1
|
Troiano M, Grignaffini F, De Stefanis C, D'Oria V, Bianchi M, Mangini F, Francalanci P, Alaggio R, Frezza F, Alisi A. Comparison between two artificial intelligence models to discriminate cancerous cell nuclei based on confocal fluorescence imaging in hepatocellular carcinoma. Dig Liver Dis 2024:S1590-8658(24)01116-2. [PMID: 39674779 DOI: 10.1016/j.dld.2024.11.026] [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: 08/21/2024] [Revised: 11/06/2024] [Accepted: 11/29/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) exhibits an exceptional intratumoral heterogeneity that might influence diagnosis and outcome. Advances in digital microscopy and artificial intelligence (AI) may improve the HCC identification of liver cancer cells. AIM Two AI algorithms were designed to perform computer-assisted discrimination of tumour from non-tumour nuclei in HCC. METHODS Healthy livers and HCCs from commercially available tissue arrays were stained with an antibody against proliferating cell nuclear antigen and DRAQ5 dye with high affinity for double-stranded DNA, acquired by confocal microscopy imaging and then used to design machine learning (ML) and deep learning (DL) algorithms. RESULTS Nuclei were segmented and then used to develop the Model 1 and Model 2 algorithms, using ML and DL respectively. Model 1 was trained with some texture nuclear features extracted using discrete wavelet transform and grey-level co-occurrence matrix. Model 2 was trained with the segmented images without any additional information. The comparative analysis of the models showed that DL was more effective than ML, achieving an average accuracy of 88 % in discriminating healthy from neoplastic nuclei in HCC samples. CONCLUSION Our research shows that AI techniques and nuclear fluorescent staining could be useful tools for automatically detecting HCC cells in liver tissues.
Collapse
Affiliation(s)
- Maurizio Troiano
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Flavia Grignaffini
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy; Department of Information Engineering, Electronics and Telecommunications (DIET), "La Sapienza" University of Rome, Rome, Italy
| | | | - Valentina D'Oria
- Core Research Facilities, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Marzia Bianchi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabio Mangini
- Department of Information Engineering, Electronics and Telecommunications (DIET), "La Sapienza" University of Rome, Rome, Italy
| | - Paola Francalanci
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Rita Alaggio
- Pathology Unit, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy
| | - Fabrizio Frezza
- Department of Information Engineering, Electronics and Telecommunications (DIET), "La Sapienza" University of Rome, Rome, Italy
| | - Anna Alisi
- Research Unit of Genetics of Complex Phenotypes, Bambino Gesù Children's Hospital, IRCCS, Rome, Italy.
| |
Collapse
|
2
|
Žigutytė L, Sorz-Nechay T, Clusmann J, Kather JN. Use of artificial intelligence for liver diseases: A survey from the EASL congress 2024. JHEP Rep 2024; 6:101209. [PMID: 39583096 PMCID: PMC11585758 DOI: 10.1016/j.jhepr.2024.101209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 08/28/2024] [Accepted: 08/29/2024] [Indexed: 11/26/2024] Open
Abstract
Artificial intelligence (AI) methods enable humans to analyse large amounts of data, which would otherwise not be feasibly quantifiable. This is especially true for unstructured visual and textual data, which can contain invaluable insights into disease. The hepatology research landscape is complex and has generated large amounts of data to be mined. Many open questions can potentially be addressed with existing data through AI methods. However, the field of AI is sometimes obscured by hype cycles and imprecise terminologies. This can conceal the fact that numerous hepatology research groups already use AI methods in their scientific studies. In this review article, we aim to assess the contemporaneous use of AI methods in hepatology in Europe. To achieve this, we systematically surveyed all scientific contributions presented at the EASL Congress 2024. Out of 1,857 accepted abstracts (1,712 posters and 145 oral presentations), 6 presentations (∼4%) and 69 posters (∼4%) utilised AI methods. Of these, 55 posters were included in this review, while the others were excluded due to missing posters or incomplete methodologies. Finally, we summarise current academic trends in the use of AI methods and outline future directions, providing guidance for scientific stakeholders in the field of hepatology.
Collapse
Affiliation(s)
- Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Thomas Sorz-Nechay
- Division of Gastroenterology and Hepatology, Department of Internal Medicine III, Medical University of Vienna, Vienna, Austria
- Center for Molecular Medicine (CeMM) of the Austrian Academy of Sciences, Vienna, Austria
- Christian Doppler Lab for Portal Hypertension and Liver Fibrosis, Medical University of Vienna, Vienna, Austria
| | - Jan Clusmann
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Gastroenterology, University Hospital RWTH Aachen, Aachen, Germany
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Department of Medicine I, Faculty of Medicine and University Hospital Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
3
|
Yang Y, Bo Z, Wang J, Chen B, Su Q, Lian Y, Guo Y, Yang J, Zheng C, Wang J, Zeng H, Zhou J, Chen Y, Chen G, Wang Y. Machine learning based on alcohol drinking-gut microbiota-liver axis in predicting the occurrence of early-stage hepatocellular carcinoma. BMC Cancer 2024; 24:1468. [PMID: 39609660 PMCID: PMC11606210 DOI: 10.1186/s12885-024-13161-1] [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: 11/09/2023] [Accepted: 11/07/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Alcohol drinking and gut microbiota are related to hepatocellular carcinoma (HCC), but the specific relationship between them remains unclear. AIMS We aimed to establish the alcohol drinking-gut microbiota-liver axis and develop machine learning (ML) models in predicting the occurrence of early-stage HCC. METHODS Two hundred sixty-nine patients with early-stage HCC and 278 controls were recruited. Alcohol drinking-gut microbiota-liver axis was established through the mediation/moderation effect analyses. Eight ML algorithms including Classification and Regression Tree (CART), Gradient Boosting Machine (GBM), K-Nearest Neighbor (KNN), Logistic Regression (LR), Neural Network (NN), Random Forest (RF), Support Vector Machine (SVM), and eXtreme Gradient Boosting (XGBoost) were applied. RESULTS A total of 160 pairs of individuals were included for analyses. The mediation effects of Genus_Catenibacterium (P = 0.024), Genus_Tyzzerella_4 (P < 0.001), and Species_Tyzzerella_4 (P = 0.020) were discovered. The moderation effects of Family_Enterococcaceae (OR = 0.741, 95%CI:0.160-0.760, P = 0.017), Family_Leuconostocaceae (OR = 0.793, 95%CI:0.486-3.593, P = 0.010), Genus_Enterococcus (OR = 0.744, 95%CI:0.161-0.753, P = 0.017), Genus_Erysipelatoclostridium (OR = 0.693, 95%CI:0.062-0.672, P = 0.032), Genus_Lactobacillus (OR = 0.655, 95%CI:0.098-0.749, P = 0.011), Species_Enterococcus_faecium (OR = 0.692, 95%CI:0.061-0.673, P = 0.013), and Species_Lactobacillus (OR = 0.653, 95%CI:0.086-0.765, P = 0.014) were uncovered. The predictive power of eight ML models was satisfactory (AUCs:0.855-0.932). The XGBoost model had the best predictive ability (AUC = 0.932). CONCLUSIONS ML models based on the alcohol drinking-gut microbiota-liver axis are valuable in predicting the occurrence of early-stage HCC.
Collapse
Affiliation(s)
- Yi Yang
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Clinical Laboratory, Second Affiliated Hospital, Department of Epidemiology and Biostatistics, School of Public Health, The Key Laboratory of Intelligent Preventive Medicine of Zhejiang Province, Zhejiang University School of Medicine, Hangzhou, China
| | - Zhiyuan Bo
- Department of Surgery, Women's Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jingxian Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Bo Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qing Su
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yiran Lian
- The Second Clinical School of Wenzhou Medical University, Wenzhou, China
| | - Yimo Guo
- Clinical Medicine, Renji College, Wenzhou Medical University, Wenzhou, China
| | - Jinhuan Yang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Chongming Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
| | - Juejin Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Hao Zeng
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Junxi Zhou
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yaqing Chen
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Hepatobiliary Pancreatic Tumor Bioengineering Cross International Joint Laboratory of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China.
- Zhejiang-Germany Interdisciplinary Joint Laboratory of Hepatobiliary-Pancreatic Tumor and Bioengineering, Wenzhou, Zhejiang, China.
| | - Yi Wang
- Department of Epidemiology and Biostatistics, School of Public Health, Wenzhou Medical University, Wenzhou, 325035, China.
| |
Collapse
|
4
|
Lu MY, Chuang WL, Yu ML. The role of artificial intelligence in the management of liver diseases. Kaohsiung J Med Sci 2024; 40:962-971. [PMID: 39440678 DOI: 10.1002/kjm2.12901] [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: 09/11/2024] [Revised: 09/24/2024] [Accepted: 09/24/2024] [Indexed: 10/25/2024] Open
Abstract
Universal neonatal hepatitis B virus (HBV) vaccination and the advent of direct-acting antivirals (DAA) against hepatitis C virus (HCV) have reshaped the epidemiology of chronic liver diseases. However, some aspects of the management of chronic liver diseases remain unresolved. Nucleotide analogs can achieve sustained HBV DNA suppression but rarely lead to a functional cure. Despite the high efficacy of DAAs, successful antiviral therapy does not eliminate the risk of hepatocellular carcinoma (HCC), highlighted the need for cost-effective identification of high-risk populations for HCC surveillance and tailored HCC treatment strategies for these populations. The accessibility of high-throughput genomic data has accelerated the development of precision medicine, and the emergence of artificial intelligence (AI) has led to a new era of precision medicine. AI can learn from complex, non-linear data and identify hidden patterns within real-world datasets. The combination of AI and multi-omics approaches can facilitate disease diagnosis, biomarker discovery, and the prediction of treatment efficacy and prognosis. AI algorithms have been implemented in various aspects, including non-invasive tests, predictive models, image diagnosis, and the interpretation of histopathology findings. AI can support clinicians in decision-making, alleviate clinical burdens, and curtail healthcare expenses. In this review, we introduce the fundamental concepts of machine learning and review the role of AI in the management of chronic liver diseases.
Collapse
Affiliation(s)
- Ming-Ying Lu
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
| | - Wan-Long Chuang
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ming-Lung Yu
- Division of Hepatobiliary, Department of Internal Medicine, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Hepatitis Research Center, College of Medicine and Center for Liquid Biopsy and Cohort Research, Kaohsiung Medical University, Kaohsiung, Taiwan
- School of Medicine and Doctoral Program of Clinical and Experimental Medicine, College of Medicine and Center of Excellence for Metabolic Associated Fatty Liver Disease, National Sun Yat-sen University, Kaohsiung, Taiwan
| |
Collapse
|
5
|
Wang Q, Tang X, Qiao W, Sun L, Shi H, Chen D, Xu B, Liu Y, Zhao J, Huang C, Jin R. Machine learning-based characterization of the gut microbiome associated with the progression of primary biliary cholangitis to cirrhosis. Microbes Infect 2024; 26:105368. [PMID: 38797428 DOI: 10.1016/j.micinf.2024.105368] [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: 11/18/2023] [Revised: 04/20/2024] [Accepted: 05/17/2024] [Indexed: 05/29/2024]
Abstract
BACKGROUND Primary biliary cholangitis (PBC) is associated closely with the gut microbiota. This study aimed to explore the characteristics of the gut microbiota after the progress of PBC to cirrhosis. METHOD This study focuses on utilizing the 16S rRNA gene sequencing method to screen for differences in gut microbiota in PBC patients who progress to cirrhosis. Then, we divided the data into training and verification sets and used seven different machine learning (ML) models to validate them respectively, calculating and comparing the accuracy, F1 score, precision, and recall, and screening the dominant intestinal flora affecting PBC cirrhosis. RESULT PBC cirrhosis patients showed decreased diversity and richness of gut microbiota. Additionally, there are alterations in the composition of gut microbiota in PBC cirrhosis patients. The abundance of Faecalibacterium and Gemmiger bacteria significantly decreases, while the abundance of Veillonella and Streptococcus significantly increases. Furthermore, machine learning methods identify Streptococcus and Gemmiger as the predominant gut microbiota in PBC patients with cirrhosis, serving as non-invasive biomarkers (AUC = 0.902). CONCLUSION Our study revealed that PBC cirrhosis patients gut microbiota composition and function have significantly changed. Streptococcus and Gemmiger may become a non-invasive biomarker for predicting the progression of PBC progress to cirrhosis.
