51
|
Lee Y, Alam MR, Park H, Yim K, Seo KJ, Hwang G, Kim D, Chung Y, Gong G, Cho NH, Yoo CW, Chong Y, Choi HJ. Improved Diagnostic Accuracy of Thyroid Fine-Needle Aspiration Cytology with Artificial Intelligence Technology. Thyroid 2024; 34:723-734. [PMID: 38874262 DOI: 10.1089/thy.2023.0384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 06/15/2024]
Abstract
Background: Artificial intelligence (AI) is increasingly being applied in pathology and cytology, showing promising results. We collected a large dataset of whole slide images (WSIs) of thyroid fine-needle aspiration cytology (FNA), incorporating z-stacking, from institutions across the nation to develop an AI model. Methods: We conducted a multicenter retrospective diagnostic accuracy study using thyroid FNA dataset from the Open AI Dataset Project that consists of digitalized images samples collected from 3 university hospitals and 215 Korean institutions through extensive quality check during the case selection, scanning, labeling, and reviewing process. Multiple z-layer images were captured using three different scanners and image patches were extracted from WSIs and resized after focus fusion and color normalization. We pretested six AI models, determining Inception ResNet v2 as the best model using a subset of dataset, and subsequently tested the final model with total datasets. Additionally, we compared the performance of AI and cytopathologists using randomly selected 1031 image patches and reevaluated the cytopathologists' performance after reference to AI results. Results: A total of 10,332 image patches from 306 thyroid FNAs, comprising 78 malignant (papillary thyroid carcinoma) and 228 benign from 86 institutions were used for the AI training. Inception ResNet v2 achieved highest accuracy of 99.7%, 97.7%, and 94.9% for training, validation, and test dataset, respectively (sensitivity 99.9%, 99.6%, and 100% and specificity 99.6%, 96.4%, and 90.4% for training, validation, and test dataset, respectively). In the comparison between AI and human, AI model showed higher accuracy and specificity than the average expert cytopathologists beyond the two-standard deviation (accuracy 99.71% [95% confidence interval (CI), 99.38-100.00%] vs. 88.91% [95% CI, 86.99-90.83%], sensitivity 99.81% [95% CI, 99.54-100.00%] vs. 87.26% [95% CI, 85.22-89.30%], and specificity 99.61% [95% CI, 99.23-99.99%] vs. 90.58% [95% CI, 88.80-92.36%]). Moreover, after referring to the AI results, the performance of all the experts (accuracy 96%, 95%, and 96%, respectively) and the diagnostic agreement (from 0.64 to 0.84) increased. Conclusions: These results suggest that the application of AI technology to thyroid FNA cytology may improve the diagnostic accuracy as well as intra- and inter-observer variability among pathologists. Further confirmatory research is needed.
Collapse
Affiliation(s)
- Yujin Lee
- Department of Hospital Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Mohammad Rizwan Alam
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Hongsik Park
- Department of Hospital Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| | - Kwangil Yim
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Kyung Jin Seo
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | | | | | | | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, Seoul, Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul, Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer Center, Ilsan, Republic of Korea
| | - Yosep Chong
- Department of Hospital Pathology, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Uijeongbu, Republic of Korea
| | - Hyun Joo Choi
- Department of Hospital Pathology, St. Vincent's Hospital, College of Medicine, The Catholic University of Korea, Suwon, Republic of Korea
| |
Collapse
|
52
|
McGenity C, Clarke EL, Jennings C, Matthews G, Cartlidge C, Freduah-Agyemang H, Stocken DD, Treanor D. Artificial intelligence in digital pathology: a systematic review and meta-analysis of diagnostic test accuracy. NPJ Digit Med 2024; 7:114. [PMID: 38704465 PMCID: PMC11069583 DOI: 10.1038/s41746-024-01106-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Accepted: 04/12/2024] [Indexed: 05/06/2024] Open
Abstract
Ensuring diagnostic performance of artificial intelligence (AI) before introduction into clinical practice is essential. Growing numbers of studies using AI for digital pathology have been reported over recent years. The aim of this work is to examine the diagnostic accuracy of AI in digital pathology images for any disease. This systematic review and meta-analysis included diagnostic accuracy studies using any type of AI applied to whole slide images (WSIs) for any disease. The reference standard was diagnosis by histopathological assessment and/or immunohistochemistry. Searches were conducted in PubMed, EMBASE and CENTRAL in June 2022. Risk of bias and concerns of applicability were assessed using the QUADAS-2 tool. Data extraction was conducted by two investigators and meta-analysis was performed using a bivariate random effects model, with additional subgroup analyses also performed. Of 2976 identified studies, 100 were included in the review and 48 in the meta-analysis. Studies were from a range of countries, including over 152,000 whole slide images (WSIs), representing many diseases. These studies reported a mean sensitivity of 96.3% (CI 94.1-97.7) and mean specificity of 93.3% (CI 90.5-95.4). There was heterogeneity in study design and 99% of studies identified for inclusion had at least one area at high or unclear risk of bias or applicability concerns. Details on selection of cases, division of model development and validation data and raw performance data were frequently ambiguous or missing. AI is reported as having high diagnostic accuracy in the reported areas but requires more rigorous evaluation of its performance.
Collapse
Affiliation(s)
- Clare McGenity
- University of Leeds, Leeds, UK.
- Leeds Teaching Hospitals NHS Trust, Leeds, UK.
| | - Emily L Clarke
- University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | - Charlotte Jennings
- University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
| | | | | | | | | | - Darren Treanor
- University of Leeds, Leeds, UK
- Leeds Teaching Hospitals NHS Trust, Leeds, UK
- Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
- Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| |
Collapse
|
53
|
Tian F, Liu D, Wei N, Fu Q, Sun L, Liu W, Sui X, Tian K, Nemeth G, Feng J, Xu J, Xiao L, Han J, Fu J, Shi Y, Yang Y, Liu J, Hu C, Feng B, Sun Y, Wang Y, Yu G, Kong D, Wang M, Li W, Chen K, Li X. Prediction of tumor origin in cancers of unknown primary origin with cytology-based deep learning. Nat Med 2024; 30:1309-1319. [PMID: 38627559 PMCID: PMC11108774 DOI: 10.1038/s41591-024-02915-w] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Accepted: 03/07/2024] [Indexed: 04/26/2024]
Abstract
Cancer of unknown primary (CUP) site poses diagnostic challenges due to its elusive nature. Many cases of CUP manifest as pleural and peritoneal serous effusions. Leveraging cytological images from 57,220 cases at four tertiary hospitals, we developed a deep-learning method for tumor origin differentiation using cytological histology (TORCH) that can identify malignancy and predict tumor origin in both hydrothorax and ascites. We examined its performance on three internal (n = 12,799) and two external (n = 14,538) testing sets. In both internal and external testing sets, TORCH achieved area under the receiver operating curve values ranging from 0.953 to 0.991 for cancer diagnosis and 0.953 to 0.979 for tumor origin localization. TORCH accurately predicted primary tumor origins, with a top-1 accuracy of 82.6% and top-3 accuracy of 98.9%. Compared with results derived from pathologists, TORCH showed better prediction efficacy (1.677 versus 1.265, P < 0.001), enhancing junior pathologists' diagnostic scores significantly (1.326 versus 1.101, P < 0.001). Patients with CUP whose initial treatment protocol was concordant with TORCH-predicted origins had better overall survival than those who were administrated discordant treatment (27 versus 17 months, P = 0.006). Our study underscores the potential of TORCH as a valuable ancillary tool in clinical practice, although further validation in randomized trials is warranted.
Collapse
Affiliation(s)
- Fei Tian
- Department of Abdominal Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Dong Liu
- Department of Radiology, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | - Na Wei
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Qianqian Fu
- Department of Pathology, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Lin Sun
- Department of Pathology, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Wei Liu
- Department of Pathology, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | - Xiaolong Sui
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Kathryn Tian
- Harvard Dunster House, Harvard University, Cambridge, MA, USA
| | | | - Jingyu Feng
- School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Jingjing Xu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Lin Xiao
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junya Han
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jingjie Fu
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yinhua Shi
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yichen Yang
- Tianjin Cancer Institute, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Jia Liu
- Department of Abdominal Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Chunhong Hu
- Department of Radiology, The First Affiliated Hospital of Suzhou University, Suzhou, China
| | - Bin Feng
- Department of Pathology, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yan Sun
- Department of Pathology, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Yunjun Wang
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Guohua Yu
- Department of Pathology, Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Dalu Kong
- Department of Abdominal Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China
| | - Meiyun Wang
- Department of Radiology, Henan Provincial People's Hospital, The People's Hospital of Zhengzhou University, Zhengzhou, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Kexin Chen
- Department of Epidemiology and Biostatistics, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Molecular Cancer Epidemiology of Tianjin, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| | - Xiangchun Li
- Tianjin Cancer Institute, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive Cancer, National Clinical Research Center for Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin Medical University, Tianjin, China.
| |
Collapse
|
54
|
Kazemi A, Rasouli-Saravani A, Gharib M, Albuquerque T, Eslami S, Schüffler PJ. A systematic review of machine learning-based tumor-infiltrating lymphocytes analysis in colorectal cancer: Overview of techniques, performance metrics, and clinical outcomes. Comput Biol Med 2024; 173:108306. [PMID: 38554659 DOI: 10.1016/j.compbiomed.2024.108306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2023] [Revised: 03/04/2024] [Accepted: 03/12/2024] [Indexed: 04/02/2024]
Abstract
The incidence of colorectal cancer (CRC), one of the deadliest cancers around the world, is increasing. Tissue microenvironment (TME) features such as tumor-infiltrating lymphocytes (TILs) can have a crucial impact on diagnosis or decision-making for treating patients with CRC. While clinical studies showed that TILs improve the host immune response, leading to a better prognosis, inter-observer agreement for quantifying TILs is not perfect. Incorporating machine learning (ML) based applications in clinical routine may promote diagnosis reliability. Recently, ML has shown potential for making progress in routine clinical procedures. We aim to systematically review the TILs analysis based on ML in CRC histological images. Deep learning (DL) and non-DL techniques can aid pathologists in identifying TILs, and automated TILs are associated with patient outcomes. However, a large multi-institutional CRC dataset with a diverse and multi-ethnic population is necessary to generalize ML methods.
Collapse
Affiliation(s)
- Azar Kazemi
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany.
| | - Ashkan Rasouli-Saravani
- Student Research Committee, Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Masoumeh Gharib
- Department of Pathology, Faculty of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran.
| | | | - Saeid Eslami
- Department of Medical Informatics, School of Medicine, Mashhad University of Medical Sciences, Mashhad, Iran; Pharmaceutical Sciences Research Center, Institute of Pharmaceutical Technology, Mashhad University of Medical Sciences, Mashhad, Iran; Department of Medical Informatics, University of Amsterdam, Amsterdam, the Netherlands.
| | - Peter J Schüffler
- Institute of General and Surgical Pathology, Technical University of Munich, Munich, Germany; TUM School of Computation, Information and Technology, Technical University of Munich, Munich, Germany; Munich Center for Machine Learning, Munich, Germany; Munich Data Science Institute, Munich, Germany.
| |
Collapse
|
55
|
Oloruntoba A, Ingvar Å, Sashindranath M, Anthony O, Abbott L, Guitera P, Caccetta T, Janda M, Soyer HP, Mar V. Examining labelling guidelines for AI-based software as a medical device: A review and analysis of dermatology mobile applications in Australia. Australas J Dermatol 2024. [PMID: 38693690 DOI: 10.1111/ajd.14269] [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: 11/21/2023] [Revised: 02/26/2024] [Accepted: 04/01/2024] [Indexed: 05/03/2024]
Abstract
In recent years, there has been a surge in the development of AI-based Software as a Medical Device (SaMD), particularly in visual specialties such as dermatology. In Australia, the Therapeutic Goods Administration (TGA) regulates AI-based SaMD to ensure its safe use. Proper labelling of these devices is crucial to ensure that healthcare professionals and the general public understand how to use them and interpret results accurately. However, guidelines for labelling AI-based SaMD in dermatology are lacking, which may result in products failing to provide essential information about algorithm development and performance metrics. This review examines existing labelling guidelines for AI-based SaMD across visual medical specialties, with a specific focus on dermatology. Common recommendations for labelling are identified and applied to currently available dermatology AI-based SaMD mobile applications to determine usage of these labels. Of the 21 AI-based SaMD mobile applications identified, none fully comply with common labelling recommendations. Results highlight the need for standardized labelling guidelines. Ensuring transparency and accessibility of information is essential for the safe integration of AI into health care and preventing potential risks associated with inaccurate clinical decisions.
Collapse
Affiliation(s)
| | - Åsa Ingvar
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Victorian Melanoma Service, Alfred Health, Melbourne, Victoria, Australia
- Department of Dermatology, Skåne University Hospital, Lund University, Lund, Sweden
- Department of Clinical Sciences, Skåne University Hospital, Lund University, Lund, Sweden
| | - Maithili Sashindranath
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
| | - Ojochonu Anthony
- Faculty of Medicine, Nursing and Health Sciences, Monash University, Melbourne, Victoria, Australia
| | - Lisa Abbott
- Melanoma Institute Australia, The University of Sydney, Sydney, New South Wales, Australia
| | - Pascale Guitera
- Faculty of Medicine and Health, The University of Sydney, Sydney, New South Wales, Australia
- Sydney Melanoma Diagnostic Centre, Royal Prince Alfred Hospital, Camperdown, New South Wales, Australia
- Perth Dermatology Clinic, Perth, Western Australia, Australia
| | - Tony Caccetta
- Perth Dermatology Clinic, Perth, Western Australia, Australia
| | - Monika Janda
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - H Peter Soyer
- Dermatology Research Centre, Frazer Institute, The University of Queensland, Brisbane, Queensland, Australia
| | - Victoria Mar
- School of Public Health and Preventive Medicine, Monash University, Melbourne, Victoria, Australia
- Victorian Melanoma Service, Alfred Health, Melbourne, Victoria, Australia
| |
Collapse
|
56
|
Hang JF, Ou YC, Yang WL, Tsao TY, Yeh CH, Li CB, Hsu EY, Hung PY, Lin MY, Hwang YT, Liu TJ, Tung MC. Evaluating Urine Cytology Slide Digitization Efficiency: A Comparative Study Using an Artificial Intelligence-Based Heuristic Scanning Simulation and Multiple Z-Plane Scanning. Acta Cytol 2024; 68:342-350. [PMID: 38648759 DOI: 10.1159/000538985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Accepted: 04/16/2024] [Indexed: 04/25/2024]
Abstract
INTRODUCTION Digitizing cytology slides presents challenges because of their three-dimensional features and uneven cell distribution. While multi-Z-plane scan is a prevalent solution, its adoption in clinical digital cytopathology is hindered by prolonged scanning times, increased image file sizes, and the requirement for cytopathologists to review multiple Z-plane images. METHODS This study presents heuristic scan as a novel solution, using an artificial intelligence (AI)-based approach specifically designed for cytology slide scanning as an alternative to the multi-Z-plane scan. Both the 21 Z-plane scan and the heuristic scan simulation methods were used on 52 urine cytology slides from three distinct cytopreparations (Cytospin, ThinPrep, and BD CytoRich™ [SurePath]), generating whole-slide images (WSIs) via the Leica Aperio AT2 digital scanner. The AI algorithm inferred the WSI from 21 Z-planes to quantitate the total number of suspicious for high-grade urothelial carcinoma or more severe cells (SHGUC+) cells. The heuristic scan simulation calculated the total number of SHGUC+ cells from the 21 Z-plane scan data. Performance metrics including SHGUC+ cell coverage rates (calculated by dividing the number of SHGUC+ cells identified in multiple Z-planes or heuristic scan simulation by the total SHGUC+ cells in the 21 Z-planes for each WSI), scanning time, and file size were analyzed to compare the performance of each scanning method. The heuristic scan's metrics were linearly estimated from the 21 Z-plane scan data. Additionally, AI-aided interpretations of WSIs with scant SHGUC+ cells followed The Paris System guidelines and were compared with original diagnoses. RESULTS The heuristic scan achieved median SHGUC+ cell coverage rates similar to 5 Z-plane scans across three cytopreparations (0.78-0.91 vs. 0.75-0.88, p = 0.451-0.578). Notably, it substantially reduced both scanning time (137.2-635.0 s vs. 332.6-1,278.8 s, p < 0.05) and image file size (0.51-2.10 GB vs. 1.16-3.10 GB, p < 0.05). Importantly, the heuristic scan yielded higher rates of accurate AI-aided interpretations compared to the single Z-plane scan (62.5% vs. 37.5%). CONCLUSION We demonstrated that the heuristic scan offers a cost-effective alternative to the conventional multi-Z-plane scan in digital cytopathology. It achieves comparable SHGUC+ cell capture rates while reducing both scanning time and image file size, promising to aid digital urine cytology interpretations with a higher accuracy rate compared to the conventional single (optimal) plane scan. Further studies are needed to assess the integration of this new technology into compatible digital scanners for practical cytology slide scanning.
