1
|
Prabhu S, Prasad K, Hoang T, Lu X. MultiSCCHisto-Net-KD: A deep network for multi-organ explainable squamous cell carcinoma diagnosis with knowledge distillation. Comput Biol Med 2025; 185:109469. [PMID: 39662318 DOI: 10.1016/j.compbiomed.2024.109469] [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: 05/13/2024] [Revised: 10/07/2024] [Accepted: 11/21/2024] [Indexed: 12/13/2024]
Abstract
Squamous cell carcinoma is a prevalent cancer type that affects various organs in the human body. Manual analysis for detecting squamous cell carcinoma in histopathological images is time-consuming and may be subjective. Squamous cell carcinoma diagnosis is typically based on the differences in the architectural arrangement of squamous epithelial layers and the presence of keratinization. However, the existing literature has predominantly concentrated on identifying cellular irregularities with high magnification images and considering specific organs of squamous cell carcinoma origin. In contrast, relatively little attention has been given to recognizing structural abnormalities observable at low magnification images. In this paper, we consider squamous cell carcinoma histopathological images across different organs of origin captured at low magnification and these images are gathered from various centers to develop a robust model. We propose a novel deep neural network model (MultiSCCHisto-Net) that can detect squamous cell carcinoma of any organ irrespective of organ of origin. In addition, deep neural networks used for histopathological image analysis typically have many parameters, making them computationally expensive. To address this research gap, we incorporate knowledge distillation, which compresses knowledge from a complex teacher model (MultiSCCHisto-Net) into a smaller student model (MultiSCCHisto-Net-KD) while preserving performance and enhancing the generalization of the student model by learning from the teacher's intermediate layer features. Moreover, an explainable deep learning technique called gradient-weighted class activation mapping is incorporated to highlight the image areas that help to classify the sample into particular classes. This explainability significantly enhances our confidence in the proposed model outcomes. We evaluate the model's robustness using private multi-centric and publicly available datasets. Our results show that accuracy rates of 97% and 93% are achieved on private and public datasets, respectively, surpassing the performance of state-of-the-art models.
Collapse
Affiliation(s)
- Swathi Prabhu
- Department of Information and Communication Technology, Manipal Institute of Technology, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Keerthana Prasad
- Manipal School of Information Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India.
| | - Thuong Hoang
- School of Information Technology, Faculty of Science Engineering and Built Environment, Deakin University, Geelong, VIC 3220, Victoria, Australia
| | - Xuequan Lu
- Department of Computer Science and IT, La Trobe University, Melbourne, VIC 3086, Victoria, Australia
| |
Collapse
|
2
|
Wang X, Zhao J, Marostica E, Yuan W, Jin J, Zhang J, Li R, Tang H, Wang K, Li Y, Wang F, Peng Y, Zhu J, Zhang J, Jackson CR, Zhang J, Dillon D, Lin NU, Sholl L, Denize T, Meredith D, Ligon KL, Signoretti S, Ogino S, Golden JA, Nasrallah MP, Han X, Yang S, Yu KH. A pathology foundation model for cancer diagnosis and prognosis prediction. Nature 2024; 634:970-978. [PMID: 39232164 DOI: 10.1038/s41586-024-07894-z] [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: 11/16/2023] [Accepted: 08/01/2024] [Indexed: 09/06/2024]
Abstract
Histopathology image evaluation is indispensable for cancer diagnoses and subtype classification. Standard artificial intelligence methods for histopathology image analyses have focused on optimizing specialized models for each diagnostic task1,2. Although such methods have achieved some success, they often have limited generalizability to images generated by different digitization protocols or samples collected from different populations3. Here, to address this challenge, we devised the Clinical Histopathology Imaging Evaluation Foundation (CHIEF) model, a general-purpose weakly supervised machine learning framework to extract pathology imaging features for systematic cancer evaluation. CHIEF leverages two complementary pretraining methods to extract diverse pathology representations: unsupervised pretraining for tile-level feature identification and weakly supervised pretraining for whole-slide pattern recognition. We developed CHIEF using 60,530 whole-slide images spanning 19 anatomical sites. Through pretraining on 44 terabytes of high-resolution pathology imaging datasets, CHIEF extracted microscopic representations useful for cancer cell detection, tumour origin identification, molecular profile characterization and prognostic prediction. We successfully validated CHIEF using 19,491 whole-slide images from 32 independent slide sets collected from 24 hospitals and cohorts internationally. Overall, CHIEF outperformed the state-of-the-art deep learning methods by up to 36.1%, showing its ability to address domain shifts observed in samples from diverse populations and processed by different slide preparation methods. CHIEF provides a generalizable foundation for efficient digital pathology evaluation for patients with cancer.
Collapse
Affiliation(s)
- Xiyue Wang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Junhan Zhao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA
| | - Wei Yuan
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Jietian Jin
- Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jiayu Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Ruijiang Li
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | - Hongping Tang
- Department of Pathology, Shenzhen Maternity & Child Healthcare Hospital, Shenzhen, China
| | - Kanran Wang
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yu Li
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China
| | - Fang Wang
- Department of Pathology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, China
| | - Yulong Peng
- Department of Pathology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Junyou Zhu
- Department of Burn, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jing Zhang
- College of Biomedical Engineering, Sichuan University, Chengdu, China
| | - Christopher R Jackson
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Pathology and Laboratory Medicine, Pennsylvania State University, Hummelstown, PA, USA
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | | | - Deborah Dillon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Nancy U Lin
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Lynette Sholl
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Thomas Denize
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - David Meredith
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Keith L Ligon
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Sabina Signoretti
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Shuji Ogino
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA, USA
| | | | - Sen Yang
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA.
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
- Harvard Data Science Initiative, Harvard University, Cambridge, MA, USA.
| |
Collapse
|
3
|
Hoang DT, Dinstag G, Shulman ED, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. A deep-learning framework to predict cancer treatment response from histopathology images through imputed transcriptomics. NATURE CANCER 2024; 5:1305-1317. [PMID: 38961276 DOI: 10.1038/s43018-024-00793-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Accepted: 06/06/2024] [Indexed: 07/05/2024]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an indirect two-step approach consisting of (1) DeepPT, a deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response to targeted and immune therapies from the inferred expression values. We show that DeepPT successfully predicts transcriptomics in all 16 The Cancer Genome Atlas cohorts tested and generalizes well to two independent datasets. ENLIGHT-DeepPT successfully predicts true responders in five independent patient cohorts involving four different treatments spanning six cancer types, with an overall odds ratio of 2.28 and a 39.5% increased response rate among predicted responders versus the baseline rate. Notably, its prediction accuracy, obtained without any training on the treatment data, is comparable to that achieved by directly predicting the response from the images, which requires specific training on the treatment evaluation cohorts.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | | | - Eldad D Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Leandro C Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- School of Clinical Medicine, University of Cambridge, Cambridge, UK
| | - James L Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, Australian Capital Territory, Australia.
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| |
Collapse
|
4
|
Dhruba SR, Sahni S, Wang B, Wu D, Rajagopal PS, Schmidt Y, Shulman ED, Sinha S, Sammut SJ, Caldas C, Wang K, Ruppin E. The expression patterns of different cell types and their interactions in the tumor microenvironment are predictive of breast cancer patient response to neoadjuvant chemotherapy. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.14.598770. [PMID: 39372749 PMCID: PMC11451622 DOI: 10.1101/2024.06.14.598770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/08/2024]
Abstract
The tumor microenvironment (TME) is a complex ecosystem of diverse cell types whose interactions govern tumor growth and clinical outcome. While the TME's impact on immunotherapy has been extensively studied, its role in chemotherapy response remains less explored. To address this, we developed DECODEM (DEcoupling Cell-type-specific Outcomes using DEconvolution and Machine learning), a generic computational framework leveraging cellular deconvolution of bulk transcriptomics to associate the gene expression of individual cell types in the TME with clinical response. Employing DECODEM to analyze the gene expression of breast cancer (BC) patients treated with neoadjuvant chemotherapy, we find that the gene expression of specific immune cells (myeloid, plasmablasts, B-cells) and stromal cells (endothelial, normal epithelial, CAFs) are highly predictive of chemotherapy response, going beyond that of the malignant cells. These findings are further tested and validated in a single-cell cohort of triple negative breast cancer. To investigate the possible role of immune cell-cell interactions (CCIs) in mediating chemotherapy response, we extended DECODEM to DECODEMi to identify such CCIs, validated in single-cell data. Our findings highlight the importance of active pre-treatment immune infiltration for chemotherapy success. The tools developed here are made publicly available and are applicable for studying the role of the TME in mediating response from readily available bulk tumor expression in a wide range of cancer treatments and indications.
Collapse
Affiliation(s)
- Saugato Rahman Dhruba
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sahil Sahni
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Binbin Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Di Wu
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Padma Sheila Rajagopal
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- Women’s Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yael Schmidt
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eldad D. Shulman
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI-Designated Cancer Center, Sanford Burnham Prebys Medical Discovery Institute, San Diego, CA, USA
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, UK
- The Royal Marsden Hospital NHS Foundation Trust, London, UK
| | - Carlos Caldas
- Institute of Metabolic Science, School of Clinical Medicine, University of Cambridge, Cambridge, UK
- Department of Clinical Biochemistry, University of Cambridge, Cambridge, UK
| | - Kun Wang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
5
|
Levenson RM, Singh Y, Rieck B, Hathaway QA, Farrelly C, Rozenblit J, Prasanna P, Erickson B, Choudhary A, Carlsson G, Sarkar D. Advancing Precision Medicine: Algebraic Topology and Differential Geometry in Radiology and Computational Pathology. J Transl Med 2024; 104:102060. [PMID: 38626875 DOI: 10.1016/j.labinv.2024.102060] [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: 12/15/2023] [Revised: 04/08/2024] [Accepted: 04/10/2024] [Indexed: 05/19/2024] Open
Abstract
Precision medicine aims to provide personalized care based on individual patient characteristics, rather than guideline-directed therapies for groups of diseases or patient demographics. Images-both radiology- and pathology-derived-are a major source of information on presence, type, and status of disease. Exploring the mathematical relationship of pixels in medical imaging ("radiomics") and cellular-scale structures in digital pathology slides ("pathomics") offers powerful tools for extracting both qualitative and, increasingly, quantitative data. These analytical approaches, however, may be significantly enhanced by applying additional methods arising from fields of mathematics such as differential geometry and algebraic topology that remain underexplored in this context. Geometry's strength lies in its ability to provide precise local measurements, such as curvature, that can be crucial for identifying abnormalities at multiple spatial levels. These measurements can augment the quantitative features extracted in conventional radiomics, leading to more nuanced diagnostics. By contrast, topology serves as a robust shape descriptor, capturing essential features such as connected components and holes. The field of topological data analysis was initially founded to explore the shape of data, with functional network connectivity in the brain being a prominent example. Increasingly, its tools are now being used to explore organizational patterns of physical structures in medical images and digitized pathology slides. By leveraging tools from both differential geometry and algebraic topology, researchers and clinicians may be able to obtain a more comprehensive, multi-layered understanding of medical images and contribute to precision medicine's armamentarium.
Collapse
Affiliation(s)
- Richard M Levenson
- Department of Pathology and Laboratory Medicine, University of California Davis, Davis, California.
| | - Yashbir Singh
- Department of Radiology, Mayo Clinic, Rochester, Minnesota.
| | - Bastian Rieck
- Helmholtz Munich and Technical University of Munich, Munich, Germany
| | - Quincy A Hathaway
- Department of Medical Education, West Virginia University, Morgantown, West Virginia
| | | | | | - Prateek Prasanna
- Department of Biomedical Informatics, Stony Brook University, Stony Brook, New York
| | | | | | - Gunnar Carlsson
- Department of Mathematics, Stanford University, Stanford, California
| | - Deepa Sarkar
- Institute of Genomic Health, Ichan school of Medicine, Mount Sinai, New York
| |
Collapse
|
6
|
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
|
7
|
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
|
8
|
Sykes JR, Denby KJ, Franks DW. Computer vision for plant pathology: A review with examples from cocoa agriculture. APPLICATIONS IN PLANT SCIENCES 2024; 12:e11559. [PMID: 38638617 PMCID: PMC11022223 DOI: 10.1002/aps3.11559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 07/04/2023] [Accepted: 07/13/2023] [Indexed: 04/20/2024]
Abstract
Plant pathogens can decimate crops and render the local cultivation of a species unprofitable. In extreme cases this has caused famine and economic collapse. Timing is vital in treating crop diseases, and the use of computer vision for precise disease detection and timing of pesticide application is gaining popularity. Computer vision can reduce labour costs, prevent misdiagnosis of disease, and prevent misapplication of pesticides. Pesticide misapplication is both financially costly and can exacerbate pesticide resistance and pollution. Here, we review the application and development of computer vision and machine learning methods for the detection of plant disease. This review goes beyond the scope of previous works to discuss important technical concepts and considerations when applying computer vision to plant pathology. We present new case studies on adapting standard computer vision methods and review techniques for acquiring training data, the use of diagnostic tools from biology, and the inspection of informative features. In addition to an in-depth discussion of convolutional neural networks (CNNs) and transformers, we also highlight the strengths of methods such as support vector machines and evolved neural networks. We discuss the benefits of carefully curating training data and consider situations where less computationally expensive techniques are advantageous. This includes a comparison of popular model architectures and a guide to their implementation.
