51
|
Schneider L, Laiouar-Pedari S, Kuntz S, Krieghoff-Henning E, Hekler A, Kather JN, Gaiser T, Fröhling S, Brinker TJ. Integration of deep learning-based image analysis and genomic data in cancer pathology: A systematic review. Eur J Cancer 2021; 160:80-91. [PMID: 34810047 DOI: 10.1016/j.ejca.2021.10.007] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Accepted: 10/11/2021] [Indexed: 02/07/2023]
Abstract
BACKGROUND Over the past decade, the development of molecular high-throughput methods (omics) increased rapidly and provided new insights for cancer research. In parallel, deep learning approaches revealed the enormous potential for medical image analysis, especially in digital pathology. Combining image and omics data with deep learning tools may enable the discovery of new cancer biomarkers and a more precise prediction of patient prognosis. This systematic review addresses different multimodal fusion methods of convolutional neural network-based image analyses with omics data, focussing on the impact of data combination on the classification performance. METHODS PubMed was screened for peer-reviewed articles published in English between January 2015 and June 2021 by two independent researchers. Search terms related to deep learning, digital pathology, omics, and multimodal fusion were combined. RESULTS We identified a total of 11 studies meeting the inclusion criteria, namely studies that used convolutional neural networks for haematoxylin and eosin image analysis of patients with cancer in combination with integrated omics data. Publications were categorised according to their endpoints: 7 studies focused on survival analysis and 4 studies on prediction of cancer subtypes, malignancy or microsatellite instability with spatial analysis. CONCLUSIONS Image-based classifiers already show high performances in prognostic and predictive cancer diagnostics. The integration of omics data led to improved performance in all studies described here. However, these are very early studies that still require external validation to demonstrate their generalisability and robustness. Further and more comprehensive studies with larger sample sizes are needed to evaluate performance and determine clinical benefits.
Collapse
Affiliation(s)
- Lucas Schneider
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Laiouar-Pedari
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Sara Kuntz
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Krieghoff-Henning
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Achim Hekler
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Jakob N Kather
- Department of Medicine III, RWTH Aachen University Hospital, Aachen, Germany; Medical Oncology, National Center for Tumour Diseases, University Hospital Heidelberg, Heidelberg, Germany
| | - Timo Gaiser
- Institute of Pathology, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Stefan Fröhling
- Translational Medical Oncology, National Center for Tumour Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
52
|
Bodelon C, Mullooly M, Pfeiffer RM, Fan S, Abubakar M, Lenz P, Vacek PM, Weaver DL, Herschorn SD, Johnson JM, Sprague BL, Hewitt S, Shepherd J, Malkov S, Keely PJ, Eliceiri KW, Sherman ME, Conklin MW, Gierach GL. Mammary collagen architecture and its association with mammographic density and lesion severity among women undergoing image-guided breast biopsy. Breast Cancer Res 2021; 23:105. [PMID: 34753492 PMCID: PMC8579610 DOI: 10.1186/s13058-021-01482-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Accepted: 10/26/2021] [Indexed: 12/20/2022] Open
Abstract
Background Elevated mammographic breast density is a strong breast cancer risk factor with poorly understood etiology. Increased deposition of collagen, one of the main fibrous proteins present in breast stroma, has been associated with increased mammographic density. Collagen fiber architecture has been linked to poor outcomes in breast cancer. However, relationships of quantitative collagen fiber features assessed in diagnostic biopsies with mammographic density and lesion severity are not well-established. Methods Clinically indicated breast biopsies from 65 in situ or invasive breast cancer cases and 73 frequency matched-controls with a benign biopsy result were used to measure collagen fiber features (length, straightness, width, alignment, orientation and density (fibers/µm2)) using second harmonic generation microscopy in up to three regions of interest (ROIs) per biopsy: normal, benign breast disease, and cancer. Local and global mammographic density volumes were quantified in the ipsilateral breast in pre-biopsy full-field digital mammograms. Associations of fibrillar collagen features with mammographic density and severity of biopsy diagnosis were evaluated using generalized estimating equation models with an independent correlation structure to account for multiple ROIs within each biopsy section. Results Collagen fiber density was positively associated with the proportion of stroma on the biopsy slide (p < 0.001) and with local percent mammographic density volume at both the biopsy target (p = 0.035) and within a 2 mm perilesional ring (p = 0.02), but not with global mammographic density measures. As severity of the breast biopsy diagnosis increased at the ROI level, collagen fibers tended to be less dense, shorter, straighter, thinner, and more aligned with one another (p < 0.05). Conclusions Collagen fiber density was positively associated with local, but not global, mammographic density, suggesting that collagen microarchitecture may not translate into macroscopic mammographic features. However, collagen fiber features may be markers of cancer risk and/or progression among women referred for biopsy based on abnormal breast imaging. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-021-01482-z.
Collapse
Affiliation(s)
- Clara Bodelon
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA.
| | - Maeve Mullooly
- Division of Population Health Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
| | - Ruth M Pfeiffer
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| | - Shaoqi Fan
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| | - Mustapha Abubakar
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| | - Petra Lenz
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| | - Pamela M Vacek
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, USA
| | - Donald L Weaver
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, USA
| | - Sally D Herschorn
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, USA
| | - Jason M Johnson
- The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Brian L Sprague
- University of Vermont College of Medicine and Vermont Cancer Center, Burlington, VT, USA
| | - Stephen Hewitt
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| | - John Shepherd
- University of Hawaii Cancer Center, Honolulu, HI, USA
| | | | - Patricia J Keely
- Department of Cell and Regenerative Biology and Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Ave., WIMR II Rm. 4528, Madison, WI, 53705, USA
| | - Kevin W Eliceiri
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | | | - Matthew W Conklin
- Department of Cell and Regenerative Biology and Carbone Cancer Center, School of Medicine and Public Health, University of Wisconsin-Madison, 1111 Highland Ave., WIMR II Rm. 4528, Madison, WI, 53705, USA.
| | - Gretchen L Gierach
- Division of Cancer Epidemiology and Genetics, National Cancer Institute, 9609 Medical Center Dr., Rm 7-E238, Bethesda, MD, 20892, USA
| |
Collapse
|
53
|
Convolutional neural network for human cancer types prediction by integrating protein interaction networks and omics data. Sci Rep 2021; 11:20691. [PMID: 34667236 PMCID: PMC8526703 DOI: 10.1038/s41598-021-98814-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 09/14/2021] [Indexed: 02/07/2023] Open
Abstract
Many studies have proven the power of gene expression profile in cancer identification, however, the explosive growth of genomics data increasing needs of tools for cancer diagnosis and prognosis in high accuracy and short times. Here, we collected 6136 human samples from 11 cancer types, and integrated their gene expression profiles and protein-protein interaction (PPI) network to generate 2D images with spectral clustering method. To predict normal samples and 11 cancer tumor types, the images of these 6136 human cancer network were separated into training and validation dataset to develop convolutional neural network (CNN). Our model showed 97.4% and 95.4% accuracies in identification of normal versus tumors and 11 cancer types, respectively. We also provided the results that tumors located in neighboring tissues or in the same cell types, would induce machine make error classification due to the similar gene expression profiles. Furthermore, we observed some patients may exhibit better prognosis if their tumors often misjudged into normal samples. As far as we know, we are the first to generate thousands of cancer networks to predict and classify multiple cancer types with CNN architecture. We believe that our model not only can be applied to cancer diagnosis and prognosis, but also promote the discovery of multiple cancer biomarkers.
Collapse
|
54
|
Tran KA, Kondrashova O, Bradley A, Williams ED, Pearson JV, Waddell N. Deep learning in cancer diagnosis, prognosis and treatment selection. Genome Med 2021; 13:152. [PMID: 34579788 PMCID: PMC8477474 DOI: 10.1186/s13073-021-00968-x] [Citation(s) in RCA: 270] [Impact Index Per Article: 90.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2020] [Accepted: 09/12/2021] [Indexed: 12/13/2022] Open
Abstract
Deep learning is a subdiscipline of artificial intelligence that uses a machine learning technique called artificial neural networks to extract patterns and make predictions from large data sets. The increasing adoption of deep learning across healthcare domains together with the availability of highly characterised cancer datasets has accelerated research into the utility of deep learning in the analysis of the complex biology of cancer. While early results are promising, this is a rapidly evolving field with new knowledge emerging in both cancer biology and deep learning. In this review, we provide an overview of emerging deep learning techniques and how they are being applied to oncology. We focus on the deep learning applications for omics data types, including genomic, methylation and transcriptomic data, as well as histopathology-based genomic inference, and provide perspectives on how the different data types can be integrated to develop decision support tools. We provide specific examples of how deep learning may be applied in cancer diagnosis, prognosis and treatment management. We also assess the current limitations and challenges for the application of deep learning in precision oncology, including the lack of phenotypically rich data and the need for more explainable deep learning models. Finally, we conclude with a discussion of how current obstacles can be overcome to enable future clinical utilisation of deep learning.
Collapse
Affiliation(s)
- Khoa A. Tran
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, 4059 Australia
| | - Olga Kondrashova
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| | - Andrew Bradley
- Faculty of Engineering, Queensland University of Technology (QUT), Brisbane, 4000 Australia
| | - Elizabeth D. Williams
- School of Biomedical Sciences, Faculty of Health, Queensland University of Technology (QUT), Brisbane, 4059 Australia
- Australian Prostate Cancer Research Centre - Queensland (APCRC-Q) and Queensland Bladder Cancer Initiative (QBCI), Brisbane, 4102 Australia
| | - John V. Pearson
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| | - Nicola Waddell
- Department of Genetics and Computational Biology, QIMR Berghofer Medical Research Institute, Brisbane, 4006 Australia
| |
Collapse
|
55
|
Xu H, Cong F, Hwang TH. Machine Learning and Artificial Intelligence-driven Spatial Analysis of the Tumor Immune Microenvironment in Pathology Slides. Eur Urol Focus 2021; 7:706-709. [PMID: 34353733 DOI: 10.1016/j.euf.2021.07.006] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Accepted: 07/21/2021] [Indexed: 12/27/2022]
Abstract
A better understanding of the tumor immune microenvironment (TIME) could lead to accurate diagnosis, prognosis, and treatment stratification. Although molecular analyses at the tissue and/or single cell level could reveal the cellular status of the tumor microenvironment, these approaches lack information related to spatial-level cellular distribution, co-organization, and cell-cell interaction in the TIME. With the emergence of computational pathology coupled with machine learning (ML) and artificial intelligence (AI), ML- and AI-driven spatial TIME analyses of pathology images could revolutionize our understanding of the highly heterogeneous and complex molecular architecture of the TIME. In this review we highlight recent studies on spatial TIME analysis of pathology slides using state-of-the-art ML and AI algorithms. PATIENT SUMMARY: This mini-review reports recent advances in machine learning and artificial intelligence for spatial analysis of the tumor immune microenvironment in pathology slides. This information can help in understanding the spatial heterogeneity and organization of cells in patient tumors.
Collapse
Affiliation(s)
- Hongming Xu
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Fengyu Cong
- School of Biomedical Engineering, Faculty of Electronic Information and Electrical Engineering, Dalian University of Technology, Dalian, China
| | - Tae Hyun Hwang
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, OH, USA.
| |
Collapse
|
56
|
Lara H, Li Z, Abels E, Aeffner F, Bui MM, ElGabry EA, Kozlowski C, Montalto MC, Parwani AV, Zarella MD, Bowman D, Rimm D, Pantanowitz L. Quantitative Image Analysis for Tissue Biomarker Use: A White Paper From the Digital Pathology Association. Appl Immunohistochem Mol Morphol 2021; 29:479-493. [PMID: 33734106 PMCID: PMC8354563 DOI: 10.1097/pai.0000000000000930] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 02/12/2021] [Indexed: 01/19/2023]
Abstract
Tissue biomarkers have been of increasing utility for scientific research, diagnosing disease, and treatment response prediction. There has been a steady shift away from qualitative assessment toward providing more quantitative scores for these biomarkers. The application of quantitative image analysis has thus become an indispensable tool for in-depth tissue biomarker interrogation in these contexts. This white paper reviews current technologies being employed for quantitative image analysis, their application and pitfalls, regulatory framework demands, and guidelines established for promoting their safe adoption in clinical practice.