Collapse
Affiliation(s)
- Jen-Fan Hang
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- School of Medicine and Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yen-Chuan Ou
- Division of Urology, Department of Surgery, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | | | - Tang-Yi Tsao
- Department of Pathology, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| | | | - Chi-Bin Li
- AIxMed, Inc., Santa Clara, California, USA
| | - En-Yu Hsu
- AIxMed, Inc., Santa Clara, California, USA
| | | | | | - Yi-Ting Hwang
- Department of Statistics, National Taipei University, Taipei, Taiwan
| | | | - Min-Che Tung
- Division of Urology, Department of Surgery, Tung's Taichung MetroHarbor Hospital, Taichung, Taiwan
| |
Collapse
|
57
|
Godson L, Alemi N, Nsengimana J, Cook GP, Clarke EL, Treanor D, Bishop DT, Newton-Bishop J, Gooya A, Magee D. Immune subtyping of melanoma whole slide images using multiple instance learning. Med Image Anal 2024; 93:103097. [PMID: 38325154 DOI: 10.1016/j.media.2024.103097] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2022] [Revised: 01/15/2024] [Accepted: 01/25/2024] [Indexed: 02/09/2024]
Abstract
Determining early-stage prognostic markers and stratifying patients for effective treatment are two key challenges for improving outcomes for melanoma patients. Previous studies have used tumour transcriptome data to stratify patients into immune subgroups, which were associated with differential melanoma specific survival and potential predictive biomarkers. However, acquiring transcriptome data is a time-consuming and costly process. Moreover, it is not routinely used in the current clinical workflow. Here, we attempt to overcome this by developing deep learning models to classify gigapixel haematoxylin and eosin (H&E) stained pathology slides, which are well established in clinical workflows, into these immune subgroups. We systematically assess six different multiple instance learning (MIL) frameworks, using five different image resolutions and three different feature extraction methods. We show that pathology-specific self-supervised models using 10x resolution patches generate superior representations for the classification of immune subtypes. In addition, in a primary melanoma dataset, we achieve a mean area under the receiver operating characteristic curve (AUC) of 0.80 for classifying histopathology images into 'high' or 'low immune' subgroups and a mean AUC of 0.82 in an independent TCGA melanoma dataset. Furthermore, we show that these models are able to stratify patients into 'high' and 'low immune' subgroups with significantly different melanoma specific survival outcomes (log rank test, P< 0.005). We anticipate that MIL methods will allow us to find new biomarkers of high importance, act as a tool for clinicians to infer the immune landscape of tumours and stratify patients, without needing to carry out additional expensive genetic tests.
Collapse
Affiliation(s)
- Lucy Godson
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom.
| | - Navid Alemi
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom
| | - Jérémie Nsengimana
- Population Health Sciences Institute, Newcastle University, Newcastle upon Tyne, NE1 7RU, United Kingdom
| | - Graham P Cook
- Leeds Institute of Medical Research, University of Leeds School of Medicine, St. James's University Hospital, Leeds, United Kingdom
| | - Emily L Clarke
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - Darren Treanor
- Department of Histopathology, Leeds Teaching Hospitals NHS Trust, Leeds, United Kingdom; Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom; Department of Clinical Pathology and Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Centre for Medical Image Science and Visualization (CMIV), Linköping University, Linköping, Sweden
| | - D Timothy Bishop
- Leeds Institute of Medical Research, University of Leeds School of Medicine, St. James's University Hospital, Leeds, United Kingdom
| | - Julia Newton-Bishop
- Division of Pathology and Data Analytics, Leeds Institute of Cancer and Pathology, University of Leeds, Beckett Street, Leeds, LS9 7TF, United Kingdom
| | - Ali Gooya
- School of Computing, University of Glasgow, Glasgow, G12 8QQ, United Kingdom
| | - Derek Magee
- School of Computing, University of Leeds, Woodhouse, Leeds, LS2 9JT, United Kingdom
| |
Collapse
|
58
|
Mhaske S, Ramalingam K, Nair P, Patel S, Menon P A, Malik N, Mhaske S. Automated Analysis of Nuclear Parameters in Oral Exfoliative Cytology Using Machine Learning. Cureus 2024; 16:e58744. [PMID: 38779230 PMCID: PMC11110917 DOI: 10.7759/cureus.58744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/22/2024] [Indexed: 05/25/2024] Open
Abstract
BACKGROUND As oral cancer remains a major worldwide health concern, sophisticated diagnostic tools are needed to aid in early diagnosis. Non-invasive methods like exfoliative cytology, albeit with the help of artificial intelligence (AI), have drawn additional interest. AIM The study aimed to harness the power of machine learning algorithms for the automated analysis of nuclear parameters in oral exfoliative cytology. Further, the analysis of two different AI systems, namely convoluted neural networks (CNN) and support vector machine (SVM), were compared for accuracy. METHODS A comparative diagnostic study was performed in two groups of patients (n=60). The control group without evidence of lesions (n=30) and the other group with clinically suspicious oral malignancy (n=30) were evaluated. All patients underwent cytological smears using an exfoliative cytology brush, followed by routine Hematoxylin and Eosin staining. Image preprocessing, data splitting, machine learning, model development, feature extraction, and model evaluation were done. An independent t-test was run on each nuclear characteristic, and Pearson's correlation coefficient test was performed with Statistical Package for the Social Sciences (SPSS) software (IBM SPSS Statistics for Windows, Version 28.0. IBM Corp, Armonk, NY, USA). RESULTS The study found substantial variations between the study and control groups in nuclear size (p<0.05), nuclear shape (p<0.01), and chromatin distribution (p<0.001). The Pearson correlation coefficient of SVM was 0.6472, and CNN was 0.7790, showing that SVM had more accuracy. CONCLUSION The availability of multidimensional datasets, combined with breakthroughs in high-performance computers and new deep-learning architectures, has resulted in an explosion of AI use in numerous areas of oncology research. The discerned diagnostic accuracy exhibited by the SVM and CNN models suggests prospective improvements in early detection rates, potentially improving patient outcomes and enhancing healthcare practices.
Collapse
Affiliation(s)
- Shubhangi Mhaske
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
- Oral and Maxillofacial Pathology, People's College Of Dental Science and Research Center, Bhopal, IND
| | - Karthikeyan Ramalingam
- Oral Pathology and Microbiology, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, IND
| | - Preeti Nair
- Oral Medicine and Radiology, People's College Of Dental Science and Research Center, Bhopal, IND
| | - Shubham Patel
- Oral and Maxillofacial Pathology, People's College Of Dental Science and Research Center, Bhopal, IND
| | - Arathi Menon P
- Dentistry, Indian Council of Medical Research, Bhopal, IND
| | - Nida Malik
- Periodontics, Kamala Nehru Hospital, Bhopal, IND
| | - Sumedh Mhaske
- Medicine, Government Medical College & Hospital, Aurangabad, IND
| |
Collapse
|
59
|
Wang C, Wang X, Gao Z, Ran C, Li C, Ding C. Multiple serous cavity effusion screening based on smear images using vision transformer. Sci Rep 2024; 14:7395. [PMID: 38548898 PMCID: PMC10978834 DOI: 10.1038/s41598-024-58151-2] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Accepted: 03/26/2024] [Indexed: 04/01/2024] Open
Abstract
Serous cavity effusion is a prevalent pathological condition encountered in clinical settings. Fluid samples obtained from these effusions are vital for diagnostic and therapeutic purposes. Traditionally, cytological examination of smears is a common method for diagnosing serous cavity effusion, renowned for its convenience. However, this technique presents limitations that can compromise its efficiency and diagnostic accuracy. This study aims to overcome these challenges and introduce an improved method for the precise detection of malignant cells in serous cavity effusions. We have developed a transformer-based classification framework, specifically employing the vision transformer (ViT) model, to fulfill this objective. Our research involved collecting smear images and corresponding cytological reports from 161 patients who underwent serous cavity drainage. We meticulously annotated 4836 patches from these images, identifying regions with and without malignant cells, thus creating a unique dataset for smear image classification. The findings of our study reveal that deep learning models, particularly the ViT model, exhibit remarkable accuracy in classifying patches as malignant or non-malignant. The ViT model achieved an impressive area under the receiver operating characteristic curve (AUROC) of 0.99, surpassing the performance of the convolutional neural network (CNN) model, which recorded an AUROC of 0.86. Additionally, we validated our models using an external cohort of 127 patients. The ViT model sustained its high-level screening performance, achieving an AUROC of 0.98 at the patient level, compared to the CNN model's AUROC of 0.84. The visualization of our ViT models confirmed their capability to precisely identify regions containing malignant cells in multiple serous cavity effusion smear images. In summary, our study demonstrates the potential of deep learning models, particularly the ViT model, in automating the screening process for serous cavity effusions. These models offer significant assistance to cytologists in enhancing diagnostic accuracy and efficiency. The ViT model stands out for its advanced self-attention mechanism, making it exceptionally suitable for tasks that necessitate detailed analysis of small, sparsely distributed targets like cellular clusters in serous cavity effusions.
Collapse
Affiliation(s)
- Chunbao Wang
- Department of Pathology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Xiangyu Wang
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Zeyu Gao
- CRUK Cambridge Centre, University of Cambridge, Cambridge, CB2 0RE, UK
| | - Caihong Ran
- Department of Pathology, Ngari Prefecture People's Hospital, Ngari of Tibet, 859000, China
| | - Chen Li
- School of Computer Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China.
| | - Caixia Ding
- Department of Pathology, Shaanxi Provincial Tumor Hospital, Xi'an, 710061, China.
| |
Collapse
|
60
|
Sweed D, Elhamed SMA, Aiad HAS, Ehsan NA, Hemida AS, Dawoud MM. STIM1/SOX2 proteins are co-expressed in the tumor and microenvironmental stromal cells of pancreatic ductal adenocarcinoma and ampullary carcinoma. World J Surg Oncol 2024; 22:84. [PMID: 38532463 DOI: 10.1186/s12957-024-03356-y] [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: 01/10/2024] [Accepted: 03/08/2024] [Indexed: 03/28/2024] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) and ampullary carcinoma (AAC) are lethal malignancies with modest benefits from surgery. SOX2 and STIM1 have been linked to anticancer activity in several human malignancies. This study included 94 tumor cases: 48 primary PDAC, 25 metastatic PDAC, and 21 primary AAC with corresponding non-tumor tissue. All cases were immunohistochemically stained for STIM1 and SOX2 and results were correlated with clinicopathologic data, patient survival, and BCL2 immunostaining results. Results revealed that STIM1 and SOX2 epithelial/stromal expressions were significantly higher in PDAC and AAC in comparison to the control groups. STIM1 and SOX2 expressions were positively correlated in the primary and metastatic PDAC (P = 0.016 and, P = 0.001, respectively). However, their expressions were not significantly associated with BCL2 expression. SOX2 epithelial/stromal expressions were positively correlated with the large tumor size in the primary AAC group (P = 0.052, P = 0.044, respectively). STIM1 stromal and SOX2 epithelial over-expressions had a bad prognostic impact on the overall survival of AAC (P = 0.002 and P = 0.001, respectively). Therefore, STIM1 and SOX2 co-expression in tumor cells and intra-tumoral stroma could contribute to the development of PDAC and AAC. STIM1/SOX2 expression is linked to a bad prognosis in AAC.
Collapse
Affiliation(s)
- Dina Sweed
- Pathology Department, National Liver Institute, Menoufia University, Shibin Al Koom, Egypt
| | | | - Hayam Abdel Samie Aiad
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt
| | - Nermine Ahmed Ehsan
- Pathology Department, National Liver Institute, Menoufia University, Shibin Al Koom, Egypt
| | - Aiat Shaban Hemida
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt
| | - Marwa Mohammed Dawoud
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin Al Koom, 32511, Egypt.
| |
Collapse
|
61
|
Kim HK, Han E, Lee J, Yim K, Abdul-Ghafar J, Seo KJ, Seo JW, Gong G, Cho NH, Kim M, Yoo CW, Chong Y. Artificial-Intelligence-Assisted Detection of Metastatic Colorectal Cancer Cells in Ascitic Fluid. Cancers (Basel) 2024; 16:1064. [PMID: 38473421 DOI: 10.3390/cancers16051064] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Revised: 02/17/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024] Open
Abstract
Ascites cytology is a cost-effective test for metastatic colorectal cancer (CRC) in the abdominal cavity. However, metastatic carcinoma of the peritoneum is difficult to diagnose based on biopsy findings, and ascitic aspiration cytology has a low sensitivity and specificity and a high inter-observer variability. The aim of the present study was to apply artificial intelligence (AI) to classify benign and malignant cells in ascites cytology patch images of metastatic CRC using a deep convolutional neural network. Datasets were collected from The OPEN AI Dataset Project, a nationwide cytology dataset for AI research. The numbers of patch images used for training, validation, and testing were 56,560, 7068, and 6534, respectively. We evaluated 1041 patch images of benign and metastatic CRC in the ascitic fluid to compare the performance of pathologists and an AI algorithm, and to examine whether the diagnostic accuracy of pathologists improved with the assistance of AI. This AI method showed an accuracy, a sensitivity, and a specificity of 93.74%, 87.76%, and 99.75%, respectively, for the differential diagnosis of malignant and benign ascites. The diagnostic accuracy and sensitivity of the pathologist with the assistance of the proposed AI method increased from 86.8% to 90.5% and from 73.3% to 79.3%, respectively. The proposed deep learning method may assist pathologists with different levels of experience in diagnosing metastatic CRC cells of ascites.
Collapse
Affiliation(s)
- Hyung Kyung Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
- Department of Pathology, Samsung Medical Center, Seoul 06351, Republic of Korea
| | - Eunkyung Han
- Department of Pathology, Soonchunyang University Hospital Bucheon, Bucheon 14584, Republic of Korea
| | - Jeonghyo Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam 13620, Republic of Korea
| | - Kwangil Yim
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Kyung Jin Seo
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Jang Won Seo
- AI Team, MTS Company Inc., Seoul 06178, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Milim Kim
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer Center, Goyang 10408, Republic of Korea
| | - Yosep Chong
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| |
Collapse
|
62
|
Neto PC, Montezuma D, Oliveira SP, Oliveira D, Fraga J, Monteiro A, Monteiro J, Ribeiro L, Gonçalves S, Reinhard S, Zlobec I, Pinto IM, Cardoso JS. An interpretable machine learning system for colorectal cancer diagnosis from pathology slides. NPJ Precis Oncol 2024; 8:56. [PMID: 38443695 PMCID: PMC10914836 DOI: 10.1038/s41698-024-00539-4] [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: 07/18/2023] [Accepted: 02/08/2024] [Indexed: 03/07/2024] Open
Abstract
Considering the profound transformation affecting pathology practice, we aimed to develop a scalable artificial intelligence (AI) system to diagnose colorectal cancer from whole-slide images (WSI). For this, we propose a deep learning (DL) system that learns from weak labels, a sampling strategy that reduces the number of training samples by a factor of six without compromising performance, an approach to leverage a small subset of fully annotated samples, and a prototype with explainable predictions, active learning features and parallelisation. Noting some problems in the literature, this study is conducted with one of the largest WSI colorectal samples dataset with approximately 10,500 WSIs. Of these samples, 900 are testing samples. Furthermore, the robustness of the proposed method is assessed with two additional external datasets (TCGA and PAIP) and a dataset of samples collected directly from the proposed prototype. Our proposed method predicts, for the patch-based tiles, a class based on the severity of the dysplasia and uses that information to classify the whole slide. It is trained with an interpretable mixed-supervision scheme to leverage the domain knowledge introduced by pathologists through spatial annotations. The mixed-supervision scheme allowed for an intelligent sampling strategy effectively evaluated in several different scenarios without compromising the performance. On the internal dataset, the method shows an accuracy of 93.44% and a sensitivity between positive (low-grade and high-grade dysplasia) and non-neoplastic samples of 0.996. On the external test samples varied with TCGA being the most challenging dataset with an overall accuracy of 84.91% and a sensitivity of 0.996.
