, specifically the United States and the United Kingdom. From the analysis, a combination of sematic segmentation and object detection (n = 10, 37%) and convolutional neural network (CNN) (n = 11, 41%), become the most frequent automated task and ML approaches applied for model development respectively. All models developed their own ground truth datasets for training and validation, and 59% of the studies assessed the prognostic value of TILs. In conclusion, this analysis contends that automated scoring methods for TILs assessment of breast cancer show significant promise for commodification and application within clinical settings.
Collapse
Affiliation(s)
- Nurkhairul Bariyah Baharun
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia.
- Department of Medical Diagnostic, Faculty of Health Sciences, Universiti Selangor, Jalan Zirkon A7/7, Seksyen 7, Shah Alam, Selangor, 40000, Malaysia.
| | - Afzan Adam
- Centre for Artificial Intelligence Technology (CAIT), Faculty of Information Science & Technology, The National University of Malaysia, Bangi, Selangor, 43600, Malaysia
| | - Mohamed Afiq Hidayat Zailani
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia
- Department of Pathology and Forensic Pathology, Faculty of Medicine, MAHSA University, Bandar Saujana Putra, Malaysia
| | - Nasir M Rajpoot
- Department of Computer Science, University of Warwick, 6 Lord Bhattacharyya Way, Coventry, CV4 7EZ, UK
| | - Qiaoyi Xu
- Centre for Artificial Intelligence Technology (CAIT), Faculty of Information Science & Technology, The National University of Malaysia, Bangi, Selangor, 43600, Malaysia
| | - Reena Rahayu Md Zin
- Department of Pathology, Faculty of Medicine, The National University of Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Wilayah Persekutuan, 56000, Malaysia
| |
Collapse
|
16
|
Hoang DT, Dinstag G, Shulman ED, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. A deep-learning framework to predict cancer treatment response from histopathology images through imputed transcriptomics. NATURE CANCER 2024; 5:1305-1317. [PMID: 38961276 DOI: 10.1038/s43018-024-00793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an indirect two-step approach consisting of (1) DeepPT, a deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response to targeted and immune therapies from the inferred expression values. We show that DeepPT successfully predicts transcriptomics in all 16 The Cancer Genome Atlas cohorts tested and generalizes well to two independent datasets. ENLIGHT-DeepPT successfully predicts true responders in five independent patient cohorts involving four different treatments spanning six cancer types, with an overall odds ratio of 2.28 and a 39.5% increased response rate among predicted responders versus the baseline rate. Notably, its prediction accuracy, obtained without any training on the treatment data, is comparable to that achieved by directly predicting the response from the images, which requires specific training on the treatment evaluation cohorts.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | | | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Leandro C Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - James L Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
17
|
Zamojski D, Gogler A, Scieglinska D, Marczyk M. EpidermaQuant: Unsupervised Detection and Quantification of Epidermal Differentiation Markers on H-DAB-Stained Images of Reconstructed Human Epidermis. Diagnostics (Basel) 2024; 14:1904. [PMID: 39272688 PMCID: PMC11394256 DOI: 10.3390/diagnostics14171904] [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: 07/15/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/15/2024] Open
Abstract
The integrity of the reconstructed human epidermis generated in vitro can be assessed using histological analyses combined with immunohistochemical staining of keratinocyte differentiation markers. Technical differences during the preparation and capture of stained images may influence the outcome of computational methods. Due to the specific nature of the analyzed material, no annotated datasets or dedicated methods are publicly available. Using a dataset with 598 unannotated images showing cross-sections of in vitro reconstructed human epidermis stained with DAB-based immunohistochemistry reaction to visualize four different keratinocyte differentiation marker proteins (filaggrin, keratin 10, Ki67, HSPA2) and counterstained with hematoxylin, we developed an unsupervised method for the detection and quantification of immunohistochemical staining. The pipeline consists of the following steps: (i) color normalization; (ii) color deconvolution; (iii) morphological operations; (iv) automatic image rotation; and (v) clustering. The most effective combination of methods includes (i) Reinhard's normalization; (ii) Ruifrok and Johnston color-deconvolution method; (iii) proposed image-rotation method based on boundary distribution of image intensity; and (iv) k-means clustering. The results of the work should enhance the performance of quantitative analyses of protein markers in reconstructed human epidermis samples and enable the comparison of their spatial distribution between different experimental conditions.
Collapse
Affiliation(s)
- Dawid Zamojski
- Department of Data Science and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
- Genetic Laboratory, Gyncentrum Sp. z o.o., 41-208 Sosnowiec, Poland
| | - Agnieszka Gogler
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-102 Gliwice, Poland
| | - Dorota Scieglinska
- Center for Translational Research and Molecular Biology of Cancer, Maria Sklodowska-Curie National Research Institute of Oncology Gliwice Branch, 44-102 Gliwice, Poland
| | - Michal Marczyk
- Department of Data Science and Engineering, Silesian University of Technology, 44-100 Gliwice, Poland
- Yale Cancer Center, Yale School of Medicine, New Haven, CT 06510, USA
| |
Collapse
|
18
|
Hussain I, Boza J, Lukande R, Ayanga R, Semeere A, Cesarman E, Martin J, Maurer T, Erickson D. Automated detection of Kaposi sarcoma-associated herpesvirus infected cells in immunohistochemical images of skin biopsies. RESEARCH SQUARE 2024:rs.3.rs-4736178. [PMID: 39184072 PMCID: PMC11343169 DOI: 10.21203/rs.3.rs-4736178/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Immunohistochemical (IHC) staining for the antigen of Kaposi sarcoma-associated herpesvirus (KSHV), latency-associated nuclear antigen (LANA), is helpful in diagnosing Kaposi sarcoma (KS). A challenge, however, lies in distinguishing anti-LANA-positive cells from morphologically similar brown counterparts. In this work, we demonstrate a framework for automated localization and quantification of LANA positivity in whole slide images (WSI) of skin biopsies, leveraging weakly supervised multiple instance learning (MIL) while reducing false positive predictions by introducing a novel morphology-based slide aggregation method. Our framework generates interpretable heatmaps, offering insights into precise anti-LANA-positive cell localization within WSIs and a quantitative value for the percentage of positive tiles, which may assist with histological subtyping. We trained and tested our framework with an anti-LANA-stained KS pathology dataset prepared by pathologists in the United States from skin biopsies of KS-suspected patients investigated in Uganda. We achieved an area under the receiver operating characteristic curve (AUC) of 0.99 with a sensitivity and specificity of 98.15% and 96.00% in predicting anti-LANA-positive WSIs in a test dataset. We believe that the framework can provide promise for automated detection of LANA in skin biopsies, which may be especially impactful in resource-limited areas that lack trained pathologists.
Collapse
|
19
|
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
|
20
|
Bakkerus L, Subtil B, Bontkes HJ, Gootjes EC, Reijm M, Vullings M, Verrijp K, Bokhorst JM, Woortman C, Nagtegaal ID, Jonker MA, van der Vliet HJ, Verhoef C, Gorris MA, de Vries IJM, de Gruijl TD, Verheul HM, Buffart TE, Tauriello DVF. Exploring immune status in peripheral blood and tumor tissue in association with survival in patients with multi-organ metastatic colorectal cancer. Oncoimmunology 2024; 13:2361971. [PMID: 38868078 PMCID: PMC11168219 DOI: 10.1080/2162402x.2024.2361971] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 05/27/2024] [Indexed: 06/14/2024] Open
Abstract
Colorectal cancer (CRC) raises considerable clinical challenges, including a high mortality rate once the tumor spreads to distant sites. At this advanced stage, more accurate prediction of prognosis and treatment outcome is urgently needed. The role of cancer immunity in metastatic CRC (mCRC) is poorly understood. Here, we explore cellular immune cell status in patients with multi-organ mCRC. We analyzed T cell infiltration in primary tumor sections, surveyed the lymphocytic landscape of liver metastases, and assessed circulating mononuclear immune cells. Besides asking whether immune cells are associated with survival at this stage of the disease, we investigated correlations between the different tissue types; as this could indicate a dominant immune phenotype. Taken together, our analyses corroborate previous observations that higher levels of CD8+ T lymphocytes link to better survival outcomes. Our findings therefore extend evidence from earlier stages of CRC to indicate an important role for cancer immunity in disease control even after metastatic spreading to multiple organs. This finding may help to improve predicting outcome of patients with mCRC and suggests a future role for immunotherapeutic strategies.
Collapse
Affiliation(s)
- Lotte Bakkerus
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Beatriz Subtil
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hetty J. Bontkes
- Department Laboratory Medicine, LGDO, Section Medical Immunology, Amsterdam, The Netherlands
| | - Elske C. Gootjes
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Martine Reijm
- Department Laboratory Medicine, LGDO, Section Medical Immunology, Amsterdam, The Netherlands
| | - Manon Vullings
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Kiek Verrijp
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - John-Melle Bokhorst
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Carmen Woortman
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Iris D. Nagtegaal
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Marianne A. Jonker
- Department of IQ Health, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Hans J. van der Vliet
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Cornelis Verhoef
- Department of Surgery, ErasmusMC Cancer Institute, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mark A.J. Gorris
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - I. Jolanda M. de Vries
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Tanja D. de Gruijl
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Henk M.W. Verheul
- Department of Medical Oncology, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| | - Tineke E. Buffart
- Department of Medical Oncology, Cancer Center Amsterdam, Amsterdam UMC, Location VUmc, Amsterdam, The Netherlands
| | - Daniele V. F. Tauriello
- Department of Medical BioSciences, Radboud University Medical Center, Nijmegen, The Netherlands
- Department of Medical Oncology, Erasmus MC Cancer Institute, University Medical Center Rotterdam, The Netherlands
| |
Collapse
|
21
|
Kazemi A, Rasouli-Saravani A, Gharib M, Albuquerque T, Eslami S, Schüffler PJ. A systematic review of machine learning-based tumor-infiltrating lymphocytes analysis in colorectal cancer: Overview of techniques, performance metrics, and clinical outcomes. Comput Biol Med 2024; 173:108306. [PMID: 38554659 DOI: 10.1016/j.compbiomed.2024.108306] [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: 10/14/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
The incidence of colorectal cancer (CRC), one of the deadliest cancers around the world, is increasing. Tissue microenvironment (TME) features such as tumor-infiltrating lymphocytes (TILs) can have a crucial impact on diagnosis or decision-making for treating patients with CRC. While clinical studies showed that TILs improve the host immune response, leading to a better prognosis, inter-observer agreement for quantifying TILs is not perfect. Incorporating machine learning (ML) based applications in clinical routine may promote diagnosis reliability. Recently, ML has shown potential for making progress in routine clinical procedures. We aim to systematically review the TILs analysis based on ML in CRC histological images. Deep learning (DL) and non-DL techniques can aid pathologists in identifying TILs, and automated TILs are associated with patient outcomes. However, a large multi-institutional CRC dataset with a diverse and multi-ethnic population is necessary to generalize ML methods.
Collapse
Affiliation(s)
- Azar Kazemi
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany.
| | - Ashkan Rasouli-Saravani
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Gharib
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Saeid Eslami
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Informatics, University of Amsterdam, Amsterdam, the Netherlands.
| | - Peter J Schüffler
- Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany; TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; Munich Center for Machine Learning, Munich, Germany; Munich Data Science Institute, Munich, Germany.
| |
Collapse
|
22
|
Zhang Q, Ren T, Cao K, Xu Z. Advances of machine learning-assisted small extracellular vesicles detection strategy. Biosens Bioelectron 2024; 251:116076. [PMID: 38340580 DOI: 10.1016/j.bios.2024.116076] [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: 09/05/2023] [Revised: 01/22/2024] [Accepted: 01/23/2024] [Indexed: 02/12/2024]
Abstract
Detection of extracellular vesicles (EVs), particularly small EVs (sEVs), is of great significance in exploring their physiological characteristics and clinical applications. The heterogeneity of sEVs plays a crucial role in distinguishing different types of cells and diseases. Machine learning, with its exceptional data processing capabilities, offers a solution to overcome the limitations of conventional detection methods for accurately classifying sEV subtypes and sources. Principal component analysis, linear discriminant analysis, partial least squares discriminant analysis, XGBoost, support vector machine, k-nearest neighbor, and deep learning, along with some combined methods such as principal component-linear discriminant analysis, have been successfully applied in the detection and identification of sEVs. This review focuses on machine learning-assisted detection strategies for cell identification and disease prediction via sEVs, and summarizes the integration of these strategies with surface-enhanced Raman scattering, electrochemistry, inductively coupled plasma mass spectrometry and fluorescence. The performance of different machine learning-based detection strategies is compared, and the advantages and limitations of various machine learning models are also evaluated. Finally, we discuss the merits and limitations of the current approaches and briefly outline the perspective of potential research directions in the field of sEV analysis based on machine learning.