Collapse
Affiliation(s)
- Haydee Lara
- GlaxoSmithKline-R&D, Cellular Biomarkers, Collegeville, PA
| | - Zaibo Li
- The Ohio State University, Columbus, OH
| | | | - Famke Aeffner
- Translational Safety and Bioanalytical Sciences, Amgen Research, Amgen Inc
| | | | | | | | | | | | | | | | - David Rimm
- Yale University School of Medicine, New Haven, CT
| | | |
Collapse
|
57
|
Klein C, Zeng Q, Arbaretaz F, Devêvre E, Calderaro J, Lomenie N, Maiuri MC. Artificial Intelligence for solid tumor diagnosis in digital pathology. Br J Pharmacol 2021; 178:4291-4315. [PMID: 34302297 DOI: 10.1111/bph.15633] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 02/05/2021] [Accepted: 02/07/2021] [Indexed: 11/30/2022] Open
Abstract
Tumor diagnosis relies on the visual examination of histological slides by pathologists through a microscope eyepiece. Digital pathology, the digitalization of histological slides at high magnification with slides scanners, has raised the opportunity to extract quantitative information thanks to image analysis. In the last decade, medical image analysis has made exceptional progress due to the development of artificial intelligence (AI) algorithms. AI has been successfully used in the field of medical imaging and more recently in digital pathology. The feasibility and usefulness of AI assisted pathology tasks have been demonstrated in the very last years and we can expect those developments to be applied on routine histopathology in the future. In this review, we will describe and illustrate this technique and present the most recent applications in the field of tumor histopathology.
Collapse
Affiliation(s)
- Christophe Klein
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Qinghe Zeng
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France.,Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Floriane Arbaretaz
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Estelle Devêvre
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| | - Julien Calderaro
- Département de pathologie, Hôpital Henri Mondor, Créteil, France
| | - Nicolas Lomenie
- Laboratoire d'informatique Paris Descartes (LIPADE), Université de Paris, Paris, France
| | - Maria Chiara Maiuri
- Centre de recherche des Cordeliers, Centre d'Imagerie, Histologie et Cytométrie (CHIC), INSERM, Sorbonne Université, Université de Paris, Paris, France
| |
Collapse
|
58
|
A Computational Tumor-Infiltrating Lymphocyte Assessment Method Comparable with Visual Reporting Guidelines for Triple-Negative Breast Cancer. EBioMedicine 2021; 70:103492. [PMID: 34280779 PMCID: PMC8318866 DOI: 10.1016/j.ebiom.2021.103492] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 06/30/2021] [Accepted: 06/30/2021] [Indexed: 12/21/2022] Open
Abstract
Background Tumor-infiltrating lymphocytes (TILs) are clinically significant in triple-negative breast cancer (TNBC). Although a standardized methodology for visual TILs assessment (VTA) exists, it has several inherent limitations. We established a deep learning-based computational TIL assessment (CTA) method broadly following VTA guideline and compared it with VTA for TNBC to determine the prognostic value of the CTA and a reasonable CTA workflow for clinical practice. Methods We trained three deep neural networks for nuclei segmentation, nuclei classification and necrosis classification to establish a CTA workflow. The automatic TIL (aTIL) score generated was compared with manual TIL (mTIL) scores provided by three pathologists in an Asian (n = 184) and a Caucasian (n = 117) TNBC cohort to evaluate scoring concordance and prognostic value. Findings The intraclass correlations (ICCs) between aTILs and mTILs varied from 0.40 to 0.70 in two cohorts. Multivariate Cox proportional hazards analysis revealed that the aTIL score was associated with disease free survival (DFS) in both cohorts, as either a continuous [hazard ratio (HR)=0.96, 95% CI 0.94–0.99] or dichotomous variable (HR=0.29, 95% CI 0.12–0.72). A higher C-index was observed in a composite mTIL/aTIL three-tier stratification model than in the dichotomous model, using either mTILs or aTILs alone. Interpretation The current study provides a useful tool for stromal TIL assessment and prognosis evaluation for patients with TNBC. A workflow integrating both VTA and CTA may aid pathologists in performing risk management and decision-making tasks.
Collapse
|
59
|
Choschzick M, Alyahiaoui M, Ciritsis A, Rossi C, Gut A, Hejduk P, Boss A. Deep learning for the standardized classification of Ki-67 in vulva carcinoma: A feasibility study. Heliyon 2021; 7:e07577. [PMID: 34386617 PMCID: PMC8346648 DOI: 10.1016/j.heliyon.2021.e07577] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Revised: 02/02/2021] [Accepted: 07/12/2021] [Indexed: 12/12/2022] Open
Abstract
Background The aim of this study is to demonstrate the feasibility of automatic classification of Ki-67 histological immunostainings in patients with squamous cell carcinoma of the vulva using a deep convolutional neural network (dCNN). Material and methods For evaluation of the dCNN, we used 55 well characterized squamous cell carcinomas of the vulva in a tissue microarray (TMA) format in this retrospective study. The tumor specimens were classified in 3 different categories C1 (0-2%), C2 (2-20%) and C3 (>20%), representing the relation of the number of KI-67 positive tumor cells to all cancer cells on the TMA spot. Representative areas of the spots were manually labeled by extracting images of 351 × 280 pixels. A dCNN with 13 convolutional layers was used for the evaluation. Two independent pathologists classified 45 labeled images in order to compare the dCNN's results to human readouts. Results Using a small labeled dataset with 1020 images with equal distribution among classes, the dCNN reached an accuracy of 90.9% (93%) for the training (validation) data. Applying a larger dataset with additional 1017 labeled images resulted in an accuracy of 96.1% (91.4%) for the training (validation) dataset. For the human readout, there were no significant differences between the pathologists and the dCNN in Ki-67 classification results. Conclusion The dCNN is capable of a standardized classification of Ki-67 staining in vulva carcinoma; therefore, it may be suitable for quality control and standardization in the assessment of tumor grading.
Collapse
Affiliation(s)
| | - Mariam Alyahiaoui
- Institute for Clinical Pathology, University Hospital Zurich, Switzerland.,Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland
| | - Alexander Ciritsis
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland
| | - Cristina Rossi
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland
| | - André Gut
- Institute for Clinical Pathology, University Hospital Zurich, Switzerland
| | - Patryk Hejduk
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland
| | - Andreas Boss
- Institute for Diagnostic and Interventional Radiology, University Hospital Zurich, Switzerland
| |
Collapse
|
60
|
Abstract
It is very important to make an objective evaluation of colorectal cancer histological images. Current approaches are generally based on the use of different combinations of textual features and classifiers to assess the classification performance, or transfer learning to classify different organizational types. However, since histological images contain multiple tissue types and characteristics, classification is still challenging. In this study, we proposed the best classification methodology based on the selected optimizer and modified the parameters of CNN methods. Then, we used deep learning technology to distinguish between healthy and diseased large intestine tissues. Firstly, we trained a neural network and compared the network architecture optimizers. Secondly, we modified the parameters of the network layer to optimize the superior architecture. Finally, we compared our well-trained deep learning methods on two different histological image open datasets, which comprised 5000 H&E images of colorectal cancer. The other dataset was composed of nine organizational categories of 100,000 images with an external validation of 7180 images. The results showed that the accuracy of the recognition of histopathological images was significantly better than that of existing methods. Therefore, this method is expected to have great potential to assist physicians to make clinical diagnoses and reduce the number of disparate assessments based on the use of artificial intelligence to classify colorectal cancer tissue.
Collapse
|
61
|
Yu H, Yang LT, Zhang Q, Armstrong D, Deen MJ. Convolutional neural networks for medical image analysis: State-of-the-art, comparisons, improvement and perspectives. Neurocomputing 2021. [DOI: 10.1016/j.neucom.2020.04.157] [Citation(s) in RCA: 46] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
62
|
Mitotic nuclei analysis in breast cancer histopathology images using deep ensemble classifier. Med Image Anal 2021; 72:102121. [PMID: 34139665 DOI: 10.1016/j.media.2021.102121] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 05/20/2021] [Accepted: 05/24/2021] [Indexed: 02/06/2023]
Abstract
Mitotic nuclei estimation in breast tumour samples has a prognostic significance in analysing tumour aggressiveness and grading system. The automated assessment of mitotic nuclei is challenging because of their high similarity with non-mitotic nuclei and heteromorphic appearance. In this work, we have proposed a new Deep Convolutional Neural Network (CNN) based Heterogeneous Ensemble technique "DHE-Mit-Classifier" for analysis of mitotic nuclei in breast histopathology images. The proposed technique in the first step detects candidate mitotic patches from the histopathological biopsy regions, whereas, in the second step, these patches are classified into mitotic and non-mitotic nuclei using the proposed DHE-Mit-Classifier. For the development of a heterogeneous ensemble, five different deep CNNs are designed and used as base-classifiers. These deep CNNs have varying architectural designs to capture the structural, textural, and morphological properties of the mitotic nuclei. The developed base-classifiers exploit different ideas, including (i) region homogeneity and feature invariance, (ii) asymmetric split-transform-merge, (iii) dilated convolution based multi-scale transformation, (iv) spatial and channel attention, and (v) residual learning. Multi-layer-perceptron is used as a meta-classifier to develop a robust and accurate classifier for providing the final decision. The performance of the proposed ensemble "DHE-Mit-Classifier" is evaluated against state-of-the-art CNNs. The performance evaluation on the test set suggests the superiority of the proposed ensemble with an F-score (0.77), recall (0.71), precision (0.83), and area under the precision-recall curve (0.80). The good generalisation of the proposed ensemble with a considerably high F-score and precision suggests its potential use in the development of an assistance tool for pathologists.
Collapse
|
63
|
Machine learning-based image analysis for accelerating the diagnosis of complicated preneoplastic and neoplastic ductal lesions in breast biopsy tissues. Breast Cancer Res Treat 2021; 188:649-659. [PMID: 33934277 DOI: 10.1007/s10549-021-06243-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 04/21/2021] [Indexed: 10/21/2022]
Abstract
PURPOSE Diagnosis of breast preneoplastic and neoplastic lesions is difficult due to their similar morphology in breast biopsy specimens. To diagnose these lesions, pathologists perform immunohistochemical analysis and consult with expert breast pathologists. These additional examinations are time-consuming and expensive. Artificial intelligence (AI)-based image analysis has recently improved, and may help in ordinal pathological diagnosis. Here, we showed the significance of machine learning-based image analysis of breast preneoplastic and neoplastic lesions for facilitating high-throughput diagnosis. METHODS Images were obtained from normal mammary glands, hyperplastic lesions, preneoplastic lesions and neoplastic lesions, such as usual ductal hyperplasia (UDH), columnar cell lesion (CCL), ductal carcinoma in situ (DCIS), and DCIS with comedo necrosis (comedo DCIS) in breast biopsy specimens. The original enhanced convoluted neural network (CNN) system was used for analyzing the pathological images. RESULTS The AI-based image analysis provided the following area under the curve values (AUC): normal lesion versus DCIS, 0.9902; DCIS versus comedo DCIS, 0.9942; normal lesion versus CCL, 0.9786; and UDH versus DCIS, 1.000. Multiple comparison analysis showed precision and recall scores similar to those of single comparison analysis. Based on the gradient-weighted class activation mapping (Grad-CAM) used to visualize the important regions reflecting the result of CNN analysis, the ratio of stromal tissue in the whole weighted area was significantly higher in UDH and CCL than that in DCIS. CONCLUSIONS These analyses may provide a more accurate and rapid pathological diagnosis of patients. Moreover, Grad-CAM identifies uncharted important histological characteristics for newer pathological findings and targets of research for understanding diseases.
Collapse
|
64
|
Improving DCIS diagnosis and predictive outcome by applying artificial intelligence. Biochim Biophys Acta Rev Cancer 2021; 1876:188555. [PMID: 33933557 DOI: 10.1016/j.bbcan.2021.188555] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 04/22/2021] [Accepted: 04/24/2021] [Indexed: 11/21/2022]
Abstract
Breast ductal carcinoma in situ (DCIS) is a preinvasive lesion that is considered to be a precursor to invasive breast cancer. Nevertheless, not all DCIS will progress to invasion. Current histopathological classification systems are unable to predict which cases will or will not progress, and therefore many women with DCIS may be overtreated. Artificial intelligence (AI) image-based analysis methods have potential to identify and analyze novel features that may facilitate tumor identification, prediction of disease outcome and response to treatment. Indeed, these methods prove promising for accurately identifying DCIS lesions, and show potential clinical utility in the therapeutic stratification of DCIS patients. Here, we review how AI techniques in histopathology may aid diagnosis and clinical decisions in regards to DCIS, and how such techniques could be incorporated into clinical practice.
