1
|
Aggarwal A, Bharadwaj S, Corredor G, Pathak T, Badve S, Madabhushi A. Artificial intelligence in digital pathology - time for a reality check. Nat Rev Clin Oncol 2025; 22:283-291. [PMID: 39934323 DOI: 10.1038/s41571-025-00991-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/24/2025] [Indexed: 02/13/2025]
Abstract
The past decade has seen the introduction of artificial intelligence (AI)-based approaches aimed at optimizing several workflows across many medical specialties. In clinical oncology, the most promising applications include those involving image analysis, such as digital pathology. In this Perspective, we provide a comprehensive examination of the developments in AI in digital pathology between 2019 and 2024. We evaluate the current landscape from the lens of technological innovations, regulatory trends, deployment and implementation, reimbursement and commercial implications. We assess the technological advances that have driven improvements in AI, enabling more robust and scalable solutions for digital pathology. We also examine regulatory developments, in particular those affecting in-house devices and laboratory-developed tests, which are shaping the landscape of AI-based tools in digital pathology. Finally, we discuss the role of reimbursement frameworks and commercial investment in the clinical adoption of AI-based technologies. In this Perspective, we highlight both the progress and challenges in AI-driven digital pathology over the past 5 years, outlining the path forward for its adoption into routine practice in clinical oncology.
Collapse
Affiliation(s)
- Arpit Aggarwal
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Satvika Bharadwaj
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Germán Corredor
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA
| | - Tilak Pathak
- Department of Biomedical Engineering, Emory University, Atlanta, GA, USA
| | - Sunil Badve
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA, USA
| | - Anant Madabhushi
- Wallace H. Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA.
- Atlanta Veterans Affairs Medical Center, Atlanta, GA, USA.
| |
Collapse
|
2
|
Li MY, Pan Y, Lv Y, Ma H, Sun PL, Gao HW. Digital pathology and artificial intelligence in renal cell carcinoma focusing on feature extraction: a literature review. Front Oncol 2025; 15:1516264. [PMID: 39926279 PMCID: PMC11802434 DOI: 10.3389/fonc.2025.1516264] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 01/06/2025] [Indexed: 02/11/2025] Open
Abstract
The integrated application of artificial intelligence (AI) and digital pathology (DP) technology has opened new avenues for advancements in oncology and molecular pathology. Consequently, studies in renal cell carcinoma (RCC) have emerged, highlighting potential in histological subtype classification, molecular aberration identification, and outcome prediction by extracting high-throughput features. However, reviews of these studies are still rare. To address this gap, we conducted a thorough literature review on DP and AI applications in RCC through database searches. Notably, we found that AI models based on deep learning achieved area under the curve (AUC) of over 0.93 in subtype classification, 0.89-0.96 in grading of clear cell RCC, 0.70-0,89 in molecular prediction, and over 0.78 in survival prediction. This review finally discussed the current state of researches and potential future directions.
Collapse
Affiliation(s)
- Ming-Yue Li
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yu Pan
- Department of Urology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Yang Lv
- Department of Orthopedics, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - He Ma
- Department of Anesthesiology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Ping-Li Sun
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Hong-Wen Gao
- Department of Pathology, The Second Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
3
|
Miller JW, Johnson JS, Guske C, Mannam G, Hatoum F, Nassar M, Potez M, Fazili A, Spiess PE, Chahoud J. Immune-Based and Novel Therapies in Variant Histology Renal Cell Carcinomas. Cancers (Basel) 2025; 17:326. [PMID: 39858107 PMCID: PMC11763753 DOI: 10.3390/cancers17020326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/13/2025] [Accepted: 01/18/2025] [Indexed: 01/27/2025] Open
Abstract
Renal cell carcinoma (RCC) is a heterogeneous disease that represents the most common type of kidney cancer. The classification of RCC is primarily based on distinct morphological and molecular characteristics, with two broad categories: clear cell RCC (ccRCC) and non-clear cell RCC (nccRCC). Clear cell RCC is the predominant subtype, representing about 70-80% of all RCC cases, while non-clear cell subtypes collectively make up the remaining 20-30%. Non-clear cell RCC encompasses many histopathological variants, each with unique biological and clinical characteristics. Additionally, any RCC subtype can undergo sarcomatoid dedifferentiation, which is associated with poor prognosis and rapid disease progression. Recent advances in molecular profiling have also led to the identification of molecularly defined variants, further highlighting the complexity of this disease. While immunotherapy has shown efficacy in some RCC variants and subpopulations, significant gaps remain in the treatment of rare subtypes. This review explores the outcomes of immunotherapy across RCC subtypes, including rare variants, and highlights opportunities for improving care through novel therapies, biomarker-driven approaches, and inclusive clinical trial designs.
Collapse
Affiliation(s)
- Justin W. Miller
- USF Health Morsani College of Medicine, Tampa, FL 33602, USA; (J.W.M.)
| | - Jeffrey S. Johnson
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Christopher Guske
- USF Health Morsani College of Medicine, Tampa, FL 33602, USA; (J.W.M.)
| | - Gowtam Mannam
- USF Health Morsani College of Medicine, Tampa, FL 33602, USA; (J.W.M.)
| | - Firas Hatoum
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | | | - Marine Potez
- Department of Immunology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Adnan Fazili
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Philippe E. Spiess
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| | - Jad Chahoud
- Department of Genitourinary Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL 33612, USA
| |
Collapse
|
4
|
Sun Y, Yang S. Autoimmune side-effect of immunotherapy in lung cancer treatment revealed from large-scale cohort. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.12.03.24318450. [PMID: 39677478 PMCID: PMC11643146 DOI: 10.1101/2024.12.03.24318450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/17/2024]
Abstract
Although immune checkpoint inhibitors have illustrated strong benefits in patient survival and have been widely acknowledged in treating lung cancer, they may be subject to increased risk of immune-related adverse effects (irAEs). Although existing literature have studied the mechanisms of irAEs of immunotherapy, it is difficult to quantify such effect, especially at a large-scale real-world population level. In this paper, the autoimmune-related risk of multiple immune checkpoint inhibitors is compared with that of chemotherapy based on Medicaid and CHIP TAF (T-MSIS Analytic File) data of over 100,000 patient samples from 2012 to 2018. Results show that the irAEs of immunotherapy is significantly higher than chemotherapy in both unadjusted and adjusted samples from the dataset. Analysis on subpopulation and specific disease types further shows that certain immunotherapy treatments are associated with higher risk of irAEs, and the risk of certain autoimmune diseases may vary. We also illustrate the robustness of our conclusion through additional sensitivity analysis, confirming the necessity of keeping track of autoimmune side effects of immune checkpoint inhibitors for medicine researchers. Our methods are also available to evaluate effectiveness and side effects of novel therapies at a large-scale population level.
Collapse
Affiliation(s)
- Yan Sun
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, 755 Ferst Dr NW, Atlanta, 30318, Georgia, USA
| | - Shihao Yang
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, 755 Ferst Dr NW, Atlanta, 30318, Georgia, USA
| |
Collapse
|
5
|
Fan J, Lv T, Wang P, Hong X, Liu Y, Jiang C, Ni J, Li L, Pan X. DCDiff: Dual-Granularity Cooperative Diffusion Models for Pathology Image Analysis. IEEE TRANSACTIONS ON MEDICAL IMAGING 2024; 43:4393-4403. [PMID: 38941198 DOI: 10.1109/tmi.2024.3420804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/30/2024]
Abstract
Whole Slide Images (WSIs) are paramount in the medical field, with extensive applications in disease diagnosis and treatment. Recently, many deep-learning methods have been used to classify WSIs. However, these methods are inadequate for accurately analyzing WSIs as they treat regions in WSIs as isolated entities and ignore contextual information. To address this challenge, we propose a novel Dual-Granularity Cooperative Diffusion Model (DCDiff) for the precise classification of WSIs. Specifically, we first design a cooperative forward and reverse diffusion strategy, utilizing fine-granularity and coarse-granularity to regulate each diffusion step and gradually improve context awareness. To exchange information between granularities, we propose a coupled U-Net for dual-granularity denoising, which efficiently integrates dual-granularity consistency information using the designed Fine- and Coarse-granularity Cooperative Aware (FCCA) model. Ultimately, the cooperative diffusion features extracted by DCDiff can achieve cross-sample perception from the reconstructed distribution of training samples. Experiments on three public WSI datasets show that the proposed method can achieve superior performance over state-of-the-art methods. The code is available at https://github.com/hemo0826/DCDiff.
Collapse
|
6
|
Chen S, Wang X, Zhang J, Jiang L, Gao F, Xiang J, Yang S, Yang W, Zheng J, Han X. Deep learning-based diagnosis and survival prediction of patients with renal cell carcinoma from primary whole slide images. Pathology 2024; 56:951-960. [PMID: 39168777 DOI: 10.1016/j.pathol.2024.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 05/06/2024] [Accepted: 05/20/2024] [Indexed: 08/23/2024]
Abstract
There is an urgent clinical demand to explore novel diagnostic and prognostic biomarkers for renal cell carcinoma (RCC). We proposed deep learning-based artificial intelligence strategies. The study included 1752 whole slide images from multiple centres. Based on the pixel-level of RCC segmentation, the diagnosis diagnostic model achieved an area under the receiver operating characteristic curve (AUC) of 0.977 (95% CI 0.969-0.984) in the external validation cohort. In addition, our diagnostic model exhibited excellent performance in the differential diagnosis of RCC from renal oncocytoma, which achieved an AUC of 0.951 (0.922-0.972). The graderisk for the recognition of high-grade tumour achieved AUCs of 0.840 (0.805-0.871) in the Cancer Genome Atlas (TCGA) cohort, 0.857 (0.813-0.894) in the Shanghai General Hospital (General) cohort, and 0.894 (0.842-0.933) in the Clinical Proteomic Tumor Analysis Consortium (CPTAC) cohort, for the recognition of high-grade tumour. The OSrisk for predicting 5-year survival status achieved an AUC of 0.784 (0.746-0.819) in the TCGA cohort, which was further verified in the independent general cohort and the CPTAC cohort, with AUCs of 0.774 (0.723-0.820) and 0.702 (0.632-0.765), respectively. Moreover, the competing-risk nomogram (CRN) showed its potential to be a prognostic indicator, with a hazard ratio (HR) of 5.664 (3.893-8.239, p<0.0001), outperforming other traditional clinical prognostic indicators. Kaplan-Meier survival analysis further illustrated that our CRN could significantly distinguish patients with high survival risk. Deep learning-based artificial intelligence could be a useful tool for clinicians to diagnose and predict the prognosis of RCC patients, thus improving the process of individualised treatment.