Collapse
Affiliation(s)
- Qi Zhang
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Tingju Ren
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Ke Cao
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China
| | - Zhangrun Xu
- Research Center for Analytical Sciences, Northeastern University, Shenyang, 110819, PR China.
| |
Collapse
|
23
|
Dawson H, Bokhorst J, Studer L, Vieth M, Oguz Erdogan AS, Kus Öztürk S, Kirsch R, Brockmoeller S, Cathomas G, Buslei R, Fink D, Roumet M, Zlobec I, van der Laak J, Nagtegaal ID, Lugli A. Lymph node metastases and recurrence in pT1 colorectal cancer: Prediction with the International Budding Consortium Score-A retrospective, multi-centric study. United European Gastroenterol J 2024; 12:299-308. [PMID: 38193866 PMCID: PMC11017758 DOI: 10.1002/ueg2.12521] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 11/03/2023] [Indexed: 01/10/2024] Open
Abstract
BACKGROUND The International Collaboration on Cancer Reporting proposes histological tumour type, lymphovascular invasion, tumour grade, perineural invasion, extent, and dimensions of invasion as risk factors for lymph node metastases and tumour progression in completely endoscopically resected pT1 colorectal cancer (CRC). OBJECTIVE The aim of the study was to propose a predictive and reliable score to optimise the clinical management of endoscopically resected pT1 CRC patients. METHODS This multi-centric, retrospective International Budding Consortium (IBC) study included an international pT1 CRC cohort of 565 patients. All cases were reviewed by eight expert gastrointestinal pathologists. All risk factors were reported according to international guidelines. Tumour budding and immune response (CD8+ T-cells) were assessed with automated models using artificial intelligence. We used the information on risk factors and least absolute shrinkage and selection operator logistic regression to develop a prediction model and generate a score to predict the occurrence of lymph node metastasis or cancer recurrence. RESULTS The IBC prediction score included the following parameters: lymphovascular invasion, tumour buds, infiltration depth and tumour grade. The score has an acceptable discrimination power (area under the curve of 0.68 [95% confidence intervals (CI) 0.61-0.75]; 0.64 [95% CI 0.57-0.71] after internal validation). At a cut-off of 6.8 points to discriminate high-and low-risk patients, the score had a sensitivity and specificity of 0.9 [95% CI 0.8-0.95] and 0.26 [95% 0.22, 0.3], respectively. CONCLUSION The IBC score is based on well-established risk factors and is a promising tool with clinical utility to support the management of pT1 CRC patients.
Collapse
Affiliation(s)
- Heather Dawson
- Institute of Tissue Medicine and PathologyUniversity of BernBernSwitzerland
| | | | - Linda Studer
- Institute of Tissue Medicine and PathologyUniversity of BernBernSwitzerland
- Institute of Artificial Intelligence and Complex SystemsUniversity of Applied Sciences and Arts Western SwitzerlandFribourgSwitzerland
| | - Michael Vieth
- Institute of PathologyFriedrich‐Alexander‐University Erlangen‐NurembergKlinikum BayreuthBayreuthGermany
| | | | | | - Richard Kirsch
- Pathology and Laboratory MedicineMount Sinai HospitalUniversity of TorontoTorontoOntarioCanada
| | - Scarlett Brockmoeller
- Pathology and Data AnalyticsLeeds Institute of Medical Research at St. James's School of MedicineLeedsUK
| | - Gieri Cathomas
- Institute of PathologyKantonsspital BasellandLiestalSwitzerland
- Present address:
Institute of Tissue Medicine and PathologyUniversity of BernBernSwitzerland.
| | - Rolf Buslei
- Institut und Praxis für Pathologie, Neuropathologie, Molekulare Diagnostik und ZytologieSozialstiftung BambergBambergGermany
| | - David Fink
- Department of Pathology and ImmunologyBaylor College of MedicineHoustonTexasUSA
| | - Marie Roumet
- Clinical Trials UnitUniversity of BernBernSwitzerland
| | - Inti Zlobec
- Institute of Tissue Medicine and PathologyUniversity of BernBernSwitzerland
| | | | | | - Alessandro Lugli
- Institute of Tissue Medicine and PathologyUniversity of BernBernSwitzerland
| |
Collapse
|
24
|
Russo C, Bria A, Marrocco C. GravityNet for end-to-end small lesion detection. Artif Intell Med 2024; 150:102842. [PMID: 38553147 DOI: 10.1016/j.artmed.2024.102842] [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: 10/27/2023] [Revised: 03/01/2024] [Accepted: 03/11/2024] [Indexed: 04/02/2024]
Abstract
This paper introduces a novel one-stage end-to-end detector specifically designed to detect small lesions in medical images. Precise localization of small lesions presents challenges due to their appearance and the diverse contextual backgrounds in which they are found. To address this, our approach introduces a new type of pixel-based anchor that dynamically moves towards the targeted lesion for detection. We refer to this new architecture as GravityNet, and the novel anchors as gravity points since they appear to be "attracted" by the lesions. We conducted experiments on two well-established medical problems involving small lesions to evaluate the performance of the proposed approach: microcalcifications detection in digital mammograms and microaneurysms detection in digital fundus images. Our method demonstrates promising results in effectively detecting small lesions in these medical imaging tasks.
Collapse
Affiliation(s)
- Ciro Russo
- Department of Electrical and Information Engineering, University of Cassino and L.M., Via G. Di Biasio 43, 03043 Cassino (FR), Italy.
| | - Alessandro Bria
- Department of Electrical and Information Engineering, University of Cassino and L.M., Via G. Di Biasio 43, 03043 Cassino (FR), Italy.
| | - Claudio Marrocco
- Department of Electrical and Information Engineering, University of Cassino and L.M., Via G. Di Biasio 43, 03043 Cassino (FR), Italy.
| |
Collapse
|
25
|
Naji H, Sancere L, Simon A, Büttner R, Eich ML, Lohneis P, Bożek K. HoLy-Net: Segmentation of histological images of diffuse large B-cell lymphoma. Comput Biol Med 2024; 170:107978. [PMID: 38237235 DOI: 10.1016/j.compbiomed.2024.107978] [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: 07/13/2023] [Revised: 10/30/2023] [Accepted: 01/08/2024] [Indexed: 02/28/2024]
Abstract
Over the last years, there has been large progress in automated segmentation and classification methods in histological whole slide images (WSIs) stained with hematoxylin and eosin (H&E). Current state-of-the-art (SOTA) techniques are based on diverse datasets of H&E-stained WSIs of different types of predominantly solid cancer. However, there is a scarcity of methods and datasets enabling segmentation of tumors of the lymphatic system (lymphomas). Here, we propose a solution for segmentation of diffuse large B-cell lymphoma (DLBCL), the most common non-Hodgkin's lymphoma. Our method applies to both H&E-stained slides and to a broad range of markers stained with immunohistochemistry (IHC). While IHC staining is an important tool in cancer diagnosis and treatment decisions, there are few automated segmentation and classification methods for IHC-stained WSIs. To address the challenges of nuclei segmentation in H&E- and IHC-stained DLBCL images, we propose HoLy-Net - a HoVer-Net-based deep learning model for lymphoma segmentation. We train two different models, one for segmenting H&E- and one for IHC-stained images and compare the test results with the SOTA methods as well as with the original version of HoVer-Net. Subsequently, we segment patient WSIs and perform single cell-level analysis of different cell types to identify patient-specific tumor characteristics such as high level of immune infiltration. Our method outperforms general-purpose segmentation methods for H&E staining in lymphoma WSIs (with an F1 score of 0.899) and is also a unique automated method for IHC slide segmentation (with an F1 score of 0.913). With our solution, we provide a new dataset we denote LyNSeC (lymphoma nuclear segmentation and classification) containing 73,931 annotated cell nuclei from H&E and 87,316 from IHC slides. Our method and dataset open up new avenues for quantitative, large-scale studies of morphology and microenvironment of lymphomas overlooked by the current automated segmentation methods.
Collapse
Affiliation(s)
- Hussein Naji
- Institute for Biomedical Informatics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany.
| | - Lucas Sancere
- Institute for Biomedical Informatics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany
| | - Adrian Simon
- Institute of Pathology, University Hospital Cologne, Germany
| | | | - Marie-Lisa Eich
- Institute of Pathology, University Hospital Cologne, Germany
| | - Philipp Lohneis
- Institute of Pathology, University Hospital Cologne, Germany; Hämatopathologie Lübeck, Germany
| | - Katarzyna Bożek
- Institute for Biomedical Informatics, Faculty of Medicine and University Hospital Cologne, University of Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Germany
| |
Collapse
|
26
|
Jiao Y, van der Laak J, Albarqouni S, Li Z, Tan T, Bhalerao A, Cheng S, Ma J, Pocock J, Pluim JPW, Koohbanani NA, Bashir RMS, Raza SEA, Liu S, Graham S, Wetstein S, Khurram SA, Liu X, Rajpoot N, Veta M, Ciompi F. LYSTO: The Lymphocyte Assessment Hackathon and Benchmark Dataset. IEEE J Biomed Health Inform 2024; 28:1161-1172. [PMID: 37878422 DOI: 10.1109/jbhi.2023.3327489] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2023]
Abstract
We introduce LYSTO, the Lymphocyte Assessment Hackathon, which was held in conjunction with the MICCAI 2019 Conference in Shenzhen (China). The competition required participants to automatically assess the number of lymphocytes, in particular T-cells, in images of colon, breast, and prostate cancer stained with CD3 and CD8 immunohistochemistry. Differently from other challenges setup in medical image analysis, LYSTO participants were solely given a few hours to address this problem. In this paper, we describe the goal and the multi-phase organization of the hackathon; we describe the proposed methods and the on-site results. Additionally, we present post-competition results where we show how the presented methods perform on an independent set of lung cancer slides, which was not part of the initial competition, as well as a comparison on lymphocyte assessment between presented methods and a panel of pathologists. We show that some of the participants were capable to achieve pathologist-level performance at lymphocyte assessment. After the hackathon, LYSTO was left as a lightweight plug-and-play benchmark dataset on grand-challenge website, together with an automatic evaluation platform.
Collapse
|
27
|
Leo M, Carcagnì P, Signore L, Corcione F, Benincasa G, Laukkanen MO, Distante C. Convolutional Neural Networks in the Diagnosis of Colon Adenocarcinoma. AI 2024; 5:324-341. [DOI: 10.3390/ai5010016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
Colorectal cancer is one of the most lethal cancers because of late diagnosis and challenges in the selection of therapy options. The histopathological diagnosis of colon adenocarcinoma is hindered by poor reproducibility and a lack of standard examination protocols required for appropriate treatment decisions. In the current study, using state-of-the-art approaches on benchmark datasets, we analyzed different architectures and ensembling strategies to develop the most efficient network combinations to improve binary and ternary classification. We propose an innovative two-stage pipeline approach to diagnose colon adenocarcinoma grading from histological images in a similar manner to a pathologist. The glandular regions were first segmented by a transformer architecture with subsequent classification using a convolutional neural network (CNN) ensemble, which markedly improved the learning efficiency and shortened the learning time. Moreover, we prepared and published a dataset for clinical validation of the developed artificial neural network, which suggested the discovery of novel histological phenotypic alterations in adenocarcinoma sections that could have prognostic value. Therefore, AI could markedly improve the reproducibility, efficiency, and accuracy of colon cancer diagnosis, which are required for precision medicine to personalize the treatment of cancer patients.