Collapse
Affiliation(s)
- Jamie R. Sykes
- Department of Computer ScienceUniversity of YorkDeramore Lane, YorkYO10 5GHYorkshireUnited Kingdom
| | - Katherine J. Denby
- Centre for Novel Agricultural Products, Department of BiologyUniversity of YorkWentworth Way, YorkYO10 5DDYorkshireUnited Kingdom
| | - Daniel W. Franks
- Department of Computer ScienceUniversity of YorkDeramore Lane, YorkYO10 5GHYorkshireUnited Kingdom
- Department of BiologyUniversity of YorkWentworth Way, YorkYO10 5DDYorkshireUnited Kingdom
| |
Collapse
|
9
|
Vocino Trucco G, Righi L, Volante M, Papotti M. Updates on lung neuroendocrine neoplasm classification. Histopathology 2024; 84:67-85. [PMID: 37794655 DOI: 10.1111/his.15058] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2023] [Revised: 09/08/2023] [Accepted: 09/14/2023] [Indexed: 10/06/2023]
Abstract
Lung neuroendocrine neoplasms (NENs) are a heterogeneous group of pulmonary neoplasms showing different morphological patterns and clinical and biological characteristics. The World Health Organisation (WHO) classification of lung NENs has been recently updated as part of the broader attempt to uniform the classification of NENs. This much-needed update has come at a time when insights from seminal molecular characterisation studies revolutionised our understanding of the biological and pathological architecture of lung NENs, paving the way for the development of novel diagnostic techniques, prognostic factors and therapeutic approaches. In this challenging and rapidly evolving landscape, the relevance of the 2021 WHO classification has been recently questioned, particularly in terms of its morphology-orientated approach and its prognostic implications. Here, we provide a state-of-the-art review on the contemporary understanding of pulmonary NEN morphology and the potential contribution of artificial intelligence, the advances in NEN molecular profiling with their impact on the classification system and, finally, the key current and upcoming prognostic factors.
Collapse
Affiliation(s)
| | - Luisella Righi
- Department of Oncology, University of Turin, Turin, Italy
| | - Marco Volante
- Department of Oncology, University of Turin, Turin, Italy
| | - Mauro Papotti
- Department of Oncology, University of Turin, Turin, Italy
| |
Collapse
|
10
|
Wagner SJ, Matek C, Shetab Boushehri S, Boxberg M, Lamm L, Sadafi A, Winter DJE, Marr C, Peng T. Built to Last? Reproducibility and Reusability of Deep Learning Algorithms in Computational Pathology. Mod Pathol 2024; 37:100350. [PMID: 37827448 DOI: 10.1016/j.modpat.2023.100350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2023] [Revised: 10/02/2023] [Accepted: 10/03/2023] [Indexed: 10/14/2023]
Abstract
Recent progress in computational pathology has been driven by deep learning. While code and data availability are essential to reproduce findings from preceding publications, ensuring a deep learning model's reusability is more challenging. For that, the codebase should be well-documented and easy to integrate into existing workflows and models should be robust toward noise and generalizable toward data from different sources. Strikingly, only a few computational pathology algorithms have been reused by other researchers so far, let alone employed in a clinical setting. To assess the current state of reproducibility and reusability of computational pathology algorithms, we evaluated peer-reviewed articles available in PubMed, published between January 2019 and March 2021, in 5 use cases: stain normalization; tissue type segmentation; evaluation of cell-level features; genetic alteration prediction; and inference of grading, staging, and prognostic information. We compiled criteria for data and code availability and statistical result analysis and assessed them in 160 publications. We found that only one-quarter (41 of 160 publications) made code publicly available. Among these 41 studies, three-quarters (30 of 41) analyzed their results statistically, half of them (20 of 41) released their trained model weights, and approximately a third (16 of 41) used an independent cohort for evaluation. Our review is intended for both pathologists interested in deep learning and researchers applying algorithms to computational pathology challenges. We provide a detailed overview of publications with published code in the field, list reusable data handling tools, and provide criteria for reproducibility and reusability.
Collapse
Affiliation(s)
- Sophia J Wagner
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Computation, Information and Technology, Technical University of Munich, Garching, Germany
| | - Christian Matek
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Institute of Pathology, University Hospital Erlangen, Erlangen, Germany
| | - Sayedali Shetab Boushehri
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Data & Analytics (D&A), Roche Pharma Research and Early Development (pRED), Roche Innovation Center Munich, Germany
| | - Melanie Boxberg
- Institute of Pathology, Technical University Munich, Munich, Germany; Institute of Pathology Munich-North, Munich, Germany
| | - Lorenz Lamm
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; Helmholtz Pioneer Campus, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Ario Sadafi
- School of Computation, Information and Technology, Technical University of Munich, Garching, Germany; Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany
| | - Dominik J E Winter
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany; School of Life Sciences, Technical University of Munich, Weihenstephan, Germany
| | - Carsten Marr
- Institute of AI for Health, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| | - Tingying Peng
- Helmholtz AI, Helmholtz Munich-German Research Center for Environmental Health, Neuherberg, Germany.
| |
Collapse
|
11
|
Xiong DD, He RQ, Huang ZG, Wu KJ, Mo YY, Liang Y, Yang DP, Wu YH, Tang ZQ, Liao ZT, Chen G. Global bibliometric mapping of the research trends in artificial intelligence-based digital pathology for lung cancer over the past two decades. Digit Health 2024; 10:20552076241277735. [PMID: 39233894 PMCID: PMC11372859 DOI: 10.1177/20552076241277735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Accepted: 08/08/2024] [Indexed: 09/06/2024] Open
Abstract
Background and Objective The rapid development of computer technology has led to a revolutionary transformation in artificial intelligence (AI)-assisted healthcare. The integration of whole-slide imaging technology with AI algorithms has facilitated the development of digital pathology for lung cancer (LC). However, there is a lack of comprehensive scientometric analysis in this field. Methods A bibliometric analysis was conducted on 197 publications related to digital pathology in LC from 502 institutions across 39 countries, published in 97 academic journals in the Web of Science Core Collection between 2004 and 2023. Results Our analysis has identified the United States and China as the primary research nations in the field of digital pathology in LC. However, it is important to note that the current research primarily consists of independent studies among countries, emphasizing the necessity of strengthening academic collaboration and data sharing between nations. The current focus and challenge of research related to digital pathology in LC lie in enhancing the accuracy of classification and prediction through improved deep learning algorithms. The integration of multi-omics studies presents a promising future research direction. Additionally, researchers are increasingly exploring the application of digital pathology in immunotherapy for LC patients. Conclusions In conclusion, this study provides a comprehensive knowledge framework for digital pathology in LC, highlighting research trends, hotspots, and gaps in this field. It also provides a theoretical basis for the application of AI in clinical decision-making for LC patients.
Collapse
Affiliation(s)
- Dan-Dan Xiong
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Rong-Quan He
- Department of Medical Oncology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Zhi-Guang Huang
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Kun-Jun Wu
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Ying-Yu Mo
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Yue Liang
- Department of Pathology, Liuzhou People's Hospital, Liuzhou, Guangxi, China
| | - Da-Ping Yang
- Department of Pathology, Guigang City People's Hospital, Guigang, Guangxi, China
| | - Ying-Hui Wu
- Department of Pathology, The First People's Hospital of Yulin, Yulin, Guangxi, China
| | - Zhong-Qing Tang
- Department of Pathology, Gongren Hospital of Wuzhou, Wuzhou, Guangxi, China
| | - Zu-Tuan Liao
- Department of Pathology, The First People's Hospital of Hechi, Hechi, Guangxi, China
| | - Gang Chen
- Department of Pathology, First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Guangxi Zhuang Autonomous Region Engineering Research Center for Artificial Intelligence Analysis of Multimodal Tumor Images, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| |
Collapse
|
12
|
Wang S, Rong R, Zhou Q, Yang DM, Zhang X, Zhan X, Bishop J, Chi Z, Wilhelm CJ, Zhang S, Pickering CR, Kris MG, Minna J, Xie Y, Xiao G. Deep learning of cell spatial organizations identifies clinically relevant insights in tissue images. Nat Commun 2023; 14:7872. [PMID: 38081823 PMCID: PMC10713592 DOI: 10.1038/s41467-023-43172-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2023] [Accepted: 11/02/2023] [Indexed: 12/18/2023] Open
Abstract
Recent advancements in tissue imaging techniques have facilitated the visualization and identification of various cell types within physiological and pathological contexts. Despite the emergence of cell-cell interaction studies, there is a lack of methods for evaluating individual spatial interactions. In this study, we introduce Ceograph, a cell spatial organization-based graph convolutional network designed to analyze cell spatial organization (for example,. the cell spatial distribution, morphology, proximity, and interactions) derived from pathology images. Ceograph identifies key cell spatial organization features by accurately predicting their influence on patient clinical outcomes. In patients with oral potentially malignant disorders, our model highlights reduced structural concordance and increased closeness in epithelial substrata as driving features for an elevated risk of malignant transformation. In lung cancer patients, Ceograph detects elongated tumor nuclei and diminished stroma-stroma closeness as biomarkers for insensitivity to EGFR tyrosine kinase inhibitors. With its potential to predict various clinical outcomes, Ceograph offers a deeper understanding of biological processes and supports the development of personalized therapeutic strategies.
Collapse
Affiliation(s)
- Shidan Wang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Ruichen Rong
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Qin Zhou
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Donghan M Yang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xinyi Zhang
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xiaowei Zhan
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Justin Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Zhikai Chi
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Clare J Wilhelm
- Department of Thoracic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Siyuan Zhang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | - Mark G Kris
- Department of Thoracic Oncology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - John Minna
- Hamon Center for Therapeutic Oncology Research, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yang Xie
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA
| | - Guanghua Xiao
- Quantitative Biomedical Research Center, Peter O'Donnell Jr. School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Simmons Comprehensive Cancer Center, UT Southwestern Medical Center, Dallas, TX, USA.
- Department of Bioinformatics, UT Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
13
|
Ullah N, Bin Naeem S, Jhatial MA, Majeed MA, Saeed SM, Muzaffar S, Imran M, Hussain M, Fasih S. Acute Tubulointerstitial Nephritis and Secondary Renal Amyloidosis: A Rare Complication of Atezolizumab. Cureus 2023; 15:e49533. [PMID: 38156149 PMCID: PMC10753143 DOI: 10.7759/cureus.49533] [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: 11/27/2023] [Indexed: 12/30/2023] Open
Abstract
Lung cancer is the second most common malignancy in both genders and the most common cause of cancer-related deaths worldwide. Broadly, lung cancer is divided into two types: small-cell lung cancer (SCLC) and non-small cell lung cancer (NSCLC). Non-small cell lung cancer accounts for 85% of the diagnoses of lung cancer. It is necessary to check for any targetable mutations, which can help in deciding the treatment plan for the patients. The patient we are reporting is a 70-year-old male with multiple co-morbidities diagnosed with non-small cell carcinoma, favoring adenocarcinoma on histopathology. He was started on Atezolizumab/Bevacizumab/Carboplatin/Paclitaxel (ABCP). He was switched to maintenance Atezolizumab/Bevacizumab after four cycles due to poor tolerance to carboplatin and paclitaxel. The patient presented with neutropenic colitis and acute kidney injury (AKI), requiring admission. workup revealed nephrotic range proteinuria with a high urinary albumin-to-creatinine ratio. He underwent a renal biopsy to ascertain the cause of his proteinuria, which showed marked acute and chronic tubulo-interstitial nephritis (TIN), amyloidosis, and global glomerulosclerosis. Secondary (AA) amyloidosis is characterized by the extracellular deposition of misfolded proteins. Although interstitial nephritis is a reported side effect of immune checkpoint inhibitors, AA amyloidosis is a rarer side effect. So, to determine the exact cause and early therapeutic intervention in immune checkpoint inhibitor-related kidney injury, large retrospective or prospective studies should be done.
