1
|
Corbalan JJ, Jagadeesan P, Frietze KK, Taylor R, Gao GL, Gallagher G, Nickels JT. Humanized monoacylglycerol acyltransferase 2 mice develop metabolic dysfunction-associated steatohepatitis. J Lipid Res 2024; 65:100695. [PMID: 39505262 DOI: 10.1016/j.jlr.2024.100695] [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: 08/19/2024] [Revised: 10/01/2024] [Accepted: 10/28/2024] [Indexed: 11/08/2024] Open
Abstract
Mice lacking monoacylglycerol acyltransferase 2 (mMGAT21) are resistant to diet-induced fatty liver, suggesting hMOGAT2 inhibition is a viable option for treating metabolic dysfunction-associated steatotic liver disease (MASLD)/metabolic dysfunction-associated steatohepatitis (MASH). We generated humanized hMOGAT2 mice (HuMgat2) for use in pre-clinical studies testing the efficacy of hMOGAT2 inhibitors for treating MASLD/MASH. HuMgat2 mice developed MASH when fed a steatotic diet. Computer-aided histology revealed the presence of hepatocyte cell ballooning, immune cell infiltration, and fibrosis. Hepatocytes accumulated Mallory-Denk bodies containing phosphorylated p62/sequestosome-1-ubiquitinated protein aggregates likely caused by defects in autophagy. Metainflammation and apoptotic cell death were seen in the livers of HuMgat2 mice. Treating HuMgat2 mice with elafibranor reduced several MASH phenotypes. RNASeq analysis predicted changes in bile acid transporter expression that correlated with altered bile acid metabolism indicative of cholestasis. Our results suggest that HuMgat2 mice will serve as a pre-clinical model for testing hMOGAT2 inhibitor efficacy and toxicity and allow for the study of hMOGAT2 in the context of MASH.
Collapse
Affiliation(s)
- J Jose Corbalan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Pranavi Jagadeesan
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Karla K Frietze
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Rulaiha Taylor
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA
| | - Grace L Gao
- Department of Pharmacology and Toxicology, Earnest Mario School of Pharmacy, Rutgers University, Piscataway, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA
| | - Grant Gallagher
- Oncoveda, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA
| | - Joseph T Nickels
- The Institute of Metabolic Disorders, Genesis Research and Development Institute, Genesis Biotechnology Group, Hamilton, NJ, USA; Rutgers Center for Lipid Research, New Jersey Institute for Food, Nutrition, and Health, Rutgers University, New Brunswick, NJ, USA.
| |
Collapse
|
2
|
Kumar A, Dixit S, Srinivasan K, M D, Vincent PMDR. Personalized cancer vaccine design using AI-powered technologies. Front Immunol 2024; 15:1357217. [PMID: 39582860 PMCID: PMC11581883 DOI: 10.3389/fimmu.2024.1357217] [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: 12/17/2023] [Accepted: 09/24/2024] [Indexed: 11/26/2024] Open
Abstract
Immunotherapy has ushered in a new era of cancer treatment, yet cancer remains a leading cause of global mortality. Among various therapeutic strategies, cancer vaccines have shown promise by activating the immune system to specifically target cancer cells. While current cancer vaccines are primarily prophylactic, advancements in targeting tumor-associated antigens (TAAs) and neoantigens have paved the way for therapeutic vaccines. The integration of artificial intelligence (AI) into cancer vaccine development is revolutionizing the field by enhancing various aspect of design and delivery. This review explores how AI facilitates precise epitope design, optimizes mRNA and DNA vaccine instructions, and enables personalized vaccine strategies by predicting patient responses. By utilizing AI technologies, researchers can navigate complex biological datasets and uncover novel therapeutic targets, thereby improving the precision and efficacy of cancer vaccines. Despite the promise of AI-powered cancer vaccines, significant challenges remain, such as tumor heterogeneity and genetic variability, which can limit the effectiveness of neoantigen prediction. Moreover, ethical and regulatory concerns surrounding data privacy and algorithmic bias must be addressed to ensure responsible AI deployment. The future of cancer vaccine development lies in the seamless integration of AI to create personalized immunotherapies that offer targeted and effective cancer treatments. This review underscores the importance of interdisciplinary collaboration and innovation in overcoming these challenges and advancing cancer vaccine development.
Collapse
Affiliation(s)
- Anant Kumar
- School of Bioscience and Technology, Vellore Institute of Technology, Vellore, India
| | - Shriniket Dixit
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Kathiravan Srinivasan
- School of Computer Science and Engineering, Vellore Institute of Technology, Vellore, India
| | - Dinakaran M
- School of Computer Science and Engineering, Vellore Institute of Technology, Chennai, India
| | - P. M. Durai Raj Vincent
- School of Computer Science Engineering and Information Systems, Vellore Institute of Technology, Vellore, India
| |
Collapse
|
3
|
Chen S, Zhang P, Duan X, Bao A, Wang B, Zhang Y, Li H, Zhang L, Liu S. Lesion Localization and Pathological Diagnosis of Ovine Pulmonary Adenocarcinoma Based on MASK R-CNN. Animals (Basel) 2024; 14:2488. [PMID: 39272273 PMCID: PMC11393988 DOI: 10.3390/ani14172488] [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: 07/26/2024] [Revised: 08/21/2024] [Accepted: 08/22/2024] [Indexed: 09/15/2024] Open
Abstract
Ovine pulmonary adenocarcinoma (OPA) is a contagious lung tumour caused by the Jaagsiekte Sheep Retrovirus (JSRV). Histopathological diagnosis is the gold standard for OPA diagnosis. However, interpretation of traditional pathology images is complex and operator dependent. The mask regional convolutional neural network (Mask R-CNN) has emerged as a valuable tool in pathological diagnosis. This study utilized 54 typical OPA whole slide images (WSI) to extract 7167 typical lesion images containing OPA to construct a Common Objects in Context (COCO) dataset for OPA pathological images. The dataset was categorized into training and test sets (8:2 ratio) for model training and validation. Mean average specificity (mASp) and average sensitivity (ASe) were used to evaluate model performance. Six WSI-level pathological images (three OPA and three non-OPA images), not included in the dataset, were used for anti-peeking model validation. A random selection of 500 images, not included in the dataset establishment, was used to compare the performance of the model with assessment by pathologists. Accuracy, sensitivity, specificity, and concordance rate were evaluated. The model achieved a mASp of 0.573 and an ASe of 0.745, demonstrating effective lesion detection and alignment with expert annotation. In Anti-Peeking verification, the model showed good performance in locating OPA lesions and distinguished OPA from non-OPA pathological images. In the random 500-image diagnosis, the model achieved 92.8% accuracy, 100% sensitivity, and 88% specificity. The agreement rates between junior and senior pathologists were 100% and 96.5%, respectively. In conclusion, the Mask R-CNN-based OPA diagnostic model developed for OPA facilitates rapid and accurate diagnosis in practical applications.
Collapse
Affiliation(s)
- Sixu Chen
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Pei Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Xujie Duan
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Anyu Bao
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Buyu Wang
- College of Computer and Information Engineering, Inner Mongolia Agricultural University, Hohhot 010010, China
| | - Yufei Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Huiping Li
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Liang Zhang
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| | - Shuying Liu
- College of Veterinary Medicine, Inner Mongolia Agricultural University, Zhao Wu Da Road No. 306, Hohhot 010018, China
- Inner Mongolia Key Laboratory of Basic Veterinary Science, Hohhot 010018, China
- Key Laboratory of Clinical Diagnosis and Treatment Technology in Animal Disease, Ministry of Agriculture, Hohhot 010018, China
| |
Collapse
|
4
|
Kildal W, Cyll K, Kalsnes J, Islam R, Julbø FM, Pradhan M, Ersvær E, Shepherd N, Vlatkovic L, Tekpli X, Garred Ø, Kristensen GB, Askautrud HA, Hveem TS, Danielsen HE. Deep learning for automated scoring of immunohistochemically stained tumour tissue sections - Validation across tumour types based on patient outcomes. Heliyon 2024; 10:e32529. [PMID: 39040241 PMCID: PMC11261074 DOI: 10.1016/j.heliyon.2024.e32529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 06/05/2024] [Indexed: 07/24/2024] Open
Abstract
We aimed to develop deep learning (DL) models to detect protein expression in immunohistochemically (IHC) stained tissue-sections, and to compare their accuracy and performance with manually scored clinically relevant proteins in common cancer types. Five cancer patient cohorts (colon, two prostate, breast, and endometrial) were included. We developed separate DL models for scoring IHC-stained tissue-sections with nuclear, cytoplasmic, and membranous staining patterns. For training, we used images with annotations of cells with positive and negative staining from the colon cohort stained for Ki-67 and PMS2 (nuclear model), the prostate cohort 1 stained for PTEN (cytoplasmic model) and β-catenin (membranous model). The nuclear DL model was validated for MSH6 in the colon, MSH6 and PMS2 in the endometrium, Ki-67 and CyclinB1 in prostate, and oestrogen and progesterone receptors in the breast cancer cohorts. The cytoplasmic DL model was validated for PTEN and Mapre2, and the membranous DL model for CD44 and Flotillin1, all in prostate cohorts. When comparing the results of manual and DL scores in the validation sets, using manual scores as the ground truth, we observed an average correct classification rate of 91.5 % (76.9-98.5 %) for the nuclear model, 85.6 % (73.3-96.6 %) for the cytoplasmic model, and 78.4 % (75.5-84.3 %) for the membranous model. In survival analyses, manual and DL scores showed similar prognostic impact, with similar hazard ratios and p-values for all DL models. Our findings demonstrate that DL models offer a promising alternative to manual IHC scoring, providing efficiency and reproducibility across various data sources and markers.