Collapse
Affiliation(s)
- Marco Leo
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council (CNR) of Italy, 73100 Lecce, Italy
| | - Pierluigi Carcagnì
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council (CNR) of Italy, 73100 Lecce, Italy
| | - Luca Signore
- Dipartimento di Ingegneria per L’Innovazione, Università del Salento, 73100 Lecce, Italy
| | | | | | - Mikko O. Laukkanen
- Department of Translational Medical Sciences, University of Naples Federico II, 80131 Naples, Italy
| | - Cosimo Distante
- Institute of Applied Sciences and Intelligent Systems (ISASI), National Research Council (CNR) of Italy, 73100 Lecce, Italy
- Dipartimento di Ingegneria per L’Innovazione, Università del Salento, 73100 Lecce, Italy
| |
Collapse
|
28
|
Tavolara TE, Su Z, Gurcan MN, Niazi MKK. One label is all you need: Interpretable AI-enhanced histopathology for oncology. Semin Cancer Biol 2023; 97:70-85. [PMID: 37832751 DOI: 10.1016/j.semcancer.2023.09.006] [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: 10/24/2022] [Revised: 09/06/2023] [Accepted: 09/25/2023] [Indexed: 10/15/2023]
Abstract
Artificial Intelligence (AI)-enhanced histopathology presents unprecedented opportunities to benefit oncology through interpretable methods that require only one overall label per hematoxylin and eosin (H&E) slide with no tissue-level annotations. We present a structured review of these methods organized by their degree of verifiability and by commonly recurring application areas in oncological characterization. First, we discuss morphological markers (tumor presence/absence, metastases, subtypes, grades) in which AI-identified regions of interest (ROIs) within whole slide images (WSIs) verifiably overlap with pathologist-identified ROIs. Second, we discuss molecular markers (gene expression, molecular subtyping) that are not verified via H&E but rather based on overlap with positive regions on adjacent tissue. Third, we discuss genetic markers (mutations, mutational burden, microsatellite instability, chromosomal instability) that current technologies cannot verify if AI methods spatially resolve specific genetic alterations. Fourth, we discuss the direct prediction of survival to which AI-identified histopathological features quantitatively correlate but are nonetheless not mechanistically verifiable. Finally, we discuss in detail several opportunities and challenges for these one-label-per-slide methods within oncology. Opportunities include reducing the cost of research and clinical care, reducing the workload of clinicians, personalized medicine, and unlocking the full potential of histopathology through new imaging-based biomarkers. Current challenges include explainability and interpretability, validation via adjacent tissue sections, reproducibility, data availability, computational needs, data requirements, domain adaptability, external validation, dataset imbalances, and finally commercialization and clinical potential. Ultimately, the relative ease and minimum upfront cost with which relevant data can be collected in addition to the plethora of available AI methods for outcome-driven analysis will surmount these current limitations and achieve the innumerable opportunities associated with AI-driven histopathology for the benefit of oncology.
Collapse
Affiliation(s)
- Thomas E Tavolara
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Ziyu Su
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - Metin N Gurcan
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA
| | - M Khalid Khan Niazi
- Center for Artificial Intelligence Research, Wake Forest University School of Medicine, Winston-Salem, NC, USA.
| |
Collapse
|
29
|
Ghahremani P, Marino J, Hernandez-Prera J, de la Iglesia JV, Slebos RJ, Chung CH, Nadeem S. An AI-Ready Multiplex Staining Dataset for Reproducible and Accurate Characterization of Tumor Immune Microenvironment. MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION : MICCAI ... INTERNATIONAL CONFERENCE ON MEDICAL IMAGE COMPUTING AND COMPUTER-ASSISTED INTERVENTION 2023; 14225:704-713. [PMID: 37841230 PMCID: PMC10571229 DOI: 10.1007/978-3-031-43987-2_68] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/15/2023]
Abstract
We introduce a new AI-ready computational pathology dataset containing restained and co-registered digitized images from eight head-and-neck squamous cell carcinoma patients. Specifically, the same tumor sections were stained with the expensive multiplex immunofluorescence (mIF) assay first and then restained with cheaper multiplex immunohistochemistry (mIHC). This is a first public dataset that demonstrates the equivalence of these two staining methods which in turn allows several use cases; due to the equivalence, our cheaper mIHC staining protocol can offset the need for expensive mIF staining/scanning which requires highly-skilled lab technicians. As opposed to subjective and error-prone immune cell annotations from individual pathologists (disagreement > 50%) to drive SOTA deep learning approaches, this dataset provides objective immune and tumor cell annotations via mIF/mIHC restaining for more reproducible and accurate characterization of tumor immune microenvironment (e.g. for immunotherapy). We demonstrate the effectiveness of this dataset in three use cases: (1) IHC quantification of CD3/CD8 tumor-infiltrating lymphocytes via style transfer, (2) virtual translation of cheap mIHC stains to more expensive mIF stains, and (3) virtual tumor/immune cellular phenotyping on standard hematoxylin images. The dataset is available at https://github.com/nadeemlab/DeepLIIF.
Collapse
Affiliation(s)
| | - Joseph Marino
- Memorial Sloan Kettering Cancer Center, New York NY 10065, USA
| | | | | | | | | | - Saad Nadeem
- Memorial Sloan Kettering Cancer Center, New York NY 10065, USA
| |
Collapse
|
30
|
Cooper M, Ji Z, Krishnan RG. Machine learning in computational histopathology: Challenges and opportunities. Genes Chromosomes Cancer 2023; 62:540-556. [PMID: 37314068 DOI: 10.1002/gcc.23177] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 05/18/2023] [Accepted: 05/20/2023] [Indexed: 06/15/2023] Open
Abstract
Digital histopathological images, high-resolution images of stained tissue samples, are a vital tool for clinicians to diagnose and stage cancers. The visual analysis of patient state based on these images are an important part of oncology workflow. Although pathology workflows have historically been conducted in laboratories under a microscope, the increasing digitization of histopathological images has led to their analysis on computers in the clinic. The last decade has seen the emergence of machine learning, and deep learning in particular, a powerful set of tools for the analysis of histopathological images. Machine learning models trained on large datasets of digitized histopathology slides have resulted in automated models for prediction and stratification of patient risk. In this review, we provide context for the rise of such models in computational histopathology, highlight the clinical tasks they have found success in automating, discuss the various machine learning techniques that have been applied to this domain, and underscore open problems and opportunities.
Collapse
Affiliation(s)
- Michael Cooper
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- University Health Network, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Zongliang Ji
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
| | - Rahul G Krishnan
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Vector Institute, Toronto, Ontario, Canada
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Ontario, Canada
| |
Collapse
|
31
|
Rauf Z, Khan AR, Sohail A, Alquhayz H, Gwak J, Khan A. Lymphocyte detection for cancer analysis using a novel fusion block based channel boosted CNN. Sci Rep 2023; 13:14047. [PMID: 37640739 PMCID: PMC10462751 DOI: 10.1038/s41598-023-40581-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/13/2023] [Indexed: 08/31/2023] Open
Abstract
Tumor-infiltrating lymphocytes, specialized immune cells, are considered an important biomarker in cancer analysis. Automated lymphocyte detection is challenging due to its heterogeneous morphology, variable distribution, and presence of artifacts. In this work, we propose a novel Boosted Channels Fusion-based CNN "BCF-Lym-Detector" for lymphocyte detection in multiple cancer histology images. The proposed network initially selects candidate lymphocytic regions at the tissue level and then detects lymphocytes at the cellular level. The proposed "BCF-Lym-Detector" generates diverse boosted channels by utilizing the feature learning capability of different CNN architectures. In this connection, a new adaptive fusion block is developed to combine and select the most relevant lymphocyte-specific features from the generated enriched feature space. Multi-level feature learning is used to retain lymphocytic spatial information and detect lymphocytes with variable appearances. The assessment of the proposed "BCF-Lym-Detector" show substantial improvement in terms of F-score (0.93 and 0.84 on LYSTO and NuClick, respectively), which suggests that the diverse feature extraction and dynamic feature selection enhanced the feature learning capacity of the proposed network. Moreover, the proposed technique's generalization on unseen test sets with a good recall (0.75) and F-score (0.73) shows its potential use for pathologists' assistance.
Collapse
Affiliation(s)
- Zunaira Rauf
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan
- PIEAS Artificial Intelligence Center (PAIC), Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan
| | - Abdul Rehman Khan
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan
| | - Anabia Sohail
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan
- Department of Electrical Engineering and Computer Science, Khalifa University of Science and Technology, Abu Dhabi, UAE
| | - Hani Alquhayz
- Department of Computer Science and Information, College of Science in Zulfi, Majmaah University, 11952, Al-Majmaah, Saudi Arabia
| | - Jeonghwan Gwak
- Department of Software, Korea National University of Transportation, Chungju, 27469, Republic of Korea.
| | - Asifullah Khan
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan.
- PIEAS Artificial Intelligence Center (PAIC), Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan.
- Center for Mathematical Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, 45650, Islamabad, Pakistan.
