1
|
Molero A, Hernandez S, Alonso M, Peressini M, Curto D, Lopez-Rios F, Conde E. Assessment of PD-L1 expression and tumour infiltrating lymphocytes in early-stage non-small cell lung carcinoma with artificial intelligence algorithms. J Clin Pathol 2024:jcp-2024-209766. [PMID: 39419594 DOI: 10.1136/jcp-2024-209766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/26/2024] [Indexed: 10/19/2024]
Abstract
AIMS To study programmed death ligand 1 (PD-L1) expression and tumour infiltrating lymphocytes (TILs) in patients with early-stage non-small cell lung carcinoma (NSCLC) with artificial intelligence (AI) algorithms. METHODS The study included samples from 50 early-stage NSCLCs. PD-L1 immunohistochemistry (IHC) stained slides (clone SP263) were scored manually and with two different AI tools (PathAI and Navify Digital Pathology) by three pathologists. TILs were digitally assessed on H&E and CD8 IHC stained sections with two different algorithms (PathAI and Navify Digital Pathology, respectively). The agreement between observers and methods for each biomarker was analysed. For PD-L1, the turn-around time (TAT) for manual versus AI-assisted scoring was recorded. RESULTS Agreement was higher in tumours with low PD-L1 expression regardless of the approach. Both AI-powered tools identified a significantly higher number of cases equal or above 1% PD-L1 tumour proportion score as compared with manual scoring (p=0.00015), a finding with potential therapeutic implications. Regarding TAT, there were significant differences between manual scoring and AI use (p value <0.0001 for all comparisons). The total TILs density with the PathAI algorithm and the total density of CD8+ cells with the Navify Digital Pathology software were significantly correlated (τ=0.49 (95% CI 0.37, 0.61), p value<0.0001). CONCLUSIONS This preliminary study supports the use of AI algorithms for the scoring of PD-L1 and TILs in patients with NSCLC.
Collapse
Affiliation(s)
- Aida Molero
- Pathology, Complejo Asistencial de Segovia, Segovia, Spain
| | - Susana Hernandez
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Marta Alonso
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Melina Peressini
- Tumor Microenvironment and Immunotherapy Research Group, Research Institute Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Daniel Curto
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
| | - Fernando Lopez-Rios
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), CIBERONC, Universidad Complutense de Madrid, Madrid, Spain
| | - Esther Conde
- Pathology, Hospital Universitario 12 de Octubre, Madrid, Spain
- Research Institute Hospital 12 de Octubre (i+12), CIBERONC, Universidad Complutense de Madrid, Madrid, Spain
| |
Collapse
|
2
|
Alnafisah KH, Ranjan A, Sahu SP, Chen J, Alhejji SM, Noël A, Gartia MR, Mukhopadhyay S. Machine learning for automated classification of lung collagen in a urethane-induced lung injury mouse model. BIOMEDICAL OPTICS EXPRESS 2024; 15:5980-5998. [PMID: 39421774 PMCID: PMC11482176 DOI: 10.1364/boe.527972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 06/18/2024] [Accepted: 07/25/2024] [Indexed: 10/19/2024]
Abstract
Dysregulation of lung tissue collagen level plays a vital role in understanding how lung diseases progress. However, traditional scoring methods rely on manual histopathological examination introducing subjectivity and inconsistency into the assessment process. These methods are further hampered by inter-observer variability, lack of quantification, and their time-consuming nature. To mitigate these drawbacks, we propose a machine learning-driven framework for automated scoring of lung collagen content. Our study begins with the collection of a lung slide image dataset from adult female mice using second harmonic generation (SHG) microscopy. In our proposed approach, first, we manually extracted features based on the 46 statistical parameters of fibrillar collagen. Subsequently, we pre-processed the images and utilized a pre-trained VGG16 model to uncover hidden features from pre-processed images. We then combined both image and statistical features to train various machine learning and deep neural network models for classification tasks. We employed advanced unsupervised techniques like K-means, principal component analysis (PCA), t-distributed stochastic neighbour embedding (t-SNE), and uniform manifold approximation and projection (UMAP) to conduct thorough image analysis for lung collagen content. Also, the evaluation of the trained models using the collagen data includes both binary and multi-label classification to predict lung cancer in a urethane-induced mouse model. Experimental validation of our proposed approach demonstrates promising results. We obtained an average accuracy of 83% and an area under the receiver operating characteristic curve (ROC AUC) values of 0.96 through the use of a support vector machine (SVM) model for binary categorization tasks. For multi-label classification tasks, to quantify the structural alteration of collagen, we attained an average accuracy of 73% and ROC AUC values of 1.0, 0.38, 0.95, and 0.86 for control, baseline, treatment_1, and treatment_2 groups, respectively. Our findings provide significant potential for enhancing diagnostic accuracy, understanding disease mechanisms, and improving clinical practice using machine learning and deep learning models.
Collapse
Affiliation(s)
| | - Amit Ranjan
- Center for Computation & Technology and Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Sushant P Sahu
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
- Amity Institute of Biotechnology and Applied Sciences, Amity University, Mumbai, Maharashtra-410206, India
| | - Jianhua Chen
- Department of Computer Science, Louisiana State University, Baton Rouge, LA 70803, USA
| | | | - Alexandra Noël
- Department of Comparative Biomedical Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Manas Ranjan Gartia
- Department of Mechanical and Industrial Engineering, Louisiana State University, Baton Rouge, LA 70803, USA
| | - Supratik Mukhopadhyay
- Center for Computation & Technology and Department of Environmental Sciences, Louisiana State University, Baton Rouge, LA 70803, USA
| |
Collapse
|
3
|
Xiong Z, Liu K, Liu S, Feng J, Wang J, Feng Z, Lai B, Zhang Q, Jiang Q, Zhang W. Precision HER2: a comprehensive AI system for accurate and consistent evaluation of HER2 expression in invasive breast Cancer. BMC Cancer 2024; 24:1204. [PMID: 39350085 PMCID: PMC11441240 DOI: 10.1186/s12885-024-12980-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND With the development of novel anti-HER2 targeted drugs, such as ADCs, it has become increasingly important to accurately interpret HER2 expression in breast cancer. Previous studies have demonstrated high intra-observer and inter-observer variabilities in evaluating HER2 staining by human eyes. There exists a strong requirement to develop artificial intelligence (AI) systems to achieve high-precision HER2 expression scoring for better clinical therapy. METHODS In the present study, we collected breast cancer tissue samples and stained consecutive sections with anti-Calponin and anti-HER2 antibodies. High-quality digital images were selected from immunohistochemical slides and interpreted as HER2 3+, 2+, 1+, and 0. AI models were trained and assessed using annotated training and testing sets. The AI model was trained to automatically identify ductal carcinoma in situ (DCIS) by Calponin staining and myoepithelial annotation and filter out DCIS components in HER2-stained slides using image-overlapping techniques. Furthermore, we organized two-phase validation studies. In phase one, pathologists interpreted 112 HER2 whole-slide images (WSIs) without AI assistance, whereas in phase two, pathologists read the same slides using the AI system after a washing period of 2 weeks. RESULTS Our AI model greatly improved the accuracy of reading (0.902 vs. 0.710). The number of HER2 1 + patients misdiagnosed as HER2 0 was significantly reduced (32/279 vs. 65/279), and they benefitted from ADC drugs. In addition, the AI algorithm improved the intra-group consistency of HER2 readings by pathologists with different years of experience (intra-class correlation coefficient [ICC]: 0.872-0.926 vs. 0.818-0.908), with the improvement most pronounced among junior pathologists (0.885 vs. 0.818). CONCLUSIONS We proposed a high-precision AI system to identify and filter out DCIS components and automatically evaluate HER2 expression in invasive breast cancer.
Collapse
Affiliation(s)
- Zhongtang Xiong
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China
| | - Kai Liu
- Cells Vision (Guangzhou) Medical Technology Inc, Guangzhou, China
| | - Shaoyan Liu
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China
| | - Jiahao Feng
- Cells Vision (Guangzhou) Medical Technology Inc, Guangzhou, China
| | - Jin Wang
- Cells Vision (Guangzhou) Medical Technology Inc, Guangzhou, China
| | - Zewen Feng
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China
| | - Boan Lai
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China
| | - Qingxin Zhang
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China
| | - Qingping Jiang
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China.
| | - Wei Zhang
- Department of Pathology; Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, NO.63, Duobao Road, Guangzhou, 510150, China.
| |
Collapse
|
4
|
Han D, Li H, Zheng X, Fu S, Wei R, Zhao Q, Liu C, Wang Z, Huang W, Hao S. Whole slide image-based weakly supervised deep learning for predicting major pathological response in non-small cell lung cancer following neoadjuvant chemoimmunotherapy: a multicenter, retrospective, cohort study. Front Immunol 2024; 15:1453232. [PMID: 39372403 PMCID: PMC11449764 DOI: 10.3389/fimmu.2024.1453232] [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: 06/22/2024] [Accepted: 08/27/2024] [Indexed: 10/08/2024] Open
Abstract
Objective Develop a predictive model utilizing weakly supervised deep learning techniques to accurately forecast major pathological response (MPR) in patients with resectable non-small cell lung cancer (NSCLC) undergoing neoadjuvant chemoimmunotherapy (NICT), by leveraging whole slide images (WSIs). Methods This retrospective study examined pre-treatment WSIs from 186 patients with non-small cell lung cancer (NSCLC), using a weakly supervised learning framework. We employed advanced deep learning architectures, including DenseNet121, ResNet50, and Inception V3, to analyze WSIs on both micro (patch) and macro (slide) levels. The training process incorporated innovative data augmentation and normalization techniques to bolster the robustness of the models. We evaluated the performance of these models against traditional clinical predictors and integrated them with a novel pathomics signature, which was developed using multi-instance learning algorithms that facilitate feature aggregation from patch-level probability distributions. Results Univariate and multivariable analyses confirmed histology as a statistically significant prognostic factor for MPR (P-value< 0.05). In patch model evaluations, DenseNet121 led in the validation set with an area under the curve (AUC) of 0.656, surpassing ResNet50 (AUC = 0.626) and Inception V3 (AUC = 0.654), and showed strong generalization in external testing (AUC = 0.611). Further evaluation through visual inspection of patch-level data integration into WSIs revealed XGBoost's superior class differentiation and generalization, achieving the highest AUCs of 0.998 in training and robust scores of 0.818 in validation and 0.805 in testing. Integrating pathomics features with clinical data into a nomogram yielded AUC of 0.819 in validation and 0.820 in testing, enhancing discriminative accuracy. Gradient-weighted Class Activation Mapping (Grad-CAM) and feature aggregation methods notably boosted the model's interpretability and feature modeling. Conclusion The application of weakly supervised deep learning to WSIs offers a powerful tool for predicting MPR in NSCLC patients treated with NICT.