Collapse
Affiliation(s)
- Wanja Kildal
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Karolina Cyll
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Joakim Kalsnes
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Rakibul Islam
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Frida M. Julbø
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Manohar Pradhan
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Elin Ersvær
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Neil Shepherd
- Gloucestershire Cellular Pathology Laboratory, Gloucester, GL53 7AN, UK
| | - Ljiljana Vlatkovic
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - OSBREAC
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
- Gloucestershire Cellular Pathology Laboratory, Gloucester, GL53 7AN, UK
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, NO-0450, Oslo, Norway
- Department of Pathology, Oslo University Hospital, NO-0424, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, OX3 9DU, UK
| | - Xavier Tekpli
- Department of Medical Genetics, Institute of Clinical Medicine, Faculty of Medicine, University of Oslo and Oslo University Hospital, NO-0450, Oslo, Norway
| | - Øystein Garred
- Department of Pathology, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Gunnar B. Kristensen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Hanne A. Askautrud
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Tarjei S. Hveem
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
| | - Håvard E. Danielsen
- Institute for Cancer Genetics and Informatics, Oslo University Hospital, NO-0424, Oslo, Norway
- Nuffield Division of Clinical Laboratory Sciences, University of Oxford, Oxford, OX3 9DU, UK
| |
Collapse
|
5
|
Osorio P, Jimenez-Perez G, Montalt-Tordera J, Hooge J, Duran-Ballester G, Singh S, Radbruch M, Bach U, Schroeder S, Siudak K, Vienenkoetter J, Lawrenz B, Mohammadi S. Latent Diffusion Models with Image-Derived Annotations for Enhanced AI-Assisted Cancer Diagnosis in Histopathology. Diagnostics (Basel) 2024; 14:1442. [PMID: 39001331 PMCID: PMC11241396 DOI: 10.3390/diagnostics14131442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 06/19/2024] [Accepted: 06/26/2024] [Indexed: 07/16/2024] Open
Abstract
Artificial Intelligence (AI)-based image analysis has immense potential to support diagnostic histopathology, including cancer diagnostics. However, developing supervised AI methods requires large-scale annotated datasets. A potentially powerful solution is to augment training data with synthetic data. Latent diffusion models, which can generate high-quality, diverse synthetic images, are promising. However, the most common implementations rely on detailed textual descriptions, which are not generally available in this domain. This work proposes a method that constructs structured textual prompts from automatically extracted image features. We experiment with the PCam dataset, composed of tissue patches only loosely annotated as healthy or cancerous. We show that including image-derived features in the prompt, as opposed to only healthy and cancerous labels, improves the Fréchet Inception Distance (FID) by 88.6. We also show that pathologists find it challenging to detect synthetic images, with a median sensitivity/specificity of 0.55/0.55. Finally, we show that synthetic data effectively train AI models.
Collapse
Affiliation(s)
- Pedro Osorio
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | | | | | - Jens Hooge
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | | | - Shivam Singh
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| | - Moritz Radbruch
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | - Ute Bach
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | | | - Krystyna Siudak
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | | | - Bettina Lawrenz
- Pathology and Clinical Pathology, Bayer AG, 13353 Berlin, Germany
| | - Sadegh Mohammadi
- Decision Science & Advanced Analytics, Bayer AG, 13353 Berlin, Germany
| |
Collapse
|
6
|
Alharbi E, Rajaram A, Côté K, Farag M, Maleki F, Gao ZH, Maedler-Kron C, Marcus V, Fiset PO. A Deep Learning-Based Approach to Estimate Paneth Cell Granule Area in Celiac Disease. Arch Pathol Lab Med 2024; 148:828-835. [PMID: 37852171 DOI: 10.5858/arpa.2023-0074-oa] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/10/2023] [Indexed: 10/20/2023]
Abstract
CONTEXT.— Changes in Paneth cell numbers can be associated with chronic inflammatory diseases of the gastrointestinal tract. So far, no consensus has been achieved on the number of Paneth cells and their relevance to celiac disease (CD). OBJECTIVES.— To compare crypt and Paneth cell granule areas between patients with CD and those without CD (non-CD) using an artificial intelligence-based solution. DESIGN.— Hematoxylin-eosin-stained sections of duodenal biopsies from 349 patients at the McGill University Health Centre were analyzed. Of these, 185 had a history of CD and 164 were controls. Slides were digitized, and NoCodeSeg, a code-free workflow using open-source software (QuPath, DeepMIB), was implemented to train deep learning models to segment crypts and Paneth cell granules. The total area of the entire analyzed tissue, epithelium, crypts, and Paneth cell granules was documented for all slides, and comparisons were performed. RESULTS.— A mean intersection-over-union score of 88.76% and 91.30% was achieved for crypt areas and Paneth cell granule segmentations, respectively. On normalization to total tissue area, the crypt to total tissue area in CD was increased and the Paneth cell granule area to total tissue area decreased when compared to non-CD controls. CONCLUSIONS.— Crypt hyperplasia was confirmed in CD compared to non-CD controls. The area of Paneth cell granules, an indirect measure of Paneth cell function, decreased with increasing severity of CD. More importantly, our study analyzed complete hematoxylin-eosin slide sections using an efficient and easy to use coding-free artificial intelligence workflow.
Collapse
Affiliation(s)
- Ebtihal Alharbi
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
- Department of Pathology, Faculty of Medicine in Rabigh, King Abdulaziz University, Saudi Arabia (Alharbi)
| | - Ajay Rajaram
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
| | - Kevin Côté
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
| | - Mina Farag
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
| | - Farhad Maleki
- Augmented Intelligence & Precision Health Laboratory, Research Institute and Department of Radiology, McGill University Health Centre, Montreal, Quebec, Canada (Maleki)
- Department of Computer Science, University of Calgary, Calgary, Alberta, Canada (Maleki)
| | - Zu-Hua Gao
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada (Gao)
| | - Chelsea Maedler-Kron
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
- Research Institute of McGill University Health Center, Montreal, Quebec, Canada (Maedler-Kron, Fiset)
| | - Victoria Marcus
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
| | - Pierre Olivier Fiset
- From the Department of Pathology, McGill University, Montreal, Quebec, Canada (Alharbi, Rajaram, Côté, Farag, Gao, Maedler-Kron, Marcus, Fiset)
- Research Institute of McGill University Health Center, Montreal, Quebec, Canada (Maedler-Kron, Fiset)
| |
Collapse
|
7
|
Al Taher RS, Abbas MA, Halahleh K, Sughayer MA. Correlation Between ImageJ and Conventional Manual Scoring Methods for Programmed Death-Ligand 1 Immuno-Histochemically Stained Sections. Technol Cancer Res Treat 2024; 23:15330338241242635. [PMID: 38562094 PMCID: PMC10989033 DOI: 10.1177/15330338241242635] [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: 09/21/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Background: One of the most frequently used methods for quantifying PD-L1 (programmed cell death-ligand 1) expression in tumor tissue is IHC (immunohistochemistry). This may predict the patient's response to anti-PD1/PD-L1 therapy in cancer. Methods: ImageJ software was used to score IHC-stained sections for PD-L1 and compare the results with the conventional manual method. Results: In diffuse large B cell lymphoma, no significant difference between the scores obtained by the conventional method and ImageJ scores obtained using the option "RGB" or "Brightness/Contrast." On the other hand, a significant difference was found between the conventional and HSB scoring methods. ImageJ faced some challenges in analyzing head and neck squamous cell carcinoma tissues because of tissue heterogenicity. A significant difference was found between the conventional and ImageJ scores using HSB or RGB but not with the "Brightness/Contrast" option. Scores obtained by ImageJ analysis after taking images using 20 × objective lens gave significantly higher readings compared to 40 × magnification. A significant difference between camera-captured images' scores and scanner whole slide images' scores was observed. Conclusion: ImageJ can be used to score homogeneous tissues. In the case of highly heterogeneous tissues, it is advised to use the conventional method rather than ImageJ scoring.
Collapse
Affiliation(s)
- Rand Suleiman Al Taher
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman, Jordan
| | - Manal A. Abbas
- Department of Medical Laboratory Sciences, Faculty of Allied Medical Sciences, Al-Ahliyya Amman University, Amman, Jordan
- Pharmacological and Diagnostic Research Laboratory, Al-Ahliyya Amman University, Amman, Jordan
| | - Khalid Halahleh
- Department of Medical Oncology, King Hussein Cancer Center, Amman, Jordan
| | - Maher A. Sughayer
- Department of Pathology and Laboratory Medicine, King Hussein Cancer Center, Amman, Jordan
| |
Collapse
|
8
|
Jesus R, Bastião Silva L, Sousa V, Carvalho L, Garcia Gonzalez D, Carias J, Costa C. Personalizable AI platform for universal access to research and diagnosis in digital pathology. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 242:107787. [PMID: 37717524 DOI: 10.1016/j.cmpb.2023.107787] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/22/2023] [Accepted: 09/01/2023] [Indexed: 09/19/2023]
Abstract
BACKGROUND AND MOTIVATION Digital pathology has been evolving over the last years, proposing significant workflow advantages that have fostered its adoption in professional environments. Patient clinical and image data are readily available in remote data banks that can be consumed efficiently over standard communication technologies. The appearance of new imaging techniques and advanced artificial intelligence algorithms has significantly reduced the burden on medical professionals by speeding up the screening process. Despite these advancements, the usage of digital pathology in professional environments has been slowed down by poor interoperability between services resulting from a lack of standard interfaces and integrative solutions. This work addresses this issue by proposing a cloud-based digital pathology platform built on standard and open interfaces. METHODS The work proposes and describes a vendor-neutral platform that provides interfaces for managing digital slides, and medical reports, and integrating digital image analysis services compatible with existing standards. The solution integrates the open-source plugin-based Dicoogle PACS for interoperability and extensibility, which grants the proposed solution great feature customization. RESULTS The solution was developed in collaboration with iPATH research project partners, including the validation by medical pathologists. The result is a pure Web collaborative framework that supports both research and production environments. A total of 566 digital slides from different pathologies were successfully uploaded to the platform. Using the integration interfaces, a mitosis detection algorithm was successfully installed into the platform, and it was trained with 2400 annotations collected from breast carcinoma images. CONCLUSION Interoperability is a key factor when discussing digital pathology solutions, as it facilitates their integration into existing institutions' information systems. Moreover, it improves data sharing and integration of third-party services such as image analysis services, which have become relevant in today's digital pathology workflow. The proposed solution fully embraces the DICOM standard for digital pathology, presenting an interoperable cloud-based solution that provides great feature customization thanks to its extensible architecture.