Collapse
Affiliation(s)
- Naqib Ullah
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Sameen Bin Naeem
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Mussadique Ali Jhatial
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | | | - Saad M Saeed
- Pathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Shakeel Muzaffar
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Maryam Imran
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Mudassar Hussain
- Histopathology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, Lahore, PAK
| | - Samir Fasih
- Medical Oncology, Shaukat Khanum Memorial Cancer Hospital and Research Centre, lahore, PAK
| |
Collapse
|
14
|
Terada K, Yoshizawa A, Liu X, Ito H, Hamaji M, Menju T, Date H, Bise R, Haga H. Deep Learning for Predicting Effect of Neoadjuvant Therapies in Non-Small Cell Lung Carcinomas With Histologic Images. Mod Pathol 2023; 36:100302. [PMID: 37580019 DOI: 10.1016/j.modpat.2023.100302] [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: 04/06/2023] [Revised: 06/23/2023] [Accepted: 08/02/2023] [Indexed: 08/16/2023]
Abstract
Neoadjuvant therapies are used for locally advanced non-small cell lung carcinomas, whereby pathologists histologically evaluate the effect using resected specimens. Major pathological response (MPR) has recently been used for treatment evaluation and as an economical survival surrogate; however, interobserver variability and poor reproducibility are often noted. The aim of this study was to develop a deep learning (DL) model to predict MPR from hematoxylin and eosin-stained tissue images and to validate its utility for clinical use. We collected data on 125 primary non-small cell lung carcinoma cases that were resected after neoadjuvant therapy. The cases were randomly divided into 55 for training/validation and 70 for testing. A total of 261 hematoxylin and eosin-stained slides were obtained from the maximum tumor beds, and whole slide images were prepared. We used a multiscale patch model that can adaptively weight multiple convolutional neural networks trained with different field-of-view images. We performed 3-fold cross-validation to evaluate the model. During testing, we compared the percentages of viable tumor evaluated by annotator pathologists (reviewed data), those evaluated by nonannotator pathologists (primary data), and those predicted by the DL-based model using 2-class confusion matrices and receiver operating characteristic curves and performed a survival analysis between MPR-achieved and non-MPR cases. In cross-validation, accuracy and mean F1 score were 0.859 and 0.805, respectively. During testing, accuracy and mean F1 score with reviewed data and those with primary data were 0.986, 0.985, 0.943, and 0.943, respectively. The areas under the receiver operating characteristic curve with reviewed and primary data were 0.999 and 0.978, respectively. The disease-free survival of MPR-achieved cases with reviewed and primary data was significantly better than that of the non-MPR cases (P<.001 and P=.001), and that predicted by the DL-based model was almost identical (P=.005). The DL model may support pathologist evaluations and can offer accurate determinations of MPR in patients.
Collapse
Affiliation(s)
- Kazuhiro Terada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan.
| | - Xiaoqing Liu
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Hiroaki Ito
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Ryoma Bise
- Department of Advanced Information Technology, Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
15
|
Rai HM, Yoo J. A comprehensive analysis of recent advancements in cancer detection using machine learning and deep learning models for improved diagnostics. J Cancer Res Clin Oncol 2023; 149:14365-14408. [PMID: 37540254 DOI: 10.1007/s00432-023-05216-w] [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: 06/20/2023] [Accepted: 07/26/2023] [Indexed: 08/05/2023]
Abstract
PURPOSE There are millions of people who lose their life due to several types of fatal diseases. Cancer is one of the most fatal diseases which may be due to obesity, alcohol consumption, infections, ultraviolet radiation, smoking, and unhealthy lifestyles. Cancer is abnormal and uncontrolled tissue growth inside the body which may be spread to other body parts other than where it has originated. Hence it is very much required to diagnose the cancer at an early stage to provide correct and timely treatment. Also, manual diagnosis and diagnostic error may cause of the death of many patients hence much research are going on for the automatic and accurate detection of cancer at early stage. METHODS In this paper, we have done the comparative analysis of the diagnosis and recent advancement for the detection of various cancer types using traditional machine learning (ML) and deep learning (DL) models. In this study, we have included four types of cancers, brain, lung, skin, and breast and their detection using ML and DL techniques. In extensive review we have included a total of 130 pieces of literature among which 56 are of ML-based and 74 are from DL-based cancer detection techniques. Only the peer reviewed research papers published in the recent 5-year span (2018-2023) have been included for the analysis based on the parameters, year of publication, feature utilized, best model, dataset/images utilized, and best accuracy. We have reviewed ML and DL-based techniques for cancer detection separately and included accuracy as the performance evaluation metrics to maintain the homogeneity while verifying the classifier efficiency. RESULTS Among all the reviewed literatures, DL techniques achieved the highest accuracy of 100%, while ML techniques achieved 99.89%. The lowest accuracy achieved using DL and ML approaches were 70% and 75.48%, respectively. The difference in accuracy between the highest and lowest performing models is about 28.8% for skin cancer detection. In addition, the key findings, and challenges for each type of cancer detection using ML and DL techniques have been presented. The comparative analysis between the best performing and worst performing models, along with overall key findings and challenges, has been provided for future research purposes. Although the analysis is based on accuracy as the performance metric and various parameters, the results demonstrate a significant scope for improvement in classification efficiency. CONCLUSION The paper concludes that both ML and DL techniques hold promise in the early detection of various cancer types. However, the study identifies specific challenges that need to be addressed for the widespread implementation of these techniques in clinical settings. The presented results offer valuable guidance for future research in cancer detection, emphasizing the need for continued advancements in ML and DL-based approaches to improve diagnostic accuracy and ultimately save more lives.
Collapse
Affiliation(s)
- Hari Mohan Rai
- School of Computing, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, 13120, Gyeonggi-do, Republic of Korea.
| | - Joon Yoo
- School of Computing, Gachon University, 1342 Seongnam-daero, Sujeong-gu, Seongnam-si, 13120, Gyeonggi-do, Republic of Korea
| |
Collapse
|
16
|
Hoang DT, Dinstag G, Hermida LC, Ben-Zvi DS, Elis E, Caley K, Sammut SJ, Sinha S, Sinha N, Dampier CH, Stossel C, Patil T, Rajan A, Lassoued W, Strauss J, Bailey S, Allen C, Redman J, Beker T, Jiang P, Golan T, Wilkinson S, Sowalsky AG, Pine SR, Caldas C, Gulley JL, Aldape K, Aharonov R, Stone EA, Ruppin E. Prediction of cancer treatment response from histopathology images through imputed transcriptomics. RESEARCH SQUARE 2023:rs.3.rs-3193270. [PMID: 37790315 PMCID: PMC10543028 DOI: 10.21203/rs.3.rs-3193270/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Advances in artificial intelligence have paved the way for leveraging hematoxylin and eosin (H&E)-stained tumor slides for precision oncology. We present ENLIGHT-DeepPT, an approach for predicting response to multiple targeted and immunotherapies from H&E-slides. In difference from existing approaches that aim to predict treatment response directly from the slides, ENLIGHT-DeepPT is an indirect two-step approach consisting of (1) DeepPT, a new deep-learning framework that predicts genome-wide tumor mRNA expression from slides, and (2) ENLIGHT, which predicts response based on the DeepPT inferred expression values. DeepPT successfully predicts transcriptomics in all 16 TCGA cohorts tested and generalizes well to two independent datasets. Our key contribution is showing that ENLIGHT-DeepPT successfully predicts true responders in five independent patients' cohorts involving four different treatments spanning six cancer types with an overall odds ratio of 2.44, increasing the baseline response rate by 43.47% among predicted responders, without the need for any treatment data for training. Furthermore, its prediction accuracy on these datasets is comparable to a supervised approach predicting the response directly from the images, which needs to be trained and tested on the same cohort. ENLIGHT-DeepPT future application could provide clinicians with rapid treatment recommendations to an array of different therapies and importantly, may contribute to advancing precision oncology in developing countries.
Collapse
Affiliation(s)
- Danh-Tai Hoang
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | | | - Leandro C. Hermida
- Department of Immunology, University of Pittsburgh, Pittsburgh, PA, USA
- Tumor Microenvironment Center, UPMC Hillman Cancer Center, University of Pittsburgh, Pittsburgh, PA, USA
| | | | | | - Katherine Caley
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Stephen-John Sammut
- Breast Cancer Now Toby Robins Research Centre, The Institute of Cancer Research, London, United Kingdom
- The Royal Marsden Hospital NHS Foundation Trust, London, United Kingdom
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - Sanju Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Neelam Sinha
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Christopher H. Dampier
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Chani Stossel
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Tejas Patil
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Arun Rajan
- Thoracic and GI Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Wiem Lassoued
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Julius Strauss
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Shania Bailey
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Clint Allen
- Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Jason Redman
- Center for Immuno-Oncology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Peng Jiang
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Talia Golan
- Oncology Institute, Sheba Medical Center at Tel-Hashomer, Tel Aviv University, Tel Aviv, Israel
| | - Scott Wilkinson
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Adam G. Sowalsky
- Laboratory of Genitourinary Cancer Pathogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Sharon R. Pine
- Division of Medical Oncology, University of Colorado Anschutz Medical Campus, Aurora, CO, USA
| | - Carlos Caldas
- Cancer Research UK Cambridge Institute, University of Cambridge, Li Ka Shing Centre, Cambridge, UK
| | - James L. Gulley
- Genitourinary Malignancy Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Kenneth Aldape
- Laboratory of Pathology, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | | | - Eric A. Stone
- Biological Data Science Institute, College of Science, Australian National University, Canberra, ACT, Australia
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
17
|
Halawani R, Buchert M, Chen YPP. Deep learning exploration of single-cell and spatially resolved cancer transcriptomics to unravel tumour heterogeneity. Comput Biol Med 2023; 164:107274. [PMID: 37506451 DOI: 10.1016/j.compbiomed.2023.107274] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Revised: 07/03/2023] [Accepted: 07/16/2023] [Indexed: 07/30/2023]
Abstract
Tumour heterogeneity is one of the critical confounding aspects in decoding tumour growth. Malignant cells display variations in their gene transcription profiles and mutation spectra even when originating from a single progenitor cell. Single-cell and spatial transcriptomics sequencing have recently emerged as key technologies for unravelling tumour heterogeneity. Single-cell sequencing promotes individual cell-type identification through transcriptome-wide gene expression measurements of each cell. Spatial transcriptomics facilitates identification of cell-cell interactions and the structural organization of heterogeneous cells within a tumour tissue through associating spatial RNA abundance of cells at distinct spots in the tissue section. However, extracting features and analyzing single-cell and spatial transcriptomics data poses challenges. Single-cell transcriptome data is extremely noisy and its sparse nature and dropouts can lead to misinterpretation of gene expression and the misclassification of cell types. Deep learning predictive power can overcome data challenges, provide high-resolution analysis and enhance precision oncology applications that involve early cancer prognosis, diagnosis, patient survival estimation and anti-cancer therapy planning. In this paper, we provide a background to and review of the recent progress of deep learning frameworks to investigate tumour heterogeneity using both single-cell and spatial transcriptomics data types.
Collapse
Affiliation(s)
- Raid Halawani
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia
| | - Michael Buchert
- School of Cancer Medicine, La Trobe University, Melbourne, Victoria, Australia; Olivia Newton-John Cancer Research Institute, Melbourne, Victoria, Australia
| | - Yi-Ping Phoebe Chen
- Department of Computer Science and Information Technology, La Trobe University, Melbourne, Australia.
| |
Collapse
|
18
|
Wang Y, Luo F, Yang X, Wang Q, Sun Y, Tian S, Feng P, Huang P, Xiao H. The Swin-Transformer network based on focal loss is used to identify images of pathological subtypes of lung adenocarcinoma with high similarity and class imbalance. J Cancer Res Clin Oncol 2023; 149:8581-8592. [PMID: 37097394 DOI: 10.1007/s00432-023-04795-y] [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: 04/12/2023] [Accepted: 04/15/2023] [Indexed: 04/26/2023]
Abstract
PURPOSE The classification of primary lung adenocarcinoma is complex and varied. Different subtypes of lung adenocarcinoma have different treatment methods and different prognosis. In this study, we collected 11 datasets comprising subtypes of lung cancer and proposed FL-STNet model to provide the assistance for improving clinical problems of pathologic classification in primary adenocarcinoma of lung. METHODS Samples were collected from 360 patients diagnosed with lung adenocarcinoma and other subtypes of lung diseases. In addition, an auxiliary diagnosis algorithm based on Swin-Transformer, which used Focal Loss for function in training, was developed. Meanwhile, the diagnostic accuracy of the Swin-Transformer was compared to pathologists. RESULTS The Swin-Transformer captures not only information in the overall tissue structure but also the local tissue details in the images of lung cancer pathology. Furthermore, training FL-STNet with the Focal Loss function can further balance the difference in the amount of data between different subtypes, improving recognition accuracy. The average classification accuracy, F1, and AUC of the proposed FL-STNet reached 85.71%, 86.57%, and 0.9903. The average accuracy of the FL-STNet was higher by 17% and 34%, respectively, than in the senior pathologist and junior pathologist group. CONCLUSION The first deep learning based on an 11-category classifier was developed for classifying lung adenocarcinoma subtypes based on WSI histopathology. Aiming at the deficiencies of the current CNN and Vit, FL-STNet model is proposed in this study by introducing Focal Loss and combining the advantages of Swin-Transformer model.