Collapse
Affiliation(s)
- Pedro C Neto
- Institute for Systems and Computer Engineering, Technology and Science (INESC TEC), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal.
- Faculty of Engineering, University of Porto (FEUP), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal.
| | - Diana Montezuma
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal.
- Cancer Biology and Epigenetics Group, Research Center of IPO Porto (CI-IPOP) / RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto) / Porto Comprehensive Cancer Center (Porto.CCC), R. Dr. António Bernardino de Almeida 865, Porto, 4200-072, Porto, Portugal.
- Doctoral Programme in Medical Sciences, School of Medicine and Biomedical Sciences - University of Porto (ICBAS-UP), R. Jorge de Viterbo Ferreira 228, Porto, 4050-313, Porto, Portugal.
| | - Sara P Oliveira
- Institute for Systems and Computer Engineering, Technology and Science (INESC TEC), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal.
- Faculty of Engineering, University of Porto (FEUP), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal.
| | - Domingos Oliveira
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - João Fraga
- Department of Pathology, IPO-Porto, R. Dr. António Bernardino de Almeida 865, Porto, 4200-072, Porto, Portugal
| | - Ana Monteiro
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - João Monteiro
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - Liliana Ribeiro
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - Sofia Gonçalves
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - Stefan Reinhard
- Institute of Pathology, University of Bern, Uni Bern, Murtenstrasse 31, Bern, 3008, Bern, Switzerland
| | - Inti Zlobec
- Institute of Pathology, University of Bern, Uni Bern, Murtenstrasse 31, Bern, 3008, Bern, Switzerland
| | - Isabel M Pinto
- IMP Diagnostics, Praça do Bom Sucesso, 61, sala 808, Porto, 4150-146, Porto, Portugal
| | - Jaime S Cardoso
- Institute for Systems and Computer Engineering, Technology and Science (INESC TEC), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal
- Faculty of Engineering, University of Porto (FEUP), R. Dr. Roberto Frias, Porto, 4200-465, Porto, Portugal
| |
Collapse
|
63
|
Yuh WT, Khil EK, Yoon YS, Kim B, Yoon H, Lim J, Lee KY, Yoo YS, An KD. Deep Learning-Assisted Quantitative Measurement of Thoracolumbar Fracture Features on Lateral Radiographs. Neurospine 2024; 21:30-43. [PMID: 38569629 PMCID: PMC10992637 DOI: 10.14245/ns.2347366.683] [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: 12/24/2023] [Revised: 01/24/2024] [Accepted: 02/02/2024] [Indexed: 04/05/2024] Open
Abstract
OBJECTIVE This study aimed to develop and validate a deep learning (DL) algorithm for the quantitative measurement of thoracolumbar (TL) fracture features, and to evaluate its efficacy across varying levels of clinical expertise. METHODS Using the pretrained Mask Region-Based Convolutional Neural Networks model, originally developed for vertebral body segmentation and fracture detection, we fine-tuned the model and added a new module for measuring fracture metrics-compression rate (CR), Cobb angle (CA), Gardner angle (GA), and sagittal index (SI)-from lumbar spine lateral radiographs. These metrics were derived from six-point labeling by 3 radiologists, forming the ground truth (GT). Training utilized 1,000 nonfractured and 318 fractured radiographs, while validations employed 213 internal and 200 external fractured radiographs. The accuracy of the DL algorithm in quantifying fracture features was evaluated against GT using the intraclass correlation coefficient. Additionally, 4 readers with varying expertise levels, including trainees and an attending spine surgeon, performed measurements with and without DL assistance, and their results were compared to GT and the DL model. RESULTS The DL algorithm demonstrated good to excellent agreement with GT for CR, CA, GA, and SI in both internal (0.860, 0.944, 0.932, and 0.779, respectively) and external (0.836, 0.940, 0.916, and 0.815, respectively) validations. DL-assisted measurements significantly improved most measurement values, particularly for trainees. CONCLUSION The DL algorithm was validated as an accurate tool for quantifying TL fracture features using radiographs. DL-assisted measurement is expected to expedite the diagnostic process and enhance reliability, particularly benefiting less experienced clinicians.
Collapse
Affiliation(s)
- Woon Tak Yuh
- Department of Neurosurgery, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Eun Kyung Khil
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
- Department of Radiology, Fastbone Orthopedic Hospital, Hwaseong, Korea
| | - Yu Sung Yoon
- Department of Radiology, Kyungpook National University Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | | | | | - Jihe Lim
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Kyoung Yeon Lee
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Yeong Seo Yoo
- Department of Radiology, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| | - Kyeong Deuk An
- Department of Neurosurgery, Hallym University Dongtan Sacred Heart Hospital, Hwaseong, Korea
| |
Collapse
|
64
|
Elkady N, Allam DM. The Role of Galectin3, Tubulinβ, and Maspin in Promoting Tumor Budding in Colorectal Carcinoma and Their Clinical Implications. Appl Immunohistochem Mol Morphol 2024; 32:143-150. [PMID: 38251657 DOI: 10.1097/pai.0000000000001183] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2023] [Accepted: 12/27/2023] [Indexed: 01/23/2024]
Abstract
Colorectal cancer (CRC) is a leading cause of death worldwide. Despite the advances in surgical and therapeutic management, tumor metastases and poor prognosis are still major problems. Tumor budding is a relevant prognostic factor in CRC, and it can predict tumor metastasis. Galectin3 is responsible for the development and progression of many cancers through the regulation of cell-cell/cell-matrix interactions and tumor cell invasion. Tubulin is a microtubule protein, and maspin is a serine protease inhibitor; both induce tumor cell invasion through the stimulation of epithelial-mesenchymal transition. This study aims to evaluate the relationship between the expression of galecin3, tubulinβ, and maspin in CRC and clinicopathological features, including tumor budding, their prognostic roles, and clinical implications using immunohistochemistry. Galectin3, tubulinβ, and maspin were detected in tumor cells in 95%, 65%, and 87.5% of cases and in stromal cells in 28.8%, 40%, and 0% of cases. High expression of galectin3 and tubulinβ expression either in tumor cells or stroma was significantly associated with aggressive tumor features such as lymph node metastasis, lymphovascular invasion, tumor budding, and advanced tumor stage. The nucleocytoplasmic expression of maspin in tumor cells showed a significant association with deeper tumor invasion, lymph node metastasis, tumor budding, and advanced tumor stage. Significant associations were found between high galectin3 tumor cell expression and nucleocytoplasmic maspin and shorter survival. High expression of galectin3, tubulinβ, and nucleocytoplasmic maspin were significantly associated with aggressive tumor features such as tumor invasion, metastasis, high tumor budding, and short survival in CRC. They could be used as biomarkers for tumor budding and tumor aggressiveness in CRC and may be considered for future target therapy.
Collapse
Affiliation(s)
- Noha Elkady
- Pathology Department, Faculty of Medicine, Menoufia University, Shibin El Kom, Egypt
| | | |
Collapse
|
65
|
Grignaffini F, Barbuto F, Troiano M, Piazzo L, Simeoni P, Mangini F, De Stefanis C, Onetti Muda A, Frezza F, Alisi A. The Use of Artificial Intelligence in the Liver Histopathology Field: A Systematic Review. Diagnostics (Basel) 2024; 14:388. [PMID: 38396427 PMCID: PMC10887838 DOI: 10.3390/diagnostics14040388] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 02/02/2024] [Accepted: 02/06/2024] [Indexed: 02/25/2024] Open
Abstract
Digital pathology (DP) has begun to play a key role in the evaluation of liver specimens. Recent studies have shown that a workflow that combines DP and artificial intelligence (AI) applied to histopathology has potential value in supporting the diagnosis, treatment evaluation, and prognosis prediction of liver diseases. Here, we provide a systematic review of the use of this workflow in the field of hepatology. Based on the PRISMA 2020 criteria, a search of the PubMed, SCOPUS, and Embase electronic databases was conducted, applying inclusion/exclusion filters. The articles were evaluated by two independent reviewers, who extracted the specifications and objectives of each study, the AI tools used, and the results obtained. From the 266 initial records identified, 25 eligible studies were selected, mainly conducted on human liver tissues. Most of the studies were performed using whole-slide imaging systems for imaging acquisition and applying different machine learning and deep learning methods for image pre-processing, segmentation, feature extractions, and classification. Of note, most of the studies selected demonstrated good performance as classifiers of liver histological images compared to pathologist annotations. Promising results to date bode well for the not-too-distant inclusion of these techniques in clinical practice.
Collapse
Affiliation(s)
- Flavia Grignaffini
- Department of Information Engineering, Electronics and Telecommunications (DIET), “La Sapienza”, University of Rome, 00184 Rome, Italy; (F.G.); (F.B.); (L.P.); (F.M.); (F.F.)
| | - Francesco Barbuto
- Department of Information Engineering, Electronics and Telecommunications (DIET), “La Sapienza”, University of Rome, 00184 Rome, Italy; (F.G.); (F.B.); (L.P.); (F.M.); (F.F.)
| | - Maurizio Troiano
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.T.); (C.D.S.)
| | - Lorenzo Piazzo
- Department of Information Engineering, Electronics and Telecommunications (DIET), “La Sapienza”, University of Rome, 00184 Rome, Italy; (F.G.); (F.B.); (L.P.); (F.M.); (F.F.)
| | - Patrizio Simeoni
- National Transport Authority (NTA), D02 WT20 Dublin, Ireland;
- Faculty of Lifelong Learning, South East Technological University (SETU), R93 V960 Carlow, Ireland
| | - Fabio Mangini
- Department of Information Engineering, Electronics and Telecommunications (DIET), “La Sapienza”, University of Rome, 00184 Rome, Italy; (F.G.); (F.B.); (L.P.); (F.M.); (F.F.)
| | - Cristiano De Stefanis
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.T.); (C.D.S.)
| | | | - Fabrizio Frezza
- Department of Information Engineering, Electronics and Telecommunications (DIET), “La Sapienza”, University of Rome, 00184 Rome, Italy; (F.G.); (F.B.); (L.P.); (F.M.); (F.F.)
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children’s Hospital, IRCCS, 00165 Rome, Italy; (M.T.); (C.D.S.)
| |
Collapse
|
66
|
Sabbaghian M, Gheitasi H, Shekarchi AA, Tavakoli A, Poortahmasebi V. The mysterious anelloviruses: investigating its role in human diseases. BMC Microbiol 2024; 24:40. [PMID: 38281930 PMCID: PMC10823751 DOI: 10.1186/s12866-024-03187-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 01/08/2024] [Indexed: 01/30/2024] Open
Abstract
Anelloviruses (AVs) that infect the human population are members of the Anelloviridae family. They are widely distributed in human populations worldwide. Torque teno virus (TTV) was the first virus of this family to be identified and is estimated to be found in the serum of 80-90% of the human population. Sometime after the identification of TTV, Torque teno mini virus (TTMV) and Torque teno midi virus (TTMDV) were also identified and classified in this family. Since identifying these viruses, have been detected in various types of biological fluids of the human body, including blood and urine, as well as vital organs such as the liver and kidney. They can be transmitted from person to person through blood transfusions, fecal-oral contact, and possibly sexual intercourse. Recent studies on these newly introduced viruses show that although they are not directly related to human disease, they may be indirectly involved in initiating or exacerbating some human population-related diseases and viral infections. Among these diseases, we can mention various types of cancers, immune system diseases, viral infections, hepatitis, and AIDS. Also, they likely use the microRNAs (miRNAs) they encode to fulfill this cooperative role. Also, in recent years, the role of proliferation and their viral load, especially TTV, has been highlighted to indicate the immune system status of immunocompromised people or people who undergo organ transplants. Here, we review the possible role of these viruses in diseases that target humans and highlight them as important viruses that require further study. This review can provide new insights to researchers.
Collapse
Affiliation(s)
- Mohammad Sabbaghian
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hamidreza Gheitasi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ali Akbar Shekarchi
- Department of Pathology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Ahmad Tavakoli
- Research Center of Pediatric Infectious Diseases, Institute of Immunology and Infectious Diseases, Iran University of Medical Sciences, Tehran, Iran
| | - Vahdat Poortahmasebi
- Department of Bacteriology and Virology, Faculty of Medicine, Tabriz University of Medical Sciences, Tabriz, Iran.
- Research Center for Clinical Virology, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
67
|
Yu JL, Li Z, Zhang B, Huang YN, Zhao TY. Case report: Kikuchi-Fujimoto disease: unveiling a case of recurrent fever and enlarged cervical lymph nodes in a young female patient with a literature review of the immune mechanism. Front Immunol 2024; 14:1279592. [PMID: 38313434 PMCID: PMC10837848 DOI: 10.3389/fimmu.2023.1279592] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 12/27/2023] [Indexed: 02/06/2024] Open
Abstract
The inflammatory response to viral infection is an important component of the antiviral response, a process that involves the activation and proliferation of CD8+ T, CD4+ T, and dendritic cells; thus, viral infection disrupts the immune homeostasis of the organism, leading to an increased release of inflammatory factors. Kikuchi-Fujimoto disease (KFD) is an inflammatory self-limited disorder of unknown etiology, and it is generally believed that the pathogenesis of this disease includes two aspects: viral infection and autoimmune response. Various immune cells, such as CD8+ T lymphocytes, CD4+ T lymphocytes, and CD123+ plasmacytoid dendritic cells, as well as the cytokines they induce and secrete, such as interferons, interleukins, and tumor necrosis factors, play a crucial role in the pathogenesis of KFD. In this article, we present a case study of a young female patient from China who exhibited typical symptoms of lymph node inflammation and fever. The diagnosis of KFD was confirmed through a lymph node biopsy. She presented with elevated ESR, IL-6, and IFN-γ. Viral markers showed elevated IgG and IgM of cytomegalovirus (CMV) and elevated IgG of Epstein-Barr virus (EBV), while changes occurred in the CD4+ T and CD8+ T cell counts. Eventually, the patient achieved disease relief through steroid treatment. Based on these findings, we conducted a comprehensive review of the involvement of viral infection-induced inflammatory response processes and autoimmunity in the pathogenesis of Kikuchi-Fujimoto disease.
Collapse
Affiliation(s)
- Jia-Li Yu
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhen Li
- Department of Gastroenterology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Bo Zhang
- Department of Pathology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Ya-Nan Huang
- Department of Infectious Disease, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Tian-Yu Zhao
- Department of Infectious Disease, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| |
Collapse
|
68
|
Pateras IS, Igea A, Nikas IP, Leventakou D, Koufopoulos NI, Ieronimaki AI, Bergonzini A, Ryu HS, Chatzigeorgiou A, Frisan T, Kittas C, Panayiotides IG. Diagnostic Challenges during Inflammation and Cancer: Current Biomarkers and Future Perspectives in Navigating through the Minefield of Reactive versus Dysplastic and Cancerous Lesions in the Digestive System. Int J Mol Sci 2024; 25:1251. [PMID: 38279253 PMCID: PMC10816510 DOI: 10.3390/ijms25021251] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 01/12/2024] [Accepted: 01/16/2024] [Indexed: 01/28/2024] Open
Abstract
In the setting of pronounced inflammation, changes in the epithelium may overlap with neoplasia, often rendering it impossible to establish a diagnosis with certainty in daily clinical practice. Here, we discuss the underlying molecular mechanisms driving tissue response during persistent inflammatory signaling along with the potential association with cancer in the gastrointestinal tract, pancreas, extrahepatic bile ducts, and liver. We highlight the histopathological challenges encountered in the diagnosis of chronic inflammation in routine practice and pinpoint tissue-based biomarkers that could complement morphology to differentiate reactive from dysplastic or cancerous lesions. We refer to the advantages and limitations of existing biomarkers employing immunohistochemistry and point to promising new markers, including the generation of novel antibodies targeting mutant proteins, miRNAs, and array assays. Advancements in experimental models, including mouse and 3D models, have improved our understanding of tissue response. The integration of digital pathology along with artificial intelligence may also complement routine visual inspections. Navigating through tissue responses in various chronic inflammatory contexts will help us develop novel and reliable biomarkers that will improve diagnostic decisions and ultimately patient treatment.