Collapse
Affiliation(s)
- Rui Jesus
- University of A. Coruña, A Coruña, Spain; BMD Software, Aveiro, Portugal.
| | | | - Vítor Sousa
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lina Carvalho
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | | | - João Carias
- Center for Computer Graphics, Braga, Portugal
| | - Carlos Costa
- IEETA/DETI, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
9
|
Nishimura Y, Ryo E, Inoue S, Kawazu M, Ueno T, Namikawa K, Takahashi A, Ogata D, Yoshida A, Yamazaki N, Mano H, Yatabe Y, Mori T. Strategic Approach to Heterogeneity Analysis of Cutaneous Adnexal Carcinomas Using Computational Pathology and Genomics. JID INNOVATIONS 2023; 3:100229. [PMID: 37965425 PMCID: PMC10641284 DOI: 10.1016/j.xjidi.2023.100229] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 07/30/2023] [Accepted: 08/07/2023] [Indexed: 11/16/2023] Open
Abstract
Cutaneous adnexal tumors are neoplasms that arise from skin appendages. Their morphologic diversity and phenotypic variability with rare progression to malignancy make them difficult to diagnose and classify, and there is currently no established treatment strategy. To overcome these difficulties, this study investigated the transcription factor SOX9 expression, morphology, and genetics of skin adnexal tumors for understanding their biology, especially their histogenesis. We showed that cutaneous adnexal tumors and their nontumor counterparts of skin and appendages exhibit expression patterns similar to that of SOX9. Its expression intensity and pattern, as well as histopathologic evaluation of tumors, were analyzed using digital images of 69 normal skin adnexal 9-type organs and 185 skin adnexal 29-type tumors as references. It was possible to distinguish basal cell carcinoma from squamous cell carcinoma, sebaceous carcinoma, and pilomatrixoma with significant differences, along with porocarcinoma from squamous cell carcinoma. Furthermore, unsupervised machine learning "computational pathology" was used to derive a multiregion whole-exome sequencing fusion method termed "genocomputed pathology." The genocomputed pathology of three representable adnexal carcinomas (porocarcinoma, hidradenocarcinoma, and spiradenocarcinoma) was evaluated for total nine cases. We showed that there was more heterogeneity than expected within the tumors as well as the coexistence of components lacking driver fusion genes. The presence or absence of potential driver genes, such as PIK3CA, YAP1, and PTEN, in each region was identified, highlighting a therapeutic strategy for cutaneous adnexal carcinoma encompassing heterogeneous tumors.
Collapse
Affiliation(s)
- Yuuki Nishimura
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Eijitsu Ryo
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Satoshi Inoue
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Masahito Kawazu
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Toshihide Ueno
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Kenjiro Namikawa
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akira Takahashi
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Dai Ogata
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Akihiko Yoshida
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoya Yamazaki
- Dermatologic Oncology, National Cancer Center Hospital, Tokyo, Japan
| | - Hiroyuki Mano
- Division of Cellular Signaling, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Yasushi Yatabe
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Course of Advanced Clinical Research of Cancer, Juntendo University Graduate School of Medicine, Tokyo, Japan
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| | - Taisuke Mori
- Department of Diagnostic Pathology, National Cancer Center Hospital, Tokyo, Japan
- Division of Molecular Pathology, National Cancer Center Reserch Institute, Tokyo, Japan
| |
Collapse
|
10
|
Baek EB, Lee J, Hwang JH, Park H, Lee BS, Kim YB, Jun SY, Her J, Son HY, Cho JW. Application of multiple-finding segmentation utilizing Mask R-CNN-based deep learning in a rat model of drug-induced liver injury. Sci Rep 2023; 13:17555. [PMID: 37845356 PMCID: PMC10579263 DOI: 10.1038/s41598-023-44897-8] [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: 03/22/2023] [Accepted: 10/13/2023] [Indexed: 10/18/2023] Open
Abstract
Drug-induced liver injury (DILI) presents significant diagnostic challenges, and recently artificial intelligence-based deep learning technology has been used to predict various hepatic findings. In this study, we trained a set of Mask R-CNN-based deep algorithms to learn and quantify typical toxicant induced-histopathological lesions, portal area, and connective tissue in Sprague Dawley rats. We compared a set of single-finding models (SFMs) and a combined multiple-finding model (MFM) for their ability to simultaneously detect, classify, and quantify multiple hepatic findings on rat liver slide images. All of the SFMs yielded mean average precision (mAP) values above 85%, suggesting that the models had been successfully established. The MFM showed better performance than the SFMs, with a total mAP value of 92.46%. We compared the model predictions for slide images with ground-truth annotations generated by an accredited pathologist. For the MFM, the overall and individual finding predictions were highly correlated with the annotated areas, with R-squared values of 0.852, 0.952, 0.999, 0.990, and 0.958 being obtained for portal area, infiltration, necrosis, vacuolation, and connective tissue (including fibrosis), respectively. Our results indicate that the proposed MFM could be a useful tool for detecting and predicting multiple hepatic findings in basic non-clinical study settings.
Collapse
Affiliation(s)
- Eun Bok Baek
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea
| | - Jaeku Lee
- Research and Development Team, LAC Inc, Seoul, Republic of Korea
| | - Ji-Hee Hwang
- Toxicologic Pathology Research Group, Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Heejin Park
- Toxicologic Pathology Research Group, Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Byoung-Seok Lee
- Toxicologic Pathology Research Group, Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea
| | - Yong-Bum Kim
- Department of Advanced Toxicology Research, Korea Institute of Toxicology, Daejeon, 34114, Republic of Korea
| | - Sang-Yeop Jun
- Research and Development Team, LAC Inc, Seoul, Republic of Korea
| | - Jun Her
- Research and Development Team, LAC Inc, Seoul, Republic of Korea
| | - Hwa-Young Son
- College of Veterinary Medicine, Chungnam National University, 99 Daehak-ro, Yuseong-gu, Daejeon, 34134, Republic of Korea.
| | - Jae-Woo Cho
- Toxicologic Pathology Research Group, Department of Advanced Toxicology Research, Korea Institute of Toxicology, 141 Gajeong-ro, Yuseong-gu, Daejeon, 34114, Republic of Korea.
| |
Collapse
|
11
|
Rudmann DG, Bertrand L, Zuraw A, Deiters J, Staup M, Rivenson Y, Kuklyte J. Building a nonclinical pathology laboratory of the future for pharmaceutical research excellence. Drug Discov Today 2023; 28:103747. [PMID: 37598916 DOI: 10.1016/j.drudis.2023.103747] [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: 07/12/2023] [Revised: 08/02/2023] [Accepted: 08/14/2023] [Indexed: 08/22/2023]
Abstract
We describe a roadmap for a fully digital artificial intelligence (AI)-augmented nonclinical pathology laboratory across three continents. Underpinning the design are Good Laboratory Practice (GLP)-validated laboratory information management systems (LIMS), whole slide-scanners (WSS), image management systems (IMS), and a digital microscope intended for use by the nonclinical pathologist. Digital diagnostics are supported by tools that include AI-based virtual staining and deep learning-based decision support. Implemented during the COVID-19 pandemic, the initial digitized workflow largely mitigated disruption of pivotal nonclinical studies required to support pharmaceutical clinical testing. We believe that this digital transformation of our nonclinical pathology laboratories will promote efficiency and innovation in the future and enhance the quality and speed of drug development decision making.
Collapse
Affiliation(s)
- D G Rudmann
- Charles River Laboratories, Digital Toxicologic Pathology, Discovery and Safety Assessment, Wilmington, DE, USA.
| | - L Bertrand
- Charles River Laboratories, Digital Toxicologic Pathology, Discovery and Safety Assessment, Wilmington, DE, USA
| | - A Zuraw
- Charles River Laboratories, Digital Toxicologic Pathology, Discovery and Safety Assessment, Wilmington, DE, USA
| | - J Deiters
- Charles River Laboratories, Digital Toxicologic Pathology, Discovery and Safety Assessment, Wilmington, DE, USA
| | - M Staup
- Charles River Laboratories, Digital Toxicologic Pathology, Discovery and Safety Assessment, Wilmington, DE, USA
| | | | | |
Collapse
|
12
|
Swillens JEM, Nagtegaal ID, Engels S, Lugli A, Hermens RPMG, van der Laak JAWM. Pathologists' first opinions on barriers and facilitators of computational pathology adoption in oncological pathology: an international study. Oncogene 2023; 42:2816-2827. [PMID: 37587332 PMCID: PMC10504072 DOI: 10.1038/s41388-023-02797-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 07/26/2023] [Indexed: 08/18/2023]
Abstract
Computational pathology (CPath) algorithms detect, segment or classify cancer in whole slide images, approaching or even exceeding the accuracy of pathologists. Challenges have to be overcome before these algorithms can be used in practice. We therefore aim to explore international perspectives on the future role of CPath in oncological pathology by focusing on opinions and first experiences regarding barriers and facilitators. We conducted an international explorative eSurvey and semi-structured interviews with pathologists utilizing an implementation framework to classify potential influencing factors. The eSurvey results showed remarkable variation in opinions regarding attitude, understandability and validation of CPath. Interview results showed that barriers focused on the quality of available evidence, while most facilitators concerned strengths of CPath. A lack of consensus was present for multiple factors, such as the determination of sufficient validation using CPath, the preferred function of CPath within the digital workflow and the timing of CPath introduction in pathology education. The diversity in opinions illustrates variety in influencing factors in CPath adoption. A next step would be to quantitatively determine important factors for adoption and initiate validation studies. Both should include clear case descriptions and be conducted among a more homogenous panel of pathologists based on sub specialization.
