1
|
Gou M, Zhang H, Qian N, Zhang Y, Sun Z, Li G, Wang Z, Dai G. Deep learning radiomics analysis for prediction of survival in patients with unresectable gastric cancer receiving immunotherapy. Eur J Radiol Open 2025; 14:100626. [PMID: 39807092 PMCID: PMC11728962 DOI: 10.1016/j.ejro.2024.100626] [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: 10/28/2024] [Revised: 12/03/2024] [Accepted: 12/14/2024] [Indexed: 01/16/2025] Open
Abstract
Objective Immunotherapy has become an option for the first-line therapy of advanced gastric cancer (GC), with improved survival. Our study aimed to investigate unresectable GC from an imaging perspective combined with clinicopathological variables to identify patients who were most likely to benefit from immunotherapy. Method Patients with unresectable GC who were consecutively treated with immunotherapy at two different medical centers of Chinese PLA General Hospital were included and divided into the training and validation cohorts, respectively. A deep learning neural network, using a multimodal ensemble approach based on CT imaging data before immunotherapy, was trained in the training cohort to predict survival, and an internal validation cohort was constructed to select the optimal ensemble model. Data from another cohort were used for external validation. The area under the receiver operating characteristic curve was analyzed to evaluate performance in predicting survival. Detailed clinicopathological data and peripheral blood prior to immunotherapy were collected for each patient. Univariate and multivariable logistic regression analysis of imaging models and clinicopathological variables was also applied to identify the independent predictors of survival. A nomogram based on multivariable logistic regression was constructed. Result A total of 79 GC patients in the training cohort and 97 patients in the external validation cohort were enrolled in this study. A multi-model ensemble approach was applied to train a model to predict the 1-year survival of GC patients. Compared to individual models, the ensemble model showed improvement in performance metrics in both the internal and external validation cohorts. There was a significant difference in overall survival (OS) among patients with different imaging models based on the optimum cutoff score of 0.5 (HR = 0.20, 95 % CI: 0.10-0.37, P < 0.001). Multivariate Cox regression analysis revealed that the imaging models, PD-L1 expression, and lung immune prognostic index were independent prognostic factors for OS. We combined these variables and built a nomogram. The calibration curves showed that the C-index of the nomogram was 0.85 and 0.78 in the training and validation cohorts. Conclusion The deep learning model in combination with several clinical factors showed predictive value for survival in patients with unresectable GC receiving immunotherapy.
Collapse
Affiliation(s)
- Miaomiao Gou
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Hongtao Zhang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Niansong Qian
- Department of Thoracic Oncology, The Eighth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Yong Zhang
- Department of Medical Oncology, The Second Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Zeyu Sun
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Guang Li
- R&D Center, Keya Medical Technology Co., Ltd, Beijing, PR China
| | - Zhikuan Wang
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| | - Guanghai Dai
- Department of Medical Oncology, The Fifth Medical Center, Chinese People’s Liberation Army General Hospital, Beijing, PR China
| |
Collapse
|
2
|
Gundavda K, Rajasimman AS, Patkar S, Chhatrala R, Baheti AD, Haria P, Kolhe M, Bhandare M, Chaudhari V, Shrikhande SV. Correlation between Tomographic and Histopathological Staging in Upfront Resected Gastric Cancer: Enhancing Diagnostic Accuracy in the Era of Perioperative Therapy. J Gastrointest Cancer 2025; 56:123. [PMID: 40425902 PMCID: PMC12116807 DOI: 10.1007/s12029-025-01245-5] [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] [Accepted: 05/05/2025] [Indexed: 05/29/2025]
Abstract
PURPOSE This study aimed to assess the diagnostic accuracy of multidetector contrast-enhanced computerised tomography (MDCT) and to establish a correlation between radiological and histopathological staging in upfront resected localised gastric cancers (GC). METHODS All consecutive patients of resectable, localised GC who underwent upfront elective resection between 2010 and 2022 were included. The initial clinical staging determined during multidisciplinary meetings was compared with the pathological stage obtained after surgery. Subsequently, a retrospective, blinded review was conducted to assign a revised clinical staging, and accuracy was correlated. RESULTS The analysis of 138 patients revealed varying accuracy of MDCT in determining the T stage (66.9% for T1/T2, 64.6% for T3, and 87.2% for T4) and N stage (60.8% for N0, 63.7% for N1, and 83.2% for N2). The accuracy for stage group ranged from 71 to 78.65%. There was weak agreement observed between the T, N, and overall stage on clinicopathological correlation. However, a blinded radiology review by oncoradiologists resulted in improved accuracy, particularly in T1/T2 disease, and also improved pathological stage correlation. CONCLUSIONS Although MDCT is a valuable initial staging tool for gastric cancer, we found weak agreement between the clinical and the pathological stages in upfront resected gastric cancers. By implementing an expert radiology review and standardising scanning and reporting protocols, we can significantly improve the accuracy and correlation of MDCT with pathology, even for T1/T2 disease. This may help in better selecting patients for upfront surgery versus perioperative chemotherapy, especially in resource-constrained settings.