Collapse
Affiliation(s)
- Yujun Wang
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Furong Luo
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Xing Yang
- College of Computer and Cyber Security, Chengdu University of Technology, Chengdu, 610059, People's Republic of China
| | - Qiushi Wang
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China
| | - Yunchun Sun
- Center of Digital Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, People's Republic of China
| | - Sukun Tian
- Center of Digital Dentistry, School and Hospital of Stomatology, Peking University, Beijing, 100081, People's Republic of China
| | - Peng Feng
- Key Laboratory of Optoelectronic Technology & Systems (Ministry of Education), College of Optoelectronic Engineering, Chongqing University, Chongqing, 400044, People's Republic of China
| | - Pan Huang
- Key Laboratory of Optoelectronic Technology & Systems (Ministry of Education), College of Optoelectronic Engineering, Chongqing University, Chongqing, 400044, People's Republic of China.
| | - Hualiang Xiao
- Department of Pathology, Daping Hospital, Army Medical University, Chongqing, 400042, People's Republic of China.
| |
Collapse
|
19
|
Nasrallah MP, Zhao J, Tsai CC, Meredith D, Marostica E, Ligon KL, Golden JA, Yu KH. Machine learning for cryosection pathology predicts the 2021 WHO classification of glioma. MED 2023; 4:526-540.e4. [PMID: 37421953 PMCID: PMC10527821 DOI: 10.1016/j.medj.2023.06.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2022] [Revised: 04/17/2023] [Accepted: 06/06/2023] [Indexed: 07/10/2023]
Abstract
BACKGROUND Timely and accurate intraoperative cryosection evaluations remain the gold standard for guiding surgical treatments for gliomas. However, the tissue-freezing process often generates artifacts that make histologic interpretation difficult. In addition, the 2021 WHO Classification of Tumors of the Central Nervous System incorporates molecular profiles in the diagnostic categories, so standard visual evaluation of cryosections alone cannot completely inform diagnoses based on the new classification system. METHODS To address these challenges, we develop the context-aware Cryosection Histopathology Assessment and Review Machine (CHARM) using samples from 1,524 glioma patients from three different patient populations to systematically analyze cryosection slides. FINDINGS Our CHARM models successfully identified malignant cells (AUROC = 0.98 ± 0.01 in the independent validation cohort), distinguished isocitrate dehydrogenase (IDH)-mutant tumors from wild type (AUROC = 0.79-0.82), classified three major types of molecularly defined gliomas (AUROC = 0.88-0.93), and identified the most prevalent subtypes of IDH-mutant tumors (AUROC = 0.89-0.97). CHARM further predicts clinically important genetic alterations in low-grade glioma, including ATRX, TP53, and CIC mutations, CDKN2A/B homozygous deletion, and 1p/19q codeletion via cryosection images. CONCLUSIONS Our approaches accommodate the evolving diagnostic criteria informed by molecular studies, provide real-time clinical decision support, and will democratize accurate cryosection diagnoses. FUNDING Supported in part by the National Institute of General Medical Sciences grant R35GM142879, the Google Research Scholar Award, the Blavatnik Center for Computational Biomedicine Award, the Partners' Innovation Discovery Grant, and the Schlager Family Award for Early Stage Digital Health Innovations.
Collapse
Affiliation(s)
- MacLean P Nasrallah
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine at the University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junhan Zhao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - Cheng Che Tsai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA
| | - David Meredith
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA 02139, USA
| | - Keith L Ligon
- Department of Pathology, Dana-Farber Cancer Institute, Boston, MA 02215, USA
| | - Jeffrey A Golden
- Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA; Department of Pathology, Cedars-Sinai Medical Center, Los Angeles, CA 90048, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA 02115, USA; Department of Pathology, Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
20
|
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
|
21
|
Xiao H, Liu Q, Li L. MFMANet: Multi-feature Multi-attention Network for efficient subtype classification on non-small cell lung cancer CT images. Biomed Signal Process Control 2023. [DOI: 10.1016/j.bspc.2023.104768] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/19/2023]
|
22
|
Czaplewska P, Bogucka A, Macur K, Rybicka M, Rychłowski M, Fiołka MJ. Proteomic response of A549 lung cancer cell line to protein-polysaccharide complex Venetin-1 isolated from earthworm coelomic fluid. Front Mol Biosci 2023; 10:1128320. [PMID: 37377864 PMCID: PMC10292018 DOI: 10.3389/fmolb.2023.1128320] [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: 12/20/2022] [Accepted: 05/16/2023] [Indexed: 06/29/2023] Open
Abstract
Earthworms' celomic fluid has long attracted scientists' interest due to their toxic properties. It has been shown that the elimination of coelomic fluid cytotoxicity to normal human cells was crucial for the generation of the non-toxic Venetin-1 protein-polysaccharide complex, which exhibits selective activity against Candida albicans cells as well as A549 non-small cell lung cancer cells. To find the molecular mechanisms behind the anti-cancer properties of the preparation, this research investigated the proteome response of A549 cells to the presence of Venetin-1. The sequential window acquisition of all theoretical mass spectra (SWATH-MS) methodology was used for the analysis, which allows for a relative quantitative analysis to be carried out without radiolabelling. The results showed that the formulation did not induce significant proteome responses in normal BEAS-2B cells. In the case of the tumour line, 31 proteins were up regulated, and 18 proteins down regulated. Proteins with increased expression in neoplastic cells are mainly associated with the mitochondrion, membrane transport and the endoplasmic reticulum. In the case of altered proteins, Venetin-1 interferes with proteins that stabilise the structures, i.e., keratin, glycolysis/gluconeogenesis and metabolic processes.
Collapse
Affiliation(s)
- Paulina Czaplewska
- Intercollegiate Faculty of Biotechnology, The University of Gdansk, Gdańsk, Poland
| | - Aleksandra Bogucka
- Intercollegiate Faculty of Biotechnology, The University of Gdansk, Gdańsk, Poland
- Institute of Biochemistry, Justus Liebig University of Giessen, Giessen, Germany
| | - Katarzyna Macur
- Intercollegiate Faculty of Biotechnology, The University of Gdansk, Gdańsk, Poland
| | - Magda Rybicka
- Intercollegiate Faculty of Biotechnology, The University of Gdansk, Gdańsk, Poland
| | - Michał Rychłowski
- Intercollegiate Faculty of Biotechnology, The University of Gdansk, Gdańsk, Poland
| | - Marta J. Fiołka
- Department of Immunobiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
23
|
Tsai PC, Lee TH, Kuo KC, Su FY, Lee TLM, Marostica E, Ugai T, Zhao M, Lau MC, Väyrynen JP, Giannakis M, Takashima Y, Kahaki SM, Wu K, Song M, Meyerhardt JA, Chan AT, Chiang JH, Nowak J, Ogino S, Yu KH. Histopathology images predict multi-omics aberrations and prognoses in colorectal cancer patients. Nat Commun 2023; 14:2102. [PMID: 37055393 PMCID: PMC10102208 DOI: 10.1038/s41467-023-37179-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Accepted: 03/03/2023] [Indexed: 04/15/2023] Open
Abstract
Histopathologic assessment is indispensable for diagnosing colorectal cancer (CRC). However, manual evaluation of the diseased tissues under the microscope cannot reliably inform patient prognosis or genomic variations crucial for treatment selections. To address these challenges, we develop the Multi-omics Multi-cohort Assessment (MOMA) platform, an explainable machine learning approach, to systematically identify and interpret the relationship between patients' histologic patterns, multi-omics, and clinical profiles in three large patient cohorts (n = 1888). MOMA successfully predicts the overall survival, disease-free survival (log-rank test P-value<0.05), and copy number alterations of CRC patients. In addition, our approaches identify interpretable pathology patterns predictive of gene expression profiles, microsatellite instability status, and clinically actionable genetic alterations. We show that MOMA models are generalizable to multiple patient populations with different demographic compositions and pathology images collected from distinctive digitization methods. Our machine learning approaches provide clinically actionable predictions that could inform treatments for colorectal cancer patients.
Collapse
Affiliation(s)
- Pei-Chen Tsai
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Tsung-Hua Lee
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Kun-Chi Kuo
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Fang-Yi Su
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC
| | - Tsung-Lu Michael Lee
- Department of Computer Science and Information Engineering, Southern Taiwan University of Science and Technology, Tainan, Taiwan ROC
| | - Eliana Marostica
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Division of Health Sciences and Technology, Harvard-Massachusetts Institute of Technology, Boston, MA, USA
| | - Tomotaka Ugai
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Melissa Zhao
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Mai Chan Lau
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Juha P Väyrynen
- Cancer and Translational Medicine Research Unit, Medical Research Center Oulu, Oulu University Hospital and University of Oulu, Oulu, Finland
| | - Marios Giannakis
- Department of Medicine, Dana Farber Cancer Institute, Boston, MA, USA
| | | | | | - Kana Wu
- Department of Nutrition, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Mingyang Song
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | | | - Andrew T Chan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital, Boston, MA, USA
| | - Jung-Hsien Chiang
- Department of Computer Science and Information Engineering, National Cheng Kung University, Tainan, Taiwan ROC.
| | - Jonathan Nowak
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
| | - Shuji Ogino
- Department of Epidemiology, Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA.
- Department of Pathology, Brigham and Women's Hospital, Boston, MA, USA.
| |
Collapse
|
24
|
Alghamdi RA, Al-Zahrani MH. Integrated bioinformatics analyses identifying key transcriptomes correlated with prognosis and immune infiltrations in lung squamous cell carcinoma. Saudi J Biol Sci 2023; 30:103596. [PMID: 36879671 PMCID: PMC9985037 DOI: 10.1016/j.sjbs.2023.103596] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2022] [Revised: 01/15/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Background Lung Squamous Cell Carcinoma (LUSC) is a major subtype of lung malignancies and is associated with the cause of cancer-mediated mortality worldwide. However, identification of transcriptomic signatures associated with survival-prognosis and immunity of tumor remains lacking. Method The GSE2088, GSE6044, GSE19188, GSE21933, GSE33479, GSE33532, and GSE74706 were integrated for identifying differentially expressed genes (DEGs) with combined effect sizes. Also, the TCGA LUSC cohort was used for further analysis. A series of bioinformatics methods were utilized for conducting the whole study. Results The 831 genes (such as DSG3, PKP1, DSC3, TPX2, and UBE2C) were found upregulated and the 731 genes (such as ABCA8, SELENBP1, FAM107A, and CACNA2D2) were downregulated in the LUSC. The functional enrichment analysis identifies the upregulated KEGG pathways, including cell cycle, DNA replication, base excision repair, proteasome, mismatch repair, and cellular senescence. Also, the key hub genes (such as EGFR, HRAS, JUN, CDH1, BRCA1, CASP3, RHOA, HDAC1, HIF1A, and CCNA2) were identified along with the eight gene modules that were significantly related to the protein-protein interaction (PPI). The clinical analyses identified that the overexpression group of CDH3, PLAU, PKP3, STIL, CALU, LOXL2, POSTN, DPP3, GALNT2, LOX, and ITPA are substantially associated with a poor survival prognosis and the downregulated group of IL18R1 showed a similar trend. Moreover, our investigation demonstrated that the survival-associated genes were correlated with the stromal and immune scores in LUSC, indicating that the survival-associated genes regulate tumor immunity. The survival-associated genes were genetically altered in 27% of LUSC patients and showed excellent diagnostic efficiency. Finally, the consistent expression level of CDH3, PLAU, PKP3, STIL, CALU, LOXL2, POSTN, DPP3, GALNT2, and ITPA were found in the TCGA LUSC cohort. Conclusions The identification of key transcriptomic signatures can be elucidated by the crucial mechanism of LUSC carcinogenesis.