Collapse
Affiliation(s)
- Julie E M Swillens
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands.
| | - Iris D Nagtegaal
- Department of Pathology, Radboud Institute for Molecular Life Sciences (RIMLS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Sam Engels
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | | | - Rosella P M G Hermens
- Scientific Center for Quality of Healthcare (IQ Healthcare), Radboud Institute for Health Sciences (RIHS), Radboud University Medical Centre, Nijmegen, The Netherlands
| | | |
Collapse
|
13
|
Park SM, Chen CJJ, Mathy JE, Lin SCY, Martin RCW, Mathy JA, Dunbar PR. Seven-colour multiplex immunochemistry/immunofluorescence and whole slide imaging of frozen sections. J Immunol Methods 2023:113490. [PMID: 37172777 DOI: 10.1016/j.jim.2023.113490] [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: 04/03/2023] [Revised: 05/09/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
Multiplex Immunochemistry/Immunofluorescence (mIHC/IF) aims to visualise multiple biomarkers in a single tissue section and is especially powerful when used on slide scanners coupled with digital analysis tools. mIHC/IF is commonly employed in immuno-oncology to characterise features of the tumour microenvironment (TME) and correlate them with clinical parameters to guide prognostication and therapy. However, mIHC/IF can be applied to a wide range of organisms in any physiological or disease context. Recent innovation has extended the number of markers that can be detected using slide scanners well beyond the 3-4 markers typically reported in traditional fluorescence microscopy. However, these methods often require sequential antibody staining and stripping, and are not compatible with frozen tissue sections. Using fluorophore-conjugated antibodies, we have established a simple mIHC/IF imaging workflow that enables simultaneous staining and detection of seven markers in a single section of frozen tissue. Coupled with automated whole slide imaging and digital quantification, our data efficiently revealed the tumour-immune complexity in metastatic melanoma. Computational image analysis quantified the immune and stromal cell populations present in the TME as well as their spatial interactions. This imaging workflow can also be performed with an indirect labelling panel consisting of primary and secondary antibodies. Our new methods, combined with digital quantification, will provide a valuable tool for high-quality mIHC/IF assays in immuno-oncology research and other translational studies, especially in circumstances where frozen sections are required for detection of particular markers, or for applications where frozen sections may be preferred, such as spatial transcriptomics.
Collapse
Affiliation(s)
- Saem Mul Park
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Chun-Jen J Chen
- School of Biological Sciences, University of Auckland, Auckland, New Zealand
| | - Joanna E Mathy
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | - Shelly C Y Lin
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand
| | | | - Jon A Mathy
- Department of Surgery, Faculty of Medical Health Sciences, Waipapa Taumata Rau - The University of Auckland, Auckland, New Zealand; Auckland Regional Plastic, Reconstructive & Hand Surgery Unit, Auckland, New Zealand
| | - P Rod Dunbar
- School of Biological Sciences, University of Auckland, Auckland, New Zealand; Maurice Wilkins Centre, University of Auckland, Auckland, New Zealand.
| |
Collapse
|
14
|
Huss R, Raffler J, Märkl B. Artificial intelligence and digital biomarker in precision pathology guiding immune therapy selection and precision oncology. Cancer Rep (Hoboken) 2023:e1796. [PMID: 36813293 PMCID: PMC10363837 DOI: 10.1002/cnr2.1796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 01/15/2023] [Accepted: 02/09/2023] [Indexed: 02/24/2023] Open
Abstract
BACKGROUND The currently available immunotherapies already changed the strategy how many cancers are treated from first to last line. Understanding even the most complex heterogeneity in tumor tissue and mapping the spatial cartography of the tumor immunity allows the best and optimized selection of immune modulating agents to (re-)activate the patient's immune system and direct it against the individual cancer in the most effective way. RECENT FINDINGS Primary cancer and metastases maintain a high degree of plasticity to escape any immune surveillance and continue to evolve depending on many intrinsic and extrinsic factors In the field of immune-oncology (IO) immune modulating agents are recognized as practice changing therapeutic modalities. Recent studies have shown that an optimal and lasting efficacy of IO therapeutics depends on the understanding of the spatial communication network and functional context of immune and cancer cells within the tumor microenvironment. Artificial intelligence (AI) provides an insight into the immune-cancer-network through the visualization of very complex tumor and immune interactions in cancer tissue specimens and allows the computer-assisted development and clinical validation of such digital biomarker. CONCLUSIONS The successful implementation of AI-supported digital biomarker solutions guides the clinical selection of effective immune therapeutics based on the retrieval and visualization of spatial and contextual information from cancer tissue images and standardized data. As such, computational pathology (CP) turns into "precision pathology" delivering individual therapy response prediction. Precision Pathology does not only include digital and computational solutions but also high levels of standardized processes in the routine histopathology workflow and the use of mathematical tools to support clinical and diagnostic decisions as the basic principle of a "precision oncology".
Collapse
Affiliation(s)
- Ralf Huss
- Medical Faculty University Augsburg, Augsburg, Germany
- Institute for Digital Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Johannes Raffler
- Institute for Digital Medicine, University Hospital Augsburg, Augsburg, Germany
| | - Bruno Märkl
- Medical Faculty University Augsburg, Augsburg, Germany
| |
Collapse
|
15
|
Kindler C, Elfwing S, Öhrvik J, Nikberg M. A Deep Neural Network-Based Decision Support Tool for the Detection of Lymph Node Metastases in Colorectal Cancer Specimens. Mod Pathol 2023; 36:100015. [PMID: 36853787 DOI: 10.1016/j.modpat.2022.100015] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/13/2022] [Accepted: 09/16/2022] [Indexed: 01/11/2023]
Abstract
The identification of lymph node metastases in colorectal cancer (CRC) specimens is crucial for the planning of postoperative treatment and can be a time-consuming task for pathologists. In this study, we developed a deep neural network (DNN) algorithm for the detection of metastatic CRC in digitized histologic sections of lymph nodes and evaluated its performance as a diagnostic support tool. First, the DNN algorithm was trained using pixel-level annotations of cancerous areas on 758 whole slide images (360 with cancerous areas). The algorithm's performance was evaluated on 74 whole slide images (43 with cancerous areas). Second, the algorithm was evaluated as a decision support tool on 288 whole slide images covering 1517 lymph node sections, randomized in 16 batches. Two senior pathologists (C.K. and C.O.) assessed each batch with and without the help of the algorithm in a 2 × 2 crossover design, with a washout period of 1 month in between. The time needed for the evaluation of each node section was recorded. The DNN algorithm achieved a median pixel-level accuracy of 0.952 on slides with cancerous areas and 0.996 on slides with benign samples. N+ disease (metastases, micrometastases, or tumor deposits) was present in 103 of the 1517 sections. The algorithm highlighted cancerous areas in 102 of these sections, with a sensitivity of 0.990. Assisted by the algorithm, the median time needed for evaluation was significantly shortened for both pathologists (median time for pathologist 1, 26 vs 14 seconds; P < .001; 95% CI, 11.0-12.0; median time for pathologist 2, 25 vs 23 seconds; P < .001; 95% CI, 2.0-4.0). Our DNN showed high accuracy for detecting metastatic CRC in digitized histologic sections of lymph nodes. This decision support tool has the potential to improve the diagnostic workflow by shortening the time needed for the evaluation of lymph nodes in CRC specimens without impairing diagnostic accuracy.
Collapse
Affiliation(s)
- Csaba Kindler
- Department of Pathology, Laboratory Medicine, Västmanlands Hospital, Västerås, Sweden; Centre for Clinical Research, Uppsala University, Västerås, Sweden.
| | | | - John Öhrvik
- Centre for Clinical Research, Uppsala University, Västerås, Sweden
| | - Maziar Nikberg
- Centre for Clinical Research, Uppsala University, Västerås, Sweden; Department of Surgery, Västmanlands Hospital, Västerås, Sweden
| |
Collapse
|
16
|
Li Z, Li B, Eliceiri KW, Narayanan V. Computationally efficient adaptive decompression for whole slide image processing. BIOMEDICAL OPTICS EXPRESS 2023; 14:667-686. [PMID: 36874494 PMCID: PMC9979681 DOI: 10.1364/boe.477515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 12/16/2022] [Accepted: 12/17/2022] [Indexed: 06/18/2023]
Abstract
Whole slide image (WSI) analysis is increasingly being adopted as an important tool in modern pathology. Recent deep learning-based methods have achieved state-of-the-art performance on WSI analysis tasks such as WSI classification, segmentation, and retrieval. However, WSI analysis requires a significant amount of computation resources and computation time due to the large dimensions of WSIs. Most of the existing analysis approaches require the complete decompression of the whole image exhaustively, which limits the practical usage of these methods, especially for deep learning-based workflows. In this paper, we present compression domain processing-based computation efficient analysis workflows for WSIs classification that can be applied to state-of-the-art WSI classification models. The approaches leverage the pyramidal magnification structure of WSI files and compression domain features that are available from the raw code stream. The methods assign different decompression depths to the patches of WSIs based on the features directly retained from compressed patches or partially decompressed patches. Patches from the low-magnification level are screened by attention-based clustering, resulting in different decompression depths assigned to the high-magnification level patches at different locations. A finer-grained selection based on compression domain features from the file code stream is applied to select further a subset of the high-magnification patches that undergo a full decompression. The resulting patches are fed to the downstream attention network for final classification. Computation efficiency is achieved by reducing unnecessary access to the high zoom level and expensive full decompression. With the number of decompressed patches reduced, the time and memory costs of downstream training and inference procedures are also significantly reduced. Our approach achieves a 7.2× overall speedup, and the memory cost is reduced by 1.1 orders of magnitudes, while the resulting model accuracy is comparable to the original workflow.
Collapse
Affiliation(s)
- Zheyu Li
- Department of Computer Science and Engineering, Pennsylvania State University, State College, PA 16801, USA
- Authors contributed equally
| | - Bin Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Authors contributed equally
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53706, USA
- Morgridge Institute for Research, Madison, WI 53715, USA
- Authors contributed equally
| | - Vijaykrishnan Narayanan
- Department of Computer Science and Engineering, Pennsylvania State University, State College, PA 16801, USA
- Authors contributed equally
| |
Collapse
|
17
|
Ko YS, Choi YM, Kim M, Park Y, Ashraf M, Quiñones Robles WR, Kim MJ, Jang J, Yun S, Hwang Y, Jang H, Yi MY. Improving quality control in the routine practice for histopathological interpretation of gastrointestinal endoscopic biopsies using artificial intelligence. PLoS One 2022; 17:e0278542. [PMID: 36520777 PMCID: PMC9754254 DOI: 10.1371/journal.pone.0278542] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 11/18/2022] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Colorectal and gastric cancer are major causes of cancer-related deaths. In Korea, gastrointestinal (GI) endoscopic biopsy specimens account for a high percentage of histopathologic examinations. Lack of a sufficient pathologist workforce can cause an increase in human errors, threatening patient safety. Therefore, we developed a digital pathology total solution combining artificial intelligence (AI) classifier models and pathology laboratory information system for GI endoscopic biopsy specimens to establish a post-analytic daily fast quality control (QC) system, which was applied in clinical practice for a 3-month trial run by four pathologists. METHODS AND FINDINGS Our whole slide image (WSI) classification framework comprised patch-generator, patch-level classifier, and WSI-level classifier. The classifiers were both based on DenseNet (Dense Convolutional Network). In laboratory tests, the WSI classifier achieved accuracy rates of 95.8% and 96.0% in classifying histopathological WSIs of colorectal and gastric endoscopic biopsy specimens, respectively, into three classes (Negative for dysplasia, Dysplasia, and Malignant). Classification by pathologic diagnosis and AI prediction were compared and daily reviews were conducted, focusing on discordant cases for early detection of potential human errors by the pathologists, allowing immediate correction, before the pathology report error is conveyed to the patients. During the 3-month AI-assisted daily QC trial run period, approximately 7-10 times the number of slides compared to that in the conventional monthly QC (33 months) were reviewed by pathologists; nearly 100% of GI endoscopy biopsy slides were double-checked by the AI models. Further, approximately 17-30 times the number of potential human errors were detected within an average of 1.2 days. CONCLUSIONS The AI-assisted daily QC system that we developed and established demonstrated notable improvements in QC, in quantitative, qualitative, and time utility aspects. Ultimately, we developed an independent AI-assisted post-analytic daily fast QC system that was clinically applicable and influential, which could enhance patient safety.