Collapse
Affiliation(s)
- Kaival Gundavda
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Aishvarya Shri Rajasimman
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Shraddha Patkar
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India.
| | - Renish Chhatrala
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Akshay D Baheti
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Purvi Haria
- Department of Radiodiagnosis, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Manjushree Kolhe
- Department of Biostatistics, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Manish Bhandare
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Vikram Chaudhari
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| | - Shailesh V Shrikhande
- Department of Gastrointestinal and Hepatobiliary-Pancreatic Surgery, Department of Surgical Oncology, Tata Memorial Hospital, Homi Bhabha National Institute (HBNI), Mumbai, Maharashtra, India
| |
Collapse
|
3
|
Zhong L, Shi L, Li W, Zhou L, Wang K, Gu L. An Ultrasound Image-Based Deep Learning Radiomics Nomogram for Differentiating Between Benign and Malignant Indeterminate Cytology (Bethesda III) Thyroid Nodules: A Retrospective Study. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025. [PMID: 40396203 DOI: 10.1002/jcu.24058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 10/29/2024] [Accepted: 03/31/2025] [Indexed: 05/22/2025]
Abstract
RATIONALE AND OBJECTIVES Our objective is to develop and validate a deep learning radiomics nomogram (DLRN) based on preoperative ultrasound images and clinical features, for predicting the malignancy of thyroid nodules with indeterminate cytology (Bethesda III). MATERIALS AND METHODS Between June 2017 and June 2022, we conducted a retrospective study on 194 patients with surgically confirmed indeterminate cytology (Bethesda III) in our hospital. The training and internal validation cohorts were comprised of 155 and 39 patients, in a 7:3 ratio. To facilitate external validation, we selected an additional 80 patients from each of the remaining two medical centers. Utilizing preoperative ultrasound data, we obtained imaging markers that encompass both deep learning and manually radiomic features. After feature selection, we developed a comprehensive diagnostic model to evaluate the predictive value for Bethesda III benign and malignant cases. The model's diagnostic accuracy, calibration, and clinical applicability were systematically assessed. RESULTS The results showed that the prediction model, which integrated 512 DTL features extracted from the pre-trained Resnet34 network, ultrasound radiomics, and clinical features, exhibited superior stability in distinguishing between benign and malignant indeterminate thyroid nodules (Bethesda Class III). In the validation set, the AUC was 0.92 (95% CI: 0.831-1.000), and the accuracy, sensitivity, specificity, precision, and recall were 0.897, 0.882, 0.909, 0.882, and 0.882, respectively. CONCLUSION The comprehensive multidimensional data model based on deep transfer learning, ultrasound radiomics features, and clinical characteristics can effectively distinguish the benign and malignant indeterminate thyroid nodules (Bethesda Class III), providing valuable guidance for treatment selection in patients with indeterminate thyroid nodules (Bethesda Class III).
