1
|
Ersöz CC, Berber H, Heper A. Grade 4 astrocytoma vs. grade 4 glioblastoma: is there any clue in H&E? Int J Neurosci 2024:1-6. [PMID: 39686561 DOI: 10.1080/00207454.2024.2441994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/08/2024] [Accepted: 12/09/2024] [Indexed: 12/18/2024]
Abstract
Objective: Gliomas are the most common primary tumors of the central nervous system. The fifth edition of the World Health Organization (WHO) Classification of Tumors of the CNS identifies IDH mutant astrocytomas grade 4 and IDH wild type glioblastomas grade 4 as distinct entities. This study aimed to identify morphological indicators that could predict IDH mutation status in grade 4 diffuse astrocytomas and grade 4 glioblastomas among fifty patients from two groups. Methods: Hematoxylin and eosin (H&E)-stained tumor slides were scanned using a digital scanner and further histopathological examinations were performed on digital images, with additional calculations and measurements. Results: The study showed that, IDH-wildtype glioblastomas and IDH-mutant grade 4 astrocytomas exhibit unique morphological features, particularly in relation to levels of necrosis, microvessel density, and the presence of "C" or "Ring" shape giant cells. Conclusion: Despite advancements in genomic biomarker technology, histology remains an essential tool for predicting patient outcomes. Therefore, pathologists must continue to investigate and document the morphological implications of molecular changes in CNS tumors.
Collapse
Affiliation(s)
| | - Havva Berber
- Department of Pathology, Ankara University Medical School, Ankara, Turkey
| | - Aylin Heper
- Department of Pathology, Ankara University Medical School, Ankara, Turkey
| |
Collapse
|
2
|
Gallo E, Guardiani D, Betti M, Arteni BAM, Di Martino S, Baldinelli S, Daralioti T, Merenda E, Ascione A, Visca P, Pescarmona E, Lavitrano M, Nisticò P, Ciliberto G, Pallocca M. AI drives the assessment of lung cancer microenvironment composition. J Pathol Inform 2024; 15:100400. [PMID: 39469280 PMCID: PMC11513621 DOI: 10.1016/j.jpi.2024.100400] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2024] [Revised: 07/24/2024] [Accepted: 09/26/2024] [Indexed: 10/30/2024] Open
Abstract
Purpose The abundance and distribution of tumor-infiltrating lymphocytes (TILs) as well as that of other components of the tumor microenvironment is of particular importance for predicting response to immunotherapy in lung cancer (LC). We describe here a pilot study employing artificial intelligence (AI) in the assessment of TILs and other cell populations, intending to reduce the inter- or intra-observer variability that commonly characterizes this evaluation. Design We developed a machine learning-based classifier to detect tumor, immune, and stromal cells on hematoxylin and eosin-stained sections, using the open-source framework QuPath. We evaluated the quantity of the aforementioned three cell populations among 37 LC whole slide images regions of interest, comparing the assessments made by five pathologists, both before and after using graphical predictions made by AI, for a total of 1110 quantitative measurements. Results Our findings indicate noteworthy variations in score distribution among pathologists and between individual pathologists and AI. The AI-guided pathologist's evaluations resulted in reduction of significant discrepancies across pathologists: three comparisons showed a loss of significance (p > 0.05), whereas other four showed a reduction in significance (p > 0.01). Conclusions We show that employing a machine learning approach in cell population quantification reduces inter- and intra-observer variability, improving reproducibility and facilitating its use in further validation studies.
Collapse
Affiliation(s)
- Enzo Gallo
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Davide Guardiani
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Martina Betti
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Rome, Italy
- Department of Computer, Control and Management Engineering, La Sapienza University of Rome, Rome, Italy
| | - Brindusa Ana Maria Arteni
- UOC Anatomy Pathology, Biobank IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici, Ospitalieri IFO, Rome, Italy
| | - Simona Di Martino
- UOC Anatomy Pathology, Biobank IRCCS Regina Elena National Cancer Institute, Istituti Fisioterapici, Ospitalieri IFO, Rome, Italy
| | - Sara Baldinelli
- Biostatistics, Bioinformatics and Clinical Trial Center, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Theodora Daralioti
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Elisabetta Merenda
- Department of Radiological, Oncological and Pathological Sciences, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Andrea Ascione
- Department of Experimental Medicine, Sapienza University of Rome, Policlinico Umberto I, Rome, Italy
| | - Paolo Visca
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Edoardo Pescarmona
- Department of Pathology, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Marialuisa Lavitrano
- School of Medicine and Surgery, University of Milano-Bicocca, 20900 Monza, Italy
| | - Paola Nisticò
- Tumor Immunology and Immunotherapy Unit, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Gennaro Ciliberto
- Scientific Direction, IRCCS Regina Elena National Cancer Institute, Rome, Italy
| | - Matteo Pallocca
- Institute of Experimental Endocrinology and Oncology, National Research Council, Naples, Italy
| |
Collapse
|
3
|
Nguyen VT, Huynh PTD, Nguyen PTB, Tran DN, Nguyen VT, Ngo DQ, Le CV. Application of digital slide scanning in external quality assessment program on intestinal parasites. Ann Saudi Med 2024; 44:369-376. [PMID: 39651924 PMCID: PMC11627036 DOI: 10.5144/0256-4947.2024.369] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 09/28/2024] [Indexed: 12/12/2024] Open
Abstract
BACKGROUND Intestinal parasitic infections remain prevalent in Viet Nam. Therefore, ensuring quality assurance in intestinal parasite testing is crucial for screening laboratories. The challenges associated with liquid or glass slide samples necessitate the exploration of digital slide applications, which can offer numerous benefits to program suppliers and participants. OBJECTIVES Compare the true and concordance rates of digital and glass slides for diagnosis. DESIGN Experimental research design. MATERIALS AND METHODS In total, 30 medical professionals from 30 hospitals participated in the trial. The sets of slides encompassed a range of densities, including negative and coinfected slides. Seven types of glass slides were selected for scanning and digital slide production. MAIN OUTCOME MEASURES The primary outcomes were true and concordance variables. Secondary outcomes included time sample sending and time completion. The digital slides were uploaded to a secure website for participant access while glass slides were sent individually by mail. Data collection involved participants analyzing specimens and reporting their results using a scoring method based on parasite detection and identification accuracy. SAMPLE SIZE 210 glass and digital slide-reading results each. RESULTS The mean true rate between original and glass slides diagnosis was 97.6% (range 90.0%-100%), and it slightly increased to 98.1% (range 90.0%-100%) when using digital slides. The average concordance diagnosis rate between glass and digital slides was 99.5%. Importantly, there were no differences in the diagnostic results between glass and digital slides. The findings revealed that the use of digital slides reduced the total time required by approximately 1.1 days compared with that of glass slides. CONCLUSION Altogether, the application of digital slides in the external quality assessment program for intestinal parasites offers convenience for users through online platforms and saves operational time process. LIMITATIONS The small sample size in this experimental study limited the statistical significance of the comparisons.
Collapse
Affiliation(s)
- Vien Tien Nguyen
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Phuc Thi Diem Huynh
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Phuong Thi Be Nguyen
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Dang Ngoc Tran
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Vien Truong Nguyen
- From the Faculty of Public Health, Pham Ngoc Thach University of Medicine, Ho Chi Minh, Vietnam
| | - Dat Quoc Ngo
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| | - Chuon Van Le
- From the University of Medicine and Pharmacy at Ho Chi Minh City, Ho Chi Minh City, Vietnam
| |
Collapse
|
4
|
Li F, Ma J, Wen T, Tian Z, Liang HN. HI-Net: A novel histopathologic image segmentation model for metastatic breast cancer via lightweight dataset construction. Heliyon 2024; 10:e38410. [PMID: 39421372 PMCID: PMC11483284 DOI: 10.1016/j.heliyon.2024.e38410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Revised: 09/22/2024] [Accepted: 09/24/2024] [Indexed: 10/19/2024] Open
Abstract
Since 2020, breast cancer has remained the most prevalent cancer worldwide and the World Health Organisation projects significant increases by 2040, with new cases expected to exceed 3 million annually (a 40% increase) and deaths to surpass 1 million (a 50% increase), highlighting the urgent need for advancements in detection and treatment. Current detection of metastasis is highly dependent on labour-intensive and error-prone pathological examination of large-scale biotissue. Given the high-resolution (100,000 × 100,000 gigapixels) but limited quantity of open-source pathological slide datasets, existing deep learning models face preprocessing challenges. This paper introduces HI-Net, a high-speed panoramic feature-extraction pyramid network for rapid and accurate detection of metastatic breast cancer, balancing panoramic segmentation and local attention. Additionally, a lightweight pathological slide dataset optimised for 512 x 512-pixel resolution, derived from downsampled and reassembled competitive datasets, accelerates training and reduces computational costs. HI-Net demonstrates superior performance on existing medical imaging competition datasets and our lightweight dataset, evidencing its effectiveness across datasets and potential for contributing to the generalisation of intelligent diagnostics.
