1
|
Zadvornyi T. Digital Pathology as an Innovative Tool for Improving Cancer Diagnosis and Treatment. Exp Oncol 2025; 46:289-294. [PMID: 39985358 DOI: 10.15407/exp-oncology.2024.04.289] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Indexed: 02/24/2025]
Abstract
For more than a century, the "gold" standard for diagnosing malignant neoplasms has been pathohistology. However, the continuous advancement of modern technologies is leading to a radical transformation of this field and the emergence of digital pathology. The main advantages of digital pathology include the convenience of the data storage and transfer, as well as the potential for automating diagnostic processes through the application of artificial intelligence technologies. Integrating digital pathology into clinical practice is expected to accelerate the analysis of histological samples, reduce the costs associated with such procedures, and enable the accumulation of large datasets for future scientific research. At the same time, the development of digital pathology faces certain challenges such as the need for technical upgrades in laboratories, ensuring data cybersecurity, and training qualified personnel.
Collapse
Affiliation(s)
- T Zadvornyi
- R.E. Kavetsky Institute of Experimental Pathology, Oncology, and Radiobiology, the NAS of Ukraine, Kyiv, Ukraine
| |
Collapse
|
2
|
Madrid MF, Mendoza EN, Padilla AL, Choquenaira-Quispe C, de Jesus Guimarães C, de Melo Pereira JV, Barros-Nepomuceno FWA, Lopes Dos Santos I, Pessoa C, de Moraes Filho MO, Rocha DD, Ferreira PMP. In vitro models to evaluate multidrug resistance in cancer cells: Biochemical and morphological techniques and pharmacological strategies. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2025; 28:1-27. [PMID: 39363148 DOI: 10.1080/10937404.2024.2407452] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2024]
Abstract
The overexpression of ATP-binding cassette (ABC) transporters contributes to the failure of chemotherapies and symbolizes a great challenge in oncology, associated with the adaptation of tumor cells to anticancer drugs such that these transporters become less effective, a mechanism known as multidrug resistance (MDR). The aim of this review is to present the most widely used methodologies for induction and comprehension of in vitro models for detection of multidrug-resistant (MDR) modulators or inhibitors, including biochemical and morphological techniques for chemosensitivity studies. The overexpression of MDR proteins, predominantly, the subfamily glycoprotein-1 (P-gp or ABCB1) multidrug resistance, multidrug resistance-associated protein 1 (MRP1 or ABCCC1), multidrug resistance-associated protein 2 (MRP2 or ABCC2) and cancer resistance protein (ABCG2), in chemotherapy-exposed cancer lines have been established/investigated by several techniques. Amongst these techniques, the most used are (i) colorimetric/fluorescent indirect bioassays, (ii) rhodamine and efflux analysis, (iii) release of 3,30-diethyloxacarbocyanine iodide by fluorescence microscopy and flow cytometry to measure P-gp function and other ABC transporters, (iv) exclusion of calcein-acetoxymethylester, (v) ATPase assays to distinguish types of interaction with ABC transporters, (vi) morphology to detail phenotypic characteristics in transformed cells, (vii) molecular testing of resistance-related proteins (RT-qPCR) and (viii) 2D and 3D models, (ix) organoids, and (x) microfluidic technology. Then, in vitro models for detecting chemotherapy MDR cells to assess innovative therapies to modulate or inhibit tumor cell growth and overcome clinical resistance. It is noteworthy that different therapies including anti-miRNAs, antibody-drug conjugates (to natural products), and epigenetic modifications were also considered as promising alternatives, since currently no anti-MDR therapies are able to improve patient quality of life. Therefore, there is also urgency for new clinical markers of resistance to more reliably reflect in vivo effectiveness of novel antitumor drugs.
Collapse
Affiliation(s)
- Maria Fernanda Madrid
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Eleicy Nathaly Mendoza
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Ana Lizeth Padilla
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
| | - Celia Choquenaira-Quispe
- Pharmaceutical Sciences, Faculty of Pharmacy, Dentistry, and Nursing, Federal University of Ceará, Fortaleza, Brazil
- Catholic University of Santa María, Arequipa, Perú
| | - Celina de Jesus Guimarães
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - João Victor de Melo Pereira
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | | | - Ingredy Lopes Dos Santos
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| | - Claudia Pessoa
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Manoel Odorico de Moraes Filho
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Danilo Damasceno Rocha
- Drug Research and Development Center (NPDM), Department of Physiology and Pharmacology, Federal University of Ceará, Fortaleza, Brazil
| | - Paulo Michel Pinheiro Ferreira
- Laboratory of Experimental Cancerology (LabCancer), Department of Biophysics and Physiology, Federal University of Piauí, Teresina, Brazil
| |
Collapse
|
3
|
Alizadeh SM, Helfroush MS, Celebi ME. An Innovative Attention-based Triplet Deep Hashing Approach to Retrieve Histopathology Images. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024:10.1007/s10278-024-01310-8. [PMID: 39528884 DOI: 10.1007/s10278-024-01310-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Revised: 10/16/2024] [Accepted: 10/19/2024] [Indexed: 11/16/2024]
Abstract
Content-based histopathology image retrieval (CBHIR) can assist in the diagnosis of different diseases. The retrieval procedure can be complex and time-consuming if high-dimensional features are required. Thus, hashing techniques are employed to address these issues by mapping the feature space into binary values of varying lengths. The performance of deep hashing approaches in image retrieval is often superior to that of traditional hashing methods. Among deep hashing approaches, triplet-based models are typically more effective than pairwise ones. Recent studies have demonstrated that incorporating the attention mechanism into a deep hashing approach can improve its effectiveness in retrieving images. This paper presents an innovative triplet deep hashing strategy based on the attention mechanism for retrieving histopathology images, called histopathology attention triplet deep hashing (HATDH). Three deep attention-based hashing models with identical architectures and weights are employed to produce binary values. The proposed attention module can aid the models in extracting features more efficiently. Moreover, we introduce an improved triplet loss function considering pair inputs separately in addition to triplet inputs for increasing efficiency during the training and retrieval steps. Based on experiments conducted on two public histopathology datasets, BreakHis and Kather, HATDH significantly outperforms state-of-the-art hashing algorithms.
Collapse
Affiliation(s)
| | | | - M Emre Celebi
- Department of Computer Science and Engineering, University of Central Arkansas, Conway, AR, 72035, USA
| |
Collapse
|
4
|
Mo W, Ke Q, Yang Q, Zhou M, Xie G, Qi D, Peng L, Wang X, Wang F, Ni S, Wang A, Huang J, Wen J, Yang Y, Du K, Wang X, Du X, Zhao Z. A Dual-Modal, Label-Free Raman Imaging Method for Rapid Virtual Staining of Large-Area Breast Cancer Tissue Sections. Anal Chem 2024; 96:13410-13420. [PMID: 38967251 DOI: 10.1021/acs.analchem.4c00870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
As one of the most common cancers, accurate, rapid, and simple histopathological diagnosis is very important for breast cancer. Raman imaging is a powerful technique for label-free analysis of tissue composition and histopathology, but it suffers from slow speed when applied to large-area tissue sections. In this study, we propose a dual-modal Raman imaging method that combines Raman mapping data with microscopy bright-field images to achieve virtual staining of breast cancer tissue sections. We validate our method on various breast tissue sections with different morphologies and biomarker expressions and compare it with the golden standard of histopathological methods. The results demonstrate that our method can effectively distinguish various types and components of tissues, and provide staining images comparable to stained tissue sections. Moreover, our method can improve imaging speed by up to 65 times compared to general spontaneous Raman imaging methods. It is simple, fast, and suitable for clinical applications.