Collapse
Affiliation(s)
- Young Sin Ko
- Pathology Center, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Yoo Mi Choi
- Pathology Center, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Mujin Kim
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Youngjin Park
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Murtaza Ashraf
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Willmer Rafell Quiñones Robles
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| | - Min-Ju Kim
- Department of Pathology, Incheon Sejong Hospital, Incheon, Republic of Korea
| | - Jiwook Jang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Seokju Yun
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Yuri Hwang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Hani Jang
- AI Research Team, Digital Innovation Sector, Seegene Medical Foundation, Seoul, Republic of Korea
| | - Mun Yong Yi
- Graduate School of Data Science, Department of Industrial & Systems Engineering, Korea Advanced Institute of Science and Technology, Daejeon, Republic of Korea
| |
Collapse
|
18
|
Li B, Nelson MS, Savari O, Loeffler AG, Eliceiri KW. Differentiation of pancreatic ductal adenocarcinoma and chronic pancreatitis using graph neural networks on histopathology and collagen fiber features. J Pathol Inform 2022; 13:100158. [PMID: 36605110 PMCID: PMC9808020 DOI: 10.1016/j.jpi.2022.100158] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 11/16/2022] [Accepted: 11/16/2022] [Indexed: 11/21/2022] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is one of the most lethal human cancers. However, the symptoms and radiographic appearance of chronic pancreatitis (CP) mimics that of PDAC, and sometimes the 2 entities can also be difficult to differentiate microscopically. The need for accurate differentiation of PDAC and CP has become a major topic in pancreatic pathology. These 2 diseases can present similar histomorphological features, such as excessive deposition of fibrotic stroma in the tissue microenvironment and inflammatory cell infiltration. In this paper, we present a quantitative analysis pipeline empowered by graph neural networks (GNN) capable of automatic detection and differentiation of PDAC and CP in human histological specimens. Modeling histological images as graphs and deploying graph convolutions can enable the capture of histomorphological features at different scales, ranging from nuclear size to the organization of ducts. The analysis pipeline combines image features computed from co-registered hematoxylin and eosin (H&E) images and Second-Harmonic Generation (SHG) microscopy images, with the SHG images enabling the extraction of collagen fiber morphological features. Evaluating the analysis pipeline on a human tissue micro-array dataset consisting of 786 cores and a tissue region dataset consisting of 268 images, it attained 86.4% accuracy with an average area under the curve (AUC) of 0.954 and 88.9% accuracy with an average AUC of 0.957, respectively. Moreover, incorporating topological features of collagen fibers computed from SHG images into the model further increases the classification accuracy on the tissue region dataset to 91.3% with an average AUC of 0.962, suggesting that collagen characteristics are diagnostic features in PDAC and CP detection and differentiation.
Collapse
Affiliation(s)
- Bin Li
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison 53706, WI, USA
- Morgridge Institute for Research, Madison 53705, WI, USA
| | - Michael S. Nelson
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison 53706, WI, USA
| | - Omid Savari
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh 15213, PA, USA
| | - Agnes G. Loeffler
- Department of Pathology, MetroHealth Medical Center, Cleveland 44109, OH, USA
| | - Kevin W. Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison 53706, WI, USA
- Morgridge Institute for Research, Madison 53705, WI, USA
- Department of Medical Physics, University of Wisconsin-Madison, Madison 53706, WI, USA
| |
Collapse
|
19
|
Kim I, Kang K, Song Y, Kim TJ. Application of Artificial Intelligence in Pathology: Trends and Challenges. Diagnostics (Basel) 2022; 12:2794. [PMID: 36428854 PMCID: PMC9688959 DOI: 10.3390/diagnostics12112794] [Citation(s) in RCA: 35] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 11/03/2022] [Accepted: 11/11/2022] [Indexed: 11/16/2022] Open
Abstract
Given the recent success of artificial intelligence (AI) in computer vision applications, many pathologists anticipate that AI will be able to assist them in a variety of digital pathology tasks. Simultaneously, tremendous advancements in deep learning have enabled a synergy with artificial intelligence (AI), allowing for image-based diagnosis on the background of digital pathology. There are efforts for developing AI-based tools to save pathologists time and eliminate errors. Here, we describe the elements in the development of computational pathology (CPATH), its applicability to AI development, and the challenges it faces, such as algorithm validation and interpretability, computing systems, reimbursement, ethics, and regulations. Furthermore, we present an overview of novel AI-based approaches that could be integrated into pathology laboratory workflows.
Collapse
Affiliation(s)
- Inho Kim
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Kyungmin Kang
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Youngjae Song
- College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Republic of Korea
| | - Tae-Jung Kim
- Department of Hospital Pathology, Yeouido St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, 10, 63-ro, Yeongdeungpo-gu, Seoul 07345, Republic of Korea
| |
Collapse
|
20
|
Su A, Lee H, Tan X, Suarez CJ, Andor N, Nguyen Q, Ji HP. A deep learning model for molecular label transfer that enables cancer cell identification from histopathology images. NPJ Precis Oncol 2022; 6:14. [PMID: 35236916 PMCID: PMC8891271 DOI: 10.1038/s41698-022-00252-0] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 12/16/2021] [Indexed: 12/19/2022] Open
Abstract
Deep-learning classification systems have the potential to improve cancer diagnosis. However, development of these computational approaches so far depends on prior pathological annotations and large training datasets. The manual annotation is low-resolution, time-consuming, highly variable and subject to observer variance. To address this issue, we developed a method, H&E Molecular neural network (HEMnet). HEMnet utilizes immunohistochemistry as an initial molecular label for cancer cells on a H&E image and trains a cancer classifier on the overlapping clinical histopathological images. Using this molecular transfer method, HEMnet successfully generated and labeled 21,939 tumor and 8782 normal tiles from ten whole-slide images for model training. After building the model, HEMnet accurately identified colorectal cancer regions, which achieved 0.84 and 0.73 of ROC AUC values compared to p53 staining and pathological annotations, respectively. Our validation study using histopathology images from TCGA samples accurately estimated tumor purity, which showed a significant correlation (regression coefficient of 0.8) with the estimation based on genomic sequencing data. Thus, HEMnet contributes to addressing two main challenges in cancer deep-learning analysis, namely the need to have a large number of images for training and the dependence on manual labeling by a pathologist. HEMnet also predicts cancer cells at a much higher resolution compared to manual histopathologic evaluation. Overall, our method provides a path towards a fully automated delineation of any type of tumor so long as there is a cancer-oriented molecular stain available for subsequent learning. Software, tutorials and interactive tools are available at: https://github.com/BiomedicalMachineLearning/HEMnet.
Collapse
Affiliation(s)
- Andrew Su
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - HoJoon Lee
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Xiao Tan
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia
| | - Carlos J Suarez
- Department of Pathology, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Noemi Andor
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
- Department of Integrated Mathematical Oncology, Moffitt Cancer Center, 12902 Magnolia Drive, Tampa, FL, 33612, USA
| | - Quan Nguyen
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD, 4072, Australia.
| | - Hanlee P Ji
- Division of Oncology, Department of Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA.
- Stanford Genome Technology Center, Stanford University, Palo Alto, CA, 94304, USA.
| |
Collapse
|
21
|
Coulter C, McKay F, Hallowell N, Browning L, Colling R, Macklin P, Sorell T, Aslam M, Bryson G, Treanor D, Verrill C. Understanding the ethical and legal considerations of Digital Pathology. JOURNAL OF PATHOLOGY CLINICAL RESEARCH 2022; 8:101-115. [PMID: 34796679 PMCID: PMC8822384 DOI: 10.1002/cjp2.251] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 09/12/2021] [Accepted: 10/12/2021] [Indexed: 12/21/2022]
Abstract
Digital Pathology (DP) is a platform which has the potential to develop a truly integrated and global pathology community. The generation of DP data at scale creates novel challenges for the histopathology community in managing, processing, and governing the use of these data. The current understanding of, and confidence in, the legal and ethical aspects of DP by pathologists is unknown. We developed an electronic survey (e-survey), comprising 22 questions, with input from the Royal College of Pathologists (RCPath) Digital Pathology Working Group. The e-survey was circulated via e-mail and social media (Twitter) through the RCPath Digital Pathology Working Group network, RCPath Trainee Committee network, the Pathology image data Lake for Analytics, Knowledge and Education (PathLAKE) digital pathology consortium, National Pathology Imaging Co-operative (NPIC), local contacts, and to the membership of both The Pathological Society of Great Britain and Ireland and the British Division of the International Academy of Pathology (BDIAP). Between 14 July 2020 and 6 September 2020, we collected 198 responses representing a cross section of histopathologists, including individuals with experience of DP research. We ascertained that, in the UK, DP is being used for diagnosis, research, and teaching, and that the platform is enabling data sharing. Our survey demonstrated that there is often a lack of confidence and understanding of the key issues of consent, legislation, and ethical guidelines. Of 198 respondents, 82 (41%) did not know when the use of digital scanned slide images would fall under the relevant legislation and 93 (47%) were 'Not confident at all' in their interpretation of consent for scanned slide images in research. With increasing uptake of DP, a working knowledge of these areas is essential but histopathologists often express a lack of confidence in these topics. The need for specific training in these areas is highlighted by the findings of this study.