Collapse
Affiliation(s)
- Dan Han
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Hao Li
- Department of Radiology, Shandong Provincial Qianfoshan Hospital, Shandong University, Jinan, Shandong, China
- Department of Radiation Oncology and Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Xin Zheng
- Department of Traditional Chinese Medicine, Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, China
| | - Shenbo Fu
- Department of Radiation Oncology, Shanxi Provincial Tumor Hospital, Xi’an, Shanxi, China
| | - Ran Wei
- Department of Radiology, Jining No.1 People’s Hospital, Jining, Shandong, China
| | - Qian Zhao
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Chengxin Liu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Zhongtang Wang
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Wei Huang
- Department of Radiation Oncology, Shandong University Cancer Center, Jinan, Shandong, China
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| | - Shaoyu Hao
- Department of Thoracic Surgery, Shandong University Cancer Center, Jinan, Shandong, China
- Department of Thoracic Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, and Shandong Academy of Medical Sciences, Jinan, Shandong, China
| |
Collapse
|
5
|
Li J, Dong P, Wang X, Zhang J, Zhao M, Shen H, Cai L, He J, Han M, Miao J, Liu H, Yang W, Han X, Liu Y. Artificial intelligence enhances whole-slide interpretation of PD-L1 CPS in triple-negative breast cancer: A multi-institutional ring study. Histopathology 2024; 85:451-467. [PMID: 38747491 DOI: 10.1111/his.15205] [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: 10/11/2023] [Revised: 03/11/2024] [Accepted: 04/21/2024] [Indexed: 08/09/2024]
Abstract
BACKGROUND AND AIMS Evaluation of the programmed cell death ligand-1 (PD-L1) combined positive score (CPS) is vital to predict the efficacy of the immunotherapy in triple-negative breast cancer (TNBC), but pathologists show substantial variability in the consistency and accuracy of the interpretation. It is of great importance to establish an objective and effective method which is highly repeatable. METHODS We proposed a model in a deep learning-based framework, which at the patch level incorporated cell analysis and tissue region analysis, followed by the whole-slide level fusion of patch results. Three rounds of ring studies (RSs) were conducted. Twenty-one pathologists of different levels from four institutions evaluated the PD-L1 CPS in TNBC specimens as continuous scores by visual assessment and our artificial intelligence (AI)-assisted method. RESULTS In the visual assessment, the interpretation results of PD-L1 (Dako 22C3) CPS by different levels of pathologists have significant differences and showed weak consistency. Using AI-assisted interpretation, there were no significant differences between all pathologists (P = 0.43), and the intraclass correlation coefficient (ICC) value was increased from 0.618 [95% confidence interval (CI) = 0.524-0.719] to 0.931 (95% CI = 0.902-0.955). The accuracy of interpretation result is further improved to 0.919 (95% CI = 0.886-0.947). Acceptance of AI results by junior pathologists was the highest among all levels, and 80% of the AI results were accepted overall. CONCLUSION With the help of the AI-assisted diagnostic method, different levels of pathologists achieved excellent consistency and repeatability in the interpretation of PD-L1 (Dako 22C3) CPS. Our AI-assisted diagnostic approach was proved to strengthen the consistency and repeatability in clinical practice.
Collapse
Affiliation(s)
- Jinze Li
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Pei Dong
- AI Lab, Tencent, Shenzhen, Guangdong, China
| | - Xinran Wang
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jun Zhang
- AI Lab, Tencent, Shenzhen, Guangdong, China
| | - Meng Zhao
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | | | - Lijing Cai
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiankun He
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Mengxue Han
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Jiaxian Miao
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hongbo Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Wei Yang
- AI Lab, Tencent, Shenzhen, Guangdong, China
| | - Xiao Han
- AI Lab, Tencent, Shenzhen, Guangdong, China
| | - Yueping Liu
- Department of Pathology, The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| |
Collapse
|
6
|
Caranfil E, Lami K, Uegami W, Fukuoka J. Artificial Intelligence and Lung Pathology. Adv Anat Pathol 2024; 31:344-351. [PMID: 38780094 DOI: 10.1097/pap.0000000000000448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
This manuscript provides a comprehensive overview of the application of artificial intelligence (AI) in lung pathology, particularly in the diagnosis of lung cancer. It discusses various AI models designed to support pathologists and clinicians. AI models supporting pathologists are to standardize diagnosis, score PD-L1 status, supporting tumor cellularity count, and indicating explainability for pathologic judgements. Several models predict outcomes beyond pathologic diagnosis and predict clinical outcomes like patients' survival and molecular alterations. The manuscript emphasizes the potential of AI to enhance accuracy and efficiency in pathology, while also addressing the challenges and future directions for integrating AI into clinical practice.
Collapse
Affiliation(s)
- Emanuel Caranfil
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Kris Lami
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
| | - Wataru Uegami
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| | - Junya Fukuoka
- Department of Pathology Informatics, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki
- Department of Pathology, Kameda Medical Center, Kamogawa, Japan
| |
Collapse
|
7
|
Cortellini A, Zampacorta C, De Tursi M, Grillo LR, Ricciardi S, Bria E, Martini M, Giusti R, Filetti M, Dal Mas A, Russano M, Dall’Olio FG, Buttitta F, Marchetti A. A preliminary study on the diagnostic performance of the uPath PD-L1 (SP263) artificial intelligence (AI) algorithm in patients with NSCLC treated with PD-1/PD-L1 checkpoint blockade. Pathologica 2024; 116:222-231. [PMID: 39377504 PMCID: PMC11460154 DOI: 10.32074/1591-951x-998] [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: 03/22/2024] [Accepted: 07/12/2024] [Indexed: 10/09/2024] Open
Abstract
Objective The uPath PD-L1 (SP263) is an AI-based platform designed to aid pathologists in identifying and quantifying PD-L1 positive tumor cells in non-small cell lung cancer (NSCLC) samples stained with the SP263 assay. Methods In this preliminary study, we explored the diagnostic performance of the uPath PD-L1 algorithm in defining PD-L1 tumor proportion score (TPS) and predict clinical outcomes in a series of patients with advanced stage NSCLC treated with single agent PD-1/PD-L1 checkpoint blockade previously assessed with the SP263 assay in clinical practice. Results 44 patients treated from August 2015 to January 2019 were included, with baseline PD-L1 TPS of ≥ 50%, 1-49% and < 1% in 38.6%, 25.0% and 36.4%, respectively. The median uPath PD-L1 score was 6 with a significant correlation with the baseline PD-L1 TPS (r: 0.83, p < 0.01). However, only 27 cases (61.4%) were scored within the same clinically relevant range of expression (≥ vs < 50%). In the study population the baseline PD-L1 TPS was not significantly associated with clinical outcomes, while the uPath PD-L1 score showed a good diagnostic ability for the risk of death at the ROC curve analysis [AUC: 0.81 (95%CI: 0.66-0.91), optimal cut-off of ≥ 3.2], resulting in 19 patients (43.2%) being u-Path low and 25 patients (56.8%) being uPath high. The objective response rate in uPath high and low was 51.6% and 25.0% (p = 0.1), respectively, although the uPath was significantly associated with overall survival (OS, HR 2.45, 95%CI: 1.19-5.05) and progression free survival (PFS, HR 3.04, 95%CI: 1.51-6.14). At the inverse probability of treatment weighting analysis used to balance baseline covariates, the uPath categories confirmed to be independently associated with OS and PFS. Conclusions This preliminary analysis suggests that AI-based, digital pathology tools such as uPath PD-L1 (SP263) can be used to optimize already available biomarkers for immune-oncology treatment in patients with NSCLC.
Collapse
Affiliation(s)
- Alessio Cortellini
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
- Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Roma, Italy
- Department of Surgery and Cancer, Hammersmith Hospital Campus, Imperial College London, London, United Kingdom
| | - Claudia Zampacorta
- Center for Advanced Studies and Technology (CAST), University Chieti-Pescara, Italy
- Diagnostic Molecular Pathology, Unit of Anatomic Pathology, SS Annunziata Hospital, Chieti, Italy
| | - Michele De Tursi
- Department of Innovative Technologies in Medicine & Dentistry, University G. D’Annunzio, Chieti-Pescara, Italy
| | - Lucia R. Grillo
- Anatomic Pathology Unit, San Camillo-Forlanini Hospitals, Rome, Italy
| | | | - Emilio Bria
- Medical Oncology, Università Cattolica del Sacro Cuore, Rome, Italy
- Comprehensive Cancer Center, Fondazione Policlinico Universitario Agostino Gemelli, Istituto di Ricovero e Cura a Carattere Scientifico, Rome, Italy
| | - Maurizio Martini
- Department of Human Pathology in Adult and Developmental Age “Gaetano Barresi”, Section of Pathology, University of Messina, Messina, Italy
| | - Raffaele Giusti
- Medical Oncology Unit, Azienda Ospedaliero Universitaria Sant’Andrea, Rome, Italy
| | - Marco Filetti
- Phase 1 Unit, Fondazione Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Department of Experimental Medicine, Sapienza University of Rome, Rome, Italy
| | | | - Marco Russano
- Operative Research Unit of Medical Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Roma, Italy
| | | | - Fiamma Buttitta
- Center for Advanced Studies and Technology (CAST), University Chieti-Pescara, Italy
- Diagnostic Molecular Pathology, Unit of Anatomic Pathology, SS Annunziata Hospital, Chieti, Italy
- Department of Medical, Oral, and Biotechnological Sciences University “G. D’Annunzio” of Chieti-Pescara
| | - Antonio Marchetti
- Center for Advanced Studies and Technology (CAST), University Chieti-Pescara, Italy
- Diagnostic Molecular Pathology, Unit of Anatomic Pathology, SS Annunziata Hospital, Chieti, Italy
- Department of Medical, Oral, and Biotechnological Sciences University “G. D’Annunzio” of Chieti-Pescara
| |
Collapse
|
8
|
Ma M, Zeng X, Qu L, Sheng X, Ren H, Chen W, Li B, You Q, Xiao L, Wang Y, Dai M, Zhang B, Lu C, Sheng W, Huang D. Advancing Automatic Gastritis Diagnosis: An Interpretable Multilabel Deep Learning Framework for the Simultaneous Assessment of Multiple Indicators. THE AMERICAN JOURNAL OF PATHOLOGY 2024; 194:1538-1549. [PMID: 38762117 DOI: 10.1016/j.ajpath.2024.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Revised: 03/17/2024] [Accepted: 04/26/2024] [Indexed: 05/20/2024]
Abstract
The evaluation of morphologic features, such as inflammation, gastric atrophy, and intestinal metaplasia, is crucial for diagnosing gastritis. However, artificial intelligence analysis for nontumor diseases like gastritis is limited. Previous deep learning models have omitted important morphologic indicators and cannot simultaneously diagnose gastritis indicators or provide interpretable labels. To address this, an attention-based multi-instance multilabel learning network (AMMNet) was developed to simultaneously achieve the multilabel diagnosis of activity, atrophy, and intestinal metaplasia with only slide-level weak labels. To evaluate AMMNet's real-world performance, a diagnostic test was designed to observe improvements in junior pathologists' diagnostic accuracy and efficiency with and without AMMNet assistance. In this study of 1096 patients from seven independent medical centers, AMMNet performed well in assessing activity [area under the curve (AUC), 0.93], atrophy (AUC, 0.97), and intestinal metaplasia (AUC, 0.93). The false-negative rates of these indicators were only 0.04, 0.08, and 0.18, respectively, and junior pathologists had lower false-negative rates with model assistance (0.15 versus 0.10). Furthermore, AMMNet reduced the time required per whole slide image from 5.46 to 2.85 minutes, enhancing diagnostic efficiency. In block-level clustering analysis, AMMNet effectively visualized task-related patches within whole slide images, improving interpretability. These findings highlight AMMNet's effectiveness in accurately evaluating gastritis morphologic indicators on multicenter data sets. Using multi-instance multilabel learning strategies to support routine diagnostic pathology deserves further evaluation.