Collapse
Affiliation(s)
- Wenbo Mo
- National Key Laboratory of Plasma Physics, Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
- Department of Engineering Physics, Tsinghua University, 100084 Beijing, China
| | - Qi Ke
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Qiang Yang
- China Academy of Engineering Physics, 621900 Mianyang, China
- Department of Engineering Physics, Tsinghua University, 100084 Beijing, China
| | - Minjie Zhou
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Gang Xie
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Daojian Qi
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Lijun Peng
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Xinming Wang
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Fei Wang
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Shuang Ni
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Anqun Wang
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Jinglin Huang
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Jiaxing Wen
- National Key Laboratory of Plasma Physics, Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Yue Yang
- National Key Laboratory of Plasma Physics, Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Kai Du
- Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| | - Xuewu Wang
- Department of Engineering Physics, Tsinghua University, 100084 Beijing, China
| | - Xiaobo Du
- Mianyang Central Hospital, 621000 Mianyang, China
| | - Zongqing Zhao
- National Key Laboratory of Plasma Physics, Laser Fusion Research Center, China Academy of Engineering Physics, 621900 Mianyang, China
| |
Collapse
|
5
|
Fisher TB, Saini G, Rekha TS, Krishnamurthy J, Bhattarai S, Callagy G, Webber M, Janssen EAM, Kong J, Aneja R. Digital image analysis and machine learning-assisted prediction of neoadjuvant chemotherapy response in triple-negative breast cancer. Breast Cancer Res 2024; 26:12. [PMID: 38238771 PMCID: PMC10797728 DOI: 10.1186/s13058-023-01752-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Accepted: 12/11/2023] [Indexed: 01/22/2024] Open
Abstract
BACKGROUND Pathological complete response (pCR) is associated with favorable prognosis in patients with triple-negative breast cancer (TNBC). However, only 30-40% of TNBC patients treated with neoadjuvant chemotherapy (NAC) show pCR, while the remaining 60-70% show residual disease (RD). The role of the tumor microenvironment in NAC response in patients with TNBC remains unclear. In this study, we developed a machine learning-based two-step pipeline to distinguish between various histological components in hematoxylin and eosin (H&E)-stained whole slide images (WSIs) of TNBC tissue biopsies and to identify histological features that can predict NAC response. METHODS H&E-stained WSIs of treatment-naïve biopsies from 85 patients (51 with pCR and 34 with RD) of the model development cohort and 79 patients (41 with pCR and 38 with RD) of the validation cohort were separated through a stratified eightfold cross-validation strategy for the first step and leave-one-out cross-validation strategy for the second step. A tile-level histology label prediction pipeline and four machine-learning classifiers were used to analyze 468,043 tiles of WSIs. The best-trained classifier used 55 texture features from each tile to produce a probability profile during testing. The predicted histology classes were used to generate a histology classification map of the spatial distributions of different tissue regions. A patient-level NAC response prediction pipeline was trained with features derived from paired histology classification maps. The top graph-based features capturing the relevant spatial information across the different histological classes were provided to the radial basis function kernel support vector machine (rbfSVM) classifier for NAC treatment response prediction. RESULTS The tile-level prediction pipeline achieved 86.72% accuracy for histology class classification, while the patient-level pipeline achieved 83.53% NAC response (pCR vs. RD) prediction accuracy of the model development cohort. The model was validated with an independent cohort with tile histology validation accuracy of 83.59% and NAC prediction accuracy of 81.01%. The histological class pairs with the strongest NAC response predictive ability were tumor and tumor tumor-infiltrating lymphocytes for pCR and microvessel density and polyploid giant cancer cells for RD. CONCLUSION Our machine learning pipeline can robustly identify clinically relevant histological classes that predict NAC response in TNBC patients and may help guide patient selection for NAC treatment.
Collapse
Affiliation(s)
- Timothy B Fisher
- Department of Biology, Georgia State University, Atlanta, GA, 30302, USA
| | - Geetanjali Saini
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - T S Rekha
- JSSAHER (JSS Academy of Higher Education and Research) Medical College, Mysuru, Karnataka, India
| | - Jayashree Krishnamurthy
- JSSAHER (JSS Academy of Higher Education and Research) Medical College, Mysuru, Karnataka, India
| | - Shristi Bhattarai
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Grace Callagy
- Discipline of Pathology, University of Galway, Galway, Ireland
| | - Mark Webber
- Discipline of Pathology, University of Galway, Galway, Ireland
| | - Emiel A M Janssen
- Department of Pathology, Stavanger University Hospital, Stavanger, Norway
- Department of Chemistry, Bioscience and Environmental Engineering, University of Stavanger, Stavanger, Norway
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, 30303, USA.
| | - Ritu Aneja
- Department of Biology, Georgia State University, Atlanta, GA, 30302, USA.
- School of Health Professions, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
6
|
Wagner SJ, Matek C, Shetab Boushehri S, Boxberg M, Lamm L, Sadafi A, Winter DJE, Marr C, Peng T. Built to Last? Reproducibility and Reusability of Deep Learning Algorithms in Computational Pathology. Mod Pathol 2024; 37:100350. [PMID: 37827448 DOI: 10.1016/j.modpat.2023.100350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Recent progress in computational pathology has been driven by deep learning. While code and data availability are essential to reproduce findings from preceding publications, ensuring a deep learning model's reusability is more challenging. For that, the codebase should be well-documented and easy to integrate into existing workflows and models should be robust toward noise and generalizable toward data from different sources. Strikingly, only a few computational pathology algorithms have been reused by other researchers so far, let alone employed in a clinical setting. To assess the current state of reproducibility and reusability of computational pathology algorithms, we evaluated peer-reviewed articles available in PubMed, published between January 2019 and March 2021, in 5 use cases: stain normalization; tissue type segmentation; evaluation of cell-level features; genetic alteration prediction; and inference of grading, staging, and prognostic information. We compiled criteria for data and code availability and statistical result analysis and assessed them in 160 publications. We found that only one-quarter (41 of 160 publications) made code publicly available. Among these 41 studies, three-quarters (30 of 41) analyzed their results statistically, half of them (20 of 41) released their trained model weights, and approximately a third (16 of 41) used an independent cohort for evaluation. Our review is intended for both pathologists interested in deep learning and researchers applying algorithms to computational pathology challenges. We provide a detailed overview of publications with published code in the field, list reusable data handling tools, and provide criteria for reproducibility and reusability.
Collapse
Affiliation(s)
- Sophia J Wagner
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Christian Matek
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Sayedali Shetab Boushehri
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany; Institute of Pathology Munich-North, Munich, Germany
| | - Lorenz Lamm
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Helmholtz Pioneer Campus, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ario Sadafi
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Dominik J E Winter
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences, Technical University of Munich, Weihenstephan, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| | - Tingying Peng
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
7
|
Martos O, Hoque MZ, Keskinarkaus A, Kemi N, Näpänkangas J, Eskuri M, Pohjanen VM, Kauppila JH, Seppänen T. Optimized detection and segmentation of nuclei in gastric cancer images using stain normalization and blurred artifact removal. Pathol Res Pract 2023; 248:154694. [PMID: 37494804 DOI: 10.1016/j.prp.2023.154694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/04/2023] [Revised: 07/03/2023] [Accepted: 07/13/2023] [Indexed: 07/28/2023]
Abstract
Histological analysis with microscopy is the gold standard to diagnose and stage cancer, where slides or whole slide images are analyzed for cell morphological and spatial features by pathologists. The nuclei of cancerous cells are characterized by nonuniform chromatin distribution, irregular shapes, and varying size. As nucleus area and shape alone carry prognostic value, detection and segmentation of nuclei are among the most important steps in disease grading. However, evaluation of nuclei is a laborious, time-consuming, and subjective process with large variation among pathologists. Recent advances in digital pathology have allowed significant applications in nuclei detection, segmentation, and classification, but automated image analysis is greatly affected by staining factors, scanner variability, and imaging artifacts, requiring robust image preprocessing, normalization, and segmentation methods for clinically satisfactory results. In this paper, we aimed to evaluate and compare the digital image analysis techniques used in clinical pathology and research in the setting of gastric cancer. A literature review was conducted to evaluate potential methods of improving nuclei detection. Digitized images of 35 patients from a retrospective cohort of gastric adenocarcinoma at Oulu University Hospital in 1987-2016 were annotated for nuclei (n = 9085) by expert pathologists and 14 images of different cancer types from public TCGA dataset with annotated nuclei (n = 7000) were used as a comparison to evaluate applicability in other cancer types. The detection and segmentation accuracy with the selected color normalization and stain separation techniques were compared between the methods. The extracted information can be supplemented by patient's medical data and fed to the existing statistical clinical tools or subjected to subsequent AI-assisted classification and prediction models. The performance of each method is evaluated by several metrics against the annotations done by expert pathologists. The F1-measure of 0.854 ± 0.068 is achieved with color normalization for the gastric cancer dataset, and 0.907 ± 0.044 with color deconvolution for the public dataset, showing comparable results to the earlier state-of-the-art works. The developed techniques serve as a basis for further research on application and interpretability of AI-assisted tools for gastric cancer diagnosis.
Collapse
Affiliation(s)
- Oleg Martos
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| | - Md Ziaul Hoque
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland.
| | - Anja Keskinarkaus
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| | - Niko Kemi
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Juha Näpänkangas
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Maarit Eskuri
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Vesa-Matti Pohjanen
- Department of Pathology, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Joonas H Kauppila
- Department of Surgery, Oulu University Hospital, Finland, and University of Oulu, Finland
| | - Tapio Seppänen
- Center for Machine Vision and Signal Analysis, Faculty of Information Technology and Electrical Engineering, University of Oulu, Finland
| |
Collapse
|
8
|
Duenweg SR, Bobholz SA, Lowman AK, Stebbins MA, Winiarz A, Nath B, Kyereme F, Iczkowski KA, LaViolette PS. Whole slide imaging (WSI) scanner differences influence optical and computed properties of digitized prostate cancer histology. J Pathol Inform 2023; 14:100321. [PMID: 37496560 PMCID: PMC10365953 DOI: 10.1016/j.jpi.2023.100321] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 06/13/2023] [Accepted: 06/28/2023] [Indexed: 07/28/2023] Open
Abstract
Purpose Digital pathology is becoming an increasingly popular area of advancement in both research and clinically. Pathologists are now able to manage and interpret slides digitally, as well as collaborate with external pathologists with digital copies of slides. Differences in slide scanners include variation in resolution, image contrast, and optical properties, which may influence downstream image processing. This study tested the hypothesis that varying slide scanners would result in differences in computed pathomic features on prostate cancer whole mount slides. Design This study collected 192 unique tissue slides from 30 patients following prostatectomy. Tissue samples were paraffin-embedded, stained for hematoxylin and eosin (H&E), and digitized using 3 different scanning microscopes at the highest available magnification rate, for a total of 3 digitized slides per tissue slide. These scanners included a (S1) Nikon microscope equipped with an automated sliding stage, an (S2) Olympus VS120 slide scanner, and a (S3) Huron TissueScope LE scanner. A color deconvolution algorithm was then used to optimize contrast by projecting the RGB image into color channels representing optical stain density. The resulting intensity standardized images were then computationally processed to segment tissue and calculate pathomic features including lumen, stroma, epithelium, and epithelial cell density, as well as second-order features including lumen area and roundness; epithelial area, roundness, and wall thickness; and cell fraction. For each tested feature, mean values of that feature per digitized slide were collected and compared across slide scanners using mixed effect models, fit to compare differences in the tested feature associated with all slide scanners for each slide, including a random effect of subject with a nested random effect of slide to account for repeated measures. Similar models were also computed for tissue densities to examine how differences in scanner impact downstream processing. Results Each mean color channel intensity (i.e., Red, Green, Blue) differed between slide scanners (all P<.001). Of the color deconvolved images, only the hematoxylin channel was similar in all 3 scanners (all P>.05). Lumen and stroma densities between S3 and S1 slides, and epithelial cell density between S3 and S2 (P>.05) were comparable but all other comparisons were significantly different (P<.05). The second-order features were found to be comparable for all scanner comparisons, except for lumen area and epithelium area. Conclusion This study demonstrates that both optical and computed properties of digitized histological samples are impacted by slide scanner differences. Future research is warranted to better understand which scanner properties influence the tissue segmentation process and to develop harmonization techniques for comparing data across multiple slide scanners.