Collapse
Affiliation(s)
- Cheryl Coulter
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.,Nuffield Division of Clinical Laboratory Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Francis McKay
- The Wellcome Centre for Ethics and Humanities and the Ethox Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Nina Hallowell
- The Wellcome Centre for Ethics and Humanities and the Ethox Centre, Nuffield Department of Population Health, University of Oxford, Oxford, UK
| | - Lisa Browning
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Richard Colling
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.,Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| | - Philip Macklin
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK
| | - Tom Sorell
- Department of Politics and International Studies, University of Warwick, Coventry, UK
| | - Muhammad Aslam
- Department of Histopathology, Glangwilli Hospital, Hywel Dda University Health Board, Carmarthen, Wales, UK
| | - Gareth Bryson
- Department of Pathology, Queen Elizabeth University Hospital, NHS Greater Glasgow and Clyde, Glasgow, Scotland, UK
| | - Darren Treanor
- Department of Pathology, Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Clare Verrill
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.,NIHR Oxford Biomedical Research Centre, Oxford University Hospitals NHS Foundation Trust, John Radcliffe Hospital, Oxford, UK.,Nuffield Department of Surgical Sciences, University of Oxford, John Radcliffe Hospital, Oxford, UK
| |
Collapse
|
22
|
A new lightweight convolutional neural network for radiation-induced liver disease classification. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103463] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
23
|
Tran KA, Kondrashova O, Bradley A, Williams ED, Pearson JV, Waddell N. Deep learning in cancer diagnosis, prognosis and treatment selection. Genome Med 2021; 13:152. [PMID: 34579788 PMCID: PMC8477474 DOI: 10.1186/s13073-021-00968-x] [Citation(s) in RCA: 318] [Impact Index Per Article: 79.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 09/12/2021] [Indexed: 12/13/2022] Open
Abstract
Deep learning is a subdiscipline of artificial intelligence that uses a machine learning technique called artificial neural networks to extract patterns and make predictions from large data sets. The increasing adoption of deep learning across healthcare domains together with the availability of highly characterised cancer datasets has accelerated research into the utility of deep learning in the analysis of the complex biology of cancer. While early results are promising, this is a rapidly evolving field with new knowledge emerging in both cancer biology and deep learning. In this review, we provide an overview of emerging deep learning techniques and how they are being applied to oncology. We focus on the deep learning applications for omics data types, including genomic, methylation and transcriptomic data, as well as histopathology-based genomic inference, and provide perspectives on how the different data types can be integrated to develop decision support tools. We provide specific examples of how deep learning may be applied in cancer diagnosis, prognosis and treatment management. We also assess the current limitations and challenges for the application of deep learning in precision oncology, including the lack of phenotypically rich data and the need for more explainable deep learning models. Finally, we conclude with a discussion of how current obstacles can be overcome to enable future clinical utilisation of deep learning.
Collapse
Affiliation(s)
- Khoa A. Tran
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, 4059 Australia
| | - Olga Kondrashova
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| | - Andrew Bradley
- Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, 4000 Australia
| | - Elizabeth D. Williams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, 4059 Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, 4102 Australia
| | - John V. Pearson
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| | - Nicola Waddell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| |
Collapse
|
24
|
Zuraw A, Aeffner F. Whole-slide imaging, tissue image analysis, and artificial intelligence in veterinary pathology: An updated introduction and review. Vet Pathol 2021; 59:6-25. [PMID: 34521285 DOI: 10.1177/03009858211040484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since whole-slide imaging has been commercially available for over 2 decades, digital pathology has become a constantly expanding aspect of the pathology profession that will continue to significantly impact how pathologists conduct their craft. While some aspects, such as whole-slide imaging for archiving, consulting, and teaching, have gained broader acceptance, other facets such as quantitative tissue image analysis and artificial intelligence-based assessments are still met with some reservations. While most vendors in this space have focused on diagnostic applications, that is, viewing one or few slides at a time, some are developing solutions tailored more specifically to the various aspects of veterinary pathology including updated diagnostic, discovery, and research applications. This has especially advanced the use of digital pathology in toxicologic pathology and drug development, for primary reads as well as peer reviews. It is crucial that pathologists gain a deeper understanding of digital pathology and tissue image analysis technology and their applications in order to fully use these tools in a way that enhances and improves the pathologist's assessment as well as work environment. This review focuses on an updated introduction to the basics of digital pathology and image analysis and introduces emerging topics around artificial intelligence and machine learning.
Collapse
Affiliation(s)
| | - Famke Aeffner
- Amgen Inc, Amgen Research, South San Francisco, CA, USA
| |
Collapse
|
25
|
[Standardized diagnosis of pancreatic head carcinoma]. DER PATHOLOGE 2021; 42:453-463. [PMID: 34357472 DOI: 10.1007/s00292-021-00971-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Most pancreatic ductal adenocarcinomas are localized in the pancreatic head. Due to the complex anatomic relationships with the surrounding organs and vascular structures in the retroperitoneal space and to the presence of numerous transection margins and dissection planes, pancreatic head resections belong to the most complex specimens concerning grossing and sampling for histopathologic analysis.Here we discuss current guidelines for standardized grossing and reporting of pancreatic cancer, with special reference to the assessment of the resection margin status. The importance of standardized reporting for the sake of completeness, comprehensibility, comparability, and quality control as well as for the integration of pathology reports in interdisciplinary digital workflows and artificial intelligence applications will be emphasized.
Collapse
|
26
|
Wharton KA, Wood D, Manesse M, Maclean KH, Leiss F, Zuraw A. Tissue Multiplex Analyte Detection in Anatomic Pathology - Pathways to Clinical Implementation. Front Mol Biosci 2021; 8:672531. [PMID: 34386519 PMCID: PMC8353449 DOI: 10.3389/fmolb.2021.672531] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 07/14/2021] [Indexed: 12/12/2022] Open
Abstract
Background: Multiplex tissue analysis has revolutionized our understanding of the tumor microenvironment (TME) with implications for biomarker development and diagnostic testing. Multiplex labeling is used for specific clinical situations, but there remain barriers to expanded use in anatomic pathology practice. Methods: We review immunohistochemistry (IHC) and related assays used to localize molecules in tissues, with reference to United States regulatory and practice landscapes. We review multiplex methods and strategies used in clinical diagnosis and in research, particularly in immuno-oncology. Within the framework of assay design and testing phases, we examine the suitability of multiplex immunofluorescence (mIF) for clinical diagnostic workflows, considering its advantages and challenges to implementation. Results: Multiplex labeling is poised to radically transform pathologic diagnosis because it can answer questions about tissue-level biology and single-cell phenotypes that cannot be addressed with traditional IHC biomarker panels. Widespread implementation will require improved detection chemistry, illustrated by InSituPlex technology (Ultivue, Inc., Cambridge, MA) that allows coregistration of hematoxylin and eosin (H&E) and mIF images, greater standardization and interoperability of workflow and data pipelines to facilitate consistent interpretation by pathologists, and integration of multichannel images into digital pathology whole slide imaging (WSI) systems, including interpretation aided by artificial intelligence (AI). Adoption will also be facilitated by evidence that justifies incorporation into clinical practice, an ability to navigate regulatory pathways, and adequate health care budgets and reimbursement. We expand the brightfield WSI system “pixel pathway” concept to multiplex workflows, suggesting that adoption might be accelerated by data standardization centered on cell phenotypes defined by coexpression of multiple molecules. Conclusion: Multiplex labeling has the potential to complement next generation sequencing in cancer diagnosis by allowing pathologists to visualize and understand every cell in a tissue biopsy slide. Until mIF reagents, digital pathology systems including fluorescence scanners, and data pipelines are standardized, we propose that diagnostic labs will play a crucial role in driving adoption of multiplex tissue diagnostics by using retrospective data from tissue collections as a foundation for laboratory-developed test (LDT) implementation and use in prospective trials as companion diagnostics (CDx).
Collapse
|
27
|
Montironi R, Cimadamore A, Cheng L, Lopez-Beltran A, Scarpelli M. Lesson from the COVID-19 pandemic: pathologists need to build their confidence on working in a digital microscopy environment. Virchows Arch 2021; 479:227-229. [PMID: 34032916 PMCID: PMC8144693 DOI: 10.1007/s00428-021-03123-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Revised: 04/06/2021] [Accepted: 05/14/2021] [Indexed: 11/30/2022]
Affiliation(s)
- Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Via Conca 71, 60126, Ancona, Italy.
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Via Conca 71, 60126, Ancona, Italy
| | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Antonio Lopez-Beltran
- Department of Morphological Sciences, Cordoba University Medical School, Cordoba, Spain
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Via Conca 71, 60126, Ancona, Italy
| |
Collapse
|
28
|
Duplancic R, Kero D. Novel approach for quantification of multiple immunofluorescent signals using histograms and 2D plot profiling of whole-section panoramic images. Sci Rep 2021; 11:8619. [PMID: 33883639 PMCID: PMC8060297 DOI: 10.1038/s41598-021-88101-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Accepted: 04/08/2021] [Indexed: 12/27/2022] Open
Abstract
We describe a novel approach for quantification and colocalization of immunofluorescence (IF) signals of multiple markers on high-resolution panoramic images of serial histological sections utilizing standard staining techniques and readily available software for image processing and analysis. Human gingiva samples stained with primary antibodies against the common leukocyte antigen CD45 and factors related to heparan sulfate glycosaminoglycans (HS GAG) were used. Expression domains and spatial gradients of IF signals were quantified by histograms and 2D plot profiles, respectively. The importance of histomorphometric profiling of tissue samples and IF signal thresholding is elaborated. This approach to quantification of IF staining utilizes pixel (px) counts and comparison of px grey value (GV) or luminance. No cell counting is applied either to determine the cellular content of a given histological section nor the number of cells positive to the primary antibody of interest. There is no selection of multiple Regions-Of-Interest (ROIs) since the entire histological section is quantified. Although the standard IF staining protocol is applied, the data output enables colocalization of multiple markers (up to 30) from a given histological sample. This can serve as an alternative for colocalization of IF staining of multiple primary antibodies based on repeating cycles of staining of the same histological section since those techniques require non standard staining protocols and sophisticated equipment that can be out of reach for small laboratories in academic settings. Combined with the data from ontological bases, this approach to quantification of IF enables creation of in silico virtual disease models.