Collapse
|
65
|
Bajaj S, Donnelly D, Call M, Johannet P, Moran U, Polsky D, Shapiro R, Berman R, Pavlick A, Weber J, Zhong J, Osman I. Melanoma Prognosis: Accuracy of the American Joint Committee on Cancer Staging Manual Eighth Edition. J Natl Cancer Inst 2021; 112:921-928. [PMID: 31977051 DOI: 10.1093/jnci/djaa008] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Revised: 11/11/2019] [Accepted: 11/20/2019] [Indexed: 12/11/2022] Open
Abstract
BACKGROUND The American Joint Committee on Cancer (AJCC) maintains that the eighth edition of its Staging Manual (AJCC8) has improved accuracy compared with the seventh (AJCC7). However, there are concerns that implementation may disrupt analysis of active clinical trials for stage III patients. We used an independent cohort of melanoma patients to test the extent to which AJCC8 has improved prognostic accuracy compared with AJCC7. METHODS We analyzed a cohort of 1315 prospectively enrolled patients. We assessed primary tumor and nodal classification of stage I-III patients using AJCC7 and AJCC8 to assign disease stages at diagnosis. We compared recurrence-free (RFS) and overall survival (OS) using Kaplan-Meier curves and log-rank tests. We then compared concordance indices of discriminatory prognostic ability and area under the curve of 5-year survival to predict RFS and OS. All statistical tests were two-sided. RESULTS Stage IIC patients continued to have worse outcomes than stage IIIA patients, with a 5-year RFS of 26.5% (95% confidence interval [CI] = 12.8% to 55.1%) vs 56.0% (95% CI = 37.0% to 84.7%) by AJCC8 (P = .002). For stage I, removing mitotic index as a T classification factor decreased its prognostic value, although not statistically significantly (RFS concordance index [C-index] = 0.63, 95% CI = 0.56 to 0.69; to 0.56, 95% CI = 0.49 to 0.63, P = .07; OS C-index = 0.48, 95% CI = 0.38 to 0.58; to 0.48, 95% CI = 0.41 to 0.56, P = .90). For stage II, prognostication remained constant (RFS C-index = 0.65, 95% CI = 0.57 to 0.72; OS C-index = 0.61, 95% CI = 0.50 to 0.72), and for stage III, AJCC8 yielded statistically significantly enhanced prognostication for RFS (C-index = 0.65, 95% CI = 0.60 to 0.70; to 0.70, 95% CI = 0.66 to 0.75, P = .01). CONCLUSIONS Compared with AJCC7, we demonstrate that AJCC8 enables more accurate prognosis for patients with stage III melanoma. Restaging a large cohort of patients can enhance the analysis of active clinical trials.
Collapse
Affiliation(s)
- Shirin Bajaj
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Douglas Donnelly
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Melissa Call
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Paul Johannet
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - Una Moran
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| | - David Polsky
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA.,Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Richard Shapiro
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Russell Berman
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Anna Pavlick
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Jeffrey Weber
- Perlmutter Cancer Center, NYU Langone Health, New York, NY, USA
| | - Judy Zhong
- Department of Population Health, NYU Langone School of Medicine, New York, NY, USA
| | - Iman Osman
- Ronald O. Perelman Department of Dermatology, NYU Langone School of Medicine, New York, NY, USA
| |
Collapse
|
66
|
Wilbur DC, Smith ML, Cornell LD, Andryushkin A, Pettus JR. Automated identification of glomeruli and synchronised review of special stains in renal biopsies by machine learning and slide registration: a cross-institutional study. Histopathology 2021; 79:499-508. [PMID: 33813779 DOI: 10.1111/his.14376] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/11/2021] [Accepted: 03/29/2021] [Indexed: 11/30/2022]
Abstract
AIMS Machine learning in digital pathology can improve efficiency and accuracy via prescreening with automated feature identification. Studies using uniform histological material have shown promise. Generalised application requires validation on slides from multiple institutions. We used machine learning to identify glomeruli on renal biopsies and compared performance between single and multiple institutions. METHODS AND RESULTS Randomly selected, adequately sampled renal core biopsy cases (71) consisting of four stains each (haematoxylin and eosin, trichrome, silver, periodic acid Schiff) from three institutions were digitised at ×40. Glomeruli were manually annotated by three renal pathologists using a digital tool. Cases were divided into training/validation (n = 52) and evaluation (n = 19) cohorts. An algorithm was trained to develop three convolutional neural network (CNN) models which tested case cohorts intra- and inter-institutionally. Raw CNN search data from each of the four slides per case were merged into composite regions of interest containing putative glomeruli. The sensitivity and modified specificity of glomerulus detection (versus annotated truth) were calculated for each model/cohort. Intra-institutional (3) sensitivity ranged from 90 to 93%, with modified specificity from 86 to 98%. Interinstitutional (1) sensitivity was 77%, with modified specificity 97%. Combined intra- and inter-institutional (1) sensitivity was 86%, with modified specificity 92%. CONCLUSIONS Feature detection sensitivity degrades when training and test material originate from different sites. Training using a combined set of digital slides from three institutions improves performance. Differing histology methods probably account for algorithm performance contrasts. Our data highlight the need for diverse training sets for the development of generalisable machine learning histology algorithms.
Collapse
Affiliation(s)
| | - Maxwell L Smith
- Department of Laboratory Medicine and Pathology, Mayo Clinic - Scottsdale, Phoenix, AZ, USA
| | - Lynn D Cornell
- Department of Laboratory Medicine and Pathology, Mayo Clinic - Rochester, Rochester, MN, USA
| | | | - Jason R Pettus
- Department of Pathology and Laboratory Medicine, Dartmouth-Hitchcock Medical Center, Lebanon, MH, USA
| |
Collapse
|
67
|
Bhinder B, Gilvary C, Madhukar NS, Elemento O. Artificial Intelligence in Cancer Research and Precision Medicine. Cancer Discov 2021; 11:900-915. [PMID: 33811123 DOI: 10.1158/2159-8290.cd-21-0090] [Citation(s) in RCA: 192] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 02/06/2021] [Accepted: 02/08/2021] [Indexed: 11/16/2022]
Abstract
Artificial intelligence (AI) is rapidly reshaping cancer research and personalized clinical care. Availability of high-dimensionality datasets coupled with advances in high-performance computing, as well as innovative deep learning architectures, has led to an explosion of AI use in various aspects of oncology research. These applications range from detection and classification of cancer, to molecular characterization of tumors and their microenvironment, to drug discovery and repurposing, to predicting treatment outcomes for patients. As these advances start penetrating the clinic, we foresee a shifting paradigm in cancer care becoming strongly driven by AI. SIGNIFICANCE: AI has the potential to dramatically affect nearly all aspects of oncology-from enhancing diagnosis to personalizing treatment and discovering novel anticancer drugs. Here, we review the recent enormous progress in the application of AI to oncology, highlight limitations and pitfalls, and chart a path for adoption of AI in the cancer clinic.
Collapse
Affiliation(s)
- Bhavneet Bhinder
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York.,Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York
| | | | | | - Olivier Elemento
- Caryl and Israel Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, New York. .,Department of Physiology and Biophysics, Weill Cornell Medicine, New York, New York.,OneThree Biotech, New York, New York
| |
Collapse
|
68
|
Wetstein SC, Stathonikos N, Pluim JPW, Heng YJ, Ter Hoeve ND, Vreuls CPH, van Diest PJ, Veta M. Deep learning-based grading of ductal carcinoma in situ in breast histopathology images. J Transl Med 2021; 101:525-533. [PMID: 33608619 PMCID: PMC7985025 DOI: 10.1038/s41374-021-00540-6] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2020] [Revised: 01/05/2021] [Accepted: 01/07/2021] [Indexed: 11/08/2022] Open
Abstract
Ductal carcinoma in situ (DCIS) is a non-invasive breast cancer that can progress into invasive ductal carcinoma (IDC). Studies suggest DCIS is often overtreated since a considerable part of DCIS lesions may never progress into IDC. Lower grade lesions have a lower progression speed and risk, possibly allowing treatment de-escalation. However, studies show significant inter-observer variation in DCIS grading. Automated image analysis may provide an objective solution to address high subjectivity of DCIS grading by pathologists. In this study, we developed and evaluated a deep learning-based DCIS grading system. The system was developed using the consensus DCIS grade of three expert observers on a dataset of 1186 DCIS lesions from 59 patients. The inter-observer agreement, measured by quadratic weighted Cohen's kappa, was used to evaluate the system and compare its performance to that of expert observers. We present an analysis of the lesion-level and patient-level inter-observer agreement on an independent test set of 1001 lesions from 50 patients. The deep learning system (dl) achieved on average slightly higher inter-observer agreement to the three observers (o1, o2 and o3) (κo1,dl = 0.81, κo2,dl = 0.53 and κo3,dl = 0.40) than the observers amongst each other (κo1,o2 = 0.58, κo1,o3 = 0.50 and κo2,o3 = 0.42) at the lesion-level. At the patient-level, the deep learning system achieved similar agreement to the observers (κo1,dl = 0.77, κo2,dl = 0.75 and κo3,dl = 0.70) as the observers amongst each other (κo1,o2 = 0.77, κo1,o3 = 0.75 and κo2,o3 = 0.72). The deep learning system better reflected the grading spectrum of DCIS than two of the observers. In conclusion, we developed a deep learning-based DCIS grading system that achieved a performance similar to expert observers. To the best of our knowledge, this is the first automated system for the grading of DCIS that could assist pathologists by providing robust and reproducible second opinions on DCIS grade.
Collapse
Affiliation(s)
- Suzanne C Wetstein
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Nikolas Stathonikos
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Josien P W Pluim
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Yujing J Heng
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Natalie D Ter Hoeve
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Celien P H Vreuls
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, University Utrecht, Utrecht, The Netherlands
| | - Mitko Veta
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
69
|
Two Ensemble-CNN Approaches for Colorectal Cancer Tissue Type Classification. J Imaging 2021; 7:jimaging7030051. [PMID: 34460707 PMCID: PMC8321410 DOI: 10.3390/jimaging7030051] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2021] [Revised: 02/16/2021] [Accepted: 02/26/2021] [Indexed: 02/06/2023] Open
Abstract
In recent years, automatic tissue phenotyping has attracted increasing interest in the Digital Pathology (DP) field. For Colorectal Cancer (CRC), tissue phenotyping can diagnose the cancer and differentiate between different cancer grades. The development of Whole Slide Images (WSIs) has provided the required data for creating automatic tissue phenotyping systems. In this paper, we study different hand-crafted feature-based and deep learning methods using two popular multi-classes CRC-tissue-type databases: Kather-CRC-2016 and CRC-TP. For the hand-crafted features, we use two texture descriptors (LPQ and BSIF) and their combination. In addition, two classifiers are used (SVM and NN) to classify the texture features into distinct CRC tissue types. For the deep learning methods, we evaluate four Convolutional Neural Network (CNN) architectures (ResNet-101, ResNeXt-50, Inception-v3, and DenseNet-161). Moreover, we propose two Ensemble CNN approaches: Mean-Ensemble-CNN and NN-Ensemble-CNN. The experimental results show that the proposed approaches outperformed the hand-crafted feature-based methods, CNN architectures and the state-of-the-art methods in both databases.