Collapse
Affiliation(s)
- Fengze Li
- University of Liverpool, Liverpool, UK
- Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Jieming Ma
- Xi'an Jiaotong-Liverpool University, Suzhou, China
| | - Tianxi Wen
- Xi'an Jiaotong-Liverpool University, Suzhou, China
| | | | | |
Collapse
|
5
|
Jain E, Patel A, Parwani AV, Shafi S, Brar Z, Sharma S, Mohanty SK. Whole Slide Imaging Technology and Its Applications: Current and Emerging Perspectives. Int J Surg Pathol 2024; 32:433-448. [PMID: 37437093 DOI: 10.1177/10668969231185089] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/14/2023]
Abstract
Background. Whole slide imaging (WSI) represents a paradigm shift in pathology, serving as a necessary first step for a wide array of digital tools to enter the field. It utilizes virtual microscopy wherein glass slides are converted into digital slides and are viewed by pathologists by automated image analysis. Its impact on pathology workflow, reproducibility, dissemination of educational material, expansion of service to underprivileged areas, and institutional collaboration exemplifies a significant innovative movement. The recent US Food and Drug Administration approval to WSI for its use in primary surgical pathology diagnosis has opened opportunities for wider application of this technology in routine practice. Main Text. The ongoing technological advances in digital scanners, image visualization methods, and the integration of artificial intelligence-derived algorithms with these systems provide avenues to exploit its applications. Its benefits are innumerable such as ease of access through the internet, avoidance of physical storage space, and no risk of deterioration of staining quality or breakage of slides to name a few. Although the benefits of WSI to pathology practices are many, the complexities of implementation remain an obstacle to widespread adoption. Some barriers including the high cost, technical glitches, and most importantly professional hesitation to adopt a new technology have hindered its use in routine pathology. Conclusions. In this review, we summarize the technical aspects of WSI, its applications in diagnostic pathology, training, and research along with future perspectives. It also highlights improved understanding of the current challenges to implementation, as well as the benefits and successes of the technology. WSI provides a golden opportunity for pathologists to guide its evolution, standardization, and implementation to better acquaint them with the key aspects of this technology and its judicial use. Also, implementation of routine digital pathology is an extra step requiring resources which (currently) does not usually result increased efficiency or payment.
Collapse
Affiliation(s)
- Ekta Jain
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Ankush Patel
- Department of Pathology, Wexner Medical Center, Columbus, OH, USA
| | - Anil V Parwani
- Department of Pathology, Wexner Medical Center, Columbus, OH, USA
| | - Saba Shafi
- Department of Pathology, Wexner Medical Center, Columbus, OH, USA
| | - Zoya Brar
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Shivani Sharma
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| | - Sambit K Mohanty
- Department of Pathology and Laboratory Medicine, CORE Diagnostics, Gurgaon, India
| |
Collapse
|
6
|
Sode M, Thagaard J, Eriksen JO, Laenkholm AV. Digital image analysis and assisted reading of the HER2 score display reduced concordance: pitfalls in the categorisation of HER2-low breast cancer. Histopathology 2023; 82:912-924. [PMID: 36737248 DOI: 10.1111/his.14877] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 01/11/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
AIMS Digital image analysis (DIA) is used increasingly as an assisting tool to evaluate biomarkers, including human epidermal growth factor receptor 2 (HER2) in invasive breast cancer (BC). DIA can assist pathologists in HER2 evaluation by presenting quantitative information about the HER2 staining in APP assisted reading (AR). Concurrently, the HER2-low category (HER2-1+/2+ without HER2 gene amplification) has gained prominence due to newly developed antibody-drug conjugates. However, major inter- and intraobserver variability have been observed for the entity. The present quality assurance study investigated the concordance between DIA and AR in clinical use, especially concerning the HER2-low category. METHODS AND RESULTS HER2 immunohistochemistry (IHC) in 761 tumours from 727 patients was evaluated in tissue microarray (TMA) cores by DIA (Visiopharm HER2-CONNECT) and AR. Overall concordance between HER2-scores were 73% (n = 552, weighted-κ: 0.66), and 88% (n = 669, weighted-κ: 0.70), when combining HER2-0/1+. A total of 205 scores were discordant by one category, while four were discordant by two categories. A heterogeneous HER2 pattern was relatively common in the discordant cases and a pitfall in the categorisation of HER2-low BC. AR more commonly reassigned a lower HER2 score (from HER2-1+ to HER2-0) within the HER2-low subgroup (n = 624) compared with DIA. CONCLUSION DIA and AR display moderate agreement with heterogeneous and aberrant staining, representing a source of discordance and a pitfall in the evaluation of HER2.
Collapse
Affiliation(s)
- Michael Sode
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Jens Ole Eriksen
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark
| | - Anne-Vibeke Laenkholm
- Department of Pathology, Zealand University Hospital, Roskilde, Denmark.,Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
7
|
Li B, Guo S, Yin X, Ni C, Gao S, Li G, Ni C, Jiang H, Lau WY, Jin G. Risk factors of positive resection margin differ in pancreaticoduodenectomy and distal pancreatosplenectomy for pancreatic ductal adenocarcinoma undergoing upfront surgery. Asian J Surg 2022; 46:1541-1549. [PMID: 36376184 DOI: 10.1016/j.asjsur.2022.09.156] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Revised: 06/13/2022] [Accepted: 09/26/2022] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVE Positive resection margin indicates worse prognosis. The present study identified the independent risk factors of R1 resection in pancreaticoduodenectomy (PD) and distal pancreatosplenectomy (DP) for patients with pancreatic ductal adenocarcinoma (PDAC). METHOD Consecutive patients who were operated from 1st December 2017 to 30th December 2018 were analyzed retrospectively. A standardized pathological examination with digital whole-mount slide images (DWMSIs) was utilized for evaluation of resection margin status. R1 was defined as microscopic tumor infiltration within 1 mm to the resection margin. The potential risk factors of R1 resection for PD and DP were analyzed separately by univariate and multivariate logistic regression analyses. RESULTS For the 192 patients who underwent PD, and the 87 patients who underwent DP, the R1 resection rates were 31.8% and 35.6%, respectively. Univariate analysis on risk factors of R1 resection for PD were tumor location, lymphovascular invasion, N staging, and TNM staging; while those for DP were T staging and TNM staging. Multivariate logistic regression analysis showed the location of tumor in the neck and uncinate process, and N1/2 staging were independent risk factors of R1 resection for PD; while those for DP were T3 staging. CONCLUSIONS The clarification of the risk factors of R1 resection might clearly make surgeons take reasonable decisions on surgical strategies for different surgical procedures in patients with PDAC, so as to obtain the first attempt of R0 resection.
Collapse
|
8
|
Zarella MD, Rivera Alvarez K. High throughput whole-slide scanning to enable large-scale data repository building. J Pathol 2022; 257:383-390. [PMID: 35511469 PMCID: PMC9327504 DOI: 10.1002/path.5923] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Revised: 04/19/2022] [Accepted: 05/02/2022] [Indexed: 11/07/2022]
Abstract
Digital pathology and artificial intelligence (AI) rely on digitization of patient material as a necessary first step. AI development benefits from large sample sizes and diverse cohorts, and therefore efforts to digitize glass slides must meet these needs in an efficient and cost-effective manner. Technical innovation in whole-slide imaging has enabled high throughput slide scanning through the coordinated increase in scanner capacity, speed, and automation. Combining these hardware innovations with automated informatics approaches has enabled more efficient workflows and the opportunity to provide higher quality imaging data using fewer personnel. Here we review several practical considerations for deploying high throughput scanning and we present strategies to increase efficiency with a focus on quality. Finally, we review remaining challenges and issue a call to vendors to innovate in the areas of automation and quality control in order to make high throughput scanning realizable to laboratories with limited resources. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Mark D Zarella
- Department of Pathology, Johns Hopkins University, Baltimore, MD, USA.,Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | | |
Collapse
|
9
|
Brozgol E, Kumar P, Necula D, Bronshtein-Berger I, Lindner M, Medalion S, Twito L, Shapira Y, Gondra H, Barshack I, Garini Y. Cancer detection from stained biopsies using high-speed spectral imaging. BIOMEDICAL OPTICS EXPRESS 2022; 13:2503-2515. [PMID: 35519262 PMCID: PMC9045910 DOI: 10.1364/boe.445782] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Revised: 01/31/2022] [Accepted: 02/06/2022] [Indexed: 06/14/2023]
Abstract
The escalating demand for diagnosing pathological biopsies requires the procedures to be expedited and automated. The existing imaging systems for measuring biopsies only measure color, and even though a lot of effort is invested in deep learning analysis, there are still serious challenges regarding the performance and validity of the data for the intended medical setting. We developed a system that rapidly acquires spectral images from biopsies, followed by spectral classification algorithms. The spectral information is remarkably more informative than the color information, and leads to very high accuracy in identifying cancer cells, as tested on tens of cancer cases. This was improved even more by using artificial intelligence algorithms that required a rather small training set, indicating the high level of information that exists in the spectral images. The most important spectral differences are observed in the nucleus and they are related to aneuploidy in tumor cells. Rapid spectral imaging measurement therefore can bridge the gap in the machine-aided diagnostics of whole biopsies, thus improving patient care, and expediting the treatment procedure.