Collapse
Affiliation(s)
- Qi Wang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China
| | - Xiaomeng Tang
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China
| | - Wenying Qiao
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Changping Laboratory, Beijing, PR China
| | - Lina Sun
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Han Shi
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Dexi Chen
- Beijing Institute of Hepatology, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Bin Xu
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Yanmin Liu
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Juan Zhao
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China
| | - Chunyang Huang
- Second Department of Liver Disease Center, Beijing You 'an Hospital, Capital Medical University, Beijing 100069, PR China.
| | - Ronghua Jin
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Beijing Institute of Infectious Diseases, Beijing, PR China; National Center for Infectious Diseases, Beijing Ditan Hospital, Capital Medical University, Beijing, PR China; Changping Laboratory, Beijing, PR China.
| |
Collapse
|
6
|
Wang H, Li J, Ouyang Y, Ren H, An C, Liu W. Multiparametric MRI based deep learning model for prediction of early recurrence of hepatocellular carcinoma after SR following TACE. J Cancer Res Clin Oncol 2024; 150:448. [PMID: 39379692 PMCID: PMC11461583 DOI: 10.1007/s00432-024-05941-w] [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: 06/06/2024] [Accepted: 09/04/2024] [Indexed: 10/10/2024]
Abstract
BACKGROUND Surgical resection (SR) following transarterial chemoembolization (TACE) is a promising treatment for unresectable hepatocellular carcinoma (uHCC). However, biomarkers for the prediction of postoperative recurrence are needed. PURPOSE To develop and validate a model combining deep learning (DL) and clinical data for early recurrence (ER) in uHCC patients after TACE. METHODS A total of 511 patients who received SR following TACE were assigned to derivation (n = 413) and validation (n = 98) cohorts. Deep learning features were taken from the largest tumor area in liver MRI. A nomogram using DL signatures and clinical data was made to forecast early recurrence risk in uHCC patients. Model performance was evaluated using area under the curve (AUC). RESULTS A total of 2278 subsequences and 31,346 slices multiparametric MRI including contrast-enhanced T1WI, T2WI and DWI were input in the DL model simultaneously. Multivariable analysis identified three independent predictors for the development of the nomogram: tumor number (hazard ratio [HR]:3.42, 95% confidence interval [CI]: 2.75-4.31, P = 0.003), microvascular invasion (HR: 9.21, 6.24-32.14; P < 0.001), and DL scores (HR: 17.46, 95% CI: 12.94-23.57, P < 0.001). The AUC of the nomogram was 0.872 and 0.862 in two cohorts, significantly outperforming single-subsequence-based DL mode and clinical model (all, P < 0.001). The nomogram provided two risk strata for cumulative overall survival in two cohorts, showing significant statistical results (P < 0.001). CONCLUSIONS The DL-based nomogram is essential to identify patients with uHCC suitable for treatment with SR following TACE and may potentially benefit personalized decision-making.
Collapse
Affiliation(s)
- Hongyu Wang
- Department of Interventional Therapy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Jinwei Li
- Department of Interventional Therapy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - Yushu Ouyang
- Department of Interventional Therapy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510080, Guangdong, People's Republic of China
| | - He Ren
- Department of Ultrasound, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Chao An
- Department of Minimal Invasive Intervention, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Wendao Liu
- Department of Interventional Therapy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, No. 111 Dade Road, Guangzhou, 510080, Guangdong, People's Republic of China.
| |
Collapse
|
7
|
Laurent-Bellue A, Sadraoui A, Claude L, Calderaro J, Posseme K, Vibert E, Cherqui D, Rosmorduc O, Lewin M, Pesquet JC, Guettier C. Deep Learning Classification and Quantification of Pejorative and Nonpejorative Architectures in Resected Hepatocellular Carcinoma from Digital Histopathologic Images. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1684-1700. [PMID: 38879083 DOI: 10.1016/j.ajpath.2024.05.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 04/17/2024] [Accepted: 05/16/2024] [Indexed: 06/27/2024]
Abstract
Liver resection is one of the best treatments for small hepatocellular carcinoma (HCC), but post-resection recurrence is frequent. Biotherapies have emerged as an efficient adjuvant treatment, making the identification of patients at high risk of recurrence critical. Microvascular invasion (mVI), poor differentiation, pejorative macrotrabecular architectures, and vessels encapsulating tumor clusters architectures are the most accurate histologic predictors of recurrence, but their evaluation is time-consuming and imperfect. Herein, a supervised deep learning-based approach with ResNet34 on 680 whole slide images (WSIs) from 107 liver resection specimens was used to build an algorithm for the identification and quantification of these pejorative architectures. This model achieved an accuracy of 0.864 at patch level and 0.823 at WSI level. To assess its robustness, it was validated on an external cohort of 29 HCCs from another hospital, with an accuracy of 0.787 at WSI level, affirming its generalization capabilities. Moreover, the largest connected areas of the pejorative architectures extracted from the model were positively correlated to the presence of mVI and the number of tumor emboli. These results suggest that the identification of pejorative architectures could be an efficient surrogate of mVI and have a strong predictive value for the risk of recurrence. This study is the first step in the construction of a composite predictive algorithm for early post-resection recurrence of HCC, including artificial intelligence-based features.
Collapse
Affiliation(s)
- Astrid Laurent-Bellue
- Department of Pathology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Aymen Sadraoui
- Centre de Vision Numérique, Paris-Saclay University, Inria, CentraleSupélec, Gif-sur-Yvette, France
| | - Laura Claude
- Department of Pathology, Charles Nicolle Hospital, Rouen, France
| | - Julien Calderaro
- Department of Pathology, Henri-Mondor Hospital, Assistance Publique-Hôpitaux de Paris, Créteil, France
| | - Katia Posseme
- Department of Pathology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France
| | - Eric Vibert
- Centre Hépato-Biliaire, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, France; Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, France; Unité Mixte de Recherche 1193, Paris-Saclay University, INSERM, Villejuif, France
| | - Daniel Cherqui
- Centre Hépato-Biliaire, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, France; Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, France; Unité Mixte de Recherche 1193, Paris-Saclay University, INSERM, Villejuif, France
| | - Olivier Rosmorduc
- Centre Hépato-Biliaire, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, France; Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, France; Unité Mixte de Recherche 1193, Paris-Saclay University, INSERM, Villejuif, France
| | - Maïté Lewin
- Centre Hépato-Biliaire, Paul-Brousse Hospital, Assistance Publique-Hôpitaux de Paris, Villejuif, France; Faculté de Médecine, Paris-Saclay University, Le Kremlin-Bicêtre, France; Unité Mixte de Recherche 1193, Paris-Saclay University, INSERM, Villejuif, France
| | - Jean-Christophe Pesquet
- Centre de Vision Numérique, Paris-Saclay University, Inria, CentraleSupélec, Gif-sur-Yvette, France
| | - Catherine Guettier
- Department of Pathology, Bicêtre Hospital, Assistance Publique-Hôpitaux de Paris, Le Kremlin-Bicêtre, France.
| |
Collapse
|
8
|
Chen M, Chen W, Sun S, Lu Y, Wu G, Xu H, Yang H, Li C, He W, Xu M, Li X, Jiang D, Cai Y, Liu C, Zhang W, He Z. CDK4/6 inhibitor PD-0332991 suppresses hepatocarcinogenesis by inducing senescence of hepatic tumor-initiating cells. J Adv Res 2024:S2090-1232(24)00374-6. [PMID: 39218249 DOI: 10.1016/j.jare.2024.08.034] [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: 04/08/2024] [Revised: 08/08/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024] Open
Abstract
INTRODUCTION Owing to the limited treatment options for hepatocellular carcinoma (HCC), interventions targeting pre-HCC stages have attracted increasing attention. In the pre-HCC stage, hepatic tumor-initiating cells (hTICs) proliferate abnormally and contribute to hepatocarcinogenesis. Numerous studies have investigated targeted senescence induction as an HCC intervention. However, it remains to be clarified whether senescence induction of hTICs could serve as a pre-HCC intervention. OBJECTIVES This study was designed to investigate whether senescence induction of hTICs in the precancerous stage inhibit HCC initiation. METHODS AND RESULTS HCC models developed from chronic liver injury (CLI) were established by using Fah-/- mice and N-Ras + AKT mice. PD-0332991, a selective CDK4/6 inhibitor that blocks the G1/S transition in proliferating cells, was used to induce senescence during the pre-HCC stage. Upon administration of PD-0332991, we observed a significant reduction in HCC incidence following selective senescence induction in hTICs, and an alleviation liver injury in the CLI-HCC models. PD-0332991 also induced senescence in vitro in cultured hTICs isolated from CLI-HCC models. Moreover, RNA sequencing (RNA-seq) analysis delineated that the "Cyclin D-CDK4/6-INK4-Rb" pathway was activated in both mouse and human liver samples during the pre-HCC stage, while PD-0332991 exhibited substantial inhibition of this pathway, thereby inducing cellular senescence in hTICs. Regarding the immune microenvironment, we demonstrated that senescent hTICs secrete key senescence-associated secretory phenotypic (SASP) factors, CXCL10 and CCL2, to activate and recruit macrophages, and contribute to immune surveillance. CONCLUSION We found that hTICs can be targeted and induced into a senescent state during the pre-HCC stage. The SASP factors released by senescent hTICs further activate the immune response, facilitating the clearance of hTICs, and consequently suppressing HCC occurrence. We highlight the importance of pre-HCC interventions and propose that senescence-inducing drugs hold promise for preventing HCC initiation under CLI.
Collapse
Affiliation(s)
- Miaomiao Chen
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Wenjian Chen
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Shiwen Sun
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Yanli Lu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Guoxiu Wu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Hongyu Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Huiru Yang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Chong Li
- Zhoupu Community Health Service Center of Pudong New Area, Shanghai, China
| | - Weizhi He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Mingyang Xu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Xiuhua Li
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Dong Jiang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Yongchao Cai
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Changcheng Liu
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Wencheng Zhang
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China
| | - Zhiying He
- Institute for Regenerative Medicine, Medical Innovation Center and State Key Laboratory of Cardiology, Shanghai East Hospital, School of Life Sciences and Technology, Tongji University, Shanghai 200123, P. R. China; Shanghai Engineering Research Center of Stem Cells Translational Medicine, Shanghai 200335, P. R. China; Shanghai Institute of Stem Cell Research and Clinical Translation, Shanghai 200120, P. R. China.
| |
Collapse
|
9
|
Wu R, Chen Z, Yu J, Lai P, Chen X, Han A, Xu M, Fan Z, Cheng B, Jiang Y, Xia J. A graph-learning based model for automatic diagnosis of Sjögren's syndrome on digital pathological images: a multicentre cohort study. J Transl Med 2024; 22:748. [PMID: 39118142 PMCID: PMC11308146 DOI: 10.1186/s12967-024-05550-8] [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: 02/05/2024] [Accepted: 07/29/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Sjögren's Syndrome (SS) is a rare chronic autoimmune disorder primarily affecting adult females, characterized by chronic inflammation and salivary and lacrimal gland dysfunction. It is often associated with systemic lupus erythematosus, rheumatoid arthritis and kidney disease, which can lead to increased mortality. Early diagnosis is critical, but traditional methods for diagnosing SS, mainly through histopathological evaluation of salivary gland tissue, have limitations. METHODS The study used 100 labial gland biopsy, creating whole-slide images (WSIs) for analysis. The proposed model, named Cell-tissue-graph-based pathological image analysis model (CTG-PAM) and based on graph theory, characterizes single-cell feature, cell-cell feature, and cell-tissue feature. Building upon these features, CTG-PAM achieves cellular-level classification, enabling lymphocyte recognition. Furthermore, it leverages connected component analysis techniques in the cell graph structure to perform SS diagnosis based on lymphocyte counts. FINDINGS CTG-PAM outperforms traditional deep learning methods in diagnosing SS. Its area under the receiver operating characteristic curve (AUC) is 1.0 for the internal validation dataset and 0.8035 for the external test dataset. This indicates high accuracy. The sensitivity of CTG-PAM for the external dataset is 98.21%, while the accuracy is 93.75%. In comparison, the sensitivity and accuracy for traditional deep learning methods (ResNet-50) are lower. The study also shows that CTG-PAM's diagnostic accuracy is closer to skilled pathologists compared to beginners. INTERPRETATION Our findings indicate that CTG-PAM is a reliable method for diagnosing SS. Additionally, CTG-PAM shows promise in enhancing the prognosis of SS patients and holds significant potential for the differential diagnosis of both non-neoplastic and neoplastic diseases. The AI model potentially extends its application to diagnosing immune cells in tumor microenvironments.
Collapse
Affiliation(s)
- Ruifan Wu
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Zhipei Chen
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Jiali Yu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Peng Lai
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China
| | - Xuanyi Chen
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Anjia Han
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Meng Xu
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhaona Fan
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Bin Cheng
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Ying Jiang
- School of Computer Science and Engineering, Sun Yat-sen University, Guangzhou, China.
| | - Juan Xia
- Department of Oral Medicine, Hospital of Stomatology, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
10
|
Calderaro J, Žigutytė L, Truhn D, Jaffe A, Kather JN. Artificial intelligence in liver cancer - new tools for research and patient management. Nat Rev Gastroenterol Hepatol 2024; 21:585-599. [PMID: 38627537 DOI: 10.1038/s41575-024-00919-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/11/2024] [Indexed: 07/31/2024]
Abstract
Liver cancer has high incidence and mortality globally. Artificial intelligence (AI) has advanced rapidly, influencing cancer care. AI systems are already approved for clinical use in some tumour types (for example, colorectal cancer screening). Crucially, research demonstrates that AI can analyse histopathology, radiology and natural language in liver cancer, and can replace manual tasks and access hidden information in routinely available clinical data. However, for liver cancer, few of these applications have translated into large-scale clinical trials or clinically approved products. Here, we advocate for the incorporation of AI in all stages of liver cancer management. We present a taxonomy of AI approaches in liver cancer, highlighting areas with academic and commercial potential, and outline a policy for AI-based liver cancer management, including interdisciplinary training of researchers, clinicians and patients. The potential of AI in liver cancer is immense, but effort is required to ensure that AI can fulfil expectations.