Collapse
|
70
|
Lépine C, Klein P, Voron T, Mandavit M, Berrebi D, Outh-Gauer S, Péré H, Tournier L, Pagès F, Tartour E, Le Meur T, Berlemont S, Teissier N, Carlevan M, Leboulanger N, Galmiche L, Badoual C. Histological Severity Risk Factors Identification in Juvenile-Onset Recurrent Respiratory Papillomatosis: How Immunohistochemistry and AI Algorithms Can Help? Front Oncol 2021; 11:596499. [PMID: 33763347 PMCID: PMC7982831 DOI: 10.3389/fonc.2021.596499] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Accepted: 02/12/2021] [Indexed: 11/23/2022] Open
Abstract
Juvenile-onset recurrent respiratory papillomatosis (JoRRP) is a condition characterized by the repeated growth of benign exophytic papilloma in the respiratory tract. The course of the disease remains unpredictable: some children experience minor symptoms, while others require multiple interventions due to florid growth. Our study aimed to identify histologic severity risk factors in patients with JoRRP. Forty-eight children from two French pediatric centers were included retrospectively. Criteria for a severe disease were: annual rate of surgical endoscopy ≥ 5, spread to the lung, carcinomatous transformation or death. We conducted a multi-stage study with image analysis. First, with Hematoxylin and eosin (HE) digital slides of papilloma, we searched for morphological patterns associated with a severe JoRRP using a deep-learning algorithm. Then, immunohistochemistry with antibody against p53 and p63 was performed on sections of FFPE samples of laryngeal papilloma obtained between 2008 and 2018. Immunostainings were quantified according to the staining intensity through two automated workflows: one using machine learning, the other using deep learning. Twenty-four patients had severe disease. For the HE analysis, no significative results were obtained with cross-validation. For immunostaining with anti-p63 antibody, we found similar results between the two image analysis methods. Using machine learning, we found 23.98% of stained nuclei for medium intensity for mild JoRRP vs. 36.1% for severe JoRRP (p = 0.041); and for medium and strong intensity together, 24.14% for mild JoRRP vs. 36.9% for severe JoRRP (p = 0.048). Using deep learning, we found 58.32% for mild JoRRP vs. 67.45% for severe JoRRP (p = 0.045) for medium and strong intensity together. Regarding p53, we did not find any significant difference in the number of nuclei stained between the two groups of patients. In conclusion, we highlighted that immunochemistry with the anti-p63 antibody is a potential biomarker to predict the severity of the JoRRP.
Collapse
Affiliation(s)
- Charles Lépine
- INSERM-U970, PARCC, Université de Paris, Paris, France.,Department of Pathology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | | | | | | | | | - Sophie Outh-Gauer
- INSERM-U970, PARCC, Université de Paris, Paris, France.,Department of Pathology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Hélène Péré
- INSERM-U970, PARCC, Université de Paris, Paris, France.,Department of Virology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Louis Tournier
- Department of Pathology, Hôpital Robert Debré, APHP, Paris, France
| | - Franck Pagès
- Department of Immunology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | - Eric Tartour
- INSERM-U970, PARCC, Université de Paris, Paris, France.,Department of Immunology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| | | | | | - Natacha Teissier
- Department of Pediatric ENT Surgery, Hôpital Robert Debré, APHP, Paris, France
| | - Mathilde Carlevan
- Department of Pediatric ENT Surgery, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Nicolas Leboulanger
- Department of Pediatric ENT Surgery, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Louise Galmiche
- Department of Pathology, Hôpital Necker-Enfants Malades, APHP, Paris, France
| | - Cécile Badoual
- INSERM-U970, PARCC, Université de Paris, Paris, France.,Department of Pathology, Hôpital Européen Georges-Pompidou, APHP, Paris, France
| |
Collapse
|
71
|
Ho DJ, Yarlagadda DVK, D'Alfonso TM, Hanna MG, Grabenstetter A, Ntiamoah P, Brogi E, Tan LK, Fuchs TJ. Deep Multi-Magnification Networks for multi-class breast cancer image segmentation. Comput Med Imaging Graph 2021; 88:101866. [PMID: 33485058 PMCID: PMC7975990 DOI: 10.1016/j.compmedimag.2021.101866] [Citation(s) in RCA: 40] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2020] [Revised: 12/18/2020] [Accepted: 12/23/2020] [Indexed: 01/17/2023]
Abstract
Pathologic analysis of surgical excision specimens for breast carcinoma is important to evaluate the completeness of surgical excision and has implications for future treatment. This analysis is performed manually by pathologists reviewing histologic slides prepared from formalin-fixed tissue. In this paper, we present Deep Multi-Magnification Network trained by partial annotation for automated multi-class tissue segmentation by a set of patches from multiple magnifications in digitized whole slide images. Our proposed architecture with multi-encoder, multi-decoder, and multi-concatenation outperforms other single and multi-magnification-based architectures by achieving the highest mean intersection-over-union, and can be used to facilitate pathologists' assessments of breast cancer.
Collapse
Affiliation(s)
- David Joon Ho
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA.
| | - Dig V K Yarlagadda
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Timothy M D'Alfonso
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Matthew G Hanna
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Anne Grabenstetter
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Peter Ntiamoah
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Edi Brogi
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Lee K Tan
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA
| | - Thomas J Fuchs
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065 USA; Weill Cornell Graduate School for Medical Sciences, New York, NY 10065 USA
| |
Collapse
|
72
|
Malherbe K. Tumor Microenvironment and the Role of Artificial Intelligence in Breast Cancer Detection and Prognosis. THE AMERICAN JOURNAL OF PATHOLOGY 2021; 191:1364-1373. [PMID: 33639101 DOI: 10.1016/j.ajpath.2021.01.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2020] [Revised: 01/02/2021] [Accepted: 01/28/2021] [Indexed: 12/21/2022]
Abstract
A critical knowledge gap has been noted in breast cancer detection, prognosis, and evaluation between tumor microenvironment and associated neoplasm. Artificial intelligence (AI) has multiple subsets or methods for data extraction and evaluation, including artificial neural networking, which allows computational foundations, similar to neurons, to make connections and new neural pathways during data set training. Deep machine learning and AI hold great potential to accurately assess tumor microenvironment models employing vast data management techniques. Despite the significant potential AI holds, there is still much debate surrounding the appropriate and ethical curation of medical data from picture archiving and communication systems. AI output's clinical significance depends on its human predecessor's data training sets. Integration between biomarkers, risk factors, and imaging data will allow the best predictor models for patient-based outcomes.
Collapse
Affiliation(s)
- Kathryn Malherbe
- Department Radiography, Faculty Health Sciences, University of Pretoria, Pretoria, South Africa.
| |
Collapse
|
73
|
Dong Y, Wan J, Si L, Meng Y, Dong Y, Liu S, He H, Ma H. Deriving Polarimetry Feature Parameters to Characterize Microstructural Features in Histological Sections of Breast Tissues. IEEE Trans Biomed Eng 2021; 68:881-892. [PMID: 32845834 DOI: 10.1109/tbme.2020.3019755] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
OBJECTIVE Mueller matrix polarimetry technique has been regarded as a powerful tool for probing the microstructural information of tissues. The multiplying of cells and remodeling of collagen fibers in breast carcinoma tissues have been reported to be related to patient survival and prognosis, and they give rise to observable patterns in hematoxylin and eosin (H&E) sections of typical breast tissues (TBTs) that the pathologist can label as three distinctive pathological features (DPFs)-cell nuclei, aligned collagen, and disorganized collagen. The aim of this paper is to propose a pixel-based extraction approach of polarimetry feature parameters (PFPs) using a linear discriminant analysis (LDA) classifier. These parameters provide quantitative characterization of the three DPFs in four types of TBTs. METHODS The LDA-based training method learns to find the most simplified linear combination from polarimetry basis parameters (PBPs) constrained under the accuracy remains constant to characterize the specific microstructural feature quantitatively in TBTs. RESULTS We present results from a cohort of 32 clinical patients with analysis of 224 regions-of-interest. The characterization accuracy for PFPs ranges from 0.82 to 0.91. CONCLUSION This work demonstrates the ability of PFPs to quantitatively characterize the DPFs in the H&E pathological sections of TBTs. SIGNIFICANCE This technique paves the way for automatic and quantitative evaluation of specific microstructural features in histopathological digitalization and computer-aided diagnosis.
Collapse
|
74
|
Park J, Chung YR, Kong ST, Kim YW, Park H, Kim K, Kim DI, Jung KH. Aggregation of cohorts for histopathological diagnosis with deep morphological analysis. Sci Rep 2021; 11:2876. [PMID: 33536550 PMCID: PMC7858624 DOI: 10.1038/s41598-021-82642-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Accepted: 01/21/2021] [Indexed: 12/13/2022] Open
Abstract
There have been substantial efforts in using deep learning (DL) to diagnose cancer from digital images of pathology slides. Existing algorithms typically operate by training deep neural networks either specialized in specific cohorts or an aggregate of all cohorts when there are only a few images available for the target cohort. A trade-off between decreasing the number of models and their cancer detection performance was evident in our experiments with The Cancer Genomic Atlas dataset, with the former approach achieving higher performance at the cost of having to acquire large datasets from the cohort of interest. Constructing annotated datasets for individual cohorts is extremely time-consuming, with the acquisition cost of such datasets growing linearly with the number of cohorts. Another issue associated with developing cohort-specific models is the difficulty of maintenance: all cohort-specific models may need to be adjusted when a new DL algorithm is to be used, where training even a single model may require a non-negligible amount of computation, or when more data is added to some cohorts. In resolving the sub-optimal behavior of a universal cancer detection model trained on an aggregate of cohorts, we investigated how cohorts can be grouped to augment a dataset without increasing the number of models linearly with the number of cohorts. This study introduces several metrics which measure the morphological similarities between cohort pairs and demonstrates how the metrics can be used to control the trade-off between performance and the number of models.
Collapse
Affiliation(s)
| | - Yul Ri Chung
- Pathology Center, Seegene Medical Foundation, Seoul, Korea
| | | | | | | | | | - Dong-Il Kim
- Department of Pathology, Green Cross Laboratories, Yong-in, Gyeonggi, Korea
| | | |
Collapse
|
75
|
Harrison JH, Gilbertson JR, Hanna MG, Olson NH, Seheult JN, Sorace JM, Stram MN. Introduction to Artificial Intelligence and Machine Learning for Pathology. Arch Pathol Lab Med 2021; 145:1228-1254. [PMID: 33493264 DOI: 10.5858/arpa.2020-0541-cp] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/10/2020] [Indexed: 11/06/2022]
Abstract
CONTEXT.— Recent developments in machine learning have stimulated intense interest in software that may augment or replace human experts. Machine learning may impact pathology practice by offering new capabilities in analysis, interpretation, and outcomes prediction using images and other data. The principles of operation and management of machine learning systems are unfamiliar to pathologists, who anticipate a need for additional education to be effective as expert users and managers of the new tools. OBJECTIVE.— To provide a background on machine learning for practicing pathologists, including an overview of algorithms, model development, and performance evaluation; to examine the current status of machine learning in pathology and consider possible roles and requirements for pathologists in local deployment and management of machine learning systems; and to highlight existing challenges and gaps in deployment methodology and regulation. DATA SOURCES.— Sources include the biomedical and engineering literature, white papers from professional organizations, government reports, electronic resources, and authors' experience in machine learning. References were chosen when possible for accessibility to practicing pathologists without specialized training in mathematics, statistics, or software development. CONCLUSIONS.— Machine learning offers an array of techniques that in recent published results show substantial promise. Data suggest that human experts working with machine learning tools outperform humans or machines separately, but the optimal form for this combination in pathology has not been established. Significant questions related to the generalizability of machine learning systems, local site verification, and performance monitoring remain to be resolved before a consensus on best practices and a regulatory environment can be established.
Collapse
Affiliation(s)
- James H Harrison
- From the Department of Pathology, University of Virginia School of Medicine, Charlottesville (Harrison)
| | - John R Gilbertson
- the Departments of Biomedical Informatics and Pathology, University of Pittsburgh, Pittsburgh, Pennsylvania (Gilbertson)
| | - Matthew G Hanna
- the Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York (Hanna)
| | - Niels H Olson
- the Defense Innovation Unit, Mountain View, California (Olson).,the Department of Pathology, Uniformed Services University, Bethesda, Maryland (Olson)
| | - Jansen N Seheult
- the Department of Pathology, University of Pittsburgh, and Vitalant Specialty Labs, Pittsburgh, Pennsylvania (Seheult)
| | - James M Sorace
- the US Department of Health and Human Services, retired, Lutherville, Maryland (Sorace)
| | - Michelle N Stram
- the Department of Forensic Medicine, New York University, and Office of Chief Medical Examiner, New York, New York (Stram)
| |
Collapse
|
76
|
Srinidhi CL, Ciga O, Martel AL. Deep neural network models for computational histopathology: A survey. Med Image Anal 2021; 67:101813. [PMID: 33049577 PMCID: PMC7725956 DOI: 10.1016/j.media.2020.101813] [Citation(s) in RCA: 212] [Impact Index Per Article: 70.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Revised: 05/12/2020] [Accepted: 08/09/2020] [Indexed: 12/14/2022]
Abstract
Histopathological images contain rich phenotypic information that can be used to monitor underlying mechanisms contributing to disease progression and patient survival outcomes. Recently, deep learning has become the mainstream methodological choice for analyzing and interpreting histology images. In this paper, we present a comprehensive review of state-of-the-art deep learning approaches that have been used in the context of histopathological image analysis. From the survey of over 130 papers, we review the field's progress based on the methodological aspect of different machine learning strategies such as supervised, weakly supervised, unsupervised, transfer learning and various other sub-variants of these methods. We also provide an overview of deep learning based survival models that are applicable for disease-specific prognosis tasks. Finally, we summarize several existing open datasets and highlight critical challenges and limitations with current deep learning approaches, along with possible avenues for future research.