Collapse
Affiliation(s)
- Roko Duplancic
- Study Program of Dental Medicine, School of Medicine, University of Split, Soltanska 2, 21000, Split, Croatia
| | - Darko Kero
- Study Program of Dental Medicine, School of Medicine, University of Split, Soltanska 2, 21000, Split, Croatia. .,Department of Anatomy, Histology and Embryology, Laboratory for Early Human Development, School of Medicine, University of Split, Soltanska 2, 21000, Split, Croatia.
| |
Collapse
|
29
|
Kazdal D, Rempel E, Oliveira C, Allgäuer M, Harms A, Singer K, Kohlwes E, Ormanns S, Fink L, Kriegsmann J, Leichsenring M, Kriegsmann K, Stögbauer F, Tavernar L, Leichsenring J, Volckmar AL, Longuespée R, Winter H, Eichhorn M, Heußel CP, Herth F, Christopoulos P, Reck M, Muley T, Weichert W, Budczies J, Thomas M, Peters S, Warth A, Schirmacher P, Stenzinger A, Kriegsmann M. Conventional and semi-automatic histopathological analysis of tumor cell content for multigene sequencing of lung adenocarcinoma. Transl Lung Cancer Res 2021; 10:1666-1678. [PMID: 34012783 PMCID: PMC8107748 DOI: 10.21037/tlcr-20-1168] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Background Targeted genetic profiling of tissue samples is paramount to detect druggable genetic aberrations in patients with non-squamous non-small cell lung cancer (NSCLC). Accurate upfront estimation of tumor cell content (TCC) is a crucial pre-analytical step for reliable testing and to avoid false-negative results. As of now, TCC is usually estimated on hematoxylin-eosin (H&E) stained tissue sections by a pathologist, a methodology that may be prone to substantial intra- and interobserver variability. Here we the investigate suitability of digital pathology for TCC estimation in a clinical setting by evaluating the concordance between semi-automatic and conventional TCC quantification. Methods TCC was analyzed in 120 H&E and thyroid transcription factor 1 (TTF-1) stained high-resolution images by 19 participants with different levels of pathological expertise as well as by applying two semi-automatic digital pathology image analysis tools (HALO and QuPath). Results Agreement of TCC estimations [intra-class correlation coefficients (ICC)] between the two software tools (H&E: 0.87; TTF-1: 0.93) was higher compared to that between conventional observers (0.48; 0.47). Digital TCC estimations were in good agreement with the average of human TCC estimations (0.78; 0.96). Conventional TCC estimators tended to overestimate TCC, especially in H&E stainings, in tumors with solid patterns and in tumors with an actual TCC close to 50%. Conclusions Our results determine factors that influence TCC estimation. Computer-assisted analysis can improve the accuracy of TCC estimates prior to molecular diagnostic workflows. In addition, we provide a free web application to support self-training and quality improvement initiatives at other institutions.
Collapse
Affiliation(s)
- Daniel Kazdal
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Eugen Rempel
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Cristiano Oliveira
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Allgäuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Alexander Harms
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Kerstin Singer
- Institute of Pathology, University Hospital Tübingen, Tübingen, Germany
| | - Elke Kohlwes
- Institute of Pathology, Johannes Gutenberg University Mainz, Mainz, Germany
| | - Steffen Ormanns
- Institute of Pathology, Ludwig-Maximilians University of Munich, Munich, Germany
| | - Ludger Fink
- Institute of Pathology, Cytopathology, and Molecular Pathology, UEGP MVZ, Giessen/Wetzlar/Limburg, Germany
| | - Jörg Kriegsmann
- MVZ for Histology, Cytology and Molecular Diagnostics, Trier, Germany
| | | | - Katharina Kriegsmann
- Department of Hematology, Oncology and Rheumatology, University Hospital Heidelberg, Heidelberg, Germany
| | - Fabian Stögbauer
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Luca Tavernar
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Jonas Leichsenring
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Anna-Lena Volckmar
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Rémi Longuespée
- Department of Clinical Pharmacology and Pharmacoepidemiology, University Hospital Heidelberg, Heidelberg, Germany
| | - Hauke Winter
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Eichhorn
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany
| | - Claus Peter Heußel
- Department of Thoracic Surgery, Thoraxklinik at University Hospital Heidelberg, Heidelberg, Germany.,Diagnostic and Interventional Radiology With Nuclear Medicine, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Felix Herth
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Pulmonology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Petros Christopoulos
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Martin Reck
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Oncology, Lung Clinic Grosshansdorf, Airway Research Center North (ARCN), German Center for Lung Research (DZL), Grosshansdorf, Germany
| | - Thomas Muley
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Translational Research Unit, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Wilko Weichert
- Institute of Pathology, Technical University of Munich, Munich, Germany
| | - Jan Budczies
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Michael Thomas
- Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany.,Department of Thoracic Oncology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Solange Peters
- Department of Oncology, Centre Hospitalier Universitaire Vaudois (CHUV) and Lausanne University, Lausanne, Switzerland
| | - Arne Warth
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Institute of Pathology, Cytopathology, and Molecular Pathology, UEGP MVZ, Giessen/Wetzlar/Limburg, Germany
| | - Peter Schirmacher
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Center for Personalized Medicine Heidelberg (ZPM), Heidelberg, Germany.,National Network Genomic Medicine Heidelberg (nNGM), Heidelberg, Germany
| | - Albrecht Stenzinger
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| | - Mark Kriegsmann
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany.,Translational Lung Research Center (TLRC) Heidelberg, German Center for Lung Research (DZL), Heidelberg, Germany
| |
Collapse
|
30
|
Cui M, Zhang DY. Artificial intelligence and computational pathology. J Transl Med 2021; 101:412-422. [PMID: 33454724 PMCID: PMC7811340 DOI: 10.1038/s41374-020-00514-0] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Revised: 11/08/2020] [Accepted: 11/10/2020] [Indexed: 02/07/2023] Open
Abstract
Data processing and learning has become a spearhead for the advancement of medicine, with pathology and laboratory medicine has no exception. The incorporation of scientific research through clinical informatics, including genomics, proteomics, bioinformatics, and biostatistics, into clinical practice unlocks innovative approaches for patient care. Computational pathology is burgeoning subspecialty in pathology that promises a better-integrated solution to whole-slide images, multi-omics data, and clinical informatics. However, computational pathology faces several challenges, including the ability to integrate raw data from different sources, limitation of hardware processing capacity, and a lack of specific training programs, as well as issues on ethics and larger societal acceptable practices that are still solidifying. The establishment of the entire industry of computational pathology requires far-reaching changes of the three essential elements connecting patients and doctors: the local laboratory, the scan center, and the central cloud hub/portal for data processing and retrieval. Computational pathology, unlocked through information integration and advanced digital communication networks, has the potential to improve clinical workflow efficiency, diagnostic quality, and ultimately create personalized diagnosis and treatment plans for patients. This review describes clinical perspectives and discusses the statistical methods, clinical applications, potential obstacles, and future directions of computational pathology.
Collapse
Affiliation(s)
- Miao Cui
- St. Luke's Roosevelt Hospital Center, Icahn School of Medicine at Mount Sinai, New York, NY, 10025, USA
| | - David Y Zhang
- Pathology and Laboratory Services, VA Medical Center, New York, NY, 10010, USA.
| |
Collapse
|
31
|
Sobhani F, Robinson R, Hamidinekoo A, Roxanis I, Somaiah N, Yuan Y. Artificial intelligence and digital pathology: Opportunities and implications for immuno-oncology. Biochim Biophys Acta Rev Cancer 2021; 1875:188520. [PMID: 33561505 PMCID: PMC9062980 DOI: 10.1016/j.bbcan.2021.188520] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 01/04/2021] [Accepted: 01/30/2021] [Indexed: 02/08/2023]
Abstract
The field of immuno-oncology has expanded rapidly over the past decade, but key questions remain. How does tumour-immune interaction regulate disease progression? How can we prospectively identify patients who will benefit from immunotherapy? Identifying measurable features of the tumour immune-microenvironment which have prognostic or predictive value will be key to making meaningful gains in these areas. Recent developments in deep learning enable big-data analysis of pathological samples. Digital approaches allow data to be acquired, integrated and analysed far beyond what is possible with conventional techniques, and to do so efficiently and at scale. This has the potential to reshape what can be achieved in terms of volume, precision and reliability of output, enabling data for large cohorts to be summarised and compared. This review examines applications of artificial intelligence (AI) to important questions in immuno-oncology (IO). We discuss general considerations that need to be taken into account before AI can be applied in any clinical setting. We describe AI methods that have been applied to the field of IO to date and present several examples of their use.
Collapse
Affiliation(s)
- Faranak Sobhani
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK; Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Ruth Robinson
- Division of Radiotherapy and Imaging, Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, UK.
| | - Azam Hamidinekoo
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK; Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| | - Ioannis Roxanis
- The Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK.
| | - Navita Somaiah
- Division of Radiotherapy and Imaging, Institute of Cancer Research, The Royal Marsden NHS Foundation Trust, London, UK.
| | - Yinyin Yuan
- Division of Molecular Pathology, The Institute of Cancer Research, London, UK; Centre for Evolution and Cancer, The Institute of Cancer Research, London, UK.
| |
Collapse
|
32
|
"Omics" in traumatic brain injury: novel approaches to a complex disease. Acta Neurochir (Wien) 2021; 163:2581-2594. [PMID: 34273044 PMCID: PMC8357753 DOI: 10.1007/s00701-021-04928-7] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Accepted: 06/23/2021] [Indexed: 11/12/2022]
Abstract
BACKGROUND To date, there is neither any pharmacological treatment with efficacy in traumatic brain injury (TBI) nor any method to halt the disease progress. This is due to an incomplete understanding of the vast complexity of the biological cascades and failure to appreciate the diversity of secondary injury mechanisms in TBI. In recent years, techniques for high-throughput characterization and quantification of biological molecules that include genomics, proteomics, and metabolomics have evolved and referred to as omics. METHODS In this narrative review, we highlight how omics technology can be applied to potentiate diagnostics and prognostication as well as to advance our understanding of injury mechanisms in TBI. RESULTS The omics platforms provide possibilities to study function, dynamics, and alterations of molecular pathways of normal and TBI disease states. Through advanced bioinformatics, large datasets of molecular information from small biological samples can be analyzed in detail and provide valuable knowledge of pathophysiological mechanisms, to include in prognostic modeling when connected to clinically relevant data. In such a complex disease as TBI, omics enables broad categories of studies from gene compositions associated with susceptibility to secondary injury or poor outcome, to potential alterations in metabolites following TBI. CONCLUSION The field of omics in TBI research is rapidly evolving. The recent data and novel methods reviewed herein may form the basis for improved precision medicine approaches, development of pharmacological approaches, and individualization of therapeutic efforts by implementing mathematical "big data" predictive modeling in the near future.