Collapse
Affiliation(s)
- Rana A. Alghamdi
- Department of Chemistry, Science and Arts College, King Abdulaziz University, Rabigh, Saudi Arabia
- Corresponding author at: Department of Chemistry, Science and Arts College, King Abdulaziz University, Rabigh, Saudi Arabia.
| | - Maryam H. Al-Zahrani
- Biochemistry Department, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
25
|
Yang D, Wu Y, Wan Z, Xu Z, Li W, Yuan P, Shang Q, Peng J, Tao L, Chen Q, Dan H, Xu H. HISMD: A Novel Immune Subtyping System for HNSCC. J Dent Res 2023; 102:270-279. [PMID: 36333876 DOI: 10.1177/00220345221134605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Immune subtyping is an important way to reveal immune heterogeneity, which may contribute to the diversity of the progression and treatment in head and neck squamous cell carcinoma (HNSCC). However, reported immune subtypes mainly focus on levels of immune infiltration and are mostly based on a mono-omics profile. This study aimed to identify a comprehensive immune subtype for HNSCC via multi-omics clustering and build a novel subtype prediction system for clinical application. Data were obtained from The Cancer Genome Atlas database and our independent multicenter cohort. Multi-omics clustering was performed to identify 3 clusters of 499 patients in The Cancer Genome Atlas based on immune-related gene expression and somatic mutations. The immune characteristics and biological features of the obtained clusters were revealed by bioinformatics, and 3 immune subtypes were identified: 1) adaptive immune activation subtype predominantly enriched in T cells, 2) innate immune activation subtype predominantly enriched in macrophages, and 3) immune desert subtype. Subsequently, the clinical implications of each subtype were analyzed per clinical epidemiology. We found that adaptive immune activation showed better survival outcomes and had a similar response to chemotherapy with innate immune activation, whereas immune desert might be relatively resistant to chemotherapy. Moreover, a subtype prediction system was developed by deep learning with whole slide images and named HISMD: HNSCC Immune Subtypes via Multi-omics and Deep Learning. We endowed HISMD with interpretability through image-based key feature extraction. The clinical implications, biological significances, and predictive stability of HISMD were successfully verified by using our independent multicenter cohort data set. In summary, this study revealed the immune heterogeneity of HNSCC and obtained a novel, highly accurate, and interpretable immune subtyping prediction system. For clinical implementation in the future, additional validation and utility studies are warranted.
Collapse
Affiliation(s)
- D Yang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Y Wu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Wan
- Department of Pathology, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Z Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - W Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - P Yuan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Q Shang
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - J Peng
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - L Tao
- College of Mathematics, Sichuan University, Chengdu, China
| | - Q Chen
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China.,Key Laboratory of Oral Biomedical Research of Zhejiang Province, Affiliated Stomatology Hospital, Zhejiang University School of Stomatology, Hangzhou, China
| | - H Dan
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - H Xu
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, Research Unit of Oral Carcinogenesis and Management, Chinese Academy of Medical Sciences, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
26
|
Eminaga O, Abbas M, Shen J, Laurie M, Brooks JD, Liao JC, Rubin DL. PlexusNet: A neural network architectural concept for medical image classification. Comput Biol Med 2023; 154:106594. [PMID: 36753979 DOI: 10.1016/j.compbiomed.2023.106594] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 01/12/2023] [Accepted: 01/22/2023] [Indexed: 01/27/2023]
Abstract
State-of-the-art (SOTA) convolutional neural network models have been widely adapted in medical imaging and applied to address different clinical problems. However, the complexity and scale of such models may not be justified in medical imaging and subject to the available resource budget. Further increasing the number of representative feature maps for the classification task decreases the model explainability. The current data normalization practice is fixed prior to model development and discounting the specification of the data domain. Acknowledging these issues, the current work proposed a new scalable model family called PlexusNet; the block architecture and model scaling by the network's depth, width, and branch regulate PlexusNet's architecture. The efficient computation costs outlined the dimensions of PlexusNet scaling and design. PlexusNet includes a new learnable data normalization algorithm for better data generalization. We applied a simple yet effective neural architecture search to design PlexusNet tailored to five clinical classification problems that achieve a performance noninferior to the SOTA models ResNet-18 and EfficientNet B0/1. It also does so with lower parameter capacity and representative feature maps in ten-fold ranges than the smallest SOTA models with comparable performance. The visualization of representative features revealed distinguishable clusters associated with categories based on latent features generated by PlexusNet. The package and source code are at https://github.com/oeminaga/PlexusNet.git.
Collapse
Affiliation(s)
- Okyaz Eminaga
- Center for Artificial Intelligence in Medicine & Imaging and Department of Urology, Stanford School of Medicine, Stanford, CA, 94305, USA; Department of Urology, Stanford School of Medicine, Stanford, CA, 94305, USA
| | - Mahmoud Abbas
- Department of Pathology, University of Muenster, Muenster, Germany.
| | - Jeanne Shen
- Department of Pathology, Stanford School of Medicine, Stanford, CA, 94305, USA.
| | - Mark Laurie
- Department of Computer Science, Stanford University, Stanford, CA, 94305, USA.
| | - James D Brooks
- Department of Urology, Stanford School of Medicine, Stanford, CA, 94305, USA.
| | - Joseph C Liao
- Department of Urology, Stanford School of Medicine, Stanford, CA, 94305, USA.
| | - Daniel L Rubin
- Department of Biomedical Data Science, Stanford School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
27
|
Jiang Y, Sui X, Ding Y, Xiao W, Zheng Y, Zhang Y. A semi-supervised learning approach with consistency regularization for tumor histopathological images analysis. Front Oncol 2023; 12:1044026. [PMID: 36698401 PMCID: PMC9870542 DOI: 10.3389/fonc.2022.1044026] [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: 09/16/2022] [Accepted: 12/06/2022] [Indexed: 01/12/2023] Open
Abstract
Introduction Manual inspection of histopathological images is important in clinical cancer diagnosis. Pathologists implement pathological diagnosis and prognostic evaluation through the microscopic examination of histopathological slices. This entire process is time-consuming, laborious, and challenging for pathologists. The modern use of whole-slide imaging, which scans histopathology slides to digital slices, and analysis using computer-aided diagnosis is an essential problem. Methods To solve the problem of difficult labeling of histopathological data, and improve the flexibility of histopathological analysis in clinical applications, we herein propose a semi-supervised learning algorithm coupled with consistency regularization strategy, called"Semi- supervised Histopathology Analysis Network"(Semi-His-Net), for automated normal-versus-tumor and subtype classifications. Specifically, when inputted disturbing versions of the same image, the model should predict similar outputs. Based on this, the model itself can assign artificial labels to unlabeled data for subsequent model training, thereby effectively reducing the labeled data required for training. Results Our Semi-His-Net is able to classify patches from breast cancer histopathological images into normal tissue and three other different tumor subtypes, achieving an accuracy was 90%. The average AUC of cross-classification between tumors reached 0.893. Discussion To overcome the limitations of visual inspection by pathologists for histopathology images, such as long time and low repeatability, we have developed a deep learning-based framework (Semi-His-Net) for automatic classification subdivision of the subtypes contained in the whole pathological images. This learning-based framework has great potential to improve the efficiency and repeatability of histopathological image diagnosis.
Collapse
Affiliation(s)
- Yanyun Jiang
- School of Mathematics and Statistics, Shandong Normal University, Jinan, China
| | - Xiaodan Sui
- School of Mathematics and Statistics, Shandong Normal University, Jinan, China
| | - Yanhui Ding
- School of Mathematics and Statistics, Shandong Normal University, Jinan, China
| | - Wei Xiao
- Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Yuanjie Zheng
- School of Mathematics and Statistics, Shandong Normal University, Jinan, China,*Correspondence: Yuanjie Zheng, ; Yongxin Zhang,
| | - Yongxin Zhang
- School of Mathematics and Statistics, Shandong Normal University, Jinan, China,*Correspondence: Yuanjie Zheng, ; Yongxin Zhang,
| |
Collapse
|
28
|
Wu B, Moeckel G. Application of digital pathology and machine learning in the liver, kidney and lung diseases. J Pathol Inform 2023; 14:100184. [PMID: 36714454 PMCID: PMC9874068 DOI: 10.1016/j.jpi.2022.100184] [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: 09/21/2022] [Revised: 11/28/2022] [Accepted: 12/28/2022] [Indexed: 01/05/2023] Open
Abstract
The development of rapid and accurate Whole Slide Imaging (WSI) has paved the way for the application of Artificial Intelligence (AI) to digital pathology. The availability of WSI in the recent years allowed the rapid development of various AI technologies to blossom. WSI-based digital pathology combined with neural networks can automate arduous and time-consuming tasks of slide evaluation. Machine Learning (ML)-based AI has been demonstrated to outperform pathologists by eliminating inter- and intra-observer subjectivity, obtaining quantitative data from slide images, and extracting hidden image patterns that are relevant to disease subtype and progression. In this review, we outline the functionality of different AI technologies such as neural networks and deep learning and discover how aspects of different diseases make them benefit from the implementation of AI. AI has proven to be valuable in many different organs, with this review focusing on the liver, kidney, and lungs. We also discuss how AI and image analysis not only can grade diseases objectively but also discover aspects of diseases that have prognostic value. In the end, we review the current status of the integration of AI in pathology and share our vision on the future of digital pathology.
Collapse
Affiliation(s)
- Benjamin Wu
- Horace Mann School, Bronx, NY, USA,Corresponding author at: 950 Post Rd., Scarsdale, NY 10583, USA.
| | - Gilbert Moeckel
- Department of Pathology, Yale University School of Medicine, New Haven, CT, USA
| |
Collapse
|
29
|
He X, Liu X, Zuo F, Shi H, Jing J. Artificial intelligence-based multi-omics analysis fuels cancer precision medicine. Semin Cancer Biol 2023; 88:187-200. [PMID: 36596352 DOI: 10.1016/j.semcancer.2022.12.009] [Citation(s) in RCA: 79] [Impact Index Per Article: 39.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 12/16/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023]
Abstract
With biotechnological advancements, innovative omics technologies are constantly emerging that have enabled researchers to access multi-layer information from the genome, epigenome, transcriptome, proteome, metabolome, and more. A wealth of omics technologies, including bulk and single-cell omics approaches, have empowered to characterize different molecular layers at unprecedented scale and resolution, providing a holistic view of tumor behavior. Multi-omics analysis allows systematic interrogation of various molecular information at each biological layer while posing tricky challenges regarding how to extract valuable insights from the exponentially increasing amount of multi-omics data. Therefore, efficient algorithms are needed to reduce the dimensionality of the data while simultaneously dissecting the mysteries behind the complex biological processes of cancer. Artificial intelligence has demonstrated the ability to analyze complementary multi-modal data streams within the oncology realm. The coincident development of multi-omics technologies and artificial intelligence algorithms has fuelled the development of cancer precision medicine. Here, we present state-of-the-art omics technologies and outline a roadmap of multi-omics integration analysis using an artificial intelligence strategy. The advances made using artificial intelligence-based multi-omics approaches are described, especially concerning early cancer screening, diagnosis, response assessment, and prognosis prediction. Finally, we discuss the challenges faced in multi-omics analysis, along with tentative future trends in this field. With the increasing application of artificial intelligence in multi-omics analysis, we anticipate a shifting paradigm in precision medicine becoming driven by artificial intelligence-based multi-omics technologies.
Collapse
Affiliation(s)
- Xiujing He
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China
| | - Xiaowei Liu
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China
| | - Fengli Zuo
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China
| | - Hubing Shi
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China
| | - Jing Jing
- Laboratory of Integrative Medicine, Clinical Research Center for Breast, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University and Collaborative Innovation Center, Chengdu, Sichuan, PR China.
| |
Collapse
|
30
|
Couture HD. Deep Learning-Based Prediction of Molecular Tumor Biomarkers from H&E: A Practical Review. J Pers Med 2022; 12:2022. [PMID: 36556243 PMCID: PMC9784641 DOI: 10.3390/jpm12122022] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 10/26/2022] [Accepted: 12/05/2022] [Indexed: 12/12/2022] Open
Abstract
Molecular and genomic properties are critical in selecting cancer treatments to target individual tumors, particularly for immunotherapy. However, the methods to assess such properties are expensive, time-consuming, and often not routinely performed. Applying machine learning to H&E images can provide a more cost-effective screening method. Dozens of studies over the last few years have demonstrated that a variety of molecular biomarkers can be predicted from H&E alone using the advancements of deep learning: molecular alterations, genomic subtypes, protein biomarkers, and even the presence of viruses. This article reviews the diverse applications across cancer types and the methodology to train and validate these models on whole slide images. From bottom-up to pathologist-driven to hybrid approaches, the leading trends include a variety of weakly supervised deep learning-based approaches, as well as mechanisms for training strongly supervised models in select situations. While results of these algorithms look promising, some challenges still persist, including small training sets, rigorous validation, and model explainability. Biomarker prediction models may yield a screening method to determine when to run molecular tests or an alternative when molecular tests are not possible. They also create new opportunities in quantifying intratumoral heterogeneity and predicting patient outcomes.