Collapse
Affiliation(s)
- Ioannis S. Pateras
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Ana Igea
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), 15706 Santiago de Compostela, Spain;
- Mobile Genomes, Centre for Research in Molecular Medicine and Chronic Diseases (CiMUS), University of Santiago de Compostela (USC), 15706 Santiago de Compostela, Spain
| | - Ilias P. Nikas
- Medical School, University of Cyprus, 2029 Nicosia, Cyprus
| | - Danai Leventakou
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Nektarios I. Koufopoulos
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Argyro Ioanna Ieronimaki
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| | - Anna Bergonzini
- Center for Infectious Medicine, Department of Medicine Huddinge, Karolinska Institutet, Karolinska University Hospital, Alfred Nobels Allé 8, 141 52 Stockholm, Sweden;
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden;
| | - Han Suk Ryu
- Department of Pathology, Seoul National University Hospital, Seoul 03080, Republic of Korea;
| | - Antonios Chatzigeorgiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, 115 27 Athens, Greece;
| | - Teresa Frisan
- Department of Molecular Biology and Umeå Centre for Microbial Research (UCMR), Umeå University, 901 87 Umeå, Sweden;
| | - Christos Kittas
- Department of Histopathology, Biomedicine Group of Health Company, 156 26 Athens, Greece;
| | - Ioannis G. Panayiotides
- 2nd Department of Pathology, “Attikon” University Hospital, Medical School, National and Kapodistrian University of Athens, 124 62 Athens, Greece; (D.L.); (N.I.K.); (A.I.I.); (I.G.P.)
| |
Collapse
|
69
|
Lee TH, Yoon K, Lee S, Choi WR, Kim KG. Comparative Sensing and Judgment Control System for Temperature Maintenance for Optimal Treatment in Hyperthermic Intraperitoneal Chemotherapy Surgery. SENSORS (BASEL, SWITZERLAND) 2024; 24:596. [PMID: 38257692 PMCID: PMC10821041 DOI: 10.3390/s24020596] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 01/05/2024] [Accepted: 01/11/2024] [Indexed: 01/24/2024]
Abstract
For tumors wherein cancer cells remain in the tissue after colorectal cancer surgery, a hyperthermic anticancer agent is injected into the abdominal cavity to necrotize the remaining cancer cells with heat using a hyperthermic intraperitoneal chemotherapy system. However, during circulation, the processing temperature is out of range and the processing result is deteriorated. This paper proposes a look-up table (LUT) module design method that can stably maintain the processing temperature range during circulation via feedback. If the temperature decreases or increases, the LUT transmits a command signal to the heat exchanger to reduce or increase heat input, thereby maintaining the treatment temperature range. The command signal for increasing and decreasing heat input is Tp and Ta, respectively. The command signal for the treatment temperature range is Ts. If drug temperatures below 41 and above 43 °C are input to the LUT, it sends a Tp or Ta signal to the heat exchanger to increase or decrease the input heat, respectively. If the drug's temperature is 41-43 °C, the LUT generates a Ts signal and proceeds with the treatment. The proposed system can automatically control drug temperature using temperature feedback to ensure rapid, accurate, and safe treatment.
Collapse
Affiliation(s)
- Tae-Hyeon Lee
- Department of Electronic Engineering, Gyeonggi University of Science and Technology, 269 Gyeonggigwagi–dearo, Gyeonggi–do, Siheung City 15073, Republic of Korea;
| | - Kicheol Yoon
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 beon–gil, Namdong–daero Namdong–gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.); (W.R.C.)
- Department of Premedicine Course, College of Medicine, Gachon University, 38–13, 3 Dokjom–ro, Namdong–gu, Incheon 21565, Republic of Korea
| | - Sangyun Lee
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 beon–gil, Namdong–daero Namdong–gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.); (W.R.C.)
- Department of Health and Safety Convergence Sciences & Health and Environmental Convergence Sciences, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
- Department of Biomedical Engineering, Gachon University, 38–13, 3 Dokjom–ro, Namdong–gu, Incheon 21565, Republic of Korea
| | - Woong Rak Choi
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 beon–gil, Namdong–daero Namdong–gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.); (W.R.C.)
- Department of Biomedical Engineering, Gachon University, 38–13, 3 Dokjom–ro, Namdong–gu, Incheon 21565, Republic of Korea
- School of Electrical Engineering, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul 02841, Republic of Korea
| | - Kwang Gi Kim
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 beon–gil, Namdong–daero Namdong–gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.); (W.R.C.)
- Department of Biomedical Engineering, Gachon University, 38–13, 3 Dokjom–ro, Namdong–gu, Incheon 21565, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, 38–13, 3 Dokjom–ro, Namdong–gu, Incheon 21565, Republic of Korea
| |
Collapse
|
70
|
Addante F, d’Amati A, Santoro A, Angelico G, Inzani F, Arciuolo D, Travaglino A, Raffone A, D’Alessandris N, Scaglione G, Valente M, Tinnirello G, Sfregola S, Padial Urtueta B, Piermattei A, Cianfrini F, Mulè A, Bragantini E, Zannoni GF. Mismatch Repair Deficiency as a Predictive and Prognostic Biomarker in Endometrial Cancer: A Review on Immunohistochemistry Staining Patterns and Clinical Implications. Int J Mol Sci 2024; 25:1056. [PMID: 38256131 PMCID: PMC10816607 DOI: 10.3390/ijms25021056] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 12/21/2023] [Accepted: 01/13/2024] [Indexed: 01/24/2024] Open
Abstract
Among the four endometrial cancer (EC) TCGA molecular groups, the MSI/hypermutated group represents an important percentage of tumors (30%), including different histotypes, and generally confers an intermediate prognosis for affected women, also providing new immunotherapeutic strategies. Immunohistochemistry for MMR proteins (MLH1, MSH2, MSH6 and PMS2) has become the optimal diagnostic MSI surrogate worldwide. This review aims to provide state-of-the-art knowledge on MMR deficiency/MSI in EC and to clarify the pathological assessment, interpretation pitfalls and reporting of MMR status.
Collapse
Affiliation(s)
- Francesca Addante
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
- Unit of Anatomical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Antonio d’Amati
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
- Unit of Anatomical Pathology, Department of Precision and Regenerative Medicine and Ionian Area, University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
- Unit of Human Anatomy and Histology, Department of Translational Biomedicine and Neuroscience (DiBraiN), University of Bari “Aldo Moro”, Piazza Giulio Cesare 11, 70124 Bari, Italy
| | - Angela Santoro
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
- Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
| | - Giuseppe Angelico
- Department of Medical and Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.A.)
| | - Frediano Inzani
- Anatomic Pathology Unit, Department of Molecular Medicine, University of Pavia and Fondazione IRCCS San Matteo Hospital, 27100 Pavia, Italy;
| | - Damiano Arciuolo
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Antonio Travaglino
- Pathology Unit, Department of Medicine and Technological Innovation, University of Insubria, 21100 Varese, Italy
| | - Antonio Raffone
- Gynecology and Obstetrics Unit, Department of Public Health, University of Naples Federico II, 80138 Naples, Italy
| | - Nicoletta D’Alessandris
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Giulia Scaglione
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Michele Valente
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Giordana Tinnirello
- Department of Medical and Surgical Sciences and Advanced Technologies “G. F. Ingrassia”, Anatomic Pathology, University of Catania, 95123 Catania, Italy; (G.A.)
| | - Stefania Sfregola
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Belen Padial Urtueta
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Alessia Piermattei
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Federica Cianfrini
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Antonino Mulè
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
| | - Emma Bragantini
- Department of Surgical Pathology, Ospedale S. Chiara, Largo Medaglie d’Oro 9, 38122 Trento, Italy
| | - Gian Franco Zannoni
- Unità di Ginecopatologia e Patologia Mammaria, Dipartimento Scienze della Salute della Donna, del Bambino e di Sanità Pubblica, Fondazione Policlinico Universitario A. Gemelli-IRCCS, Largo A. Gemelli 8, 00168 Rome, Italy (A.d.); (G.S.); (A.P.); (G.F.Z.)
- Istituto di Anatomia Patologica, Università Cattolica del Sacro Cuore, Largo A. Gemelli 8, 00168 Rome, Italy
| |
Collapse
|
71
|
Chong Y, Park G, Cha HJ, Kim HJ, Kang CS, Abdul-Ghafar J, Lee SS. Response to comment on "A stepwise approach to fine needle aspiration cytology of lymph nodes". J Pathol Transl Med 2024; 58:43-44. [PMID: 38229435 DOI: 10.4132/jptm.2023.12.04] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 12/04/2023] [Indexed: 01/18/2024] Open
Affiliation(s)
- Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Gyeongsin Park
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hee Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hyun-Jung Kim
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Chang Suk Kang
- Department of Pathology, Samkwang Medical Laboratories, Seoul, Korea
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung-Sook Lee
- Department of Pathology, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
72
|
Malik S, Zaheer S. ChatGPT as an aid for pathological diagnosis of cancer. Pathol Res Pract 2024; 253:154989. [PMID: 38056135 DOI: 10.1016/j.prp.2023.154989] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 11/26/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Diagnostic workup of cancer patients is highly reliant on the science of pathology using cytopathology, histopathology, and other ancillary techniques like immunohistochemistry and molecular cytogenetics. Data processing and learning by means of artificial intelligence (AI) has become a spearhead for the advancement of medicine, with pathology and laboratory medicine being no exceptions. ChatGPT, an artificial intelligence (AI)-based chatbot, that was recently launched by OpenAI, is currently a talk of the town, and its role in cancer diagnosis is also being explored meticulously. Pathology workflow by integration of digital slides, implementation of advanced algorithms, and computer-aided diagnostic techniques extend the frontiers of the pathologist's view beyond a microscopic slide and enables effective integration, assimilation, and utilization of knowledge that is beyond human limits and boundaries. Despite of it's numerous advantages in the pathological diagnosis of cancer, it comes with several challenges like integration of digital slides with input language parameters, problems of bias, and legal issues which have to be addressed and worked up soon so that we as a pathologists diagnosing malignancies are on the same band wagon and don't miss the train.
Collapse
Affiliation(s)
- Shaivy Malik
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India
| | - Sufian Zaheer
- Department of Pathology, Vardhman Mahavir Medical College and Safdarjung Hospital, New Delhi, India.
| |
Collapse
|
73
|
Hassan T, Li Z, Javed S, Dias J, Werghi N. Neural Graph Refinement for Robust Recognition of Nuclei Communities in Histopathological Landscape. IEEE TRANSACTIONS ON IMAGE PROCESSING : A PUBLICATION OF THE IEEE SIGNAL PROCESSING SOCIETY 2023; 33:241-256. [PMID: 38064329 DOI: 10.1109/tip.2023.3337666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/20/2023]
Abstract
Accurate classification of nuclei communities is an important step towards timely treating the cancer spread. Graph theory provides an elegant way to represent and analyze nuclei communities within the histopathological landscape in order to perform tissue phenotyping and tumor profiling tasks. Many researchers have worked on recognizing nuclei regions within the histology images in order to grade cancerous progression. However, due to the high structural similarities between nuclei communities, defining a model that can accurately differentiate between nuclei pathological patterns still needs to be solved. To surmount this challenge, we present a novel approach, dubbed neural graph refinement, that enhances the capabilities of existing models to perform nuclei recognition tasks by employing graph representational learning and broadcasting processes. Based on the physical interaction of the nuclei, we first construct a fully connected graph in which nodes represent nuclei and adjacent nodes are connected to each other via an undirected edge. For each edge and node pair, appearance and geometric features are computed and are then utilized for generating the neural graph embeddings. These embeddings are used for diffusing contextual information to the neighboring nodes, all along a path traversing the whole graph to infer global information over an entire nuclei network and predict pathologically meaningful communities. Through rigorous evaluation of the proposed scheme across four public datasets, we showcase that learning such communities through neural graph refinement produces better results that outperform state-of-the-art methods.
Collapse
|
74
|
Zhao C, Pan B, Wang T, Yang H, Vance D, Li X, Zhao H, Hu X, Yang T, Chen Z, Hao L, Liu T, Wang Y. Advances in NIR-Responsive Natural Macromolecular Hydrogel Assembly Drugs for Cancer Treatment. Pharmaceutics 2023; 15:2729. [PMID: 38140070 PMCID: PMC10747500 DOI: 10.3390/pharmaceutics15122729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 11/28/2023] [Accepted: 12/01/2023] [Indexed: 12/24/2023] Open
Abstract
Cancer is a serious disease with an abnormal proliferation of organ tissues; it is characterized by malignant infiltration and growth that affects human life. Traditional cancer therapies such as resection, radiotherapy and chemotherapy have a low cure rate and often cause irreversible damage to the body. In recent years, since the traditional treatment of cancer is still very far from perfect, researchers have begun to focus on non-invasive near-infrared (NIR)-responsive natural macromolecular hydrogel assembly drugs (NIR-NMHADs). Due to their unique biocompatibility and extremely high drug encapsulation, coupling with the spatiotemporal controllability of NIR, synergistic photothermal therapy (PTT), photothermal therapy (PDT), chemotherapy (CT) and immunotherapy (IT) has created excellent effects and good prospects for cancer treatment. In addition, some emerging bioengineering technologies can also improve the effectiveness of drug delivery systems. This review will discuss the properties of NIR light, the NIR-functional hydrogels commonly used in current research, the cancer therapy corresponding to the materials encapsulated in them and the bioengineering technology that can assist drug delivery systems. The review provides a constructive reference for the optimization of NIR-NMHAD experimental ideas and its application to human body.
Collapse
Affiliation(s)
- Chenyu Zhao
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China;
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| | - Boyue Pan
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China;
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| | - Tianlin Wang
- Department of Biophysics, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; (T.W.); (H.Y.)
| | - Huazhe Yang
- Department of Biophysics, School of Intelligent Medicine, China Medical University, Shenyang 110122, China; (T.W.); (H.Y.)
| | - David Vance
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
- School of Pharmacy, Queen’s University Belfast, Belfast BT7 1NN, UK
| | - Xiaojia Li
- Teaching Center for Basic Medical Experiment, China Medical University, Shenyang 110122, China; (X.L.); (H.Z.)
| | - Haiyang Zhao
- Teaching Center for Basic Medical Experiment, China Medical University, Shenyang 110122, China; (X.L.); (H.Z.)
| | - Xinru Hu
- The 1st Clinical Department, China Medical University, Shenyang 110122, China;
| | - Tianchang Yang
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
| | - Zihao Chen
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
| | - Liang Hao
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China;
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| | - Ting Liu
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
| | - Yang Wang
- China Medical University—The Queen’s University Belfast Joint College, China Medical University, Shenyang 110122, China; (C.Z.); (B.P.); (D.V.); (T.Y.); (Z.C.)
- Department of Chemistry, School of Forensic Medicine, China Medical University, Shenyang 110122, China;
- Liaoning Province Key Laboratory of Forensic Bio-Evidence Sciences, Shenyang 110122, China
- Center of Forensic Investigation, China Medical University, Shenyang 110122, China
| |
Collapse
|
75
|
Sano S, Akiyoshi T, Yamamoto N, Hiyoshi Y, Mukai T, Yamaguchi T, Nagasaki T, Taketomi A, Fukunaga Y, Kawachi H. Intratumoral Budding and CD8-Positive T-cell Density in Pretreatment Biopsies as a Predictor of Response to Neoadjuvant Chemoradiotherapy in Advanced Rectal Cancer. Clin Colorectal Cancer 2023; 22:411-420.e1. [PMID: 37516615 DOI: 10.1016/j.clcc.2023.07.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/17/2023] [Accepted: 07/18/2023] [Indexed: 07/31/2023]
Abstract
BACKGROUND Neoadjuvant chemoradiotherapy (CRT) is the standard treatment for advanced rectal cancer. Yet, the response to CRT varies from complete response to zero tumor regression. MATERIALS AND METHODS The impact of intratumoral budding (ITB) and intratumoral CD8+ cell density on response to CRT and survival were evaluated in biopsy samples from 266 patients with advanced rectal cancer who were treated with long-course neoadjuvant CRT. The expression of epithelial-mesenchymal transition (EMT) markers was compared between patients with high and low ITB, using data from 174 patients with RNA sequencing. RESULTS High ITB was observed in 62 patients (23.3%). There was no association between ITB and CD8+ cell density. The multivariable logistic regression analysis showed that high CD8+ cell density (OR, 2.69; 95% CI, 1.45-4.98; P = .002) was associated with good response to CRT, whereas high ITB (OR, 0.33; 95% CI, 0.14-0.80; P = .014) was associated with poor response. Multivariable Cox regression analysis for survival showed that high CD8+ cell density was associated with better recurrence-free survival (HR, 0.41; 95% CI, 0.24-0.72; P = .002) and overall survival (HR, 0.36; 95% CI, 0.17-0.74; P = .005), but significance values for ITB were marginal (P = .104 for recurrence-free survival and P = .163 for overall survival). The expression of EMT-related genes was not significantly different between patients with high and low ITB. CONCLUSION ITB and CD8+ cell density in biopsy samples may serve as useful biomarkers to predict therapy response in patients with rectal cancer treated with neoadjuvant CRT.