Collapse
Affiliation(s)
- Siteng Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiyue Wang
- Department of Radiation Oncology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Liren Jiang
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Feng Gao
- Department of Pathology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xiao Han
- Tencent AI Lab, Shenzhen, China.
| |
Collapse
|
7
|
Qixin Y, Jing H, Jiang H, Xueyang L, Lu Y, Yuehua L. Transcriptome-based network analysis related to regulatory T cells infiltration identified RCN1 as a potential biomarker for prognosis in clear cell renal cell carcinoma. BioData Min 2024; 17:51. [PMID: 39543725 PMCID: PMC11566375 DOI: 10.1186/s13040-024-00404-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/31/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND Regulatory T cells (Tregs) play a critical role in shaping the immunosuppressive microenvironment within tumors. Investigating the role of Tregs in Clear cell renal cell carcinoma (ccRCC) is crucial for identifying prognostic markers and therapeutic targets for ccRCC. METHODS Weighted gene co-expression network analysis (WGCNA) was utilized to pinpoint modules related to Treg infiltration in TCGA-KIRC samples. Following this, consensus clustering was employed to derive two clusters associated with Treg infiltration in ccRCC. A prognostic model was then developed using the gene module associated with Treg infiltration. We then evaluated the ability of the prognostic model to predict ccRCC overall survival and demonstrated that RCN1 can be used as a target to predict ccRCC prognosis. RESULTS We deduce that the two clusters associated with Treg infiltration exhibit distinct compositions of the immune microenvironment, pathway activations, prognosis, and drug sensitivities commonly utilized in ccRCC treatment. Furthermore, a 7-gene model risk score, developed based on ccRCC Treg infiltration, proved to be a reliable prognostic marker in both training and validation cohorts. Additionally, survival analysis indicated that RCN1 serves as a reliable prognostic factor for ccRCC. Single-cell sequencing analysis revealed that RCN1 is predominantly expressed in tumor cells. A pan-cancer analysis highlighted that RCN1 is linked with poor prognosis and the activation of inflammatory response pathways across various cancers. CONCLUSION We developed a prognostic model associated with Treg infiltration, which facilitates the clinical categorization of ccRCC progression. Moreover, our findings underscore the significant potential of RCN1 as a ccRCC biomarker.
Collapse
Affiliation(s)
- Yang Qixin
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Huang Jing
- Department of Rehabilitation, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - He Jiang
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Liu Xueyang
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Yu Lu
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China
| | - Li Yuehua
- Department of Urology, University-Town Hospital of Chongqing Medical University, Chongqing, 401331, P.R. China.
| |
Collapse
|
8
|
Durant AM, Medero RC, Briggs LG, Choudry MM, Nguyen M, Channar A, Ghaffar U, Banerjee I, Bin Riaz I, Abdul-Muhsin H. The Current Application and Future Potential of Artificial Intelligence in Renal Cancer. Urology 2024; 193:157-163. [PMID: 39029807 DOI: 10.1016/j.urology.2024.07.010] [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: 04/04/2024] [Revised: 06/12/2024] [Accepted: 07/06/2024] [Indexed: 07/21/2024]
Abstract
Artificial intelligence (AI) is the integration of human tasks into machine processes. The role of AI in kidney cancer evaluation, management, and outcome predictions are constantly evolving. We performed a narrative review utilizing PubMed electronic database to query AI as a method of analysis in kidney cancer research. Key search-words included: Artificial Intelligence, Supervised/Unsupervised Machine Learning, Deep Learning, Natural Language Processing, Neural Networks, radiomics, pathomics, and kidney or renal neoplasms or cancer. 72 clinically relevant and impactful studies related to imaging, histopathology, and outcomes were recognized. We anticipate the incorporation of AI tools into future clinical decision-making for kidney cancer.
Collapse
Affiliation(s)
- Adri M Durant
- Department of Urology, Mayo Clinic Arizona, Phoenix, AZ.
| | - Ramon Correa Medero
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ
| | | | | | - Mimi Nguyen
- Department of Urology, Mayo Clinic Arizona, Phoenix, AZ
| | - Aneeta Channar
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Arizona, Phoenix, AZ
| | - Umar Ghaffar
- Department of Urology, Mayo Clinic Rochester, Rochester, MN
| | - Imon Banerjee
- School of Computing and Augmented Intelligence, Arizona State University, Tempe, AZ; Department of Radiology, Mayo Clinic Arizona, Scottsdale, AZ
| | - Irbaz Bin Riaz
- Division of Hematology and Oncology, Department of Internal Medicine, Mayo Clinic Arizona, Phoenix, AZ
| | | |
Collapse
|
9
|
Leite KRM, Melo PADS. Artificial Intelligence in Uropathology. Diagnostics (Basel) 2024; 14:2279. [PMID: 39451602 PMCID: PMC11506825 DOI: 10.3390/diagnostics14202279] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 09/25/2024] [Accepted: 10/10/2024] [Indexed: 10/26/2024] Open
Abstract
The global population is currently at unprecedented levels, with an estimated 7.8 billion people inhabiting the planet. We are witnessing a rise in cancer cases, attributed to improved control of cardiovascular diseases and a growing elderly population. While this has resulted in an increased workload for pathologists, it also presents an opportunity for advancement. The accurate classification of tumors and identification of prognostic and predictive factors demand specialized expertise and attention. Fortunately, the rapid progression of artificial intelligence (AI) offers new prospects in medicine, particularly in diagnostics such as image and surgical pathology. This article explores the transformative impact of AI in the field of uropathology, with a particular focus on its application in diagnosing, grading, and prognosticating various urological cancers. AI, especially deep learning algorithms, has shown significant potential in improving the accuracy and efficiency of pathology workflows. This comprehensive review is dedicated to providing an insightful overview of the primary data concerning the utilization of AI in diagnosing, predicting prognosis, and determining drug responses for tumors of the urinary tract. By embracing these advancements, we can look forward to improved outcomes and better patient care.
Collapse
Affiliation(s)
- Katia Ramos Moreira Leite
- Laboratory of Medical Investigation, Urology Department, University of São Paulo Medical School, LIM55, Av Dr. Arnando 455, Sao Paulo 01246-903, SP, Brazil;
| | | |
Collapse
|
10
|
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
|
11
|
Chandramohan D, Garapati HN, Nangia U, Simhadri PK, Lapsiwala B, Jena NK, Singh P. Diagnostic accuracy of deep learning in detection and prognostication of renal cell carcinoma: a systematic review and meta-analysis. Front Med (Lausanne) 2024; 11:1447057. [PMID: 39301494 PMCID: PMC11412207 DOI: 10.3389/fmed.2024.1447057] [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: 06/11/2024] [Accepted: 08/07/2024] [Indexed: 09/22/2024] Open
Abstract
INTRODUCTION The prevalence of Renal cell carcinoma (RCC) is increasing among adults. Histopathologic samples obtained after surgical resection or from biopsies of a renal mass require subtype classification for diagnosis, prognosis, and to determine surveillance. Deep learning in artificial intelligence (AI) and pathomics are rapidly advancing, leading to numerous applications such as histopathological diagnosis. In our meta-analysis, we assessed the pooled diagnostic performances of deep neural network (DNN) frameworks in detecting RCC subtypes and to predicting survival. METHODS A systematic search was done in PubMed, Google Scholar, Embase, and Scopus from inception to November 2023. The random effects model was used to calculate the pooled percentages, mean, and 95% confidence interval. Accuracy was defined as the number of cases identified by AI out of the total number of cases, i.e. (True Positive + True Negative)/(True Positive + True Negative + False Positive + False Negative). The heterogeneity between study-specific estimates was assessed by the I 2 statistic. The Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) guidelines were used to conduct and report the analysis. RESULTS The search retrieved 347 studies; 13 retrospective studies evaluating 5340 patients were included in the final analysis. The pooled performance of the DNN was as follows: accuracy 92.3% (95% CI: 85.8-95.9; I 2 = 98.3%), sensitivity 97.5% (95% CI: 83.2-99.7; I 2 = 92%), specificity 89.2% (95% CI: 29.9-99.4; I 2 = 99.6%) and area under the curve 0.91 (95% CI: 0.85-0.97.3; I 2 = 99.6%). Specifically, their accuracy in RCC subtype detection was 93.5% (95% CI: 88.7-96.3; I 2 = 92%), and the accuracy in survival analysis prediction was 81% (95% CI: 67.8-89.6; I 2 = 94.4%). DISCUSSION The DNN showed excellent pooled diagnostic accuracy rates to classify RCC into subtypes and grade them for prognostic purposes. Further studies are required to establish generalizability and validate these findings on a larger scale.
Collapse
Affiliation(s)
- Deepak Chandramohan
- Department of Nephrology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Hari Naga Garapati
- Department of Nephrology, Baptist Medical Center South, Montgomery, AL, United States
| | - Udit Nangia
- Department of Medicine, University Hospital Parma Medical Center, Parma, OH, United States
| | - Prathap K. Simhadri
- Department of Nephrology, The University of Alabama at Birmingham, Birmingham, AL, United States
| | - Boney Lapsiwala
- Department of Internal Medicine, Medical City Arlington, Arlington, TX, United States
| | - Nihar K. Jena
- Department of Cardiology, Trinity Health Oakland Hospital, Pontiac, MI, United States
| | - Prabhat Singh
- Department of Nephrology, Christus Spohn Health System, Corpus Christi, TX, United States
| |
Collapse
|
12
|
Yuan H, Kido T, Hirata M, Ueno K, Imai Y, Chen K, Ren W, Yang L, Chen K, Qu L, Wu Y. New vision of HookEfficientNet deep neural network: Intelligent histopathological recognition system of non-small cell lung cancer. Comput Biol Med 2024; 178:108710. [PMID: 38843570 DOI: 10.1016/j.compbiomed.2024.108710] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Revised: 05/24/2024] [Accepted: 06/03/2024] [Indexed: 07/24/2024]
Abstract
BACKGROUND Efficient and precise diagnosis of non-small cell lung cancer (NSCLC) is quite critical for subsequent targeted therapy and immunotherapy. Since the advent of whole slide images (WSIs), the transition from traditional histopathology to digital pathology has aroused the application of convolutional neural networks (CNNs) in histopathological recognition and diagnosis. HookNet can make full use of macroscopic and microscopic information for pathological diagnosis, but it cannot integrate other excellent CNN structures. The new version of HookEfficientNet is based on a combination of HookNet structure and EfficientNet that performs well in the recognition of general objects. Here, a high-precision artificial intelligence-guided histopathological recognition system was established by HookEfficientNet to provide a basis for the intelligent differential diagnosis of NSCLC. METHODS A total of 216 WSIs of lung adenocarcinoma (LUAD) and 192 WSIs of lung squamous cell carcinoma (LUSC) were recruited from the First Affiliated Hospital of Zhengzhou University. Deep learning methods based on HookEfficientNet, HookNet and EfficientNet B4-B6 were developed and compared with each other using area under the curve (AUC) and the Youden index. Temperature scaling was used to calibrate the heatmap and highlight the cancer region of interest. Four pathologists of different levels blindly reviewed 108 WSIs of LUAD and LUSC, and the diagnostic results were compared with the various deep learning models. RESULTS The HookEfficientNet model outperformed HookNet and EfficientNet B4-B6. After temperature scaling, the HookEfficientNet model achieved AUCs of 0.973, 0.980, and 0.989 and Youden index values of 0.863, 0.899, and 0.922 for LUAD, LUSC and normal lung tissue, respectively, in the testing set. The accuracy of the model was better than the average accuracy from experienced pathologists, and the model was superior to pathologists in the diagnosis of LUSC. CONCLUSIONS HookEfficientNet can effectively recognize LUAD and LUSC with performance superior to that of senior pathologists, especially for LUSC. The model has great potential to facilitate the application of deep learning-assisted histopathological diagnosis for LUAD and LUSC in the future.