Collapse
Affiliation(s)
- Lichang Zhong
- Department of Ultrasound in Medicine, Sixth People's Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Lin Shi
- Department of Ultrasound in Medicine, Sixth People's Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Weimei Li
- Department of Ultrasound in Medicine, Sixth People's Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| | - Liang Zhou
- Department of Information, Sixth People's Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai, China
| | - Kui Wang
- The Affiliated Jiangsu Shengze Hospital of Nanjing Medical University, Suzhou, China
| | - Liping Gu
- Department of Ultrasound in Medicine, Sixth People's Hospital Affiliated to Medical College of Shanghai Jiaotong University, Shanghai Institute of Ultrasound in Medicine, Shanghai, China
| |
Collapse
|
4
|
Xu P, Yu H, Xing W, Zhang S, Hu H, Li W, Jia D, Zhi S, Peng X. Development and validation of a predictive model combining radiomics and deep learning features for spread through air spaces in stage T1 non-small cell lung cancer: a multicenter study. Front Oncol 2025; 15:1572720. [PMID: 40406248 PMCID: PMC12094994 DOI: 10.3389/fonc.2025.1572720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Purpose The goal of this paper is to compare the effectiveness of three deep learning models (2D, 3D, and 2.5D), three radiomics models(INTRA, Peri2mm, and Fusion2mm), and a combined model in predicting the spread through air spaces (STAS) in non-small cell lung cancer (NSCLC) to identify the optimal model for clinical surgery planning. Methods We included 480 patients who underwent surgery at four centers between January 2019 and August 2024, dividing them into a training cohort, an internal test cohort, and an external validation cohort. We extracted deep learning features using the ResNet50 algorithm. Least absolute shrinkage selection operator(Lasso) and spearman rank correlation were utilized to choose features. Extreme Gradient Boosting (XGboost) was used to execute deep learning and radiomics. Then, a combination model was developed, integrating both sources of data. Result The combined model showed outstanding performance, with an area under the receiver operating characteristic curve (AUC) of 0.927 (95% CI 0.870 - 0.984) in the test set and 0.867 (95% CI 0.819 - 0.915) in the validation set. This model significantly distinguished between high-risk and low-risk patients and demonstrated significant advantages in clinical application. Conclusion The combined model is adequate for preoperative prediction of STAS in patients with stage T1 NSCLC, outperforming the other six models in predicting STAS risk.
Collapse
Affiliation(s)
- Pengliang Xu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Huanming Yu
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Wenjian Xing
- Department of Radiology, Linghu Hospital, Second Medical Group of Nanxun District, Huzhou, China
| | - Shiyu Zhang
- Department of Radiology, Xishan People’s Hospital of Wuxi, Wuxi, China
| | - Haihua Hu
- Department of Radiology, Zhebei Mingzhou Hospital of Huzhou, Huzhou, China
| | - Wenhui Li
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Dan Jia
- Department of Respiratory Medicine, The First People’s Hospital of Huzhou, Huzhou, China
| | - Shengxu Zhi
- Department of Thoracic Surgery, The First People’s Hospital of Huzhou, Huzhou, China
| | - Xiuhua Peng
- Department of Radiology, The First People’s Hospital of Huzhou, Huzhou, China
| |
Collapse
|
5
|
Liang X, Ke X, Hu W, Jiang J, Li S, Xue C, Liu X, Dend J, Yan C, Gao M, Zhao L, Zhou J. Deep learning radiomic nomogram outperforms the clinical model in distinguishing intracranial solitary fibrous tumors from angiomatous meningiomas and can predict patient prognosis. Eur Radiol 2025; 35:2670-2680. [PMID: 39412667 DOI: 10.1007/s00330-024-11082-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Revised: 07/31/2024] [Accepted: 08/11/2024] [Indexed: 04/25/2025]
Abstract