Collapse
Affiliation(s)
- Savannah R. Duenweg
- Departments of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Samuel A. Bobholz
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Allison K. Lowman
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Margaret A. Stebbins
- Departments of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Aleksandra Winiarz
- Departments of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Biprojit Nath
- Departments of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Fitzgerald Kyereme
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Kenneth A. Iczkowski
- Department of Pathology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| | - Peter S. LaViolette
- Departments of Biophysics, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
- Department of Radiology, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
- Department of Biomedical Engineering, Medical College of Wisconsin, 8701 Watertown Plank Rd., Milwaukee, WI 53226, USA
| |
Collapse
|
9
|
Chen Y, Li H, Janowczyk A, Toro P, Corredor G, Whitney J, Lu C, Koyuncu CF, Mokhtari M, Buzzy C, Ganesan S, Feldman MD, Fu P, Corbin H, Harbhajanka A, Gilmore H, Goldstein LJ, Davidson NE, Desai S, Parmar V, Madabhushi A. Computational pathology improves risk stratification of a multi-gene assay for early stage ER+ breast cancer. NPJ Breast Cancer 2023; 9:40. [PMID: 37198173 PMCID: PMC10192429 DOI: 10.1038/s41523-023-00545-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 04/28/2023] [Indexed: 05/19/2023] Open
Abstract
Prognostic markers currently utilized in clinical practice for estrogen receptor-positive (ER+) and lymph node-negative (LN-) invasive breast cancer (IBC) patients include the Nottingham grading system and Oncotype Dx (ODx). However, these biomarkers are not always optimal and remain subject to inter-/intra-observer variability and high cost. In this study, we evaluated the association between computationally derived image features from H&E images and disease-free survival (DFS) in ER+ and LN- IBC. H&E images from a total of n = 321 patients with ER+ and LN- IBC from three cohorts were employed for this study (Training set: D1 (n = 116), Validation sets: D2 (n = 121) and D3 (n = 84)). A total of 343 features relating to nuclear morphology, mitotic activity, and tubule formation were computationally extracted from each slide image. A Cox regression model (IbRiS) was trained to identify significant predictors of DFS and predict a high/low-risk category using D1 and was validated on independent testing sets D2 and D3 as well as within each ODx risk category. IbRiS was significantly prognostic of DFS with a hazard ratio (HR) of 2.33 (95% confidence interval (95% CI) = 1.02-5.32, p = 0.045) on D2 and a HR of 2.94 (95% CI = 1.18-7.35, p = 0.0208) on D3. In addition, IbRiS yielded significant risk stratification within high ODx risk categories (D1 + D2: HR = 10.35, 95% CI = 1.20-89.18, p = 0.0106; D1: p = 0.0238; D2: p = 0.0389), potentially providing more granular risk stratification than offered by ODx alone.
Collapse
Grants
- R01 CA216579 NCI NIH HHS
- C06 RR012463 NCRR NIH HHS
- R43 EB028736 NIBIB NIH HHS
- U01 CA239055 NCI NIH HHS
- R01 CA249992 NCI NIH HHS
- UG1 CA233328 NCI NIH HHS
- R01 CA220581 NCI NIH HHS
- R01 CA202752 NCI NIH HHS
- R01 CA208236 NCI NIH HHS
- U01 CA248226 NCI NIH HHS
- I01 BX004121 BLRD VA
- R01 CA257612 NCI NIH HHS
- U54 CA254566 NCI NIH HHS
- Research reported in this study was supported by the National Cancer Institute under award numbers R01CA249992-01A1, R01CA202752-01A1, R01CA208236-01A1, R01CA216579-01A1, R01CA220581-01A1, R01CA257612-01A1, 1U01CA239055-01, 1U01CA248226-01, 1U54CA254566-01, National Heart, Lung and Blood Institute 1R01HL15127701A1, R01HL15807101A1, National Institute of Biomedical Imaging and Bioengineering 1R43EB028736-01, National Center for Research Resources under award number 1 C06 RR12463-01, VA Merit Review Award IBX004121A from the United States Department of Veterans Affairs Biomedical Laboratory Research and Development Service the Office of the Assistant Secretary of Defense for Health Affairs, through the Breast Cancer Research Program (W81XWH-19-1-0668), the Prostate Cancer Research Program (W81XWH-15-1-0558, W81XWH-20-1-0851), the Lung Cancer Research Program (W81XWH-18-1-0440, W81XWH-20-1-0595), the Peer Reviewed Cancer Research Program (W81XWH-18-1-0404, W81XWH-21-1-0345, W81XWH-21-1-0160), the Kidney Precision Medicine Project (KPMP) Glue Grant and sponsored research agreements from Bristol Myers-Squibb, Boehringer-Ingelheim, Eli-Lilly and Astrazeneca.
Collapse
Affiliation(s)
- Yuli Chen
- Shaanxi Normal University, School of Computer Science, Xi'an, China
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Haojia Li
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Andrew Janowczyk
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Precision Oncology Center, University of Lausanne, Lausanne, Switzerland
| | - Paula Toro
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Germán Corredor
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Jon Whitney
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Cheng Lu
- Shaanxi Normal University, School of Computer Science, Xi'an, China
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Can F Koyuncu
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Mojgan Mokhtari
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Christina Buzzy
- Department of Biomedical Engineering, Case Western Reserve University, Cleveland, OH, USA
| | - Shridar Ganesan
- Rutgers Cancer Institute of New Jersey, New Brunswick, NJ, USA
| | - Michael D Feldman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA, USA
| | - Pingfu Fu
- Department of Population and Quantitative Health Sciences, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Haley Corbin
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Hannah Gilmore
- University Hospitals Cleveland Medical Center, Cleveland, OH, USA
| | | | - Nancy E Davidson
- Fred Hutchinson Cancer Research Center, University of Washington, and Seattle Cancer Care Alliance, Seattle, WA, USA
| | - Sangeeta Desai
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Vani Parmar
- Tata Memorial Centre, Homi Bhabha National Institute, Mumbai, India
| | - Anant Madabhushi
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
- Louis Stokes VA Medical Center, Cleveland, OH, USA.
| |
Collapse
|
10
|
Moncayo R, Martel AL, Romero E. Removing non-nuclei information from histopathological images: A preprocessing step towards improving nuclei segmentation methods. J Pathol Inform 2023; 14:100315. [PMID: 37811335 PMCID: PMC10550762 DOI: 10.1016/j.jpi.2023.100315] [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: 02/26/2023] [Revised: 04/10/2023] [Accepted: 04/12/2023] [Indexed: 10/10/2023] Open
Abstract
Disease interpretation by computer-aided diagnosis systems in digital pathology depends on reliable detection and segmentation of nuclei in hematoxylin and eosin (HE) images. These 2 tasks are challenging since appearance of both cell nuclei and background structures are very variable. This paper presents a method to improve nuclei detection and segmentation in HE images by removing tiles that only contain background information. The method divides each image into smaller patches and uses their projection to the noiselet space to capture different spatial features from non-nuclei background and nuclei structures. The noiselet features are clustered by a K-means algorithm and the resultant partition, defined by the cluster centroids, is herein named the noiselet code-book. A part of an image, a tile, is divided into patches and represented by the histogram of occurrences of the projected patches in the noiselet code-book. Finally, with these histograms, a classifier learns to differentiate between nuclei and non-nuclei tiles. By applying a conventional watershed-marked method to detect and segment nuclei, evaluation consisted in comparing pure watershed method against denoising-plus-watershed in an open database with 8 different types of tissues. The averaged F-score of nuclei detection improved from 0.830 to 0.86 and the dice score after segmentation increased from 0.701 to 0.723.