Collapse
Affiliation(s)
- Huijie Yuan
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China
| | | | | | - Kengo Ueno
- KYOCERA Communication Systems Co., Ltd, Kyoto, Japan
| | - Yuji Imai
- KYOCERA Communication Systems (Shanghai) Co., Ltd, Shanghai, China
| | - Kangxuan Chen
- KYOCERA Communication Systems (Shanghai) Co., Ltd, Shanghai, China
| | - Wujie Ren
- Henan 863 Software Co., Ltd, Zhengzhou, China
| | - Liang Yang
- Henan 863 Software Co., Ltd, Zhengzhou, China
| | - Kuisheng Chen
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Lingbo Qu
- Henan Joint International Research Laboratory of Green Construction of Functional Molecules and Their Bioanalytical Applications, Zhengzhou University, Zhengzhou, 450001, China
| | - Yongjun Wu
- College of Public Health, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
13
|
Zheng Q, Wang X, Yang R, Fan J, Yuan J, Liu X, Wang L, Xiao Z, Chen Z. Predicting tumor mutation burden and VHL mutation from renal cancer pathology slides with self-supervised deep learning. Cancer Med 2024; 13:e70112. [PMID: 39166457 PMCID: PMC11336896 DOI: 10.1002/cam4.70112] [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: 01/05/2024] [Revised: 05/28/2024] [Accepted: 08/04/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND Tumor mutation burden (TMB) and VHL mutation play a crucial role in the management of patients with clear cell renal cell carcinoma (ccRCC), such as guiding adjuvant chemotherapy and improving clinical outcomes. However, the time-consuming and expensive high-throughput sequencing methods severely limit their clinical applicability. Predicting intratumoral heterogeneity poses significant challenges in biology and clinical settings. Our aimed to develop a self-supervised attention-based multiple instance learning (SSL-ABMIL) model to predict TMB and VHL mutation status from hematoxylin and eosin-stained histopathological images. METHODS We obtained whole slide images (WSIs) and somatic mutation data of 350 ccRCC patients from The Cancer Genome Atlas for developing SSL-ABMIL model. In parallel, 163 ccRCC patients from Clinical Proteomic Tumor Analysis Consortium cohort was used as independent external validation set. We systematically compared three different models (Wang-ABMIL, Ciga-ABMIL, and ImageNet-MIL) for their ability to predict TMB and VHL alterations. RESULTS We first identified two groups of populations with high- and low-TMB (cut-off point = 0.9). In two independent cohorts, the Wang-ABMIL model achieved the highest performance with decent generalization performance (AUROC = 0.83 ± 0.02 and 0.8 ± 0.04 in predicting TMB and VHL, respectively). Attention heatmaps revealed that the Wang-ABMIL model paid the highest attention to tumor regions in high-TMB patients, while in VHL mutation prediction, non-tumor regions were also assigned high attention, particularly the stromal regions infiltrated by lymphocytes. CONCLUSIONS Our results indicated that SSL-ABMIL can effectively extract histological features for predicting TMB and VHL mutation, demonstrating promising results in linking tumor morphology and molecular biology.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xinyu Wang
- Centre for Reproductive ScienceRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Rui Yang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Junjie Fan
- University of Chinese Academy of SciencesBeijingChina
- Trusted Computing and Information Assurance LaboratoryInstitute of Software, Chinese Academy of SciencesBeijingChina
| | - Jingping Yuan
- Department of PathologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Xiuheng Liu
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Lei Wang
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zhuoni Xiao
- Centre for Reproductive ScienceRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zhiyuan Chen
- Department of UrologyRenmin Hospital of Wuhan UniversityWuhanHubeiChina
- Institute of Urologic DiseaseRenmin Hospital of Wuhan UniversityWuhanHubeiChina
| |
Collapse
|
14
|
Khene ZE, Kammerer-Jacquet SF, Bigot P, Rabilloud N, Albiges L, Margulis V, De Crevoisier R, Acosta O, Rioux-Leclercq N, Lotan Y, Rouprêt M, Bensalah K. Clinical Application of Digital and Computational Pathology in Renal Cell Carcinoma: A Systematic Review. Eur Urol Oncol 2024; 7:401-411. [PMID: 37925349 DOI: 10.1016/j.euo.2023.10.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 09/26/2023] [Accepted: 10/24/2023] [Indexed: 11/06/2023]
Abstract
CONTEXT Computational pathology is a new interdisciplinary field that combines traditional pathology with modern technologies such as digital imaging and machine learning to better understand the diagnosis, prognosis, and natural history of many diseases. OBJECTIVE To provide an overview of digital and computational pathology and its current and potential applications in renal cell carcinoma (RCC). EVIDENCE ACQUISITION A systematic review of the English-language literature was conducted using the PubMed, Web of Science, and Scopus databases in December 2022 according to the Preferred Reporting Items for Systematic Reviews and Meta-Analyses guidelines (PROSPERO ID: CRD42023389282). Risk of bias was assessed according to the Prediction Model Study Risk of Bias Assessment Tool. EVIDENCE SYNTHESIS In total, 20 articles were included in the review. All the studies used a retrospective design, and all digital pathology techniques were implemented retrospectively. The studies were classified according to their primary objective: detection, tumor characterization, and patient outcome. Regarding the transition to clinical practice, several studies showed promising potential. However, none presented a comprehensive assessment of clinical utility and implementation. Notably, there was substantial heterogeneity for both the strategies used for model building and the performance metrics reported. CONCLUSIONS This review highlights the vast potential of digital and computational pathology for the detection, classification, and assessment of oncological outcomes in RCC. Preliminary work in this field has yielded promising results. However, these models have not yet reached a stage where they can be integrated into routine clinical practice. PATIENT SUMMARY Computational pathology combines traditional pathology and technologies such as digital imaging and artificial intelligence to improve diagnosis of disease and identify prognostic factors and new biomarkers. The number of studies exploring its potential in kidney cancer is rapidly increasing. However, despite the surge in research activity, computational pathology is not yet ready for widespread routine use.
Collapse
Affiliation(s)
- Zine-Eddine Khene
- Department of Urology, University of Rennes, Rennes, France; Laboratoire Traitement du Signal et de l'Image, Inserm U1099, Université de Rennes 1, Rennes, France; Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Solène-Florence Kammerer-Jacquet
- Laboratoire Traitement du Signal et de l'Image, Inserm U1099, Université de Rennes 1, Rennes, France; Department of Pathology, University of Rennes, Rennes, France
| | - Pierre Bigot
- Department of Urology, University of Angers, Rennes, France
| | - Noémie Rabilloud
- Laboratoire Traitement du Signal et de l'Image, Inserm U1099, Université de Rennes 1, Rennes, France
| | - Laurence Albiges
- Department of Medical Oncology, Gustave Roussy, Villejuif, France
| | - Vitaly Margulis
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Oscar Acosta
- Laboratoire Traitement du Signal et de l'Image, Inserm U1099, Université de Rennes 1, Rennes, France
| | | | - Yair Lotan
- Department of Urology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Morgan Rouprêt
- Department of Urology, La Pitie Salpétrière Hospital, Paris, France
| | - Karim Bensalah
- Department of Urology, University of Rennes, Rennes, France
| |
Collapse
|
15
|
Xu J. Comparing multi-class classifier performance by multi-class ROC analysis: A nonparametric approach. Neurocomputing 2024; 583:127520. [PMID: 38645687 PMCID: PMC11031188 DOI: 10.1016/j.neucom.2024.127520] [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] [Indexed: 04/23/2024]
Abstract
The area under the Receiver Operating Characteristic (ROC) curve (AUC) is a standard metric for quantifying and comparing binary classifiers. Real world applications often require classification into multiple (more than two) classes. For multi-class classifiers that produce class membership scores, a popular multi-class AUC (MAUC) variant is to average the pairwise AUC values [1]. Due to the complicated correlation patterns, the variance of MAUC is often estimated numerically using resampling techniques. This work is a generalization of DeLong's nonparameteric approach for binary AUC analysis [2] to MAUC. We first derive the closed-form expression of the covariance matrix of the pairwise AUCs within a single MAUC. Then by dropping higher order terms, we obtain an approximate covariance matrix with a compact, matrix factorization form, which then serves as the basis for variance estimation of a single MAUC. We further extend this approach to estimate the covariance of correlated MAUCs that arise from multiple competing classifiers. For the special case of binary correlated AUCs, our results coincide with that of DeLong. Our numerical studies confirm the accuracy of the variance and covariance estimates. We provide the source code of the proposed covariance estimation of correlated MAUCs on GitHub (https://tinyurl.com/euj6wvsz) for its easy adoption by machine learning and statistical analysis packages to quantify and compare multi-class classifiers.
Collapse
Affiliation(s)
- Jingyan Xu
- Department of Radiology, Johns Hopkins University, MD, USA
| |
Collapse
|
16
|
Pilva P, Bülow R, Boor P. Deep learning applications for kidney histology analysis. Curr Opin Nephrol Hypertens 2024; 33:291-297. [PMID: 38411024 DOI: 10.1097/mnh.0000000000000973] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
PURPOSE OF REVIEW Nephropathology is increasingly incorporating computational methods to enhance research and diagnostic accuracy. The widespread adoption of digital pathology, coupled with advancements in deep learning, will likely transform our pathology practices. Here, we discuss basic concepts of deep learning, recent applications in nephropathology, current challenges in implementation and future perspectives. RECENT FINDINGS Deep learning models have been developed and tested in various areas of nephropathology, for example, predicting kidney disease progression or diagnosing diseases based on imaging and clinical data. Despite their promising potential, challenges remain that hinder a wider adoption, for example, the lack of prospective evidence and testing in real-world scenarios. SUMMARY Deep learning offers great opportunities to improve quantitative and qualitative kidney histology analysis for research and clinical nephropathology diagnostics. Although exciting approaches already exist, the potential of deep learning in nephropathology is only at its beginning and we can expect much more to come.