Collapse
|
31
|
Iqbal MS, Ahmad W, Alizadehsani R, Hussain S, Rehman R. Breast Cancer Dataset, Classification and Detection Using Deep Learning. Healthcare (Basel) 2022; 10:2395. [PMID: 36553919 PMCID: PMC9778593 DOI: 10.3390/healthcare10122395] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 11/24/2022] [Accepted: 11/25/2022] [Indexed: 12/05/2022] Open
Abstract
Incorporating scientific research into clinical practice via clinical informatics, which includes genomics, proteomics, bioinformatics, and biostatistics, improves patients' treatment. Computational pathology is a growing subspecialty with the potential to integrate whole slide images, multi-omics data, and health informatics. Pathology and laboratory medicine are critical to diagnosing cancer. This work will review existing computational and digital pathology methods for breast cancer diagnosis with a special focus on deep learning. The paper starts by reviewing public datasets related to breast cancer diagnosis. Additionally, existing deep learning methods for breast cancer diagnosis are reviewed. The publicly available code repositories are introduced as well. The paper is closed by highlighting challenges and future works for deep learning-based diagnosis.
Collapse
Affiliation(s)
- Muhammad Shahid Iqbal
- Department of Computer Science and Information Technology, Women University AJK, Bagh 12500, Pakistan
| | - Waqas Ahmad
- Higher Education Department Govt, AJK, Mirpur 10250, Pakistan
| | - Roohallah Alizadehsani
- Institute for Intelligent Systems Research and Innovation (IISRI), Deakin University, Geelong, VIC 3216, Australia
| | - Sadiq Hussain
- Examination Branch, Dibrugarh University, Dibrugarh 786004, India
| | - Rizwan Rehman
- Centre for Computer Science and Applications, Dibrugarh University, Dibrugarh 786004, India
| |
Collapse
|
32
|
Civit-Masot J, Bañuls-Beaterio A, Domínguez-Morales M, Rivas-Pérez M, Muñoz-Saavedra L, Rodríguez Corral JM. Non-small cell lung cancer diagnosis aid with histopathological images using Explainable Deep Learning techniques. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2022; 226:107108. [PMID: 36113183 DOI: 10.1016/j.cmpb.2022.107108] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/26/2022] [Accepted: 09/01/2022] [Indexed: 06/15/2023]
Abstract
BACKGROUND Lung cancer has the highest mortality rate in the world, twice as high as the second highest. On the other hand, pathologists are overworked and this is detrimental to the time spent on each patient, diagnostic turnaround time, and their success rate. OBJECTIVE In this work, we design, implement, and evaluate a diagnostic aid system for non-small cell lung cancer detection, using Deep Learning techniques. METHODS The classifier developed is based on Artificial Intelligence techniques, obtaining an automatic classification result between healthy, adenocarcinoma and squamous cell carcinoma, given an histopathological image from lung tissue. Moreover, a report module based on Explainable Deep Learning techniques is included and gives the pathologist information about the image's areas used to classify the sample and the confidence of belonging to each class. RESULTS The results show a system accuracy between 97.11 and 99.69%, depending on the number of classes classified, and a value of the area under ROC curve between 99.77 and 99.94%. CONCLUSIONS The classification results obtain a substantial improvement according to previous works. Thanks to the given report, the time spent by the pathologist and the diagnostic turnaround time can be reduced.
Collapse
Affiliation(s)
- Javier Civit-Masot
- Architecture and Computer Technology department (ATC), Robotics and Technology of Computers Lab (RTC), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain
| | - Alejandro Bañuls-Beaterio
- Architecture and Computer Technology department (ATC), Robotics and Technology of Computers Lab (RTC), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain
| | - Manuel Domínguez-Morales
- Architecture and Computer Technology department (ATC), Robotics and Technology of Computers Lab (RTC), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain; Computer Engineering Research Institute (I3US), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain.
| | - Manuel Rivas-Pérez
- Architecture and Computer Technology department (ATC), Robotics and Technology of Computers Lab (RTC), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain
| | - Luis Muñoz-Saavedra
- Architecture and Computer Technology department (ATC), Robotics and Technology of Computers Lab (RTC), E.T.S. Ingeniería Informática, Avda. Reina Mercedes s/n, Universidad de Sevilla, Seville, 41012, Spain
| | - José M Rodríguez Corral
- Computer Science department, School of Engineering, Avda. Universidad de Cádiz 10, Universidad de Cádiz, Puerto Real (Cádiz), 11519, Spain
| |
Collapse
|
33
|
Lee SH, Jang HJ. Deep learning-based prediction of molecular cancer biomarkers from tissue slides: A new tool for precision oncology. Clin Mol Hepatol 2022; 28:754-772. [PMID: 35443570 PMCID: PMC9597228 DOI: 10.3350/cmh.2021.0394] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 04/17/2022] [Indexed: 02/06/2023] Open
Abstract
Molecular tests are necessary to stratify cancer patients for targeted therapy. However, high cost and technical barriers limit the application of these tests, hindering optimal treatment. Recently, deep learning (DL) has been applied to predict molecular test results from digitized images of tissue slides. Furthermore, treatment response and prognosis can be predicted from tissue slides using DL. In this review, we summarized DL-based studies regarding the prediction of genetic mutation, microsatellite instability, tumor mutational burden, molecular subtypes, gene expression, treatment response, and prognosis directly from hematoxylin- and eosin-stained tissue slides. Although performance needs to be improved, these studies clearly demonstrated the feasibility of DL-based prediction of key molecular features in cancer tissues. With the accumulation of data and technical advances, the performance of the DL system could be improved in the near future. Therefore, we expect that DL could provide cost- and time-effective alternative tools for patient stratification in the era of precision oncology.
Collapse
Affiliation(s)
- Sung Hak Lee
- Department of Hospital Pathology, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hyun-Jong Jang
- Catholic Big Data Integration Center, Department of Physiology, College of Medicine, The Catholic University of Korea, Seoul, Korea,Corresponding author : Hyun-Jong Jang Department of Physiology, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul 06591, Korea Tel: +82-2-2258-7274, Fax: +82-2-532-9575, E-mail:
| |
Collapse
|
34
|
Li Y, Dong Y, Zhao S, Gao J, Hao X, Wang Z, Li M, Wang M, Liu Y, Yu X, Xu W. Serum-derived piR-hsa-164586 of extracellular vesicles as a novel biomarker for early diagnosis of non-small cell lung cancer. Front Oncol 2022; 12:850363. [PMID: 36249068 PMCID: PMC9559724 DOI: 10.3389/fonc.2022.850363] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Accepted: 08/10/2022] [Indexed: 11/13/2022] Open
Abstract
Non-small cell lung cancer (NSCLC) is a major cause of death in those with malignant tumors. To achieve the early diagnosis of NSCLC, we investigated serum-derived Piwi-interacting RNA (piRNA) of extracellular vesicles to filter diagnostic biomarkers for NSCLC. High-throughput sequencing from cancerous tissues and adjacent noncancerous tissues in patients with NSCLC was first applied to recognize candidate piRNAs as diagnostic biomarkers. These screened piRNAs were further validated in 115 patients (including 95 cases in stage I) and 47 healthy individuals using quantitative real-time PCR (qRT-PCR). We showed that piR-hsa-164586 was significantly upregulated compared with paracancerous tissues and extracellular vesicles from the serum samples of healthy individuals. Moreover, the area under the curve (AUC) value of piR-hsa-164586 was 0.623 and 0.624 to distinguish patients with all stages or stage I of NSCLC, respectively, from healthy individuals. The diagnostic performance of piR-hsa-164586 was greatly improved compared with the cytokeratin-19-fragment (CYFRA21-1). Additionally, piR-hs-164586 was associated with the clinical characteristics of patients with NSCLC. Its expression was associated with the age and TNM stage of patients with NSCLC, indicating that it can serve as an effective and promising biomarker for the early diagnosis of NSCLC.
Collapse
Affiliation(s)
- Yanli Li
- Department of Pathology and Pathophysiology, The Medical Faculty of Qingdao University, Qingdao, China
| | - Yanhan Dong
- Institute of Translational Medicine, Qingdao University, Qingdao, China
| | - Shupeng Zhao
- Asset and Laboratory Management Office, Qingdao University, Qingdao, China
| | - Jinning Gao
- Institute of Translational Medicine, Qingdao University, Qingdao, China
| | - Xiaodan Hao
- Institute of Translational Medicine, Qingdao University, Qingdao, China
| | - Zibo Wang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Meng Li
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Mengyuan Wang
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Yiming Liu
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
| | - Xiaoling Yu
- Department of Pathology and Pathophysiology, The Medical Faculty of Qingdao University, Qingdao, China
- *Correspondence: Xiaoling Yu, ; Wenhua Xu,
| | - Wenhua Xu
- Department of Inspection, The Medical Faculty of Qingdao University, Qingdao, China
- *Correspondence: Xiaoling Yu, ; Wenhua Xu,
| |
Collapse
|
35
|
Shmatko A, Ghaffari Laleh N, Gerstung M, Kather JN. Artificial intelligence in histopathology: enhancing cancer research and clinical oncology. NATURE CANCER 2022; 3:1026-1038. [PMID: 36138135 DOI: 10.1038/s43018-022-00436-4] [Citation(s) in RCA: 158] [Impact Index Per Article: 52.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Accepted: 08/03/2022] [Indexed: 06/16/2023]
Abstract
Artificial intelligence (AI) methods have multiplied our capabilities to extract quantitative information from digital histopathology images. AI is expected to reduce workload for human experts, improve the objectivity and consistency of pathology reports, and have a clinical impact by extracting hidden information from routinely available data. Here, we describe how AI can be used to predict cancer outcome, treatment response, genetic alterations and gene expression from digitized histopathology slides. We summarize the underlying technologies and emerging approaches, noting limitations, including the need for data sharing and standards. Finally, we discuss the broader implications of AI in cancer research and oncology.
Collapse
Affiliation(s)
- Artem Shmatko
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK
| | | | - Moritz Gerstung
- Division of AI in Oncology, German Cancer Research Center (DKFZ), Heidelberg, Germany.
- European Molecular Biology Laboratory, European Bioinformatics Institute, Cambridge, UK.
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.
- Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany.
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
36
|
Xiao W, Jiang Y, Yao Z, Zhou X, Sui X, Zheng Y. LAD-GCN: Automatic diagnostic framework for quantitative estimation of growth patterns during clinical evaluation of lung adenocarcinoma. Front Physiol 2022; 13:946099. [PMID: 36035486 PMCID: PMC9399647 DOI: 10.3389/fphys.2022.946099] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Accepted: 07/04/2022] [Indexed: 11/27/2022] Open
Abstract
Quantitative estimation of growth patterns is important for diagnosis of lung adenocarcinoma and prediction of prognosis. However, the growth patterns of lung adenocarcinoma tissue are very dependent on the spatial organization of cells. Deep learning for lung tumor histopathological image analysis often uses convolutional neural networks to automatically extract features, ignoring this spatial relationship. In this paper, a novel fully automated framework is proposed for growth pattern evaluation in lung adenocarcinoma. Specifically, the proposed method uses graph convolutional networks to extract cell structural features; that is, cells are extracted and graph structures are constructed based on histopathological image data without graph structure. A deep neural network is then used to extract the global semantic features of histopathological images to complement the cell structural features obtained in the previous step. Finally, the structural features and semantic features are fused to achieve growth pattern prediction. Experimental studies on several datasets validate our design, demonstrating that methods based on the spatial organization of cells are appropriate for the analysis of growth patterns.
Collapse
Affiliation(s)
- Wei Xiao
- Shandong Provincial Hospital, Shandong University, Jinan, China
- *Correspondence: Wei Xiao,
| | - Yanyun Jiang
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| | - Zhigang Yao
- Department of Pathology, Shandong Provincial Hospital Affiliated to Shandong First Medical University Department of Pathology, Shandong Provincial Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Xiaoming Zhou
- Shandong Provincial Hospital, Shandong University, Jinan, China
| | - Xiaodan Sui
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| | - Yuanjie Zheng
- School of Information Science and Engineering, Shandong Normal University, Jinan, China
| |
Collapse
|
37
|
Zhou X, Wang H, Feng C, Xu R, He Y, Li L, Tu C. Emerging Applications of Deep Learning in Bone Tumors: Current Advances and Challenges. Front Oncol 2022; 12:908873. [PMID: 35928860 PMCID: PMC9345628 DOI: 10.3389/fonc.2022.908873] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/15/2022] [Indexed: 12/12/2022] Open
Abstract
Deep learning is a subfield of state-of-the-art artificial intelligence (AI) technology, and multiple deep learning-based AI models have been applied to musculoskeletal diseases. Deep learning has shown the capability to assist clinical diagnosis and prognosis prediction in a spectrum of musculoskeletal disorders, including fracture detection, cartilage and spinal lesions identification, and osteoarthritis severity assessment. Meanwhile, deep learning has also been extensively explored in diverse tumors such as prostate, breast, and lung cancers. Recently, the application of deep learning emerges in bone tumors. A growing number of deep learning models have demonstrated good performance in detection, segmentation, classification, volume calculation, grading, and assessment of tumor necrosis rate in primary and metastatic bone tumors based on both radiological (such as X-ray, CT, MRI, SPECT) and pathological images, implicating a potential for diagnosis assistance and prognosis prediction of deep learning in bone tumors. In this review, we first summarized the workflows of deep learning methods in medical images and the current applications of deep learning-based AI for diagnosis and prognosis prediction in bone tumors. Moreover, the current challenges in the implementation of the deep learning method and future perspectives in this field were extensively discussed.