Collapse
Affiliation(s)
- Shuhei Sano
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Takashi Akiyoshi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan.
| | - Noriko Yamamoto
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Yukiharu Hiyoshi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiki Mukai
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Tomohiro Yamaguchi
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Toshiya Nagasaki
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Yosuke Fukunaga
- Department of Colorectal Surgery, Gastroenterological Center, Cancer Institute Hospital, Japanese Foundation for Cancer Research, Tokyo, Japan
| | - Hiroshi Kawachi
- Division of Pathology, Cancer Institute, Japanese Foundation for Cancer Research, Tokyo, Japan
| |
Collapse
|
76
|
Mou P, Ge QH, Sheng R, Zhu TF, Liu Y, Ding K. Research progress on the immune microenvironment and immunotherapy in gastric cancer. Front Immunol 2023; 14:1291117. [PMID: 38077373 PMCID: PMC10701536 DOI: 10.3389/fimmu.2023.1291117] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Accepted: 10/24/2023] [Indexed: 12/18/2023] Open
Abstract
The tumor microenvironment, particularly the immune microenvironment, plays an indispensable role in the malignant progression and metastasis of gastric cancer (GC). As our understanding of the GC microenvironment continues to evolve, we are gaining deeper insights into the biological mechanisms at the single-cell level. This, in turn, has offered fresh perspectives on GC therapy. Encouragingly, there are various monotherapy and combination therapies in use, such as immune checkpoint inhibitors, adoptive cell transfer therapy, chimeric antigen receptor T cell therapy, antibody-drug conjugates, and cancer vaccines. In this paper, we review the current research progress regarding the GC microenvironment and summarize promising immunotherapy research and targeted therapies.
Collapse
Affiliation(s)
- Pei Mou
- Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Qing-hua Ge
- Department of Otolaryngology, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Rong Sheng
- Department of Outpatient, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Teng-fei Zhu
- Department of Anesthesiology, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Ye Liu
- Department of Blood Transfusion, Changzheng Hospital of Naval Medical University, Shanghai, China
| | - Kai Ding
- Department of Gastroenterology, Changzheng Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
77
|
Kim T, Chang H, Kim B, Yang J, Koo D, Lee J, Chang JW, Hwang G, Gong G, Cho NH, Yoo CW, Pyo JY, Chong Y. Deep learning-based diagnosis of lung cancer using a nationwide respiratory cytology image set: improving accuracy and inter-observer variability. Am J Cancer Res 2023; 13:5493-5503. [PMID: 38058836 PMCID: PMC10695775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2023] [Accepted: 09/18/2023] [Indexed: 12/08/2023] Open
Abstract
Deep learning (DL)-based image analysis has recently seen widespread application in digital pathology. Recent studies utilizing DL in cytopathology have shown promising results, however, the development of DL models for respiratory specimens is limited. In this study, we designed a DL model to improve lung cancer diagnosis accuracy using cytological images from the respiratory tract. This retrospective, multicenter study used digital cytology images of respiratory specimens from a quality-controlled national dataset collected from over 200 institutions. The image processing involves generating extended z-stack images to reduce the phase difference of cell clusters, color normalizing, and cropping image patches to 256 × 256 pixels. The accuracy of diagnosing lung cancer in humans from image patches before and after receiving AI assistance was compared. 30,590 image patches (1,273 whole slide images [WSIs]) were divided into 27,362 (1,146 WSIs) for training, 2,928 (126 WSIs) for validation, and 1,272 (1,272 WSIs) for testing. The Densenet121 model, which showed the best performance among six convolutional neural network models, was used for analysis. The results of sensitivity, specificity, and accuracy were 95.9%, 98.2%, and 96.9% respectively, outperforming the average of three experienced pathologists. The accuracy of pathologists after receiving AI assistance improved from 82.9% to 95.9%, and the inter-rater agreement of Fleiss' Kappa value was improved from 0.553 to 0.908. In conclusion, this study demonstrated that a DL model was effective in diagnosing lung cancer in respiratory cytology. By increasing diagnostic accuracy and reducing inter-observer variability, AI has the potential to enhance the diagnostic capabilities of pathologists.
Collapse
Affiliation(s)
- Taehee Kim
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Internal Medicine, Hallym University Kangnam Sacred Heart Hospital, Hallym University College of MedicineSeoul, South Korea
| | - Hyun Chang
- Division of Medical Oncology and Hematology, Department of Internal Medicine, International St. Mary’s Hospital, Catholic Kwandong University College of MedicineIncheon, South Korea
| | - Binna Kim
- Department of Pathology, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of KoreaGyeonggi-do, South Korea
| | - Jaeho Yang
- Department of Pathology, Asan Medical Center, University of Ulsan College of MedicineSeoul, South Korea
| | - Dongjun Koo
- Interdisciplinary Program in Bioengineering, College of Engineering, Seoul National UniversitySeoul, South Korea
| | - Jeongwon Lee
- Osong Medical Innovation FoundationChungcheongbuk-do, South Korea
| | | | - Gisu Hwang
- AI Team, DeepNoid Inc.Seoul, South Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of MedicineSeoul, South Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of MedicineSeoul, South Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer CenterIlsan, Gyeonggi-do, South Korea
| | - Ju-Yeon Pyo
- Department of Pathology, Yonsei University College of MedicineSeoul, South Korea
| | - Yosep Chong
- Department of Pathology, Uijeongbu St. Mary’s Hospital, College of Medicine, The Catholic University of KoreaGyeonggi-do, South Korea
| |
Collapse
|
78
|
Mohanty S, Shivanna DB, Rao RS, Astekar M, Chandrashekar C, Radhakrishnan R, Sanjeevareddygari S, Kotrashetti V, Kumar P. Building Automation Pipeline for Diagnostic Classification of Sporadic Odontogenic Keratocysts and Non-Keratocysts Using Whole-Slide Images. Diagnostics (Basel) 2023; 13:3384. [PMID: 37958281 PMCID: PMC10648794 DOI: 10.3390/diagnostics13213384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 10/13/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The microscopic diagnostic differentiation of odontogenic cysts from other cysts is intricate and may cause perplexity for both clinicians and pathologists. Of particular interest is the odontogenic keratocyst (OKC), a developmental cyst with unique histopathological and clinical characteristics. Nevertheless, what distinguishes this cyst is its aggressive nature and high tendency for recurrence. Clinicians encounter challenges in dealing with this frequently encountered jaw lesion, as there is no consensus on surgical treatment. Therefore, the accurate and early diagnosis of such cysts will benefit clinicians in terms of treatment management and spare subjects from the mental agony of suffering from aggressive OKCs, which impact their quality of life. The objective of this research is to develop an automated OKC diagnostic system that can function as a decision support tool for pathologists, whether they are working locally or remotely. This system will provide them with additional data and insights to enhance their decision-making abilities. This research aims to provide an automation pipeline to classify whole-slide images of OKCs and non-keratocysts (non-KCs: dentigerous and radicular cysts). OKC diagnosis and prognosis using the histopathological analysis of tissues using whole-slide images (WSIs) with a deep-learning approach is an emerging research area. WSIs have the unique advantage of magnifying tissues with high resolution without losing information. The contribution of this research is a novel, deep-learning-based, and efficient algorithm that reduces the trainable parameters and, in turn, the memory footprint. This is achieved using principal component analysis (PCA) and the ReliefF feature selection algorithm (ReliefF) in a convolutional neural network (CNN) named P-C-ReliefF. The proposed model reduces the trainable parameters compared to standard CNN, achieving 97% classification accuracy.
Collapse
Affiliation(s)
- Samahit Mohanty
- Department of Computer Science and Engineering, M S Ramaiah University of Applied Sciences, Bengaluru 560054, India;
| | - Divya B. Shivanna
- Department of Computer Science and Engineering, M S Ramaiah University of Applied Sciences, Bengaluru 560054, India;
| | - Roopa S. Rao
- Department of Oral Pathology and Microbiology, Faculty of Dental Sciences, M S Ramaiah University of Applied Sciences, Bengaluru 560054, India;
| | - Madhusudan Astekar
- Department of Oral Pathology, Institute of Dental Sciences, Bareilly 243006, India;
| | - Chetana Chandrashekar
- Department of Oral & Maxillofacial Pathology & Microbiology, Manipal College of Dental Sciences, Manipal 576104, India; (C.C.); (R.R.)
| | - Raghu Radhakrishnan
- Department of Oral & Maxillofacial Pathology & Microbiology, Manipal College of Dental Sciences, Manipal 576104, India; (C.C.); (R.R.)
| | | | - Vijayalakshmi Kotrashetti
- Department of Oral & Maxillofacial Pathology & Microbiology, Maratha Mandal’s Nathajirao G Halgekar, Institute of Dental Science & Research Centre, Belgaum 590010, India;
| | - Prashant Kumar
- Department of Oral & Maxillofacial Pathology, Nijalingappa Institute of Dental Science & Research, Gulbarga 585105, India;
| |
Collapse
|
79
|
Kang D, Sohn SY. CTM and QFD analysis: Framework for fintech adoption priority in commercial banks. PLoS One 2023; 18:e0287826. [PMID: 37910545 PMCID: PMC10619835 DOI: 10.1371/journal.pone.0287826] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Accepted: 06/14/2023] [Indexed: 11/03/2023] Open
Abstract
As financial technology (fintech) is developing rapidly, many commercial banks experience difficulty deciding what kind of fintech to primarily focus on when managing their business. Owing to limited resources and assets, there is a practical need for guidelines for banks' investments in fintech. This study provides a systemic procedure to identify promising fintech groups and their investment priorities. We propose a QFD-based decision support framework for banks by considering both aspects of the emerging fintech push identified using patent topic modeling and the market pull of banking services obtained from a survey of the literature and experts. An empirical application of the proposed QFD framework to major South Korean banks shows that transaction support technology, secure transactions, and trading platforms are the three most important fintech categories. The QFD results are utilized to guide individual banks for further investment strategies such as mergers and acquisitions, strategic partnerships, and spin-off operations. The proposed framework can be generalized and applied to other financial service firms.
Collapse
Affiliation(s)
- Donghoon Kang
- Graduate Program in Technology Policy, Yonsei University, Seoul, Republic of Korea
| | - So Young Sohn
- Department of Industrial Engineering, Yonsei University, Seoul, Republic of Korea
| |
Collapse
|
80
|
Yamamoto G, Ito T, Suzuki O, Kamae N, Kakuta M, Takahashi A, Iuchi K, Arai T, Ishida H, Akagi K. Concordance between microsatellite instability testing and immunohistochemistry for mismatch repair proteins and efficient screening of mismatch repair deficient gastric cancer. Oncol Lett 2023; 26:494. [PMID: 37854865 PMCID: PMC10579988 DOI: 10.3892/ol.2023.14081] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Accepted: 09/07/2023] [Indexed: 10/20/2023] Open
Abstract
Microsatellite instability (MSI) testing, an established technique that has gained prominence in recent years for its predictive potential regarding the efficacy of immune checkpoint inhibitors, is used to evaluate DNA mismatch repair (MMR) deficiency (dMMR). As with other methods, the immunohistochemistry (IHC) of MMR proteins is also widely adopted. Although both techniques have been validated, their concordance rate remains unknown, particularly regarding non-colorectal cancer. Therefore, the aim of the present study was to explore and elucidate their concordance in the context of gastric cancer (GC). A total of 489 surgically resected primary GC tissues were analyzed to compare the results yielded by the MSI test and those from IHC. Of 488 GC cases, 56 (11.5%) exhibited a loss of MMR proteins, whereas 52 (10.7%) were classified as high-frequency MSI (MSI-H). The concordance rate between these two categories was 99.2%. The microsatellite markers BAT26 and MONO27 demonstrated 100% sensitivity and 99.5% specificity in detecting dMMR GC. In addition, histopathological analysis revealed that MSI-H was more prevalent in GCs exhibiting coexisting Tub2 and Por1 subtypes. However, four discordant cases were observed. All four cases were microsatellite-stable cases but exhibited loss of MLH1 protein expression with hypermethylation of the MLH1 promoter. The results of the present study highlight that while there is a strong concordance between MSI and IHC testing results for determining dMMR status, IHC testing may offer superior efficacy in detecting dMMR.
Collapse
Affiliation(s)
- Gou Yamamoto
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Tetsuya Ito
- Department of Digestive Tract and General Surgery, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| | - Okihide Suzuki
- Department of Digestive Tract and General Surgery, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
- Department of Clinical Genetics, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| | - Nao Kamae
- Department of Clinical Genetics, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| | - Miho Kakuta
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Akemi Takahashi
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Katsuya Iuchi
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama 362-0806, Japan
| | - Tomio Arai
- Department of Pathology, Tokyo Metropolitan Geriatric Hospital, Tokyo 173-0015, Japan
| | - Hideyuki Ishida
- Department of Digestive Tract and General Surgery, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
- Department of Clinical Genetics, Saitama Medical Center, Saitama Medical University, Saitama 350-8550, Japan
| | - Kiwamu Akagi
- Department of Molecular Diagnosis and Cancer Prevention, Saitama Cancer Center, Saitama 362-0806, Japan
| |
Collapse
|
81
|
Ramzan M, Raza M, Sharif MI, Azam F, Kim J, Kadry S. Gastrointestinal tract disorders classification using ensemble of InceptionNet and proposed GITNet based deep feature with ant colony optimization. PLoS One 2023; 18:e0292601. [PMID: 37831692 PMCID: PMC10575542 DOI: 10.1371/journal.pone.0292601] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Accepted: 09/24/2023] [Indexed: 10/15/2023] Open
Abstract
Computer-aided classification of diseases of the gastrointestinal tract (GIT) has become a crucial area of research. Medical science and artificial intelligence have helped medical experts find GIT diseases through endoscopic procedures. Wired endoscopy is a controlled procedure that helps the medical expert in disease diagnosis. Manual screening of the endoscopic frames is a challenging and time taking task for medical experts that also increases the missed rate of the GIT disease. An early diagnosis of GIT disease can save human beings from fatal diseases. An automatic deep feature learning-based system is proposed for GIT disease classification. The adaptive gamma correction and weighting distribution (AGCWD) preprocessing procedure is the first stage of the proposed work that is used for enhancing the intensity of the frames. The deep features are extracted from the frames by deep learning models including InceptionNetV3 and GITNet. Ant Colony Optimization (ACO) procedure is employed for feature optimization. Optimized features are fused serially. The classification operation is performed by variants of support vector machine (SVM) classifiers, including the Cubic SVM (CSVM), Coarse Gaussian SVM (CGSVM), Quadratic SVM (QSVM), and Linear SVM (LSVM) classifiers. The intended model is assessed on two challenging datasets including KVASIR and NERTHUS that consist of eight and four classes respectively. The intended model outperforms as compared with existing methods by achieving an accuracy of 99.32% over the KVASIR dataset and 99.89% accuracy using the NERTHUS dataset.