Collapse
Affiliation(s)
- Julien Calderaro
- Département de Pathologie, Assistance Publique Hôpitaux de Paris, Groupe Hospitalier Henri Mondor, Créteil, France
- Institut Mondor de Recherche Biomédicale, MINT-HEP Mondor Integrative Hepatology, Université Paris Est Créteil, Créteil, France
| | - Laura Žigutytė
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany
| | - Daniel Truhn
- Department of Diagnostic and Interventional Radiology, University Hospital RWTH Aachen, Aachen, Germany
| | - Ariel Jaffe
- Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, Section of Digestive Diseases, Yale School of Medicine, New Haven, CT, USA
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health (EKFZ), Medical Faculty Carl Gustav Carus, TUD Dresden University of Technology, Dresden, Germany.
- Department of Medicine I, University Hospital Dresden, Dresden, Germany.
- Medical Oncology, National Center for Tumour Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.
| |
Collapse
|
11
|
Wu L, Li S, Wu C, Wu S, Lin Y, Wei D. Ultrasound-based deep learning radiomics nomogram for differentiating mass mastitis from invasive breast cancer. BMC Med Imaging 2024; 24:189. [PMID: 39060962 PMCID: PMC11282842 DOI: 10.1186/s12880-024-01353-x] [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: 07/28/2023] [Accepted: 07/02/2024] [Indexed: 07/28/2024] Open
Abstract
BACKGROUND The purpose of this study is to develop and validate the potential value of the deep learning radiomics nomogram (DLRN) based on ultrasound to differentiate mass mastitis (MM) and invasive breast cancer (IBC). METHODS 50 cases of MM and 180 cases of IBC with ultrasound Breast Imaging Reporting and Data System 4 category were recruited (training cohort, n = 161, validation cohort, n = 69). Based on PyRadiomics and ResNet50 extractors, radiomics and deep learning features were extracted, respectively. Based on supervised machine learning methods such as logistic regression, random forest, and support vector machine, as well as unsupervised machine learning methods using K-means clustering analysis, the differences in features between MM and IBC were analyzed to develop DLRN. The performance of DLRN had been evaluated by receiver operating characteristic curve, calibration, and clinical practicality. RESULTS Supervised machine learning results showed that compared with radiomics models, especially random forest models, deep learning models were better at recognizing MM and IBC. The area under the curve (AUC) of the validation cohort was 0.84, the accuracy was 0.83, the sensitivity was 0.73, and the specificity was 0.83. Compared to radiomics or deep learning models, DLRN even further improved discrimination ability (AUC of 0.90 and 0.90, accuracy of 0.83 and 0.88 for training and validation cohorts), which had better clinical benefits and good calibratability. In addition, the information heterogeneity of deep learning features in MM and IBC was validated again through unsupervised machine learning clustering analysis, indicating that MM had a unique features phenotype. CONCLUSION The DLRN developed based on radiomics and deep learning features of ultrasound images has potential clinical value in effectively distinguishing between MM and IBC. DLRN breaks through visual limitations and quantifies more image information related to MM based on computers, further utilizing machine learning to effectively utilize this information for clinical decision-making. As DLRN becomes an autonomous screening system, it will improve the recognition rate of MM in grassroots hospitals and reduce the possibility of incorrect treatment and overtreatment.
Collapse
Affiliation(s)
- Linyong Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China
| | - Songhua Li
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China
| | - Chaojun Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China
| | - Shaofeng Wu
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China
| | - Yan Lin
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China
| | - Dayou Wei
- Department of Medical Ultrasound, Maoming People's Hospital, Maoming, 525011, Guangdong, P. R. China.
| |
Collapse
|
12
|
Laohawetwanit T, Apornvirat S, Namboonlue C. Thinking like a pathologist: Morphologic approach to hepatobiliary tumors by ChatGPT. Am J Clin Pathol 2024:aqae087. [PMID: 39030695 DOI: 10.1093/ajcp/aqae087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Accepted: 06/22/2024] [Indexed: 07/21/2024] Open
Abstract
OBJECTIVES This research aimed to evaluate the effectiveness of ChatGPT in accurately diagnosing hepatobiliary tumors using histopathologic images. METHODS The study compared the diagnostic accuracies of the GPT-4 model, providing the same set of images and 2 different input prompts. The first prompt, the morphologic approach, was designed to mimic pathologists' approach to analyzing tissue morphology. In contrast, the second prompt functioned without incorporating this morphologic analysis feature. Diagnostic accuracy and consistency were analyzed. RESULTS A total of 120 photomicrographs, composed of 60 images of each hepatobiliary tumor and nonneoplastic liver tissue, were used. The findings revealed that the morphologic approach significantly enhanced the diagnostic accuracy and consistency of the artificial intelligence (AI). This version was particularly more accurate in identifying hepatocellular carcinoma (mean accuracy: 62.0% vs 27.3%), bile duct adenoma (10.7% vs 3.3%), and cholangiocarcinoma (68.7% vs 16.0%), as well as in distinguishing nonneoplastic liver tissues (77.3% vs 37.5%) (Ps ≤ .01). It also demonstrated higher diagnostic consistency than the other model without a morphologic analysis (κ: 0.46 vs 0.27). CONCLUSIONS This research emphasizes the importance of incorporating pathologists' diagnostic approaches into AI to enhance accuracy and consistency in medical diagnostics. It mainly showcases the AI's histopathologic promise when replicating expert diagnostic processes.
Collapse
Affiliation(s)
- Thiyaphat Laohawetwanit
- Division of Pathology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Division of Pathology, Thammasat University Hospital, Pathum Thani, Thailand
| | - Sompon Apornvirat
- Division of Pathology, Chulabhorn International College of Medicine, Thammasat University, Pathum Thani, Thailand
- Division of Pathology, Thammasat University Hospital, Pathum Thani, Thailand
| | | |
Collapse
|
13
|
Ye Y, Xia L, Yang S, Luo Y, Tang Z, Li Y, Han L, Xie H, Ren Y, Na N. Deep learning-enabled classification of kidney allograft rejection on whole slide histopathologic images. Front Immunol 2024; 15:1438247. [PMID: 39034991 PMCID: PMC11257957 DOI: 10.3389/fimmu.2024.1438247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 06/21/2024] [Indexed: 07/23/2024] Open
Abstract
Background Diagnosis of kidney transplant rejection currently relies on manual histopathological assessment, which is subjective and susceptible to inter-observer variability, leading to limited reproducibility. We aim to develop a deep learning system for automated assessment of whole-slide images (WSIs) from kidney allograft biopsies to enable detection and subtyping of rejection and to predict the prognosis of rejection. Method We collected H&E-stained WSIs of kidney allograft biopsies at 400x magnification from January 2015 to September 2023 at two hospitals. These biopsy specimens were classified as T cell-mediated rejection, antibody-mediated rejection, and other lesions based on the consensus reached by two experienced transplant pathologists. To achieve feature extraction, feature aggregation, and global classification, we employed multi-instance learning and common convolution neural networks (CNNs). The performance of the developed models was evaluated using various metrics, including confusion matrix, receiver operating characteristic curves, the area under the curve (AUC), classification map, heat map, and pathologist-machine confrontations. Results In total, 906 WSIs from 302 kidney allograft biopsies were included for analysis. The model based on multi-instance learning enables detection and subtyping of rejection, named renal rejection artificial intelligence model (RRAIM), with the overall 3-category AUC of 0.798 in the independent test set, which is superior to that of three transplant pathologists under nearly routine assessment conditions. Moreover, the prognosis models accurately predicted graft loss within 1 year following rejection and treatment response for rejection, achieving AUC of 0.936 and 0.756, respectively. Conclusion We first developed deep-learning models utilizing multi-instance learning for the detection and subtyping of rejection and prediction of rejection prognosis in kidney allograft biopsies. These models performed well and may be useful in assisting the pathological diagnosis.
Collapse
Affiliation(s)
- Yongrong Ye
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liubing Xia
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Shicong Yang
- Department of Pathology, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - You Luo
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zuofu Tang
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yuanqing Li
- School of Automation Science and Engineering, South China University of Technology, Guangzhou, China
- Research Center for Brain-Computer Interface, Pazhou Lab, Guangzhou, China
| | - Lanqing Han
- Center for Artificial Intelligence in Medicine, Research Institute of Tsinghua, Pearl River Delta, Guangzhou, China
| | - Hanbin Xie
- Department of Anesthesiology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Yong Ren
- Scientific Research Project Department, Guangdong Artificial Intelligence and Digital Economy Laboratory (Guangzhou), Pazhou Lab, Guangzhou, China
- Shensi lab, Shenzhen Institute for Advanced Study, University of Electronic Science and Technology of China (UESTC), Shenzhen, China
- The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen, China
| | - Ning Na
- Department of Kidney Transplantation, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Jung JO, Pisula JI, Beyerlein X, Lukomski L, Knipper K, Abu Hejleh AP, Fuchs HF, Tolkach Y, Chon SH, Nienhüser H, Büchler MW, Bruns CJ, Quaas A, Bozek K, Popp F, Schmidt T. Deep Learning Histology for Prediction of Lymph Node Metastases and Tumor Regression after Neoadjuvant FLOT Therapy of Gastroesophageal Adenocarcinoma. Cancers (Basel) 2024; 16:2445. [PMID: 39001507 PMCID: PMC11240557 DOI: 10.3390/cancers16132445] [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: 06/04/2024] [Revised: 06/27/2024] [Accepted: 07/02/2024] [Indexed: 07/16/2024] Open
Abstract
BACKGROUND The aim of this study was to establish a deep learning prediction model for neoadjuvant FLOT chemotherapy response. The neural network utilized clinical data and visual information from whole-slide images (WSIs) of therapy-naïve gastroesophageal cancer biopsies. METHODS This study included 78 patients from the University Hospital of Cologne and 59 patients from the University Hospital of Heidelberg used as external validation. RESULTS After surgical resection, 33 patients from Cologne (42.3%) were ypN0 and 45 patients (57.7%) were ypN+, while 23 patients from Heidelberg (39.0%) were ypN0 and 36 patients (61.0%) were ypN+ (p = 0.695). The neural network had an accuracy of 92.1% to predict lymph node metastasis and the area under the curve (AUC) was 0.726. A total of 43 patients from Cologne (55.1%) had less than 50% residual vital tumor (RVT) compared to 34 patients from Heidelberg (57.6%, p = 0.955). The model was able to predict tumor regression with an error of ±14.1% and an AUC of 0.648. CONCLUSIONS This study demonstrates that visual features extracted by deep learning from therapy-naïve biopsies of gastroesophageal adenocarcinomas correlate with positive lymph nodes and tumor regression. The results will be confirmed in prospective studies to achieve early allocation of patients to the most promising treatment.