Collapse
Affiliation(s)
- Mengke Ma
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Xixi Zeng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Linhao Qu
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China
| | - Xia Sheng
- Department of Pathology, Minhang Hospital, Fudan University, Shanghai, China
| | - Hongzheng Ren
- Department of Pathology, Gongli Hospital, Naval Medical University, Shanghai, China
| | - Weixiang Chen
- Department of Pathology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Bin Li
- Department of Pathology, Shanghai Xu-Hui Central Hospital, Shanghai, China
| | - Qinghua You
- Department of Pathology, Shanghai Pudong Hospital, Fudan University Pudong Medical Center, Shanghai, China
| | - Li Xiao
- Department of Pathology, Huadong Hospital, Shanghai, China
| | - Yi Wang
- Information Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mei Dai
- Information Center, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Boqiang Zhang
- Shanghai Foremost Medical Technology Co. Ltd., Shanghai, China
| | - Changqing Lu
- Shanghai Foremost Medical Technology Co. Ltd., Shanghai, China
| | - Weiqi Sheng
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China.
| | - Dan Huang
- Department of Pathology, Fudan University Shanghai Cancer Center, Shanghai, China; Department of Oncology, Fudan University Shanghai Medical College, Shanghai, China; Institute of Pathology, Fudan University, Shanghai, China.
| |
Collapse
|
9
|
Gharaibeh NY, De Fazio R, Al-Naami B, Al-Hinnawi AR, Visconti P. Automated Lung Cancer Diagnosis Applying Butterworth Filtering, Bi-Level Feature Extraction, and Sparce Convolutional Neural Network to Luna 16 CT Images. J Imaging 2024; 10:168. [PMID: 39057739 PMCID: PMC11277772 DOI: 10.3390/jimaging10070168] [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: 04/27/2024] [Revised: 06/12/2024] [Accepted: 07/08/2024] [Indexed: 07/28/2024] Open
Abstract
Accurate prognosis and diagnosis are crucial for selecting and planning lung cancer treatments. As a result of the rapid development of medical imaging technology, the use of computed tomography (CT) scans in pathology is becoming standard practice. An intricate interplay of requirements and obstacles characterizes computer-assisted diagnosis, which relies on the precise and effective analysis of pathology images. In recent years, pathology image analysis tasks such as tumor region identification, prognosis prediction, tumor microenvironment characterization, and metastasis detection have witnessed the considerable potential of artificial intelligence, especially deep learning techniques. In this context, an artificial intelligence (AI)-based methodology for lung cancer diagnosis is proposed in this research work. As a first processing step, filtering using the Butterworth smooth filter algorithm was applied to the input images from the LUNA 16 lung cancer dataset to remove noise without significantly degrading the image quality. Next, we performed the bi-level feature selection step using the Chaotic Crow Search Algorithm and Random Forest (CCSA-RF) approach to select features such as diameter, margin, spiculation, lobulation, subtlety, and malignancy. Next, the Feature Extraction step was performed using the Multi-space Image Reconstruction (MIR) method with Grey Level Co-occurrence Matrix (GLCM). Next, the Lung Tumor Severity Classification (LTSC) was implemented by using the Sparse Convolutional Neural Network (SCNN) approach with a Probabilistic Neural Network (PNN). The developed method can detect benign, normal, and malignant lung cancer images using the PNN algorithm, which reduces complexity and efficiently provides classification results. Performance parameters, namely accuracy, precision, F-score, sensitivity, and specificity, were determined to evaluate the effectiveness of the implemented hybrid method and compare it with other solutions already present in the literature.
Collapse
Affiliation(s)
- Nasr Y. Gharaibeh
- Department of Electrical Engineering, Al-Balqa Applied University, Salt 21163, Jordan;
| | - Roberto De Fazio
- Department of Innovation Engineering, University of Salento, 73100 Lecce, Italy;
| | - Bassam Al-Naami
- Department of Biomedical Engineering, Faculty of Engineering, The Hashemite University, Zarqa 13133, Jordan;
| | - Abdel-Razzak Al-Hinnawi
- Department of Medical Imaging, Faculty of Allied Medical Sciences, Isra University, Amman 11622, Jordan;
| | - Paolo Visconti
- Department of Innovation Engineering, University of Salento, 73100 Lecce, Italy;
| |
Collapse
|
10
|
Huang HN, Kuo CW, Hung YL, Yang CH, Hsieh YH, Lin YC, Chang MDT, Lin YY, Ko JC. Optimizing immunofluorescence with high-dynamic-range imaging to enhance PD-L1 expression evaluation for 3D pathology assessment from NSCLC tumor tissue. Sci Rep 2024; 14:15176. [PMID: 38956114 PMCID: PMC11219731 DOI: 10.1038/s41598-024-65187-x] [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: 03/29/2024] [Accepted: 06/18/2024] [Indexed: 07/04/2024] Open
Abstract
Assessing programmed death ligand 1 (PD-L1) expression through immunohistochemistry (IHC) is the golden standard in predicting immunotherapy response of non-small cell lung cancer (NSCLC). However, observation of heterogeneous PD-L1 distribution in tumor space is a challenge using IHC only. Meanwhile, immunofluorescence (IF) could support both planar and three-dimensional (3D) histological analyses by combining tissue optical clearing with confocal microscopy. We optimized clinical tissue preparation for the IF assay focusing on staining, imaging, and post-processing to achieve quality identical to traditional IHC assay. To overcome limited dynamic range of the fluorescence microscope's detection system, we incorporated a high dynamic range (HDR) algorithm to restore the post imaging IF expression pattern and further 3D IF images. Following HDR processing, a noticeable improvement in the accuracy of diagnosis (85.7%) was achieved using IF images by pathologists. Moreover, 3D IF images revealed a 25% change in tumor proportion score for PD-L1 expression at various depths within tumors. We have established an optimal and reproducible process for PD-L1 IF images in NSCLC, yielding high quality data comparable to traditional IHC assays. The ability to discern accurate spatial PD-L1 distribution through 3D pathology analysis could provide more precise evaluation and prediction for immunotherapy targeting advanced NSCLC.
Collapse
Affiliation(s)
- Hsien-Neng Huang
- Department of Pathology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
- Department and Graduate Institute of Pathology, College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Chun-Wei Kuo
- Department of Pathology, National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | | | | | | | | | | | | | - Jen-Chung Ko
- Department of Internal Medicine, National Taiwan University HospitalHsin-Chu Branch, No. 25, Ln. 442, Sec. 1, Jingguo Rd., North Dist., Hsinchu City, 300, Taiwan, ROC.
| |
Collapse
|
11
|
Lee KS, Choi E, Cho SI, Park S, Ryu J, Puche AV, Ma M, Park J, Jung W, Ro J, Kim S, Park G, Song S, Ock CY, Choe G, Park JH. An artificial intelligence-powered PD-L1 combined positive score (CPS) analyser in urothelial carcinoma alleviating interobserver and intersite variability. Histopathology 2024; 85:81-91. [PMID: 38477366 DOI: 10.1111/his.15176] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Revised: 02/20/2024] [Accepted: 02/29/2024] [Indexed: 03/14/2024]
Abstract
AIMS Immune checkpoint inhibitors targeting programmed death-ligand 1 (PD-L1) have shown promising clinical outcomes in urothelial carcinoma (UC). The combined positive score (CPS) quantifies PD-L1 22C3 expression in UC, but it can vary between pathologists due to the consideration of both immune and tumour cell positivity. METHODS AND RESULTS An artificial intelligence (AI)-powered PD-L1 CPS analyser was developed using 1,275,907 cells and 6175.42 mm2 of tissue annotated by pathologists, extracted from 400 PD-L1 22C3-stained whole slide images of UC. We validated the AI model on 543 UC PD-L1 22C3 cases collected from three institutions. There were 446 cases (82.1%) where the CPS results (CPS ≥10 or <10) were in complete agreement between three pathologists, and 486 cases (89.5%) where the AI-powered CPS results matched the consensus of two or more pathologists. In the pathologist's assessment of the CPS, statistically significant differences were noted depending on the source hospital (P = 0.003). Three pathologists reevaluated discrepancy cases with AI-powered CPS results. After using the AI as a guide and revising, the complete agreement increased to 93.9%. The AI model contributed to improving the concordance between pathologists across various factors including hospital, specimen type, pathologic T stage, histologic subtypes, and dominant PD-L1-positive cell type. In the revised results, the evaluation discordance among slides from different hospitals was mitigated. CONCLUSION This study suggests that AI models can help pathologists to reduce discrepancies between pathologists in quantifying immunohistochemistry including PD-L1 22C3 CPS, especially when evaluating data from different institutions, such as in a telepathology setting.
Collapse
Affiliation(s)
- Kyu Sang Lee
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Euno Choi
- Department of Pathology, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | | | | - Gheeyoung Choe
- Department of Pathology, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam-si, Republic of Korea
| | - Jeong Hwan Park
- Department of Pathology, SMG-SNU Boramae Medical Center, Seoul National University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
12
|
Hartman DJ. Applications of Artificial Intelligence in Lung Pathology. Surg Pathol Clin 2024; 17:321-328. [PMID: 38692814 DOI: 10.1016/j.path.2023.11.013] [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] [Indexed: 05/03/2024]
Abstract
Artificial intelligence/machine learning tools are being created for use in pathology. Some examples related to lung pathology include acid-fast stain evaluation, programmed death ligand-1 (PDL-1) interpretation, evaluating histologic patterns of non-small-cell lung carcinoma, evaluating histologic features in mesothelioma associated with adverse outcomes, predicting response to anti-PDL-1 therapy from hematoxylin and eosin-stained slides, evaluation of tumor microenvironment, evaluating patterns of interstitial lung disease, nondestructive methods for tissue evaluation, and others. There are still some frameworks (regulatory, workflow, and payment) that need to be established for these tools to be integrated into pathology.
Collapse
Affiliation(s)
- Douglas J Hartman
- University of Pittsburgh Medical Center, 200 Lothrop Street C-620, Pittsburgh, PA 15213, USA.
| |
Collapse
|
13
|
Knudsen BS, Jadhav A, Perry LJ, Thagaard J, Deftereos G, Ying J, Brintz BJ, Zhang W. A Pipeline for Evaluation of Machine Learning/Artificial Intelligence Models to Quantify Programmed Death Ligand 1 Immunohistochemistry. J Transl Med 2024; 104:102070. [PMID: 38677590 DOI: 10.1016/j.labinv.2024.102070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 04/08/2024] [Accepted: 04/18/2024] [Indexed: 04/29/2024] Open
Abstract
Immunohistochemistry (IHC) is used to guide treatment decisions in multiple cancer types. For treatment with checkpoint inhibitors, programmed death ligand 1 (PD-L1) IHC is used as a companion diagnostic. However, the scoring of PD-L1 is complicated by its expression in cancer and immune cells. Separation of cancer and noncancer regions is needed to calculate tumor proportion scores (TPS) of PD-L1, which is based on the percentage of PD-L1-positive cancer cells. Evaluation of PD-L1 expression requires highly experienced pathologists and is often challenging and time-consuming. Here, we used a multi-institutional cohort of 77 lung cancer cases stained centrally with the PD-L1 22C3 clone. We developed a 4-step pipeline for measuring TPS that includes the coregistration of hematoxylin and eosin, PD-L1, and negative control (NC) digital slides for exclusion of necrosis, segmentation of cancer regions, and quantification of PD-L1+ cells. As cancer segmentation is a challenging step for TPS generation, we trained DeepLab V3 in the Visiopharm software package to outline cancer regions in PD-L1 and NC images and evaluated the model performance by mean intersection over union (mIoU) against manual outlines. Only 14 cases were required to accomplish a mIoU of 0.82 for cancer segmentation in hematoxylin-stained NC cases. For PD-L1-stained slides, a model trained on PD-L1 tiles augmented by registered NC tiles achieved a mIoU of 0.79. In segmented cancer regions from whole slide images, the digital TPS achieved an accuracy of 75% against the manual TPS scores from the pathology report. Major reasons for algorithmic inaccuracies include the inclusion of immune cells in cancer outlines and poor nuclear segmentation of cancer cells. Our transparent and stepwise approach and performance metrics can be applied to any IHC assay to provide pathologists with important insights on when to apply and how to evaluate commercial automated IHC scoring systems.