Collapse
Affiliation(s)
- Ricardo Moncayo
- Computer Imaging and Medical Applications Laboratory (CIM@LAB), Universidad Nacional de Colombia, Bogotá, Colombia
| | - Anne L. Martel
- Department of Medical Biophysics, University of Toronto, Ontario, Canada
- Physical Sciences, Sunnybrook Research Institute, Toronto, Ontario, Canada
| | - Eduardo Romero
- Computer Imaging and Medical Applications Laboratory (CIM@LAB), Universidad Nacional de Colombia, Bogotá, Colombia
| |
Collapse
|
11
|
Cinar U, Cetin Atalay R, Cetin YY. Human Hepatocellular Carcinoma Classification from H&E Stained Histopathology Images with 3D Convolutional Neural Networks and Focal Loss Function. J Imaging 2023; 9:jimaging9020025. [PMID: 36826944 PMCID: PMC9959324 DOI: 10.3390/jimaging9020025] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Revised: 01/13/2023] [Accepted: 01/18/2023] [Indexed: 01/26/2023] Open
Abstract
This paper proposes a new Hepatocellular Carcinoma (HCC) classification method utilizing a hyperspectral imaging system (HSI) integrated with a light microscope. Using our custom imaging system, we have captured 270 bands of hyperspectral images of healthy and cancer tissue samples with HCC diagnosis from a liver microarray slide. Convolutional Neural Networks with 3D convolutions (3D-CNN) have been used to build an accurate classification model. With the help of 3D convolutions, spectral and spatial features within the hyperspectral cube are incorporated to train a strong classifier. Unlike 2D convolutions, 3D convolutions take the spectral dimension into account while automatically collecting distinctive features during the CNN training stage. As a result, we have avoided manual feature engineering on hyperspectral data and proposed a compact method for HSI medical applications. Moreover, the focal loss function, utilized as a CNN cost function, enables our model to tackle the class imbalance problem residing in the dataset effectively. The focal loss function emphasizes the hard examples to learn and prevents overfitting due to the lack of inter-class balancing. Our empirical results demonstrate the superiority of hyperspectral data over RGB data for liver cancer tissue classification. We have observed that increased spectral dimension results in higher classification accuracy. Both spectral and spatial features are essential in training an accurate learner for cancer tissue classification.
Collapse
|
12
|
Mamonto L, Nelwan BJ, Sungowati NK, Miskad UA, Cangara MH, Zainuddin AA. Association of chemokine (CXC motif) receptor 4 expression with lymphovascular invasion and lymph node metastasis of invasive breast cancer. Breast Dis 2023; 41:447-453. [PMID: 36617771 DOI: 10.3233/bd-229003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
BACKGROUND The histological tumor grade influences the prognosis of breast cancer. In metastatic breast cancer, stromal cells produce chemokine (CXC motif) ligand 12 or stromal cell-derived factor-1 as a chemoattractant, which binds to chemokine (CXC motif) receptor 4 (CXCR4) expressed by breast cancer cells. OBJECTIVE This study aimed to determine the expression of CXCR4 in invasive breast cancer in relation to lymphovascular invasion (LVI) and lymph node metastasis. METHODS This observational study retrospectively investigated a paraffin block archived sample diagnosed with invasive breast cancer. The results of immunohistochemical staining with CXCR4 antibody and expression analysis were evaluated using light microscopy. The data were statistically analyzed using the chi-square test and presented in a table using SPSS version 18. P-values of <0.05 were considered statistically significant. RESULTS The expression of CXCR4 was significantly associated with the incidence of LVI and lymph node metastasis in invasive breast cancer (both p = 0.001). CONCLUSIONS The results show that the expression of CXCR4 varies and support its decisive role in the incidence of LVI and lymph node metastasis in invasive breast cancer.
Collapse
Affiliation(s)
- Lidya Mamonto
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Berti J Nelwan
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Ni Ketut Sungowati
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Upik A Miskad
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Muh Husni Cangara
- Department of Anatomical Pathology, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| | - Andi Alfian Zainuddin
- Department of Public Health and Community Medicine Science, Faculty of Medicine, Hasanuddin University, Makassar, Indonesia
| |
Collapse
|
13
|
An Approach toward Automatic Specifics Diagnosis of Breast Cancer Based on an Immunohistochemical Image. J Imaging 2023; 9:jimaging9010012. [PMID: 36662110 PMCID: PMC9866917 DOI: 10.3390/jimaging9010012] [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: 11/27/2022] [Revised: 12/30/2022] [Accepted: 01/01/2023] [Indexed: 01/06/2023] Open
Abstract
The paper explored the problem of automatic diagnosis based on immunohistochemical image analysis. The issue of automated diagnosis is a preliminary and advisory statement for a diagnostician. The authors studied breast cancer histological and immunohistochemical images using the following biomarkers progesterone, estrogen, oncoprotein, and a cell proliferation biomarker. The authors developed a breast cancer diagnosis method based on immunohistochemical image analysis. The proposed method consists of algorithms for image preprocessing, segmentation, and the determination of informative indicators (relative area and intensity of cells) and an algorithm for determining the molecular genetic breast cancer subtype. An adaptive algorithm for image preprocessing was developed to improve the quality of the images. It includes median filtering and image brightness equalization techniques. In addition, the authors developed a software module part of the HIAMS software package based on the Java programming language and the OpenCV computer vision library. Four molecular genetic breast cancer subtypes could be identified using this solution: subtype Luminal A, subtype Luminal B, subtype HER2/neu amplified, and basalt-like subtype. The developed algorithm for the quantitative characteristics of the immunohistochemical images showed sufficient accuracy in determining the cancer subtype "Luminal A". It was experimentally established that the relative area of the nuclei of cells covered with biomarkers of progesterone, estrogen, and oncoprotein was more than 85%. The given approach allows for automating and accelerating the process of diagnosis. Developed algorithms for calculating the quantitative characteristics of cells on immunohistochemical images can increase the accuracy of diagnosis.
Collapse
|
14
|
Sadeghi A, Naghavi SMH, Mozafari M, Afshari E. Nanoscale biomaterials for terahertz imaging: A non-invasive approach for early cancer detection. Transl Oncol 2023; 27:101565. [PMID: 36343417 PMCID: PMC9643578 DOI: 10.1016/j.tranon.2022.101565] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2022] [Revised: 09/12/2022] [Accepted: 10/09/2022] [Indexed: 11/06/2022] Open
Abstract
Terahertz (THz) technology is developing a non-invasive imaging system for biosensing and clinical diagnosis. THz medical imaging mainly benefits from great sensitivity in detecting changes in water content and structural variations in diseased cells versus normal tissues. Compared to healthy tissues, cancerous tumors contain a higher level of water molecules and show structural changes, resulting in different THz absorption. Here we described the principle of THz imaging and advancement in the field of translational biomedicine and early detection of pathologic tissue, with a particular focus on oncology. In addition, although the main forte of THz imaging relies on detecting differences in water content to distinguish the exact margin of tumor, THz displays limited contrast in living tissue for in-vivo clinical imaging. In the last few years, nanotechnology has attracted attention to aid THz medical imaging and various nanoparticles have been investigated as contrast enhancements to improve the accuracy, sensitivity, and specificity of THz images. Most of these multimodal contrast agents take advantage of the temperature-dependent of THz spectrum to the conformational variation of the water molecule. We discuss advances in developing THz contrast agents to accelerate the advancement of non-invasive THz imaging with improved sensitivity and specificity for translational clinical oncology.
Collapse
Affiliation(s)
- Ali Sadeghi
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA.
| | - S M Hossein Naghavi
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA
| | - Masoud Mozafari
- Mount Sinai Hospital, University of Toronto, Toronto, ON, Canada
| | - Ehsan Afshari
- Department of Electrical Engineering and Computer Science, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
15
|
Michielli N, Caputo A, Scotto M, Mogetta A, Pennisi OAM, Molinari F, Balmativola D, Bosco M, Gambella A, Metovic J, Tota D, Carpenito L, Gasparri P, Salvi M. Stain normalization in digital pathology: Clinical multi-center evaluation of image quality. J Pathol Inform 2022; 13:100145. [PMID: 36268060 PMCID: PMC9577129 DOI: 10.1016/j.jpi.2022.100145] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2022] [Revised: 09/14/2022] [Accepted: 09/22/2022] [Indexed: 11/20/2022] Open
Abstract
In digital pathology, the final appearance of digitized images is affected by several factors, resulting in stain color and intensity variation. Stain normalization is an innovative solution to overcome stain variability. However, the validation of color normalization tools has been assessed only from a quantitative perspective, through the computation of similarity metrics between the original and normalized images. To the best of our knowledge, no works investigate the impact of normalization on the pathologist's evaluation. The objective of this paper is to propose a multi-tissue (i.e., breast, colon, liver, lung, and prostate) and multi-center qualitative analysis of a stain normalization tool with the involvement of pathologists with different years of experience. Two qualitative studies were carried out for this purpose: (i) a first study focused on the analysis of the perceived image quality and absence of significant image artifacts after the normalization process; (ii) a second study focused on the clinical score of the normalized image with respect to the original one. The results of the first study prove the high quality of the normalized image with a low impact artifact generation, while the second study demonstrates the superiority of the normalized image with respect to the original one in clinical practice. The normalization process can help both to reduce variability due to tissue staining procedures and facilitate the pathologist in the histological examination. The experimental results obtained in this work are encouraging and can justify the use of a stain normalization tool in clinical routine.