Collapse
Affiliation(s)
- Eugene Brozgol
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Contributed equally
| | - Pramod Kumar
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Contributed equally
| | - Daniela Necula
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | | | - Moshe Lindner
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | | | - Lee Twito
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | - Yotam Shapira
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
| | - Helena Gondra
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
| | - Iris Barshack
- Department of Pathology, Sheba Medical Center, Ramat Gan, Israel
- Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv, Israel
- Equal supervision
| | - Yuval Garini
- Physics Department and Nanotechnology Institute, Bar Ilan University, Ramat Gan, Israel
- Biomedical Engineering Faculty, Technion − Israel Institute of Technology, Haifa, Israel
- Equal supervision
| |
Collapse
|
10
|
Zuraw A, Aeffner F. Whole-slide imaging, tissue image analysis, and artificial intelligence in veterinary pathology: An updated introduction and review. Vet Pathol 2021; 59:6-25. [PMID: 34521285 DOI: 10.1177/03009858211040484] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Since whole-slide imaging has been commercially available for over 2 decades, digital pathology has become a constantly expanding aspect of the pathology profession that will continue to significantly impact how pathologists conduct their craft. While some aspects, such as whole-slide imaging for archiving, consulting, and teaching, have gained broader acceptance, other facets such as quantitative tissue image analysis and artificial intelligence-based assessments are still met with some reservations. While most vendors in this space have focused on diagnostic applications, that is, viewing one or few slides at a time, some are developing solutions tailored more specifically to the various aspects of veterinary pathology including updated diagnostic, discovery, and research applications. This has especially advanced the use of digital pathology in toxicologic pathology and drug development, for primary reads as well as peer reviews. It is crucial that pathologists gain a deeper understanding of digital pathology and tissue image analysis technology and their applications in order to fully use these tools in a way that enhances and improves the pathologist's assessment as well as work environment. This review focuses on an updated introduction to the basics of digital pathology and image analysis and introduces emerging topics around artificial intelligence and machine learning.
Collapse
Affiliation(s)
| | - Famke Aeffner
- Amgen Inc, Amgen Research, South San Francisco, CA, USA
| |
Collapse
|
11
|
Lujan G, Quigley JC, Hartman D, Parwani A, Roehmholdt B, Meter BV, Ardon O, Hanna MG, Kelly D, Sowards C, Montalto M, Bui M, Zarella MD, LaRosa V, Slootweg G, Retamero JA, Lloyd MC, Madory J, Bowman D. Dissecting the Business Case for Adoption and Implementation of Digital Pathology: A White Paper from the Digital Pathology Association. J Pathol Inform 2021; 12:17. [PMID: 34221633 PMCID: PMC8240548 DOI: 10.4103/jpi.jpi_67_20] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2020] [Revised: 09/20/2020] [Accepted: 11/02/2020] [Indexed: 12/13/2022] Open
Abstract
We believe the switch to a digital pathology (DP) workflow is imminent and it is essential to understand the economic implications of conversion. Many aspects of the adoption of DP will be disruptive and have a direct financial impact, both in short term costs, such as investment in equipment and personnel, and long term revenue potential, such as improved productivity and novel tests. The focus of this whitepaper is to educate pathologists, laboratorians and other stakeholders about the business and monetary considerations of converting to a digital pathology workflow. The components of a DP business plan will be thoroughly summarized, and guidance will be provided on how to build a case for adoption and implementation as well as a roadmap for transitioning from an analog to a digital pathology workflow in various laboratory settings. It is important to clarify that this publication is not intended to list prices although some financials will be mentioned as examples. The authors encourage readers who are evaluating conversion to a DP workflow to use this paper as a foundational guide for conducting a thorough and complete assessment while incorporating in current market pricing. Contributors to this paper analyzed peer-reviewed literature and data collected from various institutions, some of which are mentioned. Digital pathology will change the way we practice through facilitating patient access to expert pathology services and enabling image analysis tools and assays to aid in diagnosis, prognosis, risk stratification and therapeutic selection. Together, they will result in the delivery of valuable information from which to make better decisions and improve the health of patients.
Collapse
Affiliation(s)
- Giovanni Lujan
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | | | - Douglas Hartman
- Department of Pathology, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Anil Parwani
- Department of Pathology, The Ohio State University Wexner Medical Center, Columbus, OH, USA
| | - Brian Roehmholdt
- Department of Pathology, Southern California Permanente Medical Group, La Canada Flintridge, CA, USA
| | | | - Orly Ardon
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Matthew G. Hanna
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | | | | | | | - Marilyn Bui
- Department of Anatomic Pathology, H. Lee Moffitt Cancer Center, Tampa, FL, USA
| | - Mark D. Zarella
- Johns Hopkins Medicine Pathology Informatics, Baltimore, MD 21287, USA
| | - Victoria LaRosa
- Education Services Department, Oracle Corp, Austin, Texas, USA
| | | | | | | | - James Madory
- Department of Pathology, Medical University of South Carolina, Charleston, SC, USA
| | | |
Collapse
|
12
|
Farris AB, Vizcarra J, Amgad M, Cooper LAD, Gutman D, Hogan J. Artificial intelligence and algorithmic computational pathology: an introduction with renal allograft examples. Histopathology 2021; 78:791-804. [PMID: 33211332 DOI: 10.1111/his.14304] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Whole slide imaging, which is an important technique in the field of digital pathology, has recently been the subject of increased interest and avenues for utilisation, and with more widespread whole slide image (WSI) utilisation, there will also be increased interest in and implementation of image analysis (IA) techniques. IA includes artificial intelligence (AI) and targeted or hypothesis-driven algorithms. In the overall pathology field, the number of citations related to these topics has increased in recent years. Renal pathology is one anatomical pathology subspecialty that has utilised WSIs and IA algorithms; it can be argued that renal transplant pathology could be particularly suited for whole slide imaging and IA, as renal transplant pathology is frequently classified by use of the semiquantitative Banff classification of renal allograft pathology. Hypothesis-driven/targeted algorithms have been used in the past for the assessment of a variety of features in the kidney (e.g. interstitial fibrosis, tubular atrophy, inflammation); in recent years, the amount of research has particularly increased in the area of AI/machine learning for the identification of glomeruli, for histological segmentation, and for other applications. Deep learning is the form of machine learning that is most often used for such AI approaches to the 'big data' of pathology WSIs, and deep learning methods such as artificial neural networks (ANNs)/convolutional neural networks (CNNs) are utilised. Unsupervised and supervised AI algorithms can be employed to accomplish image or semantic classification. In this review, AI and other IA algorithms applied to WSIs are discussed, and examples from renal pathology are covered, with an emphasis on renal transplant pathology.
Collapse
Affiliation(s)
- Alton B Farris
- Department of Pathology and Laboratory Medicine, Atlanta, GA, USA
| | - Juan Vizcarra
- Department of Bioinformatics, Emory University, Atlanta, GA, USA
| | - Mohamed Amgad
- Department of Pathology and Center for Computational Imaging and Signal Analytics, Northwestern University, Chicago, IL, USA
| | - Lee A D Cooper
- Department of Pathology and Center for Computational Imaging and Signal Analytics, Northwestern University, Chicago, IL, USA
| | - David Gutman
- Department of Bioinformatics, Emory University, Atlanta, GA, USA
| | - Julien Hogan
- Department of Surgery, Emory University, Atlanta, GA, USA
| |
Collapse
|
13
|
Roy M, Wang F, Vo H, Teng D, Teodoro G, Farris AB, Castillo-Leon E, Vos MB, Kong J. Deep-learning-based accurate hepatic steatosis quantification for histological assessment of liver biopsies. J Transl Med 2020; 100:1367-1383. [PMID: 32661341 PMCID: PMC7502534 DOI: 10.1038/s41374-020-0463-y] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2020] [Revised: 01/30/2020] [Accepted: 06/22/2020] [Indexed: 12/17/2022] Open
Abstract
Hepatic steatosis droplet quantification with histology biopsies has high clinical significance for risk stratification and management of patients with fatty liver diseases and in the decision to use donor livers for transplantation. However, pathology reviewing processes, when conducted manually, are subject to a high inter- and intra-reader variability, due to the overwhelmingly large number and significantly varying appearance of steatosis instances. This process is challenging as there is a large number of overlapped steatosis droplets with either missing or weak boundaries. In this study, we propose a deep-learning-based region-boundary integrated network for precise steatosis quantification with whole slide liver histopathology images. The proposed model consists of two sequential steps: a region extraction and a boundary prediction module for foreground regions and steatosis boundary prediction, followed by an integrated prediction map generation. Missing steatosis boundaries are next recovered from the predicted map and assembled from adjacent image patches to generate results for the whole slide histopathology image. The resulting steatosis measures both at the pixel level and steatosis object-level present strong correlation with pathologist annotations, radiology readouts and clinical data. In addition, the segregated steatosis object count is shown as a promising alternative measure to the traditional metrics at the pixel level. These results suggest a high potential of artificial intelligence-assisted technology to enhance liver disease decision support using whole slide images.