Collapse
Affiliation(s)
- Beatrice S Knudsen
- Department of Pathology, University of Utah, Salt Lake City, Utah; Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| | | | - Lindsey J Perry
- Department of Pathology, University of Utah, Salt Lake City, Utah
| | | | - Georgios Deftereos
- Department of Pathology, University of California San Francisco, San Francisco, California
| | - Jian Ying
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Ben J Brintz
- Department of Internal Medicine, University of Utah, Salt Lake City, Utah
| | - Wei Zhang
- Department of Pathology, University of Utah, Salt Lake City, Utah; Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah.
| |
Collapse
|
14
|
Ito H, Yoshizawa A, Terada K, Nakakura A, Rokutan-Kurata M, Sugimoto T, Nishimura K, Nakajima N, Sumiyoshi S, Hamaji M, Menju T, Date H, Morita S, Bise R, Haga H. A Deep Learning-Based Assay for Programmed Death Ligand 1 Immunohistochemistry Scoring in Non-Small Cell Lung Carcinoma: Does it Help Pathologists Score? Mod Pathol 2024; 37:100485. [PMID: 38588885 DOI: 10.1016/j.modpat.2024.100485] [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: 08/10/2023] [Revised: 02/08/2024] [Accepted: 04/01/2024] [Indexed: 04/10/2024]
Abstract
Several studies have developed various artificial intelligence (AI) models for immunohistochemical analysis of programmed death ligand 1 (PD-L1) in patients with non-small cell lung carcinoma; however, none have focused on specific ways by which AI-assisted systems could help pathologists determine the tumor proportion score (TPS). In this study, we developed an AI model to calculate the TPS of the PD-L1 22C3 assay and evaluated whether and how this AI-assisted system could help pathologists determine the TPS and analyze how AI-assisted systems could affect pathologists' assessment accuracy. We assessed the 4 methods of the AI-assisted system: (1 and 2) pathologists first assessed and then referred to automated AI scoring results (1, positive tumor cell percentage; 2, positive tumor cell percentage and visualized overlay image) for final confirmation, and (3 and 4) pathologists referred to the automated AI scoring results (3, positive tumor cell percentage; 4, positive tumor cell percentage and visualized overlay image) while determining TPS. Mixed-model analysis was used to calculate the odds ratios (ORs) with 95% CI for AI-assisted TPS methods 1 to 4 compared with pathologists' scoring. For all 584 samples of the tissue microarray, the OR for AI-assisted TPS methods 1 to 4 was 0.94 to 1.07 and not statistically significant. Of them, we found 332 discordant cases, on which the pathologists' judgments were inconsistent; the ORs for AI-assisted TPS methods 1, 2, 3, and 4 were 1.28 (1.06-1.54; P = .012), 1.29 (1.06-1.55; P = .010), 1.28 (1.06-1.54; P = .012), and 1.29 (1.06-1.55; P = .010), respectively, which were statistically significant. For discordant cases, the OR for each AI-assisted TPS method compared with the others was 0.99 to 1.01 and not statistically significant. This study emphasized the usefulness of the AI-assisted system for cases in which pathologists had difficulty determining the PD-L1 TPS.
Collapse
Affiliation(s)
- Hiroaki Ito
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akihiko Yoshizawa
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Nara Medical University, Nara, Japan.
| | - Kazuhiro Terada
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| | - Akiyoshi Nakakura
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | - Tatsuhiko Sugimoto
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Kazuya Nishimura
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Naoki Nakajima
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Toyooka Hospital, Hyogo, Japan
| | - Shinji Sumiyoshi
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan; Department of Diagnostic Pathology, Tenri Hospital, Nara, Japan
| | - Masatsugu Hamaji
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Toshi Menju
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Hiroshi Date
- Department of Thoracic Surgery, Kyoto University Hospital, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | - Ryoma Bise
- Department of Advanced Information Technology, Kyushu University, Fukuoka, Japan
| | - Hironori Haga
- Department of Diagnostic Pathology, Kyoto University Hospital, Kyoto, Japan
| |
Collapse
|
15
|
Kim H, Kim S, Choi S, Park C, Park S, Pereira S, Ma M, Yoo D, Paeng K, Jung W, Park S, Ock CY, Lee SH, Choi YL, Chung JH. Clinical Validation of Artificial Intelligence-Powered PD-L1 Tumor Proportion Score Interpretation for Immune Checkpoint Inhibitor Response Prediction in Non-Small Cell Lung Cancer. JCO Precis Oncol 2024; 8:e2300556. [PMID: 38723233 DOI: 10.1200/po.23.00556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2023] [Revised: 12/11/2023] [Accepted: 04/03/2024] [Indexed: 05/15/2024] Open
Abstract
PURPOSE Evaluation of PD-L1 tumor proportion score (TPS) by pathologists has been very impactful but is limited by factors such as intraobserver/interobserver bias and intratumor heterogeneity. We developed an artificial intelligence (AI)-powered analyzer to assess TPS for the prediction of immune checkpoint inhibitor (ICI) response in advanced non-small cell lung cancer (NSCLC). MATERIALS AND METHODS The AI analyzer was trained with 393,565 tumor cells annotated by board-certified pathologists for PD-L1 expression in 802 whole-slide images (WSIs) stained by 22C3 pharmDx immunohistochemistry. The clinical performance of the analyzer was validated in an external cohort of 430 WSIs from patients with NSCLC. Three pathologists performed annotations of this external cohort, and their consensus TPS was compared with AI-based TPS. RESULTS In comparing PD-L1 TPS assessed by AI analyzer and by pathologists, a significant positive correlation was observed (Spearman coefficient = 0.925; P < .001). The concordance of TPS between AI analyzer and pathologists according to TPS ≥50%, 1%-49%, and <1% was 85.7%, 89.3%, and 52.4%, respectively. In median progression-free survival (PFS), AI-based TPS predicted prognosis in the TPS 1%-49% or TPS <1% group better than the pathologist's reading, with the TPS ≥50% group as a reference (hazard ratio [HR], 1.49 [95% CI, 1.19 to 1.86] v HR, 1.36 [95% CI, 1.08 to 1.71] for TPS 1%-49% group, and HR, 2.38 [95% CI, 1.69 to 3.35] v HR, 1.62 [95% CI, 1.23 to 2.13] for TPS <1% group). CONCLUSION PD-L1 TPS assessed by AI analyzer correlates with that of pathologists, with clinical performance also being comparable when referenced to PFS. The AI model can accurately predict tumor response and PFS of ICI in advanced NSCLC via assessment of PD-L1 TPS.
Collapse
Affiliation(s)
- Hyojin Kim
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Sangjoon Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Changhee Park
- Department of Internal Medicine, Seoul National University Hospital, Seoul, Republic of Korea
| | | | | | - Minuk Ma
- Lunit Inc., Seoul, Republic of Korea
| | | | | | | | - Sehhoon Park
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | | | - Se-Hoon Lee
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Yoon-La Choi
- Department of Pathology and Translational Genomics, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Republic of Korea
| | - Jin-Haeng Chung
- Department of Pathology, Seoul National University Bundang Hospital, Seongnam, Republic of Korea
| |
Collapse
|
16
|
Ahmed AA, Fawi M, Brychcy A, Abouzid M, Witt M, Kaczmarek E. Development and Validation of a Deep Learning Model for Histopathological Slide Analysis in Lung Cancer Diagnosis. Cancers (Basel) 2024; 16:1506. [PMID: 38672588 PMCID: PMC11048051 DOI: 10.3390/cancers16081506] [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: 03/06/2024] [Revised: 04/04/2024] [Accepted: 04/12/2024] [Indexed: 04/28/2024] Open
Abstract
Lung cancer is the leading cause of cancer-related deaths worldwide. Two of the crucial factors contributing to these fatalities are delayed diagnosis and suboptimal prognosis. The rapid advancement of deep learning (DL) approaches provides a significant opportunity for medical imaging techniques to play a pivotal role in the early detection of lung tumors and subsequent monitoring during treatment. This study presents a DL-based model for efficient lung cancer detection using whole-slide images. Our methodology combines convolutional neural networks (CNNs) and separable CNNs with residual blocks, thereby improving classification performance. Our model improves accuracy (96% to 98%) and robustness in distinguishing between cancerous and non-cancerous lung cell images in less than 10 s. Moreover, the model's overall performance surpassed that of active pathologists, with an accuracy of 100% vs. 79%. There was a significant linear correlation between pathologists' accuracy and years of experience (r Pearson = 0.71, 95% CI 0.14 to 0.93, p = 0.022). We conclude that this model enhances the accuracy of cancer detection and can be used to train junior pathologists.