Collapse
|
33
|
Montironi R, Cimadamore A, Lopez-Beltran A, Cheng L, Scarpelli M. Update on Prostate Cancer Diagnosis, Prognosis, and Prediction to Response to Therapy. Cells 2020; 10:cells10010020. [PMID: 33374303 PMCID: PMC7824536 DOI: 10.3390/cells10010020] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2020] [Accepted: 12/18/2020] [Indexed: 12/18/2022] Open
Affiliation(s)
- Rodolfo Montironi
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
- Correspondence: ; Tel.: +39-071-596-4830; Fax: +39-071-889-985
| | - Alessia Cimadamore
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
| | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Marina Scarpelli
- Section of Pathological Anatomy, School of Medicine, United Hospitals, Polytechnic University of the Marche Region, 60126 Ancona, Italy; (A.C.); (M.S.)
| |
Collapse
|
34
|
Cheng JY, Abel JT, Balis UGJ, McClintock DS, Pantanowitz L. Challenges in the Development, Deployment, and Regulation of Artificial Intelligence in Anatomic Pathology. THE AMERICAN JOURNAL OF PATHOLOGY 2020; 191:1684-1692. [PMID: 33245914 DOI: 10.1016/j.ajpath.2020.10.018] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 10/08/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
Significant advances in artificial intelligence (AI), deep learning, and other machine-learning approaches have been made in recent years, with applications found in almost every industry, including health care. AI has proved to be capable of completing a spectrum of mundane to complex medically oriented tasks previously performed only by boarded physicians, most recently assisting with the detection of cancers difficult to find on histopathology slides. Although computers will not replace pathologists any time soon, properly designed AI-based tools hold great potential for increasing workflow efficiency and diagnostic accuracy in the practice of pathology. Recent trends, such as data augmentation, crowdsourcing for generating annotated data sets, and unsupervised learning with molecular and/or clinical outcomes versus human diagnoses as a source of ground truth, are eliminating the direct role of pathologists in algorithm development. Proper integration of AI-based systems into anatomic-pathology practice will necessarily require fully digital imaging platforms, an overhaul of legacy information-technology infrastructures, modification of laboratory/pathologist workflows, appropriate reimbursement/cost-offsetting models, and ultimately, the active participation of pathologists to encourage buy-in and oversight. Regulations tailored to the nature and limitations of AI are currently in development and, when instituted, are expected to promote safe and effective use. This review addresses the challenges in AI development, deployment, and regulation to be overcome prior to its widespread adoption in anatomic pathology.
Collapse
Affiliation(s)
- Jerome Y Cheng
- Department of Pathology, University of Michigan, Ann Arbor, Michigan.
| | - Jacob T Abel
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | - Ulysses G J Balis
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| | | | - Liron Pantanowitz
- Department of Pathology, University of Michigan, Ann Arbor, Michigan
| |
Collapse
|
35
|
Montironi R, Cimadamore A, Lopez-Beltran A, Cheng L, Scarpelli M. Exciting experiences in the ' Rocky road to digital diagnostics'. J Clin Pathol 2020; 74:5-6. [PMID: 33132214 DOI: 10.1136/jclinpath-2020-207161] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2020] [Accepted: 10/07/2020] [Indexed: 11/04/2022]
Affiliation(s)
- Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of Marche, Ancona, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of Marche, Ancona, Italy
| | - Antonio Lopez-Beltran
- Pathology and Surgery, Universidad de Cordoba Facultad de Medicina y Enfermeria, Cordoba, Spain
| | - Liang Cheng
- Pathology & Laboratory Medicine, Indiana University School of Medicine, Indianapolis, Indiana, USA
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of Marche, Ancona, Italy
| |
Collapse
|
36
|
Herrington CS, Poulsom R, Coates PJ. Recent Advances in Pathology: the 2020 Annual Review Issue of The Journal of Pathology. J Pathol 2020; 250:475-479. [PMID: 32346919 DOI: 10.1002/path.5425] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Accepted: 03/13/2020] [Indexed: 12/13/2022]
Abstract
This year's Annual Review Issue of The Journal of Pathology contains 18 invited reviews on current research areas in pathology. The subject areas reflect the broad range of topics covered by the journal and this year encompass the development and application of software in digital histopathology, implementation of biomarkers in pathology practice; genetics and epigenetics, and stromal influences in disease. The reviews are authored by experts in their field and provide comprehensive updates in the chosen areas, in which there has been considerable recent progress in our understanding of disease. © 2020 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- C Simon Herrington
- Edinburgh Cancer Research Centre, Institute of Genetics and Molecular Medicine, University of Edinburgh, Edinburgh, UK
| | - Richard Poulsom
- The Pathological Society of Great Britain and Ireland, London, UK
| | - Philip J Coates
- RECAMO, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| |
Collapse
|
37
|
Zhang H, Kalirai H, Acha-Sagredo A, Yang X, Zheng Y, Coupland SE. Piloting a Deep Learning Model for Predicting Nuclear BAP1 Immunohistochemical Expression of Uveal Melanoma from Hematoxylin-and-Eosin Sections. Transl Vis Sci Technol 2020; 9:50. [PMID: 32953248 PMCID: PMC7476670 DOI: 10.1167/tvst.9.2.50] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/28/2020] [Indexed: 12/20/2022] Open
Abstract
Background Uveal melanoma (UM) is the most common primary intraocular malignancy in adults. Monosomy 3 and BAP1 mutation are strong prognostic factors predicting metastatic risk in UM. Nuclear BAP1 (nBAP1) expression is a close immunohistochemical surrogate for both genetic alterations. Not all laboratories perform routine BAP1 immunohistochemistry or genetic testing, and rely mainly on clinical information and anatomic/morphologic analyses for UM prognostication. The purpose of our study was to pilot deep learning (DL) techniques to predict nBAP1 expression on whole slide images (WSIs) of hematoxylin and eosin (H&E) stained UM sections. Methods One hundred forty H&E-stained UMs were scanned at 40 × magnification, using commercially available WSI image scanners. The training cohort comprised 66 BAP1+ and 74 BAP1− UM, with known chromosome 3 status and clinical outcomes. Nonoverlapping areas of three different dimensions (512 × 512, 1024 × 1024, and 2048 × 2048 pixels) for comparison were extracted from tumor regions in each WSI, and were resized to 256 × 256 pixels. Deep convolutional neural networks (Resnet18 pre-trained on Imagenet) and auto-encoder-decoders (U-Net) were trained to predict nBAP1 expression of these patches. Trained models were tested on the patches cropped from a test cohort of WSIs of 16 BAP1+ and 28 BAP1− UM cases. Results The trained model with best performance achieved area under the curve values of 0.90 for patches and 0.93 for slides on the test set. Conclusions Our results show the effectiveness of DL for predicting nBAP1 expression in UM on the basis of H&E sections only. Translational Relevance Our pilot demonstrates a high capacity of artificial intelligence-related techniques for automated prediction on the basis of histomorphology, and may be translatable into routine histology laboratories.
Collapse
Affiliation(s)
- Hongrun Zhang
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Helen Kalirai
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| | - Amelia Acha-Sagredo
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Xiaoyun Yang
- Chinese Academy of Sciences (CAS) IntelliCloud Technology Co., Ltd., Shanghai, China
| | - Yalin Zheng
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, UK
| | - Sarah E Coupland
- Liverpool Ocular Oncology Research Group, Department of Molecular and Clinical Cancer Medicine, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK.,Liverpool Clinical Laboratories, Liverpool University Hospitals NHS Foundation Trust, Liverpool, UK
| |
Collapse
|
38
|
Browning L, Fryer E, Roskell D, White K, Colling R, Rittscher J, Verrill C. Role of digital pathology in diagnostic histopathology in the response to COVID-19: results from a survey of experience in a UK tertiary referral hospital. J Clin Pathol 2020; 74:129-132. [PMID: 32616541 PMCID: PMC7841475 DOI: 10.1136/jclinpath-2020-206786] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2020] [Accepted: 06/05/2020] [Indexed: 12/21/2022]
Abstract
The COVID-19 pandemic has challenged our diagnostic services at a time when many histopathology departments already faced a diminishing workforce and increasing workload. Digital pathology (DP) has been hailed as a potential solution to at least some of the challenges faced. We present a survey of pathologists within a UK National Health Service cellular pathology department with access to DP, in which we ascertain the role of DP in clinical services during this current pandemic and explore challenges encountered. This survey indicates an increase in uptake of diagnostic DP during this period, with increased remote access. Half of respondents agreed that DP had facilitated maintenance of diagnostic practice. While challenges have been encountered, these are remediable, and none have impacted on the uptake of DP during this period. We conclude that in our institution, DP has demonstrated current and future potential to increase resilience in diagnostic practice and have highlighted some of the challenges that need to be considered.
Collapse
Affiliation(s)
- Lisa Browning
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK .,NIHR Oxford Biomedical Research Centre, Oxford, Oxfordshire, UK
| | - Eve Fryer
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
| | - Derek Roskell
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
| | - Kieron White
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK
| | - Richard Colling
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
| | - Jens Rittscher
- NIHR Oxford Biomedical Research Centre, Oxford, Oxfordshire, UK.,Department of Engineering Science, University of Oxford, Oxford, Oxfordshire, UK
| | - Clare Verrill
- Department of Cellular Pathology, Oxford University Hospitals NHS Foundation Trust, Oxford, Oxfordshire, UK.,NIHR Oxford Biomedical Research Centre, Oxford, Oxfordshire, UK.,Nuffield Department of Surgical Sciences, University of Oxford, Oxford, Oxfordshire, UK
| |
Collapse
|