| |
Collapse
|
32
|
Thagaard J, Broeckx G, Page DB, Jahangir CA, Verbandt S, Kos Z, Gupta R, Khiroya R, Abduljabbar K, Acosta Haab G, Acs B, Akturk G, Almeida JS, Alvarado‐Cabrero I, Amgad M, Azmoudeh‐Ardalan F, Badve S, Baharun NB, Balslev E, Bellolio ER, Bheemaraju V, Blenman KRM, Botinelly Mendonça Fujimoto L, Bouchmaa N, Burgues O, Chardas A, Chon U Cheang M, Ciompi F, Cooper LAD, Coosemans A, Corredor G, Dahl AB, Dantas Portela FL, Deman F, Demaria S, Doré Hansen J, Dudgeon SN, Ebstrup T, Elghazawy M, Fernandez‐Martín C, Fox SB, Gallagher WM, Giltnane JM, Gnjatic S, Gonzalez‐Ericsson PI, Grigoriadis A, Halama N, Hanna MG, Harbhajanka A, Hart SN, Hartman J, Hauberg S, Hewitt S, Hida AI, Horlings HM, Husain Z, Hytopoulos E, Irshad S, Janssen EAM, Kahila M, Kataoka TR, Kawaguchi K, Kharidehal D, Khramtsov AI, Kiraz U, Kirtani P, Kodach LL, Korski K, Kovács A, Laenkholm A, Lang‐Schwarz C, Larsimont D, Lennerz JK, Lerousseau M, Li X, Ly A, Madabhushi A, Maley SK, Manur Narasimhamurthy V, Marks DK, McDonald ES, Mehrotra R, Michiels S, Minhas FUAA, Mittal S, Moore DA, Mushtaq S, Nighat H, Papathomas T, Penault‐Llorca F, Perera RD, Pinard CJ, Pinto‐Cardenas JC, Pruneri G, Pusztai L, Rahman A, Rajpoot NM, Rapoport BL, Rau TT, Reis‐Filho JS, et alThagaard J, Broeckx G, Page DB, Jahangir CA, Verbandt S, Kos Z, Gupta R, Khiroya R, Abduljabbar K, Acosta Haab G, Acs B, Akturk G, Almeida JS, Alvarado‐Cabrero I, Amgad M, Azmoudeh‐Ardalan F, Badve S, Baharun NB, Balslev E, Bellolio ER, Bheemaraju V, Blenman KRM, Botinelly Mendonça Fujimoto L, Bouchmaa N, Burgues O, Chardas A, Chon U Cheang M, Ciompi F, Cooper LAD, Coosemans A, Corredor G, Dahl AB, Dantas Portela FL, Deman F, Demaria S, Doré Hansen J, Dudgeon SN, Ebstrup T, Elghazawy M, Fernandez‐Martín C, Fox SB, Gallagher WM, Giltnane JM, Gnjatic S, Gonzalez‐Ericsson PI, Grigoriadis A, Halama N, Hanna MG, Harbhajanka A, Hart SN, Hartman J, Hauberg S, Hewitt S, Hida AI, Horlings HM, Husain Z, Hytopoulos E, Irshad S, Janssen EAM, Kahila M, Kataoka TR, Kawaguchi K, Kharidehal D, Khramtsov AI, Kiraz U, Kirtani P, Kodach LL, Korski K, Kovács A, Laenkholm A, Lang‐Schwarz C, Larsimont D, Lennerz JK, Lerousseau M, Li X, Ly A, Madabhushi A, Maley SK, Manur Narasimhamurthy V, Marks DK, McDonald ES, Mehrotra R, Michiels S, Minhas FUAA, Mittal S, Moore DA, Mushtaq S, Nighat H, Papathomas T, Penault‐Llorca F, Perera RD, Pinard CJ, Pinto‐Cardenas JC, Pruneri G, Pusztai L, Rahman A, Rajpoot NM, Rapoport BL, Rau TT, Reis‐Filho JS, Ribeiro JM, Rimm D, Roslind A, Vincent‐Salomon A, Salto‐Tellez M, Saltz J, Sayed S, Scott E, Siziopikou KP, Sotiriou C, Stenzinger A, Sughayer MA, Sur D, Fineberg S, Symmans F, Tanaka S, Taxter T, Tejpar S, Teuwen J, Thompson EA, Tramm T, Tran WT, van der Laak J, van Diest PJ, Verghese GE, Viale G, Vieth M, Wahab N, Walter T, Waumans Y, Wen HY, Yang W, Yuan Y, Zin RM, Adams S, Bartlett J, Loibl S, Denkert C, Savas P, Loi S, Salgado R, Specht Stovgaard E. Pitfalls in machine learning-based assessment of tumor-infiltrating lymphocytes in breast cancer: A report of the International Immuno-Oncology Biomarker Working Group on Breast Cancer. J Pathol 2023; 260:498-513. [PMID: 37608772 PMCID: PMC10518802 DOI: 10.1002/path.6155] [Show More Authors] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 06/07/2023] [Indexed: 08/24/2023]
Abstract
The clinical significance of the tumor-immune interaction in breast cancer is now established, and tumor-infiltrating lymphocytes (TILs) have emerged as predictive and prognostic biomarkers for patients with triple-negative (estrogen receptor, progesterone receptor, and HER2-negative) breast cancer and HER2-positive breast cancer. How computational assessments of TILs might complement manual TIL assessment in trial and daily practices is currently debated. Recent efforts to use machine learning (ML) to automatically evaluate TILs have shown promising results. We review state-of-the-art approaches and identify pitfalls and challenges of automated TIL evaluation by studying the root cause of ML discordances in comparison to manual TIL quantification. We categorize our findings into four main topics: (1) technical slide issues, (2) ML and image analysis aspects, (3) data challenges, and (4) validation issues. The main reason for discordant assessments is the inclusion of false-positive areas or cells identified by performance on certain tissue patterns or design choices in the computational implementation. To aid the adoption of ML for TIL assessment, we provide an in-depth discussion of ML and image analysis, including validation issues that need to be considered before reliable computational reporting of TILs can be incorporated into the trial and routine clinical management of patients with triple-negative breast cancer. © 2023 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Jeppe Thagaard
- Technical University of DenmarkKongens LyngbyDenmark
- Visiopharm A/SHørsholmDenmark
| | - Glenn Broeckx
- Department of PathologyGZA‐ZNA HospitalsAntwerpBelgium
- Centre for Oncological Research (CORE), MIPPRO, Faculty of MedicineAntwerp UniversityAntwerpBelgium
| | - David B Page
- Earle A Chiles Research InstituteProvidence Cancer InstitutePortlandORUSA
| | - Chowdhury Arif Jahangir
- UCD School of Biomolecular and Biomedical Science, UCD Conway InstituteUniversity College DublinDublinIreland
| | - Sara Verbandt
- Digestive Oncology, Department of OncologyKU LeuvenLeuvenBelgium
| | - Zuzana Kos
- Department of Pathology and Laboratory MedicineBC Cancer Vancouver Centre, University of British ColumbiaVancouverBritish ColumbiaCanada
| | - Rajarsi Gupta
- Department of Biomedical InformaticsStony Brook UniversityStony BrookNYUSA
| | - Reena Khiroya
- Department of Cellular PathologyUniversity College Hospital LondonLondonUK
| | | | | | - Balazs Acs
- Department of Oncology and PathologyKarolinska InstitutetStockholmSweden
- Department of Clinical Pathology and Cancer DiagnosticsKarolinska University HospitalStockholmSweden
| | - Guray Akturk
- Translational Molecular Biomarkers, Merck & Co IncRahwayNJUSA
| | - Jonas S Almeida
- Division of Cancer Epidemiology and Genetics (DCEG)National Cancer Institute (NCI)Rockville, MDUSA
| | | | - Mohamed Amgad
- Department of PathologyNorthwestern University Feinberg School of MedicineChicagoILUSA
| | | | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Emory University School of MedicineEmory University Winship Cancer InstituteAtlantaGAUSA
| | | | - Eva Balslev
- Department of PathologyHerlev and Gentofte HospitalHerlevDenmark
| | - Enrique R Bellolio
- Departamento de Anatomía Patológica, Facultad de MedicinaUniversidad de La FronteraTemucoChile
| | | | - Kim RM Blenman
- Department of Internal Medicine Section of Medical Oncology and Yale Cancer CenterYale School of MedicineNew HavenCTUSA
- Department of Computer ScienceYale School of Engineering and Applied ScienceNew HavenCTUSA
| | | | - Najat Bouchmaa
- Institute of Biological Sciences, Faculty of Medical SciencesMohammed VI Polytechnic University (UM6P)Ben‐GuerirMorocco
| | - Octavio Burgues
- Pathology DepartmentHospital Cliníco Universitario de Valencia/InclivaValenciaSpain
| | - Alexandros Chardas
- Department of Pathobiology & Population SciencesThe Royal Veterinary CollegeLondonUK
| | - Maggie Chon U Cheang
- Head of Integrative Genomics Analysis in Clinical Trials, ICR‐CTSU, Division of Clinical StudiesThe Institute of Cancer ResearchLondonUK
| | - Francesco Ciompi
- Radboud University Medical CenterDepartment of PathologyNijmegenThe Netherlands
| | - Lee AD Cooper
- Department of PathologyNorthwestern Feinberg School of MedicineChicagoILUSA
| | - An Coosemans
- Department of Oncology, Laboratory of Tumor Immunology and ImmunotherapyKU LeuvenLeuvenBelgium
| | - Germán Corredor
- Biomedical Engineering DepartmentEmory UniversityAtlantaGAUSA
| | - Anders B Dahl
- Technical University of DenmarkKongens LyngbyDenmark
| | | | | | - Sandra Demaria
- Department of Radiation OncologyWeill Cornell MedicineNew YorkNYUSA
- Department of Pathology and Laboratory MedicineWeill Cornell MedicineNew YorkNYUSA
| | | | - Sarah N Dudgeon
- Conputational Biology and BioinformaticsYale UniversityNew HavenCTUSA
| | | | | | - Claudio Fernandez‐Martín
- Instituto Universitario de Investigación en Tecnología Centrada en el Ser Humano, HUMAN‐techUniversitat Politècnica de ValènciaValenciaSpain
| | - Stephen B Fox
- Pathology, Peter MacCallum Cancer Centre and Sir Peter MacCallum Department of OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - William M Gallagher
- UCD School of Biomolecular and Biomedical Science, UCD Conway InstituteUniversity College DublinDublinIreland
| | | | - Sacha Gnjatic
- Department of Oncological Sciences, Medicine Hem/Onc, and Pathology, Tisch Cancer Institute – Precision Immunology InstituteIcahn School of Medicine at Mount SinaiNew YorkNYUSA
| | | | - Anita Grigoriadis
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- The Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Niels Halama
- Department of Translational ImmunotherapyGerman Cancer Research CenterHeidelbergGermany
| | - Matthew G Hanna
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkUSA
| | | | - Steven N Hart
- Department of Laboratory Medicine and PathologyMayo ClinicRochester, MNUSA
| | - Johan Hartman
- Department of Oncology and PathologyKarolinska InstitutetStockholmSweden
- Department of Clinical Pathology and Cancer DiagnosticsKarolinska University HospitalStockholmSweden
| | - Søren Hauberg
- Technical University of DenmarkKongens LyngbyDenmark
| | - Stephen Hewitt
- Laboratory of Pathology, Center for Cancer Research, National Cancer InstituteNational Institutes of HealthBethesdaMDUSA
| | - Akira I Hida
- Department of PathologyMatsuyama Shimin HospitalMatsuyamaJapan
| | - Hugo M Horlings
- Division of PathologyNetherlands Cancer Institute (NKI)AmsterdamThe Netherlands
| | | | | | - Sheeba Irshad
- King's College London & Guy's & St Thomas’ NHS TrustLondonUK
| | - Emiel AM Janssen
- Department of PathologyStavanger University HospitalStavangerNorway
- Department of Chemistry, Bioscience and Environmental TechnologyUniversity of StavangerStavangerNorway
| | | | | | - Kosuke Kawaguchi
- Department of Breast SurgeryKyoto University Graduate School of MedicineKyotoJapan
| | | | - Andrey I Khramtsov
- Department of Pathology and Laboratory MedicineAnn & Robert H. Lurie Children's Hospital of ChicagoChicagoILUSA
| | - Umay Kiraz
- Department of PathologyStavanger University HospitalStavangerNorway
- Department of Chemistry, Bioscience and Environmental TechnologyUniversity of StavangerStavangerNorway
| | - Pawan Kirtani
- Department of HistopathologyAakash Healthcare Super Speciality HospitalNew DelhiIndia
| | - Liudmila L Kodach
- Department of PathologyNetherlands Cancer Institute – Antoni van Leeuwenhoek HospitalAmsterdamThe Netherlands
| | - Konstanty Korski
- Data, Analytics and Imaging, Product DevelopmentF. Hoffmann‐La Roche AGBaselSwitzerland
| | - Anikó Kovács
- Department of Clinical PathologySahlgrenska University HospitalGothenburgSweden
- Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anne‐Vibeke Laenkholm
- Department of Surgical PathologyZealand University HospitalRoskildeDenmark
- Department of Surgical PathologyUniversity of CopenhagenCopenhagenDenmark
| | - Corinna Lang‐Schwarz
- Institute of Pathology, Klinikum Bayreuth GmbHFriedrich‐Alexander‐University Erlangen‐NurembergBayreuthGermany
| | - Denis Larsimont
- Institut Jules BordetUniversité Libre de BruxellesBrusselsBelgium
| | - Jochen K Lennerz
- Center for Integrated DiagnosticsMassachusetts General Hospital/Harvard Medical SchoolBostonMAUSA
| | - Marvin Lerousseau
- Centre for Computational Biology (CBIO)Mines Paris, PSL UniversityParisFrance
- Institut CuriePSL UniversityParisFrance
- INSERMParisFrance
| | - Xiaoxian Li
- Department of Pathology and Laboratory MedicineEmory UniversityAtlantaGAUSA
| | - Amy Ly
- Department of PathologyMassachusetts General HospitalBostonMAUSA
| | - Anant Madabhushi
- Department of Biomedical Engineering, Radiology and Imaging Sciences, Biomedical Informatics, PathologyGeorgia Institute of Technology and Emory UniversityAtlantaGAUSA
| | - Sai K Maley
- NRG Oncology/NSABP FoundationPittsburghPAUSA
| | | | | | - Elizabeth S McDonald
- Breast Cancer Translational Research GroupUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Ravi Mehrotra