Collapse
Affiliation(s)
- Jin-On Jung
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Juan I. Pisula
- Data Science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Xenia Beyerlein
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Leandra Lukomski
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Karl Knipper
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Aram P. Abu Hejleh
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Hans F. Fuchs
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Yuri Tolkach
- Institute of Pathology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Seung-Hun Chon
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Henrik Nienhüser
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Markus W. Büchler
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| | - Christiane J. Bruns
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Alexander Quaas
- Institute of Pathology, University Hospital of Cologne, 50937 Cologne, Germany
| | - Katarzyna Bozek
- Data Science of Bioimages Lab, Center for Molecular Medicine Cologne (CMMC), Faculty of Medicine, University Hospital of Cologne, Robert-Koch-Straße 21, 50937 Cologne, Germany
| | - Felix Popp
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
| | - Thomas Schmidt
- Department of General, Visceral, Tumor and Transplantation Surgery, University Hospital of Cologne, Kerpener Straße 62, 50937 Cologne, Germany
- Department of General, Visceral and Transplantation Surgery, University Hospital of Heidelberg, Im Neuenheimer Feld 420, 69120 Heidelberg, Germany
| |
Collapse
|
15
|
He L, Yang Z, Wang Y, Chen W, Diao L, Wang Y, Yuan W, Li X, Zhang Y, He Y, Shen E. A deep learning algorithm to identify carotid plaques and assess their stability. Front Artif Intell 2024; 7:1321884. [PMID: 38952409 PMCID: PMC11215125 DOI: 10.3389/frai.2024.1321884] [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: 10/15/2023] [Accepted: 05/23/2024] [Indexed: 07/03/2024] Open
Abstract
Background Carotid plaques are major risk factors for stroke. Carotid ultrasound can help to assess the risk and incidence rate of stroke. However, large-scale carotid artery screening is time-consuming and laborious, the diagnostic results inevitably involve the subjectivity of the diagnostician to a certain extent. Deep learning demonstrates the ability to solve the aforementioned challenges. Thus, we attempted to develop an automated algorithm to provide a more consistent and objective diagnostic method and to identify the presence and stability of carotid plaques using deep learning. Methods A total of 3,860 ultrasound images from 1,339 participants who underwent carotid plaque assessment between January 2021 and March 2023 at the Shanghai Eighth People's Hospital were divided into a 4:1 ratio for training and internal testing. The external test included 1,564 ultrasound images from 674 participants who underwent carotid plaque assessment between January 2022 and May 2023 at Xinhua Hospital affiliated with Dalian University. Deep learning algorithms, based on the fusion of a bilinear convolutional neural network with a residual neural network (BCNN-ResNet), were used for modeling to detect carotid plaques and assess plaque stability. We chose AUC as the main evaluation index, along with accuracy, sensitivity, and specificity as auxiliary evaluation indices. Results Modeling for detecting carotid plaques involved training and internal testing on 1,291 ultrasound images, with 617 images showing plaques and 674 without plaques. The external test comprised 470 ultrasound images, including 321 images with plaques and 149 without. Modeling for assessing plaque stability involved training and internal testing on 764 ultrasound images, consisting of 494 images with unstable plaques and 270 with stable plaques. The external test was composed of 279 ultrasound images, including 197 images with unstable plaques and 82 with stable plaques. For the task of identifying the presence of carotid plaques, our model achieved an AUC of 0.989 (95% CI: 0.840, 0.998) with a sensitivity of 93.2% and a specificity of 99.21% on the internal test. On the external test, the AUC was 0.951 (95% CI: 0.962, 0.939) with a sensitivity of 95.3% and a specificity of 82.24%. For the task of identifying the stability of carotid plaques, our model achieved an AUC of 0.896 (95% CI: 0.865, 0.922) on the internal test with a sensitivity of 81.63% and a specificity of 87.27%. On the external test, the AUC was 0.854 (95% CI: 0.889, 0.830) with a sensitivity of 68.52% and a specificity of 89.49%. Conclusion Deep learning using BCNN-ResNet algorithms based on routine ultrasound images could be useful for detecting carotid plaques and assessing plaque instability.
Collapse
Affiliation(s)
- Lan He
- Department of Ultrasound Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Department of Ultrasound Medicine, Shanghai Eighth People’s Hospital, Shanghai, China
| | | | | | | | | | - Yitong Wang
- Department of Ultrasound Medicine, Xinhua Hospital, Dalian University, Dalian, China
| | - Wei Yuan
- Department of Ultrasound Medicine, Caohejing Street Community Health Service Centre, Shanghai, China
| | - Xu Li
- Department of Cardiology, The First Hospital of Soochow University, Suzhou, China
| | - Ying Zhang
- Department of Ultrasound Medicine, Xinhua Hospital, Dalian University, Dalian, China
| | - Yongming He
- Department of Cardiology, The First Hospital of Soochow University, Suzhou, China
| | - E. Shen
- Department of Ultrasound Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| |
Collapse
|
16
|
Yan Z, Li X, Li Z, Liu S, Chang H. Prognostic significance of TNFRSF4 expression and development of a pathomics model to predict expression in hepatocellular carcinoma. Heliyon 2024; 10:e31882. [PMID: 38841483 PMCID: PMC11152671 DOI: 10.1016/j.heliyon.2024.e31882] [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: 09/11/2023] [Revised: 05/16/2024] [Accepted: 05/23/2024] [Indexed: 06/07/2024] Open
Abstract
Background TNFRSF4 plays a significant role in cancer progression, especially in hepatocellular carcinoma (HCC). This study aims to investigate the prognostic value of TNFRSF4 expression in patients with HCC and to develop a predictive pathomics model for its expression. Methods A cohort of patients with HCC retrieved from the TCGA database was analyzed using RNA-seq analysis to determine TNFRSF4 expression and its impact on overall survival (OS). Additionally, hematoxylin-eosin staining analysis was performed to construct a pathomics model for predicting TNFRSF4 expression. Then, pathway enrichment analysis was conducted, immune checkpoint markers were investigated, and immune cell infiltration was examined to explore the underlying biological mechanism of the pathomics score. Results TNFRSF4 expression was significantly higher in tumor tissues than in normal tissues. TNFRSF4 expression also exhibited significant correlations with various clinical variables, including pathologic stage III/IV and R1/R2/RX residual tumor. Furthermore, elevated TNFRSF4 expression was associated with unfavorable OS. Interestingly, in the subgroup analysis, elevated TNFRSF4 expression was identified as a significant risk factor for OS in male patients. The newly developed pathomics model successfully predicted TNFRSF4 expression with good performance and revealed a significant association between high pathomics scores and worse OS. In male patients, high pathomics scores were also associated with a higher risk of mortality. Moreover, pathomics scores were also involved in specific hallmarks, immune-related characteristics, and apoptosis-related genes in HCC, such as epithelial-mesenchymal transition, Tregs, and BAX expression. Conclusions Our findings suggest that TNFRSF4 expression and the newly devised pathomics scores hold potential as prognostic markers for OS in patients with HCC. Additionally, gender influenced the association between these markers and patient outcomes.
Collapse
Affiliation(s)
- Zhaoyong Yan
- Department of Interventional Radiology, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Xiang Li
- Department of Immunology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science & Technology, Wuhan, 430000, China
| | - Zeyu Li
- Department of General Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Sinan Liu
- Department of SICU, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, China
| | - Hulin Chang
- Department of Hepatobiliary Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| |
Collapse
|
17
|
Codipilly DC, Faghani S, Hagan C, Lewis J, Erickson BJ, Iyer PG. The Evolving Role of Artificial Intelligence in Gastrointestinal Histopathology: An Update. Clin Gastroenterol Hepatol 2024; 22:1170-1180. [PMID: 38154727 DOI: 10.1016/j.cgh.2023.11.044] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/28/2023] [Revised: 11/20/2023] [Accepted: 11/21/2023] [Indexed: 12/30/2023]
Abstract
Significant advances in artificial intelligence (AI) over the past decade potentially may lead to dramatic effects on clinical practice. Digitized histology represents an area ripe for AI implementation. We describe several current needs within the world of gastrointestinal histopathology, and outline, using currently studied models, how AI potentially can address them. We also highlight pitfalls as AI makes inroads into clinical practice.
Collapse
Affiliation(s)
- D Chamil Codipilly
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, Minnesota
| | - Shahriar Faghani
- Mayo Artificial Intelligence Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Catherine Hagan
- Department of Laboratory Medicine and Pathology, Mayo Clinic, Rochester, Minnesota
| | - Jason Lewis
- Department of Pathology, Mayo Clinic, Jacksonville, Florida
| | - Bradley J Erickson
- Mayo Artificial Intelligence Laboratory, Department of Radiology, Mayo Clinic, Rochester, Minnesota
| | - Prasad G Iyer
- Barrett's Esophagus Unit, Division of Gastroenterology and Hepatology, Mayo Clinic Rochester, Rochester, Minnesota.
| |
Collapse
|
18
|
Liu Y, Chen W, Ruan R, Zhang Z, Wang Z, Guan T, Lin Q, Tang W, Deng J, Wang Z, Li G. Deep learning based digital pathology for predicting treatment response to first-line PD-1 blockade in advanced gastric cancer. J Transl Med 2024; 22:438. [PMID: 38720336 PMCID: PMC11077733 DOI: 10.1186/s12967-024-05262-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 04/29/2024] [Indexed: 05/12/2024] Open
Abstract
BACKGROUND Advanced unresectable gastric cancer (GC) patients were previously treated with chemotherapy alone as the first-line therapy. However, with the Food and Drug Administration's (FDA) 2022 approval of programmed cell death protein 1 (PD-1) inhibitor combined with chemotherapy as the first-li ne treatment for advanced unresectable GC, patients have significantly benefited. However, the significant costs and potential adverse effects necessitate precise patient selection. In recent years, the advent of deep learning (DL) has revolutionized the medical field, particularly in predicting tumor treatment responses. Our study utilizes DL to analyze pathological images, aiming to predict first-line PD-1 combined chemotherapy response for advanced-stage GC. METHODS In this multicenter retrospective analysis, Hematoxylin and Eosin (H&E)-stained slides were collected from advanced GC patients across four medical centers. Treatment response was evaluated according to iRECIST 1.1 criteria after a comprehensive first-line PD-1 immunotherapy combined with chemotherapy. Three DL models were employed in an ensemble approach to create the immune checkpoint inhibitors Response Score (ICIsRS) as a novel histopathological biomarker derived from Whole Slide Images (WSIs). RESULTS Analyzing 148,181 patches from 313 WSIs of 264 advanced GC patients, the ensemble model exhibited superior predictive accuracy, leading to the creation of ICIsNet. The model demonstrated robust performance across four testing datasets, achieving AUC values of 0.92, 0.95, 0.96, and 1 respectively. The boxplot, constructed from the ICIsRS, reveals statistically significant disparities between the well response and poor response (all p-values < = 0.001). CONCLUSION ICIsRS, a DL-derived biomarker from WSIs, effectively predicts advanced GC patients' responses to PD-1 combined chemotherapy, offering a novel approach for personalized treatment planning and allowing for more individualized and potentially effective treatment strategies based on a patient's unique response situations.
Collapse
Affiliation(s)
- Yifan Liu
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Wei Chen
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Ruiwen Ruan
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China
| | - Zhimei Zhang
- Department of Pathology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Zhixiong Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Tianpei Guan
- Department of Gastrointestinal Surgery, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Qi Lin
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Wei Tang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China
| | - Jun Deng
- Department of Oncology, First Affiliated Hospital of Nanchang University, Nanchang, 330006, Jiangxi Province, China.
| | - Zhao Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China.
| | - Guanghua Li
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Sun Yat-sen University, Zhongshan 2nd Street, No. 58, Guangzhou, 510080, 86, Guangdong, China.
| |
Collapse
|
19
|
Lehrich BM, Zhang J, Monga SP, Dhanasekaran R. Battle of the biopsies: Role of tissue and liquid biopsy in hepatocellular carcinoma. J Hepatol 2024; 80:515-530. [PMID: 38104635 PMCID: PMC10923008 DOI: 10.1016/j.jhep.2023.11.030] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2023] [Revised: 10/27/2023] [Accepted: 11/27/2023] [Indexed: 12/19/2023]
Abstract
The diagnosis and management of hepatocellular carcinoma (HCC) have improved significantly in recent years. With the introduction of immunotherapy-based combination therapy, there has been a notable expansion in treatment options for patients with unresectable HCC. Simultaneously, innovative molecular tests for early detection and management of HCC are emerging. This progress prompts a key question: as liquid biopsy techniques rise in prominence, will they replace traditional tissue biopsies, or will both techniques remain relevant? Given the ongoing challenges of early HCC detection, including issues with ultrasound sensitivity, accessibility, and patient adherence to surveillance, the evolution of diagnostic techniques is more relevant than ever. Furthermore, the accurate stratification of HCC is limited by the absence of reliable biomarkers which can predict response to therapies. While the advantages of molecular diagnostics are evident, their potential has not yet been fully harnessed, largely because tissue biopsies are not routinely performed for HCC. Liquid biopsies, analysing components such as circulating tumour cells, DNA, and extracellular vesicles, provide a promising alternative, though they are still associated with challenges related to sensitivity, cost, and accessibility. The early results from multi-analyte liquid biopsy panels are promising and suggest they could play a transformative role in HCC detection and management; however, comprehensive clinical validation is still ongoing. In this review, we explore the challenges and potential of both tissue and liquid biopsy, highlighting that these diagnostic methods, while distinct in their approaches, are set to jointly reshape the future of HCC management.