OBJECTIVES To evaluate the value of a magnetic resonance imaging (MRI)-based deep learning radiomic nomogram (DLRN) for distinguishing intracranial solitary fibrous tumors (ISFTs) from angiomatous meningioma (AMs) and predicting overall survival (OS) for ISFT patients. METHODS In total, 1090 patients from Beijing Tiantan Hospital, Capital Medical University, and 131 from Lanzhou University Second Hospital were categorized as primary cohort (PC) and external validation cohort (EVC), respectively. An MRI-based DLRN was developed in PC to distinguish ISFTs from AMs. We validated the DLRN and compared it with a clinical model (CM) in EVC. In total, 149 ISFT patients were followed up. We carried out Cox regression analysis on DLRN score, clinical characteristics, and histological stratification. Besides, we evaluated the association between independent risk factors and OS in the follow-up patients using Kaplan-Meier curves. RESULTS The DLRN outperformed CM in distinguishing ISFTs from AMs (area under the curve [95% confidence interval (CI)]: 0.86 [0.84-0.88] for DLRN and 0.70 [0.67-0.72] for CM, p < 0.001) in EVC. Patients with high DLRN score [per 1 increase; hazard ratio (HR) 1.079, 95% CI: 1.009-1.147, p = 0.019] and subtotal resection (STR) [per 1 increase; HR 2.573, 95% CI: 1.337-4.932, p = 0.004] were associated with a shorter OS. A statistically significant difference in OS existed between the high and low DLRN score groups with a cutoff value of 12.19 (p < 0.001). There is also a difference in OS between total excision (GTR) and STR groups (p < 0.001). CONCLUSION The proposed DLRN outperforms the CM in distinguishing ISFTs from AMs and can predict OS for ISFT patients. CLINICAL RELEVANCE STATEMENT The proposed MRI-based deep learning radiomic nomogram outperforms the clinical model in distinguishing ISFTs from AMs and can predict OS of ISFT patients, which could guide the surgical strategy and predict prognosis for patients. KEY POINTS Distinguishing ISFTs from AMs based on conventional radiological signs is challenging. The DLRN outperformed the CM in our study. The DLRN can predict OS for ISFT patients.
Collapse
Affiliation(s)
- Xiaohong Liang
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Xiaoai Ke
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Wanjun Hu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Jian Jiang
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Shenglin Li
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Caiqiang Xue
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Xianwang Liu
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Juan Dend
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China
| | - Cheng Yan
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Mingzi Gao
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Liqin Zhao
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China.
| | - Junlin Zhou
- Department of Radiology, Lanzhou University Second Hospital, Lanzhou, China.
- Key Laboratory of Medical Imaging of Gansu Province, Lanzhou University Second Hospital, Lanzhou, China.
- Gansu International Scientific and Technological Cooperation Base of Medical Imaging Artificial Intelligence, Lanzhou, China.
| |
Collapse
|
6
|
Zhao T, Zhang X, Liu X, Wang Q, Hu X, Luo Z. Advancements in Diagnostics and Therapeutics for Cancer of Unknown Primary in the Era of Precision Medicine. MedComm (Beijing) 2025; 6:e70161. [PMID: 40242159 PMCID: PMC12000684 DOI: 10.1002/mco2.70161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Cancer of unknown primary (CUP), a set of histologically confirmed metastases that cannot be identified or traced back to its primary despite comprehensive investigations, accounts for 2-5% of all malignancies. CUP is the fourth leading cause of cancer-related deaths worldwide, with a median overall survival (OS) of 3-16 months. CUP has long been challenging to diagnose principally due to the occult properties of primary site. In the current era of molecular diagnostics, advancements in methodologies based on cytology, histology, gene expression profiling (GEP), and genomic and epigenomic analysis have greatly improved the diagnostic accuracy of CUP, surpassing 90%. Our center conducted the world's first phase III trial and demonstrated improved progression-free survival and favorable OS by GEP-guided site-specific treatment of CUP, setting the foundation of site-specific treatment in first-line management for CUP. In this review, we detailed the epidemiology, etiology, pathogenesis, as well as the histologic, genetic, and clinical characteristics of CUP. We also provided an overview of the advancements in the diagnostics and therapeutics of CUP over the past 50 years. Moving forward, we propose optimizing diagnostic modalities and exploring further-line treatment regimens as two focus areas for future studies on CUP.