Collapse
Affiliation(s)
- Alhassan Ali Ahmed
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, 61-806 Poznan, Poland;
- Doctoral School, Poznan University of Medical Sciences, 61-806 Poznan, Poland;
| | - Muhammad Fawi
- Spider Silk Security DMCC, Dubai 282945, United Arab Emirates
| | - Agnieszka Brychcy
- Department of Clinical Patomorphology, Heliodor Swiecicki Clinical Hospital of the Poznan University of Medical Sciences, 61-806 Poznan, Poland
| | - Mohamed Abouzid
- Doctoral School, Poznan University of Medical Sciences, 61-806 Poznan, Poland;
- Department of Physical Pharmacy and Pharmacokinetics, Poznan University of Medical Sciences, 60-806 Poznan, Poland
| | - Martin Witt
- Department of Anatomy, Poznan University of Medical Sciences, 60-806 Poznan, Poland;
- Department of Anatomy, Technische Universität Dresden, 01307 Dresden, Germany
| | - Elżbieta Kaczmarek
- Department of Bioinformatics and Computational Biology, Poznan University of Medical Sciences, 61-806 Poznan, Poland;
| |
Collapse
|
17
|
van Eekelen L, Spronck J, Looijen-Salamon M, Vos S, Munari E, Girolami I, Eccher A, Acs B, Boyaci C, de Souza GS, Demirel-Andishmand M, Meesters LD, Zegers D, van der Woude L, Theelen W, van den Heuvel M, Grünberg K, van Ginneken B, van der Laak J, Ciompi F. Comparing deep learning and pathologist quantification of cell-level PD-L1 expression in non-small cell lung cancer whole-slide images. Sci Rep 2024; 14:7136. [PMID: 38531958 DOI: 10.1038/s41598-024-57067-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/14/2024] [Indexed: 03/28/2024] Open
Abstract
Programmed death-ligand 1 (PD-L1) expression is currently used in the clinic to assess eligibility for immune-checkpoint inhibitors via the tumor proportion score (TPS), but its efficacy is limited by high interobserver variability. Multiple papers have presented systems for the automatic quantification of TPS, but none report on the task of determining cell-level PD-L1 expression and often reserve their evaluation to a single PD-L1 monoclonal antibody or clinical center. In this paper, we report on a deep learning algorithm for detecting PD-L1 negative and positive tumor cells at a cellular level and evaluate it on a cell-level reference standard established by six readers on a multi-centric, multi PD-L1 assay dataset. This reference standard also provides for the first time a benchmark for computer vision algorithms. In addition, in line with other papers, we also evaluate our algorithm at slide-level by measuring the agreement between the algorithm and six pathologists on TPS quantification. We find a moderately low interobserver agreement at cell-level level (mean reader-reader F1 score = 0.68) which our algorithm sits slightly under (mean reader-AI F1 score = 0.55), especially for cases from the clinical center not included in the training set. Despite this, we find good AI-pathologist agreement on quantifying TPS compared to the interobserver agreement (mean reader-reader Cohen's kappa = 0.54, 95% CI 0.26-0.81, mean reader-AI kappa = 0.49, 95% CI 0.27-0.72). In conclusion, our deep learning algorithm demonstrates promise in detecting PD-L1 expression at a cellular level and exhibits favorable agreement with pathologists in quantifying the tumor proportion score (TPS). We publicly release our models for use via the Grand-Challenge platform.
Collapse
Affiliation(s)
- Leander van Eekelen
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands.
| | - Joey Spronck
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Monika Looijen-Salamon
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Shoko Vos
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Enrico Munari
- Pathology Unit, Department of Molecular and Translational Medicine, University of Brescia, Brescia, Italy
| | - Ilaria Girolami
- Department of Pathology, Provincial Hospital of Bolzano (SABES-ASDAA), Bolzano-Bozen, Italy
| | - Albino Eccher
- Department of Pathology and Diagnostics, University and Hospital Trust of Verona, Verona, Italy
| | - Balazs Acs
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Ceren Boyaci
- Department of Clinical Pathology and Cancer Diagnostics, Karolinska University Hospital, Stockholm, Sweden
| | - Gabriel Silva de Souza
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Muradije Demirel-Andishmand
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Luca Dulce Meesters
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Daan Zegers
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Lieke van der Woude
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Willemijn Theelen
- Department of Thoracic Oncology, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Michel van den Heuvel
- Respiratory Diseases Department, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Katrien Grünberg
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Bram van Ginneken
- Department of Radiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Jeroen van der Laak
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| | - Francesco Ciompi
- Department of Pathology, Radboud University Medical Center, P.O.Box 9101, 6500 HB, Nijmegen, The Netherlands
| |
Collapse
|
18
|
Kanan M, Alharbi H, Alotaibi N, Almasuood L, Aljoaid S, Alharbi T, Albraik L, Alothman W, Aljohani H, Alzahrani A, Alqahtani S, Kalantan R, Althomali R, Alameen M, Mufti A. AI-Driven Models for Diagnosing and Predicting Outcomes in Lung Cancer: A Systematic Review and Meta-Analysis. Cancers (Basel) 2024; 16:674. [PMID: 38339425 PMCID: PMC10854661 DOI: 10.3390/cancers16030674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/20/2024] [Accepted: 01/25/2024] [Indexed: 02/12/2024] Open
Abstract
(1) Background: Lung cancer's high mortality due to late diagnosis highlights a need for early detection strategies. Artificial intelligence (AI) in healthcare, particularly for lung cancer, offers promise by analyzing medical data for early identification and personalized treatment. This systematic review evaluates AI's performance in early lung cancer detection, analyzing its techniques, strengths, limitations, and comparative edge over traditional methods. (2) Methods: This systematic review and meta-analysis followed the PRISMA guidelines rigorously, outlining a comprehensive protocol and employing tailored search strategies across diverse databases. Two reviewers independently screened studies based on predefined criteria, ensuring the selection of high-quality data relevant to AI's role in lung cancer detection. The extraction of key study details and performance metrics, followed by quality assessment, facilitated a robust analysis using R software (Version 4.3.0). The process, depicted via a PRISMA flow diagram, allowed for the meticulous evaluation and synthesis of the findings in this review. (3) Results: From 1024 records, 39 studies met the inclusion criteria, showcasing diverse AI model applications for lung cancer detection, emphasizing varying strengths among the studies. These findings underscore AI's potential for early lung cancer diagnosis but highlight the need for standardization amidst study variations. The results demonstrate promising pooled sensitivity and specificity of 0.87, signifying AI's accuracy in identifying true positives and negatives, despite the observed heterogeneity attributed to diverse study parameters. (4) Conclusions: AI demonstrates promise in early lung cancer detection, showing high accuracy levels in this systematic review. However, study variations underline the need for standardized protocols to fully leverage AI's potential in revolutionizing early diagnosis, ultimately benefiting patients and healthcare professionals. As the field progresses, validated AI models from large-scale perspective studies will greatly benefit clinical practice and patient care in the future.
Collapse
Affiliation(s)
- Mohammed Kanan
- Department of Clinical Pharmacy, King Fahad Medical City, Riyadh 12211, Saudi Arabia
| | - Hajar Alharbi
- Department of Medicine, Gdansk Medical University, 80210 Gdansk, Poland
| | - Nawaf Alotaibi
- Department of Clinical Pharmacy, Northern Border University, Rafha 73213, Saudi Arabia
| | - Lubna Almasuood
- Department of Pharmacy, Qassim University, Buraydah 52571, Saudi Arabia
| | - Shahad Aljoaid
- Department of Medicine, University of Tabuk, Tabuk 47911, Saudi Arabia
| | - Tuqa Alharbi
- Department of Medicine, Qassim University, Buraydah 52571, Saudi Arabia
| | - Leen Albraik
- Department of Medicine, Al-Faisal University, Riyadh 12385, Saudi Arabia;
| | - Wojod Alothman
- Department of Medicine, Imam Abdulrahman Bin Faisal University, Dammam 31411, Saudi Arabia
| | - Hadeel Aljohani
- Department of Medicine and Surgery, King Abdulaziz University, Jeddah 22230, Saudi Arabia; (H.A.); (R.K.)
| | - Aghnar Alzahrani
- Department of Medicine, Al-Baha University, Al Bahah 65964, Saudi Arabia
| | - Sadeem Alqahtani
- Department of Pharmacy, King Khalid University, Abha 62217, Saudi Arabia
| | - Razan Kalantan
- Department of Medicine and Surgery, King Abdulaziz University, Jeddah 22230, Saudi Arabia; (H.A.); (R.K.)
| | - Raghad Althomali
- Department of Medicine, Taif University, Taif 26311, Saudi Arabia
| | - Maram Alameen
- Department of Medicine, Taif University, Taif 26311, Saudi Arabia
| | - Ahdab Mufti
- Department of Medicine, Ibn Sina National College, Jeddah 22230, Saudi Arabia
| |
Collapse
|
19
|
Ligero M, Serna G, El Nahhas OS, Sansano I, Mauchanski S, Viaplana C, Calderaro J, Toledo RA, Dienstmann R, Vanguri RS, Sauter JL, Sanchez-Vega F, Shah SP, Ramón y Cajal S, Garralda E, Nuciforo P, Perez-Lopez R, Kather JN. Weakly Supervised Deep Learning Predicts Immunotherapy Response in Solid Tumors Based on PD-L1 Expression. CANCER RESEARCH COMMUNICATIONS 2024; 4:92-102. [PMID: 38126740 PMCID: PMC10782919 DOI: 10.1158/2767-9764.crc-23-0287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/11/2023] [Accepted: 12/12/2023] [Indexed: 12/23/2023]
Abstract
Programmed death-ligand 1 (PD-L1) IHC is the most commonly used biomarker for immunotherapy response. However, quantification of PD-L1 status in pathology slides is challenging. Neither manual quantification nor a computer-based mimicking of manual readouts is perfectly reproducible, and the predictive performance of both approaches regarding immunotherapy response is limited. In this study, we developed a deep learning (DL) method to predict PD-L1 status directly from raw IHC image data, without explicit intermediary steps such as cell detection or pigment quantification. We trained the weakly supervised model on PD-L1-stained slides from the non-small cell lung cancer (NSCLC)-Memorial Sloan Kettering (MSK) cohort (N = 233) and validated it on the pan-cancer-Vall d'Hebron Institute of Oncology (VHIO) cohort (N = 108). We also investigated the performance of the model to predict response to immune checkpoint inhibitors (ICI) in terms of progression-free survival. In the pan-cancer-VHIO cohort, the performance was compared with tumor proportion score (TPS) and combined positive score (CPS). The DL model showed good performance in predicting PD-L1 expression (TPS ≥ 1%) in both NSCLC-MSK and pan-cancer-VHIO cohort (AUC 0.88 ± 0.06 and 0.80 ± 0.03, respectively). The predicted PD-L1 status showed an improved association with response to ICIs [HR: 1.5 (95% confidence interval: 1-2.3), P = 0.049] compared with TPS [HR: 1.4 (0.96-2.2), P = 0.082] and CPS [HR: 1.2 (0.79-1.9), P = 0.386]. Notably, our explainability analysis showed that the model does not just look at the amount of brown pigment in the IHC slides, but also considers morphologic factors such as lymphocyte conglomerates. Overall, end-to-end weakly supervised DL shows potential for improving patient stratification for cancer immunotherapy by analyzing PD-L1 IHC, holistically integrating morphology and PD-L1 staining intensity. SIGNIFICANCE The weakly supervised DL model to predict PD-L1 status from raw IHC data, integrating tumor staining intensity and morphology, enables enhanced patient stratification in cancer immunotherapy compared with traditional pathologist assessment.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Garazi Serna
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Omar S.M. El Nahhas
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
| | - Irene Sansano
- Pathology Department, Vall d'Hebron University Hospital (VHUH), Barcelona, Spain
| | - Siarhei Mauchanski
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Cristina Viaplana
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Julien Calderaro
- Assistance Publique-Hôpitaux de Paris, Département de Pathologie, CHU Henri Mondor, Créteil, France
- Université Paris-Est Créteil, Faculté de Médecine, Créteil, France
| | - Rodrigo A. Toledo
- Biomakers and Clonal Dynamics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rami S. Vanguri
- Department of Pathology and Laboratory Medicine, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania
| | - Jennifer L. Sauter
- Department of Pathology and Laboratory Medicine, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Sohrab P. Shah
- Computational Oncology, Memorial Sloan Kettering Cancer Center, New York, New York
| | | | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Jakob Nikolas Kather
- Else Kroener Fresenius Center for Digital Health, Technical University Dresden, Dresden, Germany
- Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom
- Department of Medicine I, University Hospital Dresden, Dresden, Germany
- Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Nibid L, Greco C, Cordelli E, Sabarese G, Fiore M, Liu CZ, Ippolito E, Sicilia R, Miele M, Tortora M, Taffon C, Rakaee M, Soda P, Ramella S, Perrone G. Deep pathomics: A new image-based tool for predicting response to treatment in stage III non-small cell lung cancer. PLoS One 2023; 18:e0294259. [PMID: 38015944 PMCID: PMC10684067 DOI: 10.1371/journal.pone.0294259] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2023] [Accepted: 10/26/2023] [Indexed: 11/30/2023] Open
Abstract
Despite the advantages offered by personalized treatments, there is presently no way to predict response to chemoradiotherapy in patients with non-small cell lung cancer (NSCLC). In this exploratory study, we investigated the application of deep learning techniques to histological tissue slides (deep pathomics), with the aim of predicting the response to therapy in stage III NSCLC. We evaluated 35 digitalized tissue slides (biopsies or surgical specimens) obtained from patients with stage IIIA or IIIB NSCLC. Patients were classified as responders (12/35, 34.7%) or non-responders (23/35, 65.7%) based on the target volume reduction shown on weekly CT scans performed during chemoradiation treatment. Digital tissue slides were tested by five pre-trained convolutional neural networks (CNNs)-AlexNet, VGG, MobileNet, GoogLeNet, and ResNet-using a leave-two patient-out cross validation approach, and we evaluated the networks' performances. GoogLeNet was globally found to be the best CNN, correctly classifying 8/12 responders and 10/11 non-responders. Moreover, Deep-Pathomics was found to be highly specific (TNr: 90.1) and quite sensitive (TPr: 0.75). Our data showed that AI could surpass the capabilities of all presently available diagnostic systems, supplying additional information beyond that currently obtainable in clinical practice. The ability to predict a patient's response to treatment could guide the development of new and more effective therapeutic AI-based approaches and could therefore be considered an effective and innovative step forward in personalised medicine.