Collapse
Affiliation(s)
- Brandon M Lehrich
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA
| | - Josephine Zhang
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA
| | - Satdarshan P Monga
- Department of Pathology and Pittsburgh Liver Institute, University of Pittsburgh, School of Medicine and University of Pittsburgh Medical Center, Pittsburgh, Pennsylvania, USA.
| | - Renumathy Dhanasekaran
- Division of Gastroenterology and Hepatology, Department of Medicine, Stanford University, Staford, CA, 94303, USA.
| |
Collapse
|
20
|
Beaufrère A, Ouzir N, Zafar PE, Laurent-Bellue A, Albuquerque M, Lubuela G, Grégory J, Guettier C, Mondet K, Pesquet JC, Paradis V. Primary liver cancer classification from routine tumour biopsy using weakly supervised deep learning. JHEP Rep 2024; 6:101008. [PMID: 38379584 PMCID: PMC10877109 DOI: 10.1016/j.jhepr.2024.101008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 12/09/2023] [Accepted: 12/17/2023] [Indexed: 02/22/2024] Open
Abstract
Background & Aims The diagnosis of primary liver cancers (PLCs) can be challenging, especially on biopsies and for combined hepatocellular-cholangiocarcinoma (cHCC-CCA). We automatically classified PLCs on routine-stained biopsies using a weakly supervised learning method. Method We selected 166 PLC biopsies divided into training, internal and external validation sets: 90, 29 and 47 samples, respectively. Two liver pathologists reviewed each whole-slide hematein eosin saffron (HES)-stained image (WSI). After annotating the tumour/non-tumour areas, tiles of 256x256 pixels were extracted from the WSIs and used to train a ResNet18 neural network. The tumour/non-tumour annotations served as labels during training, and the network's last convolutional layer was used to extract new tumour tile features. Without knowledge of the precise labels of the malignancies, we then applied an unsupervised clustering algorithm. Results Pathological review classified the training and validation sets into hepatocellular carcinoma (HCC, 33/90, 11/29 and 26/47), intrahepatic cholangiocarcinoma (iCCA, 28/90, 9/29 and 15/47), and cHCC-CCA (29/90, 9/29 and 6/47). In the two-cluster model, Clusters 0 and 1 contained mainly HCC and iCCA histological features. The diagnostic agreement between the pathological diagnosis and the two-cluster model predictions (major contingent) in the internal and external validation sets was 100% (11/11) and 96% (25/26) for HCC and 78% (7/9) and 87% (13/15) for iCCA, respectively. For cHCC-CCA, we observed a highly variable proportion of tiles from each cluster (cluster 0: 5-97%; cluster 1: 2-94%). Conclusion Our method applied to PLC HES biopsy could identify specific morphological features of HCC and iCCA. Although no specific features of cHCC-CCA were recognized, assessing the proportion of HCC and iCCA tiles within a slide could facilitate the identification of cHCC-CCA. Impact and implications The diagnosis of primary liver cancers can be challenging, especially on biopsies and for combined hepatocellular-cholangiocarcinoma (cHCC-CCA). We automatically classified primary liver cancers on routine-stained biopsies using a weakly supervised learning method. Our model identified specific features of hepatocellular carcinoma and intrahepatic cholangiocarcinoma. Despite no specific features of cHCC-CCA being recognized, the identification of hepatocellular carcinoma and intrahepatic cholangiocarcinoma tiles within a slide could facilitate the diagnosis of primary liver cancers, and particularly cHCC-CCA.
Collapse
Affiliation(s)
- Aurélie Beaufrère
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Université Paris Cité, Paris, France
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| | - Nora Ouzir
- University of Paris-Saclay, CentraleSupélec, CVN, OPIS Inria, Gif-sur-Yvette 91190, France
| | - Paul Emile Zafar
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- University of Paris-Saclay, CentraleSupélec, CVN, OPIS Inria, Gif-sur-Yvette 91190, France
| | - Astrid Laurent-Bellue
- AP-HP, Department of Pathology, Hôpital Bicêtre, Le Kremlin- Bicêtre, France; UMR-S 1193, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Miguel Albuquerque
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
| | | | - Jules Grégory
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Université Paris Cité, Paris, France
- AP-HP.Nord, Department of Imaging, Beaujon Hospital, Clichy, France
| | - Catherine Guettier
- AP-HP, Department of Pathology, Hôpital Bicêtre, Le Kremlin- Bicêtre, France; UMR-S 1193, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Kévin Mondet
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
| | | | - Valérie Paradis
- AP-HP. Nord, Department of Pathology, FHU MOSAIC, Beaujon Hospital, Clichy, France
- Université Paris Cité, Paris, France
- Centre de Recherche sur l'Inflammation, INSERM UMR 1149, Paris, France
| |
Collapse
|
21
|
Yang X, Yang C, Zhang S, Geng H, Zhu AX, Bernards R, Qin W, Fan J, Wang C, Gao Q. Precision treatment in advanced hepatocellular carcinoma. Cancer Cell 2024; 42:180-197. [PMID: 38350421 DOI: 10.1016/j.ccell.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 12/01/2023] [Accepted: 01/17/2024] [Indexed: 02/15/2024]
Abstract
The past decade has witnessed significant advances in the systemic treatment of advanced hepatocellular carcinoma (HCC). Nevertheless, the newly developed treatment strategies have not achieved universal success and HCC patients frequently exhibit therapeutic resistance to these therapies. Precision treatment represents a paradigm shift in cancer treatment in recent years. This approach utilizes the unique molecular characteristics of individual patient to personalize treatment modalities, aiming to maximize therapeutic efficacy while minimizing side effects. Although precision treatment has shown significant success in multiple cancer types, its application in HCC remains in its infancy. In this review, we discuss key aspects of precision treatment in HCC, including therapeutic biomarkers, molecular classifications, and the heterogeneity of the tumor microenvironment. We also propose future directions, ranging from revolutionizing current treatment methodologies to personalizing therapy through functional assays, which will accelerate the next phase of advancements in this area.
Collapse
Affiliation(s)
- Xupeng Yang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Chen Yang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China; Immune Regulation in Cancer Group, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Shu Zhang
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China
| | - Haigang Geng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Andrew X Zhu
- I-Mab Biopharma, Shanghai, China; Jiahui International Cancer Center, Jiahui Health, Shanghai, China
| | - René Bernards
- Division of Molecular Carcinogenesis, Oncode Institute, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Cun Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Qiang Gao
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China; Key Laboratory of Medical Epigenetics and Metabolism, Institutes of Biomedical Sciences, Fudan University, Shanghai, China.
| |
Collapse
|
22
|
Su L, Liu S, Long Y, Chen C, Chen K, Chen M, Chen Y, Cheng Y, Cui Y, Ding Q, Ding R, Duan M, Gao T, Gu X, He H, He J, Hu B, Hu C, Huang R, Huang X, Jiang H, Jiang J, Lan Y, Li J, Li L, Li L, Li W, Li Y, Lin J, Luo X, Lyu F, Mao Z, Miao H, Shang X, Shang X, Shang Y, Shen Y, Shi Y, Sun Q, Sun W, Tang Z, Wang B, Wang H, Wang H, Wang L, Wang L, Wang S, Wang Z, Wang Z, Wei D, Wu J, Wu Q, Xing X, Yang J, Yang X, Yu J, Yu W, Yu Y, Yuan H, Zhai Q, Zhang H, Zhang L, Zhang M, Zhang Z, Zhao C, Zheng R, Zhong L, Zhou F, Zhu W. Chinese experts' consensus on the application of intensive care big data. Front Med (Lausanne) 2024; 10:1174429. [PMID: 38264049 PMCID: PMC10804886 DOI: 10.3389/fmed.2023.1174429] [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: 02/26/2023] [Accepted: 11/09/2023] [Indexed: 01/25/2024] Open
Abstract
The development of intensive care medicine is inseparable from the diversified monitoring data. Intensive care medicine has been closely integrated with data since its birth. Critical care research requires an integrative approach that embraces the complexity of critical illness and the computational technology and algorithms that can make it possible. Considering the need of standardization of application of big data in intensive care, Intensive Care Medicine Branch of China Health Information and Health Care Big Data Society, Standard Committee has convened expert group, secretary group and the external audit expert group to formulate Chinese Experts' Consensus on the Application of Intensive Care Big Data (2022). This consensus makes 29 recommendations on the following five parts: Concept of intensive care big data, Important scientific issues, Standards and principles of database, Methodology in solving big data problems, Clinical application and safety consideration of intensive care big data. The consensus group believes this consensus is the starting step of application big data in the field of intensive care. More explorations and big data based retrospective research should be carried out in order to enhance safety and reliability of big data based models of critical care field.
Collapse
Affiliation(s)
- Longxiang Su
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Shengjun Liu
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Yun Long
- Department of Critical Care Medicine, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Chaodong Chen
- Department of Surgical Intensive Critical Unit, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Kai Chen
- Department of Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fuzhou, Fujian, China
| | - Ming Chen
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yaolong Chen
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yisong Cheng
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Yating Cui
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Qi Ding
- Department of Surgical Intensive Critical Unit, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Renyu Ding
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Meili Duan
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Tao Gao
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xiaohua Gu
- Department of Critical Care Medicine, Northern Jiangsu People’s Hospital; Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Hongli He
- Intensive Care Unit, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology, Chengdu, China
| | - Jiawei He
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Bo Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Chang Hu
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Rui Huang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xiaobo Huang
- Intensive Care Unit, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology, Chengdu, China
| | - Huizhen Jiang
- Department of Information Center, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| | - Jing Jiang
- Department of Critical Care Medicine, Chongqing General Hospital, Chongqing, China
| | - Yunping Lan
- Intensive Care Unit, Sichuan Academy of Medical Sciences & Sichuan Provincial People’s Hospital, School of Medicine of University of Electronic Science and Technology, Chengdu, China
| | - Jun Li
- Department of Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fuzhou, Fujian, China
| | - Linfeng Li
- Medical Data Research Institute, Chongqing Medical University, Chongqing, China
| | - Lu Li
- Department of Critical Care Medicine, Zhongnan Hospital of Wuhan University, Wuhan, Hubei, China
| | - Wenxiong Li
- Department of Surgical Intensive Critical Unit, Beijing Chao-yang Hospital, Capital Medical University, Beijing, China
| | - Yongzai Li
- Information Network Center, QiLu Hospital, ShanDong University, Jinan, China
| | - Jin Lin
- Department of Critical Care Medicine, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Xufei Luo
- Evidence-based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Feng Lyu
- Department of Computer Science and Engineering, Central South University, Changsha, China
| | - Zhi Mao
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - He Miao
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Xiaopu Shang
- Department of Information Management, Beijing Jiaotong University, Beijing, China
| | - Xiuling Shang
- Department of Critical Care Medicine, Fujian Provincial Key Laboratory of Critical Care Medicine, Shengli Clinical Medical College of Fujian Medical University, Fujian Provincial Hospital, Fujian Provincial Center for Critical Care Medicine, Fuzhou, Fujian, China
| | - You Shang
- Department of Critical Care Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yuwen Shen
- Intensive Care Unit of Cardiovascular Surgery Department, Qilu Hospital of Shandong University, Jinan, China
| | - Yinghuan Shi
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, China
| | - Qihang Sun
- British Chinese Society of Health Informatics, Beijing, China
| | - Weijun Sun
- Faculty of Automation, Guangdong University of Technology, Guangzhou, China
| | - Zhiyun Tang
- Department of Intensive Care Unit, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Emergency and Intensive Care Unit Center, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Bo Wang
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Haijun Wang
- Department of Intensive Care Unit, National Cancer Center/National Clinical Research Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Hongliang Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Li Wang
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences; School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Luhao Wang
- Department of Critical Care Medicine, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, China
| | - Sicong Wang
- Department of Critical Care Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhanwen Wang
- Intensive Care Unit, XiangYa Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiang Ya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiang Ya Hospital, Central South University, Changsha, China
| | - Zhong Wang
- Department of Intensive Care Unit, The First Hospital of China Medical University, Shenyang, Liaoning, China
| | - Dong Wei
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, China
| | - Jianfeng Wu
- Intensive Care Unit, XiangYa Hospital, Central South University, Changsha, China
| | - Qin Wu
- Department of Critical Care Medicine, West China Hospital of Sichuan University, Chengdu, China
| | - Xuezhong Xing
- Department of Epidemiology and Biostatistics, Institute of Basic Medical Sciences Chinese Academy of Medical Sciences; School of Basic Medicine Peking Union Medical College, Beijing, China
| | - Jin Yang
- Department of Critical Care Medicine, Chongqing General Hospital, Chongqing, China
| | - Xianghong Yang
- Department of Intensive Care Unit, National Cancer Center/National Clinical Research Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jiangquan Yu
- Department of Critical Care Medicine, Northern Jiangsu People’s Hospital; Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Wenkui Yu
- Department of Critical Care Medicine, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Yuan Yu
- Intensive Care Unit of Cardiovascular Surgery Department, Qilu Hospital of Shandong University, Jinan, China
| | - Hao Yuan
- Department of Critical Care Medicine, Sun Yat-Sen University First Affiliated Hospital, Guangzhou, China
| | - Qian Zhai
- National Institute of Healthcare Data Science, Nanjing University, Nanjing, China
| | - Hao Zhang
- Department of Intensive Care Unit, National Cancer Center/National Clinical Research Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lina Zhang
- Intensive Care Unit, XiangYa Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiang Ya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiang Ya Hospital, Central South University, Changsha, China
| | - Meng Zhang
- Department of Critical Care Medicine, Chongqing General Hospital, Chongqing, China
| | - Zhongheng Zhang
- Department of Emergency Medicine, Key Laboratory of Precision Medicine in Diagnosis and Monitoring Research of Zhejiang Province, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chunguang Zhao
- Intensive Care Unit, XiangYa Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiang Ya Hospital, Central South University, Changsha, China
- Hunan Provincial Clinical Research Center for Critical Care Medicine, Xiang Ya Hospital, Central South University, Changsha, China
| | - Ruiqiang Zheng
- Department of Critical Care Medicine, Northern Jiangsu People’s Hospital; Clinical Medical College, Yangzhou University, Yangzhou, China
| | - Lei Zhong
- Department of Intensive Care Unit, National Cancer Center/National Clinical Research Center, Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Feihu Zhou
- Department of Critical Care Medicine, The First Medical Center, Chinese PLA General Hospital, Beijing, China
| | - Weiguo Zhu
- Department of General Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
23
|
Jia W, Shi W, Yao Q, Mao Z, Chen C, Fan AQ, Wang Y, Zhao Z, Li J, Song W. Identifying immune infiltration by deep learning to assess the prognosis of patients with hepatocellular carcinoma. J Cancer Res Clin Oncol 2023; 149:12621-12635. [PMID: 37450030 DOI: 10.1007/s00432-023-05097-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 06/30/2023] [Indexed: 07/18/2023]
Abstract
BACKGROUND The treatment situation for hepatocellular carcinoma remains critical. The use of deep learning algorithms to assess immune infiltration is a promising new diagnostic tool. METHODS Patient data and whole slide images (WSIs) were obtained for the Xijing Hospital (XJH) cohort and TCGA cohort. We wrote programs using Visual studio 2022 with C# language to segment the WSI into tiles. Pathologists classified the tiles and later trained deep learning models using the ResNet 101V2 network via ML.NET with the TensorFlow framework. Model performance was evaluated using AccuracyMicro versus AccuracyMacro. Model performance was examined using ROC curves versus PR curves. The percentage of immune infiltration was calculated using the R package survminer to calculate the intergroup cutoff, and the Kaplan‒Meier method was used to plot the overall survival curve of patients. Cox regression was used to determine whether the percentage of immune infiltration was an independent risk factor for prognosis. A nomogram was constructed, and its accuracy was verified using time-dependent ROC curves with calibration curves. The CIBERSORT algorithm was used to assess immune infiltration between groups. Gene Ontology was used to explore the pathways of differentially expressed genes. RESULTS There were 100 WSIs and 165,293 tiles in the training set. The final deep learning models had an AccuracyMicro of 97.46% and an AccuracyMacro of 82.28%. The AUCs of the ROC curves on both the training and validation sets exceeded 0.95. The areas under the classification PR curves exceeded 0.85, except that of the TLS on the validation set, which might have had poor results (0.713) due to too few samples. There was a significant difference in OS between the TIL classification groups (p < 0.001), while there was no significant difference in OS between the TLS groups (p = 0.294). Cox regression showed that TIL percentage was an independent risk factor for prognosis in HCC patients (p = 0.015). The AUCs according to the nomogram were 0.714, 0.690, and 0.676 for the 1-year, 2-year, and 5-year AUCs in the TCGA cohort and 0.756, 0.797, and 0.883 in the XJH cohort, respectively. There were significant differences in the levels of infiltration of seven immune cell types between the two groups of samples, and gene ontology showed that the differentially expressed genes between the groups were immune related. Their expression levels of PD-1 and CTLA4 were also significantly different. CONCLUSION We constructed and tested a deep learning model that evaluates the immune infiltration of liver cancer tissue in HCC patients. Our findings demonstrate the value of the model in assessing patient prognosis, immune infiltration and immune checkpoint expression levels.