Collapse
Affiliation(s)
- Ting Zhao
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xiaowei Zhang
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Xin Liu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Qifeng Wang
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
| | - Xichun Hu
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| | - Zhiguo Luo
- Department of Medical OncologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghaiChina
| |
Collapse
|
7
|
Zhang X, Zhang X, Luo QK, Fu Q, Liu P, Pan CJ, Liu CJ, Zhang HW, Qin T. Pretreatment radiomic imaging features combined with immunological indicators to predict targeted combination immunotherapy response in advanced hepatocellular carcinoma. World J Clin Oncol 2025; 16:102735. [PMID: 40290677 PMCID: PMC12019258 DOI: 10.5306/wjco.v16.i4.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Early symptoms of hepatocellular carcinoma (HCC) are not obvious, and more than 70% of which does not receive radical hepatectomy, when first diagnosed. In recent years, molecular-targeted drugs combined with immunotherapy and other therapeutic methods have provided new treatment options for middle and advanced HCC (aHCC). Predicting the effect of targeted combined immunotherapy has become a hot topic in current research. AIM To explore the relationship between nodule enhancement in hepatobiliary phase and the efficacy of combined targeted immunotherapy for aHCC. METHODS Data from 56 patients with aHCC for magnetic resonance imaging with gadolinium-ethoxybenzyl-diethylenetriamine pentaacetic acid were retrospectively collected. Signal intensity of intrahepatic nodules was measured, and the hepatobiliary relative enhancement ratio (RER) was calculated. Progression-free survival (PFS) of patients with high and low reinforcement of HCC nodules was compared. The model was validated using receiver operating characteristic curves. Univariate and multivariate logistic regression and Kaplan-Meier analysis were performed to explore factors influencing the efficacy of targeted immunization and PFS. RESULTS Univariate and multivariate analyses revealed that the RER, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and prognostic nutritional index were significantly associated with the efficacy of tyrosine kinase inhibitors combined with immunotherapy (P < 0.05). The area under the curve of the RER for predicting the efficacy of tyrosine kinase inhibitors combined with anti-programmed death 1 antibody in patients with aHCC was 0.876 (95% confidence interval: 0.781-0.971, P < 0.05), the optimal cutoff value was 0.904, diagnostic sensitivity was 87.5%, and specificity was 79.2%. Kaplan-Meier analysis showed that neutrophil-to-lymphocyte ratio < 5, platelet-to-lymphocyte ratio < 300, prognostic nutritional index < 45, and RER < 0.9 significantly improved PFS. CONCLUSION AHCC nodules enhancement in the hepatobiliary stage was significantly correlated with PFS. Imaging information and immunological indicators had high predictive efficacy for targeted combined immunotherapy and were associated with PFS.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Xu Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Qian-Kun Luo
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Qiang Fu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Pan Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Chang-Jie Pan
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Chuan-Jiang Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Hong-Wei Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Tao Qin
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
8
|
Ma D, Fan C, Sano T, Kawabata K, Nishikubo H, Imanishi D, Sakuma T, Maruo K, Yamamoto Y, Matsuoka T, Yashiro M. Beyond Biomarkers: Machine Learning-Driven Multiomics for Personalized Medicine in Gastric Cancer. J Pers Med 2025; 15:166. [PMID: 40423038 DOI: 10.3390/jpm15050166] [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: 03/06/2025] [Revised: 04/15/2025] [Accepted: 04/22/2025] [Indexed: 05/28/2025] Open
Abstract
Gastric cancer (GC) remains one of the leading causes of cancer-related mortality worldwide, with most cases diagnosed at advanced stages. Traditional biomarkers provide only partial insights into GC's heterogeneity. Recent advances in machine learning (ML)-driven multiomics technologies, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, pathomics, and radiomics, have facilitated a deeper understanding of GC by integrating molecular and imaging data. In this review, we summarize the current landscape of ML-based multiomics integration for GC, highlighting its role in precision diagnosis, prognosis prediction, and biomarker discovery for achieving personalized medicine.