Collapse
Affiliation(s)
- Mousumi Roy
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Fusheng Wang
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA.
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, 11794, USA.
| | - Hoang Vo
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA
| | - Dejun Teng
- Department of Computer Science, Stony Brook University, Stony Brook, NY, 11794, USA
| | - George Teodoro
- Department of Computer Science, Federal University of Minas Gerais, Belo Horizonte, MG, 31270, USA
| | - Alton B Farris
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, 30322, USA
| | - Eduardo Castillo-Leon
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
| | - Miriam B Vos
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Pediatrics, Emory University, Atlanta, GA, 30322, USA
- Children's Healthcare of Atlanta, Atlanta, GA, 30322, USA
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, GA, 30303, USA.
- Department of Computer Science, Emory University, Atlanta, GA, 30322, USA.
- Department of Biomedical Informatics, Emory University, Atlanta, GA, 30322, USA.
- Winship Cancer Institute, Emory University, Atlanta, GA, 30322, USA.
| |
Collapse
|
14
|
Jiang K, Mohammad MK, Dar WA, Kong J, Farris AB. Quantitative assessment of liver fibrosis by digital image analysis reveals correlation with qualitative clinical fibrosis staging in liver transplant patients. PLoS One 2020; 15:e0239624. [PMID: 32986732 PMCID: PMC7521727 DOI: 10.1371/journal.pone.0239624] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Accepted: 09/10/2020] [Indexed: 12/19/2022] Open
Abstract
Technologies for digitizing tissues provide important quantitative data for liver histopathology investigation. We aimed to assess liver fibrosis degree with quantitative morphometric measurements of histopathological sections utilizing digital image analysis (DIA) and to further investigate if a correlation with histopathologic scoring (Scheuer staging) exists. A retrospective study of patients with at least two post-liver transplant biopsies having a Scheuer stage of ≤ 2 at baseline were gathered. Portal tract fibrotic percentage (%) and size (μm2) were measured by DIA, while clinical fibrosis score was measured by the Scheuer system. Correlations between DIA measurements and Scheuer scores were computed by Spearman correlation analysis. Differences between mean levels of fibrosis (score, size, and percentage) at baseline versus second visit were computed by Student’s t-test. P values < 0.05 were considered significant. Of 22 patients who met the study criteria, 54 biopsies were included for analysis. Average levels ±standard error [S.E.] of portal tract fibrotic percentage (%) and size (μm2) progressed from 46.5 ± 3.6% at baseline to 61.8 ± 3.8% at the second visit (P = 0.005 by Student’s t-test), and from 28,075 ± 3,232 μm2 at base line to 67,146 ± 10,639 μm2 at the second visit (P = 0.002 by Student’s t-test), respectively. Average levels of Scheuer fibrosis scores progressed from 0.55±0.19 at baseline to 1.14±0.26 at the second visit (P = 0.02 by Student’s t-test). Portal tract fibrotic percentage (%) and portal tract fibrotic size were directly correlated with clinical Scheuer fibrosis stage, with Spearman correlation coefficient and P value computed as r = 0.70, P < 0.0001 and r = 0.41, P = 0.002, respectively. Digital quantitative assessment of portal triad size and fibrosis percentage demonstrates a strong correlation with visually assessed histologic stage of liver fibrosis and complements the standard assessment for allograft monitoring, suggesting the utility of future WSI analysis.
Collapse
Affiliation(s)
- Kun Jiang
- Department of Pathology, Emory University, Atlanta, Georgia, United States of America
- Department of Pathology, University of South Florida, Tampa, Florida, United States of America
| | - Mohammad K. Mohammad
- Department of Pathology, Emory University, Atlanta, Georgia, United States of America
| | - Wasim A. Dar
- Department of Surgery, The University of Texas Health Science Center, Houston, Texas, United States of America
| | - Jun Kong
- Department of Mathematics and Statistics, Georgia State University, Atlanta, Georgia, United States of America
- Department of Computer Science, Georgia State University, Atlanta, Georgia, United States of America
- Department of Computer Science, Emory University, Atlanta, Georgia, United States of America
- Department of Biomedical Informatics, Emory University, Atlanta, Georgia, United States of America
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
| | - Alton B. Farris
- Department of Pathology, Emory University, Atlanta, Georgia, United States of America
- Winship Cancer Institute, Emory University, Atlanta, Georgia, United States of America
- * E-mail:
| |
Collapse
|
15
|
Farris AB, Moghe I, Wu S, Hogan J, Cornell LD, Alexander MP, Kers J, Demetris AJ, Levenson RM, Tomaszewski J, Barisoni L, Yagi Y, Solez K. Banff Digital Pathology Working Group: Going digital in transplant pathology. Am J Transplant 2020; 20:2392-2399. [PMID: 32185875 PMCID: PMC7496838 DOI: 10.1111/ajt.15850] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2020] [Revised: 02/25/2020] [Accepted: 02/27/2020] [Indexed: 01/25/2023]
Abstract
The Banff Digital Pathology Working Group (DPWG) was formed in the time leading up to and during the joint American Society for Histocompatibility and Immunogenetics/Banff Meeting, September 23-27, 2019, held in Pittsburgh, Pennsylvania. At the meeting, the 14th Banff Conference, presentations directly and peripherally related to the topic of "digital pathology" were presented; and discussions before, during, and after the meeting have resulted in a list of issues to address for the DPWG. Included are practice standardization, integrative approaches for study classification, scoring of histologic parameters (eg, interstitial fibrosis and tubular atrophy and inflammation), algorithm classification, and precision diagnosis (eg, molecular pathways and therapeutics). Since the meeting, a survey with international participation of mostly pathologists (81%) was conducted, showing that whole slide imaging is available at the majority of centers (71%) but that artificial intelligence (AI)/machine learning was only used in ≈12% of centers, with a wide variety of programs/algorithms employed. Digitalization is not just an end in itself. It also is a necessary precondition for AI and other approaches. Discussions at the meeting and the survey highlight the unmet need for a Banff DPWG and point the way toward future contributions that can be made.
Collapse
Affiliation(s)
| | | | - Simon Wu
- University of AlbertaEdmontonCanada
| | | | | | | | - Jesper Kers
- Amsterdam University Medical CentersAmsterdamthe Netherlands,Leiden University Medical CenterLeidenthe Netherlands
| | | | | | - John Tomaszewski
- University at BuffaloState University of New YorkBuffaloNew York
| | | | - Yukako Yagi
- Memorial Sloan Kettering Cancer CenterNew YorkNew York
| | | |
Collapse
|
16
|
Convergence of Digital Pathology and Artificial Intelligence Tools in Anatomic Pathology Practice: Current Landscape and Future Directions. Adv Anat Pathol 2020; 27:221-226. [PMID: 32541593 DOI: 10.1097/pap.0000000000000271] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
17
|
Ishihara S, Okada S, Ogi H, Kodama Y, Shimomura M, Tsunezuka H, Itoh K, Marx A, Inoue M. Programmed death-ligand 1 expression profiling in thymic epithelial cell tumors: Clinicopathological features and quantitative digital image analyses. Lung Cancer 2020; 145:40-47. [PMID: 32402921 DOI: 10.1016/j.lungcan.2020.04.038] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Revised: 04/26/2020] [Accepted: 04/28/2020] [Indexed: 12/22/2022]
Abstract
OBJECTIVES Whether the extent of expression of programmed death-ligand 1 (PD-L1) is clinically significant remains uncertain, although immuno-oncological features have been studied in thymic epithelial cell tumors (TETs). We investigated the histological features of PD-L1 expression in TETs, and assessed PD-L1 expression using digital image analysis. MATERIALS AND METHODS Participants comprised 66 patients with TET who underwent surgical resection between 2001 and 2016. We calculated tumor cell-positive ratio as total proportion score (TPS) with immunohistochemistry using SP263 anti-PD-L1 monoclonal antibody. PD-L1 expression was also quantified using digital image analysis of whole-slide images. We evaluated the relationship between conventional visual TPS using optical microscopy (TPS-V) and TPS from digital image analysis (TPS-IA). We further classified all TETs into high or low PD-L1 expression groups and assessed the clinical significance of PD-L1 expression level using TPS-V and TPS-IA. RESULTS WHO histological types were Type A (n = 8), AB (n = 18), B1 (n = 5), B2 (n = 16), B3 (n = 6), metaplastic thymoma (n = 2), and thymic carcinoma (TC) (n = 11). Median TPS-Vs were 2%, 2%, 10 %, 65 %, 90 %, 1%, and 20 %, respectively. TPS-IAs correlated with TPS-Vs in TETs overall and in thymomas, but not in TCs. PD-L1 expression levels in TETs differed significantly among histological types. Whether TPS-V or TPS-IA were used, the PD-L1high group included more cases of the more aggressive histological types. Recurrence-free survival (RFS) was shorter in the PD-L1high group than in the PD-L1low group in thymoma using TPS-IA, whereas RFS of the PD-L1high group was shorter in all TETs using TPS-V. CONCLUSION PD-L1 expression levels depended on the histological type of TET. Extensive PD-L1 expression in TETs was associated with poor prognosis. Digital image analysis is feasible for evaluating PD-L1 expression in TETs and might offer clinically relevant features of thymomas.