- Indian Cancer Genomic AtlasPuneIndia
- Centre for Health, Innovation and Policy FoundationNoidaIndia
| | - Stefan Michiels
- Office of Biostatistics and Epidemiology, Gustave Roussy, Oncostat U1018, InsermUniversity Paris‐Saclay, Ligue Contre le Cancer labeled TeamVillejuifFrance
| | - Fayyaz ul Amir Afsar Minhas
- Tissue Image Analytics Centre, Warwick Cancer Research Centre, PathLAKE Consortium, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Shachi Mittal
- Department of Chemical Engineering, Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattle, WAUSA
| | - David A Moore
- CRUK Lung Cancer Centre of Excellence, UCL and Cellular Pathology DepartmentUCLHLondonUK
| | - Shamim Mushtaq
- Department of BiochemistryZiauddin UniversityKarachiPakistan
| | - Hussain Nighat
- Pathology and Laboratory MedicineAll India Institute of Medical sciencesRaipurIndia
| | - Thomas Papathomas
- Institute of Metabolism and Systems ResearchUniversity of BirminghamBirminghamUK
- Department of Clinical PathologyDrammen Sykehus, Vestre Viken HFDrammenNorway
| | - Frederique Penault‐Llorca
- Centre Jean Perrin, Université Clermont Auvergne, INSERM, U1240 Imagerie Moléculaire et Stratégies ThéranostiquesClermont FerrandFrance
| | - Rashindrie D Perera
- School of Electrical, Mechanical and Infrastructure EngineeringUniversity of MelbourneMelbourneVictoriaAustralia
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Christopher J Pinard
- Radiogenomics LaboratorySunnybrook Health Sciences CentreTorontoOntarioCanada
- Department of Clinical Studies, Ontario Veterinary CollegeUniversity of GuelphGuelphOntarioCanada
- Department of OncologyLakeshore Animal Health PartnersMississaugaOntarioCanada
- Centre for Advancing Responsible and Ethical Artificial Intelligence (CARE‐AI)University of GuelphGuelphOntarioCanada
| | | | - Giancarlo Pruneri
- Department of Pathology and Laboratory MedicineFondazione IRCCS Istituto Nazionale dei TumoriMilanItaly
- Faculty of Medicine and SurgeryUniversity of MilanMilanItaly
| | - Lajos Pusztai
- Yale Cancer CenterYale UniversityNew HavenCTUSA
- Department of Medical Oncology, Yale School of MedicineYale UniversityNew HavenCTUSA
| | - Arman Rahman
- UCD School of Biomolecular and Biomedical Science, UCD Conway InstituteUniversity College DublinDublinIreland
| | | | - Bernardo Leon Rapoport
- The Medical Oncology Centre of RosebankJohannesburgSouth Africa
- Department of Immunology, Faculty of Health SciencesUniversity of PretoriaPretoriaSouth Africa
| | - Tilman T Rau
- Institute of PathologyUniversity Hospital Düsseldorf and Heinrich‐Heine‐University DüsseldorfDüsseldorfGermany
| | - Jorge S Reis‐Filho
- Department of Pathology and Laboratory MedicineMemorial Sloan Kettering Cancer CenterNew YorkNYUSA
| | - Joana M Ribeiro
- Département de Médecine OncologiqueGustave RoussyVillejuifFrance
| | - David Rimm
- Department of PathologyYale University School of MedicineNew HavenCTUSA
- Department of MedicineYale University School of MedicineNew HavenCTUSA
| | - Anne Roslind
- Department of PathologyHerlev and Gentofte HospitalHerlevDenmark
| | - Anne Vincent‐Salomon
- Department of Diagnostic and Theranostic Medicine, Institut CurieUniversity Paris‐Sciences et LettresParisFrance
| | - Manuel Salto‐Tellez
- Integrated Pathology UnitThe Institute of Cancer ResearchLondonUK
- Precision Medicine CentreQueen's University BelfastBelfastUK
| | - Joel Saltz
- Department of Biomedical InformaticsStony Brook UniversityStony BrookNYUSA
| | - Shahin Sayed
- Department of PathologyAga Khan UniversityNairobiKenya
| | - Ely Scott
- Translational PathologyTranslational Sciences and Diagnostics/Translational Medicine/R&D, Bristol Myers SquibbPrincetonNJUSA
| | - Kalliopi P Siziopikou
- Department of Pathology, Section of Breast PathologyNorthwestern University Feinberg School of MedicineChicagoILUSA
| | - Christos Sotiriou
- Breast Cancer Translational Research Laboratory J.‐C. Heuson, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB)Université Libre de Bruxelles (ULB)BrusselsBelgium
- Medical Oncology Department, Institut Jules Bordet, Hôpital Universitaire de Bruxelles (HUB)Université Libre de Bruxelles (ULB)BrusselsBelgium
| | - Albrecht Stenzinger
- Institute of PathologyUniversity Hospital HeidelbergHeidelbergGermany
- Centers for Personalized Medicine (ZPM)HeidelbergGermany
| | | | - Daniel Sur
- Department of Medical OncologyUniversity of Medicine and Pharmacy “Iuliu Hatieganu”Cluj‐NapocaRomania
| | - Susan Fineberg
- Montefiore Medical CenterBronxNYUSA
- Albert Einstein College of MedicineBronxNYUSA
| | - Fraser Symmans
- University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | | | | | - Sabine Tejpar
- Digestive Oncology, Department of OncologyKU LeuvenLeuvenBelgium
| | - Jonas Teuwen
- AI for Oncology Lab, The Netherlands Cancer InstituteAmsterdamThe Netherlands
| | | | - Trine Tramm
- Department of PathologyAarhus University HospitalAarhusDenmark
- Institute of Clinical MedicineAarhus UniversityAarhusDenmark
| | - William T Tran
- Department of Radiation OncologyUniversity of Toronto and Sunnybrook Health Sciences CentreTorontoOntarioCanada
| | - Jeroen van der Laak
- Department of PathologyRadboud University Medical CenterNijmegenThe Netherlands
| | - Paul J van Diest
- Department of PathologyUniversity Medical Center UtrechtThe Netherlands
- Johns Hopkins Oncology CenterBaltimoreMDUSA
| | - Gregory E Verghese
- Cancer Bioinformatics, School of Cancer & Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
- The Breast Cancer Now Research Unit, School of Cancer and Pharmaceutical Sciences, Faculty of Life Sciences and MedicineKing's College LondonLondonUK
| | - Giuseppe Viale
- Department of PathologyEuropean Institute of OncologyMilanItaly
- Department of PathologyUniversity of MilanMilanItaly
| | - Michael Vieth
- Institute of Pathology, Klinikum Bayreuth GmbHFriedrich‐Alexander‐University Erlangen‐NurembergBayreuthGermany
| | - Noorul Wahab
- Tissue Image Analytics Centre, Department of Computer ScienceUniversity of WarwickCoventryUK
| | - Thomas Walter
- Centre for Computational Biology (CBIO)Mines Paris, PSL UniversityParisFrance
- Institut CuriePSL UniversityParisFrance
- INSERMParisFrance
| | | | - Hannah Y Wen
- Department of PathologyMemorial Sloan Kettering Cancer CenterNew YorkUSA
| | - Wentao Yang
- Fudan Medical University Shanghai Cancer CenterShanghaiPR China
| | - Yinyin Yuan
- Department of Translational Molecular Pathology, Division of Pathology and Laboratory MedicineThe University of Texas MD Anderson Cancer CenterHoustonTXUSA
| | - Reena Md Zin
- Department of Pathology, Faculty of MedicineUniversiti Kebangsaan MalaysiaKuala LumpurMalaysia
| | - Sylvia Adams
- Perlmutter Cancer CenterNYU Langone HealthNew YorkNYUSA
- Department of MedicineNYU Grossman School of MedicineManhattanNYUSA
| | | | - Sibylle Loibl
- Department of Medicine and ResearchGerman Breast GroupNeu‐IsenburgGermany
| | - Carsten Denkert
- Institut für PathologiePhilipps‐Universität Marburg und Universitätsklinikum MarburgMarburgGermany
| | - Peter Savas
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of Medical OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Sherene Loi
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
- The Sir Peter MacCallum Department of Medical OncologyUniversity of MelbourneMelbourneVictoriaAustralia
| | - Roberto Salgado
- Department of PathologyGZA‐ZNA HospitalsAntwerpBelgium
- Division of Cancer ResearchPeter MacCallum Cancer CentreMelbourneVictoriaAustralia
| | - Elisabeth Specht Stovgaard
- Department of PathologyHerlev and Gentofte HospitalHerlevDenmark
- Department of Clinical MedicineUniversity of CopenhagenCopenhagenDenmark
| |
Collapse
|
33
|
Mandair D, Reis-Filho JS, Ashworth A. Biological insights and novel biomarker discovery through deep learning approaches in breast cancer histopathology. NPJ Breast Cancer 2023; 9:21. [PMID: 37024522 PMCID: PMC10079681 DOI: 10.1038/s41523-023-00518-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 02/27/2023] [Indexed: 04/08/2023] Open
Abstract
Breast cancer remains a highly prevalent disease with considerable inter- and intra-tumoral heterogeneity complicating prognostication and treatment decisions. The utilization and depth of genomic, transcriptomic and proteomic data for cancer has exploded over recent times and the addition of spatial context to this information, by understanding the correlating morphologic and spatial patterns of cells in tissue samples, has created an exciting frontier of research, histo-genomics. At the same time, deep learning (DL), a class of machine learning algorithms employing artificial neural networks, has rapidly progressed in the last decade with a confluence of technical developments - including the advent of modern graphic processing units (GPU), allowing efficient implementation of increasingly complex architectures at scale; advances in the theoretical and practical design of network architectures; and access to larger datasets for training - all leading to sweeping advances in image classification and object detection. In this review, we examine recent developments in the application of DL in breast cancer histology with particular emphasis of those producing biologic insights or novel biomarkers, spanning the extraction of genomic information to the use of stroma to predict cancer recurrence, with the aim of suggesting avenues for further advancing this exciting field.
Collapse
Affiliation(s)
- Divneet Mandair
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA
| | | | - Alan Ashworth
- UCSF Helen Diller Family Comprehensive Cancer Center, San Francisco, CA, 94158, USA.
| |
Collapse
|
34
|
Verdicchio M, Brancato V, Cavaliere C, Isgrò F, Salvatore M, Aiello M. A pathomic approach for tumor-infiltrating lymphocytes classification on breast cancer digital pathology images. Heliyon 2023; 9:e14371. [PMID: 36950640 PMCID: PMC10025040 DOI: 10.1016/j.heliyon.2023.e14371] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/03/2023] [Accepted: 03/03/2023] [Indexed: 03/11/2023] Open
Abstract
Background and objectives The detection of tumor-infiltrating lymphocytes (TILs) could aid in the development of objective measures of the infiltration grade and can support decision-making in breast cancer (BC). However, manual quantification of TILs in BC histopathological whole slide images (WSI) is currently based on a visual assessment, thus resulting not standardized, not reproducible, and time-consuming for pathologists. In this work, a novel pathomic approach, aimed to apply high-throughput image feature extraction techniques to analyze the microscopic patterns in WSI, is proposed. In fact, pathomic features provide additional information concerning the underlying biological processes compared to the WSI visual interpretation, thus providing more easily interpretable and explainable results than the most frequently investigated Deep Learning based methods in the literature. Methods A dataset containing 1037 regions of interest with tissue compartments and TILs annotated on 195 TNBC and HER2+ BC hematoxylin and eosin (H&E)-stained WSI was used. After segmenting nuclei within tumor-associated stroma using a watershed-based approach, 71 pathomic features were extracted from each nucleus and reduced using a Spearman's correlation filter followed by a nonparametric Wilcoxon rank-sum test and least absolute shrinkage and selection operator. The relevant features were used to classify each candidate nucleus as either TILs or non-TILs using 5 multivariable machine learning classification models trained using 5-fold cross-validation (1) without resampling, (2) with the synthetic minority over-sampling technique and (3) with downsampling. The prediction performance of the models was assessed using ROC curves. Results 21 features were selected, with most of them related to the well-known TILs properties of having regular shape, clearer margins, high peak intensity, more homogeneous enhancement and different textural pattern than other cells. The best performance was obtained by Random-Forest with ROC AUC of 0.86, regardless of resampling technique. Conclusions The presented approach holds promise for the classification of TILs in BC H&E-stained WSI and could provide support to pathologists for a reliable, rapid and interpretable clinical assessment of TILs in BC.