Collapse
Affiliation(s)
- Chetan L Srinidhi
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Canada.
| | - Ozan Ciga
- Department of Medical Biophysics, University of Toronto, Canada
| | - Anne L Martel
- Physical Sciences, Sunnybrook Research Institute, Toronto, Canada; Department of Medical Biophysics, University of Toronto, Canada
| |
Collapse
|
77
|
Naik N, Madani A, Esteva A, Keskar NS, Press MF, Ruderman D, Agus DB, Socher R. Deep learning-enabled breast cancer hormonal receptor status determination from base-level H&E stains. Nat Commun 2020; 11:5727. [PMID: 33199723 PMCID: PMC7670411 DOI: 10.1038/s41467-020-19334-3] [Citation(s) in RCA: 115] [Impact Index Per Article: 28.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2020] [Accepted: 09/25/2020] [Indexed: 12/09/2022] Open
Abstract
For newly diagnosed breast cancer, estrogen receptor status (ERS) is a key molecular marker used for prognosis and treatment decisions. During clinical management, ERS is determined by pathologists from immunohistochemistry (IHC) staining of biopsied tissue for the targeted receptor, which highlights the presence of cellular surface antigens. This is an expensive, time-consuming process which introduces discordance in results due to variability in IHC preparation and pathologist subjectivity. In contrast, hematoxylin and eosin (H&E) staining—which highlights cellular morphology—is quick, less expensive, and less variable in preparation. Here we show that machine learning can determine molecular marker status, as assessed by hormone receptors, directly from cellular morphology. We develop a multiple instance learning-based deep neural network that determines ERS from H&E-stained whole slide images (WSI). Our algorithm—trained strictly with WSI-level annotations—is accurate on a varied, multi-country dataset of 3,474 patients, achieving an area under the curve (AUC) of 0.92 for sensitivity and specificity. Our approach has the potential to augment clinicians’ capabilities in cancer prognosis and theragnosis by harnessing biological signals imperceptible to the human eye. Determination of estrogen receptor status (ERS) in breast cancer tissue requires immunohistochemistry, which is sensitive to the vagaries of sample processing and the subjectivity of pathologists. Here the authors present a deep learning model that determines ERS from H&E stained tissue, which could improve oncology decisions in under-resourced settings.
Collapse
Affiliation(s)
- Nikhil Naik
- Salesforce Research, 575 High St, Palo Alto, CA, 94301, USA.
| | - Ali Madani
- Salesforce Research, 575 High St, Palo Alto, CA, 94301, USA
| | - Andre Esteva
- Salesforce Research, 575 High St, Palo Alto, CA, 94301, USA
| | | | - Michael F Press
- Department of Pathology, Keck School of Medicine, University of Southern California, 2011 Zonal Ave, Los Angeles, CA, 90033, USA
| | - Daniel Ruderman
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, 12414 Exposition Blvd, Los Angeles, CA, 90064, USA
| | - David B Agus
- Lawrence J. Ellison Institute for Transformative Medicine, University of Southern California, 12414 Exposition Blvd, Los Angeles, CA, 90064, USA
| | - Richard Socher
- Salesforce Research, 575 High St, Palo Alto, CA, 94301, USA
| |
Collapse
|
78
|
Kikkisetti S, Zhu J, Shen B, Li H, Duong TQ. Deep-learning convolutional neural networks with transfer learning accurately classify COVID-19 lung infection on portable chest radiographs. PeerJ 2020; 8:e10309. [PMID: 33194447 PMCID: PMC7649013 DOI: 10.7717/peerj.10309] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022] Open
Abstract
Portable chest X-ray (pCXR) has become an indispensable tool in the management of Coronavirus Disease 2019 (COVID-19) lung infection. This study employed deep-learning convolutional neural networks to classify COVID-19 lung infections on pCXR from normal and related lung infections to potentially enable more timely and accurate diagnosis. This retrospect study employed deep-learning convolutional neural network (CNN) with transfer learning to classify based on pCXRs COVID-19 pneumonia (N = 455) on pCXR from normal (N = 532), bacterial pneumonia (N = 492), and non-COVID viral pneumonia (N = 552). The data was randomly split into 75% training and 25% testing, randomly. A five-fold cross-validation was used for the testing set separately. Performance was evaluated using receiver-operating curve analysis. Comparison was made with CNN operated on the whole pCXR and segmented lungs. CNN accurately classified COVID-19 pCXR from those of normal, bacterial pneumonia, and non-COVID-19 viral pneumonia patients in a multiclass model. The overall sensitivity, specificity, accuracy, and AUC were 0.79, 0.93, and 0.79, 0.85 respectively (whole pCXR), and were 0.91, 0.93, 0.88, and 0.89 (CXR of segmented lung). The performance was generally better using segmented lungs. Heatmaps showed that CNN accurately localized areas of hazy appearance, ground glass opacity and/or consolidation on the pCXR. Deep-learning convolutional neural network with transfer learning accurately classifies COVID-19 on portable chest X-ray against normal, bacterial pneumonia or non-COVID viral pneumonia. This approach has the potential to help radiologists and frontline physicians by providing more timely and accurate diagnosis.
Collapse
Affiliation(s)
- Shreeja Kikkisetti
- Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Jocelyn Zhu
- Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| | - Beiyi Shen
- Radiology, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Haifang Li
- Radiology, State University of New York at Stony Brook, Stony Brook, NY, USA
| | - Tim Q Duong
- Radiology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
79
|
Moxley-Wyles B, Colling R, Verrill C. Artificial intelligence in pathology: an overview. ACTA ACUST UNITED AC 2020. [DOI: 10.1016/j.mpdhp.2020.08.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
80
|
Bera K, Katz I, Madabhushi A. Reimagining T Staging Through Artificial Intelligence and Machine Learning Image Processing Approaches in Digital Pathology. JCO Clin Cancer Inform 2020; 4:1039-1050. [PMID: 33166198 PMCID: PMC7713520 DOI: 10.1200/cci.20.00110] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/21/2020] [Indexed: 02/06/2023] Open
Abstract
Tumor stage and grade, visually assessed by pathologists from evaluation of pathology images in conjunction with radiographic imaging techniques, have been linked to outcome, progression, and survival for a number of cancers. The gold standard of staging in oncology has been the TNM (tumor-node-metastasis) staging system. Though histopathological grading has shown prognostic significance, it is subjective and limited by interobserver variability even among experienced surgical pathologists. Recently, artificial intelligence (AI) approaches have been applied to pathology images toward diagnostic-, prognostic-, and treatment prediction-related tasks in cancer. AI approaches have the potential to overcome the limitations of conventional TNM staging and tumor grading approaches, providing a direct prognostic prediction of disease outcome independent of tumor stage and grade. Broadly speaking, these AI approaches involve extracting patterns from images that are then compared against previously defined disease signatures. These patterns are typically categorized as either (1) handcrafted, which involve domain-inspired attributes, such as nuclear shape, or (2) deep learning (DL)-based representations, which tend to be more abstract. DL approaches have particularly gained considerable popularity because of the minimal domain knowledge needed for training, mostly only requiring annotated examples corresponding to the categories of interest. In this article, we discuss AI approaches for digital pathology, especially as they relate to disease prognosis, prediction of genomic and molecular alterations in the tumor, and prediction of treatment response in oncology. We also discuss some of the potential challenges with validation, interpretability, and reimbursement that must be addressed before widespread clinical deployment. The article concludes with a brief discussion of potential future opportunities in the field of AI for digital pathology and oncology.
Collapse
Affiliation(s)
- Kaustav Bera
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH
- Maimonides Medical Center, Department of Internal Medicine, Brooklyn, NY
| | - Ian Katz
- Southern Sun Pathology, Sydney, Australia, and University of Queensland, Brisbane, Australia
| | - Anant Madabhushi
- Case Western Reserve University, Department of Biomedical Engineering, Cleveland, OH
- Louis Stokes Veterans Affairs Medical Center, Cleveland, OH
| |
Collapse
|
81
|
Liu W, Juhas M, Zhang Y. Fine-Grained Breast Cancer Classification With Bilinear Convolutional Neural Networks (BCNNs). Front Genet 2020; 11:547327. [PMID: 33101377 PMCID: PMC7500315 DOI: 10.3389/fgene.2020.547327] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/17/2020] [Indexed: 12/24/2022] Open
Abstract
Classification of histopathological images of cancer is challenging even for well-trained professionals, due to the fine-grained variability of the disease. Deep Convolutional Neural Networks (CNNs) showed great potential for classification of a number of the highly variable fine-grained objects. In this study, we introduce a Bilinear Convolutional Neural Networks (BCNNs) based deep learning method for fine-grained classification of breast cancer histopathological images. We evaluated our model by comparison with several deep learning algorithms for fine-grained classification. We used bilinear pooling to aggregate a large number of orderless features without taking into consideration the disease location. The experimental results on BreaKHis, a publicly available breast cancer dataset, showed that our method is highly accurate with 99.24% and 95.95% accuracy in binary and in fine-grained classification, respectively.
Collapse
Affiliation(s)
- Weihuang Liu
- College of Science, Harbin Institute of Technology, Shenzhen, China
| | - Mario Juhas
- Faculty of Science and Medicine, University of Fribourg, Fribourg, Switzerland
| | - Yang Zhang
- College of Science, Harbin Institute of Technology, Shenzhen, China
| |
Collapse
|
82
|
Russo DP, Yan X, Shende S, Huang H, Yan B, Zhu H. Virtual Molecular Projections and Convolutional Neural Networks for the End-to-End Modeling of Nanoparticle Activities and Properties. Anal Chem 2020; 92:13971-13979. [PMID: 32970421 DOI: 10.1021/acs.analchem.0c02878] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Digitalizing complex nanostructures into data structures suitable for machine learning modeling without losing nanostructure information has been a major challenge. Deep learning frameworks, particularly convolutional neural networks (CNNs), are especially adept at handling multidimensional and complex inputs. In this study, CNNs were applied for the modeling of nanoparticle activities exclusively from nanostructures. The nanostructures were represented by virtual molecular projections, a multidimensional digitalization of nanostructures, and used as input data to train CNNs. To this end, 77 nanoparticles with various activities and/or physicochemical property results were used for modeling. The resulting CNN model predictions show high correlations with the experimental results. An analysis of a trained CNN quantitatively showed that neurons were able to recognize distinct nanostructure features critical to activities and physicochemical properties. This "end-to-end" deep learning approach is well suited to digitalize complex nanostructures for data-driven machine learning modeling and can be broadly applied to rationally design nanoparticles with desired activities.