Collapse
Affiliation(s)
- Weili Jia
- Xi'an Medical University, Xi'an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Wen Shi
- Xi'an Medical University, Xi'an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | | | - Zhenzhen Mao
- Xi'an Medical University, Xi'an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Chao Chen
- Xi'an Medical University, Xi'an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - AQiang Fan
- Xi'an Medical University, Xi'an, China
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Yanfang Wang
- Xi'an Medical University, Xi'an, China
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Zihao Zhao
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Jipeng Li
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Wenjie Song
- Department of Hepatobiliary Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
24
|
Albrecht T, Rossberg A, Albrecht JD, Nicolay JP, Straub BK, Gerber TS, Albrecht M, Brinkmann F, Charbel A, Schwab C, Schreck J, Brobeil A, Flechtenmacher C, von Winterfeld M, Köhler BC, Springfeld C, Mehrabi A, Singer S, Vogel MN, Neumann O, Stenzinger A, Schirmacher P, Weis CA, Roessler S, Kather JN, Goeppert B. Deep Learning-Enabled Diagnosis of Liver Adenocarcinoma. Gastroenterology 2023; 165:1262-1275. [PMID: 37562657 DOI: 10.1053/j.gastro.2023.07.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2023] [Revised: 07/18/2023] [Accepted: 07/20/2023] [Indexed: 08/12/2023]
Abstract
BACKGROUND & AIMS Diagnosis of adenocarcinoma in the liver is a frequent scenario in routine pathology and has a critical impact on clinical decision making. However, rendering a correct diagnosis can be challenging, and often requires the integration of clinical, radiologic, and immunohistochemical information. We present a deep learning model (HEPNET) to distinguish intrahepatic cholangiocarcinoma from colorectal liver metastasis, as the most frequent primary and secondary forms of liver adenocarcinoma, with clinical grade accuracy using H&E-stained whole-slide images. METHODS HEPNET was trained on 714,589 image tiles from 456 patients who were randomly selected in a stratified manner from a pool of 571 patients who underwent surgical resection or biopsy at Heidelberg University Hospital. Model performance was evaluated on a hold-out internal test set comprising 115 patients and externally validated on 159 patients recruited at Mainz University Hospital. RESULTS On the hold-out internal test set, HEPNET achieved an area under the receiver operating characteristic curve of 0.994 (95% CI, 0.989-1.000) and an accuracy of 96.522% (95% CI, 94.521%-98.694%) at the patient level. Validation on the external test set yielded an area under the receiver operating characteristic curve of 0.997 (95% CI, 0.995-1.000), corresponding to an accuracy of 98.113% (95% CI, 96.907%-100.000%). HEPNET surpassed the performance of 6 pathology experts with different levels of experience in a reader study of 50 patients (P = .0005), boosted the performance of resident pathologists to the level of senior pathologists, and reduced potential downstream analyses. CONCLUSIONS We provided a ready-to-use tool with clinical grade performance that may facilitate routine pathology by rendering a definitive diagnosis and guiding ancillary testing. The incorporation of HEPNET into pathology laboratories may optimize the diagnostic workflow, complemented by test-related labor and cost savings.
Collapse
Affiliation(s)
- Thomas Albrecht
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Liver Cancer Center Heidelberg, Heidelberg, Germany.
| | - Annik Rossberg
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Jan Peter Nicolay
- Department of Dermatology, University Medical Centre Mannheim, Mannheim, Germany
| | - Beate Katharina Straub
- Institute of Pathology, University Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Tiemo Sven Gerber
- Institute of Pathology, University Medicine, Johannes Gutenberg University, Mainz, Germany
| | - Michael Albrecht
- European Center for Angioscience, Medical Faculty of Mannheim, Mannheim, Germany
| | - Fritz Brinkmann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Alphonse Charbel
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Constantin Schwab
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Johannes Schreck
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Alexander Brobeil
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | | | - Bruno Christian Köhler
- Liver Cancer Center Heidelberg, Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Christoph Springfeld
- Liver Cancer Center Heidelberg, Heidelberg, Germany; Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany
| | - Arianeb Mehrabi
- Liver Cancer Center Heidelberg, Heidelberg, Germany; Department of General, Visceral and Transplantation Surgery, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephan Singer
- Institute of Pathology and Neuropathology, Eberhard-Karls University, Tübingen, Germany
| | - Monika Nadja Vogel
- Diagnostic and Interventional Radiology, Thoraxklinik at Heidelberg University Hospital, Heidelberg, Germany
| | - Olaf Neumann
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | | | - Peter Schirmacher
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Liver Cancer Center Heidelberg, Heidelberg, Germany
| | - Cleo-Aron Weis
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Liver Cancer Center Heidelberg, Heidelberg, Germany
| | - Jakob Nikolas Kather
- Department of Medical Oncology, National Center for Tumor Diseases, Heidelberg University Hospital, Heidelberg, Germany; Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Benjamin Goeppert
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany; Institute of Pathology and Neuropathology, RKH Hospital Ludwigsburg, Ludwigsburg, Germany; Institute of Tissue Medicine and Pathology, University of Bern, Bern, Switzerland
| |
Collapse
|
25
|
Burt AD. Augmented liver pathology: artificial intelligence and the assessment of hepatocellular neoplasms. Histopathology 2023; 83:509-511. [PMID: 37698049 DOI: 10.1111/his.15020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 07/17/2023] [Indexed: 09/13/2023]
Affiliation(s)
- Alastair D Burt
- Translational and Clinical Research Institute, Newcastle NIHR Biomedical Research Centre, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
26
|
Li K, Cheng Z, Zeng J, Shu Y, He X, Peng H, Zheng Y. Real-time and accurate estimation of surgical hemoglobin loss using deep learning-based medical sponges image analysis. Sci Rep 2023; 13:15504. [PMID: 37726378 PMCID: PMC10509143 DOI: 10.1038/s41598-023-42572-6] [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: 06/05/2023] [Accepted: 09/12/2023] [Indexed: 09/21/2023] Open
Abstract
Real-time and accurate estimation of surgical hemoglobin (Hb) loss is essential for fluid resuscitation management and evaluation of surgical techniques. In this study, we aimed to explore a novel surgical Hb loss estimation method using deep learning-based medical sponges image analysis. Whole blood samples of pre-measured Hb concentration were collected, and normal saline was added to simulate varying levels of Hb concentration. These blood samples were distributed across blank medical sponges to generate blood-soaked sponges. Eight hundred fifty-one blood-soaked sponges representing a wide range of blood dilutions were randomly divided 7:3 into a training group (n = 595) and a testing group (n = 256). A deep learning model based on the YOLOv5 network was used as the target region extraction and detection, and the three models (Feature extraction technology, ResNet-50, and SE-ResNet50) were trained to predict surgical Hb loss. Mean absolute error (MAE), mean absolute percentage error (MAPE), coefficient (R2) value, and the Bland-Altman analysis were calculated to evaluate the predictive performance in the testing group. The deep learning model based on SE-ResNet50 could predict surgical Hb loss with the best performance (R2 = 0.99, MAE = 11.09 mg, MAPE = 8.6%) compared with other predictive models, and Bland-Altman analysis also showed a bias of 1.343 mg with narrow limits of agreement (- 29.81 to 32.5 mg) between predictive and actual Hb loss. The interactive interface was also designed to display the real-time prediction of surgical Hb loss more intuitively. Thus, it is feasible for real-time estimation of surgical Hb loss using deep learning-based medical sponges image analysis, which was helpful for clinical decisions and technical evaluation.
Collapse
Affiliation(s)
- Kai Li
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Zexin Cheng
- College of Informatics, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Junjie Zeng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Ying Shu
- Department of Laboratory Medicine, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Xiaobo He
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China
| | - Hui Peng
- College of Informatics, Huazhong Agricultural University, Wuhan, Hubei, China.
| | - Yongbin Zheng
- Department of Gastrointestinal Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.
| |
Collapse
|
27
|
Kim GJ, Lee T, Ahn S, Uh Y, Kim SH. Efficient diagnosis of IDH-mutant gliomas: 1p/19qNET assesses 1p/19q codeletion status using weakly-supervised learning. NPJ Precis Oncol 2023; 7:94. [PMID: 37717080 PMCID: PMC10505231 DOI: 10.1038/s41698-023-00450-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2023] [Accepted: 09/05/2023] [Indexed: 09/18/2023] Open
Abstract
Accurate identification of molecular alterations in gliomas is crucial for their diagnosis and treatment. Although, fluorescence in situ hybridization (FISH) allows for the observation of diverse and heterogeneous alterations, it is inherently time-consuming and challenging due to the limitations of the molecular method. Here, we report the development of 1p/19qNET, an advanced deep-learning network designed to predict fold change values of 1p and 19q chromosomes and classify isocitrate dehydrogenase (IDH)-mutant gliomas from whole-slide images. We trained 1p/19qNET on next-generation sequencing data from a discovery set (DS) of 288 patients and utilized a weakly-supervised approach with slide-level labels to reduce bias and workload. We then performed validation on an independent validation set (IVS) comprising 385 samples from The Cancer Genome Atlas, a comprehensive cancer genomics resource. 1p/19qNET outperformed traditional FISH, achieving R2 values of 0.589 and 0.547 for the 1p and 19q arms, respectively. As an IDH-mutant glioma classifier, 1p/19qNET attained AUCs of 0.930 and 0.837 in the DS and IVS, respectively. The weakly-supervised nature of 1p/19qNET provides explainable heatmaps for the results. This study demonstrates the successful use of deep learning for precise determination of 1p/19q codeletion status and classification of IDH-mutant gliomas as astrocytoma or oligodendroglioma. 1p/19qNET offers comparable results to FISH and provides informative spatial information. This approach has broader applications in tumor classification.