Collapse
Affiliation(s)
| | | | - Carlo Cavaliere
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| | - Francesco Isgrò
- Department of Electrical Engineering and Information Technologies, University of Naples Federico II, Claudio 21, Naples, 80125, Italy
| | - Marco Salvatore
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| | - Marco Aiello
- IRCCS SYNLAB SDN, Via E. Gianturco 113, Naples, 80143, Italy
| |
Collapse
|
35
|
Ding Y, Dhawan G, Jones C, Ness T, Nichols E, Krasnogor N, Reynolds NJ. An open source pipeline for quantitative immunohistochemistry image analysis of inflammatory skin disease using artificial intelligence. J Eur Acad Dermatol Venereol 2023; 37:605-614. [PMID: 36367625 PMCID: PMC10947200 DOI: 10.1111/jdv.18726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 08/17/2022] [Indexed: 11/13/2022]
Abstract
BACKGROUND The application of artificial intelligence (AI) to whole slide images has the potential to improve research reliability and ultimately diagnostic efficiency and service capacity. Image annotation plays a key role in AI and digital pathology. However, the work-streams required for tissue-specific (skin) and immunostain-specific annotation has not been extensively studied compared with the development of AI algorithms. OBJECTIVES The objective of this study is to develop a common workflow for annotating whole slide images of biopsies from inflammatory skin disease immunostained with a variety of epidermal and dermal markers prior to the development of the AI-assisted analysis pipeline. METHODS A total of 45 slides containing 3-5 sections each were scanned using Aperio AT2 slide scanner (Leica Biosystems). These slides were annotated by hand using a commonly used image analysis tool which resulted in more than 4000 images blocks. We used deep learning (DL) methodology to first sequentially segment (epidermis and upper dermis), with the exclusion of common artefacts and second to quantify the immunostained signal in those two compartments of skin biopsies and the ratio of positive cells. RESULTS We validated two DL models using 10-fold validation runs and by comparing to ground truth manually annotated data. The models achieved an average (global) accuracy of 95.0% for the segmentation of epidermis and dermis and 86.1% for the segmentation of positive/negative cells. CONCLUSIONS The application of two DL models in sequence facilitates accurate segmentation of epidermal and dermal structures, exclusion of common artefacts and enables the quantitative analysis of the immunostained signal. However, inaccurate annotation of the slides for training the DL model can decrease the accuracy of the output. Our open source code will facilitate further external validation across different immunostaining platforms and slide scanners.
Collapse
Affiliation(s)
- Yuchun Ding
- Interdisciplinary Computing and Complex Biosystems Research Group, School of Computing ScienceNewcastle UniversityNewcastle upon TyneUK
| | - Gaurav Dhawan
- Institute of Translational and Clinical MedicineNewcastle University Medical SchoolNewcastle upon TyneUK
- Department of Dermatology, Royal Victoria InfirmaryNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Claire Jones
- MRC/EPSRC, Molecular Pathology Node, Department of PathologyNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Thomas Ness
- MRC/EPSRC, Molecular Pathology Node, Department of PathologyNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Esme Nichols
- Institute of Translational and Clinical MedicineNewcastle University Medical SchoolNewcastle upon TyneUK
- Department of Dermatology, Royal Victoria InfirmaryNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| | - Natalio Krasnogor
- Interdisciplinary Computing and Complex Biosystems Research Group, School of Computing ScienceNewcastle UniversityNewcastle upon TyneUK
| | - Nick J. Reynolds
- Institute of Translational and Clinical MedicineNewcastle University Medical SchoolNewcastle upon TyneUK
- Department of Dermatology, Royal Victoria InfirmaryNewcastle Hospitals NHS Foundation TrustNewcastle upon TyneUK
| |
Collapse
|
36
|
Wilm F, Ihling C, Méhes G, Terracciano L, Puget C, Klopfleisch R, Schüffler P, Aubreville M, Maier A, Mrowiec T, Breininger K. Pan-tumor T-lymphocyte detection using deep neural networks: Recommendations for transfer learning in immunohistochemistry. J Pathol Inform 2023; 14:100301. [PMID: 36994311 PMCID: PMC10040882 DOI: 10.1016/j.jpi.2023.100301] [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: 11/25/2022] [Revised: 02/01/2023] [Accepted: 02/11/2023] [Indexed: 03/02/2023] Open
Abstract
The success of immuno-oncology treatments promises long-term cancer remission for an increasing number of patients. The response to checkpoint inhibitor drugs has shown a correlation with the presence of immune cells in the tumor and tumor microenvironment. An in-depth understanding of the spatial localization of immune cells is therefore critical for understanding the tumor's immune landscape and predicting drug response. Computer-aided systems are well suited for efficiently quantifying immune cells in their spatial context. Conventional image analysis approaches are often based on color features and therefore require a high level of manual interaction. More robust image analysis methods based on deep learning are expected to decrease this reliance on human interaction and improve the reproducibility of immune cell scoring. However, these methods require sufficient training data and previous work has reported low robustness of these algorithms when they are tested on out-of-distribution data from different pathology labs or samples from different organs. In this work, we used a new image analysis pipeline to explicitly evaluate the robustness of marker-labeled lymphocyte quantification algorithms depending on the number of training samples before and after being transferred to a new tumor indication. For these experiments, we adapted the RetinaNet architecture for the task of T-lymphocyte detection and employed transfer learning to bridge the domain gap between tumor indications and reduce the annotation costs for unseen domains. On our test set, we achieved human-level performance for almost all tumor indications with an average precision of 0.74 in-domain and 0.72-0.74 cross-domain. From our results, we derive recommendations for model development regarding annotation extent, training sample selection, and label extraction for the development of robust algorithms for immune cell scoring. By extending the task of marker-labeled lymphocyte quantification to a multi-class detection task, the pre-requisite for subsequent analyses, e.g., distinguishing lymphocytes in the tumor stroma from tumor-infiltrating lymphocytes, is met.
Collapse
Affiliation(s)
- Frauke Wilm
- Pattern Recognition Lab, Department of Computer Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Merck Healthcare KGaA, Darmstadt, Germany
- Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Gábor Méhes
- Department of Pathology, University of Debrecen, Debrecen, Hungary
| | - Luigi Terracciano
- Research Department Pathology, Universitätsspital Basel, Basel, Switzerland
| | - Chloé Puget
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Robert Klopfleisch
- Institute of Veterinary Pathology, Freie Universität Berlin, Berlin, Germany
| | - Peter Schüffler
- Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany
- School of Computation, Information and Technology, Technical University of Munich, Munich, Germany
| | | | - Andreas Maier
- Pattern Recognition Lab, Department of Computer Science, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | | - Katharina Breininger
- Department Artificial Intelligence in Biomedical Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
37
|
Rauf Z, Sohail A, Khan SH, Khan A, Gwak J, Maqbool M. Attention-guided multi-scale deep object detection framework for lymphocyte analysis in IHC histological images. Microscopy (Oxf) 2023; 72:27-42. [PMID: 36239597 DOI: 10.1093/jmicro/dfac051] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 09/21/2022] [Accepted: 10/13/2022] [Indexed: 11/14/2022] Open
Abstract
Tumor-infiltrating lymphocytes are specialized lymphocytes that can detect and kill cancerous cells. Their detection poses many challenges due to significant morphological variations, overlapping occurrence, artifact regions and high-class resemblance between clustered areas and artifacts. In this regard, a Lymphocyte Analysis Framework based on Deep Convolutional neural network (DC-Lym-AF) is proposed to analyze lymphocytes in immunohistochemistry images. The proposed framework comprises (i) pre-processing, (ii) screening phase, (iii) localization phase and (iv) post-processing. In the screening phase, a custom convolutional neural network architecture (lymphocyte dilated network) is developed to screen lymphocytic regions by performing a patch-level classification. This proposed architecture uses dilated convolutions and shortcut connections to capture multi-level variations and ensure reference-based learning. In contrast, the localization phase utilizes an attention-guided multi-scale lymphocyte detector to detect lymphocytes. The proposed detector extracts refined and multi-scale features by exploiting dilated convolutions, attention mechanism and feature pyramid network (FPN) using its custom attention-aware backbone. The proposed DC-Lym-AF shows exemplary performance on the NuClick dataset compared with the existing detection models, with an F-score and precision of 0.84 and 0.83, respectively. We verified the generalizability of our proposed framework by participating in a publically open LYON'19 challenge. Results in terms of detection rate (0.76) and F-score (0.73) suggest that the proposed DC-Lym-AF can effectively detect lymphocytes in immunohistochemistry-stained images collected from different laboratories. In addition, its promising generalization on several datasets implies that it can be turned into a medical diagnostic tool to investigate various histopathological problems. Graphical Abstract.
Collapse
Affiliation(s)
- Zunaira Rauf
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan.,PIEAS Artificial Intelligence Center, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan
| | - Anabia Sohail
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan.,Department of Computer Science, Faculty of Computing and Artificial Intelligence, Air University, E-9, Islamabad 44230, Pakistan
| | - Saddam Hussain Khan
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan.,Department of Computer Systems Engineering, University of Engineering and Applied Sciences, Swat, Khyber Pakhtunkhwa 19130, Pakistan
| | - Asifullah Khan
- Pattern Recognition Lab, Department of Computer and Information Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan.,PIEAS Artificial Intelligence Center, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan.,Center for Mathematical Sciences, Pakistan Institute of Engineering and Applied Sciences, Nilore, Islamabad 45650, Pakistan
| | - Jeonghwan Gwak
- Department of Software, Korea National University of Transportation, Chungju 27469, Republic of Korea
| | - Muhammad Maqbool
- The University of Alabama at Birmingham, 1720 2nd Ave South, Birmingham, AL 35294, USA
| |
Collapse
|
38
|
Chan RC, To CKC, Cheng KCT, Yoshikazu T, Yan LLA, Tse GM. Artificial intelligence in breast cancer histopathology. Histopathology 2023; 82:198-210. [PMID: 36482271 DOI: 10.1111/his.14820] [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: 08/01/2022] [Revised: 09/22/2022] [Accepted: 09/28/2022] [Indexed: 12/13/2022]
Abstract
This is a review on the use of artificial intelligence for digital breast pathology. A systematic search on PubMed was conducted, identifying 17,324 research papers related to breast cancer pathology. Following a semimanual screening, 664 papers were retrieved and pursued. The papers are grouped into six major tasks performed by pathologists-namely, molecular and hormonal analysis, grading, mitotic figure counting, ki-67 indexing, tumour-infiltrating lymphocyte assessment, and lymph node metastases identification. Under each task, open-source datasets for research to build artificial intelligence (AI) tools are also listed. Many AI tools showed promise and demonstrated feasibility in the automation of routine pathology investigations. We expect continued growth of AI in this field as new algorithms mature.
Collapse
Affiliation(s)
- Ronald Ck Chan
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Chun Kit Curtis To
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Ka Chuen Tom Cheng
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Tada Yoshikazu
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Lai Ling Amy Yan
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| | - Gary M Tse
- Department of Anatomical and Cellular Pathology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, Hong Kong
| |
Collapse
|
39
|
Yavuz A, Alpsoy A, Gedik EO, Celik MY, Bassorgun CI, Unal B, Elpek GO. Artificial intelligence applications in predicting the behavior of gastrointestinal cancers in pathology. Artif Intell Gastroenterol 2022; 3:142-162. [DOI: 10.35712/aig.v3.i5.142] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 11/25/2022] [Accepted: 12/14/2022] [Indexed: 12/28/2022] Open
Abstract
Recent research has provided a wealth of data supporting the application of artificial intelligence (AI)-based applications in routine pathology practice. Indeed, it is clear that these methods can significantly support an accurate and rapid diagnosis by eliminating errors, increasing reliability, and improving workflow. In addition, the effectiveness of AI in the pathological evaluation of prognostic parameters associated with behavior, course, and treatment in many types of tumors has also been noted. Regarding gastrointestinal system (GIS) cancers, the contribution of AI methods to pathological diagnosis has been investigated in many studies. On the other hand, studies focusing on AI applications in evaluating parameters to determine tumor behavior are relatively few. For this purpose, the potential of AI models has been studied over a broad spectrum, from tumor subtyping to the identification of new digital biomarkers. The capacity of AI to infer genetic alterations of cancer tissues from digital slides has been demonstrated. Although current data suggest the merit of AI-based approaches in assessing tumor behavior in GIS cancers, a wide range of challenges still need to be solved, from laboratory infrastructure to improving the robustness of algorithms, before incorporating AI applications into real-life GIS pathology practice. This review aims to present data from AI applications in evaluating pathological parameters related to the behavior of GIS cancer with an overview of the opportunities and challenges encountered in implementing AI in pathology.