Collapse
Affiliation(s)
- Daniel P Russo
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States
| | - Xiliang Yan
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Institute of Environmental Research at Greater Bay, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China
| | - Sunil Shende
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Department of Computer Science, Rutgers University, 227 Penn Street, Camden, New Jersey 08102, United States
| | | | - Bing Yan
- Institute of Environmental Research at Greater Bay, Key Laboratory for Water Quality and Conservation of the Pearl River Delta, Ministry of Education, Guangzhou University, Guangzhou 510006, China.,School of Environmental Science and Engineering, Shandong University, Jinan 250100, China
| | - Hao Zhu
- Center for Computational and Integrative Biology, Rutgers University, 201 S Broadway, Camden, New Jersey 08103, United States.,Department of Chemistry, Rutgers University, 315 Penn Street, Camden, New Jersey 08102, United States
| |
Collapse
|
83
|
Kendall TJ, Duff CM, Thomson AM, Iredale JP. Integration of geoscience frameworks into digital pathology analysis permits quantification of microarchitectural relationships in histological landscapes. Sci Rep 2020; 10:17572. [PMID: 33067578 PMCID: PMC7567886 DOI: 10.1038/s41598-020-74691-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2020] [Accepted: 10/05/2020] [Indexed: 12/27/2022] Open
Abstract
Although gold-standard histological assessment is subjective it remains central to diagnosis and clinical trial protocols and is crucial for the evaluation of any preclinical disease model. Objectivity and reproducibility are enhanced by quantitative analysis of histological images but current methods require application-specific algorithm training and fail to extract understanding from the histological context of observable features. We reinterpret histopathological images as disease landscapes to describe a generalisable framework defining topographic relationships in tissue using geoscience approaches. The framework requires no user-dependent training to operate on all image datasets in a classifier-agnostic manner but is adaptable and scalable, able to quantify occult abnormalities, derive mechanistic insights, and define a new feature class for machine-learning diagnostic classification. We demonstrate application to inflammatory, fibrotic and neoplastic disease in multiple organs, including the detection and quantification of occult lobular enlargement in the liver secondary to hilar obstruction. We anticipate this approach will provide a robust class of histological data for trial stratification or endpoints, provide quantitative endorsement of experimental models of disease, and could be incorporated within advanced approaches to clinical diagnostic pathology.
Collapse
Affiliation(s)
- Timothy J Kendall
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK.
- Edinburgh Pathology, The Royal Infirmary of Edinburgh, The University of Edinburgh, 51 Little France Crescent, Edinburgh, EH16 4SA, UK.
| | - Catherine M Duff
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
- Centre for Cardiovascular Sciences, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
| | - Andrew M Thomson
- NHS Lothian University Hospitals Division, Pathology Department, The Royal Infirmary of Edinburgh, 51 Little France Crescent, Edinburgh, EH16 4SA, UK
| | - John P Iredale
- University of Edinburgh Centre for Inflammation Research, Queen's Medical Research Institute, The University of Edinburgh, 47 Little France Crescent, Edinburgh, EH16 4TJ, UK
- Senate House, University of Bristol, Tyndall Avenue, Bristol, BS8 1TH, UK
| |
Collapse
|
84
|
Van Bockstal MR, Berlière M, Duhoux FP, Galant C. Interobserver Variability in Ductal Carcinoma In Situ of the Breast. Am J Clin Pathol 2020; 154:596-609. [PMID: 32566938 DOI: 10.1093/ajcp/aqaa077] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
OBJECTIVES Since most patients with ductal carcinoma in situ (DCIS) of the breast are treated upon diagnosis, evidence on its natural progression to invasive carcinoma is limited. It is estimated that around half of the screen-detected DCIS lesions would have remained indolent if they had never been detected. Many patients with DCIS are therefore probably overtreated. Four ongoing randomized noninferiority trials explore active surveillance as a treatment option. Eligibility for these trials is mainly based on histopathologic features. Hence, the call for reproducible histopathologic assessment has never sounded louder. METHODS Here, the available classification systems for DCIS are discussed in depth. RESULTS This comprehensive review illustrates that histopathologic evaluation of DCIS is characterized by significant interobserver variability. Future digitalization of pathology, combined with development of deep learning algorithms or so-called artificial intelligence, may be an innovative solution to tackle this problem. However, implementation of digital pathology is not within reach for each laboratory worldwide. An alternative classification system could reduce the disagreement among histopathologists who use "conventional" light microscopy: the introduction of dichotomous histopathologic assessment is likely to increase interobserver concordance. CONCLUSIONS Reproducible histopathologic assessment is a prerequisite for robust risk stratification and adequate clinical decision-making. Two-tier histopathologic assessment might enhance the quality of care.
Collapse
Affiliation(s)
- Mieke R Van Bockstal
- Department of Pathology, Brussels, Belgium
- Breast Clinic, Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
| | - Martine Berlière
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
| | - Francois P Duhoux
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
- Laboratory of Experimental Cancer Research, Department of Radiation Oncology and Experimental Cancer Research, Ghent University, Ghent, Belgium
- Institut de Recherche Expérimentale et Clinique, Université Catholique de Louvain, Brussels, Belgium
| | - Christine Galant
- Department of Pathology, Brussels, Belgium
- Department of Medical Oncology, King Albert II Cancer Institute, Cliniques universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
85
|
Javed S, Mahmood A, Werghi N, Benes K, Rajpoot N. Multiplex Cellular Communities in Multi-Gigapixel Colorectal Cancer Histology Images for Tissue Phenotyping. IEEE TRANSACTIONS ON IMAGE PROCESSING : A PUBLICATION OF THE IEEE SIGNAL PROCESSING SOCIETY 2020; PP:9204-9219. [PMID: 32966218 DOI: 10.1109/tip.2020.3023795] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
In computational pathology, automated tissue phenotyping in cancer histology images is a fundamental tool for profiling tumor microenvironments. Current tissue phenotyping methods use features derived from image patches which may not carry biological significance. In this work, we propose a novel multiplex cellular community-based algorithm for tissue phenotyping integrating cell-level features within a graph-based hierarchical framework. We demonstrate that such integration offers better performance compared to prior deep learning and texture-based methods as well as to cellular community based methods using uniplex networks. To this end, we construct celllevel graphs using texture, alpha diversity and multi-resolution deep features. Using these graphs, we compute cellular connectivity features which are then employed for the construction of a patch-level multiplex network. Over this network, we compute multiplex cellular communities using a novel objective function. The proposed objective function computes a low-dimensional subspace from each cellular network and subsequently seeks a common low-dimensional subspace using the Grassmann manifold. We evaluate our proposed algorithm on three publicly available datasets for tissue phenotyping, demonstrating a significant improvement over existing state-of-the-art methods.
Collapse
|
86
|
Pan Y, Sun Z, Wang W, Yang Z, Jia J, Feng X, Wang Y, Fang Q, Li J, Dai H, Ku C, Wang S, Liu C, Xue L, Lyu N, Zou S. Automatic detection of squamous cell carcinoma metastasis in esophageal lymph nodes using semantic segmentation. Clin Transl Med 2020; 10:e129. [PMID: 32722861 PMCID: PMC7418811 DOI: 10.1002/ctm2.129] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 06/30/2020] [Accepted: 07/01/2020] [Indexed: 12/16/2022] Open
Abstract
Esophageal squamous cell carcinoma (ESCC) is more prevalent than esophageal adenocarcinoma in Asia, especially in China, where more than half of ESCC cases occur worldwide. Many studies have reported that the automatic detection of lymph node metastasis using semantic segmentation shows good performance in breast cancer and other adenocarcinomas. However, the detection of squamous cell carcinoma metastasis in hematoxylin-eosin (H&E)-stained slides has never been reported. We collected a training set of 110 esophageal lymph node slides with metastasis and 132 lymph node slides without metastasis. An iPad-based annotation system was used to draw the contours of the cancer metastasis region. A DeepLab v3 model was trained to achieve the best fit with the training data. The learned model could estimate the probability of metastasis. To evaluate the effectiveness of the detection model of learned metastasis, we used another large cohort of clinical H&E-stained esophageal lymph node slides containing 795 esophageal lymph nodes from 154 esophageal cancer patients. The basic authenticity label for each slide was confirmed by experienced pathologists. After filtering isolated noise in the prediction, we obtained an accuracy of 94%. Furthermore, we applied the learned model to throat and lung lymph node squamous cell carcinoma metastases and achieved the following promising results: an accuracy of 96.7% in throat cancer and an accuracy of 90% in lung cancer. In this work, we organized an annotated dataset of H&E-stained esophageal lymph node and trained a deep neural network to detect lymph node metastasis in H&E-stained slides of squamous cell carcinoma automatically. Moreover, it is possible to use this model to detect lymph nodes metastasis in squamous cell carcinoma from other organs. This study directly demonstrates the potential for determining the localization of squamous cell carcinoma metastases in lymph node and assisting in pathological diagnosis.
Collapse
Affiliation(s)
- Yi Pan
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | - Wenmiao Wang
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhaoyang Yang
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jia Jia
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaolong Feng
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yaxi Wang
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qing Fang
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangtao Li
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongtian Dai
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | - Liyan Xue
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Ning Lyu
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shuangmei Zou
- Department of Pathology, National Cancer Center/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
87
|
Zhu J, Shen B, Abbasi A, Hoshmand-Kochi M, Li H, Duong TQ. Deep transfer learning artificial intelligence accurately stages COVID-19 lung disease severity on portable chest radiographs. PLoS One 2020; 15:e0236621. [PMID: 32722697 PMCID: PMC7386587 DOI: 10.1371/journal.pone.0236621] [Citation(s) in RCA: 76] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Accepted: 07/10/2020] [Indexed: 12/12/2022] Open
Abstract
This study employed deep-learning convolutional neural networks to stage lung disease severity of Coronavirus Disease 2019 (COVID-19) infection on portable chest x-ray (CXR) with radiologist score of disease severity as ground truth. This study consisted of 131 portable CXR from 84 COVID-19 patients (51M 55.1±14.9yo; 29F 60.1±14.3yo; 4 missing information). Three expert chest radiologists scored the left and right lung separately based on the degree of opacity (0-3) and geographic extent (0-4). Deep-learning convolutional neural network (CNN) was used to predict lung disease severity scores. Data were split into 80% training and 20% testing datasets. Correlation analysis between AI-predicted versus radiologist scores were analyzed. Comparison was made with traditional and transfer learning. The average opacity score was 2.52 (range: 0-6) with a standard deviation of 0.25 (9.9%) across three readers. The average geographic extent score was 3.42 (range: 0-8) with a standard deviation of 0.57 (16.7%) across three readers. The inter-rater agreement yielded a Fleiss' Kappa of 0.45 for opacity score and 0.71 for extent score. AI-predicted scores strongly correlated with radiologist scores, with the top model yielding a correlation coefficient (R2) of 0.90 (range: 0.73-0.90 for traditional learning and 0.83-0.90 for transfer learning) and a mean absolute error of 8.5% (ranges: 17.2-21.0% and 8.5%-15.5, respectively). Transfer learning generally performed better. In conclusion, deep-learning CNN accurately stages disease severity on portable chest x-ray of COVID-19 lung infection. This approach may prove useful to stage lung disease severity, prognosticate, and predict treatment response and survival, thereby informing risk management and resource allocation.
Collapse
Affiliation(s)
- Jocelyn Zhu
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Beiyi Shen
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Almas Abbasi
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Mahsa Hoshmand-Kochi
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Haifang Li
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| | - Tim Q. Duong
- Department of Radiology, Renaissance School of Medicine, Stony Brook University, Stony Brook, New York, United States of America
| |
Collapse
|
88
|
Abstract
Pathologists are adopting whole slide images (WSIs) for diagnosis, thanks to recent FDA approval of WSI systems as class II medical devices. In response to new market forces and recent technology advances outside of pathology, a new field of computational pathology has emerged that applies artificial intelligence (AI) and machine learning algorithms to WSIs. Computational pathology has great potential for augmenting pathologists' accuracy and efficiency, but there are important concerns regarding trust of AI due to the opaque, black-box nature of most AI algorithms. In addition, there is a lack of consensus on how pathologists should incorporate computational pathology systems into their workflow. To address these concerns, building computational pathology systems with explainable AI (xAI) mechanisms is a powerful and transparent alternative to black-box AI models. xAI can reveal underlying causes for its decisions; this is intended to promote safety and reliability of AI for critical tasks such as pathology diagnosis. This article outlines xAI enabled applications in anatomic pathology workflow that improves efficiency and accuracy of the practice. In addition, we describe HistoMapr-Breast, an initial xAI enabled software application for breast core biopsies. HistoMapr-Breast automatically previews breast core WSIs and recognizes the regions of interest to rapidly present the key diagnostic areas in an interactive and explainable manner. We anticipate xAI will ultimately serve pathologists as an interactive computational guide for computer-assisted primary diagnosis.