Collapse
Affiliation(s)
- Nicola Michielli
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Alessandro Caputo
- Department of Medicine and Surgery, University Hospital of Salerno, Salerno, Italy
| | - Manuela Scotto
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Alessandro Mogetta
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Orazio Antonino Maria Pennisi
- Technology Transfer and Industrial Liaison Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Filippo Molinari
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| | - Davide Balmativola
- Pathology Unit, Humanitas Gradenigo Hospital, Corso Regina Margherita 8, 10153 Turin, Italy
| | - Martino Bosco
- Department of Pathology, Michele and Pietro Ferrero Hospital, 12060 Verduno, Italy
| | - Alessandro Gambella
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Jasna Metovic
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Daniele Tota
- Pathology Unit, Department of Medical Sciences, University of Turin, Via Santena 7, 10126 Turin, Italy
| | - Laura Carpenito
- Department of Pathology, Fondazione IRCCS Istituto Nazionale dei Tumori, Milan, Italy
- University of Milan, Milan, Italy
| | - Paolo Gasparri
- UOC di Anatomia Patologica, ASP Catania P.O. “Gravina”, Caltagirone, Italy
| | - Massimo Salvi
- Biolab, PolitoMed Lab, Department of Electronics and Telecommunications, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Turin, Italy
| |
Collapse
|
16
|
Beyond the colors: enhanced deep learning on invasive ductal carcinoma. Neural Comput Appl 2022. [DOI: 10.1007/s00521-022-07478-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
|
17
|
Umer MJ, Sharif M, Kadry S, Alharbi A. Multi-Class Classification of Breast Cancer Using 6B-Net with Deep Feature Fusion and Selection Method. J Pers Med 2022; 12:683. [PMID: 35629106 PMCID: PMC9146727 DOI: 10.3390/jpm12050683] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 01/17/2023] Open
Abstract
Breast cancer has now overtaken lung cancer as the world's most commonly diagnosed cancer, with thousands of new cases per year. Early detection and classification of breast cancer are necessary to overcome the death rate. Recently, many deep learning-based studies have been proposed for automatic diagnosis and classification of this deadly disease, using histopathology images. This study proposed a novel solution for multi-class breast cancer classification from histopathology images using deep learning. For this purpose, a novel 6B-Net deep CNN model, with feature fusion and selection mechanism, was developed for multi-class breast cancer classification. For the evaluation of the proposed method, two large, publicly available datasets, namely, BreaKHis, with eight classes containing 7909 images, and a breast cancer histopathology dataset, containing 3771 images of four classes, were used. The proposed method achieves a multi-class average accuracy of 94.20%, with a classification training time of 226 s in four classes of breast cancer, and a multi-class average accuracy of 90.10%, with a classification training time of 147 s in eight classes of breast cancer. The experimental outcomes show that the proposed method achieves the highest multi-class average accuracy for breast cancer classification, and hence, the proposed method can effectively be applied for early detection and classification of breast cancer to assist the pathologists in early and accurate diagnosis of breast cancer.
Collapse
Affiliation(s)
- Muhammad Junaid Umer
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Rawalpindi 47000, Pakistan;
| | - Muhammad Sharif
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Rawalpindi 47000, Pakistan;
| | - Seifedine Kadry
- Department of Applied Data Science, Noroff University College, 4612 Kristiansand, Norway;
| | - Abdullah Alharbi
- Department of Information Technology, College of Computers and Information Technology, Taif University, P.O. Box 11099, Taif 21944, Saudi Arabia;
| |
Collapse
|
18
|
Liu H, Xu WD, Shang ZH, Wang XD, Zhou HY, Ma KW, Zhou H, Qi JL, Jiang JR, Tan LL, Zeng HM, Cai HJ, Wang KS, Qian YL. Breast Cancer Molecular Subtype Prediction on Pathological Images with Discriminative Patch Selection and Multi-Instance Learning. Front Oncol 2022; 12:858453. [PMID: 35494021 PMCID: PMC9046851 DOI: 10.3389/fonc.2022.858453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 03/14/2022] [Indexed: 11/13/2022] Open
Abstract
Molecular subtypes of breast cancer are important references to personalized clinical treatment. For cost and labor savings, only one of the patient’s paraffin blocks is usually selected for subsequent immunohistochemistry (IHC) to obtain molecular subtypes. Inevitable block sampling error is risky due to the tumor heterogeneity and could result in a delay in treatment. Molecular subtype prediction from conventional H&E pathological whole slide images (WSI) using the AI method is useful and critical to assist pathologists to pre-screen proper paraffin block for IHC. It is a challenging task since only WSI-level labels of molecular subtypes from IHC can be obtained without detailed local region information. Gigapixel WSIs are divided into a huge amount of patches to be computationally feasible for deep learning, while with coarse slide-level labels, patch-based methods may suffer from abundant noise patches, such as folds, overstained regions, or non-tumor tissues. A weakly supervised learning framework based on discriminative patch selection and multi-instance learning was proposed for breast cancer molecular subtype prediction from H&E WSIs. Firstly, co-teaching strategy using two networks was adopted to learn molecular subtype representations and filter out some noise patches. Then, a balanced sampling strategy was used to handle the imbalance in subtypes in the dataset. In addition, a noise patch filtering algorithm that used local outlier factor based on cluster centers was proposed to further select discriminative patches. Finally, a loss function integrating local patch with global slide constraint information was used to fine-tune MIL framework on obtained discriminative patches and further improve the prediction performance of molecular subtyping. The experimental results confirmed the effectiveness of the proposed AI method and our models outperformed even senior pathologists, which has the potential to assist pathologists to pre-screen paraffin blocks for IHC in clinic.
Collapse
Affiliation(s)
- Hong Liu
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- *Correspondence: Hong Liu, ; Kuan-Song Wang,
| | - Wen-Dong Xu
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Zi-Hao Shang
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Xiang-Dong Wang
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| | - Hai-Yan Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Ke-Wen Ma
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Huan Zhou
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Lin Qi
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Jia-Rui Jiang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Li-Lan Tan
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui-Min Zeng
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hui-Juan Cai
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Kuan-Song Wang
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
- School of Basic Medical Science, Central South University, Changsha, China
- *Correspondence: Hong Liu, ; Kuan-Song Wang,
| | - Yue-Liang Qian
- Beijing Key Laboratory of Mobile Computing and Pervasive Device, Institute of Computing Technology, Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
19
|
Al Noumah W, Jafar A, Al Joumaa K. Using parallel pre-trained types of DCNN model to predict breast cancer with color normalization. BMC Res Notes 2022; 15:14. [PMID: 35012681 PMCID: PMC8751220 DOI: 10.1186/s13104-021-05902-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Accepted: 12/23/2021] [Indexed: 11/23/2022] Open
Abstract
Objective Breast cancer is the most common among women, and it causes many deaths every year. Early diagnosis increases the chance of cure through treatment. The traditional manual diagnosis requires effort and time from pathological experts, as it needs a joint experience of a number of pathologists. Diagnostic mistakes can lead to catastrophic results and endanger the lives of patients. The presence of an expert system that is able to specify whether the examined tissue is healthy or not, thus improves the quality of diagnosis and saves the time of experts. In this paper, a model capable of classifying breast cancer anatomy by making use of a pre-trained DCNN has been proposed. To build this model, first of all the image should be color stained by using Vahadane algorithm, then the model which combines three pre-trained DCNN (Xception, NASNet and Inceptoin_Resnet_V2) should be built in parallel, then the three branches should be aggregated to take advantage of each other. The suggested model was tested under different values of threshold ratios and also compared with other models. Results The proposed model on the BreaKHis dataset achieved 98% accuracy, which is better than the accuracy of other models used in this field. Supplementary Information The online version contains supplementary material available at 10.1186/s13104-021-05902-3.
Collapse
Affiliation(s)
- William Al Noumah
- Department of Informatics, Higher Institute for Applied Sciences and Technology, Damascus, Syria.
| | - Assef Jafar
- Department of Informatics, Higher Institute for Applied Sciences and Technology, Damascus, Syria
| | - Kadan Al Joumaa
- Department of Informatics, Higher Institute for Applied Sciences and Technology, Damascus, Syria
| |
Collapse
|
20
|
Lagree A, Shiner A, Alera MA, Fleshner L, Law E, Law B, Lu FI, Dodington D, Gandhi S, Slodkowska EA, Shenfield A, Jerzak KJ, Sadeghi-Naini A, Tran WT. Assessment of Digital Pathology Imaging Biomarkers Associated with Breast Cancer Histologic Grade. Curr Oncol 2021; 28:4298-4316. [PMID: 34898544 PMCID: PMC8628688 DOI: 10.3390/curroncol28060366] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 10/17/2021] [Accepted: 10/23/2021] [Indexed: 12/31/2022] Open
Abstract
Background: Evaluating histologic grade for breast cancer diagnosis is standard and associated with prognostic outcomes. Current challenges include the time required for manual microscopic evaluation and interobserver variability. This study proposes a computer-aided diagnostic (CAD) pipeline for grading tumors using artificial intelligence. Methods: There were 138 patients included in this retrospective study. Breast core biopsy slides were prepared using standard laboratory techniques, digitized, and pre-processed for analysis. Deep convolutional neural networks (CNNs) were developed to identify the regions of interest containing malignant cells and to segment tumor nuclei. Imaging-based features associated with spatial parameters were extracted from the segmented regions of interest (ROIs). Clinical datasets and pathologic biomarkers (estrogen receptor, progesterone receptor, and human epidermal growth factor 2) were collected from all study subjects. Pathologic, clinical, and imaging-based features were input into machine learning (ML) models to classify histologic grade, and model performances were tested against ground-truth labels at the patient-level. Classification performances were evaluated using receiver-operating characteristic (ROC) analysis. Results: Multiparametric feature sets, containing both clinical and imaging-based features, demonstrated high classification performance. Using imaging-derived markers alone, the classification performance demonstrated an area under the curve (AUC) of 0.745, while modeling these features with other pathologic biomarkers yielded an AUC of 0.836. Conclusion: These results demonstrate an association between tumor nuclear spatial features and tumor grade. If further validated, these systems may be implemented into pathology CADs and can assist pathologists to expeditiously grade tumors at the time of diagnosis and to help guide clinical decisions.