Collapse
Affiliation(s)
- Muhammad Ramzan
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Pakistan
| | - Mudassar Raza
- Department of Computer Science, HITEC University Taxila, Taxila, Pakistan
| | - Muhammad Irfan Sharif
- Department of Information Sciences, University of Education Lahore, Jauharabad Campus, Jauharabad, Pakistan
| | - Faisal Azam
- Department of Computer Science, COMSATS University Islamabad, Wah Campus, Pakistan
| | - Jungeun Kim
- Department of Software and CMPSI, Kongju National University, Cheonan, Korea
| | - Seifedine Kadry
- Department of Applied Data Science, Noroff University College, Kristiansand, Norway
- Artificial Intelligence Research Center (AIRC), Ajman University, Ajman, United Arab Emirates
- Department of Electrical and Computer Engineering, Lebanese American University, Byblos, Lebanon
- MEU Research Unit, Middle East University, Amman, Jordan
| |
Collapse
|
82
|
Beniwal SS, Lamo P, Kaushik A, Lorenzo-Villegas DL, Liu Y, MohanaSundaram A. Current Status and Emerging Trends in Colorectal Cancer Screening and Diagnostics. BIOSENSORS 2023; 13:926. [PMID: 37887119 PMCID: PMC10605407 DOI: 10.3390/bios13100926] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2023] [Revised: 09/27/2023] [Accepted: 10/07/2023] [Indexed: 10/28/2023]
Abstract
Colorectal cancer (CRC) is a prevalent and potentially fatal disease categorized based on its high incidences and mortality rates, which raised the need for effective diagnostic strategies for the early detection and management of CRC. While there are several conventional cancer diagnostics available, they have certain limitations that hinder their effectiveness. Significant research efforts are currently being dedicated to elucidating novel methodologies that aim at comprehending the intricate molecular mechanism that underlies CRC. Recently, microfluidic diagnostics have emerged as a pivotal solution, offering non-invasive approaches to real-time monitoring of disease progression and treatment response. Microfluidic devices enable the integration of multiple sample preparation steps into a single platform, which speeds up processing and improves sensitivity. Such advancements in diagnostic technologies hold immense promise for revolutionizing the field of CRC diagnosis and enabling efficient detection and monitoring strategies. This article elucidates several of the latest developments in microfluidic technology for CRC diagnostics. In addition to the advancements in microfluidic technology for CRC diagnostics, the integration of artificial intelligence (AI) holds great promise for further enhancing diagnostic capabilities. Advancements in microfluidic systems and AI-driven approaches can revolutionize colorectal cancer diagnostics, offering accurate, efficient, and personalized strategies to improve patient outcomes and transform cancer management.
Collapse
Affiliation(s)
| | - Paula Lamo
- Escuela Superior de Ingeniería y Tecnología, Universidad Internacional de La Rioja, 26006 Logroño, Spain
| | - Ajeet Kaushik
- NanoBioTech Laboratory, Department of Environmental Engineering, Florida Polytechnic University, Lakeland, FL 33805, USA
| | | | - Yuguang Liu
- Departments of Physiology and Biomedical Engineering, Immunology and Surgery, Microbiome Program, Center for Individualized Medicine, Mayo Clinic, Rochester, MN 55905, USA
| | | |
Collapse
|
83
|
Keshtkar K, Reza Safarpour A, Heshmat R, Sotoudehmanesh R, Keshtkar A. A Systematic Review and Meta-analysis of Convolutional Neural Network in the Diagnosis of Colorectal Polyps and Cancer. THE TURKISH JOURNAL OF GASTROENTEROLOGY : THE OFFICIAL JOURNAL OF TURKISH SOCIETY OF GASTROENTEROLOGY 2023; 34:985-997. [PMID: 37681266 PMCID: PMC10645297 DOI: 10.5152/tjg.2023.22491] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Accepted: 03/22/2023] [Indexed: 09/09/2023]
Abstract
Convolutional neural networks are a class of deep neural networks used for different clinical purposes, including improving the detection rate of colorectal lesions. This systematic review and meta-analysis aimed to assess the performance of convolutional neural network-based models in the detection or classification of colorectal polyps and colorectal cancer. A systematic search was performed in MEDLINE, SCOPUS, Web of Science, and other related databases. The performance measures of the convolutional neural network models in the detection of colorectal polyps and colorectal cancer were calculated in the 2 scenarios of the best and worst accuracy. Stata and R software were used for conducting the meta-analysis. From 3368 searched records, 24 primary studies were included. The sensitivity and specificity of convolutional neural network models in predicting colorectal polyps in worst and best scenarios ranged from 84.7% to 91.6% and from 86.0% to 93.8%, respectively. These values in predicting colorectal cancer varied between 93.2% and 94.1% and between 94.6% and 97.7%. The positive and negative likelihood ratios varied between 6.2 and 14.5 and 0.09 and 0.17 in these scenarios, respectively, in predicting colorectal polyps, and 17.1-41.2 and 0.07-0.06 in predicting colorectal polyps. The diagnostic odds ratio and accuracy measures of convolutional neural network models in predicting colorectal polyps in worst and best scenarios ranged between 36% and 162% and between 80.5% and 88.6%, respectively. These values in predicting colorectal cancer in the worst and the best scenarios varied between 239.63% and 677.47% and between 88.2% and 96.4%. The area under the receiver operating characteristic varied between 0.92 and 0.97 in the worst and the best scenarios in colorectal polyps, respectively, and between 0.98 and 0.99 in colorectal polyps prediction. Convolutional neural network-based models showed an acceptable accuracy in detecting colorectal polyps and colorectal cancer.
Collapse
Affiliation(s)
- Kamyab Keshtkar
- University of Tehran School of Electrical and Computer Engineering, Tehran, Iran
| | - Ali Reza Safarpour
- Gastroenterohepatology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ramin Heshmat
- Chronic Diseases Research Center, Endocrinology and Metabolism Population Sciences Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Rasoul Sotoudehmanesh
- Department of Gastroenterology, Digestive Disease Research Center, Digestive Disease Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| | - Abbas Keshtkar
- Department of Health Sciences Education Development, Tehran University of Medical Sciences School of Public Health, Tehran, Iran
| |
Collapse
|
84
|
Jing Y, Li C, Du T, Jiang T, Sun H, Yang J, Shi L, Gao M, Grzegorzek M, Li X. A comprehensive survey of intestine histopathological image analysis using machine vision approaches. Comput Biol Med 2023; 165:107388. [PMID: 37696178 DOI: 10.1016/j.compbiomed.2023.107388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/06/2023] [Accepted: 08/25/2023] [Indexed: 09/13/2023]
Abstract
Colorectal Cancer (CRC) is currently one of the most common and deadly cancers. CRC is the third most common malignancy and the fourth leading cause of cancer death worldwide. It ranks as the second most frequent cause of cancer-related deaths in the United States and other developed countries. Histopathological images contain sufficient phenotypic information, they play an indispensable role in the diagnosis and treatment of CRC. In order to improve the objectivity and diagnostic efficiency for image analysis of intestinal histopathology, Computer-aided Diagnosis (CAD) methods based on machine learning (ML) are widely applied in image analysis of intestinal histopathology. In this investigation, we conduct a comprehensive study on recent ML-based methods for image analysis of intestinal histopathology. First, we discuss commonly used datasets from basic research studies with knowledge of intestinal histopathology relevant to medicine. Second, we introduce traditional ML methods commonly used in intestinal histopathology, as well as deep learning (DL) methods. Then, we provide a comprehensive review of the recent developments in ML methods for segmentation, classification, detection, and recognition, among others, for histopathological images of the intestine. Finally, the existing methods have been studied, and the application prospects of these methods in this field are given.
Collapse
Affiliation(s)
- Yujie Jing
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Chen Li
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China.
| | - Tianming Du
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Tao Jiang
- School of Intelligent Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China; International Joint Institute of Robotics and Intelligent Systems, Chengdu University of Information Technology, Chengdu, China
| | - Hongzan Sun
- Shengjing Hospital of China Medical University, Shenyang, China
| | - Jinzhu Yang
- Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Liyu Shi
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Minghe Gao
- Microscopic Image and Medical Image Analysis Group, College of Medicine and Biological Information Engineering, Northeastern University, China; Key Laboratory of Intelligent Computing in Medical Image, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Marcin Grzegorzek
- Institute for Medical Informatics, University of Luebeck, Luebeck, Germany; Department of Knowledge Engineering, University of Economics in Katowice, Katowice, Poland
| | - Xiaoyan Li
- Cancer Hospital of China Medical University, Liaoning Cancer Hospital, Shenyang, China.
| |
Collapse
|
85
|
Tan J, Feng R. A pan-cancer analysis of STAT3 expression and genetic alterations in human tumors. Open Med (Wars) 2023; 18:20230792. [PMID: 37724127 PMCID: PMC10505358 DOI: 10.1515/med-2023-0792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 03/30/2023] [Accepted: 08/08/2023] [Indexed: 09/20/2023] Open
Abstract
Combined cancer immunotherapy and targeted therapy have proven to be effective against various cancers and therefore have recently become the focus of cancer research. Signal transducer and activator of transcription 3 (STAT3) is a member of the STAT protein family of transcription factors. Several studies have shown that STAT3 can affect the prognosis of cancer patients by regulating immune microenvironment (IME). Therefore, STAT3 may have high research value for the development of combined immunotherapy/targeted therapy approaches for the treatment of cancer patients. We found differences in STAT3 expression between tumor and normal tissues. Kaplan-Meier survival and Cox regression analyses showed that high expression of STAT3 is associated with poor prognosis in low-grade glioma (LGG) patients. The results of the analysis of the area under the curve of the receiver operating characteristic curve further suggested that the expression of STAT3 is an effective way to evaluate the prognosis of patients with glioma. The results of the IME analysis revealed that the immune and matrix scores of LGGs were positively correlated with the expression of STAT3 (P < 0.05). The results of immune cell infiltration analysis showed that STAT3 was positively correlated with resting dendritic cells, eosinophils, neutrophils, M0 macrophages, M1 macrophages, CD4 memory resting T cells, and CD8 T cells in LGG patients, but negatively correlated with activated mast cells and M2 macrophages (P < 0.05). Our gene set enrichment analysis identified 384 enriched pathways. According to the enrichment scores, the top ten most significantly upregulated pathways were related to immune response. The top ten most significantly downregulated pathways were related to cell signal transduction and the regulation of cell survival, proliferation, and metabolism. Genetic alteration analysis showed that missense mutations in STAT3 account for the majority of mutations, and STAT3 mutations mostly occur in the Src homology domain. In conclusion overexpression of STAT3 can promote the development and growth of tumors by regulating IME, which is significantly related to the poor prognosis of cancer patients. Therefore, targeted inhibition of STAT3 expression may have high research value for the development of combined immunotherapy/targeted therapy approaches for the treatment of cancer patients.
Collapse
Affiliation(s)
- Junyin Tan
- Department of Oncology, Guigang People’s Hospital of Guangxi/The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, China
| | - Ronghao Feng
- Department of Oncology, Guigang People’s Hospital of Guangxi/The Eighth Affiliated Hospital of Guangxi Medical University, Guigang, Guangxi, China
| |
Collapse
|
86
|
Shao Z, Dai L, Jonnagaddala J, Chen Y, Wang Y, Fang Z, Zhang Y. Generalizability of Self-Supervised Training Models for Digital Pathology: A Multicountry Comparison in Colorectal Cancer. JCO Clin Cancer Inform 2023; 7:e2200178. [PMID: 37703507 DOI: 10.1200/cci.22.00178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 06/15/2023] [Accepted: 06/27/2023] [Indexed: 09/15/2023] Open
Abstract
PURPOSE In this multicountry study, we aim to explore the effectiveness of self-supervised learning (SSL) in colorectal cancer (CRC)-related predictive tasks using large amount of unlabeled digital pathology imaging data. METHODS We adopted SimSiam to conduct self-supervised pretraining on two large whole-slide image CRC data sets from the United States and Australia. The SSL pretrained encoder is then used in several predictive tasks, including supervised predictive tasks (tissue classification, microsatellite instability v microsatellite stability classification), and weakly supervised predictive tasks (polyp type classification and adenoma grading, and 5-year survival prediction). Performance on the tasks was compared between models using SSL pretraining and those using ImageNet pretraining, and performance for one-country pretraining was compared with two-country pretraining. RESULTS We demonstrate that SSL pretraining outperforms ImageNet pretraining in predictive tasks, that is, SSL pretraining outperforms the ImageNet pretraining by 3.01% of F 1 score on average over supervised predictive tasks and 1.53% of AUC on average over weakly supervised predictive tasks. Furthermore, two-country SSL pretraining has shown more stable performance than single-country pretraining, that is, two-country pretraining outperforms at least one of the single-country pretrainings by 1.93% of F 1 on average over supervised predictive tasks and 1.36% of AUC on average over weakly-supervised predictive tasks. CONCLUSION We find that using unlabeled image data for SSL pretraining in CRC related tasks is more effective than using ImageNet pretraining. Furthermore, SSL pretraining using data from multiple countries achieve more stable performance and better generalization than single-country pretraining.
Collapse
Affiliation(s)
- Zhuchen Shao
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Liuxi Dai
- School of Computer Science and Technology, Harbin Institute of Technology, Shenzhen, China
| | | | - Yang Chen
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Yifeng Wang
- School of Science, Harbin Institute of Technology, Shenzhen, China
| | - Zijie Fang
- Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen, China
| | - Yongbing Zhang
- School of Computer Science and Technology, Harbin Institute of Technology, Shenzhen, China
| |
Collapse
|
87
|
Kaczmarzyk JR, Gupta R, Kurc TM, Abousamra S, Saltz JH, Koo PK. ChampKit: A framework for rapid evaluation of deep neural networks for patch-based histopathology classification. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2023; 239:107631. [PMID: 37271050 PMCID: PMC11093625 DOI: 10.1016/j.cmpb.2023.107631] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2023] [Revised: 04/23/2023] [Accepted: 05/28/2023] [Indexed: 06/06/2023]
Abstract
BACKGROUND AND OBJECTIVE Histopathology is the gold standard for diagnosis of many cancers. Recent advances in computer vision, specifically deep learning, have facilitated the analysis of histopathology images for many tasks, including the detection of immune cells and microsatellite instability. However, it remains difficult to identify optimal models and training configurations for different histopathology classification tasks due to the abundance of available architectures and the lack of systematic evaluations. Our objective in this work is to present a software tool that addresses this need and enables robust, systematic evaluation of neural network models for patch classification in histology in a light-weight, easy-to-use package for both algorithm developers and biomedical researchers. METHODS Here we present ChampKit (Comprehensive Histopathology Assessment of Model Predictions toolKit): an extensible, fully reproducible evaluation toolkit that is a one-stop-shop to train and evaluate deep neural networks for patch classification. ChampKit curates a broad range of public datasets. It enables training and evaluation of models supported by timm directly from the command line, without the need for users to write any code. External models are enabled through a straightforward API and minimal coding. As a result, Champkit facilitates the evaluation of existing and new models and deep learning architectures on pathology datasets, making it more accessible to the broader scientific community. To demonstrate the utility of ChampKit, we establish baseline performance for a subset of possible models that could be employed with ChampKit, focusing on several popular deep learning models, namely ResNet18, ResNet50, and R26-ViT, a hybrid vision transformer. In addition, we compare each model trained either from random weight initialization or with transfer learning from ImageNet pretrained models. For ResNet18, we also consider transfer learning from a self-supervised pretrained model. RESULTS The main result of this paper is the ChampKit software. Using ChampKit, we were able to systemically evaluate multiple neural networks across six datasets. We observed mixed results when evaluating the benefits of pretraining versus random intialization, with no clear benefit except in the low data regime, where transfer learning was found to be beneficial. Surprisingly, we found that transfer learning from self-supervised weights rarely improved performance, which is counter to other areas of computer vision. CONCLUSIONS Choosing the right model for a given digital pathology dataset is nontrivial. ChampKit provides a valuable tool to fill this gap by enabling the evaluation of hundreds of existing (or user-defined) deep learning models across a variety of pathology tasks. Source code and data for the tool are freely accessible at https://github.com/SBU-BMI/champkit.