Collapse
Affiliation(s)
- Lorenzo Nibid
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Carlo Greco
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Ermanno Cordelli
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Giovanna Sabarese
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Michele Fiore
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Charles Z. Liu
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Edy Ippolito
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Rosa Sicilia
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Marianna Miele
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Matteo Tortora
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
| | - Chiara Taffon
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Mehrdad Rakaee
- Department of Clinical Pathology, University Hospital of North Norway, Tromsø, Norway
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, Massachusetts, United States of America
| | - Paolo Soda
- Unit of Computer Systems and Bioinformatics, Department of Engineering, Università Campus Bio-Medico di Roma, Rome, Italy
- Department of Radiation Sciences, Radiation Physics, Biomedical Engineering, Umeå University, Umeå, Sweden
| | - Sara Ramella
- Radiation Oncology, Department of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Radiation Oncology, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| | - Giuseppe Perrone
- Research Unit of Anatomical Pathology, Department of of Medicine and Surgery, Università Campus Bio-Medico di Roma, Rome, Italy
- Anatomical Pathology Operative Research Unit, Fondazione Policlinico Universitario Campus Bio-Medico, Rome, Italy
| |
Collapse
|
21
|
Davri A, Birbas E, Kanavos T, Ntritsos G, Giannakeas N, Tzallas AT, Batistatou A. Deep Learning for Lung Cancer Diagnosis, Prognosis and Prediction Using Histological and Cytological Images: A Systematic Review. Cancers (Basel) 2023; 15:3981. [PMID: 37568797 PMCID: PMC10417369 DOI: 10.3390/cancers15153981] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/27/2023] [Accepted: 08/03/2023] [Indexed: 08/13/2023] Open
Abstract
Lung cancer is one of the deadliest cancers worldwide, with a high incidence rate, especially in tobacco smokers. Lung cancer accurate diagnosis is based on distinct histological patterns combined with molecular data for personalized treatment. Precise lung cancer classification from a single H&E slide can be challenging for a pathologist, requiring most of the time additional histochemical and special immunohistochemical stains for the final pathology report. According to WHO, small biopsy and cytology specimens are the available materials for about 70% of lung cancer patients with advanced-stage unresectable disease. Thus, the limited available diagnostic material necessitates its optimal management and processing for the completion of diagnosis and predictive testing according to the published guidelines. During the new era of Digital Pathology, Deep Learning offers the potential for lung cancer interpretation to assist pathologists' routine practice. Herein, we systematically review the current Artificial Intelligence-based approaches using histological and cytological images of lung cancer. Most of the published literature centered on the distinction between lung adenocarcinoma, lung squamous cell carcinoma, and small cell lung carcinoma, reflecting the realistic pathologist's routine. Furthermore, several studies developed algorithms for lung adenocarcinoma predominant architectural pattern determination, prognosis prediction, mutational status characterization, and PD-L1 expression status estimation.
Collapse
Affiliation(s)
- Athena Davri
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| | - Effrosyni Birbas
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Theofilos Kanavos
- Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece; (E.B.); (T.K.)
| | - Georgios Ntritsos
- Department of Hygiene and Epidemiology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45110 Ioannina, Greece;
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Nikolaos Giannakeas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Alexandros T. Tzallas
- Department of Informatics and Telecommunications, University of Ioannina, 47100 Arta, Greece;
| | - Anna Batistatou
- Department of Pathology, Faculty of Medicine, School of Health Sciences, University of Ioannina, 45500 Ioannina, Greece;
| |
Collapse
|
22
|
Wang R, Xiong K, Wang Z, Wu D, Hu B, Ruan J, Sun C, Ma D, Li L, Liao S. Immunodiagnosis - the promise of personalized immunotherapy. Front Immunol 2023; 14:1216901. [PMID: 37520576 PMCID: PMC10372420 DOI: 10.3389/fimmu.2023.1216901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Accepted: 06/21/2023] [Indexed: 08/01/2023] Open
Abstract
Immunotherapy showed remarkable efficacy in several cancer types. However, the majority of patients do not benefit from immunotherapy. Evaluating tumor heterogeneity and immune status before treatment is key to identifying patients that are more likely to respond to immunotherapy. Demographic characteristics (such as sex, age, and race), immune status, and specific biomarkers all contribute to response to immunotherapy. A comprehensive immunodiagnostic model integrating all these three dimensions by artificial intelligence would provide valuable information for predicting treatment response. Here, we coined the term "immunodiagnosis" to describe the blueprint of the immunodiagnostic model. We illustrated the features that should be included in immunodiagnostic model and the strategy of constructing the immunodiagnostic model. Lastly, we discussed the incorporation of this immunodiagnosis model in clinical practice in hopes of improving the prognosis of tumor immunotherapy.
Collapse
Affiliation(s)
- Renjie Wang
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Kairong Xiong
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhimin Wang
- Division of Endocrinology and Metabolic Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Di Wu
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Bai Hu
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jinghan Ruan
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Chaoyang Sun
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Ding Ma
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li Li
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shujie Liao
- Department of Obstetrics and Gynecology, Cancer Biology Research Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
23
|
Choi S, Kim S. Artificial Intelligence in the Pathology of Gastric Cancer. J Gastric Cancer 2023; 23:410-427. [PMID: 37553129 PMCID: PMC10412971 DOI: 10.5230/jgc.2023.23.e25] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 07/09/2023] [Accepted: 07/14/2023] [Indexed: 08/10/2023] Open
Abstract
Recent advances in artificial intelligence (AI) have provided novel tools for rapid and precise pathologic diagnosis. The introduction of digital pathology has enabled the acquisition of scanned slide images that are essential for the application of AI. The application of AI for improved pathologic diagnosis includes the error-free detection of potentially negligible lesions, such as a minute focus of metastatic tumor cells in lymph nodes, the accurate diagnosis of potentially controversial histologic findings, such as very well-differentiated carcinomas mimicking normal epithelial tissues, and the pathological subtyping of the cancers. Additionally, the utilization of AI algorithms enables the precise decision of the score of immunohistochemical markers for targeted therapies, such as human epidermal growth factor receptor 2 and programmed death-ligand 1. Studies have revealed that AI assistance can reduce the discordance of interpretation between pathologists and more accurately predict clinical outcomes. Several approaches have been employed to develop novel biomarkers from histologic images using AI. Moreover, AI-assisted analysis of the cancer microenvironment showed that the distribution of tumor-infiltrating lymphocytes was related to the response to the immune checkpoint inhibitor therapy, emphasizing its value as a biomarker. As numerous studies have demonstrated the significance of AI-assisted interpretation and biomarker development, the AI-based approach will advance diagnostic pathology.
Collapse
Affiliation(s)
- Sangjoon Choi
- Department of Pathology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Korea
| | - Seokhwi Kim
- Department of Pathology, Ajou University School of Medicine, Suwon, Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Korea.
| |
Collapse
|
24
|
Liu W, Huo G, Chen P. Efficacy of PD-1/PD-L1 inhibitors in advanced gastroesophageal cancer based on characteristics: a meta-analysis. Immunotherapy 2023. [PMID: 37190983 DOI: 10.2217/imt-2022-0305] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/17/2023] Open
Abstract
Objective: Evaluate the potency of anti-PD-1/PD-L1 antibodies in advanced gastroesophageal cancer patients with different clinical features. Methods: Randomized, controlled trials comparing anti-PD-1/PD-L1 antibodies with chemotherapy in individuals with gastroesophageal cancer were retrieved. Results: 15 trials involving 9194 individuals were included. PD-1/PD-L1 inhibitors significantly improved overall survival (OS) but not progression-free survival. Significantly improved OS was observed in PD-L1 combined positive score ≥1, primary esophageal cancer, primary gastric cancer and Asian patients. Subgroup analysis revealed significant OS benefit achieved for esophageal squamous cell carcinoma, but not for esophageal adenocarcinoma. Conclusion: PD-1/PD-L1 inhibitors improved OS in advanced gastroesophageal carcinoma, especially in patients with esophageal cancer. Race, primary tumor sites and PD-L1 combined positive score can be used to predict the potency of immune checkpoint inhibitors.
Collapse
Affiliation(s)
- Wenjie Liu
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| | - Gengwei Huo
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| | - Peng Chen
- Department of Thoracic Oncology, Tianjin Medical University Cancer Institute & Hospital; National Clinical Research Center for Cancer; Key Laboratory of Cancer Prevention & Therapy of Tianjin; Tianjin's Clinical Research Center for Cancer; Tianjin, 300060, China
| |
Collapse
|
25
|
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: 1.0] [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
|
26
|
Dacic S. State of the Art of Pathologic and Molecular Testing. Hematol Oncol Clin North Am 2023; 37:463-473. [PMID: 36964109 DOI: 10.1016/j.hoc.2023.02.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/26/2023]
Abstract
Advances in the treatment of non-small cell lung carcinoma have resulted in improved histologic classification and the implementation of molecular testing for predictive biomarkers into the routine diagnostic workflow. Over the past decade, molecular testing has evolved from single-gene assays to high-thoroughput comprehensive next-generation sequencing. Economic barriers, suboptimal turnaround time to obtain the results, and limited tissue available for molecular assays resulted in adoption of liquid biopsies (ctDNA) into clinical practice. Multiplex immunohistochemical/immunofluorescence assays evaluating tumor microenvironment together with the AI approaches are anticipated to translate from research into clinical care.