Collapse
Affiliation(s)
| | | | - Peter Boor
- Institute of Pathology
- Department of Nephrology and Clinical Immunology, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
17
|
Chen S, Song D, Chen L, Guo T, Jiang B, Liu A, Pan X, Wang T, Tang H, Chen G, Xue Z, Wang X, Zhang N, Zheng J. Artificial intelligence-based non-invasive tumor segmentation, grade stratification and prognosis prediction for clear-cell renal-cell carcinoma. PRECISION CLINICAL MEDICINE 2023; 6:pbad019. [PMID: 38025974 PMCID: PMC10680020 DOI: 10.1093/pcmedi/pbad019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Accepted: 08/07/2023] [Indexed: 12/01/2023] Open
Abstract
Due to the complicated histopathological characteristics of clear-cell renal-cell carcinoma (ccRCC), non-invasive prognosis before operative treatment is crucial in selecting the appropriate treatment. A total of 126 345 computerized tomography (CT) images from four independent patient cohorts were included for analysis in this study. We propose a V Bottleneck multi-resolution and focus-organ network (VB-MrFo-Net) using a cascade framework for deep learning analysis. The VB-MrFo-Net achieved better performance than VB-Net in tumor segmentation, with a Dice score of 0.87. The nuclear-grade prediction model performed best in the logistic regression classifier, with area under curve values from 0.782 to 0.746. Survival analysis revealed that our prediction model could significantly distinguish patients with high survival risk, with a hazard ratio (HR) of 2.49 [95% confidence interval (CI): 1.13-5.45, P = 0.023] in the General cohort. Excellent performance had also been verified in the Cancer Genome Atlas cohort, the Clinical Proteomic Tumor Analysis Consortium cohort, and the Kidney Tumor Segmentation Challenge cohort, with HRs of 2.77 (95%CI: 1.58-4.84, P = 0.0019), 3.83 (95%CI: 1.22-11.96, P = 0.029), and 2.80 (95%CI: 1.05-7.47, P = 0.025), respectively. In conclusion, we propose a novel VB-MrFo-Net for the renal tumor segmentation and automatic diagnosis of ccRCC. The risk stratification model could accurately distinguish patients with high tumor grade and high survival risk based on non-invasive CT images before surgical treatments, which could provide practical advice for deciding treatment options.
Collapse
Affiliation(s)
- Siteng Chen
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| | - Dandan Song
- Department of Radiology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Lei Chen
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai 201807, China
| | - Tuanjie Guo
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Beibei Jiang
- Department of Radiology, Erasmus University Medical Center, Rotterdam, P.O. Box 2040, 3000 CA, The Netherlands
| | - Aie Liu
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai 201807, China
| | - Xianpan Pan
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai 201807, China
| | - Tao Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Heting Tang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Guihua Chen
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Zhong Xue
- Shanghai United Imaging Intelligence Co., Ltd., Shanghai 201807, China
| | - Xiang Wang
- Department of Urology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200080, China
| | - Ning Zhang
- Department of Urology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Junhua Zheng
- Department of Urology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200001, China
| |
Collapse
|
18
|
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: 22] [Impact Index Per Article: 11.0] [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
|
19
|
Lee M. Recent Advancements in Deep Learning Using Whole Slide Imaging for Cancer Prognosis. Bioengineering (Basel) 2023; 10:897. [PMID: 37627783 PMCID: PMC10451210 DOI: 10.3390/bioengineering10080897] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Revised: 07/21/2023] [Accepted: 07/27/2023] [Indexed: 08/26/2023] Open
Abstract
This review furnishes an exhaustive analysis of the latest advancements in deep learning techniques applied to whole slide images (WSIs) in the context of cancer prognosis, focusing specifically on publications from 2019 through 2023. The swiftly maturing field of deep learning, in combination with the burgeoning availability of WSIs, manifests significant potential in revolutionizing the predictive modeling of cancer prognosis. In light of the swift evolution and profound complexity of the field, it is essential to systematically review contemporary methodologies and critically appraise their ramifications. This review elucidates the prevailing landscape of this intersection, cataloging major developments, evaluating their strengths and weaknesses, and providing discerning insights into prospective directions. In this paper, a comprehensive overview of the field aims to be presented, which can serve as a critical resource for researchers and clinicians, ultimately enhancing the quality of cancer care outcomes. This review's findings accentuate the need for ongoing scrutiny of recent studies in this rapidly progressing field to discern patterns, understand breakthroughs, and navigate future research trajectories.
Collapse
Affiliation(s)
- Minhyeok Lee
- School of Electrical and Electronics Engineering, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
20
|
Distante A, Marandino L, Bertolo R, Ingels A, Pavan N, Pecoraro A, Marchioni M, Carbonara U, Erdem S, Amparore D, Campi R, Roussel E, Caliò A, Wu Z, Palumbo C, Borregales LD, Mulders P, Muselaers CHJ. Artificial Intelligence in Renal Cell Carcinoma Histopathology: Current Applications and Future Perspectives. Diagnostics (Basel) 2023; 13:2294. [PMID: 37443687 DOI: 10.3390/diagnostics13132294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 07/01/2023] [Accepted: 07/04/2023] [Indexed: 07/15/2023] Open
Abstract
Renal cell carcinoma (RCC) is characterized by its diverse histopathological features, which pose possible challenges to accurate diagnosis and prognosis. A comprehensive literature review was conducted to explore recent advancements in the field of artificial intelligence (AI) in RCC pathology. The aim of this paper is to assess whether these advancements hold promise in improving the precision, efficiency, and objectivity of histopathological analysis for RCC, while also reducing costs and interobserver variability and potentially alleviating the labor and time burden experienced by pathologists. The reviewed AI-powered approaches demonstrate effective identification and classification abilities regarding several histopathological features associated with RCC, facilitating accurate diagnosis, grading, and prognosis prediction and enabling precise and reliable assessments. Nevertheless, implementing AI in renal cell carcinoma generates challenges concerning standardization, generalizability, benchmarking performance, and integration of data into clinical workflows. Developing methodologies that enable pathologists to interpret AI decisions accurately is imperative. Moreover, establishing more robust and standardized validation workflows is crucial to instill confidence in AI-powered systems' outcomes. These efforts are vital for advancing current state-of-the-art practices and enhancing patient care in the future.
Collapse
Affiliation(s)
- Alfredo Distante
- Department of Urology, Catholic University of the Sacred Heart, 00168 Roma, Italy
- Department of Urology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Laura Marandino
- Department of Medical Oncology, IRCCS Ospedale San Raffaele, 20132 Milan, Italy
| | - Riccardo Bertolo
- Department of Urology, San Carlo Di Nancy Hospital, 00165 Rome, Italy
| | - Alexandre Ingels
- Department of Urology, University Hospital Henri Mondor, APHP (Assistance Publique-Hôpitaux de Paris), 94000 Créteil, France
| | - Nicola Pavan
- Department of Surgical, Oncological and Oral Sciences, Section of Urology, University of Palermo, 90133 Palermo, Italy
| | - Angela Pecoraro
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10043 Turin, Italy
| | - Michele Marchioni
- Department of Medical, Oral and Biotechnological Sciences, G. d'Annunzio University of Chieti, 66100 Chieti, Italy
| | - Umberto Carbonara
- Andrology and Kidney Transplantation Unit, Department of Emergency and Organ Transplantation-Urology, University of Bari, 70121 Bari, Italy
| | - Selcuk Erdem
- Division of Urologic Oncology, Department of Urology, Istanbul University Istanbul Faculty of Medicine, Istanbul 34093, Turkey
| | - Daniele Amparore
- Department of Urology, San Luigi Gonzaga Hospital, University of Turin, Orbassano, 10043 Turin, Italy
| | - Riccardo Campi
- Urological Robotic Surgery and Renal Transplantation Unit, Careggi Hospital, University of Florence, 50121 Firenze, Italy
| | - Eduard Roussel
- Department of Urology, University Hospitals Leuven, 3000 Leuven, Belgium
| | - Anna Caliò
- Section of Pathology, Department of Diagnostic and Public Health, University of Verona, 37134 Verona, Italy
| | - Zhenjie Wu
- Department of Urology, Changhai Hospital, Naval Medical University, Shanghai 200433, China
| | - Carlotta Palumbo
- Division of Urology, Maggiore della Carità Hospital of Novara, Department of Translational Medicine, University of Eastern Piedmont, 13100 Novara, Italy
| | - Leonardo D Borregales
- Department of Urology, Well Cornell Medicine, New York-Presbyterian Hospital, New York, NY 10032, USA
| | - Peter Mulders
- Department of Urology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| | - Constantijn H J Muselaers
- Department of Urology, Radboud University Medical Center, Geert Grooteplein 10, 6525 GA Nijmegen, The Netherlands
| |
Collapse
|
21
|
Mannas MP, Deng FM, Belanger EC, Jones D, Ren J, Huang W, Orringer DA, Taneja SS. Stimulated Raman histology as a method to determine the adequacy of renal mass biopsy and identify malignant subtypes of renal cell carcinoma. Urol Oncol 2023; 41:328.e9-328.e13. [PMID: 37225634 DOI: 10.1016/j.urolonc.2023.04.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2022] [Revised: 03/01/2023] [Accepted: 04/04/2023] [Indexed: 05/26/2023]
Abstract
INTRODUCTION Renal tumor biopsy requires adequate tissue sampling to aid in the investigation of small renal masses. In some centers the contemporary nondiagnostic renal mass biopsy rate may be as high as 22% and may be as high as 42% in challenging cases. Stimulated Raman Histology (SRH) is a novel microscopic technique which has created the possibility for rapid, label-free, high-resolution images of unprocessed tissue which may be viewed on standard radiology viewing platforms. The application of SRH to renal biopsy may provide the benefits of routine pathologic evaluation during the procedure, thereby reducing nondiagnostic results. We conducted a pilot feasibility study, to assess if renal cell carcinoma (RCC) subtypes may be imaged and to see if high-quality hematoxylin and eosin (H&E) could subsequently be generated. METHODS/MATERIALS An 18-gauge core needle biopsy was taken from a series of 25 ex vivo radical or partial nephrectomy specimens. Histologic images of the fresh, unstained biopsy samples were obtained using a SRH microscope using 2 Raman shifts: 2,845 cm-1 and 2,930 cm-1. The cores were then processed as per routine pathologic protocols. The SRH images and hematoxylin and eosin (H&E) slides were then viewed by a genitourinary pathologist. RESULTS The SRH microscope took 8 to 11 minutes to produce high-quality images of the renal biopsies. Total of 25 renal tumors including 1 oncocytoma, 3 chromophobe RCC, 16 clear cells RCC, 4 papillary RCC, and 1 medullary RCC were included. All renal tumor subtypes were captured, and the SRH images were easily differentiated from adjacent normal renal parenchyma. High quality H&E slides were produced from each of the renal biopsies after SRH was completed. Immunostains were performed on selected cases and the staining was not affected by the SRH image process. CONCLUSION SRH produces high quality images of all renal cell subtypes that can be rapidly produced and easily interpreted to determine renal mass biopsy adequacy, and on occasion, may allow renal tumor subtype identification. Renal biopsies remained available to produce high quality H&E slides and immunostains for confirmation of diagnosis. Procedural application has promise to decrease the known rate of renal mass nondiagnostic biopsies, and application of convolutional neural network methodology may further improve diagnostic capability and increase utilization of renal mass biopsy among urologists.