Collapse
Affiliation(s)
- Jakub R Kaczmarzyk
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA; Simons Center for Quantitative Biology, 1 Bungtown Rd, Cold Spring Harbor, 11724, NY, USA.
| | - Rajarsi Gupta
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA
| | - Tahsin M Kurc
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA
| | - Shahira Abousamra
- Department of Computer Science, Stony Brook University, Stony Brook, NY, USA
| | - Joel H Saltz
- Department of Biomedical Informatics, Stony Brook Medicine, 101 Nicolls Rd, Stony Brook, 11794, NY, USA.
| | - Peter K Koo
- Simons Center for Quantitative Biology, 1 Bungtown Rd, Cold Spring Harbor, 11724, NY, USA.
| |
Collapse
|
88
|
Chong Y, Hong SA, Oh HK, Jung SJ, Kim BS, Jeong JY, Lee HC, Gong G, The Committee of Quality Improvement of Korean Society for Cytopathology. Diagnostic proficiency test using digital cytopathology and comparative assessment of whole slide images of cytologic samples for quality assurance program in Korea. J Pathol Transl Med 2023; 57:251-264. [PMID: 37608552 PMCID: PMC10518242 DOI: 10.4132/jptm.2023.07.17] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Revised: 07/11/2023] [Accepted: 07/17/2023] [Indexed: 08/24/2023] Open
Abstract
BACKGROUND The Korean Society for Cytopathology introduced a digital proficiency test (PT) in 2021. However, many doubtful opinions remain on whether digitally scanned images can satisfactorily present subtle differences in the nuclear features and chromatin patterns of cytological samples. METHODS We prepared 30 whole-slide images (WSIs) from the conventional PT archive by a selection process for digital PT. Digital and conventional PT were performed in parallel for volunteer institutes, and the results were compared using feedback. To assess the quality of cytological assessment WSIs, 12 slides were collected and scanned using five different scanners, with four cytopathologists evaluating image quality through a questionnaire. RESULTS Among the 215 institutes, 108 and 107 participated in glass and digital PT, respectively. No significant difference was noted in category C (major discordance), although the number of discordant cases was slightly higher in the digital PT group. Leica, 3DHistech Pannoramic 250 Flash, and Hamamatsu NanoZoomer 360 systems showed comparable results in terms of image quality, feature presentation, and error rates for most cytological samples. Overall satisfaction was observed with the general convenience and image quality of digital PT. CONCLUSIONS As three-dimensional clusters are common and nuclear/chromatin features are critical for cytological interpretation, careful selection of scanners and optimal conditions are mandatory for the successful establishment of digital quality assurance programs in cytology.
Collapse
Affiliation(s)
- Yosep Chong
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Soon Auck Hong
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
| | - Hoon Kyu Oh
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
| | - Soo Jin Jung
- Department of Pathology, Inje University Busan Paik Hospital, Busan, Korea
| | - Bo-Sung Kim
- Department of Pathology, Green Cross Laboratories, Yongin, Korea
| | - Ji Yun Jeong
- Department of Pathology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
| | - Ho-Chang Lee
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - The Committee of Quality Improvement of Korean Society for Cytopathology
- Department of Hospital Pathology, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Pathology, Chung-Ang University College of Medicine, Seoul, Korea
- Department of Pathology, Daegu Catholic University School of Medicine, Daegu, Korea
- Department of Pathology, Inje University Busan Paik Hospital, Busan, Korea
- Department of Pathology, Green Cross Laboratories, Yongin, Korea
- Department of Pathology, Kyungpook National University Chilgok Hospital, School of Medicine, Kyungpook National University, Daegu, Korea
- Department of Pathology, Chungbuk National University College of Medicine, Cheongju, Korea
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| |
Collapse
|
89
|
Chlorogiannis DD, Verras GI, Tzelepi V, Chlorogiannis A, Apostolos A, Kotis K, Anagnostopoulos CN, Antzoulas A, Davakis S, Vailas M, Schizas D, Mulita F. Tissue classification and diagnosis of colorectal cancer histopathology images using deep learning algorithms. Is the time ripe for clinical practice implementation? PRZEGLAD GASTROENTEROLOGICZNY 2023; 18:353-367. [PMID: 38572457 PMCID: PMC10985751 DOI: 10.5114/pg.2023.130337] [Citation(s) in RCA: 30] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2023] [Accepted: 05/20/2023] [Indexed: 04/05/2024]
Abstract
Colorectal cancer is one of the most prevalent types of cancer, with histopathologic examination of biopsied tissue samples remaining the gold standard for diagnosis. During the past years, artificial intelligence (AI) has steadily found its way into the field of medicine and pathology, especially with the introduction of whole slide imaging (WSI). The main outcome of interest was the composite balanced accuracy (ACC) as well as the F1 score. The average reported ACC from the collected studies was 95.8 ±3.8%. Reported F1 scores reached as high as 0.975, with an average of 89.7 ±9.8%, indicating that existing deep learning algorithms can achieve in silico distinction between malignant and benign. Overall, the available state-of-the-art algorithms are non-inferior to pathologists for image analysis and classification tasks. However, due to their inherent uniqueness in their training and lack of widely accepted external validation datasets, their generalization potential is still limited.
Collapse
Affiliation(s)
| | | | - Vasiliki Tzelepi
- Department of Pathology, School of Medicine, University of Patras, Patras, Greece
| | | | - Anastasios Apostolos
- First Department of Cardiology, Hippokration Hospital, University of Athens, Athens, Greece
| | - Konstantinos Kotis
- Intelligent Systems Lab, Department of Cultural Technology and Communication, University of the Aegean, Mytilene, Greece
| | | | - Andreas Antzoulas
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| | - Spyridon Davakis
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Michail Vailas
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Dimitrios Schizas
- Upper Gastrointestinal and General Surgery Unit, First Department of Surgery, National and Kapodistrian University of Athens, Laiko General Hospital, Athens, Greece
| | - Francesk Mulita
- Department of Surgery, General University Hospital of Patras, Patras, Greece
| |
Collapse
|
90
|
Davri A, Birbas E, Kanavos T, Ntritsos G, Giannakeas N, Tzallas AT, Batistatou A. Deep Learning for Lung Cancer Diagnosis, Prognosis and Prediction Using Histological and Cytological Images: A Systematic Review. Cancers (Basel) 2023; 15:3981. [PMID: 37568797 PMCID: PMC10417369 DOI: 10.3390/cancers15153981] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is one of the deadliest cancers worldwide, with a high incidence rate, especially in tobacco smokers. Lung cancer accurate diagnosis is based on distinct histological patterns combined with molecular data for personalized treatment. Precise lung cancer classification from a single H&E slide can be challenging for a pathologist, requiring most of the time additional histochemical and special immunohistochemical stains for the final pathology report. According to WHO, small biopsy and cytology specimens are the available materials for about 70% of lung cancer patients with advanced-stage unresectable disease. Thus, the limited available diagnostic material necessitates its optimal management and processing for the completion of diagnosis and predictive testing according to the published guidelines. During the new era of Digital Pathology, Deep Learning offers the potential for lung cancer interpretation to assist pathologists' routine practice. Herein, we systematically review the current Artificial Intelligence-based approaches using histological and cytological images of lung cancer. Most of the published literature centered on the distinction between lung adenocarcinoma, lung squamous cell carcinoma, and small cell lung carcinoma, reflecting the realistic pathologist's routine. Furthermore, several studies developed algorithms for lung adenocarcinoma predominant architectural pattern determination, prognosis prediction, mutational status characterization, and PD-L1 expression status estimation.
Collapse
Affiliation(s)
- Athena Davri
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| | - Effrosyni Birbas
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Theofilos Kanavos
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Nikolaos Giannakeas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Alexandros T. Tzallas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Anna Batistatou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| |
Collapse
|
91
|
Zhou J, Foroughi Pour A, Deirawan H, Daaboul F, Aung TN, Beydoun R, Ahmed FS, Chuang JH. Integrative deep learning analysis improves colon adenocarcinoma patient stratification at risk for mortality. EBioMedicine 2023; 94:104726. [PMID: 37499603 PMCID: PMC10388166 DOI: 10.1016/j.ebiom.2023.104726] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2023] [Revised: 06/19/2023] [Accepted: 07/10/2023] [Indexed: 07/29/2023] Open
Abstract
BACKGROUND Colorectal cancers are the fourth most diagnosed cancer and the second leading cancer in number of deaths. Many clinical variables, pathological features, and genomic signatures are associated with patient risk, but reliable patient stratification in the clinic remains a challenging task. Here we assess how image, clinical, and genomic features can be combined to predict risk. METHODS We developed and evaluated integrative deep learning models combining formalin-fixed, paraffin-embedded (FFPE) whole slide images (WSIs), clinical variables, and mutation signatures to stratify colon adenocarcinoma (COAD) patients based on their risk of mortality. Our models were trained using a dataset of 108 patients from The Cancer Genome Atlas (TCGA), and were externally validated on newly generated dataset from Wayne State University (WSU) of 123 COAD patients and rectal adenocarcinoma (READ) patients in TCGA (N = 52). FINDINGS We first observe that deep learning models trained on FFPE WSIs of TCGA-COAD separate high-risk (OS < 3 years, N = 38) and low-risk (OS > 5 years, N = 25) patients (AUC = 0.81 ± 0.08, 5 year survival p < 0.0001, 5 year relative risk = 1.83 ± 0.04) though such models are less effective at predicting overall survival (OS) for moderate-risk (3 years < OS < 5 years, N = 45) patients (5 year survival p-value = 0.5, 5 year relative risk = 1.05 ± 0.09). We find that our integrative models combining WSIs, clinical variables, and mutation signatures can improve patient stratification for moderate-risk patients (5 year survival p < 0.0001, 5 year relative risk = 1.87 ± 0.07). Our integrative model combining image and clinical variables is also effective on an independent pathology dataset (WSU-COAD, N = 123) generated by our team (5 year survival p < 0.0001, 5 year relative risk = 1.52 ± 0.08), and the TCGA-READ data (5 year survival p < 0.0001, 5 year relative risk = 1.18 ± 0.17). Our multicenter integrative image and clinical model trained on combined TCGA-COAD and WSU-COAD is effective in predicting risk on TCGA-READ (5 year survival p < 0.0001, 5 year relative risk = 1.82 ± 0.13) data. Pathologist review of image-based heatmaps suggests that nuclear size pleomorphism, intense cellularity, and abnormal structures are associated with high-risk, while low-risk regions have more regular and small cells. Quantitative analysis shows high cellularity, high ratios of tumor cells, large tumor nuclei, and low immune infiltration are indicators of high-risk tiles. INTERPRETATION The improved stratification of colorectal cancer patients from our computational methods can be beneficial for treatment plans and enrollment of patients in clinical trials. FUNDING This study was supported by the National Cancer Institutes (Grant No. R01CA230031 and P30CA034196). The funders had no roles in study design, data collection and analysis or preparation of the manuscript.
Collapse
Affiliation(s)
- Jie Zhou
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, Farmington, CT, USA
| | | | - Hany Deirawan
- Department of Pathology, Wayne State University, Detroit, MI, USA; Department of Dermatology, Wayne State University, Detroit, MI, USA
| | - Fayez Daaboul
- Department of Pathology, Wayne State University, Detroit, MI, USA
| | - Thazin Nwe Aung
- Department of Pathology, Yale University, New Haven, CT, USA
| | - Rafic Beydoun
- Department of Pathology, Wayne State University, Detroit, MI, USA
| | | | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA; Department of Genetics and Genome Sciences, UCONN Health, Farmington, CT, USA.
| |
Collapse
|
92
|
Yoon K, Lee S, Lee TH, Kim KG. The Design of an Automatic Temperature Compensation System through Smart Heat Comparison/Judgment and Control for Stable Thermal Treatment in Hyperthermic Intraperitoneal Chemotherapy (HIPEC) Surgery. SENSORS (BASEL, SWITZERLAND) 2023; 23:6722. [PMID: 37571507 PMCID: PMC10422214 DOI: 10.3390/s23156722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/17/2023] [Revised: 07/06/2023] [Accepted: 07/18/2023] [Indexed: 08/13/2023]
Abstract
After surgery for ovarian cancer or colorectal cancer, residual tumors are left around. A practical way to treat residual tumors is to destroy them with heat by injecting high-temperature drugs into the abdominal cavity. The injected medicinal substances are induced to flow out of the abdominal cavity; then, the spilled drug flows back into the abdominal cavity through feedback. During this process, the heat starts to decrease; thus, the treatment performance reduces. To overcome this problem, this study compares and assesses the temperature needed to maintain the heat for treatment and transmits a command signal to the heat exchanger through a look-up table (LUT). When the temperature decreases during the circulation of medications leaking out of the abdominal cavity, the LUT transmits a control signal (Tp) to the heat exchanger, which increases or vice versa. However, if the temperature (To) is within the treatment range, the LUT sends a Ts signal to the heat exchanger. This principle generates a pulse signal for the temperature difference (Tdif) in TC by comparing and determining the temperature (To) of the substance flowing out of the abdominal cavity with the reference temperature (Tref) through the temperature comparator (TC). At this time, if the signal is 41 °C or less, the LUT generates (heats) a Tp signal so that the temperature of the heat exchanger can be maintained in the range of 41 °C to 43 °C. If the Tdif is 44 °C or higher, the LUT generates (cools) the Ta signal and maintains the temperature of the heat exchanger at 41-43 °C. If the Tdif is maintained at 41-43 °C, the LUT generates a Tx signal to stop the system performance. At this time, the TC operation performance and Tdif generation process for comparing and determining the signal of To and Tref for drugs leaking out of the abdominal cavity is very important. It was observed that the faster the response signal, the lower the comparison and judgment error was; therefore, the response signal was confirmed to be 0.209 μs. The proposed method can guarantee rapid/accurate/safe treatment and automatically induce temperature adjustment; thus, it could be applied to the field of surgery.
Collapse
Affiliation(s)
- Kicheol Yoon
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 Beon-gil, Namdong-daero, Namdong-gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.)
- Department of Biomedical Engineering, College of Medicine, Gachon University, 38–13, 3 Beon-gil, Dokjom-ro 3, Namdong-gu, Incheon 21565, Republic of Korea
| | - Sangyun Lee
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 Beon-gil, Namdong-daero, Namdong-gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.)
- Department of Biomedical Engineering, College of Medicine, Gachon University, 38–13, 3 Beon-gil, Dokjom-ro 3, Namdong-gu, Incheon 21565, Republic of Korea
| | - Tae-Hyeon Lee
- Department of Electronic Engineering, Gyeonggi University of Science and Technology, Gyeonggigwagi-dearo 269, Siheung City 15073, Gyeonggi-do, Republic of Korea;
| | - Kwang Gi Kim
- Medical Devices R&D Center, Gachon University Gil Medical Center, 21, 774 Beon-gil, Namdong-daero, Namdong-gu, Incheon 21565, Republic of Korea; (K.Y.); (S.L.)
- Department of Biomedical Engineering, College of Medicine, Gachon University, 38–13, 3 Beon-gil, Dokjom-ro 3, Namdong-gu, Incheon 21565, Republic of Korea
- Department of Biomedical Engineering, College of Health Science, Gachon University, 191 Hambak-moero, Yeonsu-gu, Incheon 21936, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, 38–13, 3 Beon-gil, Dokjom-ro, Namdong-gu, Incheon 21565, Republic of Korea
| |
Collapse
|
93
|
Reyes-Aldasoro CC. Modelling the Tumour Microenvironment, but What Exactly Do We Mean by "Model"? Cancers (Basel) 2023; 15:3796. [PMID: 37568612 PMCID: PMC10416922 DOI: 10.3390/cancers15153796] [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: 06/28/2023] [Revised: 07/19/2023] [Accepted: 07/25/2023] [Indexed: 08/13/2023] Open
Abstract
The Oxford English Dictionary includes 17 definitions for the word "model" as a noun and another 11 as a verb. Therefore, context is necessary to understand the meaning of the word model. For instance, "model railways" refer to replicas of railways and trains at a smaller scale and a "model student" refers to an exemplary individual. In some cases, a specific context, like cancer research, may not be sufficient to provide one specific meaning for model. Even if the context is narrowed, specifically, to research related to the tumour microenvironment, "model" can be understood in a wide variety of ways, from an animal model to a mathematical expression. This paper presents a review of different "models" of the tumour microenvironment, as grouped by different definitions of the word into four categories: model organisms, in vitro models, mathematical models and computational models. Then, the frequencies of different meanings of the word "model" related to the tumour microenvironment are measured from numbers of entries in the MEDLINE database of the United States National Library of Medicine at the National Institutes of Health. The frequencies of the main components of the microenvironment and the organ-related cancers modelled are also assessed quantitatively with specific keywords. Whilst animal models, particularly xenografts and mouse models, are the most commonly used "models", the number of these entries has been slowly decreasing. Mathematical models, as well as prognostic and risk models, follow in frequency, and these have been growing in use.