Collapse
Affiliation(s)
- Xiaowen Zhou
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Hua Wang
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Chengyao Feng
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Ruilin Xu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Yu He
- Department of Radiology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Lan Li
- Department of Pathology, The Second Xiangya Hospital, Central South University, Changsha, China
| | - Chao Tu
- Department of Orthopaedics, The Second Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Tumor Models and Individualized Medicine, The Second Xiangya Hospital, Central South University, Changsha, China
- *Correspondence: Chao Tu,
| |
Collapse
|
38
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
39
|
Court A, Laville D, Dagher S, Grosjean V, Dal-Col P, Yvorel V, Casteillo F, Bayle-Bleuez S, Vergnon JM, Forest F. Anti-CK7/CK20 Immunohistochemistry Did Not Associate with the Metastatic Site in TTF-1-Negative Lung Cancer. Diagnostics (Basel) 2022; 12:diagnostics12071589. [PMID: 35885495 PMCID: PMC9317980 DOI: 10.3390/diagnostics12071589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Revised: 06/27/2022] [Accepted: 06/28/2022] [Indexed: 11/27/2022] Open
Abstract
Anti-CK7 and anti-CK20 immunohistochemistry is sometimes used to establish a diagnosis of primary lung cancer. We performed a retrospective study on the value of anti-CK7 and anti-CK20 immunohistochemistry in 359 biopsies of patients with suspected lung carcinoma in order to assess the usefulness of these antibodies in the evaluation of lung tumors in biopsies. Our results showed TTF-1 positivity in 73.3% of patients. EGFR mutations and ALK rearrangements were significantly different between TTF-1 positive and TTF-1 negative tumors (p < 0.001 and p = 0.023, respectively). Our results show a significant difference (p < 0.001) between TTF-1 positive and TTF-1 negative carcinomas with a median survival of 21.97 months (CI95% = 17.48−30.9 months) and 6.52 months (CI95% = 3.34−10.3 months), respectively. In the group of TTF-1 negative patients, anti-CK7 and CK20 immunohistochemistry was performed in 70 patients and showed CK7+/CK20- staining in 61 patients (87.1%), CK7-/CK20- in 4 patients (5.7%), CK7+/CK20+ in 3 patients (4.3%), and CK7-/CK20- in 2 patients (2.8%). No specific or molecular pattern was found in these groups of CK7/CK20 combinations. In total, this work brings arguments concerning the uselessness of anti-CK7/CK20 immunohistochemistry in the case of suspicion of primary lung cancer in biopsies.
Collapse
Affiliation(s)
- Alice Court
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - David Laville
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - Sami Dagher
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - Vincent Grosjean
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - Pierre Dal-Col
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - Violaine Yvorel
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
- Department of Molecular Biology of Solid Tumors, University Hospital of Saint Etienne, 42270 Saint Etienne, France
| | - François Casteillo
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
| | - Sophie Bayle-Bleuez
- Department of Pneumology, University Hospital of Saint Etienne, 42270 Saint Etienne, France; (S.B.-B.); (J.-M.V.)
| | - Jean-Michel Vergnon
- Department of Pneumology, University Hospital of Saint Etienne, 42270 Saint Etienne, France; (S.B.-B.); (J.-M.V.)
| | - Fabien Forest
- Department of Pathology, University Hospital of Saint Etienne, 42055 Saint Etienne, France; (A.C.); (D.L.); (S.D.); (V.G.); (P.D.-C.); (V.Y.); (F.C.)
- Department of Molecular Biology of Solid Tumors, University Hospital of Saint Etienne, 42270 Saint Etienne, France
- Correspondence: ; Tel.: +33-(0)-4-77-12-77-34; Fax: +33-(0)-4-77-82-82-96
| |
Collapse
|
40
|
Giannikopoulos P, Parham DM. Pediatric Sarcomas: The Next Generation of Molecular Studies. Cancers (Basel) 2022; 14:2515. [PMID: 35626119 PMCID: PMC9139929 DOI: 10.3390/cancers14102515] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/13/2022] [Accepted: 05/13/2022] [Indexed: 02/04/2023] Open
Abstract
Pediatric sarcomas constitute one of the largest groups of childhood cancers, following hematopoietic, neural, and renal lesions. Partly because of their diversity, they continue to offer challenges in diagnosis and treatment. In spite of the diagnostic, nosologic, and therapeutic gains made with genetic technology, newer means for investigation are needed. This article reviews emerging technology being used to study human neoplasia and how these methods might be applicable to pediatric sarcomas. Methods reviewed include single cell RNA sequencing (scRNAseq), spatial multi-omics, high-throughput functional genomics, and clustered regularly interspersed short palindromic sequence-Cas9 (CRISPR-Cas9) technology. In spite of these advances, the field continues to be challenged by a dearth of properly annotated materials, particularly from recurrences and metastases and pre- and post-treatment samples.
Collapse
Affiliation(s)
| | - David M. Parham
- Department of Anatomic Pathology, Children’s Hospital Los Angeles, Los Angeles, CA 90027, USA
- Department of Pathology, University of Southern California Keck School of Medicine, Los Angeles, CA 90033, USA
| |
Collapse
|
41
|
Communicator-Driven Data Preprocessing Improves Deep Transfer Learning of Histopathological Prediction of Pancreatic Ductal Adenocarcinoma. Cancers (Basel) 2022; 14:cancers14081964. [PMID: 35454869 PMCID: PMC9031738 DOI: 10.3390/cancers14081964] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 03/31/2022] [Accepted: 04/01/2022] [Indexed: 12/02/2022] Open
Abstract
Simple Summary Pancreatic cancer has a dismal prognosis and its diagnosis can be challenging. Histopathological slides can be digitalized and their analysis can then be supported by computer algorithms. For this purpose, computer algorithms (neural networks) need to be trained to detect the desired tissue type (e.g., pancreatic cancer). However, raw training data often contain many different tissue types. Here we show a preprocessing step using two communicators that sort unfitting tissue tiles into a new dataset class. Using the improved dataset neural networks distinguished pancreatic cancer from other tissue types on digitalized histopathological slides including lymph node metastases. Abstract Pancreatic cancer is a fatal malignancy with poor prognosis and limited treatment options. Early detection in primary and secondary locations is critical, but fraught with challenges. While digital pathology can assist with the classification of histopathological images, the training of such networks always relies on a ground truth, which is frequently compromised as tissue sections contain several types of tissue entities. Here we show that pancreatic cancer can be detected on hematoxylin and eosin (H&E) sections by convolutional neural networks using deep transfer learning. To improve the ground truth, we describe a preprocessing data clean-up process using two communicators that were generated through existing and new datasets. Specifically, the communicators moved image tiles containing adipose tissue and background to a new data class. Hence, the original dataset exhibited improved labeling and, consequently, a higher ground truth accuracy. Deep transfer learning of a ResNet18 network resulted in a five-class accuracy of about 94% on test data images. The network was validated with independent tissue sections composed of healthy pancreatic tissue, pancreatic ductal adenocarcinoma, and pancreatic cancer lymph node metastases. The screening of different models and hyperparameter fine tuning were performed to optimize the performance with the independent tissue sections. Taken together, we introduce a step of data preprocessing via communicators as a means of improving the ground truth during deep transfer learning and hyperparameter tuning to identify pancreatic ductal adenocarcinoma primary tumors and metastases in histological tissue sections.
Collapse
|
42
|
Deep Learning Facilitates Distinguishing Histologic Subtypes of Pulmonary Neuroendocrine Tumors on Digital Whole-Slide Images. Cancers (Basel) 2022; 14:cancers14071740. [PMID: 35406511 PMCID: PMC8996915 DOI: 10.3390/cancers14071740] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Revised: 03/13/2022] [Accepted: 03/25/2022] [Indexed: 02/01/2023] Open
Abstract
Simple Summary Challenges persist in diagnosing pulmonary neuroendocrine tumors. Our case study shows that deep learning combined with convolutional neural networks has the potential to assist in the diagnosis of pulmonary neuroendocrine tumors from digital whole-slide images. Abstract The histological distinction of lung neuroendocrine carcinoma, including small cell lung carcinoma (SCLC), large cell neuroendocrine carcinoma (LCNEC) and atypical carcinoid (AC), can be challenging in some cases, while bearing prognostic and therapeutic significance. To assist pathologists with the differentiation of histologic subtyping, we applied a deep learning classifier equipped with a convolutional neural network (CNN) to recognize lung neuroendocrine neoplasms. Slides of primary lung SCLC, LCNEC and AC were obtained from the Laboratory of Clinical and Experimental Pathology (University Hospital Nice, France). Three thoracic pathologists blindly established gold standard diagnoses. The HALO-AI module (Indica Labs, UK) trained with 18,752 image tiles extracted from 60 slides (SCLC = 20, LCNEC = 20, AC = 20 cases) was then tested on 90 slides (SCLC = 26, LCNEC = 22, AC = 13 and combined SCLC with LCNEC = 4 cases; NSCLC = 25 cases) by F1-score and accuracy. A HALO-AI correct area distribution (AD) cutoff of 50% or more was required to credit the CNN with the correct diagnosis. The tumor maps were false colored and displayed side by side to original hematoxylin and eosin slides with superimposed pathologist annotations. The trained HALO-AI yielded a mean F1-score of 0.99 (95% CI, 0.939–0.999) on the testing set. Our CNN model, providing further larger validation, has the potential to work side by side with the pathologist to accurately differentiate between the different lung neuroendocrine carcinoma in challenging cases.
Collapse
|
43
|
Prabhu S, Prasad K, Robels-Kelly A, Lu X. AI-based carcinoma detection and classification using histopathological images: A systematic review. Comput Biol Med 2022; 142:105209. [DOI: 10.1016/j.compbiomed.2022.105209] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/01/2022] [Accepted: 01/01/2022] [Indexed: 02/07/2023]
|
44
|
Abstract
This overview of the molecular pathology of lung cancer includes a review of the most salient molecular alterations of the genome, transcriptome, and the epigenome. The insights provided by the growing use of next-generation sequencing (NGS) in lung cancer will be discussed, and interrelated concepts such as intertumor heterogeneity, intratumor heterogeneity, tumor mutational burden, and the advent of liquid biopsy will be explored. Moreover, this work describes how the evolving field of molecular pathology refines the understanding of different histologic phenotypes of non-small-cell lung cancer (NSCLC) and the underlying biology of small-cell lung cancer. This review will provide an appreciation for how ongoing scientific findings and technologic advances in molecular pathology are crucial for development of biomarkers, therapeutic agents, clinical trials, and ultimately improved patient care.
Collapse
Affiliation(s)
- James J Saller
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| | - Theresa A Boyle
- Departments of Pathology and Thoracic Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, Florida 33612, USA
| |
Collapse
|
45
|
Li Y, Chen D, Wu X, Yang W, Chen Y. A narrative review of artificial intelligence-assisted histopathologic diagnosis and decision-making for non-small cell lung cancer: achievements and limitations. J Thorac Dis 2022; 13:7006-7020. [PMID: 35070383 PMCID: PMC8743410 DOI: 10.21037/jtd-21-806] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2021] [Accepted: 12/01/2021] [Indexed: 12/12/2022]
Abstract
Objective To summarize the current evidence regarding the applications, workflow, and limitations of artificial intelligence (AI) in the management of patients pathologically-diagnosed with lung cancer. Background Lung cancer is one of the most common cancers and the leading cause of cancer-related deaths worldwide. AI technologies have been applied to daily medical workflow and have achieved an excellent performance in predicting histopathologic subtypes, analyzing gene mutation profiles, and assisting in clinical decision-making for lung cancer treatment. More advanced deep learning for classifying pathologic images with minimal human interactions has been developed in addition to the conventional machine learning scheme. Methods Studies were identified by searching databases, including PubMed, EMBASE, Web of Science, and Cochrane Library, up to February 2021 without language restrictions. Conclusions A number of studies have evaluated AI pipelines and confirmed that AI is robust and efficacious in lung cancer diagnosis and decision-making, demonstrating that AI models are a useful tool for assisting oncologists in health management. Although several limitations that pose an obstacle for the widespread use of AI schemes persist, the unceasing refinement of AI techniques is poised to overcome such problems. Thus, AI technology is a promising tool for use in diagnosing and managing lung cancer.