Collapse
Affiliation(s)
- Miles P Mannas
- Department of Urology, NYU Langone Health, New York, NY; Department of Urologic Sciences, University of British Columbia, Vancouver Prostate Centre, Vancouver, British Columbia, Canada.
| | - Fang-Ming Deng
- Department of Pathology, NYU Langone Health, New York, NY
| | - Eric C Belanger
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Derek Jones
- Department of Pathology, NYU Langone Health, New York, NY
| | - Joyce Ren
- Department of Pathology, NYU Langone Health, New York, NY
| | - William Huang
- Department of Urology, NYU Langone Health, New York, NY
| | - Daniel A Orringer
- Department of Pathology, NYU Langone Health, New York, NY; Department of Neurosurgery, NYU Langone Health, New York, NY
| | - Samir S Taneja
- Department of Urology, NYU Langone Health, New York, NY; Department of Radiology, NYU Langone Health, New York, NY; Department of Biomedical Engineering, NYU Langone Health, New York, NY
| |
Collapse
|
22
|
Zheng Q, Yang R, Xu H, Fan J, Jiao P, Ni X, Yuan J, Wang L, Chen Z, Liu X. A Weakly Supervised Deep Learning Model and Human-Machine Fusion for Accurate Grading of Renal Cell Carcinoma from Histopathology Slides. Cancers (Basel) 2023; 15:3198. [PMID: 37370808 DOI: 10.3390/cancers15123198] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Revised: 05/23/2023] [Accepted: 06/14/2023] [Indexed: 06/29/2023] Open
Abstract
(1) Background: The Fuhrman grading (FG) system is widely used in the management of clear cell renal cell carcinoma (ccRCC). However, it is affected by observer variability and irreproducibility in clinical practice. We aimed to use a deep learning multi-class model called SSL-CLAM to assist in diagnosing the FG status of ccRCC patients using digitized whole slide images (WSIs). (2) Methods: We recruited 504 eligible ccRCC patients from The Cancer Genome Atlas (TCGA) cohort and obtained 708 hematoxylin and eosin-stained WSIs for the development and internal validation of the SSL-CLAM model. Additionally, we obtained 445 WSIs from 188 ccRCC eligible patients in the Clinical Proteomic Tumor Analysis Consortium (CPTAC) cohort as an independent external validation set. A human-machine fusion approach was used to validate the added value of the SSL-CLAM model for pathologists. (3) Results: The SSL-CLAM model successfully diagnosed the five FG statuses (Grade-0, 1, 2, 3, and 4) of ccRCC, and achieved AUCs of 0.917 and 0.887 on the internal and external validation sets, respectively, outperforming a junior pathologist. For the normal/tumor classification (Grade-0, Grade-1/2/3/4) task, the SSL-CLAM model yielded AUCs close to 1 on both the internal and external validation sets. The SSL-CLAM model achieved a better performance for the two-tiered FG (Grade-0, Grade-1/2, and Grade-3/4) task, with AUCs of 0.936 and 0.915 on the internal and external validation sets, respectively. The human-machine diagnostic performance was superior to that of the SSL-CLAM model, showing promising prospects. In addition, the high-attention regions of the SSL-CLAM model showed that with an increasing FG status, the cell nuclei in the tumor region become larger, with irregular contours and increased cellular pleomorphism. (4) Conclusions: Our findings support the feasibility of using deep learning and human-machine fusion methods for FG classification on WSIs from ccRCC patients, which may assist pathologists in making diagnostic decisions.
Collapse
Affiliation(s)
- Qingyuan Zheng
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Rui Yang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Huazhen Xu
- Department of Pharmacology, School of Basic Medical Sciences, Wuhan University, Wuhan 430072, China
| | - Junjie Fan
- University of Chinese Academy of Sciences, Beijing 100049, China
- Trusted Computing and Information Assurance Laboratory, Institute of Software, Chinese Academy of Sciences, Beijing 100190, China
| | - Panpan Jiao
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xinmiao Ni
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jingping Yuan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Lei Wang
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Zhiyuan Chen
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Xiuheng Liu
- Department of Urology, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Institute of Urologic Disease, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
23
|
Yang Y, Pang Q, Hua M, Huangfu Z, Yan R, Liu W, Zhang W, Shi X, Xu Y, Shi J. Excavation of diagnostic biomarkers and construction of prognostic model for clear cell renal cell carcinoma based on urine proteomics. Front Oncol 2023; 13:1170567. [PMID: 37260987 PMCID: PMC10228721 DOI: 10.3389/fonc.2023.1170567] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2023] [Accepted: 04/21/2023] [Indexed: 06/02/2023] Open
Abstract
Purpose Clear cell renal cell carcinoma (ccRCC) is the most common pathology type in kidney cancer. However, the prognosis of advanced ccRCC is unsatisfactory. Thus, early diagnosis becomes one of the most important research priorities of ccRCC. However, currently available studies about ccRCC lack urine-related further studies. In this study, we applied proteomics to search urinary biomarkers to assist early diagnosis of ccRCC. In addition, we constructed a prognostic model to assist judge patients' prognosis. Materials and methods Urine which was used to perform 4D label-free quantitative proteomics was collected from 12 ccRCC patients and 11 non-tumor patients with no urinary system diseases. The urine of 12 patients with ccRCC confirmed by pathological examination after surgery was collected before operatoin. Bioinformatics analysis was used to describe the urinary proteomics landscape of these patients with ccRCC. The top ten proteins with the highest expression content were selected as the basis for subsequent validation. Urine from 46 ccRCC patients and 45 control patients were collected to use for verification by enzyme linked immunosorbent assay (ELISA). In order to assess the prognostic value of urine proteomics, a prognostic model was constructed by COX regression analysis on the intersection of RNA-sequencing data in The Cancer Genome Atlas (TCGA) database and our urine proteomic data. Results 133 proteins differentially expressed in the urinary samples were found and 85 proteins (Fold Change, FC>1.5) were identified up-regulated while 48 down-regulated (FC<0.5). Top 10 proteins including S100A14, PKHD1L1, FABP4, ITIH2, C3, C8G, C2, ATF6, ANGPTL6, F13B were performed ELISA to verify. The results showed that PKHD1L1, ANGPTL6, FABP4 and C3 were statistically significant (P<0.05). We performed multivariate logistic regression analysis and plotted a nomogram. Receiver operating characteristic (ROC) curve indicted that the diagnostic efficiency of combined indicators is satisfactory (Aare under curve, AUC=0.835). Furthermore, the prognostic value of the urine proteomics was explored through the intersection between urine proteomics and TCGA RNA-seq data. Thus, COX regression analysis showed that VSIG4, HLA-DRA, SERPINF1, and IGLV2-23 were statistically significant (P<0.05). Conclusion Our study indicated that the application of urine proteomics to explore diagnostic biomarkers and to construct prognostic models of renal clear cell carcinoma is of certain clinical value. PKHD1L1, ANGPTL6, FABP4 and C3 can assist to diagnose ccRCC. The prognostic model constituted of VSIG4, HLA-DRA, SERPINF1, and IGLV2-23 can significantly predict the prognosis of ccRCC patients, but this still needs more clinical trials to verify.
Collapse
Affiliation(s)
- Yiren Yang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Qingyang Pang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Meimian Hua
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Zhao Huangfu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Rui Yan
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wenqiang Liu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Wei Zhang
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Xiaolei Shi
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Yifan Xu
- Department of Urology, Changhai Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| | - Jiazi Shi
- Department of Urology, Changzheng Hospital, Naval Medical University (Second Military Medical University), Shanghai, China
| |
Collapse
|
24
|
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: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [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
|
25
|
Ohe C, Yoshida T, Amin MB, Uno R, Atsumi N, Yasukochi Y, Ikeda J, Nakamoto T, Noda Y, Kinoshita H, Tsuta K, Higasa K. Deep learning-based predictions of clear and eosinophilic phenotypes in clear cell renal cell carcinoma. Hum Pathol 2023; 131:68-78. [PMID: 36372298 DOI: 10.1016/j.humpath.2022.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Revised: 11/03/2022] [Accepted: 11/07/2022] [Indexed: 11/13/2022]
Abstract
We have recently shown that histological phenotypes focusing on clear and eosinophilic cytoplasm in clear cell renal cell carcinoma (ccRCC) correlated with prognosis and the response to angiogenesis inhibition and checkpoint blockade. This study aims to objectively show the diagnostic utility of clear or eosinophilic phenotypes of ccRCC by developing an artificial intelligence (AI) model using the TCGA-ccRCC dataset and to demonstrate if the clear or eosinophilic predicted phenotypes correlate with pathological factors and gene signatures associated with angiogenesis and cancer immunity. Before the development of the AI model, histological evaluation using hematoxylin and eosin whole-slide images of the TCGA-ccRCC cohort (n = 435) was performed by a urologic pathologist. The AI model was developed as follows. First, the highest-grade area on each whole slide image was captured for image processing. Second, the selected regions were cropped into tiles. Third, the AI model was trained using transfer learning on a deep convolutional neural network, and clear or eosinophilic predictions were scaled as AI scores. Next, we verified the AI model using a validation cohort (n = 95). Finally, we evaluated the accuracy of the prognostic predictions of the AI model and revealed that the AI model detected clear and eosinophilic phenotypes with high accuracy. The AI model stratified the patients' outcomes, and the predicted eosinophilic phenotypes correlated with adverse clinicopathological characteristics and high immune-related gene signatures. In conclusion, the AI-based histologic subclassification accurately predicted clear or eosinophilic phenotypes of ccRCC, allowing for consistently reproducible stratification for prognostic and therapeutic stratification.
Collapse
Affiliation(s)
- Chisato Ohe
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan.
| | - Takashi Yoshida
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Mahul B Amin
- Department of Pathology and Laboratory Medicine, University of Tennessee Health Sciences Center, 930 Madison Avenue, Memphis, TN 38163, USA; Department of Urology, University of Southern California, 1441 Eastlake Avenue, Los Angeles, CA 90033, USA
| | - Rena Uno
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan; Department of Pathology, Hyogo Cancer Center, Akashi, Hyogo 673-8558, Japan
| | - Naho Atsumi
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Yoshiki Yasukochi
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1191, Japan
| | - Junichi Ikeda
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan; Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Takahiro Nakamoto
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan; Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Yuri Noda
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Hidefumi Kinoshita
- Department of Urology and Andrology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Koji Tsuta
- Department of Pathology, Kansai Medical University, 2-3-1 Shin-machi, Hirakata, Osaka 573-1191, Japan
| | - Koichiro Higasa
- Department of Genome Analysis, Institute of Biomedical Science, Kansai Medical University, Hirakata, Osaka 573-1191, Japan
| |
Collapse
|
26
|
Prediction of early-stage melanoma recurrence using clinical and histopathologic features. NPJ Precis Oncol 2022; 6:79. [PMID: 36316482 PMCID: PMC9622809 DOI: 10.1038/s41698-022-00321-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Accepted: 10/13/2022] [Indexed: 11/07/2022] Open
Abstract
Prognostic analysis for early-stage (stage I/II) melanomas is of paramount importance for customized surveillance and treatment plans. Since immune checkpoint inhibitors have recently been approved for stage IIB and IIC melanomas, prognostic tools to identify patients at high risk of recurrence have become even more critical. This study aims to assess the effectiveness of machine-learning algorithms in predicting melanoma recurrence using clinical and histopathologic features from Electronic Health Records (EHRs). We collected 1720 early-stage melanomas: 1172 from the Mass General Brigham healthcare system (MGB) and 548 from the Dana-Farber Cancer Institute (DFCI). We extracted 36 clinicopathologic features and used them to predict the recurrence risk with supervised machine-learning algorithms. Models were evaluated internally and externally: (1) five-fold cross-validation of the MGB cohort; (2) the MGB cohort for training and the DFCI cohort for testing independently. In the internal and external validations, respectively, we achieved a recurrence classification performance of AUC: 0.845 and 0.812, and a time-to-event prediction performance of time-dependent AUC: 0.853 and 0.820. Breslow tumor thickness and mitotic rate were identified as the most predictive features. Our results suggest that machine-learning algorithms can extract predictive signals from clinicopathologic features for early-stage melanoma recurrence prediction, which will enable the identification of patients that may benefit from adjuvant immunotherapy.