Collapse
|
89
|
Acs B, Rantalainen M, Hartman J. Artificial intelligence as the next step towards precision pathology. J Intern Med 2020; 288:62-81. [PMID: 32128929 DOI: 10.1111/joim.13030] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 12/16/2019] [Accepted: 12/30/2019] [Indexed: 12/13/2022]
Abstract
Pathology is the cornerstone of cancer care. The need for accuracy in histopathologic diagnosis of cancer is increasing as personalized cancer therapy requires accurate biomarker assessment. The appearance of digital image analysis holds promise to improve both the volume and precision of histomorphological evaluation. Recently, machine learning, and particularly deep learning, has enabled rapid advances in computational pathology. The integration of machine learning into routine care will be a milestone for the healthcare sector in the next decade, and histopathology is right at the centre of this revolution. Examples of potential high-value machine learning applications include both model-based assessment of routine diagnostic features in pathology, and the ability to extract and identify novel features that provide insights into a disease. Recent groundbreaking results have demonstrated that applications of machine learning methods in pathology significantly improves metastases detection in lymph nodes, Ki67 scoring in breast cancer, Gleason grading in prostate cancer and tumour-infiltrating lymphocyte (TIL) scoring in melanoma. Furthermore, deep learning models have also been demonstrated to be able to predict status of some molecular markers in lung, prostate, gastric and colorectal cancer based on standard HE slides. Moreover, prognostic (survival outcomes) deep neural network models based on digitized HE slides have been demonstrated in several diseases, including lung cancer, melanoma and glioma. In this review, we aim to present and summarize the latest developments in digital image analysis and in the application of artificial intelligence in diagnostic pathology.
Collapse
Affiliation(s)
- B Acs
- From the, Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| | - M Rantalainen
- Department of Medical Epidemiology and Biostatistics, Karolinska Institutet, Stockholm, Sweden
| | - J Hartman
- From the, Department of Oncology and Pathology, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
90
|
Sultan AS, Elgharib MA, Tavares T, Jessri M, Basile JR. The use of artificial intelligence, machine learning and deep learning in oncologic histopathology. J Oral Pathol Med 2020; 49:849-856. [PMID: 32449232 DOI: 10.1111/jop.13042] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 04/29/2020] [Accepted: 05/09/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Recently, there has been a momentous drive to apply advanced artificial intelligence (AI) technologies to diagnostic medicine. The introduction of AI has provided vast new opportunities to improve health care and has introduced a new wave of heightened precision in oncologic pathology. The impact of AI on oncologic pathology has now become apparent, and its use with respect to oral oncology is still in the nascent stage. DISCUSSION A foundational overview of AI classification systems used in medicine and a review of common terminology used in machine learning and computational pathology will be presented. This paper provides a focused review on the recent advances in AI and deep learning in oncologic histopathology and oral oncology. In addition, specific emphasis on recent studies that have applied these technologies to oral cancer prognostication will also be discussed. CONCLUSION Machine and deep learning methods designed to enhance prognostication of oral cancer have been proposed with much of the work focused on prediction models on patient survival and locoregional recurrences in patients with oral squamous cell carcinomas (OSCC). Few studies have explored machine learning methods on OSCC digital histopathologic images. It is evident that further research at the whole slide image level is needed and future collaborations with computer scientists may progress the field of oral oncology.
Collapse
Affiliation(s)
- Ahmed S Sultan
- School of Dentistry, University of Maryland, Baltimore, MD, USA
| | | | - Tiffany Tavares
- School of Dentistry, University of Missouri-Kansas City, Kansas City, MO, USA
| | - Maryam Jessri
- Oral Health Centre of Western Australia, Perth, WA, Australia
| | - John R Basile
- School of Dentistry, University of Maryland, Baltimore, MD, USA.,University of Maryland Greenebaum Cancer Center, Baltimore, MD, USA
| |
Collapse
|
91
|
Classification and mutation prediction based on histopathology H&E images in liver cancer using deep learning. NPJ Precis Oncol 2020; 4:14. [PMID: 32550270 PMCID: PMC7280520 DOI: 10.1038/s41698-020-0120-3] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 05/07/2020] [Indexed: 12/24/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common subtype of liver cancer, and assessing its histopathological grade requires visual inspection by an experienced pathologist. In this study, the histopathological H&E images from the Genomic Data Commons Databases were used to train a neural network (inception V3) for automatic classification. According to the evaluation of our model by the Matthews correlation coefficient, the performance level was close to the ability of a 5-year experience pathologist, with 96.0% accuracy for benign and malignant classification, and 89.6% accuracy for well, moderate, and poor tumor differentiation. Furthermore, the model was trained to predict the ten most common and prognostic mutated genes in HCC. We found that four of them, including CTNNB1, FMN2, TP53, and ZFX4, could be predicted from histopathology images, with external AUCs from 0.71 to 0.89. The findings demonstrated that convolutional neural networks could be used to assist pathologists in the classification and detection of gene mutation in liver cancer.
Collapse
|
92
|
Inge L, Dennis E. Development and applications of computer image analysis algorithms for scoring of PD-L1 immunohistochemistry. ACTA ACUST UNITED AC 2020; 6:2-8. [PMID: 35757235 PMCID: PMC9216464 DOI: 10.1016/j.iotech.2020.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Immune checkpoint inhibitors targeting programmed cell death 1 (PD-1) and programmed cell death ligand 1 (PD-L1) have rapidly become integral to standard-of-care therapy for non-small cell lung cancer and other cancers. Immunohistochemical (IHC) staining of PD-L1 is currently the accepted and approved diagnostic assay for selecting patients for PD-L1/PD-1 axis therapies in certain indications. However, the inherent biological complexity of PD-L1 and the availability of several PD-L1 assays – each with different detection systems, platforms, scoring algorithms and cut-offs – have created challenges to ensure reliable and reproducible results based on subjective visual assessment by pathologists. The increasing adoption of computer technologies into the daily workflow of pathology provides an opportunity to leverage these tools towards improving the clinical value of PD-L1 IHC assays. This review describes several image analysis software programs of computer-aided PD-L1 scoring in the hope of driving further discussion and technological advancement in digital pathology and artificial intelligence approaches, particularly as precision medicine evolves to encompass accurate simultaneous assessment of multiple features of cancer cells and their interactions with the tumor microenvironment.
Collapse
|
93
|
Girolami I, Marletta S, Pantanowitz L, Torresani E, Ghimenton C, Barbareschi M, Scarpa A, Brunelli M, Barresi V, Trimboli P, Eccher A. Impact of image analysis and artificial intelligence in thyroid pathology, with particular reference to cytological aspects. Cytopathology 2020; 31:432-444. [PMID: 32248583 DOI: 10.1111/cyt.12828] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 03/27/2020] [Accepted: 03/27/2020] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Thyroid pathology has great potential for automated/artificial intelligence algorithm application as the incidence of thyroid nodules is increasing and the indeterminate interpretation rate of fine-needle aspiration remains relatively high. The aim of the study is to review the published literature on automated image analysis and artificial intelligence applications to thyroid pathology with whole-slide imaging. METHODS Systematic search was carried out in electronic databases. Studies dealing with thyroid pathology and use of automated algorithms applied to whole-slide imaging were included. Quality of studies was assessed with a modified QUADAS-2 tool. RESULTS Of 919 retrieved articles, 19 were included. The main themes addressed were the comparison of automated assessment of immunohistochemical staining with manual pathologist's assessment, quantification of differences in cellular and nuclear parameters among tumour entities, and discrimination between benign and malignant nodules. Correlation coefficients with manual assessment were higher than 0.76 and diagnostic performance of automated models was comparable with an expert pathologist diagnosis. Computational difficulties were related to the large size of whole-slide images. CONCLUSIONS Overall, the results are promising and it is likely that, with the resolution of technical issues, the application of automated algorithms in thyroid pathology will increase and be adopted following suitable validation studies.
Collapse
Affiliation(s)
- Ilaria Girolami
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Stefano Marletta
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Liron Pantanowitz
- Department of Pathology, UPMC Shadyside Hospital, University of Pittsburgh, Pittsburgh, PA, USA
| | - Evelin Torresani
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Claudio Ghimenton
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | | | - Aldo Scarpa
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Matteo Brunelli
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Valeria Barresi
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Pierpaolo Trimboli
- Clinic for Nuclear Medicine and Competence Centre for Thyroid Disease, Imaging Institute of Southern Switzerland, Ente Ospedaliero Cantonale, Bellinzona, Switzerland
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| |
Collapse
|
94
|
Chang MC, Mrkonjic M. Review of the current state of digital image analysis in breast pathology. Breast J 2020; 26:1208-1212. [PMID: 32342590 DOI: 10.1111/tbj.13858] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2019] [Accepted: 11/05/2019] [Indexed: 01/10/2023]
Abstract
Advances in digital image analysis have the potential to transform the practice of breast pathology. In the near future, a move to a digital workflow offers improvements in efficiency. Coupled with artificial intelligence (AI), digital pathology can assist pathologist interpretation, automate time-consuming tasks, and discover novel morphologic patterns. Opportunities for digital enhancements abound in breast pathology, from increasing reproducibility in grading and biomarker interpretation, to discovering features that correlate with patient outcome and treatment. Our objective is to review the most recent developments in digital pathology with clear impact to breast pathology practice. Although breast pathologists currently undertake limited adoption of digital methods, the field is rapidly evolving. Care is needed to validate emerging technologies for effective patient care.
Collapse
Affiliation(s)
- Martin C Chang
- University of Vermont Cancer Center, Burlington, VT, USA.,Department of Pathology and Laboratory Medicine, Larner College of Medicine at the University of Vermont, Burlington, VT, USA
| | - Miralem Mrkonjic
- Sinai Health System, Toronto, ON, Canada.,Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
95
|
Wetstein SC, Onken AM, Luffman C, Baker GM, Pyle ME, Kensler KH, Liu Y, Bakker B, Vlutters R, van Leeuwen MB, Collins LC, Schnitt SJ, Pluim JPW, Tamimi RM, Heng YJ, Veta M. Deep learning assessment of breast terminal duct lobular unit involution: Towards automated prediction of breast cancer risk. PLoS One 2020; 15:e0231653. [PMID: 32294107 PMCID: PMC7159218 DOI: 10.1371/journal.pone.0231653] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/27/2020] [Indexed: 02/07/2023] Open
Abstract
Terminal duct lobular unit (TDLU) involution is the regression of milk-producing structures in the breast. Women with less TDLU involution are more likely to develop breast cancer. A major bottleneck in studying TDLU involution in large cohort studies is the need for labor-intensive manual assessment of TDLUs. We developed a computational pathology solution to automatically capture TDLU involution measures. Whole slide images (WSIs) of benign breast biopsies were obtained from the Nurses' Health Study. A set of 92 WSIs was annotated for acini, TDLUs and adipose tissue to train deep convolutional neural network (CNN) models for detection of acini, and segmentation of TDLUs and adipose tissue. These networks were integrated into a single computational method to capture TDLU involution measures including number of TDLUs per tissue area, median TDLU span and median number of acini per TDLU. We validated our method on 40 additional WSIs by comparing with manually acquired measures. Our CNN models detected acini with an F1 score of 0.73±0.07, and segmented TDLUs and adipose tissue with Dice scores of 0.84±0.13 and 0.87±0.04, respectively. The inter-observer ICC scores for manual assessments on 40 WSIs of number of TDLUs per tissue area, median TDLU span, and median acini count per TDLU were 0.71, 0.81 and 0.73, respectively. Intra-observer reliability was evaluated on 10/40 WSIs with ICC scores of >0.8. Inter-observer ICC scores between automated results and the mean of the two observers were: 0.80 for number of TDLUs per tissue area, 0.57 for median TDLU span, and 0.80 for median acini count per TDLU. TDLU involution measures evaluated by manual and automated assessment were inversely associated with age and menopausal status. We developed a computational pathology method to measure TDLU involution. This technology eliminates the labor-intensiveness and subjectivity of manual TDLU assessment, and can be applied to future breast cancer risk studies.