Collapse
Affiliation(s)
- Gi Jeong Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Department of Medicine, Yonsei University Graduate School, Seoul, Republic of Korea
| | - Tonghyun Lee
- Department of Artificial Intelligence, Yonsei University College of Computing, Seoul, Republic of Korea
| | - Sangjeong Ahn
- Department of Pathology, Korea University Anam Hospital, Korea University College of Medicine, Seoul, Republic of Korea
| | - Youngjung Uh
- Department of Artificial Intelligence, Yonsei University College of Computing, Seoul, Republic of Korea.
| | - Se Hoon Kim
- Department of Pathology, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
28
|
Hatanaka S, Takahashi M, Nakano M. Discrimination of early HCC using single cell nucleus image and visualization of feature distribution in whole slide images. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2023; 2023:1-4. [PMID: 38082928 DOI: 10.1109/embc40787.2023.10340502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2023]
Abstract
Among hepatocellular carcinoma (HCC), early HCC such as well-differentiated hepatocellular carcinoma is more difficult to distinguish from non-cancer than other cancers. In particular, very well-differentiated hepatocellular carcinoma is even more difficult to distinguish, and it is difficult for pathologists to distinguish between cancer and non-cancer from a single nucleus image. If a function to distinguish cancer with a single cell nucleus image is realized, it may be possible to find new features related to nuclei that are useful for differentiating early HCC. The function will also be very helpful in needle biopsy where the area that can be observed is limited.In this study, we investigated the potential to discriminate cancer/non-cancer from an image of a single hepatocyte nucleus using CNN. The results indicated that discrimination was achievable with a correct rate of around 70%.The probability of cancer/non-cancer was visualized on WSI. The visualization results indicated a difference between cancerous and non-cancerous areas in 71% of the cases, which will help pathologists distinguish region of interest. Grouping sections with similar features also proved useful in improving accuracy and visualization results.
Collapse
|
29
|
Allaume P, Rabilloud N, Turlin B, Bardou-Jacquet E, Loréal O, Calderaro J, Khene ZE, Acosta O, De Crevoisier R, Rioux-Leclercq N, Pecot T, Kammerer-Jacquet SF. Artificial Intelligence-Based Opportunities in Liver Pathology-A Systematic Review. Diagnostics (Basel) 2023; 13:diagnostics13101799. [PMID: 37238283 DOI: 10.3390/diagnostics13101799] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 05/04/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
BACKGROUND Artificial Intelligence (AI)-based Deep Neural Networks (DNNs) can handle a wide range of applications in image analysis, ranging from automated segmentation to diagnostic and prediction. As such, they have revolutionized healthcare, including in the liver pathology field. OBJECTIVE The present study aims to provide a systematic review of applications and performances provided by DNN algorithms in liver pathology throughout the Pubmed and Embase databases up to December 2022, for tumoral, metabolic and inflammatory fields. RESULTS 42 articles were selected and fully reviewed. Each article was evaluated through the Quality Assessment of Diagnostic Accuracy Studies (QUADAS-2) tool, highlighting their risks of bias. CONCLUSIONS DNN-based models are well represented in the field of liver pathology, and their applications are diverse. Most studies, however, presented at least one domain with a high risk of bias according to the QUADAS-2 tool. Hence, DNN models in liver pathology present future opportunities and persistent limitations. To our knowledge, this review is the first one solely focused on DNN-based applications in liver pathology, and to evaluate their bias through the lens of the QUADAS2 tool.
Collapse
Affiliation(s)
- Pierre Allaume
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
| | - Noémie Rabilloud
- Impact TEAM, Laboratoire Traitement du Signal et de l'Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
| | - Bruno Turlin
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
- Research Unit n°UMR1341 NuMeCan-Nutrition, Métabolismes et Cancer, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
| | - Edouard Bardou-Jacquet
- Research Unit n°UMR1341 NuMeCan-Nutrition, Métabolismes et Cancer, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
- Department of Liver Diseases CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
| | - Olivier Loréal
- Research Unit n°UMR1341 NuMeCan-Nutrition, Métabolismes et Cancer, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
| | - Julien Calderaro
- Assistance Publique-Hôpitaux de Paris, Department of Pathology Henri Mondor, 94000 Créteil, France
- INSERM U955, Team Pathophysiology and Therapy of Chronic Viral Hepatitis and Related Cancers, 94000 Créteil, France
| | - Zine-Eddine Khene
- Impact TEAM, Laboratoire Traitement du Signal et de l'Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
- Department of Urology, CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
| | - Oscar Acosta
- Impact TEAM, Laboratoire Traitement du Signal et de l'Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
| | - Renaud De Crevoisier
- Impact TEAM, Laboratoire Traitement du Signal et de l'Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
- Department of Radiotherapy, Centre Eugène Marquis, 35033 Rennes, France
| | - Nathalie Rioux-Leclercq
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
| | - Thierry Pecot
- Biosit Platform UAR 3480 CNRS US18 INSERM U955, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
| | - Solène-Florence Kammerer-Jacquet
- Department of Pathology CHU de Rennes, Rennes 1 University, Pontchaillou Hospital, 2 rue Henri Le Guilloux, CEDEX 09, 35033 Rennes, France
- Impact TEAM, Laboratoire Traitement du Signal et de l'Image (LTSI) INSERM, Rennes 1 University, Pontchaillou Hospital, 35033 Rennes, France
| |
Collapse
|
30
|
Lan J, Chen M, Wang J, Du M, Wu Z, Zhang H, Xue Y, Wang T, Chen L, Xu C, Han Z, Hu Z, Zhou Y, Zhou X, Tong T, Chen G. Using less annotation workload to establish a pathological auxiliary diagnosis system for gastric cancer. Cell Rep Med 2023; 4:101004. [PMID: 37044091 PMCID: PMC10140598 DOI: 10.1016/j.xcrm.2023.101004] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 10/20/2022] [Accepted: 03/17/2023] [Indexed: 04/14/2023]
Abstract
Pathological diagnosis of gastric cancer requires pathologists to have extensive clinical experience. To help pathologists improve diagnostic accuracy and efficiency, we collected 1,514 cases of stomach H&E-stained specimens with complete diagnostic information to establish a pathological auxiliary diagnosis system based on deep learning. At the slide level, our system achieves a specificity of 0.8878 while maintaining a high sensitivity close to 1.0 on 269 biopsy specimens (147 malignancies) and 163 surgical specimens (80 malignancies). The classified accuracy of our system is 0.9034 at the slide level for 352 biopsy specimens (201 malignancies) from 50 medical centers. With the help of our system, the pathologists' average false-negative rate and average false-positive rate on 100 biopsy specimens (50 malignancies) are reduced to 1/5 and 1/2 of the original rates, respectively. At the same time, the average uncertainty rate and the average diagnosis time are reduced by approximately 22% and 20%, respectively.
Collapse
Affiliation(s)
- Junlin Lan
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Musheng Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Jianchao Wang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Min Du
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zhida Wu
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Hejun Zhang
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Yuyang Xue
- School of Engineering, University of Edinburgh, Edinburgh EH8 9JU, UK
| | - Tao Wang
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Lifan Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China
| | - Chaohui Xu
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Zixin Han
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Ziwei Hu
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Yuanbo Zhou
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Xiaogen Zhou
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China
| | - Tong Tong
- College of Physics and Information Engineering, Fuzhou University, Fuzhou, Fujian 350108, China; Key Lab of Medical Instrumentation & Pharmaceutical Technology of Fujian Province, Fuzhou University, Fuzhou, Fujian 350108, China; Imperial Vision Technology, Fuzhou, Fujian 350100, China.
| | - Gang Chen
- Department of Pathology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian 350014, China; Fujian Key Laboratory of Translational Cancer Medicine, Fuzhou, Fujian 350014, China.
| |
Collapse
|
31
|
Luo C, Yang J, Liu Z, Jing D. Predicting the recurrence and overall survival of patients with glioma based on histopathological images using deep learning. Front Neurol 2023; 14:1100933. [PMID: 37064206 PMCID: PMC10102594 DOI: 10.3389/fneur.2023.1100933] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Accepted: 03/13/2023] [Indexed: 04/03/2023] Open
Abstract
BackgroundA deep learning (DL) model based on representative biopsy tissues can predict the recurrence and overall survival of patients with glioma, leading to optimized personalized medicine. This research aimed to develop a DL model based on hematoxylin-eosin (HE) stained pathological images and verify its diagnostic accuracy.MethodsOur study retrospectively collected 162 patients with glioma and randomly divided them into a training set (n = 113) and a validation set (n = 49) to build a DL model. The HE-stained slide was segmented into a size of 180 × 180 pixels without overlapping. The patch-level features were extracted by the pre-trained ResNet50 to predict the recurrence and overall survival. Additionally, a light-strategy was introduced where low-size digital biopsy images with clinical information were inputted into the DL model to ensure minimum memory occupation.ResultsOur study extracted 512 histopathological features from the HE-stained slides of each glioma patient. We identified 36 and 18 features as significantly related to disease-free survival (DFS) and overall survival (OS), respectively, (P < 0.05) using the univariate Cox proportional-hazards model. Pathomics signature showed a C-index of 0.630 and 0.652 for DFS and OS prediction, respectively. The time-dependent receiver operating characteristic (ROC) curves, along with nomograms, were used to assess the diagnostic accuracy at a fixed time point. In the validation set (n = 49), the area under the curve (AUC) in the 1- and 2-year DFS was 0.955 and 0.904, respectively, and the 2-, 3-, and 5-year OS were 0.969, 0.955, and 0.960, respectively. We stratified the patients into low- and high-risk groups using the median hazard score (0.083 for DFS and−0.177 for OS) and showed significant differences between these groups (P < 0.001).ConclusionOur results demonstrated that the DL model based on the HE-stained slides showed the predictability of recurrence and survival in patients with glioma. The results can be used to assist oncologists in selecting the optimal treatment strategy in clinical practice.
Collapse
Affiliation(s)
- Chenhua Luo
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Jiyan Yang
- Department of Oncology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Zhengzheng Liu
- Xiangya School of Medicine, Central South University, Changsha, China
| | - Di Jing
- Xiangya School of Medicine, Central South University, Changsha, China
- *Correspondence: Di Jing
| |
Collapse
|
32
|
Pan J, Hong G, Zeng H, Liao C, Li H, Yao Y, Gan Q, Wang Y, Wu S, Lin T. An artificial intelligence model for the pathological diagnosis of invasion depth and histologic grade in bladder cancer. J Transl Med 2023; 21:42. [PMID: 36691055 PMCID: PMC9869632 DOI: 10.1186/s12967-023-03888-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Accepted: 01/12/2023] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Accurate pathological diagnosis of invasion depth and histologic grade is key for clinical management in patients with bladder cancer (BCa), but it is labour-intensive, experience-dependent and subject to interobserver variability. Here, we aimed to develop a pathological artificial intelligence diagnostic model (PAIDM) for BCa diagnosis. METHODS A total of 854 whole slide images (WSIs) from 692 patients were included and divided into training and validation sets. The PAIDM was developed using the training set based on the deep learning algorithm ScanNet, and the performance was verified at the patch level in validation set 1 and at the WSI level in validation set 2. An independent validation cohort (validation set 3) was employed to compare the PAIDM and pathologists. Model performance was evaluated using the area under the curve (AUC), accuracy, sensitivity, specificity, positive predictive value and negative predictive value. RESULTS The AUCs of the PAIDM were 0.878 (95% CI 0.875-0.881) at the patch level in validation set 1 and 0.870 (95% CI 0.805-0.923) at the WSI level in validation set 2. In comparing the PAIDM and pathologists, the PAIDM achieved an AUC of 0.847 (95% CI 0.779-0.905), which was non-inferior to the average diagnostic level of pathologists. There was high consistency between the model-predicted and manually annotated areas, improving the PAIDM's interpretability. CONCLUSIONS We reported an artificial intelligence-based diagnostic model for BCa that performed well in identifying invasion depth and histologic grade. Importantly, the PAIDM performed admirably in patch-level recognition, with a promising application for transurethral resection specimens.
Collapse
Affiliation(s)
- Jiexin Pan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Guibin Hong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hong Zeng
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Chengxiao Liao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Huarun Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Yuhui Yao
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Qinghua Gan
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Yun Wang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China
| | - Shaoxu Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, China.
| | - Tianxin Lin
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, 107th Yanjiangxi Road, Guangzhou, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China.