Collapse
|
27
|
Wessels F, Kuntz S, Krieghoff-Henning E, Schmitt M, Braun V, Worst TS, Neuberger M, Steeg M, Gaiser T, Fröhling S, Michel MS, Nuhn P, Brinker TJ. Artificial intelligence to predict oncological outcome directly from hematoxylin and eosin-stained slides in urology. Minerva Urol Nephrol 2022; 74:538-550. [PMID: 35274903 DOI: 10.23736/s2724-6051.22.04758-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
INTRODUCTION Artificial intelligence (AI) has been successfully applied for automatic tumor detection and grading in histopathological image analysis in urologic oncology. The aim of this review was to assess the applicability of these approaches in image-based oncological outcome prediction. EVIDENCE ACQUISITION A systematic literature search was conducted using the databases MEDLINE through PubMed and Web of Science up to April 20, 2021. Studies investigating AI approaches to determine the risk of recurrence, metastasis, or survival directly from H&E-stained tissue sections in prostate, renal cell or urothelial carcinoma were included. Characteristics of the AI approach and performance metrics were extracted and summarized. Risk of bias (RoB) was assessed using the PROBAST tool. EVIDENCE SYNTHESIS 16 studies yielding a total of 6658 patients and reporting on 17 outcome predictions were included. Six studies focused on renal cell, six on prostate and three on urothelial carcinoma while one study investigated renal cell and urothelial carcinoma. Handcrafted feature extraction was used in five, a convolutional neural network (CNN) in six and a deep feature extraction in four studies. One study compared a CNN with handcrafted feature extraction. In seven outcome predictions, a multivariable comparison with clinicopathological parameters was reported. Five of them showed statistically significant hazard ratios for the AI's model's-prediction. However, RoB was high in 15 outcome predictions and unclear in two. CONCLUSIONS The included studies are promising but predominantly early pilot studies, therefore primarily highlighting the potential of AI approaches. Additional well-designed studies are needed to assess the actual clinical applicability.
Collapse
Affiliation(s)
- Frederik Wessels
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany.,Department of Urology and Urological Surgery, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Sara Kuntz
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Eva Krieghoff-Henning
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Max Schmitt
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Volker Braun
- Library for the Medical Faculty Mannheim of Heidelberg University, Mannheim, Germany
| | - Thomas S Worst
- Department of Urology and Urological Surgery, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Manuel Neuberger
- Department of Urology and Urological Surgery, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Matthias Steeg
- Institute of Pathology, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Timo Gaiser
- Institute of Pathology, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Stefan Fröhling
- National Center for Tumor Diseases, German Cancer Research Center, Heidelberg, Germany
| | - Maurice-Stephan Michel
- Department of Urology and Urological Surgery, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Philipp Nuhn
- Department of Urology and Urological Surgery, Medical Faculty Mannheim of Heidelberg University, University Medical Center Mannheim, Mannheim, Germany
| | - Titus J Brinker
- Digital Biomarkers for Oncology Group, National Center for Tumor Diseases (NCT), German Cancer Research Center (DKFZ), Heidelberg, Germany -
| |
Collapse
|
28
|
Ghaffari Laleh N, Truhn D, Veldhuizen GP, Han T, van Treeck M, Buelow RD, Langer R, Dislich B, Boor P, Schulz V, Kather JN. Adversarial attacks and adversarial robustness in computational pathology. Nat Commun 2022; 13:5711. [PMID: 36175413 PMCID: PMC9522657 DOI: 10.1038/s41467-022-33266-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2022] [Accepted: 09/09/2022] [Indexed: 11/09/2022] Open
Abstract
Artificial Intelligence (AI) can support diagnostic workflows in oncology by aiding diagnosis and providing biomarkers directly from routine pathology slides. However, AI applications are vulnerable to adversarial attacks. Hence, it is essential to quantify and mitigate this risk before widespread clinical use. Here, we show that convolutional neural networks (CNNs) are highly susceptible to white- and black-box adversarial attacks in clinically relevant weakly-supervised classification tasks. Adversarially robust training and dual batch normalization (DBN) are possible mitigation strategies but require precise knowledge of the type of attack used in the inference. We demonstrate that vision transformers (ViTs) perform equally well compared to CNNs at baseline, but are orders of magnitude more robust to white- and black-box attacks. At a mechanistic level, we show that this is associated with a more robust latent representation of clinically relevant categories in ViTs compared to CNNs. Our results are in line with previous theoretical studies and provide empirical evidence that ViTs are robust learners in computational pathology. This implies that large-scale rollout of AI models in computational pathology should rely on ViTs rather than CNN-based classifiers to provide inherent protection against perturbation of the input data, especially adversarial attacks. Artificial Intelligence can support diagnostic workflows in oncology, but they are vulnerable to adversarial attacks. Here, the authors show that convolutional neural networks are highly susceptible to white- and black-box adversarial attacks in clinically relevant classification tasks.
Collapse
Affiliation(s)
- Narmin Ghaffari Laleh
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital Aachen, Aachen, Germany
| | - Gregory Patrick Veldhuizen
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Tianyu Han
- Department of Physics of Molecular Imaging Systems, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany
| | - Marko van Treeck
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany
| | - Roman D Buelow
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Rupert Langer
- Institute of Pathology, University of Bern, Bern, Switzerland.,Institute of Pathology and Molecular Pathology, Kepler University Hospital, Johannes Kepler University Linz, Linz, Austria
| | - Bastian Dislich
- Institute of Pathology, University of Bern, Bern, Switzerland
| | - Peter Boor
- Institute of Pathology, University Hospital RWTH Aachen, Aachen, Germany
| | - Volkmar Schulz
- Department of Physics of Molecular Imaging Systems, Institute for Experimental Molecular Imaging, RWTH Aachen University, Aachen, Germany.,Physics Institute III B, RWTH Aachen University, Aachen, Germany.,Fraunhofer Institute for Digital Medicine MEVIS, Aachen, Germany.,Hyperion Hybrid Imaging Systems GmbH, Aachen, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, RWTH Aachen university, Aachen, Germany. .,Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany. .,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany. .,Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK. .,Department of Medicine 1, University Hospital and Faculty of Medicine Carl Gustav Carus, Technical University Dresden, Dresden, Germany.
| |
Collapse
|
29
|
Büttner FA, Winter S, Stühler V, Rausch S, Hennenlotter J, Füssel S, Zastrow S, Meinhardt M, Toma M, Jerónimo C, Henrique R, Miranda-Gonçalves V, Kröger N, Ribback S, Hartmann A, Agaimy A, Stöhr C, Polifka I, Fend F, Scharpf M, Comperat E, Wasinger G, Moch H, Stenzl A, Gerlinger M, Bedke J, Schwab M, Schaeffeler E. A novel molecular signature identifies mixed subtypes in renal cell carcinoma with poor prognosis and independent response to immunotherapy. Genome Med 2022; 14:105. [PMID: 36109798 PMCID: PMC9476269 DOI: 10.1186/s13073-022-01105-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/10/2022] [Indexed: 12/30/2022] Open
Abstract
Background Renal cell carcinoma (RCC) is a heterogeneous disease comprising histologically defined subtypes. For therapy selection, precise subtype identification and individualized prognosis are mandatory, but currently limited. Our aim was to refine subtyping and outcome prediction across main subtypes, assuming that a tumor is composed of molecular features present in distinct pathological subtypes. Methods Individual RCC samples were modeled as linear combination of the main subtypes (clear cell (ccRCC), papillary (pRCC), chromophobe (chRCC)) using computational gene expression deconvolution. The new molecular subtyping was compared with histological classification of RCC using the Cancer Genome Atlas (TCGA) cohort (n = 864; ccRCC: 512; pRCC: 287; chRCC: 65) as well as 92 independent histopathologically well-characterized RCC. Predicted continuous subtypes were correlated to cancer-specific survival (CSS) in the TCGA cohort and validated in 242 independent RCC. Association with treatment-related progression-free survival (PFS) was studied in the JAVELIN Renal 101 (n = 726) and IMmotion151 trials (n = 823). CSS and PFS were analyzed using the Kaplan–Meier and Cox regression analysis. Results One hundred seventy-four signature genes enabled reference-free molecular classification of individual RCC. We unambiguously assign tumors to either ccRCC, pRCC, or chRCC and uncover molecularly heterogeneous tumors (e.g., with ccRCC and pRCC features), which are at risk of worse outcome. Assigned proportions of molecular subtype-features significantly correlated with CSS (ccRCC (P = 4.1E − 10), pRCC (P = 6.5E − 10), chRCC (P = 8.6E − 06)) in TCGA. Translation into a numerical RCC-R(isk) score enabled prognosis in TCGA (P = 9.5E − 11). Survival modeling based on the RCC-R score compared to pathological categories was significantly improved (P = 3.6E − 11). The RCC-R score was validated in univariate (P = 3.2E − 05; HR = 3.02, 95% CI: 1.8–5.08) and multivariate analyses including clinicopathological factors (P = 0.018; HR = 2.14, 95% CI: 1.14–4.04). Heterogeneous PD-L1-positive RCC determined by molecular subtyping showed increased PFS with checkpoint inhibition versus sunitinib in the JAVELIN Renal 101 (P = 3.3E − 04; HR = 0.52, 95% CI: 0.36 − 0.75) and IMmotion151 trials (P = 0.047; HR = 0.69, 95% CI: 0.48 − 1). The prediction of PFS significantly benefits from classification into heterogeneous and unambiguous subtypes in both cohorts (P = 0.013 and P = 0.032). Conclusion Switching from categorical to continuous subtype classification across most frequent RCC subtypes enables outcome prediction and fosters personalized treatment strategies.
Supplementary Information The online version contains supplementary material available at 10.1186/s13073-022-01105-y.
Collapse
|
30
|
Artificial Intelligence-Assisted Renal Pathology: Advances and Prospects. J Clin Med 2022; 11:jcm11164918. [PMID: 36013157 PMCID: PMC9410196 DOI: 10.3390/jcm11164918] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 07/30/2022] [Accepted: 08/11/2022] [Indexed: 11/17/2022] Open
Abstract
Digital imaging and advanced microscopy play a pivotal role in the diagnosis of kidney diseases. In recent years, great achievements have been made in digital imaging, providing novel approaches for precise quantitative assessments of nephropathology and relieving burdens of renal pathologists. Developing novel methods of artificial intelligence (AI)-assisted technology through multidisciplinary interaction among computer engineers, renal specialists, and nephropathologists could prove beneficial for renal pathology diagnoses. An increasing number of publications has demonstrated the rapid growth of AI-based technology in nephrology. In this review, we offer an overview of AI-assisted renal pathology, including AI concepts and the workflow of processing digital image data, focusing on the impressive advances of AI application in disease-specific backgrounds. In particular, this review describes the applied computer vision algorithms for the segmentation of kidney structures, diagnosis of specific pathological changes, and prognosis prediction based on images. Lastly, we discuss challenges and prospects to provide an objective view of this topic.