Collapse
Affiliation(s)
- Sanja Dacic
- Department of Pathology, Yale School of Medicine, 200 South Frontage Road, EP2-631, New Haven, CT 06510, USA.
| |
Collapse
|
27
|
Li T, Li Y, Zhu X, He Y, Wu Y, Ying T, Xie Z. Artificial intelligence in cancer immunotherapy: Applications in neoantigen recognition, antibody design and immunotherapy response prediction. Semin Cancer Biol 2023; 91:50-69. [PMID: 36870459 DOI: 10.1016/j.semcancer.2023.02.007] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2022] [Revised: 02/13/2023] [Accepted: 02/28/2023] [Indexed: 03/06/2023]
Abstract
Cancer immunotherapy is a method of controlling and eliminating tumors by reactivating the body's cancer-immunity cycle and restoring its antitumor immune response. The increased availability of data, combined with advancements in high-performance computing and innovative artificial intelligence (AI) technology, has resulted in a rise in the use of AI in oncology research. State-of-the-art AI models for functional classification and prediction in immunotherapy research are increasingly used to support laboratory-based experiments. This review offers a glimpse of the current AI applications in immunotherapy, including neoantigen recognition, antibody design, and prediction of immunotherapy response. Advancing in this direction will result in more robust predictive models for developing better targets, drugs, and treatments, and these advancements will eventually make their way into the clinical setting, pushing AI forward in the field of precision oncology.
Collapse
Affiliation(s)
- Tong Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yupeng Li
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Xiaoyi Zhu
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, China
| | - Yao He
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China
| | - Yanling Wu
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, China
| | - Tianlei Ying
- MOE/NHC Key Laboratory of Medical Molecular Virology, Shanghai Institute of Infectious Disease and Biosecurity, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai, China; Shanghai Engineering Research Center for Synthetic Immunology, Shanghai, China.
| | - Zhi Xie
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center, Sun Yat-sen University, Guangzhou, China; Center for Precision Medicine, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
28
|
Bidzińska J, Szurowska E. See Lung Cancer with an AI. Cancers (Basel) 2023; 15:1321. [PMID: 36831662 PMCID: PMC9954317 DOI: 10.3390/cancers15041321] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2023] [Revised: 02/13/2023] [Accepted: 02/17/2023] [Indexed: 02/22/2023] Open
Abstract
A lot has happened in the field of lung cancer screening in recent months. The ongoing discussion and documentation published by the scientific community and policymakers are of great importance to the entire European community and perhaps beyond. Lung cancer is the main worldwide killer. Low-dose computed tomography-based screening, together with smoking cessation, is the only tool to fight lung cancer, as it has already been proven in the United States of America but also European randomized controlled trials. Screening requires a lot of well-organized specialized work, but it can be supported by artificial intelligence (AI). Here we discuss whether and how to use AI for patients, radiologists, pulmonologists, thoracic surgeons, and all hospital staff supporting screening process benefits.
Collapse
Affiliation(s)
- Joanna Bidzińska
- Second Department of Radiology, Medical University of Gdansk, 80-210 Gdańsk, Poland
| | | |
Collapse
|
29
|
Ghaffari Laleh N, Ligero M, Perez-Lopez R, Kather JN. Facts and Hopes on the Use of Artificial Intelligence for Predictive Immunotherapy Biomarkers in Cancer. Clin Cancer Res 2023; 29:316-323. [PMID: 36083132 DOI: 10.1158/1078-0432.ccr-22-0390] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/26/2022] [Accepted: 08/29/2022] [Indexed: 01/19/2023]
Abstract
Immunotherapy by immune checkpoint inhibitors has become a standard treatment strategy for many types of solid tumors. However, the majority of patients with cancer will not respond, and predicting response to this therapy is still a challenge. Artificial intelligence (AI) methods can extract meaningful information from complex data, such as image data. In clinical routine, radiology or histopathology images are ubiquitously available. AI has been used to predict the response to immunotherapy from radiology or histopathology images, either directly or indirectly via surrogate markers. While none of these methods are currently used in clinical routine, academic and commercial developments are pointing toward potential clinical adoption in the near future. Here, we summarize the state of the art in AI-based image biomarkers for immunotherapy response based on radiology and histopathology images. We point out limitations, caveats, and pitfalls, including biases, generalizability, and explainability, which are relevant for researchers and health care providers alike, and outline key clinical use cases of this new class of predictive biomarkers.
Collapse
Affiliation(s)
| | - Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain.,Department of Radiology, Vall d'Hebron University Hospital, Barcelona, Spain
| | - Jakob Nikolas Kather
- Department of Medicine III, University Hospital RWTH Aachen, Aachen, Germany.,Division of Pathology and Data Analytics, Leeds Institute of Medical Research at St James's, University of Leeds, Leeds, United Kingdom.,Medical Oncology, National Center for Tumor Diseases (NCT), University Hospital Heidelberg, Heidelberg, Germany.,Else Kroener Fresenius Center for Digital Health, Medical Faculty Carl Gustav Carus, Technical University Dresden, Dresden, Germany
| |
Collapse
|
30
|
Artificial intelligence for prediction of response to cancer immunotherapy. Semin Cancer Biol 2022; 87:137-147. [PMID: 36372326 DOI: 10.1016/j.semcancer.2022.11.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 11/02/2022] [Accepted: 11/08/2022] [Indexed: 11/13/2022]
Abstract
Artificial intelligence (AI) indicates the application of machines to imitate intelligent behaviors for solving complex tasks with minimal human intervention, including machine learning and deep learning. The use of AI in medicine improves health-care systems in multiple areas such as diagnostic confirmation, risk stratification, analysis, prognosis prediction, treatment surveillance, and virtual health support, which has considerable potential to revolutionize and reshape medicine. In terms of immunotherapy, AI has been applied to unlock underlying immune signatures to associate with responses to immunotherapy indirectly as well as predict responses to immunotherapy responses directly. The AI-based analysis of high-throughput sequences and medical images can provide useful information for management of cancer immunotherapy considering the excellent abilities in selecting appropriate subjects, improving therapeutic regimens, and predicting individualized prognosis. In present review, we aim to evaluate a broad framework about AI-based computational approaches for prediction of response to cancer immunotherapy on both indirect and direct manners. Furthermore, we summarize our perspectives about challenges and opportunities of further AI applications on cancer immunotherapy relating to clinical practicability.
Collapse
|
31
|
McDonald OG, Montgomery EA. Artificial intelligence for dysplasia grading in Barrett's esophagus: hematoxylin and eosin is here to stay. Gastrointest Endosc 2022; 96:926-928. [PMID: 36253189 PMCID: PMC10594988 DOI: 10.1016/j.gie.2022.08.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Accepted: 08/07/2022] [Indexed: 02/08/2023]
Affiliation(s)
- Oliver G McDonald
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| | - Elizabeth A Montgomery
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, Florida, USA
| |
Collapse
|
32
|
Zheng S, Liang Y, Tan Y, Li L, Liu Q, Liu T, Lu X. Small Tweaks, Major Changes: Post-Translational Modifications That Occur within M2 Macrophages in the Tumor Microenvironment. Cancers (Basel) 2022; 14:5532. [PMID: 36428622 PMCID: PMC9688270 DOI: 10.3390/cancers14225532] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Revised: 10/21/2022] [Accepted: 11/07/2022] [Indexed: 11/12/2022] Open
Abstract
The majority of proteins are subjected to post-translational modifications (PTMs), regardless of whether they occur in or after biosynthesis of the protein. Capable of altering the physical and chemical properties and functions of proteins, PTMs are thus crucial. By fostering the proliferation, migration, and invasion of cancer cells with which they communicate in the tumor microenvironment (TME), M2 macrophages have emerged as key cellular players in the TME. Furthermore, growing evidence illustrates that PTMs can occur in M2 macrophages as well, possibly participating in molding the multifaceted characteristics and physiological behaviors in the TME. Hence, there is a need to review the PTMs that have been reported to occur within M2 macrophages. Although there are several reviews available regarding the roles of M2 macrophages, the majority of these reviews overlooked PTMs occurring within M2 macrophages. Considering this, in this review, we provide a review focusing on the advancement of PTMs that have been reported to take place within M2 macrophages, mainly in the TME, to better understand the performance of M2 macrophages in the tumor microenvironment. Incidentally, we also briefly cover the advances in developing inhibitors that target PTMs and the application of artificial intelligence (AI) in the prediction and analysis of PTMs at the end of the review.
Collapse
Affiliation(s)
- Shutao Zheng
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Yan Liang
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Yiyi Tan
- Department of Pathology, Basic Medicine College, Xinjiang Medical University, Urumqi 830017, China
| | - Lu Li
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Qing Liu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Tao Liu
- Department of Clinical Laboratory, First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| | - Xiaomei Lu
- State Key Laboratory of Pathogenesis, Prevention and Treatment of High Incidence Diseases in Central Asia, Clinical Medical Research Institute, The First Affiliated Hospital of Xinjiang Medical University, Urumqi 830054, China
| |
Collapse
|
33
|
Abbasian MH, Ardekani AM, Sobhani N, Roudi R. The Role of Genomics and Proteomics in Lung Cancer Early Detection and Treatment. Cancers (Basel) 2022; 14:5144. [PMID: 36291929 PMCID: PMC9600051 DOI: 10.3390/cancers14205144] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Revised: 10/10/2022] [Accepted: 10/18/2022] [Indexed: 08/17/2023] Open
Abstract
Lung cancer is the leading cause of cancer-related death worldwide, with non-small-cell lung cancer (NSCLC) being the primary type. Unfortunately, it is often diagnosed at advanced stages, when therapy leaves patients with a dismal prognosis. Despite the advances in genomics and proteomics in the past decade, leading to progress in developing tools for early diagnosis, targeted therapies have shown promising results; however, the 5-year survival of NSCLC patients is only about 15%. Low-dose computed tomography or chest X-ray are the main types of screening tools. Lung cancer patients without specific, actionable mutations are currently treated with conventional therapies, such as platinum-based chemotherapy; however, resistances and relapses often occur in these patients. More noninvasive, inexpensive, and safer diagnostic methods based on novel biomarkers for NSCLC are of paramount importance. In the current review, we summarize genomic and proteomic biomarkers utilized for the early detection and treatment of NSCLC. We further discuss future opportunities to improve biomarkers for early detection and the effective treatment of NSCLC.
Collapse
Affiliation(s)
- Mohammad Hadi Abbasian
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Ali M. Ardekani
- Department of Medical Biotechnology, National Institute of Genetic Engineering and Biotechnology (NIGEB), Tehran 1497716316, Iran
| | - Navid Sobhani
- Department of Medicine, Section of Epidemiology and Population Sciences, Baylor College of Medicine, Houston, TX 77030, USA
| | - Raheleh Roudi
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
34
|
Zhao X, Bao Y, Meng B, Xu Z, Li S, Wang X, Hou R, Ma W, Liu D, Zheng J, Shi M. From rough to precise: PD-L1 evaluation for predicting the efficacy of PD-1/PD-L1 blockades. Front Immunol 2022; 13:920021. [PMID: 35990664 PMCID: PMC9382880 DOI: 10.3389/fimmu.2022.920021] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 07/04/2022] [Indexed: 11/13/2022] Open
Abstract
Developing biomarkers for accurately predicting the efficacy of immune checkpoint inhibitor (ICI) therapies is conducive to avoiding unwanted side effects and economic burden. At the moment, the quantification of programmed cell death ligand 1 (PD-L1) in tumor tissues is clinically used as one of the combined diagnostic assays of response to anti-PD-1/PD-L1 therapy. However, the current assays for evaluating PD-L1 remain imperfect. Recent studies are promoting the methodologies of PD-L1 evaluation from rough to precise. Standardization of PD-L1 immunohistochemistry tests is being promoted by using optimized reagents, platforms, and cutoff values. Combining novel in vivo probes with PET or SPECT will probably be of benefit to map the spatio-temporal heterogeneity of PD-L1 expression. The dynamic change of PD-L1 in the circulatory system can also be realized by liquid biopsy. Consider PD-L1 expressed on non-tumor (immune and non-immune) cells, and optimized combination detection indexes are further improving the accuracy of PD-L1 in predicting the efficacy of ICIs. The combinations of artificial intelligence with novel technologies are conducive to the intelligence of PD-L1 as a predictive biomarker. In this review, we will provide an overview of the recent progress in this rapidly growing area and discuss the clinical and technical challenges.