- Guangdong Provincial Clinical Research Center for Urological Diseases, Guangzhou, Guangdong, China.
| |
Collapse
|
33
|
Stulpinas R, Zilenaite-Petrulaitiene D, Rasmusson A, Gulla A, Grigonyte A, Strupas K, Laurinavicius A. Prognostic Value of CD8+ Lymphocytes in Hepatocellular Carcinoma and Perineoplastic Parenchyma Assessed by Interface Density Profiles in Liver Resection Samples. Cancers (Basel) 2023; 15:cancers15020366. [PMID: 36672317 PMCID: PMC9857181 DOI: 10.3390/cancers15020366] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/28/2022] [Accepted: 01/04/2023] [Indexed: 01/08/2023] Open
Abstract
Hepatocellular carcinoma (HCC) often emerges in the setting of long-standing inflammatory liver disease. CD8 lymphocytes are involved in both the antitumoral response and hepatocyte damage in the remaining parenchyma. We investigated the dual role of CD8 lymphocytes by assessing density profiles at the interfaces of both HCC and perineoplastic liver parenchyma with surrounding stroma in whole-slide immunohistochemistry images of surgical resection samples. We applied a hexagonal grid-based digital image analysis method to sample the interface zones and compute the CD8 density profiles within them. The prognostic value of the indicators was explored in the context of clinicopathological, peripheral blood testing, and surgery data. Independent predictors of worse OS were a low standard deviation of CD8+ density along the tumor edge, high mean CD8+ density within the epithelial aspect of the perineoplastic liver-stroma interface, longer duration of surgery, a higher level of aspartate transaminase (AST), and a higher basophil count in the peripheral blood. A combined score, derived from these five independent predictors, enabled risk stratification of the patients into three prognostic categories with a 5-year OS probability of 76%, 40%, and 8%. Independent predictors of longer RFS were stage pT1, shorter duration of surgery, larger tumor size, wider tumor-free margin, and higher mean CD8+ density in the epithelial aspect of the tumor-stroma interface. We conclude that (1) our computational models reveal independent and opposite prognostic impacts of CD8+ cell densities at the interfaces of the malignant and non-malignant epithelium interfaces with the surrounding stroma; and (2) together with pathology, surgery, and laboratory data, comprehensive prognostic models can be constructed to predict patient outcomes after liver resection due to HCC.
Collapse
Affiliation(s)
- Rokas Stulpinas
- Faculty of Medicine, Institute of Biomedical Sciences, Department of Pathology, Forensic Medicine and Pharmacology, Vilnius University, 03101 Vilnius, Lithuania
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, 08406 Vilnius, Lithuania
- Correspondence:
| | - Dovile Zilenaite-Petrulaitiene
- Faculty of Medicine, Institute of Biomedical Sciences, Department of Pathology, Forensic Medicine and Pharmacology, Vilnius University, 03101 Vilnius, Lithuania
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, 08406 Vilnius, Lithuania
| | - Allan Rasmusson
- Faculty of Medicine, Institute of Biomedical Sciences, Department of Pathology, Forensic Medicine and Pharmacology, Vilnius University, 03101 Vilnius, Lithuania
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, 08406 Vilnius, Lithuania
| | - Aiste Gulla
- Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 03101 Vilnius, Lithuania
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Agne Grigonyte
- Faculty of Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Kestutis Strupas
- Faculty of Medicine, Institute of Clinical Medicine, Vilnius University, 03101 Vilnius, Lithuania
| | - Arvydas Laurinavicius
- Faculty of Medicine, Institute of Biomedical Sciences, Department of Pathology, Forensic Medicine and Pharmacology, Vilnius University, 03101 Vilnius, Lithuania
- National Center of Pathology, Affiliate of Vilnius University Hospital Santaros Clinics, 08406 Vilnius, Lithuania
| |
Collapse
|
34
|
D'Amico G, Colli A, Malizia G, Casazza G. The potential role of machine learning in modelling advanced chronic liver disease. Dig Liver Dis 2022; 55:704-713. [PMID: 36586769 DOI: 10.1016/j.dld.2022.12.002] [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: 11/03/2022] [Revised: 12/05/2022] [Accepted: 12/08/2022] [Indexed: 01/02/2023]
Abstract
The use of artificial intelligence is rapidly increasing in medicine to support clinical decision making mostly through diagnostic and prediction models. Such models derive from huge databases (big data) including a large variety of health-related individual patient data (input) and the corresponding diagnosis and/or outcome (labels). Various types of algorithms (e.g. neural networks) based on powerful computational ability (machine), allow to detect the relationship between input and labels (learning). More complex algorithms, like recurrent neural network can learn from previous as well as actual input (deep learning) and are used for more complex tasks like imaging analysis and personalized (bespoke) medicine. The prompt availability of big data makes that artificial intelligence can provide rapid answers to questions that would require years of traditional clinical research. It may therefore be a key tool to overcome several major gaps in the model of advanced chronic liver disease, mostly transition from mild to clinically significant portal hypertension, the impact of acute decompensation and the role of further decompensation and treatment efficiency. However, several limitations of artificial intelligence should be overcome before its application in clinical practice. Assessment of the risk of bias, understandability of the black boxes developing the models and models' validation are the most important areas deserving clarification for artificial intelligence to be widely accepted from physicians and patients.
Collapse
Affiliation(s)
- Gennaro D'Amico
- Gatroenterology Unit, Azienda Ospedaliera Ospedali Riuniti Villa Sofia-Cervello, Palermo, Italy; Gastroenterology Unit, Clinica La Maddalena, Palermo, Italy.
| | - Agostino Colli
- Department of Transfusion Medicine and Haematology Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | | | - Giovanni Casazza
- Department of Clinical Sciences and Community Health - Laboratory of Medical Statistics, Biometry and Epidemiology "G.A. Maccacaro", Università degli Studi di Milano, Milan, Italy; Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
35
|
Zeng Z, Wang Q, Yu Y, Zhang Y, Chen Q, Lou W, Wang Y, Yan L, Cheng Z, Xu L, Yi Y, Fan G, Deng L. Assessing electrocardiogram changes after ischemic stroke with artificial intelligence. PLoS One 2022; 17:e0279706. [PMID: 36574427 PMCID: PMC9794063 DOI: 10.1371/journal.pone.0279706] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Accepted: 12/13/2022] [Indexed: 12/28/2022] Open
Abstract
OBJECTIVE Ischemic stroke (IS) with subsequent cerebrocardiac syndrome (CCS) has a poor prognosis. We aimed to investigate electrocardiogram (ECG) changes after IS with artificial intelligence (AI). METHODS We collected ECGs from a healthy population and patients with IS, and then analyzed participant demographics and ECG parameters to identify abnormal features in post-IS ECGs. Next, we trained the convolutional neural network (CNN), random forest (RF) and support vector machine (SVM) models to automatically detect the changes in the ECGs; Additionally, We compared the CNN scores of good prognosis (mRS ≤ 2) and poor prognosis (mRS > 2) to assess the prognostic value of CNN model. Finally, we used gradient class activation map (Grad-CAM) to localize the key abnormalities. RESULTS Among the 3506 ECGs of the IS patients, 2764 ECGs (78.84%) led to an abnormal diagnosis. Then we divided ECGs in the primary cohort into three groups, normal ECGs (N-Ns), abnormal ECGs after the first ischemic stroke (A-ISs), and normal ECGs after the first ischemic stroke (N-ISs). Basic demographic and ECG parameter analyses showed that heart rate, QT interval, and P-R interval were significantly different between 673 N-ISs and 3546 N-Ns (p < 0.05). The CNN has the best performance among the three models in distinguishing A-ISs and N-Ns (AUC: 0.88, 95%CI = 0.86-0.90). The prediction scores of the A-ISs and N-ISs obtained from the all three models are statistically different from the N-Ns (p < 0.001). Futhermore, the CNN scores of the two groups (mRS > 2 and mRS ≤ 2) were significantly different (p < 0.05). Finally, Grad-CAM revealed that the V4 lead may harbor the highest probability of abnormality. CONCLUSION Our study showed that a high proportion of post-IS ECGs harbored abnormal changes. Our CNN model can systematically assess anomalies in and prognosticate post-IS ECGs.
Collapse
Affiliation(s)
- Ziqiang Zeng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
| | - Qixuan Wang
- Queen Mary School, Medical College of Nanchang University, Nanchang, China
| | - Yingjing Yu
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
| | - Yichu Zhang
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Qi Chen
- Department of Cardiovascular Medicine, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Weiming Lou
- Institute of Translational Medicine, Nanchang University, Nanchang, China
| | - Yuting Wang
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
| | - Lingyu Yan
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
| | - Zujue Cheng
- Department of Neurosurgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Institute of Neuroscience, Nanchang University, Nanchang, China
| | - Lijun Xu
- Department of Neurology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yingping Yi
- Department of Medical Big Data Center, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Guangqin Fan
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
| | - Libin Deng
- Jiangxi Provincial Key Laboratory of Preventive Medicine, Nanchang University, Nanchang, P.R. China
- School of Public Health, Nanchang University, Nanchang, China
- The Institute of Periodontal Disease, Nanchang University, Nanchang, China
- * E-mail:
| |
Collapse
|
36
|
Mit künstlicher Intelligenz Lebertumoren differenzieren. ZEITSCHRIFT FÜR GASTROENTEROLOGIE 2022. [DOI: 10.1055/a-1873-3877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
37
|
Gerussi A, Scaravaglio M, Cristoferi L, Verda D, Milani C, De Bernardi E, Ippolito D, Asselta R, Invernizzi P, Kather JN, Carbone M. Artificial intelligence for precision medicine in autoimmune liver disease. Front Immunol 2022; 13:966329. [PMID: 36439097 PMCID: PMC9691668 DOI: 10.3389/fimmu.2022.966329] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 10/13/2022] [Indexed: 09/10/2023] Open
Abstract
Autoimmune liver diseases (AiLDs) are rare autoimmune conditions of the liver and the biliary tree with unknown etiology and limited treatment options. AiLDs are inherently characterized by a high degree of complexity, which poses great challenges in understanding their etiopathogenesis, developing novel biomarkers and risk-stratification tools, and, eventually, generating new drugs. Artificial intelligence (AI) is considered one of the best candidates to support researchers and clinicians in making sense of biological complexity. In this review, we offer a primer on AI and machine learning for clinicians, and discuss recent available literature on its applications in medicine and more specifically how it can help to tackle major unmet needs in AiLDs.
Collapse
Affiliation(s)
- Alessio Gerussi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Miki Scaravaglio
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Laura Cristoferi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4, School of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
| | | | - Chiara Milani
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Elisabetta De Bernardi
- Department of Medicine and Surgery and Tecnomed Foundation, University of Milano - Bicocca, Monza, Italy
| | | | - Rosanna Asselta
- Humanitas Clinical and Research Center, Rozzano, Milan, Italy
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy
| | - Pietro Invernizzi
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany
- Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| | - Marco Carbone
- Division of Gastroenterology, Center for Autoimmune Liver Diseases, Department of Medicine and Surgery, University of Milano-Bicocca, Monza, Italy
- European Reference Network on Hepatological Diseases (ERN RARE-LIVER), San Gerardo Hospital, Monza, Italy
| |
Collapse
|
38
|
Wang K, Ren Y, Ma L, Fan Y, Yang Z, Yang Q, Shi J, Sun Y. Deep Learning-Based Prediction of Treatment Prognosis from Nasal Polyp Histology Slides. Int Forum Allergy Rhinol 2022; 13:886-898. [PMID: 36066094 DOI: 10.1002/alr.23083] [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: 05/18/2022] [Revised: 08/29/2022] [Accepted: 08/31/2022] [Indexed: 11/11/2022]
Abstract
BACKGROUND Histopathology of nasal polyps contains rich prognostic information, which is difficult to objectively extract. In the present study, we aimed to develop a prognostic indicator of patient outcomes by analyzing scanned conventional haematoxylin and eosin (H&E) -stained slides alone using deep learning. METHODS An interpretable supervised deep learning model was developed using 185 H&E-stained whole-slide images (WSIs) of nasal polyps, each from a patient randomly selected from the pool of 232 patients who underwent endoscopic sinus surgery at the First Affiliated Hospital of Sun Yat-sen University (internal cohort). We internally validated the model on a holdout dataset from the internal cohort (47 H&E-stained WSIs) and externally validated the model on 122 H&E-stained WSIs from the Seventh Affiliated Hospital of Sun Yat-sen University and the University of Hong Kong-Shenzhen Hospital (external cohort). A poor prognosis score (PPS) was established to evaluate patient outcomes, and then risk activation mapping was applied to visualize the histopathological features underlying PPS. RESULTS The model yielded a patient-level sensitivity of 79.5%, and specificity of 92.3%, with areas under the receiver operating characteristic curve of 0.943, on the multi-center external cohort. The predictive ability of PPS was superior to that of conventional tissue eosinophil number. Notably, eosinophil infiltration, goblet cell hyperplasia, glandular hyperplasia, squamous metaplasia, and fibrin deposition were identified as the main underlying features of PPS. CONCLUSIONS Our deep learning model is an effective method for decoding pathological images of nasal polyps, providing a valuable solution for disease prognosis prediction and precise patient treatment. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Kanghua Wang
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yong Ren
- Center for Digestive Disease, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Guangdong Provincial Key Laboratory of Digestive Cancer Research, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Ling Ma
- Department of Otorhinolaryngology, the University of Hong Kong-Shenzhen Hospital, Shenzhen, 518053, China
| | - Yunping Fan
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Zheng Yang
- Department of Pathology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China
| | - Qintai Yang
- Department of Otorhinolaryngology-Head and Neck Surgery, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Jianbo Shi
- Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Yueqi Sun
- Department of Otolaryngology, the Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, 518107, China.,Department of Otolaryngology, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| |
Collapse
|