Collapse
|
31
|
Deep learning can predict survival directly from histology in clear cell renal cell carcinoma. PLoS One 2022; 17:e0272656. [PMID: 35976907 PMCID: PMC9385058 DOI: 10.1371/journal.pone.0272656] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2022] [Accepted: 07/24/2022] [Indexed: 11/19/2022] Open
Abstract
For clear cell renal cell carcinoma (ccRCC) risk-dependent diagnostic and therapeutic algorithms are routinely implemented in clinical practice. Artificial intelligence-based image analysis has the potential to improve outcome prediction and thereby risk stratification. Thus, we investigated whether a convolutional neural network (CNN) can extract relevant image features from a representative hematoxylin and eosin-stained slide to predict 5-year overall survival (5y-OS) in ccRCC. The CNN was trained to predict 5y-OS in a binary manner using slides from TCGA and validated using an independent in-house cohort. Multivariable logistic regression was used to combine of the CNNs prediction and clinicopathological parameters. A mean balanced accuracy of 72.0% (standard deviation [SD] = 7.9%), sensitivity of 72.4% (SD = 10.6%), specificity of 71.7% (SD = 11.9%) and area under receiver operating characteristics curve (AUROC) of 0.75 (SD = 0.07) was achieved on the TCGA training set (n = 254 patients / WSIs) using 10-fold cross-validation. On the external validation cohort (n = 99 patients / WSIs), mean accuracy, sensitivity, specificity and AUROC were 65.5% (95%-confidence interval [CI]: 62.9–68.1%), 86.2% (95%-CI: 81.8–90.5%), 44.9% (95%-CI: 40.2–49.6%), and 0.70 (95%-CI: 0.69–0.71). A multivariable model including age, tumor stage and metastasis yielded an AUROC of 0.75 on the TCGA cohort. The inclusion of the CNN-based classification (Odds ratio = 4.86, 95%-CI: 2.70–8.75, p < 0.01) raised the AUROC to 0.81. On the validation cohort, both models showed an AUROC of 0.88. In univariable Cox regression, the CNN showed a hazard ratio of 3.69 (95%-CI: 2.60–5.23, p < 0.01) on TCGA and 2.13 (95%-CI: 0.92–4.94, p = 0.08) on external validation. The results demonstrate that the CNN’s image-based prediction of survival is promising and thus this widely applicable technique should be further investigated with the aim of improving existing risk stratification in ccRCC.
Collapse
|
32
|
Acosta PH, Panwar V, Jarmale V, Christie A, Jasti J, Margulis V, Rakheja D, Cheville J, Leibovich BC, Parker A, Brugarolas J, Kapur P, Rajaram S. Intratumoral Resolution of Driver Gene Mutation Heterogeneity in Renal Cancer Using Deep Learning. Cancer Res 2022; 82:2792-2806. [PMID: 35654752 PMCID: PMC9373732 DOI: 10.1158/0008-5472.can-21-2318] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 11/16/2021] [Accepted: 05/27/2022] [Indexed: 02/05/2023]
Abstract
Intratumoral heterogeneity arising from tumor evolution poses significant challenges biologically and clinically. Dissecting this complexity may benefit from deep learning (DL) algorithms, which can infer molecular features from ubiquitous hematoxylin and eosin (H&E)-stained tissue sections. Although DL algorithms have been developed to predict some driver mutations from H&E images, the ability of these DL algorithms to resolve intratumoral mutation heterogeneity at subclonal spatial resolution is unexplored. Here, we apply DL to a paradigm of intratumoral heterogeneity, clear cell renal cell carcinoma (ccRCC), the most common type of kidney cancer. Matched IHC and H&E images were leveraged to develop DL models for predicting intratumoral genetic heterogeneity of the three most frequently mutated ccRCC genes, BAP1, PBRM1, and SETD2. DL models were generated on a large cohort (N = 1,282) and tested on several independent cohorts, including a TCGA cohort (N = 363 patients) and two tissue microarray (TMA) cohorts (N = 118 and 365 patients). These models were also expanded to a patient-derived xenograft (PDX) TMA, affording analysis of homotopic and heterotopic interactions of tumor and stroma. The status of all three genes could be inferred by DL, with BAP1 showing the highest sensitivity and performance within and across tissue samples (AUC = 0.87-0.89 on holdout). BAP1 results were validated on independent human (AUC = 0.77-0.84) and PDX (AUC = 0.80) cohorts. Finally, BAP1 predictions correlated with clinical outputs such as disease-specific survival. Overall, these data show that DL models can resolve intratumoral heterogeneity in cancer with potential diagnostic, prognostic, and biological implications. SIGNIFICANCE This work demonstrates the potential for deep learning analysis of histopathologic images to serve as a fast, low-cost method to assess genetic intratumoral heterogeneity. See related commentary by Song et al., p. 2672.
Collapse
Affiliation(s)
- Paul H. Acosta
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vandana Panwar
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vipul Jarmale
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Alana Christie
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jay Jasti
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Vitaly Margulis
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA
| | - Dinesh Rakheja
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - John Cheville
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, MN, USA
| | | | - Alexander Parker
- Office of Research Affairs, University of Florida, College of Medicine, Jacksonville, FL, USA
| | - James Brugarolas
- Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Internal Medicine (Hematology-Oncology), University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Payal Kapur
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Urology, University of Texas Southwestern Medical Center at Dallas, Dallas, TX, USA.,Corresponding Authors: 1) Payal Kapur, MD, Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, Ph: 214.590.6592, 2) Satwik Rajaram, PhD, Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX - 75390-9365, Ph: 214-645-1727,
| | - Satwik Rajaram
- Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, Dallas, TX, USA,Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX, USA,Kidney Cancer Program, Simmons Comprehensive Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX, USA,Corresponding Authors: 1) Payal Kapur, MD, Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, Ph: 214.590.6592, 2) Satwik Rajaram, PhD, Lyda Hill Department of Bioinformatics, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX - 75390-9365, Ph: 214-645-1727,
| |
Collapse
|
33
|
Ye X, Liu X, Yin N, Song W, Lu J, Yang Y, Chen X. Successful first-line treatment of simultaneous multiple primary malignancies of lung adenocarcinoma and renal clear cell carcinoma: A case report. Front Immunol 2022; 13:956519. [PMID: 35979370 PMCID: PMC9376962 DOI: 10.3389/fimmu.2022.956519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
Background Multiple Primary Malignancies (MPMs) refer to the occurrence of two or more primary malignancies in the same organ or multiple organs and tissues of the same patient simultaneously or sequentially, with an incidence rate ranging from 2-17%. According to the difference in the time of occurrence of each primary tumor, MPMs can be classified as simultaneous malignancies and heterochronic malignancies. The former refers to the occurrence of two or more malignancies one after another within 6 months, while the latter refers to the occurrence of two malignancies at an interval of more than 6 months. Currently, there is a lack of effective treatment options for MPMs both nationally and internationally. Case presentation The patient was a 65-year-old male smoker with a definite diagnosis of advanced lung adenocarcinoma with kirsten rat sarcoma viral oncogene (KRAS) mutation, concomitant with primary renal clear cell carcinoma (RCCC), who had a progression-free survival (PFS) for 7 months after first-line treatment with albumin-bound paclitaxel and cisplatin in combination with sintilimab. Conclusion In this paper, we report a case of advanced lung adenocarcinoma combined with RCCC as a concurrent double primary malignancy, which achieved a satisfactory outcome after first-line chemotherapy combined with immunotherapy, with the aim of exploring effective treatment modalities for this type of MPMs, in order to improve the survival and prognosis of the patient.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Xiao Chen
- Cancer Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
34
|
Chen J, Li W, Liu B, Xie X. Low LINC02147 expression promotes the malignant progression of oral submucous fibrosis. BMC Oral Health 2022; 22:316. [PMID: 35906577 PMCID: PMC9338683 DOI: 10.1186/s12903-022-02346-4] [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: 04/26/2022] [Accepted: 07/21/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Key lncRNAs associated with the malignant progression of oral submucous fibrosis (OSF) to oral squamous cell carcinoma (OSCC) were identified. METHODS Key lncRNAs with sequential changes from normal oral mucosa (NOM) to OSF to OSCC were identified based on the GEO database. Kaplan-Meier analysis was used to screen lncRNAs related to OSCC prognosis. Cox regression analysis was used to validate the independent prognostic value. qPCR was used to confirm the expression of the candidate lncRNAs. Gene set enrichment analysis (GSEA), nucleocytoplasmic separation assay, fluorescence in situ hybridization, RNA knockdown, western blot, and cell viability assay were performed to investigate the biological functions of the candidate lncRNA. A nomogram was constructed to quantitatively predict OSCC prognosis based on TCGA. RESULTS Bioinformatics methods indicated that LINC02147 was sequentially downregulated from NOM to OSF to OSCC, as confirmed by clinical tissues and cells. Meanwhile, low LINC02147 expression, as an independent prognostic factor, predicted a poor prognosis for OSCC. GSEA and in vitro studies suggested that low LINC02147 expression promoted OSF malignant progression by promoting cell proliferation and differentiation. A LINC02147 signature-based nomogram successfully quantified each indicator's contribution to the overall survival of OSCC. CONCLUSIONS Low LINC02147 expression promoted OSF malignant progression and predicted poor OSCC prognosis.
Collapse
Affiliation(s)
- Jun Chen
- Hunan Key Laboratory of Oral Health Research & Hunan 3D, Printing Engineering Research Center of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China
| | - Wenjie Li
- Hunan Key Laboratory of Oral Health Research & Hunan 3D, Printing Engineering Research Center of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China. .,State Key Laboratory of Powder Metallurgy, Central South University, Changsha, 410083, People's Republic of China. .,Department of Oral Health Science, School of Dentistry, University of Washington, Seattle, WA, 98195, USA.
| | - Binjie Liu
- Hunan Key Laboratory of Oral Health Research & Hunan 3D, Printing Engineering Research Center of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China.
| | - Xiaoli Xie
- Hunan Key Laboratory of Oral Health Research & Hunan 3D, Printing Engineering Research Center of Oral Care and Hunan Clinical Research Center of Oral Major Diseases and Oral Health and Xiangya Stomatological Hospital and Xiangya School of Stomatology, Central South University, 72 Xiangya Road, Kaifu District, Changsha, 410008, People's Republic of China.
| |
Collapse
|
35
|
Chai D, Shi SY, Sobhani N, Ding J, Zhang Z, Jiang N, Wang G, Li M, Li H, Zheng J, Bai J. IFI35 Promotes Renal Cancer Progression by Inhibiting pSTAT1/pSTAT6-Dependent Autophagy. Cancers (Basel) 2022; 14:cancers14122861. [PMID: 35740527 PMCID: PMC9221357 DOI: 10.3390/cancers14122861] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2022] [Revised: 06/06/2022] [Accepted: 06/07/2022] [Indexed: 02/04/2023] Open
Abstract
Interferon-induced protein 35 (IFI35), is currently acknowledged to govern the virus-related immune inflammatory responses. However, the biological significance and function of IFI35 in renal cell cancer (RCC) is still not well understood. Here, IFI35 expression and function were investigated in RCC tissues, renal cancer cells, and animal models. The results showed that IFI35 expression was significantly increased in 200 specimens of RCC patients. We found that higher IFI35 levels were significantly correlated with poor RCC prognosis. In human cell lines, the knockdown of IFI35 suppressed the malignant behavior of renal cancer cells. Similarly, the IFI35 knockdown resulted in significant inhibition of tumor progression in the subcutaneous or lung metastasis mouse model. Furthermore, the knockdown of IFI35 promoted the induction of autophagy by enhancing the autophagy-related gene expression (LC3-II, Beclin-1, and ATG-5). Additionally, blockade of STAT1/STAT6 phosphorylation (pSTAT1/pSTAT6) abrogated the induced autophagy by IFI35 knockdown in renal cancer cells. The autophagy inhibitor 3-MA also abolished the prevention of tumor growth by deleting IFI35 in renal cancer models. The above results suggest that the knockdown of IFI35 suppressed tumor progression of renal cancer by pSTAT1/pSTAT6-dependent autophagy. Our research revealed that IFI35 may serve as a potential diagnosis and therapeutic target for RCC.