Collapse
Affiliation(s)
- Andrew Lagree
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Temerty Centre for AI Research and Education, University of Toronto, Toronto, ON M5S 1A8, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Audrey Shiner
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Marie Angeli Alera
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Lauren Fleshner
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Ethan Law
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Brianna Law
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
| | - Fang-I Lu
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (D.D.); (E.A.S.)
| | - David Dodington
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (D.D.); (E.A.S.)
| | - Sonal Gandhi
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada;
| | - Elzbieta A. Slodkowska
- Department of Laboratory Medicine and Molecular Diagnostics, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (D.D.); (E.A.S.)
| | - Alex Shenfield
- Department of Engineering and Mathematics, Sheffield Hallam University, Howard St, Sheffield S1 1WB, UK;
| | - Katarzyna J. Jerzak
- Division of Medical Oncology, Department of Medicine, University of Toronto, Toronto, ON M5S 3H2, Canada;
| | - Ali Sadeghi-Naini
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Department of Electrical Engineering and Computer Science, York University, Toronto, ON M3J 2S5, Canada
| | - William T. Tran
- Department of Radiation Oncology, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (A.L.); (A.S.); (M.A.A.); (L.F.); (E.L.); (B.L.); (A.S.-N.)
- Biological Sciences Platform, Sunnybrook Research Institute, Toronto, ON M4N 3M5, Canada
- Temerty Centre for AI Research and Education, University of Toronto, Toronto, ON M5S 1A8, Canada
- Radiogenomics Laboratory, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada; (F.-I.L.); (S.G.)
- Department of Radiation Oncology, University of Toronto, Toronto, ON M5T 1P5, Canada
- Correspondence: ; Tel.: +1-416-480-6100 (ext. 3746)
| |
Collapse
|
21
|
Zhang J, Mu N, Liu L, Xie J, Feng H, Yao J, Chen T, Zhu W. Highly sensitive detection of malignant glioma cells using metamaterial-inspired THz biosensor based on electromagnetically induced transparency. Biosens Bioelectron 2021; 185:113241. [PMID: 33905964 DOI: 10.1016/j.bios.2021.113241] [Citation(s) in RCA: 59] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 04/06/2021] [Accepted: 04/07/2021] [Indexed: 12/30/2022]
Abstract
Metamaterial-inspired biosensors have been extensively studied recently years for fast and low-cost THz detection. However, only the variation of the resonance frequency has been closely concerned in such sensors so far, whiles the magnitude variation, which also provide important information of the analyte, has not been sufficiently analyzed. In this paper, by the observation of two degree of variations, we propose a label-free biosensing approach for molecular classification of glioma cells. The metamaterial biosensor consisting of cut wires and split ring resonators are proposed to realize polarization-independent electromagnetic induced transparency (EIT) at THz frequencies. Simulated results show that the EIT-like resonance experiences both resonance frequency and magnitude variations when the properties of analyte change, which is further explained with coupled oscillators model theory. The theoretical sensitivity of the biosensor is evaluated up to 496.01 GHz/RIU. In experiments, two types of glioma cells (mutant and wild-type) are cultured on the biosensor surface. The dependences of frequency shifts and the peak magnitude variations on the cells concentrations for different types give new perspective for molecular classification of glioma cells. The measured results indicate that the mutant and wild-type glioma cells can be distinguished directly by observing both the variations of EIT resonance frequency and magnitude at any cells concentrations without antibody introduction. Our metamaterial-based biosensor shows a great potential in the recognition of molecule types of glioma cells, opening alternative way to sensitive biosensing technology.
Collapse
Affiliation(s)
- Jin Zhang
- Department of Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Ning Mu
- Department of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Collaborative Innovation Center for Brain Science, Chongqing, China
| | - Longhai Liu
- Tianjin University, Tianjin, 300072, China; Advantest (China) Co., Ltd, Shanghai, 201203, China
| | - Jianhua Xie
- Advantest (China) Co., Ltd, Shanghai, 201203, China
| | - Hua Feng
- Department of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Collaborative Innovation Center for Brain Science, Chongqing, China
| | | | - Tunan Chen
- Department of Neurosurgery, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China; Collaborative Innovation Center for Brain Science, Chongqing, China.
| | - Weiren Zhu
- Department of Electronic Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
22
|
Ramot Y, Deshpande A, Morello V, Michieli P, Shlomov T, Nyska A. Microscope-Based Automated Quantification of Liver Fibrosis in Mice Using a Deep Learning Algorithm. Toxicol Pathol 2021; 49:1126-1133. [PMID: 33769147 DOI: 10.1177/01926233211003866] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
In preclinical studies that involve animal models for hepatic fibrosis, accurate quantification of the fibrosis is of utmost importance. The use of digital image analysis based on deep learning artificial intelligence (AI) algorithms can facilitate accurate evaluation of liver fibrosis in these models. In the present study, we compared the quantitative evaluation of collagen proportionate area in the carbon tetrachloride model of liver fibrosis in the mouse by a newly developed AI algorithm to the semiquantitative assessment of liver fibrosis performed by a board-certified toxicologic pathologist. We found an excellent correlation between the 2 methods of assessment, most evident in the higher magnification (×40) as compared to the lower magnification (×10). These findings strengthen the confidence of using digital tools in the toxicologic pathology field as an adjunct to an expert toxicologic pathologist.
Collapse
Affiliation(s)
- Yuval Ramot
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Department of Dermatology, 58884Hadassah Medical Center, Jerusalem, Israel
| | | | | | - Paolo Michieli
- AgomAb Therapeutics NV, Gent, Belgium.,Molecular Biotechnology Center, University of Torino, Torino, Italy
| | - Tehila Shlomov
- Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel.,Hadassah Medical Center, Jerusalem, Israel
| | - Abraham Nyska
- Consultant in Toxicologic Pathology, 26745Tel Aviv and Tel Aviv University, Israel
| |
Collapse
|
23
|
Abstract
Histopathological images (HIs) are the gold standard for evaluating some types of tumors for cancer diagnosis. The analysis of such images is time and resource-consuming and very challenging even for experienced pathologists, resulting in inter-observer and intra-observer disagreements. One of the ways of accelerating such an analysis is to use computer-aided diagnosis (CAD) systems. This paper presents a review on machine learning methods for histopathological image analysis, including shallow and deep learning methods. We also cover the most common tasks in HI analysis, such as segmentation and feature extraction. Besides, we present a list of publicly available and private datasets that have been used in HI research.
Collapse
|
24
|
Drabent P, Mitri R, Le Naour G, Hermine O, Rossignol J, Molina TJ, Barete S, Fraitag S. A New Digital Method for Counting Mast Cells in Cutaneous Specific Lesions of Mastocytosis: A Series of Adult Cases of Mastocytosis With Clinical-Pathological Correlations. Am J Dermatopathol 2021; 43:35-41. [PMID: 32568831 DOI: 10.1097/dad.0000000000001705] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
ABSTRACT Cutaneous mastocytosis is characterized by the abnormal accumulation of mast cells in the skin. However, mast cell counting is not always easy and reproducible with classical methods. This work aims to demonstrate the reliability, usability, and virtues of a new software used on digital tablets for counting mast cells in cutaneous specific lesions of mastocytosis, to assess differences in mast cell counts between clinical subtypes of mastocytosis in the skin, and to consider the feasibility of applying a diagnostic mast cell count cutoff to urticaria pigmentosa, which is the most frequent form of cutaneous mastocytosis. Using a new digital tablet software that was accessible by multiple observers through its own wireless network and allowed high resolution of the image without data compression, we counted the number of mast cells on slides of patients and control skins immunostained for CD117. We found that our counting method was highly reproducible and that the new software allowed very quick counting. We evidenced strong differences in the mast cell count between most of the clinical subtypes of mastocytosis in the skin. However, when applied to a subset of patients with urticaria pigmentosa, a diagnostic cutoff in the mast cell count lacked sensitivity. Thus, our digital method for counting CD117-immunostained mast cells was highly accurate and was of a significant value for the diagnosis of mastocytosis in the skin. However, some subtypes with low mast cell counts will still require the application of additional diagnostic criteria.