Collapse
|
94
|
Park HS, Chong Y, Lee Y, Yim K, Seo KJ, Hwang G, Kim D, Gong G, Cho NH, Yoo CW, Choi HJ. Deep Learning-Based Computational Cytopathologic Diagnosis of Metastatic Breast Carcinoma in Pleural Fluid. Cells 2023; 12:1847. [PMID: 37508511 PMCID: PMC10377793 DOI: 10.3390/cells12141847] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 07/07/2023] [Accepted: 07/11/2023] [Indexed: 07/30/2023] Open
Abstract
A Pleural effusion cytology is vital for treating metastatic breast cancer; however, concerns have arisen regarding the low accuracy and inter-observer variability in cytologic diagnosis. Although artificial intelligence-based image analysis has shown promise in cytopathology research, its application in diagnosing breast cancer in pleural fluid remains unexplored. To overcome these limitations, we evaluate the diagnostic accuracy of an artificial intelligence-based model using a large collection of cytopathological slides, to detect the malignant pleural effusion cytology associated with breast cancer. This study includes a total of 569 cytological slides of malignant pleural effusion of metastatic breast cancer from various institutions. We extracted 34,221 augmented image patches from whole-slide images and trained and validated a deep convolutional neural network model (DCNN) (Inception-ResNet-V2) with the images. Using this model, we classified 845 randomly selected patches, which were reviewed by three pathologists to compare their accuracy. The DCNN model outperforms the pathologists by demonstrating higher accuracy, sensitivity, and specificity compared to the pathologists (81.1% vs. 68.7%, 95.0% vs. 72.5%, and 98.6% vs. 88.9%, respectively). The pathologists reviewed the discordant cases of DCNN. After re-examination, the average accuracy, sensitivity, and specificity of the pathologists improved to 87.9, 80.2, and 95.7%, respectively. This study shows that DCNN can accurately diagnose malignant pleural effusion cytology in breast cancer and has the potential to support pathologists.
Collapse
Affiliation(s)
- Hong Sik Park
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Yosep Chong
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Yujin Lee
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Kwangil Yim
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Kyung Jin Seo
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| | - Gisu Hwang
- AI Team, DeepNoid Inc., Seoul 08376, Republic of Korea
| | - Dahyeon Kim
- AI Team, DeepNoid Inc., Seoul 08376, Republic of Korea
| | - Gyungyub Gong
- Department of Pathology, Asan Medical Center, Seoul 05505, Republic of Korea
| | - Nam Hoon Cho
- Department of Pathology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Chong Woo Yoo
- Department of Pathology, National Cancer Center, Ilsan, Goyang-si 10408, Gyeonggi-do, Republic of Korea
| | - Hyun Joo Choi
- Department of Hospital Pathology, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea
| |
Collapse
|
95
|
Ferro M, Falagario UG, Barone B, Maggi M, Crocetto F, Busetto GM, Giudice FD, Terracciano D, Lucarelli G, Lasorsa F, Catellani M, Brescia A, Mistretta FA, Luzzago S, Piccinelli ML, Vartolomei MD, Jereczek-Fossa BA, Musi G, Montanari E, Cobelli OD, Tataru OS. Artificial Intelligence in the Advanced Diagnosis of Bladder Cancer-Comprehensive Literature Review and Future Advancement. Diagnostics (Basel) 2023; 13:2308. [PMID: 37443700 DOI: 10.3390/diagnostics13132308] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 07/03/2023] [Accepted: 07/05/2023] [Indexed: 07/15/2023] Open
Abstract
Artificial intelligence is highly regarded as the most promising future technology that will have a great impact on healthcare across all specialties. Its subsets, machine learning, deep learning, and artificial neural networks, are able to automatically learn from massive amounts of data and can improve the prediction algorithms to enhance their performance. This area is still under development, but the latest evidence shows great potential in the diagnosis, prognosis, and treatment of urological diseases, including bladder cancer, which are currently using old prediction tools and historical nomograms. This review focuses on highly significant and comprehensive literature evidence of artificial intelligence in the management of bladder cancer and investigates the near introduction in clinical practice.
Collapse
Affiliation(s)
- Matteo Ferro
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
| | - Ugo Giovanni Falagario
- Department of Urology and Organ Transplantation, University of Foggia, 71121 Foggia, Italy
| | - Biagio Barone
- Urology Unit, Department of Surgical Sciences, AORN Sant'Anna e San Sebastiano, 81100 Caserta, Italy
| | - Martina Maggi
- Department of Maternal Infant and Urologic Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy
| | - Felice Crocetto
- Department of Neurosciences and Reproductive Sciences and Odontostomatology, University of Naples Federico II, 80131 Naples, Italy
| | - Gian Maria Busetto
- Department of Urology and Organ Transplantation, University of Foggia, 71121 Foggia, Italy
| | - Francesco Del Giudice
- Department of Maternal Infant and Urologic Sciences, Policlinico Umberto I Hospital, Sapienza University of Rome, 00161 Rome, Italy
| | - Daniela Terracciano
- Department of Translational Medical Sciences, University of Naples "Federico II", 80131 Naples, Italy
| | - Giuseppe Lucarelli
- Urology, Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Francesco Lasorsa
- Urology, Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation, University of Bari, 70124 Bari, Italy
| | - Michele Catellani
- Department of Urology, ASST Papa Giovanni XXIII, 24127 Bergamo, Italy
| | - Antonio Brescia
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
| | - Francesco Alessandro Mistretta
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Stefano Luzzago
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Mattia Luca Piccinelli
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
| | | | - Barbara Alicja Jereczek-Fossa
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
- Division of Radiation Oncology, IEO-European Institute of Oncology IRCCS, 20141 Milan, Italy
| | - Gennaro Musi
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Emanuele Montanari
- Department of Urology, Foundation IRCCS Ca' Granda-Ospedale Maggiore Policlinico, 20122 Milan, Italy
- Department of Clinical Sciences and Community Health, University of Milan, 20122 Milan, Italy
| | - Ottavio de Cobelli
- Department of Urology, IEO-European Institute of Oncology, IRCCS-Istituto di Ricovero e Cura a Carattere Scientifico, 20141 Milan, Italy
- Department of Oncology and Hemato-Oncology, University of Milan, 20122 Milan, Italy
| | - Octavian Sabin Tataru
- Department of Simulation Applied in Medicine, George Emil Palade University of Medicine, Pharmacy, Science and Technology of Târgu Mures, 540142 Târgu Mures, Romania
| |
Collapse
|
96
|
Pyo JS, Kim NY, Min KW, Kang DW. Significance of Tumor-Stroma Ratio (TSR) in Predicting Outcomes of Malignant Tumors. MEDICINA (KAUNAS, LITHUANIA) 2023; 59:1258. [PMID: 37512068 PMCID: PMC10384099 DOI: 10.3390/medicina59071258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 06/17/2023] [Accepted: 06/27/2023] [Indexed: 07/30/2023]
Abstract
Background and Objectives: The present study aimed to elucidate the distribution and the prognostic implications of tumor-stroma ratio (TSR) in various malignant tumors through a meta-analysis. Materials and Methods: This meta-analysis included 51 eligible studies with information for overall survival (OS) or disease-free survival (DFS), according to TSR. In addition, subgroup analysis was performed based on criteria for high TSR. Results: The estimated rate of high TSR was 0.605 (95% confidence interval (CI) 0.565-0.644) in overall malignant tumors. The rates of high TSR ranged from 0.276 to 0.865. The highest rate of high TSR was found in endometrial cancer (0.865, 95% CI 0.827-0.895). The estimated high TSR rates of colorectal, esophageal, and stomach cancers were 0.622, 0.529, and 0.448, respectively. In overall cases, patients with high TSR had better OS and DFS than those with low TSR (hazard ratio (HR) 0.631, 95% CI 0.542-0.734, and HR 0.564, 95% CI 0.0.476-0.669, respectively). Significant correlations with OS were found in the breast, cervical, colorectal, esophagus, head and neck, ovary, stomach, and urinary tract cancers. In addition, there were significant correlations of DFS in breast, cervical, colorectal, esophageal, larynx, lung, and stomach cancers. In endometrial cancers, high TSR was significantly correlated with worse OS and DFS. Conclusions: The rate of high TSR was different in various malignant tumors. TSR can be useful for predicting prognosis through a routine microscopic examination of malignant tumors.
Collapse
Affiliation(s)
- Jung-Soo Pyo
- Department of Pathology, Uijeongbu Eulji University Hospital, Eulji University School of Medicine, Uijeongbu-si 11759, Republic of Korea
| | - Nae Yu Kim
- Department of Internal Medicine, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Uijeongbu-si 11759, Republic of Korea
| | - Kyueng-Whan Min
- Department of Pathology, Uijeongbu Eulji University Hospital, Eulji University School of Medicine, Uijeongbu-si 11759, Republic of Korea
| | - Dong-Wook Kang
- Department of Pathology, Chungnam National University Sejong Hospital, 20 Bodeum 7-ro, Sejong 30099, Republic of Korea
- Department of Pathology, Chungnam National University School of Medicine, 266 Munhwa Street, Daejeon 35015, Republic of Korea
| |
Collapse
|
97
|
Bae H. Biomimicry Industry and Patent Trends. Biomimetics (Basel) 2023; 8:288. [PMID: 37504176 PMCID: PMC10807642 DOI: 10.3390/biomimetics8030288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 06/21/2023] [Accepted: 06/29/2023] [Indexed: 07/29/2023] Open
Abstract
This study examines the current technological level and industrial/technical trends in the field of biomimicry technology, as well as recent technological and research and development trends. Patent analysis was conducted, focusing on technology that uses design elements and biological/ecological characteristics to provide solutions to technological problems. The technological scope of the analysis included the field of technologies and materials that apply to the conditions found in ecology, as well as robot machines and devices designed to mimic certain animals and ecological elements. The search for patents was conducted in Korea, the United States, Japan, and Europe from 1975 to 2021, resulting in a total of 8278 raw data cases, from which 940 valid patents were selected. The percentage of patent document and the status of both domestic and foreign applicants varied among the countries of Korea, the United States, Japan, and Europe. Based on the results of the patent analysis, it was found that biomimicry technology is in a growth phase that is expected to continue in the future and that Korea and the United States are leading the development of this technology.
Collapse
Affiliation(s)
- Haejin Bae
- Ecological Technology Research Team, Division of Ecological Application Research, National Institute of Ecology, Seocheon-gun 33657, Republic of Korea
| |
Collapse
|
98
|
Chong Y, Park G, Cha HJ, Kim HJ, Kang CS, Abdul-Ghafar J, Lee SS. A stepwise approach to fine needle aspiration cytology of lymph nodes. J Pathol Transl Med 2023; 57:196-207. [PMID: 37460394 PMCID: PMC10369138 DOI: 10.4132/jptm.2023.06.12] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 06/12/2023] [Indexed: 07/28/2023] Open
Abstract
The cytological diagnosis of lymph node lesions is extremely challenging because of the diverse diseases that cause lymph node enlargement, including both benign and malignant or metastatic lymphoid lesions. Furthermore, the cytological findings of different lesions often resemble one another. A stepwise diagnostic approach is essential for a comprehensive diagnosis that combines: clinical findings, including age, sex, site, multiplicity, and ultrasonography findings; low-power reactive, metastatic, and lymphoma patterns; high-power population patterns, including two populations of continuous range, small monotonous pattern and large monotonous pattern; and disease-specific diagnostic clues including granulomas and lymphoglandular granules. It is also important to remember the histological features of each diagnostic category that are common in lymph node cytology and to compare them with cytological findings. It is also essential to identify a few categories of diagnostic pitfalls that often resemble lymphomas and easily lead to misdiagnosis, particularly in malignant small round cell tumors, poorly differentiated squamous cell carcinomas, and nasopharyngeal undifferentiated carcinoma. Herein, we review a stepwise approach for fine needle aspiration cytology of lymphoid diseases and suggest a diagnostic algorithm that uses this approach and the Sydney classification system.
Collapse
Affiliation(s)
- Yosep Chong
- Department of Hospital Pathology, The Catholic University of Korea, College of Medicine, Seoul, Korea
| | - Gyeongsin Park
- Department of Hospital Pathology, The Catholic University of Korea, College of Medicine, Seoul, Korea
| | - Hee Jeong Cha
- Department of Pathology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, Korea
| | - Hyun-Jung Kim
- Department of Pathology, Inje University Sanggye Paik Hospital, Seoul, Korea
| | - Chang Suk Kang
- Department of Pathology, Samkwang Medical Laboratories, Seoul, Korea
| | - Jamshid Abdul-Ghafar
- Department of Hospital Pathology, The Catholic University of Korea, College of Medicine, Seoul, Korea
| | - Seung-Sook Lee
- Department of Pathology, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| |
Collapse
|
99
|
Gedefaw L, Liu CF, Ip RKL, Tse HF, Yeung MHY, Yip SP, Huang CL. Artificial Intelligence-Assisted Diagnostic Cytology and Genomic Testing for Hematologic Disorders. Cells 2023; 12:1755. [PMID: 37443789 PMCID: PMC10340428 DOI: 10.3390/cells12131755] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/21/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
Artificial intelligence (AI) is a rapidly evolving field of computer science that involves the development of computational programs that can mimic human intelligence. In particular, machine learning and deep learning models have enabled the identification and grouping of patterns within data, leading to the development of AI systems that have been applied in various areas of hematology, including digital pathology, alpha thalassemia patient screening, cytogenetics, immunophenotyping, and sequencing. These AI-assisted methods have shown promise in improving diagnostic accuracy and efficiency, identifying novel biomarkers, and predicting treatment outcomes. However, limitations such as limited databases, lack of validation and standardization, systematic errors, and bias prevent AI from completely replacing manual diagnosis in hematology. In addition, the processing of large amounts of patient data and personal information by AI poses potential data privacy issues, necessitating the development of regulations to evaluate AI systems and address ethical concerns in clinical AI systems. Nonetheless, with continued research and development, AI has the potential to revolutionize the field of hematology and improve patient outcomes. To fully realize this potential, however, the challenges facing AI in hematology must be addressed and overcome.
Collapse
Affiliation(s)
- Lealem Gedefaw
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (C.-F.L.); (M.H.Y.Y.)
| | - Chia-Fei Liu
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (C.-F.L.); (M.H.Y.Y.)
| | - Rosalina Ka Ling Ip
- Department of Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China; (R.K.L.I.); (H.-F.T.)
| | - Hing-Fung Tse
- Department of Pathology, Pamela Youde Nethersole Eastern Hospital, Hong Kong, China; (R.K.L.I.); (H.-F.T.)
| | - Martin Ho Yin Yeung
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (C.-F.L.); (M.H.Y.Y.)
| | - Shea Ping Yip
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (C.-F.L.); (M.H.Y.Y.)
| | - Chien-Ling Huang
- Department of Health Technology and Informatics, The Hong Kong Polytechnic University, Hong Kong, China; (L.G.); (C.-F.L.); (M.H.Y.Y.)
| |
Collapse
|
100
|
Gamsiz Uzun ED. Introducing JMIR Bioinformatics and Biotechnology: A Platform for Interdisciplinary Collaboration and Cutting-Edge Research. JMIR BIOINFORMATICS AND BIOTECHNOLOGY 2023; 4:e48631. [PMID: 38935954 PMCID: PMC11135224 DOI: 10.2196/48631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Accepted: 05/04/2023] [Indexed: 06/29/2024]
Abstract
JMIR Bioinformatics and Biotechnology supports interdisciplinary research and welcomes contributions that push the boundaries of bioinformatics, genomics, artificial intelligence, and pathology informatics.
Collapse
Affiliation(s)
- Ece Dilber Gamsiz Uzun
- Department of Pathology and Laboratory Medicine, Rhode Island Hospital, Providence, RI, United States
- Department of Pathology and Laboratory Medicine, Brown University, Providence, RI, United States
| |
Collapse
|