Collapse
Affiliation(s)
- Shunta Ishihara
- Division of Thoracic Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Satoru Okada
- Division of Thoracic Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hiroshi Ogi
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yoshinori Kodama
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Masanori Shimomura
- Division of Thoracic Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Hiroaki Tsunezuka
- Division of Thoracic Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Kyoko Itoh
- Department of Pathology and Applied Neurobiology, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Alexander Marx
- Institute of Pathology, University Medical Centre Mannheim, Heidelberg University, Mannheim, Germany
| | - Masayoshi Inoue
- Division of Thoracic Surgery, Department of Surgery, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, 465 Kajii-cho, Kamigyo-ku, Kyoto 602-8566, Japan.
| |
Collapse
|
18
|
Abstract
Current conventional endoscopes have restricted the accuracy of treatment delivery and monitoring. Over the past decade, there have been major developments in nanotechnology and light triggered therapy, potentially allowing a better detection of challenging lesions and targeted treatment of malignancies in the gastrointestinal tract. Theranostics is a developing form of personalized medicine because it combines diagnosis and targeted treatment delivered in one step using advances in nanotechnology. This review describes the light-triggered therapies (including photodynamic, photothermal, and photoimmunotherapies), nanotechnological advances with nanopowder, nanostent, nanogels, and nanoparticles, enhancements brought to endoscopic ultrasound, in addition to experimental endoscopic techniques, combining both enhanced diagnoses and therapies, including a developed prototype of a “smart” multifunctional endoscope for localized colorectal cancer, near-infrared laser endoscope targeting the gastrointestinal stromal tumors, the concept of endocapsule for obscure gastrointestinal bleed, and a proof-of-concept therapeutic capsule using ultrasound-mediated targeted drug delivery. Hence, the following term has been proposed encompassing these technologies: “Theranostic gastrointestinal endoscopy.” Future efforts for integration of these technologies into clinical practice would be directed toward translational and clinical trials translating into a more personalized and interdisciplinary diagnosis and treatment, shorter procedural time, higher precision, higher cost-effectiveness, and less need for repetitive procedures.
Collapse
|
19
|
Morriss NJ, Conley GM, Ospina SM, Meehan III WP, Qiu J, Mannix R. Automated Quantification of Immunohistochemical Staining of Large Animal Brain Tissue Using QuPath Software. Neuroscience 2020; 429:235-244. [DOI: 10.1016/j.neuroscience.2020.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2019] [Revised: 12/20/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022]
|
20
|
Melo RCN, Raas MWD, Palazzi C, Neves VH, Malta KK, Silva TP. Whole Slide Imaging and Its Applications to Histopathological Studies of Liver Disorders. Front Med (Lausanne) 2020; 6:310. [PMID: 31970160 PMCID: PMC6960181 DOI: 10.3389/fmed.2019.00310] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2019] [Accepted: 12/09/2019] [Indexed: 12/11/2022] Open
Abstract
Histological analysis of hepatic tissue specimens is essential for evaluating the pathology of several liver disorders such as chronic liver diseases, hepatocellular carcinomas, liver steatosis, and infectious liver diseases. Manual examination of histological slides on the microscope is a classically used method to study these disorders. However, it is considered time-consuming, limited, and associated with intra- and inter-observer variability. Emerging technologies such as whole slide imaging (WSI), also termed virtual microscopy, have increasingly been used to improve the assessment of histological features with applications in both clinical and research laboratories. WSI enables the acquisition of the tissue morphology/pathology from glass slides and translates it into a digital form comparable to a conventional microscope, but with several advantages such as easy image accessibility and storage, portability, sharing, annotation, qualitative and quantitative image analysis, and use for educational purposes. WSI-generated images simultaneously provide high resolution and a wide field of observation that can cover the entire section, extending any single field of view. In this review, we summarize current knowledge on the application of WSI to histopathological analyses of liver disorders as well as to understand liver biology. We address how WSI may improve the assessment and quantification of multiple histological parameters in the liver, and help diagnose several hepatic conditions with important clinical implications. The WSI technical limitations are also discussed.
Collapse
Affiliation(s)
- Rossana C N Melo
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Maximilian W D Raas
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil.,Faculty of Medical Sciences, Radboud University, Nijmegen, Netherlands
| | - Cinthia Palazzi
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Vitor H Neves
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Kássia K Malta
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| | - Thiago P Silva
- Laboratory of Cellular Biology, Department of Biology, Federal University of Juiz de Fora, Juiz de Fora, Brazil
| |
Collapse
|
21
|
Zarella MD, Jakubowski J. Video compression to support the expansion of whole-slide imaging into cytology. J Med Imaging (Bellingham) 2019; 6:047502. [PMID: 31890747 PMCID: PMC6921690 DOI: 10.1117/1.jmi.6.4.047502] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 11/20/2019] [Indexed: 12/31/2022] Open
Abstract
Digital screening and diagnosis from cytology slides can be aided by capturing multiple focal planes. However, using conventional methods, the large file sizes of high-resolution whole-slide images increase linearly with the number of focal planes acquired, leading to significant data storage and bandwidth requirements for the efficient storage and transfer of cytology virtual slides. We investigated whether a sequence of focal planes contained sufficient redundancy to efficiently compress virtual slides across focal planes by applying a commonly available video compression standard, high-efficiency video coding (HEVC). By developing an adaptive algorithm that applied compression to achieve a target image quality, we found that the compression ratio of HEVC exceeded that obtained using JPEG and JPEG2000 compression while maintaining a comparable level of image quality. These results suggest an alternative method for the efficient storage and transfer of whole-slide images that contain multiple focal planes, expanding the utility of this rapidly evolving imaging technology into cytology.
Collapse
Affiliation(s)
- Mark D Zarella
- Drexel University, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States
| | - Jennifer Jakubowski
- Drexel University, Department of Pathology and Laboratory Medicine, Philadelphia, Pennsylvania, United States.,Drexel University, School of Biomedical Engineering, Science and Health Systems, Philadelphia, Pennsylvania, United States
| |
Collapse
|
22
|
Larghi A, Fornelli A, Lega S, Ragazzi M, Carlinfante G, Baccarini P, Fabbri C, Pierotti P, Tallini G, Bondi A, de Biase D. Concordance, intra- and inter-observer agreements between light microscopy and whole slide imaging for samples acquired by EUS in pancreatic solid lesions. Dig Liver Dis 2019; 51:1574-1579. [PMID: 31147212 DOI: 10.1016/j.dld.2019.04.019] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/10/2019] [Accepted: 04/29/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND No study has compared the performance of light microscopy (LM) and whole slide imaging (WSI) for endoscopic ultrasound (EUS) histological acquired tissue samples from pancreatic solid lesions (PSLs). We evaluated the concordance between LM and WSI and the inter- and intra-observer agreements among pathologists on PSLs EUS acquired samples. METHODS LM and WSI from 60 patients with PSLs were evaluated by five expert pathologists to define: diagnostic classification, presence of a core, number and percentage of lesional cells. Washout period between evaluations was 3 months. Time of the procedures was also assessed. RESULTS Forty-eight cell-block and 12 biopsy samples were evaluated. A high concordance between LM and WSI was found. Inter- and intra-observer agreements for diagnostic classification were substantial and complete, respectively. For all the other parameters, the inter-observer agreement was usually higher for LM. For the intra-observer, a substantial agreement was reached regarding the presence of tissue core and the number and the percentage of malignant cells. Median time for performing LM was significantly shorter than for WSI (p < 0.0001). CONCLUSIONS LM and WSI of cell-block and biopsy samples acquired by EUS in PSLs were highly concordant, with a substantial inter-observer and a complete intra-observer agreements regarding diagnostic classification.