Collapse
Affiliation(s)
- Xuan Zhao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Yulin Bao
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Bi Meng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Zijian Xu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Sijin Li
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Xu Wang
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Rui Hou
- College of Pharmacy, Xuzhou Medical University, Xuzhou, China
| | - Wen Ma
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
| | - Dan Liu
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| | - Junnian Zheng
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| | - Ming Shi
- Jiangsu Center for the Collaboration and Innovation of Cancer Biotherapy, Cancer Institute, Xuzhou Medical University, Xuzhou, China
- *Correspondence: Dan Liu, ; Junnian Zheng, ; Ming Shi,
| |
Collapse
|
35
|
Cheng G, Zhang F, Xing Y, Hu X, Zhang H, Chen S, Li M, Peng C, Ding G, Zhang D, Chen P, Xia Q, Wu M. Artificial Intelligence-Assisted Score Analysis for Predicting the Expression of the Immunotherapy Biomarker PD-L1 in Lung Cancer. Front Immunol 2022; 13:893198. [PMID: 35844508 PMCID: PMC9286729 DOI: 10.3389/fimmu.2022.893198] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Accepted: 05/27/2022] [Indexed: 12/12/2022] Open
Abstract
Programmed cell death ligand 1 (PD-L1) is a critical biomarker for predicting the response to immunotherapy. However, traditional quantitative evaluation of PD-L1 expression using immunohistochemistry staining remains challenging for pathologists. Here we developed a deep learning (DL)-based artificial intelligence (AI) model to automatically analyze the immunohistochemical expression of PD-L1 in lung cancer patients. A total of 1,288 patients with lung cancer were included in the study. The diagnostic ability of three different AI models (M1, M2, and M3) was assessed in both PD-L1 (22C3) and PD-L1 (SP263) assays. M2 and M3 showed improved performance in the evaluation of PD-L1 expression in the PD-L1 (22C3) assay, especially at 1% cutoff. Highly accurate performance in the PD-L1 (SP263) was also achieved, with accuracy and specificity of 96.4 and 96.8% in both M2 and M3, respectively. Moreover, the diagnostic results of these three AI-assisted models were highly consistent with those from the pathologist. Similar performances of M1, M2, and M3 in the 22C3 dataset were also obtained in lung adenocarcinoma and lung squamous cell carcinoma in both sampling methods. In conclusion, these results suggest that AI-assisted diagnostic models in PD-L1 expression are a promising tool for improving the efficiency of clinical pathologists.
Collapse
Affiliation(s)
- Guoping Cheng
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | | | | | - Xingyi Hu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
- The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, China
| | - He Zhang
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
| | | | | | | | - Guangtai Ding
- School of Computer Engineering and Science, Shanghai University, Shanghai, China
| | - Dadong Zhang
- 3D Medicines Inc., Shanghai, China
- *Correspondence: Dadong Zhang, ; Peilin Chen, ; Qingxin Xia, ; Meijuan Wu,
| | - Peilin Chen
- 3D Medicines Inc., Shanghai, China
- *Correspondence: Dadong Zhang, ; Peilin Chen, ; Qingxin Xia, ; Meijuan Wu,
| | - Qingxin Xia
- Department of Pathology, Affiliated Cancer Hospital of Zhengzhou University, Zhengzhou, China
- *Correspondence: Dadong Zhang, ; Peilin Chen, ; Qingxin Xia, ; Meijuan Wu,
| | - Meijuan Wu
- Department of Pathology, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
- Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
- *Correspondence: Dadong Zhang, ; Peilin Chen, ; Qingxin Xia, ; Meijuan Wu,
| |
Collapse
|
36
|
Viswanathan VS, Toro P, Corredor G, Mukhopadhyay S, Madabhushi A. The state of the art for artificial intelligence in lung digital pathology. J Pathol 2022; 257:413-429. [PMID: 35579955 PMCID: PMC9254900 DOI: 10.1002/path.5966] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 04/26/2022] [Accepted: 05/15/2022] [Indexed: 12/03/2022]
Abstract
Lung diseases carry a significant burden of morbidity and mortality worldwide. The advent of digital pathology (DP) and an increase in computational power have led to the development of artificial intelligence (AI)-based tools that can assist pathologists and pulmonologists in improving clinical workflow and patient management. While previous works have explored the advances in computational approaches for breast, prostate, and head and neck cancers, there has been a growing interest in applying these technologies to lung diseases as well. The application of AI tools on radiology images for better characterization of indeterminate lung nodules, fibrotic lung disease, and lung cancer risk stratification has been well documented. In this article, we discuss methodologies used to build AI tools in lung DP, describing the various hand-crafted and deep learning-based unsupervised feature approaches. Next, we review AI tools across a wide spectrum of lung diseases including cancer, tuberculosis, idiopathic pulmonary fibrosis, and COVID-19. We discuss the utility of novel imaging biomarkers for different types of clinical problems including quantification of biomarkers like PD-L1, lung disease diagnosis, risk stratification, and prediction of response to treatments such as immune checkpoint inhibitors. We also look briefly at some emerging applications of AI tools in lung DP such as multimodal data analysis, 3D pathology, and transplant rejection. Lastly, we discuss the future of DP-based AI tools, describing the challenges with regulatory approval, developing reimbursement models, planning clinical deployment, and addressing AI biases. © 2022 The Authors. The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
| | - Paula Toro
- Department of PathologyCleveland ClinicClevelandOHUSA
| | - Germán Corredor
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| | | | - Anant Madabhushi
- Department of Biomedical EngineeringCase Western Reserve UniversityClevelandOHUSA
- Louis Stokes Cleveland VA Medical CenterClevelandOHUSA
| |
Collapse
|
37
|
Wang C, Ma J, Shao J, Zhang S, Li J, Yan J, Zhao Z, Bai C, Yu Y, Li W. Non-Invasive Measurement Using Deep Learning Algorithm Based on Multi-Source Features Fusion to Predict PD-L1 Expression and Survival in NSCLC. Front Immunol 2022; 13:828560. [PMID: 35464416 PMCID: PMC9022118 DOI: 10.3389/fimmu.2022.828560] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2021] [Accepted: 02/17/2022] [Indexed: 02/05/2023] Open
Abstract
Background Programmed death-ligand 1 (PD-L1) assessment of lung cancer in immunohistochemical assays was only approved diagnostic biomarker for immunotherapy. But the tumor proportion score (TPS) of PD-L1 was challenging owing to invasive sampling and intertumoral heterogeneity. There was a strong demand for the development of an artificial intelligence (AI) system to measure PD-L1 expression signature (ES) non-invasively. Methods We developed an AI system using deep learning (DL), radiomics and combination models based on computed tomography (CT) images of 1,135 non-small cell lung cancer (NSCLC) patients with PD-L1 status. The deep learning feature was obtained through a 3D ResNet as the feature map extractor and the specialized classifier was constructed for the prediction and evaluation tasks. Then, a Cox proportional-hazards model combined with clinical factors and PD-L1 ES was utilized to evaluate prognosis in survival cohort. Results The combination model achieved a robust high-performance with area under the receiver operating characteristic curves (AUCs) of 0.950 (95% CI, 0.938-0.960), 0.934 (95% CI, 0.906-0.964), and 0.946 (95% CI, 0.933-0.958), for predicting PD-L1ES <1%, 1-49%, and ≥50% in validation cohort, respectively. Additionally, when combination model was trained on multi-source features the performance of overall survival evaluation (C-index: 0.89) could be superior compared to these of the clinical model alone (C-index: 0.86). Conclusion A non-invasive measurement using deep learning was proposed to access PD-L1 expression and survival outcomes of NSCLC. This study also indicated that deep learning model combined with clinical characteristics improved prediction capabilities, which would assist physicians in making rapid decision on clinical treatment options.
Collapse
Affiliation(s)
- Chengdi Wang
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Jiechao Ma
- AI Lab, Deepwise Healthcare, Beijing, China
| | - Jun Shao
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Shu Zhang
- AI Lab, Deepwise Healthcare, Beijing, China
| | - Jingwei Li
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | | | - Zhehao Zhao
- West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| | - Congchen Bai
- Department of Medical Informatics, West China Hospital, Sichuan University, Chengdu, China
| | - Yizhou Yu
- AI Lab, Deepwise Healthcare, Beijing, China
- Department of Computer Science, The University of Hong Kong, Pokfulam, Hong Kong SAR, China
| | - Weimin Li
- Department of Respiratory and Critical Care Medicine, Med-X Center for Manufacturing, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, West China School of Medicine, Sichuan University, Chengdu, China
| |
Collapse
|
38
|
Wu J, Lin D. A Review of Artificial Intelligence in Precise Assessment of Programmed Cell Death-ligand 1 and Tumor-infiltrating Lymphocytes in Non-Small Cell Lung Cancer. Adv Anat Pathol 2021; 28:439-445. [PMID: 34623343 DOI: 10.1097/pap.0000000000000322] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Advances in immunotherapy have increased the need for stratified predictive biomarkers in patients with non-small cell lung cancer. However, precise evaluation of tumor tissue-based immune biomarkers, such as programmed cell death-ligand 1 (PD-L1) and the characteristics of tumor infiltrating lymphocytes (TILs), is a challenge in clinical practice. In recent years, the digitization of whole-slide images of tissue has accelerated the implementation of artificial intelligence (AI) approaches in tumor pathology and provided an opportunity to use AI tools to improve the interpretation of immune biomarkers. This review describes the current challenges in the assessment of PD-L1 scoring and TILs and demonstrates the role of AI in helping pathologists integrate PD-L1 and biomarkers of the tumor immune microenvironment. Computer-aided PD-L1 scoring is highly consistent with pathologists and reduces the variation among interobservers, providing a promising diagnostic tool in pathology clinics. In addition, applications of image analysis algorithms, in combination with multiplex staining, enable in-depth quantitative and spatial analysis of the broader tumor microenvironment. Upon combining digital pathology and AI, an automatic analysis system of PD-L1 and TILs, which was established using a set of digital staining images and deep learning algorithms, might be an effective way to overcome the challenges in the precise assessment of immune biomarkers.
Collapse
Affiliation(s)
- Jianghua Wu
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital
- National Clinical Research Center of Cancer
- Key Laboratory of Cancer Prevention and Therapy
- Tianjin's Clinical Research Center of Cancer, Tianjin
| | - Dongmei Lin
- Department of Pathology, Peking University Cancer Hospital and Institute
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Beijing, China
| |
Collapse
|