Collapse
Affiliation(s)
- Philippe Drabent
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Necker-Enfants Malades, APHP, Paris, France
- Sorbonne Université, Paris, France
| | - Rana Mitri
- Service d'Anatomie et Cytologie Pathologiques, Centre Hospitalier Intercommunal de Créteil, Créteil, France
| | - Gilles Le Naour
- Sorbonne Université, Paris, France
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Pitié Salpêtrière, APHP, Paris France
| | - Olivier Hermine
- Service d'Hématologie Adulte, Hôpital Necker-Enfants Malades, APHP, Paris, France
- CEREMAST-Centre de Référence des Mastocytoses, Hôpital Necker-Enfants Malades, Paris, France
| | - Julien Rossignol
- CEREMAST-Centre de Référence des Mastocytoses, Hôpital Necker-Enfants Malades, Paris, France
| | - Thierry Jo Molina
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Necker-Enfants Malades, APHP, Paris, France
- Université Paris-Descartes, Université de Paris, Paris, France ; and
| | - Stéphane Barete
- CEREMAST-Centre de Référence des Mastocytoses, Hôpital Necker-Enfants Malades, Paris, France
- Service de Médecine Interne, Hôpital Pitié Salpêtrière, APHP, Paris, France
| | - Sylvie Fraitag
- Service d'Anatomie et Cytologie Pathologiques, Hôpital Necker-Enfants Malades, APHP, Paris, France
- CEREMAST-Centre de Référence des Mastocytoses, Hôpital Necker-Enfants Malades, Paris, France
- Université Paris-Descartes, Université de Paris, Paris, France ; and
| |
Collapse
|
25
|
Wan T, Zhao L, Feng H, Li D, Tong C, Qin Z. Robust nuclei segmentation in histopathology using ASPPU-Net and boundary refinement. Neurocomputing 2020. [DOI: 10.1016/j.neucom.2019.08.103] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
|
26
|
Pantanowitz L, Hartman D, Qi Y, Cho EY, Suh B, Paeng K, Dhir R, Michelow P, Hazelhurst S, Song SY, Cho SY. Accuracy and efficiency of an artificial intelligence tool when counting breast mitoses. Diagn Pathol 2020; 15:80. [PMID: 32622359 PMCID: PMC7335442 DOI: 10.1186/s13000-020-00995-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2020] [Accepted: 06/25/2020] [Indexed: 12/17/2022] Open
Abstract
BACKGROUND The mitotic count in breast carcinoma is an important prognostic marker. Unfortunately substantial inter- and intra-laboratory variation exists when pathologists manually count mitotic figures. Artificial intelligence (AI) coupled with whole slide imaging offers a potential solution to this problem. The aim of this study was to accordingly critique an AI tool developed to quantify mitotic figures in whole slide images of invasive breast ductal carcinoma. METHODS A representative H&E slide from 320 breast invasive ductal carcinoma cases was scanned at 40x magnification. Ten expert pathologists from two academic medical centers labeled mitotic figures in whole slide images to train and validate an AI algorithm to detect and count mitoses. Thereafter, 24 readers of varying expertise were asked to count mitotic figures with and without AI support in 140 high-power fields derived from a separate dataset. Their accuracy and efficiency of performing these tasks were calculated and statistical comparisons performed. RESULTS For each experience level the accuracy, precision and sensitivity of counting mitoses by users improved with AI support. There were 21 readers (87.5%) that identified more mitoses using AI support and 13 reviewers (54.2%) that decreased the quantity of falsely flagged mitoses with AI. More time was spent on this task for most participants when not provided with AI support. AI assistance resulted in an overall time savings of 27.8%. CONCLUSIONS This study demonstrates that pathology end-users were more accurate and efficient at quantifying mitotic figures in digital images of invasive breast carcinoma with the aid of AI. Higher inter-pathologist agreement with AI assistance suggests that such algorithms can also help standardize practice. Not surprisingly, there is much enthusiasm in pathology regarding the prospect of using AI in routine practice to perform mundane tasks such as counting mitoses.
Collapse
Affiliation(s)
- Liron Pantanowitz
- Department of Pathology, University of Pittsburgh Medical Center Cancer Pavilion, Suite 201, 5150 Centre Ave, Pittsburgh, PA, 15232, USA.
- Department of Anatomical Pathology, University of the Witwatersrand and National Health Laboratory Services, Johannesburg, South Africa.
| | - Douglas Hartman
- Department of Pathology, University of Pittsburgh Medical Center Cancer Pavilion, Suite 201, 5150 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Yan Qi
- School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Eun Yoon Cho
- Department of Pathology, Samsung Medical Center, Seoul, South Korea
| | | | | | - Rajiv Dhir
- Department of Pathology, University of Pittsburgh Medical Center Cancer Pavilion, Suite 201, 5150 Centre Ave, Pittsburgh, PA, 15232, USA
| | - Pamela Michelow
- Department of Anatomical Pathology, University of the Witwatersrand and National Health Laboratory Services, Johannesburg, South Africa
| | - Scott Hazelhurst
- School of Electrical & Information Engineering and Sydney Brenner Institute for Molecular Bioscience, University of the Witwatersrand, Johannesburg, South Africa
| | - Sang Yong Song
- Department of Pathology, Samsung Medical Center, Seoul, South Korea
| | - Soo Youn Cho
- Department of Pathology, Samsung Medical Center, Seoul, South Korea
| |
Collapse
|
27
|
Troy K, Liu Y, Hainer SJ. PathSTORM: a road to early cancer detection. Mol Cell Oncol 2020; 7:1776086. [PMID: 32944634 PMCID: PMC7469673 DOI: 10.1080/23723556.2020.1776086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Disruption of chromatin structure could enable early carcinogenesis by facilitating malignant transformation. Using stochastic optical reconstruction microscopy optimized for pathological tissue (PathSTORM), we uncovered a gradual decompaction of higher-order chromatin folding through progressive stages of carcinogenesis. We demonstrated potential detection of pre-cancerous genomic architecture not easily discernible by conventional pathology.
Collapse
Affiliation(s)
- Kris Troy
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| | - Yang Liu
- Biomedical Optical Imaging Laboratory, Departments of Medicine and Bioengineering, University of Pittsburgh, Pittsburgh, PA, USA
| | - Sarah J Hainer
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
28
|
Aeffner F, Adissu HA, Boyle MC, Cardiff RD, Hagendorn E, Hoenerhoff MJ, Klopfleisch R, Newbigging S, Schaudien D, Turner O, Wilson K. Digital Microscopy, Image Analysis, and Virtual Slide Repository. ILAR J 2019; 59:66-79. [PMID: 30535284 DOI: 10.1093/ilar/ily007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Advancements in technology and digitization have ushered in novel ways of enhancing tissue-based research via digital microscopy and image analysis. Whole slide imaging scanners enable digitization of histology slides to be stored in virtual slide repositories and to be viewed via computers instead of microscopes. Easier and faster sharing of histologic images for teaching and consultation, improved storage and preservation of quality of stained slides, and annotation of features of interest in the digital slides are just a few of the advantages of this technology. Combined with the development of software for digital image analysis, digital slides further pave the way for the development of tools that extract quantitative data from tissue-based studies. This review introduces digital microscopy and pathology, and addresses technical and scientific considerations in slide scanning, quantitative image analysis, and slide repositories. It also highlights the current state of the technology and factors that need to be taken into account to insure optimal utility, including preanalytical considerations and the importance of involving a pathologist in all major steps along the digital microscopy and pathology workflow.
Collapse
Affiliation(s)
- Famke Aeffner
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Hibret A Adissu
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Michael C Boyle
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Robert D Cardiff
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Erik Hagendorn
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Mark J Hoenerhoff
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Robert Klopfleisch
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Susan Newbigging
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Dirk Schaudien
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Oliver Turner
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Kristin Wilson
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| |
Collapse
|
29
|
Zheng H, Gasbarrino K, Veinot JP, Lai C, Daskalopoulou SS. New Quantitative Digital Image Analysis Method of Histological Features of Carotid Atherosclerotic Plaques. Eur J Vasc Endovasc Surg 2019; 58:654-663. [PMID: 31543397 DOI: 10.1016/j.ejvs.2019.07.015] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2018] [Revised: 07/03/2019] [Accepted: 07/13/2019] [Indexed: 11/19/2022]
Abstract
OBJECTIVE Atherosclerosis and its thrombotic complications are major causes of morbidity and mortality worldwide. Plaque stability assessment is considered to be important for both clinical and fundamental applications. The current gold standard method to investigate plaque stability is performed by histological assessment of plaque features using semi-quantitative classifications. However, these assessments can be limited by subjectivity and variability. Thus, the aim was to develop a new digital image analysis method to measure quantitatively individual plaque features that is more precise than existing semi-quantitative methods. METHODS A quantitative method was developed using Image Pro Primer software. Carotid plaque specimens were obtained from patients who underwent carotid endarterectomy and categorised according to stability (definitely stable, probably stable, probably unstable, definitely unstable) based on the gold standard semi-quantitative method that assesses 10 histological plaque features. Using the new quantitative method, plaque features (n = 15) from each stability grade were then analysed by two independent raters. For the semi-quantitative analysis, quadratic weighted Cohen's kappa was used to test intra- and inter-rater reliability, while for the quantitative analysis, intraclass correlation coefficients (ICCs) were assessed. RESULTS Intra-rater reliability demonstrated almost perfect agreement between both methods (Cohen's kappa range 0.831-0.969, ICC range 0.848-1.000). However, inter-rater reliability demonstrated mainly fair to moderate agreement (Cohen's kappa range 0.341-0.778) for the semi-quantitative analysis, while the digital image analysis method performed most optimally regarding reproducibility, yielding high ICCs close to 1 (ICC range 0.816-0.999). Using quantitative measurements, a statistically significant proportion of the individual plaque features (p < .05) were re-classified from one grade to another (shift by one) under the semi-quantitative classification. CONCLUSION A new quantitative digital image analysis was developed for the accurate assessment of histological plaque features, which demonstrated higher precision than the gold standard semi-quantitative methods, as measured by between and within rater analysis. Moreover, quantitative image analysis of histological plaque features provided more detailed insight into plaque morphology and composition.