Collapse
Affiliation(s)
- Alberto Larghi
- Digestive Endoscopy Unit, Foundation University Hospital Policlinico A. Gemelli IRCCS, CERTT, Center for Endoscopic Research Therapeutics and Training, Catholic University, Rome, Italy
| | | | | | - Moira Ragazzi
- Pathology Unit, S. Maria Nuova Hospital, IRCSS-AUSL Reggio Emilia, Italy
| | | | | | - Carlo Fabbri
- Digestive Endoscopy and Gastroenterology, Forlì and Cesena Hospitals, Italy
| | | | - Giovanni Tallini
- Anatomical Pathology, Molecular Diagnostic Unit, University of Bologna School of Medicine, Bologna, Italy
| | - Arrigo Bondi
- Pathology Unit, Maggiore Hospital, Bologna, Italy
| | - Dario de Biase
- Department of Pharmacy and Biotechnology, Molecular Diagnostic Unit, University of Bologna, Bologna, Italy
| |
Collapse
|
23
|
Aeffner F, Adissu HA, Boyle MC, Cardiff RD, Hagendorn E, Hoenerhoff MJ, Klopfleisch R, Newbigging S, Schaudien D, Turner O, Wilson K. Digital Microscopy, Image Analysis, and Virtual Slide Repository. ILAR J 2019; 59:66-79. [PMID: 30535284 DOI: 10.1093/ilar/ily007] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2017] [Revised: 05/03/2018] [Indexed: 02/07/2023] Open
Abstract
Advancements in technology and digitization have ushered in novel ways of enhancing tissue-based research via digital microscopy and image analysis. Whole slide imaging scanners enable digitization of histology slides to be stored in virtual slide repositories and to be viewed via computers instead of microscopes. Easier and faster sharing of histologic images for teaching and consultation, improved storage and preservation of quality of stained slides, and annotation of features of interest in the digital slides are just a few of the advantages of this technology. Combined with the development of software for digital image analysis, digital slides further pave the way for the development of tools that extract quantitative data from tissue-based studies. This review introduces digital microscopy and pathology, and addresses technical and scientific considerations in slide scanning, quantitative image analysis, and slide repositories. It also highlights the current state of the technology and factors that need to be taken into account to insure optimal utility, including preanalytical considerations and the importance of involving a pathologist in all major steps along the digital microscopy and pathology workflow.
Collapse
Affiliation(s)
- Famke Aeffner
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Hibret A Adissu
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Michael C Boyle
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Robert D Cardiff
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Erik Hagendorn
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Mark J Hoenerhoff
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Robert Klopfleisch
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Susan Newbigging
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Dirk Schaudien
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Oliver Turner
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| | - Kristin Wilson
- Famke Aeffner, DVM PhD DACVP, is a principal pathologist in the Comparative Biology and Safety Sciences Department at Amgen Inc. in South San Francisco, California. Hibret Adissu, DVM PhD DVSc DACVP, is an investigative pathologist in the Laboratory of Cancer Biology and Genetics, Center for Cancer Research, at the National Cancer Institute in Bethesda, Maryland. Michael C. Boyle, DVM PhD DACVP DABT, is a principal pathologist in the Comparative Biology and Safety Sciences at Amgen Inc. in Thousand Oaks, California. Robert D. Cardiff, MD PhD, is a distinguished professor of pathology (emeritus) at the Center for Comparative Medicine at the University of California in Davis, California. Erik Hagendorn is a senior scientist of informatics at AbbVie Bioresearch in Worcester, Massachusetts. Mark J. Hoenerhoff, DVM PhD DACVP, is an associate professor and veterinary pathologist at the In Vivo Animal Core, Unit for Laboratory Animal Medicine, at the University of Michigan in Ann Arbor, Michigan. Robert Klopfleisch, DVM PhD DACVP, is an associate professor at the Institute of Veterinary Pathology of the Freie Universitaet Berlin, in Berlin, Germany. Susan Newbigging, BSc MSc DVM DVSc, is a pathologist and Director of The Pathology Core at the Toronto Center of Phenogenomics in Toronto, Ontario, Canada. Dirk Schaudien, DVM PhD DACVP, is a veterinary pathologist at the Fraunhofer Institute for Toxicology and Experimental Medicine, in Hannover, Germany. Oliver Turner, BSC(Hons), BVSc MRCVS PhD DACVP DABT, is a senior pathologist in the Preclinical Safety department of Novartis Pharmaceuticals in East Hanover, New Jersey. Kristin Wilson, DVM PhD DACVP, is a pathologist at Flagship Biosciences Inc. in Westminster, Colorado
| |
Collapse
|
24
|
Digital versus light microscopy assessment of extraprostatic extension in radical prostatectomy samples. Virchows Arch 2019; 475:735-744. [PMID: 31588959 DOI: 10.1007/s00428-019-02666-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2019] [Revised: 08/29/2019] [Accepted: 09/10/2019] [Indexed: 10/25/2022]
Abstract
Focal or non-focal/extensive extraprostatic extension of prostate carcinoma is an important pathologic prognostic parameter to be reported after radical prostatectomy. Currently, there is no agreement on how to measure and what are the best cutoff points to be used in practice. We hypothesized that digital microscopy would potentially provide more objective measurements of extraprostatic extension, thus better defining its clinical significance. To further our knowledge on digital prostate pathology, we evaluated the status of extraprostatic extension in 107 consecutive laparoscopic radical prostatectomy samples, using digital and conventional light microscopy. Mean linear and radial measurements of extraprostatic extension by digital microscopy significantly correlated to pT status (p = 0.022 and p = 0.050, respectively) but only radial measurements correlated to biochemical recurrence (p = 0.042) and grade groups (p = 0.022). None of the measurements, whether conventional or digital, were associated with lymph node status. Receiving operating characteristic analysis showed a potential cutoff point to assess linear measurements by conventional (< vs. > 24.21 mm) or digital microscopy (< vs. > 15 mm) or by radial measurement (< vs. > 1.6 mm). Finally, we observed an association between the number of paraffin blocks bearing EPE with pT (p = 0.041) status (digital microscopy), and linear measurements by conventional (p = 0.044) or digital microscopy (p = 0.045) with lymph node status. Reporting EPE measurements by digital microscopy, both linear and radial, and the number of paraffin blocks with EPE, might provide additional prognostic features after radical prostatectomy.
Collapse
|
25
|
Chong T, Palma-Diaz MF, Fisher C, Gui D, Ostrzega NL, Sempa G, Sisk AE, Valasek M, Wang BY, Zuckerman J, Khacherian C, Binder S, Wallace WD. The California Telepathology Service: UCLA's Experience in Deploying a Regional Digital Pathology Subspecialty Consultation Network. J Pathol Inform 2019; 10:31. [PMID: 31620310 PMCID: PMC6788184 DOI: 10.4103/jpi.jpi_22_19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Accepted: 07/01/2019] [Indexed: 11/25/2022] Open
Abstract
Background: The need for extending pathology diagnostic expertise to more areas is now being met by the maturation of technology that can effectively deliver this level of care. The experience and lessons learned from our successfully deployed International Telepathology Service (ITS) to a hospital system in China guided us in starting a domestic telepathology network, the California Telepathology Service (CTS). Many of the lessons learned from the ITS project informed our decision-making for the CTS. New challenges were recognized and overcome, such as addressing the complexity and cost–benefit tradeoffs involved in setting up a digital consultation system that competes with an established conventional glass slide delivery system. Methods: The CTS is based on a hub-and-spoke telepathology network using Leica Biosystems whole-slide image scanners and the eSlide Manager (eSM Version 12.3.3.7055, Leica Biosystems) digital image management software solution. The service currently comprises six spoke sites (UC San Diego [UCSD], UC Irvine [UCI], UC Davis, Northridge Hospital Medical Center [NHMC], Olive View Medical Center [OVMC], and Children's Hospital Los Angeles) and one central hub site (UCLA Medical Center). So far, five sites have been validated for telepathology case consultations following established practice guidelines, and four sites (UCI, UCSD, NHMC, and OVMC) have activated the service. Results: For the active spoke sites, we reviewed the volume, turnaround time (TAT), and case types and evaluated for utility and value. From May 2017 to July 2018, a total of 165 cases were submitted. Of note, digital consultations were particularly advantageous for preliminary kidney biopsy diagnoses (avg TAT 0.7 day). Conclusion: For spoke sites, telepathology provided shortened TAT and significant financial savings over hiring faculty with expertise to support a potentially low-volume service. For the hub site, the value includes exposure to educationally valuable cases, additional caseload volume to support specialized services, and improved communication with referring facilities over traditional carrier mail. The creation of a hub-and-spoke telepathology network is an expensive undertaking, and careful consideration needs to be given to support the needs of the clinical services, acquisition and effective deployment of the appropriate equipment, network requirements, and laboratory workflows to ensure a successful and cost-effective system.