Collapse
Affiliation(s)
- Aysen Yavuz
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Anil Alpsoy
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Elif Ocak Gedik
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | | | | | - Betul Unal
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| | - Gulsum Ozlem Elpek
- Department of Pathology, Akdeniz University Medical School, Antalya 07070, Turkey
| |
Collapse
|
40
|
Nakanishi R, Morooka K, Omori K, Toyota S, Tanaka Y, Hasuda H, Koga N, Nonaka K, Hu Q, Nakaji Y, Nakanoko T, Ando K, Ota M, Kimura Y, Oki E, Oda Y, Yoshizumi T. Artificial Intelligence-Based Prediction of Recurrence after Curative Resection for Colorectal Cancer from Digital Pathological Images. Ann Surg Oncol 2022; 30:3506-3514. [PMID: 36512260 DOI: 10.1245/s10434-022-12926-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Accepted: 10/19/2022] [Indexed: 12/15/2022]
Abstract
BACKGROUND To develop an artificial intelligence-based model to predict recurrence after curative resection for stage I-III colorectal cancer from digitized pathological slides. PATIENTS AND METHODS In this retrospective study, 471 consecutive patients who underwent curative resection for stage I-III colorectal cancer at our institution from 2004 to 2015 were enrolled, and 512 randomly selected tiles from digitally scanned images of hematoxylin and eosin-stained tumor tissue sections were used to train a convolutional neural network. Five-fold cross-validation was used to validate the model. The association between recurrence and the model's output scores were analyzed in the test cohorts. RESULTS The area under the receiver operating characteristic curve of the cross-validation was 0.7245 [95% confidence interval (CI) 0.6707-0.7783; P < 0.0001]. The score successfully classified patients into those with better and worse recurrence free survival (P < 0.0001). Multivariate analysis revealed that a high score was significantly associated with worse recurrence free survival [odds ratio (OR) 1.857; 95% CI 1.248-2.805; P = 0.0021], which was independent from other predictive factors: male sex (P = 0.0238), rectal cancer (P = 0.0396), preoperative abnormal carcinoembryonic antigen (CEA) level (P = 0.0216), pathological T3/T4 stage (P = 0.0162), and pathological positive lymph node metastasis (P < 0.0001). CONCLUSIONS The artificial intelligence-based prediction model discriminated patients with a high risk of recurrence. This approach could help decision-makers consider the benefits of adjuvant chemotherapy.
Collapse
|
41
|
Faghani S, Codipilly DC, David Vogelsang, Moassefi M, Rouzrokh P, Khosravi B, Agarwal S, Dhaliwal L, Katzka DA, Hagen C, Lewis J, Leggett CL, Erickson BJ, Iyer PG. Development of a deep learning model for the histologic diagnosis of dysplasia in Barrett's esophagus. Gastrointest Endosc 2022; 96:918-925.e3. [PMID: 35718071 DOI: 10.1016/j.gie.2022.06.013] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 06/02/2022] [Accepted: 06/08/2022] [Indexed: 12/11/2022]
Abstract
BACKGROUND AND AIMS The risk of progression in Barrett's esophagus (BE) increases with development of dysplasia. There is a critical need to improve the diagnosis of BE dysplasia, given substantial interobserver disagreement among expert pathologists and overdiagnosis of dysplasia by community pathologists. We developed a deep learning model to predict dysplasia grade on whole-slide imaging. METHODS We digitized nondysplastic BE (NDBE), low-grade dysplasia (LGD), and high-grade dysplasia (HGD) histology slides. Two expert pathologists confirmed all histology and digitally annotated areas of dysplasia. Training, validation, and test sets were created (by a random 70/20/10 split). We used an ensemble approach combining a "you only look once" model to identify regions of interest and histology class (NDBE, LGD, or HGD) followed by a ResNet101 model pretrained on ImageNet applied to the regions of interest. Diagnostic performance was determined for the whole slide. RESULTS We included slides from 542 patients (164 NDBE, 226 LGD, and 152 HGD) yielding 8596 bounding boxes in the training set, 1946 bounding boxes in the validation set, and 840 boxes in the test set. When the ensemble model was used, sensitivity and specificity for LGD was 81.3% and 100%, respectively, and >90% for NDBE and HGD. The overall positive predictive value and sensitivity metric (calculated as F1 score) was .91 for NDBE, .90 for LGD, and 1.0 for HGD. CONCLUSIONS We successfully trained and validated a deep learning model to accurately identify dysplasia on whole-slide images. This model can potentially help improve the histologic diagnosis of BE dysplasia and the appropriate application of endoscopic therapy.
Collapse
Affiliation(s)
- Shahriar Faghani
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - D Chamil Codipilly
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - David Vogelsang
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Mana Moassefi
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Pouria Rouzrokh
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bardia Khosravi
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Siddharth Agarwal
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Lovekirat Dhaliwal
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - David A Katzka
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Catherine Hagen
- Department of Pathology, Mayo Clinic, Rochester, Minnesota, USA; (5)Department of Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Jason Lewis
- Department of Pathology, Mayo Clinic, Jacksonville, Florida, USA
| | - Cadman L Leggett
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| | - Bradley J Erickson
- Artificial Intelligence Laboratory, Mayo Clinic, Rochester, Minnesota, USA; Department of Radiology, Mayo Clinic, Rochester, Minnesota, USA
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota, USA
| |
Collapse
|
42
|
Tavolara TE, Gurcan MN, Niazi MKK. Contrastive Multiple Instance Learning: An Unsupervised Framework for Learning Slide-Level Representations of Whole Slide Histopathology Images without Labels. Cancers (Basel) 2022; 14:5778. [PMID: 36497258 PMCID: PMC9738801 DOI: 10.3390/cancers14235778] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Revised: 11/16/2022] [Accepted: 11/19/2022] [Indexed: 11/25/2022] Open
Abstract
Recent methods in computational pathology have trended towards semi- and weakly-supervised methods requiring only slide-level labels. Yet, even slide-level labels may be absent or irrelevant to the application of interest, such as in clinical trials. Hence, we present a fully unsupervised method to learn meaningful, compact representations of WSIs. Our method initially trains a tile-wise encoder using SimCLR, from which subsets of tile-wise embeddings are extracted and fused via an attention-based multiple-instance learning framework to yield slide-level representations. The resulting set of intra-slide-level and inter-slide-level embeddings are attracted and repelled via contrastive loss, respectively. This resulted in slide-level representations with self-supervision. We applied our method to two tasks- (1) non-small cell lung cancer subtyping (NSCLC) as a classification prototype and (2) breast cancer proliferation scoring (TUPAC16) as a regression prototype-and achieved an AUC of 0.8641 ± 0.0115 and correlation (R2) of 0.5740 ± 0.0970, respectively. Ablation experiments demonstrate that the resulting unsupervised slide-level feature space can be fine-tuned with small datasets for both tasks. Overall, our method approaches computational pathology in a novel manner, where meaningful features can be learned from whole-slide images without the need for annotations of slide-level labels. The proposed method stands to benefit computational pathology, as it theoretically enables researchers to benefit from completely unlabeled whole-slide images.
Collapse
Affiliation(s)
- Thomas E. Tavolara
- Center for Biomedical Informatics, Wake Forest School of Medicine, Winston-Salem, NC 27101, USA
| | | | | |
Collapse
|
43
|
Huang K, Lin B, Liu J, Liu Y, Li J, Tian G, Yang J. Predicting colorectal cancer tumor mutational burden from histopathological images and clinical information using multi-modal deep learning. Bioinformatics 2022; 38:5108-5115. [PMID: 36130268 DOI: 10.1093/bioinformatics/btac641] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 08/31/2022] [Accepted: 09/20/2022] [Indexed: 12/24/2022] Open
Abstract
MOTIVATION Tumor mutational burden (TMB) is an indicator of the efficacy and prognosis of immune checkpoint therapy in colorectal cancer (CRC). In general, patients with higher TMB values are more likely to benefit from immunotherapy. Though whole-exome sequencing is considered the gold standard for determining TMB, it is difficult to be applied in clinical practice due to its high cost. There are also a few DNA panel-based methods to estimate TMB; however, their detection cost is also high, and the associated wet-lab experiments usually take days, which emphasize the need for faster and cheaper alternatives. RESULTS In this study, we propose a multi-modal deep learning model based on a residual network (ResNet) and multi-modal compact bilinear pooling to predict TMB status (i.e. TMB high (TMB_H) or TMB low(TMB_L)) directly from histopathological images and clinical data. We applied the model to CRC data from The Cancer Genome Atlas and compared it with four other popular methods, namely, ResNet18, ResNet50, VGG19 and AlexNet. We tested different TMB thresholds, namely, percentiles of 10%, 14.3%, 15%, 16.3%, 20%, 30% and 50%, to differentiate TMB_H and TMB_L.For the percentile of 14.3% (i.e. TMB value 20) and ResNet18, our model achieved an area under the receiver operating characteristic curve of 0.817 after 5-fold cross-validation, which was better than that of other compared models. In addition, we also found that TMB values were significantly associated with the tumor stage and N and M stages. Our study shows that deep learning models can predict TMB status from histopathological images and clinical information only, which is worth clinical application.
Collapse
Affiliation(s)
- Kaimei Huang
- Department of Mathematics, Zhejiang Normal University, Jinghua 321004, China.,Department of Sciences, Geneis (Beijing) Co., Ltd, Beijing 100102, China.,Department of Sciences, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Binghu Lin
- Department of General Surgery of Third Ward, Xiangyang No.1 People's Hospital, Hubei University of Medicine, Xiangyang 441000, China
| | - Jinyang Liu
- Department of Sciences, Geneis (Beijing) Co., Ltd, Beijing 100102, China.,Department of Sciences, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Yankun Liu
- Cancer Institute, Tangshan People's Hospital, Tangshan 063001, China
| | - Jingwu Li
- Cancer Institute, Tangshan People's Hospital, Tangshan 063001, China
| | - Geng Tian
- Department of Sciences, Geneis (Beijing) Co., Ltd, Beijing 100102, China.,Department of Sciences, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| | - Jialiang Yang
- Department of Sciences, Geneis (Beijing) Co., Ltd, Beijing 100102, China.,Department of Sciences, Qingdao Geneis Institute of Big Data Mining and Precision Medicine, Qingdao 266000, China
| |
Collapse
|
44
|
Hermsen M, Ciompi F, Adefidipe A, Denic A, Dendooven A, Smith BH, van Midden D, Bräsen JH, Kers J, Stegall MD, Bándi P, Nguyen T, Swiderska-Chadaj Z, Smeets B, Hilbrands LB, van der Laak JAWM. Convolutional Neural Networks for the Evaluation of Chronic and Inflammatory Lesions in Kidney Transplant Biopsies. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:1418-1432. [PMID: 35843265 DOI: 10.1016/j.ajpath.2022.06.009] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 06/13/2022] [Accepted: 06/22/2022] [Indexed: 06/15/2023]
Abstract
In kidney transplant biopsies, both inflammation and chronic changes are important features that predict long-term graft survival. Quantitative scoring of these features is important for transplant diagnostics and kidney research. However, visual scoring is poorly reproducible and labor intensive. The goal of this study was to investigate the potential of convolutional neural networks (CNNs) to quantify inflammation and chronic features in kidney transplant biopsies. A structure segmentation CNN and a lymphocyte detection CNN were applied on 125 whole-slide image pairs of periodic acid-Schiff- and CD3-stained slides. The CNN results were used to quantify healthy and sclerotic glomeruli, interstitial fibrosis, tubular atrophy, and inflammation within both nonatrophic and atrophic tubuli, and in areas of interstitial fibrosis. The computed tissue features showed high correlation with Banff lesion scores of five pathologists (A.A., A.Dend., J.H.B., J.K., and T.N.). Analyses on a small subset showed a moderate correlation toward higher CD3+ cell density within scarred regions and higher CD3+ cell count inside atrophic tubuli correlated with long-term change of estimated glomerular filtration rate. The presented CNNs are valid tools to yield objective quantitative information on glomeruli number, fibrotic tissue, and inflammation within scarred and non-scarred kidney parenchyma in a reproducible manner. CNNs have the potential to improve kidney transplant diagnostics and will benefit the community as a novel method to generate surrogate end points for large-scale clinical studies.