Collapse
Affiliation(s)
- Huaien Zheng
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre (MUHC), McGill University, Montreal, QC, Canada
| | - Karina Gasbarrino
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre (MUHC), McGill University, Montreal, QC, Canada
| | - John P Veinot
- Department of Pathology and Laboratory Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Chi Lai
- Department of Pathology and Laboratory Medicine, University of Ottawa Heart Institute, Ottawa, ON, Canada
| | - Stella S Daskalopoulou
- Division of Experimental Medicine, Department of Medicine, Faculty of Medicine, Research Institute of the McGill University Health Centre (MUHC), McGill University, Montreal, QC, Canada.
| |
Collapse
|
30
|
Sukhia K, Riaz M, Ghafoor A, Ali S, Iltaf N. Content-based histopathological image retrieval using multi-scale and multichannel decoder based LTP. Biomed Signal Process Control 2019. [DOI: 10.1016/j.bspc.2019.101582] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
31
|
Murtaza G, Shuib L, Abdul Wahab AW, Mujtaba G, Mujtaba G, Nweke HF, Al-garadi MA, Zulfiqar F, Raza G, Azmi NA. Deep learning-based breast cancer classification through medical imaging modalities: state of the art and research challenges. Artif Intell Rev 2019. [DOI: 10.1007/s10462-019-09716-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
32
|
Deep Learning in the Biomedical Applications: Recent and Future Status. APPLIED SCIENCES-BASEL 2019. [DOI: 10.3390/app9081526] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Deep neural networks represent, nowadays, the most effective machine learning technology in biomedical domain. In this domain, the different areas of interest concern the Omics (study of the genome—genomics—and proteins—transcriptomics, proteomics, and metabolomics), bioimaging (study of biological cell and tissue), medical imaging (study of the human organs by creating visual representations), BBMI (study of the brain and body machine interface) and public and medical health management (PmHM). This paper reviews the major deep learning concepts pertinent to such biomedical applications. Concise overviews are provided for the Omics and the BBMI. We end our analysis with a critical discussion, interpretation and relevant open challenges.
Collapse
|
33
|
Aeffner F, Zarella MD, Buchbinder N, Bui MM, Goodman MR, Hartman DJ, Lujan GM, Molani MA, Parwani AV, Lillard K, Turner OC, Vemuri VNP, Yuil-Valdes AG, Bowman D. Introduction to Digital Image Analysis in Whole-slide Imaging: A White Paper from the Digital Pathology Association. J Pathol Inform 2019; 10:9. [PMID: 30984469 PMCID: PMC6437786 DOI: 10.4103/jpi.jpi_82_18] [Citation(s) in RCA: 203] [Impact Index Per Article: 33.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2018] [Accepted: 12/11/2018] [Indexed: 12/22/2022] Open
Abstract
The advent of whole-slide imaging in digital pathology has brought about the advancement of computer-aided examination of tissue via digital image analysis. Digitized slides can now be easily annotated and analyzed via a variety of algorithms. This study reviews the fundamentals of tissue image analysis and aims to provide pathologists with basic information regarding the features, applications, and general workflow of these new tools. The review gives an overview of the basic categories of software solutions available, potential analysis strategies, technical considerations, and general algorithm readouts. Advantages and limitations of tissue image analysis are discussed, and emerging concepts, such as artificial intelligence and machine learning, are introduced. Finally, examples of how digital image analysis tools are currently being used in diagnostic laboratories, translational research, and drug development are discussed.
Collapse
Affiliation(s)
- Famke Aeffner
- Amgen Inc., Amgen Research, Comparative Biology and Safety Sciences, South San Francisco, CA, USA
| | - Mark D Zarella
- Department of Pathology and Laboratory Medicine, Drexel University, College of Medicine, Philadelphia, PA, USA
| | | | - Marilyn M Bui
- Department of Pathology, Moffitt Cancer Center, Tampa, FL, USA
| | | | | | | | - Mariam A Molani
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | - Anil V Parwani
- The Ohio State University Medical Center, Columbus, OH, USA
| | | | - Oliver C Turner
- Novartis, Novartis Institutes for BioMedical Research, Preclinical Safety, East Hannover, NJ, USA
| | | | - Ana G Yuil-Valdes
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE, USA
| | | |
Collapse
|
34
|
Glotsos D, Kostopoulos S, Ravazoula P, Cavouras D. Image quilting and wavelet fusion for creation of synthetic microscopy nuclei images. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2018; 162:177-186. [PMID: 29903484 DOI: 10.1016/j.cmpb.2018.05.023] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Revised: 05/09/2018] [Accepted: 05/16/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND AND OBJECTIVE In this study a texture simulation methodology is proposed for composing synthetic tissue microscopy images that could serve as a quantitative gold standard for the evaluation of the reliability, accuracy and performance of segmentation algorithms in computer-aided diagnosis. METHODS A library of background and nuclei regions was generated using pre-segmented Haematoxylin and Eosin images of brain tumours. Background image samples were used as input to an image quilting algorithm that produced the synthetic background image. Randomly selected pre-segmented nuclei were randomly fused on the synthetic background using a wavelet-based fusion approach. To investigate whether the produced synthetic images are meaningful and similar to real world images, two different tests were performed, one qualitative by an experienced histopathologist and one quantitative using the normalized mutual information and the Kullback-Leibler tests. To illustrate the challenges that synthetic images may pose to object recognition algorithms, two segmentation methodologies were utilized for nuclei detection, one based on the Otsu thresholding and another based on the seeded region growing approach. RESULTS Results showed a satisfactory to good resemblance of the synthetic with the real world images according to both qualitative and quantitative tests. The segmentation accuracy was slightly higher for the seeded region growing algorithm (87.2%) than the Otsu's algorithm (86.3%). CONCLUSIONS Since we know the exact coordinates of the regions of interest within the synthesised images, these images could then serve as a 'gold standard' for evaluation of segmentation algorithms in computer-aided diagnosis in tissue microscopy.
Collapse
Affiliation(s)
- Dimitris Glotsos
- Medical Image and Signal Processing (medisp) Lab, Department of Biomedical Engineering, Technological Educational Institute of Athens, Ag. Spyridonos Street, Egaleo, 122 10 Athens, Greece.
| | - Spiros Kostopoulos
- Medical Image and Signal Processing (medisp) Lab, Department of Biomedical Engineering, Technological Educational Institute of Athens, Ag. Spyridonos Street, Egaleo, 122 10 Athens, Greece
| | | | - Dionisis Cavouras
- Medical Image and Signal Processing (medisp) Lab, Department of Biomedical Engineering, Technological Educational Institute of Athens, Ag. Spyridonos Street, Egaleo, 122 10 Athens, Greece
| |
Collapse
|
35
|
Robertson S, Azizpour H, Smith K, Hartman J. Digital image analysis in breast pathology-from image processing techniques to artificial intelligence. Transl Res 2018; 194:19-35. [PMID: 29175265 DOI: 10.1016/j.trsl.2017.10.010] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/01/2017] [Revised: 10/28/2017] [Accepted: 10/30/2017] [Indexed: 01/04/2023]
Abstract
Breast cancer is the most common malignant disease in women worldwide. In recent decades, earlier diagnosis and better adjuvant therapy have substantially improved patient outcome. Diagnosis by histopathology has proven to be instrumental to guide breast cancer treatment, but new challenges have emerged as our increasing understanding of cancer over the years has revealed its complex nature. As patient demand for personalized breast cancer therapy grows, we face an urgent need for more precise biomarker assessment and more accurate histopathologic breast cancer diagnosis to make better therapy decisions. The digitization of pathology data has opened the door to faster, more reproducible, and more precise diagnoses through computerized image analysis. Software to assist diagnostic breast pathology through image processing techniques have been around for years. But recent breakthroughs in artificial intelligence (AI) promise to fundamentally change the way we detect and treat breast cancer in the near future. Machine learning, a subfield of AI that applies statistical methods to learn from data, has seen an explosion of interest in recent years because of its ability to recognize patterns in data with less need for human instruction. One technique in particular, known as deep learning, has produced groundbreaking results in many important problems including image classification and speech recognition. In this review, we will cover the use of AI and deep learning in diagnostic breast pathology, and other recent developments in digital image analysis.
Collapse
Affiliation(s)
- Stephanie Robertson
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden
| | - Hossein Azizpour
- School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden; Science for Life Laboratory, Stockholm, Sweden
| | - Kevin Smith
- School of Computer Science and Communication, KTH Royal Institute of Technology, Stockholm, Sweden; Science for Life Laboratory, Stockholm, Sweden
| | - Johan Hartman
- Department of Oncology-Pathology, Karolinska Institutet, Stockholm, Sweden; Department of Clinical Pathology and Cytology, Karolinska University Laboratory, Stockholm, Sweden; Stockholm South General Hospital, Stockholm, Sweden.
| |
Collapse
|