Collapse
Affiliation(s)
- Thomas Chong
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - M Fernando Palma-Diaz
- Kaiser Permanente Los Angeles Medical Center, Department of Pathology, Los Angeles, CA, USA
| | - Craig Fisher
- UCSD Medical Center Pathology, San Diego, CA, USA
| | - Dorina Gui
- Department of Pathology and Laboratory Medicine, University of California, Sacramento, CA, USA
| | - Nora L Ostrzega
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Geoffrey Sempa
- Department of Pathology and Laboratory Medicine, UC Irvine School of Medicine, Irvine, CA, USA
| | - Anthony E Sisk
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Mark Valasek
- UCSD Medical Center Pathology, San Diego, CA, USA
| | - Beverly Y Wang
- Department of Pathology and Laboratory Medicine, UC Irvine School of Medicine, Irvine, CA, USA
| | - Jonathan Zuckerman
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Chris Khacherian
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| | - Scott Binder
- Affiliated Pathologists Medical Group, Inc., Rancho Dominguez, CA, USA
| | - W Dean Wallace
- Department of Pathology and Laboratory Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
| |
Collapse
|
26
|
Dealing with Lack of Training Data for Convolutional Neural Networks: The Case of Digital Pathology. ELECTRONICS 2019. [DOI: 10.3390/electronics8030256] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Thanks to their capability to learn generalizable descriptors directly from images, deep Convolutional Neural Networks (CNNs) seem the ideal solution to most pattern recognition problems. On the other hand, to learn the image representation, CNNs need huge sets of annotated samples that are unfeasible in many every-day scenarios. This is the case, for example, of Computer-Aided Diagnosis (CAD) systems for digital pathology, where additional challenges are posed by the high variability of the cancerous tissue characteristics. In our experiments, state-of-the-art CNNs trained from scratch on histological images were less accurate and less robust to variability than a traditional machine learning framework, highlighting all the issues of fully training deep networks with limited data from real patients. To solve this problem, we designed and compared three transfer learning frameworks, leveraging CNNs pre-trained on non-medical images. This approach obtained very high accuracy, requiring much less computational resource for the training. Our findings demonstrate that transfer learning is a solution to the automated classification of histological samples and solves the problem of designing accurate and computationally-efficient CAD systems with limited training data.
Collapse
|
27
|
Heeke S, Delingette H, Fanjat Y, Long-Mira E, Lassalle S, Hofman V, Benzaquen J, Marquette CH, Hofman P, Ilié M. [The age of artificial intelligence in lung cancer pathology: Between hope, gloom and perspectives]. Ann Pathol 2019; 39:130-136. [PMID: 30772062 DOI: 10.1016/j.annpat.2019.01.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2018] [Accepted: 01/16/2019] [Indexed: 12/14/2022]
Abstract
Histopathology is the fundamental tool of pathology used for more than a century to establish the final diagnosis of lung cancer. In addition, the phenotypic data contained in the histological images reflects the overall effect of molecular alterations on the behavior of cancer cells and provides a practical visual reading of the aggressiveness of the disease. However, the human evaluation of the histological images is sometimes subjective and may lack reproducibility. Therefore, computational analysis of histological imaging using so-called "artificial intelligence" (AI) approaches has recently received considerable attention to improve this diagnostic accuracy. Thus, computational analysis of lung cancer images has recently been evaluated for the optimization of histological or cytological classification, prognostic prediction or genomic profile of patients with lung cancer. This rapidly growing field constantly demonstrates great power in the field of computing medical imaging by producing highly accurate detection, segmentation or recognition tasks. However, there are still several challenges or issues to be addressed in order to successfully succeed the actual transfer into clinical routine. The objective of this review is to emphasize recent applications of AI in pulmonary cancer pathology, but also to clarify the advantages and limitations of this approach, as well as the perspectives to be implemented for a potential transfer into clinical routine.
Collapse
Affiliation(s)
- Simon Heeke
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France
| | - Hervé Delingette
- Équipe Asclepios, Inria Sophia-Antipolis, université Côte-d'Azur, 2004, route des Lucioles, 06902 Sophia-Antipolis, France
| | - Youta Fanjat
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France
| | - Elodie Long-Mira
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France
| | - Sandra Lassalle
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France
| | - Véronique Hofman
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France
| | - Jonathan Benzaquen
- Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France; Service de pneumologie, Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France
| | - Charles-Hugo Marquette
- Service de pneumologie, Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France
| | - Paul Hofman
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France
| | - Marius Ilié
- Laboratoire de pathologie clinique et expérimentale/biobanque (BB 0033-00025), Fédération hospitalo-universitaire OncoAge, CHU de Nice, université Côte-d'Azur, 30, voie Romaine, 06000 Nice, France; Équipe 4, CNRS UMR7284, Inserm U1081, faculté de médecine, institut de recherche sur le cancer et le vieillissement de Nice (Ircan), 28, avenue de Valombrose, 06107 Nice, France.
| |
Collapse
|
28
|
Zarella MD, Bowman; D, Aeffner F, Farahani N, Xthona; A, Absar SF, Parwani A, Bui M, Hartman DJ. A Practical Guide to Whole Slide Imaging: A White Paper From the Digital Pathology Association. Arch Pathol Lab Med 2018; 143:222-234. [DOI: 10.5858/arpa.2018-0343-ra] [Citation(s) in RCA: 150] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Context.—
Whole slide imaging (WSI) represents a paradigm shift in pathology, serving as a necessary first step for a wide array of digital tools to enter the field. Its basic function is to digitize glass slides, but its impact on pathology workflows, reproducibility, dissemination of educational material, expansion of service to underprivileged areas, and intrainstitutional and interinstitutional collaboration exemplifies a significant innovative movement with far-reaching effects. Although the benefits of WSI to pathology practices, academic centers, and research institutions are many, the complexities of implementation remain an obstacle to widespread adoption. In the wake of the first regulatory clearance of WSI for primary diagnosis in the United States, some barriers to adoption have fallen. Nevertheless, implementation of WSI remains a difficult prospect for many institutions, especially those with stakeholders unfamiliar with the technologies necessary to implement a system or who cannot effectively communicate to executive leadership and sponsors the benefits of a technology that may lack clear and immediate reimbursement opportunity.
Objectives.—
To present an overview of WSI technology—present and future—and to demonstrate several immediate applications of WSI that support pathology practice, medical education, research, and collaboration.
Data Sources.—
Peer-reviewed literature was reviewed by pathologists, scientists, and technologists who have practical knowledge of and experience with WSI.
Conclusions.—
Implementation of WSI is a multifaceted and inherently multidisciplinary endeavor requiring contributions from pathologists, technologists, and executive leadership. Improved understanding of the current challenges to implementation, as well as the benefits and successes of the technology, can help prospective users identify the best path for success.
Collapse
Affiliation(s)
- Mark D. Zarella
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Douglas Bowman;
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Famke Aeffner
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Navid Farahani
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Albert Xthona;
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Syeda Fatima Absar
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Anil Parwani
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Marilyn Bui
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| | - Douglas J. Hartman
- From the Department of Pathology & Laboratory Medicine, Drexel University College of Medicine, Philadelphia, Pennsylvania (Drs Zarella and Absar); Pharma Services, Indica Labs, Inc, Corrales, New Mexico (Mr Bowman); Comparative Biology and Safety Sciences, Amgen, Inc, South San Francisco, California (Dr Aeffner); 3Scan, San Francisco, California (Dr Farahani); Barco, Inc, Beaverton, Oregon (Mr Xt
| |
Collapse
|
29
|
Digital pathology in nephrology clinical trials, research, and pathology practice. Curr Opin Nephrol Hypertens 2018; 26:450-459. [PMID: 28858910 DOI: 10.1097/mnh.0000000000000360] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
PURPOSE OF REVIEW In this review, we will discuss (i) how the recent advancements in digital technology and computational engineering are currently applied to nephropathology in the setting of clinical research, trials, and practice; (ii) the benefits of the new digital environment; (iii) how recognizing its challenges provides opportunities for transformation; and (iv) nephropathology in the upcoming era of kidney precision and predictive medicine. RECENT FINDINGS Recent studies highlighted how new standardized protocols facilitate the harmonization of digital pathology database infrastructure and morphologic, morphometric, and computer-aided quantitative analyses. Digital pathology enables robust protocols for clinical trials and research, with the potential to identify previously underused or unrecognized clinically useful parameters. The integration of digital pathology with molecular signatures is leading the way to establishing clinically relevant morpho-omic taxonomies of renal diseases. SUMMARY The introduction of digital pathology in clinical research and trials, and the progressive implementation of the modern software ecosystem, opens opportunities for the development of new predictive diagnostic paradigms and computer-aided algorithms, transforming the practice of renal disease into a modern computational science.