Collapse
Affiliation(s)
- Meyke Hermsen
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Adeyemi Adefidipe
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands
| | - Aleksandar Denic
- Division of Nephrology and Hypertension, Mayo Clinic, Rochester, Minnesota
| | - Amélie Dendooven
- Department of Pathology, Ghent University Hospital, Ghent, Belgium; Faculty of Medicine, University of Antwerp, Wilrijk, Antwerp, Belgium
| | - Byron H Smith
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, Minnesota; Division of Biomedical Statistics and Informatics, Mayo Clinic, Rochester, Minnesota
| | - Dominique van Midden
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jan Hinrich Bräsen
- Nephropathology Unit, Institute of Pathology, Hannover Medical School, Hannover, Germany
| | - Jesper Kers
- Department of Pathology, Amsterdam University Medical Centers, University of Amsterdam, Amsterdam, the Netherlands; Department of Pathology, Leiden University Medical Center, Leiden, the Netherlands; Center for Analytical Sciences Amsterdam, Van 't Hoff Institute for Molecular Sciences, University of Amsterdam, Amsterdam, the Netherlands
| | - Mark D Stegall
- Division of Transplantation Surgery, Mayo Clinic, Rochester, Minnesota
| | - Péter Bándi
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Tri Nguyen
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Zaneta Swiderska-Chadaj
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands; Faculty of Electrical Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Bart Smeets
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Luuk B Hilbrands
- Department of Nephrology, Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen A W M van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, the Netherlands; Center for Medical Image Science and Visualization, Linköping University, Linköping, Sweden.
| |
Collapse
|
45
|
Da Q, Huang X, Li Z, Zuo Y, Zhang C, Liu J, Chen W, Li J, Xu D, Hu Z, Yi H, Guo Y, Wang Z, Chen L, Zhang L, He X, Zhang X, Mei K, Zhu C, Lu W, Shen L, Shi J, Li J, S S, Krishnamurthi G, Yang J, Lin T, Song Q, Liu X, Graham S, Bashir RMS, Yang C, Qin S, Tian X, Yin B, Zhao J, Metaxas DN, Li H, Wang C, Zhang S. DigestPath: A benchmark dataset with challenge review for the pathological detection and segmentation of digestive-system. Med Image Anal 2022; 80:102485. [DOI: 10.1016/j.media.2022.102485] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 04/08/2022] [Accepted: 05/20/2022] [Indexed: 12/19/2022]
|
46
|
Lefèvre P, Guizzetti L, McKee TD, Zou G, van Viegen T, McFarlane SC, Shackelton L, Feagan BG, Jairath V, Pai RK, Casteele NV. Development and Validation of a Digital Analysis Method to Quantify CD3-immunostained T Lymphocytes in Whole Slide Images of Crohn's Disease Biopsies. Appl Immunohistochem Mol Morphol 2022; 30:486-492. [PMID: 35587994 DOI: 10.1097/pai.0000000000001035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2021] [Accepted: 04/18/2022] [Indexed: 11/25/2022]
Abstract
The T-lymphocyte-mediated inflammation in Crohn's disease can be assessed by quantifying CD3-positive T-lymphocyte counts in colonic sections. We developed and validated a process to reliably quantify immunohistochemical marker-positive cells in a high-throughput setting using whole slide images (WSIs) of CD3-immunostained colonic and ileal tissue sections. In regions of interest (ROIs) and/or whole tissue sections of 40 WSIs from 36 patients with Crohn's disease, CD3-positive cells were quantified by an expert gastrointestinal pathologist (gold standard) and by image analysis algorithms developed with software from 3 independent vendors. Semiautomated quantification of CD3-positive cell counts estimated in 1 ROI per section were accurate when compared with manual analysis (Pearson correlation coefficient, 0.877 to 0.925). Biological variability was acceptable in digitally determined CD3-positive cell measures between 2 to 5 ROIs annotated on the same tissue section (coefficient of variation <25%). Results from computer-aided analysis of CD3-positive T lymphocytes in a whole tissue section and the average of results from 2 to 5 ROIs per tissue section lacked reliability (overestimation or underestimation and systematic bias), suggesting that absolute quantification of CD3-positive T lymphocytes in a whole tissue section may be more accurate. Semiautomated image analysis in WSIs demonstrated reproducible CD3-positive cell measures across 3 independent algorithms. A computer-aided digital image analysis method was developed and validated to quantify CD3-positive T lymphocytes in colonic and ileal biopsy sections from patients with Crohn's disease. Results support consideration of this digital analysis method for use in future Crohn's disease clinical studies.
Collapse
Affiliation(s)
| | | | - Trevor D McKee
- STTARR Innovation Core Facility, Princess Margaret Cancer Centre, University Health Network
- Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| | - Guangyong Zou
- Alimentiv Inc
- Robarts Research Institute, Schulich School of Medicine and Dentistry
- Department of Epidemiology and Biostatistics
| | | | | | | | - Brian G Feagan
- Alimentiv Inc
- Department of Epidemiology and Biostatistics
- Division of Gastroenterology, Western University, London
| | - Vipul Jairath
- Alimentiv Inc
- Department of Epidemiology and Biostatistics
- Division of Gastroenterology, Western University, London
| | - Rish K Pai
- Department of Laboratory Medicine & Pathology, Mayo Clinic Arizona, Scottsdale, AZ
| | - Niels Vande Casteele
- Alimentiv Inc
- Department of Medicine, IBD Center, University of California San Diego, La Jolla, CA
| |
Collapse
|
47
|
Zhang H, Liu J, Wang P, Yu Z, Liu W, Chen H. Cross-Boosted Multi-Target Domain Adaptation for Multi-Modality Histopathology Image Translation and Segmentation. IEEE J Biomed Health Inform 2022; 26:3197-3208. [PMID: 35196252 DOI: 10.1109/jbhi.2022.3153793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Recent digital pathology workflows mainly focus on mono-modality histopathology image analysis. However, they ignore the complementarity between Haematoxylin & Eosin (H&E) and Immunohistochemically (IHC) stained images, which can provide comprehensive gold standard for cancer diagnosis. To resolve this issue, we propose a cross-boosted multi-target domain adaptation pipeline for multi-modality histopathology images, which contains Cross-frequency Style-auxiliary Translation Network (CSTN) and Dual Cross-boosted Segmentation Network (DCSN). Firstly, CSTN achieves the one-to-many translation from fluorescence microscopy images to H&E and IHC images for providing source domain training data. To generate images with realistic color and texture, Cross-frequency Feature Transfer Module (CFTM) is developed to pertinently restructure and normalize high-frequency content and low-frequency style features from different domains. Then, DCSN fulfills multi-target domain adaptive segmentation, where a dual-branch encoder is introduced, and Bidirectional Cross-domain Boosting Module (BCBM) is designed to implement cross-modality information complementation through bidirectional inter-domain collaboration. Finally, we establish Multi-modality Thymus Histopathology (MThH) dataset, which is the largest publicly available H&E and IHC image benchmark. Experiments on MThH dataset and several public datasets show that the proposed pipeline outperforms state-of-the-art methods on both histopathology image translation and segmentation.
Collapse
|
48
|
Chen X, Wang X, Zhang K, Fung KM, Thai TC, Moore K, Mannel RS, Liu H, Zheng B, Qiu Y. Recent advances and clinical applications of deep learning in medical image analysis. Med Image Anal 2022; 79:102444. [PMID: 35472844 PMCID: PMC9156578 DOI: 10.1016/j.media.2022.102444] [Citation(s) in RCA: 275] [Impact Index Per Article: 91.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 03/09/2022] [Accepted: 04/01/2022] [Indexed: 02/07/2023]
Abstract
Deep learning has received extensive research interest in developing new medical image processing algorithms, and deep learning based models have been remarkably successful in a variety of medical imaging tasks to support disease detection and diagnosis. Despite the success, the further improvement of deep learning models in medical image analysis is majorly bottlenecked by the lack of large-sized and well-annotated datasets. In the past five years, many studies have focused on addressing this challenge. In this paper, we reviewed and summarized these recent studies to provide a comprehensive overview of applying deep learning methods in various medical image analysis tasks. Especially, we emphasize the latest progress and contributions of state-of-the-art unsupervised and semi-supervised deep learning in medical image analysis, which are summarized based on different application scenarios, including classification, segmentation, detection, and image registration. We also discuss major technical challenges and suggest possible solutions in the future research efforts.
Collapse
Affiliation(s)
- Xuxin Chen
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Ximin Wang
- School of Information Science and Technology, ShanghaiTech University, Shanghai 201210, China
| | - Ke Zhang
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Kar-Ming Fung
- Department of Pathology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Theresa C Thai
- Department of Radiology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Kathleen Moore
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Robert S Mannel
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, OK 73104, USA
| | - Hong Liu
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Bin Zheng
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA
| | - Yuchen Qiu
- School of Electrical and Computer Engineering, University of Oklahoma, Norman, OK 73019, USA.
| |
Collapse
|
49
|
van der Kamp A, Waterlander TJ, de Bel T, van der Laak J, van den Heuvel-Eibrink MM, Mavinkurve-Groothuis AMC, de Krijger RR. Artificial Intelligence in Pediatric Pathology: The Extinction of a Medical Profession or the Key to a Bright Future? Pediatr Dev Pathol 2022; 25:380-387. [PMID: 35238696 DOI: 10.1177/10935266211059809] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Artificial Intelligence (AI) has become of increasing interest over the past decade. While digital image analysis (DIA) is already being used in radiology, it is still in its infancy in pathology. One of the reasons is that large-scale digitization of glass slides has only recently become available. With the advent of digital slide scanners, that digitize glass slides into whole slide images, many labs are now in a transition phase towards digital pathology. However, only few departments worldwide are currently fully digital. Digital pathology provides the ability to annotate large datasets and train computers to develop and validate robust algorithms, similar to radiology. In this opinionated overview, we will give a brief introduction into AI in pathology, discuss the potential positive and negative implications and speculate about the future role of AI in the field of pediatric pathology.
Collapse
Affiliation(s)
- Ananda van der Kamp
- 541199Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Tomas J Waterlander
- 541199Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands
| | - Thomas de Bel
- Department of Pathology, 234134Radboud University Medical Center, Nijmegen, the Netherlands
| | - Jeroen van der Laak
- Department of Pathology, 234134Radboud University Medical Center, Nijmegen, the Netherlands.,Center for Medical Image Science and Visualization, 4566Linköping University, Linköping, Sweden
| | | | | | - Ronald R de Krijger
- 541199Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.,Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands
| |
Collapse
|
50
|
Peeples JK, Jameson JF, Kotta NM, Grasman JM, Stoppel WL, Zare A. Jointly Optimized Spatial Histogram UNET Architecture (JOSHUA) for Adipose Tissue Segmentation. BME FRONTIERS 2022; 2022:9854084. [PMID: 37850183 PMCID: PMC10521712 DOI: 10.34133/2022/9854084] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2021] [Accepted: 03/31/2022] [Indexed: 10/19/2023] Open
Abstract
Objective. We aim to develop a machine learning algorithm to quantify adipose tissue deposition at surgical sites as a function of biomaterial implantation. Impact Statement. To our knowledge, this study is the first investigation to apply convolutional neural network (CNN) models to identify and segment adipose tissue in histological images from silk fibroin biomaterial implants. Introduction. When designing biomaterials for the treatment of various soft tissue injuries and diseases, one must consider the extent of adipose tissue deposition. In this work, we analyzed adipose tissue accumulation in histological images of sectioned silk fibroin-based biomaterials excised from rodents following subcutaneous implantation for 1, 2, 4, or 8 weeks. Current strategies for quantifying adipose tissue after biomaterial implantation are often tedious and prone to human bias during analysis. Methods. We used CNN models with novel spatial histogram layer(s) that can more accurately identify and segment regions of adipose tissue in hematoxylin and eosin (H&E) and Masson's trichrome stained images, allowing for determination of the optimal biomaterial formulation. We compared the method, Jointly Optimized Spatial Histogram UNET Architecture (JOSHUA), to the baseline UNET model and an extension of the baseline model, attention UNET, as well as to versions of the models with a supplemental attention-inspired mechanism (JOSHUA+ and UNET+). Results. The inclusion of histogram layer(s) in our models shows improved performance through qualitative and quantitative evaluation. Conclusion. Our results demonstrate that the proposed methods, JOSHUA and JOSHUA+, are highly beneficial for adipose tissue identification and localization. The new histological dataset and code used in our experiments are publicly available.
Collapse
Affiliation(s)
- Joshua K. Peeples
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Julie F. Jameson
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Nisha M. Kotta
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Jonathan M. Grasman
- Department of Biomedical Engineering, New Jersey Institute of Technology, University Heights, Newark, NJ 07102, USA
| | - Whitney L. Stoppel
- Department of Chemical Engineering, University of Florida, Gainesville, FL 32611, USA
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, USA
| | - Alina Zare
- Department of Electrical and Computer Engineering, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|