Collapse
Affiliation(s)
- Dafei Chai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Shang Yuchen Shi
- Department of Stereotactic Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Xuzhou 221006, China;
| | - Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Jiage Ding
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Zichun Zhang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Nan Jiang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Gang Wang
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Minle Li
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
| | - Hailong Li
- Department of Urology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China;
| | - Junnian Zheng
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Correspondence: (J.Z.); (J.B.)
| | - Jin Bai
- Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China; (D.C.); (J.D.); (Z.Z.); (N.J.); (G.W.); (M.L.)
- Center of Clinical Oncology, Affiliated Hospital of Xuzhou Medical University, Xuzhou 221002, China
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou 221002, China
- Correspondence: (J.Z.); (J.B.)
| |
Collapse
|
36
|
Cifci D, Foersch S, Kather JN. Artificial intelligence to identify genetic alterations in conventional histopathology. J Pathol 2022; 257:430-444. [PMID: 35342954 DOI: 10.1002/path.5898] [Citation(s) in RCA: 69] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2021] [Revised: 03/09/2022] [Accepted: 03/23/2022] [Indexed: 11/10/2022]
Abstract
Precision oncology relies on the identification of targetable molecular alterations in tumor tissues. In many tumor types, a limited set of molecular tests is currently part of standard diagnostic workflows. However, universal testing for all targetable alterations, especially rare ones, is limited by the cost and availability of molecular assays. From 2017 to 2021, multiple studies have shown that artificial intelligence (AI) methods can predict the probability of specific genetic alterations directly from conventional hematoxylin and eosin (H&E) tissue slides. Although these methods are currently less accurate than gold-standard testing (e.g. immunohistochemistry, polymerase chain reaction or next-generation sequencing), they could be used as pre-screening tools to reduce the workload of genetic analyses. In this systematic literature review, we summarize the state of the art in predicting molecular alterations from H&E using AI. We found that AI methods perform reasonably well across multiple tumor types, although few algorithms have been broadly validated. In addition, we found that genetic alterations in FGFR, IDH, PIK3CA, BRAF, TP53 and DNA repair pathways are predictable from H&E in multiple tumor types, while many other genetic alterations have rarely been investigated or were only poorly predictable. Finally, we discuss the next steps for the implementation of AI-based surrogate tests in diagnostic workflows. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Didem Cifci
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
| | - Sebastian Foersch
- Institute of Pathology, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, UK.,Medical Oncology, National Center for Tumor Diseases, University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
37
|
Zhu Z, Lu S, Wang SH, Górriz JM, Zhang YD. BCNet: A Novel Network for Blood Cell Classification. Front Cell Dev Biol 2022; 9:813996. [PMID: 35047515 PMCID: PMC8762289 DOI: 10.3389/fcell.2021.813996] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2021] [Accepted: 12/03/2021] [Indexed: 11/17/2022] Open
Abstract
Aims: Most blood diseases, such as chronic anemia, leukemia (commonly known as blood cancer), and hematopoietic dysfunction, are caused by environmental pollution, substandard decoration materials, radiation exposure, and long-term use certain drugs. Thus, it is imperative to classify the blood cell images. Most cell classification is based on the manual feature, machine learning classifier or the deep convolution network neural model. However, manual feature extraction is a very tedious process, and the results are usually unsatisfactory. On the other hand, the deep convolution neural network is usually composed of massive layers, and each layer has many parameters. Therefore, each deep convolution neural network needs a lot of time to get the results. Another problem is that medical data sets are relatively small, which may lead to overfitting problems. Methods: To address these problems, we propose seven models for the automatic classification of blood cells: BCARENet, BCR5RENet, BCMV2RENet, BCRRNet, BCRENet, BCRSNet, and BCNet. The BCNet model is the best model among the seven proposed models. The backbone model in our method is selected as the ResNet-18, which is pre-trained on the ImageNet set. To improve the performance of the proposed model, we replace the last four layers of the trained transferred ResNet-18 model with the three randomized neural networks (RNNs), which are RVFL, ELM, and SNN. The final outputs of our BCNet are generated by the ensemble of the predictions from the three randomized neural networks by the majority voting. We use four multi-classification indexes for the evaluation of our model. Results: The accuracy, average precision, average F1-score, and average recall are 96.78, 97.07, 96.78, and 96.77%, respectively. Conclusion: We offer the comparison of our model with state-of-the-art methods. The results of the proposed BCNet model are much better than other state-of-the-art methods.
Collapse
Affiliation(s)
- Ziquan Zhu
- School of Computing and Mathematical Sciences, University of Leicester, Leicester, United Kingdom
| | - Siyuan Lu
- School of Computing and Mathematical Sciences, University of Leicester, Leicester, United Kingdom
| | - Shui-Hua Wang
- School of Computing and Mathematical Sciences, University of Leicester, Leicester, United Kingdom
| | - Juan Manuel Górriz
- Department of Signal Theory, Networking and Communications, University of Granada, Granada, Spain
| | - Yu-Dong Zhang
- School of Computing and Mathematical Sciences, University of Leicester, Leicester, United Kingdom
- Guangxi Key Laboratory of Trusted Software, Guilin University of Electronic Technology, Guilin, China
| |
Collapse
|
38
|
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
|
39
|
Gao J, Ye F, Han F, Wang X, Jiang H, Zhang J. A Novel Radiogenomics Biomarker Based on Hypoxic-Gene Subset: Accurate Survival and Prognostic Prediction of Renal Clear Cell Carcinoma. Front Oncol 2021; 11:739815. [PMID: 34692518 PMCID: PMC8529272 DOI: 10.3389/fonc.2021.739815] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/22/2021] [Indexed: 01/21/2023] Open
Abstract
Purpose To construct a novel radiogenomics biomarker based on hypoxic-gene subset for the accurate prognostic prediction of clear cell renal cell carcinoma (ccRCC). Materials and Methods Initially, we screened for the desired hypoxic-gene subset by analysis using the GSEA database. Through univariate and multivariate cox regression hazard ratio analysis, survival-related hypoxia genes were identified, and a genomics signature was constructed in the TCGA database. Building on this, a hypoxia-gene related radiogenomics biomarker (prediction of hypoxia-genes signature by contrast-enhanced CT radiomics) was constructed in the TCIA-KIRC database by extracting features in the venous phase of contrast-enhanced CT images, selecting features using the mRMR and LASSO algorithms, and building logistic regression models. Finally, we validated the prognostic capability of the new biomarker for patients with ccRCC in an independent validation cohort at Huashan Hospital of Fudan University, Shanghai, China. Results The hypoxia-related genomics signature consisting of five genes (IFT57, PABPN1, RNF10, RNF19B and UBE2T) was shown to be significantly associated with survival for patients with ccRCC in the TCGA database, delineated by grouping of the signature expression as either low- or high-risk. In the TCIA database, we constructed a radiogenomics biomarker consisting of 13 radiomics features that were optimal predictors of hypoxia-gene signature expression levels (low- or high-risk) in patients at each institution, that demonstrated AUC values of 0.91 and 0.91 in the training and validation groups, respectively. In the independent validation cohort at Huashan Hospital, our radiogenomics biomarker was significantly associated with prognosis in patients with ccRCC (p=0.0059). Conclusions The novel prognostic radiogenomics biomarker that was constructed achieved excellent correlation with prognosis in both the cohort of TCGA/TCIA-KIRC database and the independent validation cohort of Huashan hospital patients with ccRCC. It is anticipated that this work may assist in clinical preferential treatment decisions and promote the process of precision theranostics in the future.
Collapse
Affiliation(s)
- Jiahao Gao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fangdie Ye
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China
| | - Fang Han
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Xiaoshuang Wang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| | - Haowen Jiang
- Department of Urology, Huashan Hospital, Fudan University, Shanghai, China.,Fudan Institute of Urology, Huashan Hospital, Fudan University, Shanghai, China.,National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, China
| | - Jiawen Zhang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
40
|
Wang F, Yang S, Palmer N, Fox K, Kohane IS, Liao KP, Yu KH, Kou SC. Real-world data analyses unveiled the immune-related adverse effects of immune checkpoint inhibitors across cancer types. NPJ Precis Oncol 2021; 5:82. [PMID: 34508179 PMCID: PMC8433190 DOI: 10.1038/s41698-021-00223-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Accepted: 08/20/2021] [Indexed: 12/17/2022] Open
Abstract
Immune checkpoint inhibitors have demonstrated significant survival benefits in treating many types of cancers. However, their immune-related adverse events (irAEs) have not been systematically evaluated across cancer types in large-scale real-world populations. To address this gap, we conducted real-world data analyses using nationwide insurance claims data with 85.97 million enrollees across 8 years. We identified a significantly increased risk of developing irAEs among patients receiving immunotherapy agents in all seven cancer types commonly treated with immune checkpoint inhibitors. By six months after treatment initialization, those receiving immunotherapy were 1.50-4.00 times (95% CI, lower bound from 1.15 to 2.16, upper bound from 1.69 to 20.36) more likely to develop irAEs in the first 6 months of treatment, compared to matched chemotherapy or targeted therapy groups, with a total of 92,858 patients. The risk of developing irAEs among patients using nivolumab is higher compared to those using pembrolizumab. These results confirmed the need for clinicians to assess irAEs among cancer patients undergoing immunotherapy as part of management. Our methods are extensible to characterizing the effectiveness and adverse effects of novel treatments in large populations in an efficient and economical fashion.
Collapse
Affiliation(s)
- Feicheng Wang
- Department of Statistics, Harvard University, Cambridge, MA, USA
| | - Shihao Yang
- Department of Statistics, Harvard University, Cambridge, MA, USA
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- H. Milton Stewart School of Industrial and Systems Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Nathan Palmer
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Kathe Fox
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Isaac S Kohane
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
| | - Katherine P Liao
- Department of Biomedical Informatics, Harvard Medical School, Boston, MA, USA
- Department of Medicine, Brigham and Women's Hospital, Boston, 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.
| | - S C Kou
- Department of Statistics, Harvard University, Cambridge, MA, USA.
| |
Collapse
|