Collapse
|
30
|
Cooper LAD, Demicco EG, Saltz JH, Powell RT, Rao A, Lazar AJ. PanCancer insights from The Cancer Genome Atlas: the pathologist's perspective. J Pathol 2018; 244:512-524. [PMID: 29288495 PMCID: PMC6240356 DOI: 10.1002/path.5028] [Citation(s) in RCA: 112] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2017] [Revised: 12/19/2017] [Accepted: 12/22/2017] [Indexed: 02/06/2023]
Abstract
The Cancer Genome Atlas (TCGA) represents one of several international consortia dedicated to performing comprehensive genomic and epigenomic analyses of selected tumour types to advance our understanding of disease and provide an open-access resource for worldwide cancer research. Thirty-three tumour types (selected by histology or tissue of origin, to include both common and rare diseases), comprising >11 000 specimens, were subjected to DNA sequencing, copy number and methylation analysis, and transcriptomic, proteomic and histological evaluation. Each cancer type was analysed individually to identify tissue-specific alterations, and make correlations across different molecular platforms. The final dataset was then normalized and combined for the PanCancer Initiative, which seeks to identify commonalities across different cancer types or cells of origin/lineage, or within anatomically or morphologically related groups. An important resource generated along with the rich molecular studies is an extensive digital pathology slide archive, composed of frozen section tissue directly related to the tissues analysed as part of TCGA, and representative formalin-fixed paraffin-embedded, haematoxylin and eosin (H&E)-stained diagnostic slides. These H&E image resources have primarily been used to verify diagnoses and histological subtypes with some limited extraction of standard pathological variables such as mitotic activity, grade, and lymphocytic infiltrates. Largely overlooked is the richness of these scanned images for more sophisticated feature extraction approaches coupled with machine learning, and ultimately correlation with molecular features and clinical endpoints. Here, we document initial attempts to exploit TCGA imaging archives, and describe some of the tools, and the rapidly evolving image analysis/feature extraction landscape. Our hope is to inform, and ultimately inspire and challenge, the pathology and cancer research communities to exploit these imaging resources so that the full potential of this integral platform of TCGA can be used to complement and enhance the insightful integrated analyses from the genomic and epigenomic platforms. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Lee AD Cooper
- Department of Biomedical Informatics, Emory University, Atlanta, GA, USA
- Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
- Winship Cancer Institute, Emory University, Atlanta, GA, USA
| | - Elizabeth G Demicco
- Department of Pathology and Laboratory Medicine, Sinai Health System, Toronto, Ontario, Canada
| | - Joel H Saltz
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, NY, USA
| | - Reid T Powell
- Center for Translational Cancer Research, Institute of Biosciences and Technology, Texas A&M University, Houston, TX, USA
| | - Arvind Rao
- Department of Bioinformatics and Computational Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Radiation Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Alexander J Lazar
- Departments of Pathology, Genomic Medicine, and Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
31
|
Abstract
Whole-slide imaging revolutionizes the field of pathology especially in the areas of facilitation of research, long-term storages, exchange of information, and image analysis. In this process, a scanning equipment (scanner) scans the whole glass slide into a digital file. In research in esophageal adenocarcinoma or other cancers, whole-slide imaging could help in production of high-resolution images for studying and sharing of research information, assessment of tissue microarray slides as well as allowing digital image analysis of the tissue information such as level of staining (e.g., HER2) in a more efficient and objective manner. In this chapter, we will elaborate the concepts, advantages, barriers, and the operations of whole-slide imaging scanning.
Collapse
Affiliation(s)
- Alfred K Lam
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia.
| | - Melissa Leung
- Cancer Molecular Pathology of School of Medicine, Griffith University, Gold Coast, Australia
| |
Collapse
|
32
|
Sugita S, Hirano H, Hatanaka Y, Fujita H, Kubo T, Kikuchi N, Ito Y, Sugawara T, Segawa K, Hisai H, Yamashita K, Nobuoka T, Matsuno Y, Hasegawa T. Image analysis is an excellent tool for quantifying Ki-67 to predict the prognosis of gastrointestinal stromal tumor patients. Pathol Int 2017; 68:7-11. [DOI: 10.1111/pin.12611] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2017] [Accepted: 08/02/2017] [Indexed: 12/23/2022]
Affiliation(s)
- Shintaro Sugita
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Hiroshi Hirano
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Yutaka Hatanaka
- Department of Surgical Pathology; Hokkaido University Hospital; Sapporo Hokkaido Japan
| | - Hiromi Fujita
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Terufumi Kubo
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Noriaki Kikuchi
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Yumika Ito
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Taro Sugawara
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Keiko Segawa
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Hiroyuki Hisai
- Department of Gastroenterology; Japanese Red Cross Date General Hospital; Date Hokkaido Japan
| | - Kentaro Yamashita
- Department of Gastroenterology; Rheumatology and Clinical Immunology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Takayuki Nobuoka
- Department of Surgery; Oncology and Science; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| | - Yoshihiro Matsuno
- Department of Surgical Pathology; Hokkaido University Hospital; Sapporo Hokkaido Japan
| | - Tadashi Hasegawa
- Department of Surgial Pathology; Sapporo Medical University School of Medicine; Sapporo Hokkaido Japan
| |
Collapse
|
33
|
Development of CD3 cell quantitation algorithms for renal allograft biopsy rejection assessment utilizing open source image analysis software. Virchows Arch 2017; 472:259-269. [DOI: 10.1007/s00428-017-2260-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2017] [Revised: 09/29/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022]
|
34
|
Montironi R, Cimadamore A, Massari F, Montironi MA, Lopez-Beltran A, Cheng L, Montorsi F, Scarpelli M. Whole Slide Imaging of Large Format Histology in Prostate Pathology: Potential for Information Fusion. Arch Pathol Lab Med 2017; 141:1460-1461. [DOI: 10.5858/arpa.2017-0198-le] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Affiliation(s)
- Rodolfo Montironi
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | - Alessia Cimadamore
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| | | | | | | | - Liang Cheng
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis
| | - Francesco Montorsi
- Unit of Urology/Division of Oncology, Urological Research Institute, Instituto di Ricovero e Cura a Carattere Scientifico, Ospedale San Raffaele, Milan, Italy
| | - Marina Scarpelli
- Section of Pathological Anatomy, Polytechnic University of the Marche Region, School of Medicine, United Hospitals, Ancona, Italy
| |
Collapse
|
35
|
Qaiser T, Mukherjee A, Reddy PB C, Munugoti SD, Tallam V, Pitkäaho T, Lehtimäki T, Naughton T, Berseth M, Pedraza A, Mukundan R, Smith M, Bhalerao A, Rodner E, Simon M, Denzler J, Huang CH, Bueno G, Snead D, Ellis IO, Ilyas M, Rajpoot N. HER2 challenge contest: a detailed assessment of automated HER2 scoring algorithms in whole slide images of breast cancer tissues. Histopathology 2017; 72:227-238. [DOI: 10.1111/his.13333] [Citation(s) in RCA: 62] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2017] [Accepted: 07/29/2017] [Indexed: 12/19/2022]
Affiliation(s)
- Talha Qaiser
- Department of Computer Science; University of Warwick; Coventry UK
| | - Abhik Mukherjee
- Department of Histopathology; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham UK
| | - Chaitanya Reddy PB
- Department of Electronics and Electrical Engineering; Indian Institute of Technology; Guwahati India
| | - Sai D Munugoti
- Department of Electronics and Electrical Engineering; Indian Institute of Technology; Guwahati India
| | - Vamsi Tallam
- Department of Electronics and Electrical Engineering; Indian Institute of Technology; Guwahati India
| | - Tomi Pitkäaho
- Department of Computer Science; Maynooth University; Maynooth Ireland
| | - Taina Lehtimäki
- Department of Computer Science; Maynooth University; Maynooth Ireland
| | - Thomas Naughton
- Department of Computer Science; Maynooth University; Maynooth Ireland
| | | | - Aníbal Pedraza
- VISILAB, E.T.S.I.I; University of Castilla-La Mancha; Ciudad Real Spain
| | - Ramakrishnan Mukundan
- Department of Computer Science and Software Engineering; University of Canterbury; Canterbury New Zealand
| | - Matthew Smith
- Department of Statistics; University of Warwick; Coventry UK
| | - Abhir Bhalerao
- Department of Computer Science; University of Warwick; Coventry UK
| | - Erik Rodner
- Computer Vision Group; Friedrich Schiller University of Jena; Jena Germany
| | - Marcel Simon
- Computer Vision Group; Friedrich Schiller University of Jena; Jena Germany
| | - Joachim Denzler
- Computer Vision Group; Friedrich Schiller University of Jena; Jena Germany
| | - Chao-Hui Huang
- MSD International GmbH; Singapore Singapore
- Singapore Agency for Science, Technology and Research; Singapore Singapore
| | - Gloria Bueno
- VISILAB, E.T.S.I.I; University of Castilla-La Mancha; Ciudad Real Spain
| | - David Snead
- Department of Pathology; University Hospitals Coventry and Warwickshire; Coventry UK
| | - Ian O Ellis
- Department of Histopathology; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham UK
| | - Mohammad Ilyas
- Department of Histopathology; Division of Cancer and Stem Cells; School of Medicine; University of Nottingham; Nottingham UK
- Nottingham Molecular Pathology Node; University of Nottingham; Nottingham UK
| | - Nasir Rajpoot
- Department of Computer Science; University of Warwick; Coventry UK
- Department of Pathology; University Hospitals Coventry and Warwickshire; Coventry UK
| |
Collapse
|
36
|
Affiliation(s)
- Umesh Kapur
- Associate Editor, JOH Department of Pathology Silver Cross Hospital, New Lenox, IL, USA
| |
Collapse
|