Collapse
Affiliation(s)
- Suzanne C. Wetstein
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Allison M. Onken
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Christina Luffman
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Gabrielle M. Baker
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Michael E. Pyle
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Kevin H. Kensler
- Division of Population Sciences, Dana Farber Cancer Institute, Boston, Massachusetts, United States of America
| | - Ying Liu
- Division of Public Health Sciences, Department of Surgery, Washington University School of Medicine and Alvin J. Siteman Cancer Center, St Louis, Missouri, United States of America
| | - Bart Bakker
- Philips Research Europe, High Tech Campus, Eindhoven, The Netherlands
| | - Ruud Vlutters
- Philips Research Europe, High Tech Campus, Eindhoven, The Netherlands
| | | | - Laura C. Collins
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Stuart J. Schnitt
- Dana-Farber/Brigham and Women’s Cancer Center, Harvard Medical School, Dana-Farber Cancer Institute-Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Josien P. W. Pluim
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Rulla M. Tamimi
- Channing Division of Network Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, Massachusetts, United States of America
| | - Yujing J. Heng
- Department of Pathology, Harvard Medical School, Beth Israel Deaconess Medical Center, Boston, Massachusetts, United States of America
| | - Mitko Veta
- Medical Image Analysis Group, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
| |
Collapse
|
96
|
Jiang Y, Yang M, Wang S, Li X, Sun Y. Emerging role of deep learning-based artificial intelligence in tumor pathology. Cancer Commun (Lond) 2020; 40:154-166. [PMID: 32277744 PMCID: PMC7170661 DOI: 10.1002/cac2.12012] [Citation(s) in RCA: 176] [Impact Index Per Article: 44.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2020] [Accepted: 02/06/2020] [Indexed: 12/11/2022] Open
Abstract
The development of digital pathology and progression of state-of-the-art algorithms for computer vision have led to increasing interest in the use of artificial intelligence (AI), especially deep learning (DL)-based AI, in tumor pathology. The DL-based algorithms have been developed to conduct all kinds of work involved in tumor pathology, including tumor diagnosis, subtyping, grading, staging, and prognostic prediction, as well as the identification of pathological features, biomarkers and genetic changes. The applications of AI in pathology not only contribute to improve diagnostic accuracy and objectivity but also reduce the workload of pathologists and subsequently enable them to spend additional time on high-level decision-making tasks. In addition, AI is useful for pathologists to meet the requirements of precision oncology. However, there are still some challenges relating to the implementation of AI, including the issues of algorithm validation and interpretability, computing systems, the unbelieving attitude of pathologists, clinicians and patients, as well as regulators and reimbursements. Herein, we present an overview on how AI-based approaches could be integrated into the workflow of pathologists and discuss the challenges and perspectives of the implementation of AI in tumor pathology.
Collapse
Affiliation(s)
- Yahui Jiang
- Department of PathologyKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P. R. China
| | - Meng Yang
- Department Epidemiology and BiostatisticsKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P.R. China
| | - Shuhao Wang
- Institute for Interdisciplinary Information SciencesTsinghua UniversityBeijing100084P. R. China
| | - Xiangchun Li
- Department Epidemiology and BiostatisticsKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P.R. China
| | - Yan Sun
- Department of PathologyKey Laboratory of Cancer Prevention and TherapyTianjin's Clinical Research Center for CancerNational Clinical Research Center for CancerTianjin Cancer Institute and HospitalTianjin Medical UniversityTianjin300060P. R. China
| |
Collapse
|
97
|
Smith KP, Kirby JE. Image analysis and artificial intelligence in infectious disease diagnostics. Clin Microbiol Infect 2020; 26:1318-1323. [PMID: 32213317 DOI: 10.1016/j.cmi.2020.03.012] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2019] [Revised: 03/06/2020] [Accepted: 03/13/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Microbiologists are valued for their time-honed skills in image analysis, including identification of pathogens and inflammatory context in Gram stains, ova and parasite preparations, blood smears and histopathologic slides. They also must classify colony growth on a variety of agar plates for triage and assessment. Recent advances in image analysis, in particular application of artificial intelligence (AI), have the potential to automate these processes and support more timely and accurate diagnoses. OBJECTIVES To review current AI-based image analysis as applied to clinical microbiology; and to discuss future trends in the field. SOURCES Material sourced for this review included peer-reviewed literature annotated in the PubMed or Google Scholar databases and preprint articles from bioRxiv. Articles describing use of AI for analysis of images used in infectious disease diagnostics were reviewed. CONTENT We describe application of machine learning towards analysis of different types of microbiologic image data. Specifically, we outline progress in smear and plate interpretation as well as the potential for AI diagnostic applications in the clinical microbiology laboratory. IMPLICATIONS Combined with automation, we predict that AI algorithms will be used in the future to prescreen and preclassify image data, thereby increasing productivity and enabling more accurate diagnoses through collaboration between the AI and the microbiologist. Once developed, image-based AI analysis is inexpensive and amenable to local and remote diagnostic use.
Collapse
Affiliation(s)
- K P Smith
- Department of Pathology, Beth Israel Deaconess Medical Center, USA; Harvard Medical School, Boston, MA, USA
| | - J E Kirby
- Department of Pathology, Beth Israel Deaconess Medical Center, USA; Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
98
|
Chuang WY, Chang SH, Yu WH, Yang CK, Yeh CJ, Ueng SH, Liu YJ, Chen TD, Chen KH, Hsieh YY, Hsia Y, Wang TH, Hsueh C, Kuo CF, Yeh CY. Successful Identification of Nasopharyngeal Carcinoma in Nasopharyngeal Biopsies Using Deep Learning. Cancers (Basel) 2020; 12:cancers12020507. [PMID: 32098314 PMCID: PMC7072217 DOI: 10.3390/cancers12020507] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2020] [Revised: 02/16/2020] [Accepted: 02/19/2020] [Indexed: 12/14/2022] Open
Abstract
Pathologic diagnosis of nasopharyngeal carcinoma (NPC) can be challenging since most cases are nonkeratinizing carcinoma with little differentiation and many admixed lymphocytes. Our aim was to evaluate the possibility to identify NPC in nasopharyngeal biopsies using deep learning. A total of 726 nasopharyngeal biopsies were included. Among them, 100 cases were randomly selected as the testing set, 20 cases as the validation set, and all other 606 cases as the training set. All three datasets had equal numbers of NPC cases and benign cases. Manual annotation was performed. Cropped square image patches of 256 × 256 pixels were used for patch-level training, validation, and testing. The final patch-level algorithm effectively identified NPC patches, with an area under the receiver operator characteristic curve (AUC) of 0.9900. Using gradient-weighted class activation mapping, we demonstrated that the identification of NPC patches was based on morphologic features of tumor cells. At the second stage, whole-slide images were sequentially cropped into patches, inferred with the patch-level algorithm, and reconstructed into images with a smaller size for training, validation, and testing. Finally, the AUC was 0.9848 for slide-level identification of NPC. Our result shows for the first time that deep learning algorithms can identify NPC.
Collapse
Affiliation(s)
- Wen-Yu Chuang
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan
| | - Shang-Hung Chang
- Center for Big Data Analytics and Statistics, Chang Gung Memorial Hospital, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan;
| | - Wei-Hsiang Yu
- aetherAI, Co., Ltd., No. 3-2, Yuan-Qu Street, Nangang District, Taipei City 115, Taiwan; (W.-H.Y.); (C.-K.Y.)
| | - Cheng-Kun Yang
- aetherAI, Co., Ltd., No. 3-2, Yuan-Qu Street, Nangang District, Taipei City 115, Taiwan; (W.-H.Y.); (C.-K.Y.)
| | - Chi-Ju Yeh
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
| | - Shir-Hwa Ueng
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
- Chang Gung Molecular Medicine Research Center, Chang Gung University, No. 259, Wenhua First Road, Guishan District, Taoyuan City 333, Taiwan
| | - Yu-Jen Liu
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
| | - Tai-Di Chen
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
| | - Kuang-Hua Chen
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
| | - Yi-Yin Hsieh
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
| | - Yi Hsia
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
- Department of Pathology, MacKay Memorial Hospital, No. 92, Section 2, Zhongshan North Road, Zhongshan District, Taipei City 104, Taiwan
| | - Tong-Hong Wang
- Tissue Bank, Chang Gung Memorial Hospital, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan;
| | - Chuen Hsueh
- Department of Pathology, Chang Gung Memorial Hospital and Chang Gung University, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan; (W.-Y.C.); (C.-J.Y.); (S.-H.U.); (Y.-J.L.); (T.-D.C.); (K.-H.C.); (Y.-Y.H.); (Y.H.); (C.H.)
- Chang Gung Molecular Medicine Research Center, Chang Gung University, No. 259, Wenhua First Road, Guishan District, Taoyuan City 333, Taiwan
| | - Chang-Fu Kuo
- Center for Artificial Intelligence in Medicine, Chang Gung Memorial Hospital, No. 5, Fuxing Street, Guishan District, Taoyuan City 333, Taiwan;
| | - Chao-Yuan Yeh
- aetherAI, Co., Ltd., No. 3-2, Yuan-Qu Street, Nangang District, Taipei City 115, Taiwan; (W.-H.Y.); (C.-K.Y.)
- Correspondence: ; Tel.: +886-2-27856892
| |
Collapse
|
99
|
Ianni JD, Soans RE, Sankarapandian S, Chamarthi RV, Ayyagari D, Olsen TG, Bonham MJ, Stavish CC, Motaparthi K, Cockerell CJ, Feeser TA, Lee JB. Tailored for Real-World: A Whole Slide Image Classification System Validated on Uncurated Multi-Site Data Emulating the Prospective Pathology Workload. Sci Rep 2020; 10:3217. [PMID: 32081956 PMCID: PMC7035316 DOI: 10.1038/s41598-020-59985-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 02/06/2020] [Indexed: 11/10/2022] Open
Abstract
Standard of care diagnostic procedure for suspected skin cancer is microscopic examination of hematoxylin & eosin stained tissue by a pathologist. Areas of high inter-pathologist discordance and rising biopsy rates necessitate higher efficiency and diagnostic reproducibility. We present and validate a deep learning system which classifies digitized dermatopathology slides into 4 categories. The system is developed using 5,070 images from a single lab, and tested on an uncurated set of 13,537 images from 3 test labs, using whole slide scanners manufactured by 3 different vendors. The system's use of deep-learning-based confidence scoring as a criterion to consider the result as accurate yields an accuracy of up to 98%, and makes it adoptable in a real-world setting. Without confidence scoring, the system achieved an accuracy of 78%. We anticipate that our deep learning system will serve as a foundation enabling faster diagnosis of skin cancer, identification of cases for specialist review, and targeted diagnostic classifications.
Collapse
Affiliation(s)
| | | | | | | | | | - Thomas G Olsen
- Department of Dermatology, Boonshoft School of Medicine, Wright State University School of Medicine, Dayton, Ohio, USA
- Dermatopathology Laboratory of Central States, Dayton, Ohio, USA
| | | | | | - Kiran Motaparthi
- Department of Dermatology, University of Florida College of Medicine, Gainesville, Florida, USA
| | | | | | - Jason B Lee
- Departments of Dermatology and Cutaneous Biology, Sidney Kimmel Medical College at Thomas Jefferson University, Philadelphia, Pennsylvania, USA
| |
Collapse
|
100
|
Bándi P, Balkenhol M, van Ginneken B, van der Laak J, Litjens G. Resolution-agnostic tissue segmentation in whole-slide histopathology images with convolutional neural networks. PeerJ 2019; 7:e8242. [PMID: 31871843 PMCID: PMC6924324 DOI: 10.7717/peerj.8242] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Modern pathology diagnostics is being driven toward large scale digitization of microscopic tissue sections. A prerequisite for its safe implementation is the guarantee that all tissue present on a glass slide can also be found back in the digital image. Whole-slide scanners perform a tissue segmentation in a low resolution overview image to prevent inefficient high-resolution scanning of empty background areas. However, currently applied algorithms can fail in detecting all tissue regions. In this study, we developed convolutional neural networks to distinguish tissue from background. We collected 100 whole-slide images of 10 tissue samples—staining categories from five medical centers for development and testing. Additionally, eight more images of eight unfamiliar categories were collected for testing only. We compared our fully-convolutional neural networks to three traditional methods on a range of resolution levels using Dice score and sensitivity. We also tested whether a single neural network can perform equivalently to multiple networks, each specialized in a single resolution. Overall, our solutions outperformed the traditional methods on all the tested resolutions. The resolution-agnostic network achieved average Dice scores between 0.97 and 0.98 across the tested resolution levels, only 0.0069 less than the resolution-specific networks. Finally, its excellent generalization performance was demonstrated by achieving averages of 0.98 Dice score and 0.97 sensitivity on the eight unfamiliar images. A future study should test this network prospectively.
Collapse
Affiliation(s)
- Péter Bándi
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maschenka Balkenhol
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Bram van Ginneken
- Department of Radiology and Nuclear Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Geert Litjens
- Department of Pathology, Radboud University Medical Center, Nijmegen, The Netherlands
| |
Collapse
|