Collapse
Affiliation(s)
- Yongzhong Li
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Donglai Chen
- Department of Thoracic Surgery, Shanghai Pulmonary Hospital, Tongji University, School of Medicine, Shanghai, China
| | - Xuejie Wu
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Wentao Yang
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Yongbing Chen
- Department of Thoracic Surgery, the Second Affiliated Hospital of Soochow University, Suzhou, China
| |
Collapse
|
46
|
Farahmand S, Fernandez AI, Ahmed FS, Rimm DL, Chuang JH, Reisenbichler E, Zarringhalam K. Deep learning trained on hematoxylin and eosin tumor region of Interest predicts HER2 status and trastuzumab treatment response in HER2+ breast cancer. Mod Pathol 2022; 35:44-51. [PMID: 34493825 DOI: 10.1038/s41379-021-00911-w] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2021] [Revised: 08/13/2021] [Accepted: 08/13/2021] [Indexed: 12/19/2022]
Abstract
The current standard of care for many patients with HER2-positive breast cancer is neoadjuvant chemotherapy in combination with anti-HER2 agents, based on HER2 amplification as detected by in situ hybridization (ISH) or protein immunohistochemistry (IHC). However, hematoxylin & eosin (H&E) tumor stains are more commonly available, and accurate prediction of HER2 status and anti-HER2 treatment response from H&E would reduce costs and increase the speed of treatment selection. Computational algorithms for H&E have been effective in predicting a variety of cancer features and clinical outcomes, including moderate success in predicting HER2 status. In this work, we present a novel convolutional neural network (CNN) approach able to predict HER2 status with increased accuracy over prior methods. We trained a CNN classifier on 188 H&E whole slide images (WSIs) manually annotated for tumor Regions of interest (ROIs) by our pathology team. Our classifier achieved an area under the curve (AUC) of 0.90 in cross-validation of slide-level HER2 status and 0.81 on an independent TCGA test set. Within slides, we observed strong agreement between pathologist annotated ROIs and blinded computational predictions of tumor regions / HER2 status. Moreover, we trained our classifier on pre-treatment samples from 187 HER2+ patients that subsequently received trastuzumab therapy. Our classifier achieved an AUC of 0.80 in a five-fold cross validation. Our work provides an H&E-based algorithm that can predict HER2 status and trastuzumab response in breast cancer at an accuracy that may benefit clinical evaluations.
Collapse
Affiliation(s)
- Saman Farahmand
- University of Massachusetts-Boston, Department of Mathematics, Boston, MA, USA.,University of Massachusetts-Boston, Computational Sciences PhD Program, Boston, MA, USA
| | - Aileen I Fernandez
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Fahad Shabbir Ahmed
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - David L Rimm
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA
| | - Jeffrey H Chuang
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, USA. .,UCONN Health, Department of Genetics and Genome Sciences, Farmington, CT, USA.
| | - Emily Reisenbichler
- Yale University, Yale School of Medicine, Department of Pathology, New Haven, CT, USA.
| | - Kourosh Zarringhalam
- University of Massachusetts-Boston, Department of Mathematics, Boston, MA, USA. .,University of Massachusetts-Boston, Computational Sciences PhD Program, Boston, MA, USA.
| |
Collapse
|
47
|
Wang D, Zhao J, Zhang R, Yan Q, Zhou L, Han X, Qi Y, Yu D. The value of CT radiomic in differentiating mycoplasma pneumoniae pneumonia from streptococcus pneumoniae pneumonia with similar consolidation in children under 5 years. Front Pediatr 2022; 10:953399. [PMID: 36245722 PMCID: PMC9554402 DOI: 10.3389/fped.2022.953399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 08/17/2022] [Indexed: 02/03/2023] Open
Abstract
OBJECTIVE To investigate the value of CT radiomics in the differentiation of mycoplasma pneumoniae pneumonia (MPP) from streptococcus pneumoniae pneumonia (SPP) with similar CT manifestations in children under 5 years. METHODS A total of 102 children with MPP (n = 52) or SPP (n = 50) with similar consolidation and surrounding halo on CT images in Qilu Hospital and Qilu Children's Hospital between January 2017 and March 2022 were enrolled in the retrospective study. Radiomic features of the both lesions on plain CT images were extracted including the consolidation part of the pneumonia or both consolidation and surrounding halo area which were respectively delineated at region of interest (ROI) areas on the maximum axial image. The training cohort (n = 71) and the validation cohort (n = 31) were established by stratified random sampling at a ratio of 7:3. By means of variance threshold, the effective radiomics features, SelectKBest and least absolute shrinkage and selection operator (LASSO) regression method were employed for feature selection and combined to calculate the radiomics score (Rad-score). Six classifiers, including k-nearest neighbor (KNN), support vector machine (SVM), extreme gradient boosting (XGBoost), random forest (RF), logistic regression (LR), and decision tree (DT) were used to construct the models based on radiomic features. The diagnostic performance of these models and the radiomic nomogram was estimated and compared using the area under the receiver operating characteristic (ROC) curve (AUC), and the decision curve analysis (DCA) was used to evaluate which model achieved the most net benefit. RESULTS RF outperformed other classifiers and was selected as the backbone in the classifier with the consolidation + the surrounding halo was taken as ROI to differentiate MPP from SPP in validation cohort. The AUC value of MPP in validation cohort was 0.822, the sensitivity and specificity were 0.81 and 0.81, respectively. CONCLUSION The RF model has the best classification efficiency in the identification of MPP from SPP in children, and the ROI with both consolidation and surrounding halo is most suitable for the delineation.
Collapse
Affiliation(s)
- Dongdong Wang
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Jianshe Zhao
- Department of Radiology, Children's Hospital Affiliated to Shandong University, Jinan, China
| | - Ran Zhang
- Huiying Medical Technology (Beijing) Co., Ltd., Beijing, China
| | - Qinghu Yan
- Department of Ultrasound, Shandong Public Health Clinical Center, Jinan, China
| | - Lu Zhou
- Department of Cardiac Surgery ICU, Qilu Hospital of Shandong University, Jinan, China
| | - Xiaoyu Han
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Yafei Qi
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| | - Dexin Yu
- Department of Radiology, Qilu Hospital, Cheeloo College of Medicine, Shandong University, Jinan, China
| |
Collapse
|
48
|
Using a convolutional neural network for classification of squamous and non-squamous non-small cell lung cancer based on diagnostic histopathology HES images. Sci Rep 2021; 11:23912. [PMID: 34903781 PMCID: PMC8669012 DOI: 10.1038/s41598-021-03206-x] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Accepted: 11/22/2021] [Indexed: 02/07/2023] Open
Abstract
Histological stratification in metastatic non-small cell lung cancer (NSCLC) is essential to properly guide therapy. Morphological evaluation remains the basis for subtyping and is completed by additional immunohistochemistry labelling to confirm the diagnosis, which delays molecular analysis and utilises precious sample. Therefore, we tested the capacity of convolutional neural networks (CNNs) to classify NSCLC based on pathologic HES diagnostic biopsies. The model was estimated with a learning cohort of 132 NSCLC patients and validated on an external validation cohort of 65 NSCLC patients. Based on image patches, a CNN using InceptionV3 architecture was trained and optimized to classify NSCLC between squamous and non-squamous subtypes. Accuracies of 0.99, 0.87, 0.85, 0.85 was reached in the training, validation and test sets and in the external validation cohort. At the patient level, the CNN model showed a capacity to predict the tumour histology with accuracy of 0.73 and 0.78 in the learning and external validation cohorts respectively. Selecting tumour area using virtual tissue micro-array improved prediction, with accuracy of 0.82 in the external validation cohort. This study underlines the capacity of CNN to predict NSCLC subtype with good accuracy and to be applied to small pathologic samples without annotation.
Collapse
|
49
|
Zhan X, Long H, Gou F, Duan X, Kong G, Wu J. A Convolutional Neural Network-Based Intelligent Medical System with Sensors for Assistive Diagnosis and Decision-Making in Non-Small Cell Lung Cancer. SENSORS 2021; 21:s21237996. [PMID: 34884000 PMCID: PMC8659811 DOI: 10.3390/s21237996] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Revised: 11/26/2021] [Accepted: 11/28/2021] [Indexed: 12/15/2022]
Abstract
In many regions of the world, early diagnosis of non-small cell lung cancer (NSCLC) is a major challenge due to the large population and lack of medical resources, which is difficult toeffectively address via limited physician manpower alone. Therefore, we developed a convolutional neural network (CNN)-based assisted diagnosis and decision-making intelligent medical system with sensors. This system analyzes NSCLC patients' medical records using sensors to assist staging a diagnosis and provides recommended treatment plans to physicians. To address the problem of unbalanced case samples across pathological stages, we used transfer learning and dynamic sampling techniques to reconstruct and iteratively train the model to improve the accuracy of the prediction system. In this paper, all data for training and testing the system were obtained from the medical records of 2,789,675 patients with NSCLC, which were recorded in three hospitals in China over a five-year period. When the number of case samples reached 8000, the system achieved an accuracy rate of 0.84, which is already close to that of the doctors (accuracy: 0.86). The experimental results proved that the system can quickly and accurately analyze patient data and provide decision information support for physicians.
Collapse
Affiliation(s)
- Xiangbing Zhan
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China; (X.Z.); (X.D.); (G.K.)
| | - Huiyun Long
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China; (X.Z.); (X.D.); (G.K.)
- Correspondence: (H.L.); (J.W.)
| | - Fangfang Gou
- School of Computer Science and Engineering, Central South University, Changsha 410083, China;
| | - Xun Duan
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China; (X.Z.); (X.D.); (G.K.)
| | - Guangqian Kong
- State Key Laboratory of Public Big Data, College of Computer Science and Technology, Guizhou University, Guiyang 550025, China; (X.Z.); (X.D.); (G.K.)
| | - Jia Wu
- School of Computer Science and Engineering, Central South University, Changsha 410083, China;
- Research Center for Artificial Intelligence, Monash University, Clayton, VIC 3800, Australia
- Correspondence: (H.L.); (J.W.)
| |
Collapse
|
50
|
Ektefaie Y, Yuan W, Dillon DA, Lin NU, Golden JA, Kohane IS, Yu KH. Integrative multiomics-histopathology analysis for breast cancer classification. NPJ Breast Cancer 2021; 7:147. [PMID: 34845230 PMCID: PMC8630188 DOI: 10.1038/s41523-021-00357-y] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Accepted: 11/03/2021] [Indexed: 02/07/2023] Open
Abstract
Histopathologic evaluation of biopsy slides is a critical step in diagnosing and subtyping breast cancers. However, the connections between histology and multi-omics status have never been systematically explored or interpreted. We developed weakly supervised deep learning models over hematoxylin-and-eosin-stained slides to examine the relations between visual morphological signal, clinical subtyping, gene expression, and mutation status in breast cancer. We first designed fully automated models for tumor detection and pathology subtype classification, with the results validated in independent cohorts (area under the receiver operating characteristic curve ≥ 0.950). Using only visual information, our models achieved strong predictive performance in estrogen/progesterone/HER2 receptor status, PAM50 status, and TP53 mutation status. We demonstrated that these models learned lymphocyte-specific morphological signals to identify estrogen receptor status. Examination of the PAM50 cohort revealed a subset of PAM50 genes whose expression reflects cancer morphology. This work demonstrates the utility of deep learning-based image models in both clinical and research regimes, through its ability to uncover connections between visual morphology and genetic statuses.
Collapse
Affiliation(s)
- Yasha Ektefaie
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck Street, Boston, MA, 02115, USA
| | - William Yuan
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck Street, Boston, MA, 02115, USA
| | - Deborah A Dillon
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA
| | - Nancy U Lin
- Department of Medicine, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA, 02215, USA
| | - Jeffrey A Golden
- Department of Pathology, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
- Burns and Allen Research Institute, Cedars-Sinai Medical Center, 8700 Beverly Blvd, Los Angeles, CA, 90048, USA
| | - Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck Street, Boston, MA, 02115, USA
| | - Kun-Hsing Yu
- Department of Biomedical Informatics, Harvard Medical School, 10 Shattuck Street, Boston, MA, 02115, USA.
- Department of Pathology, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, 02115, USA.
| |
Collapse
|