Collapse
Affiliation(s)
- Dongheng Ma
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Canfeng Fan
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Tomoya Sano
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Kyoka Kawabata
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Hinano Nishikubo
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Daiki Imanishi
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Takashi Sakuma
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Koji Maruo
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Yurie Yamamoto
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Tasuku Matsuoka
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| | - Masakazu Yashiro
- Molecular Oncology and Therapeutics, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Cancer Center for Translational Research, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
- Department of Gastroenterological Surgery, Osaka Metropolitan University Graduate School of Medicine, 1-4-3 Asahimachi, Abeno-ku, Osaka 545-8585, Japan
| |
Collapse
|
9
|
Dai XR, Zhang MZ, Chen L, Guo XW, Li ZX, Yan KF, He QQ, Cheng HW. Diagnostic value of systemic immune-inflammation index and prognostic nutritional index combined with CEA in gastric cancer with lymph node metastasis. Front Endocrinol (Lausanne) 2025; 16:1522349. [PMID: 40297178 PMCID: PMC12034562 DOI: 10.3389/fendo.2025.1522349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/10/2025] [Indexed: 04/30/2025] Open
Abstract
Background Carcinoembryonic antigen (CEA), systemic immune-inflammation index(SII), and prognostic nutritional index (PNI) are diagnostic markers for cancer, but their combined significance in gastric cancer (GC) with lymph node metastasis remains unclear. The aim of this study was to evaluate the association between these serum biomarkers and lymph node metastasis in patients with GC. Methods Records of patients with GC were reviewed retrospectively. Univariate and multivariate logistic regression were performed to examine the association between tumor markers, serum biomarkers and lymph node metastasis in GC. Based on the results of multivariate regression, a nomogram was developed and verified. Results Of the 395 patients aged 68.5 ± 9.1 years, 192 (48.6%) were diagnosed with lymphatic node metastasis. After adjusting for confounding factors, CEA (Odd ratio (OR):2.21; 95%CI: 1.17-3.81) and SII (OR:1.02; 95%CI: 1.01-1.04) was identified as significant risk factors, while PNI (OR:0.90; 95%CI: 0.85~0.96) was a protective factor for lymph node metastasis. The established nomogram by incorporating CEA, SII, PNI, differentiation, and tumor diameter can effectively predict lymph node metastasis in GC. Conclusion CEA, SII, PNI, differentiation, and tumor diameter were significantly associated with lymph node metastasis in patients with GC, and the combination of CEA, SII, PNI, differentiation, and tumor diameter has a better diagnostic value than either index alone.
Collapse
Affiliation(s)
- Xiao-rong Dai
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Min-zhe Zhang
- School of Public Health, Wuhan University, Wuhan, China
| | - Lei Chen
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Xin-wei Guo
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Zhen-xing Li
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Kun-feng Yan
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| | - Qi-qiang He
- School of Public Health, Wuhan University, Wuhan, China
- Hubei Biomass-Resource Chemistry and Environmental Biotechnology Key Laboratory, Wuhan University, Wuhan, China
| | - Hong-wei Cheng
- Department of Gastroenterology, Taixing People’s Hospital, Taixing, China
| |
Collapse
|
10
|
Guo X, Song J, Zhu L, Liu S, Huang C, Zhou L, Chen W, Lin G, Zhao Z, Tu J, Chen M, Chen F, Zheng L, Ji J. Multiparametric MRI-based radiomics and clinical nomogram predicts the recurrence of hepatocellular carcinoma after postoperative adjuvant transarterial chemoembolization. BMC Cancer 2025; 25:683. [PMID: 40229712 PMCID: PMC11995621 DOI: 10.1186/s12885-025-14079-y] [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: 03/02/2024] [Accepted: 04/03/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND This study was undertaken to develop and validate a radiomics model based on multiparametric magnetic resonance imaging (MRI) for predicting recurrence in patients with hepatocellular carcinoma (HCC) following postoperative adjuvant transarterial chemoembolization (PA-TACE). METHODS In this retrospective study, 149 HCC patients (81 for training, 36 for internal validation, 32 for external validation) treated with PA-TACE were included in two medical centers. Multiparametric radiomics features were extracted from three MRI sequences. Least absolute shrinkage and selection operator (LASSO)-COX regression was utilized to select radiomics features. Optimal clinical characteristics selected by multivariate Cox analysis were integrated with Rad-score to develop a recurrence-free survival (RFS) prediction model. The model performance was evaluated by time-dependent receiver operating characteristic (ROC) curves, Harrell's concordance index (C-index), and calibration curve. RESULTS Fifteen optimal radiomic features were selected and the median Rad-score value was 0.434. Multivariate Cox analysis indicated that neutrophil-to-lymphocyte ratio (NLR) (hazard ratio (HR) = 1.49, 95% confidence interval (CI): 1.1-2.1, P = 0.022) and tumor size (HR = 1.28, 95% CI: 1.1-1.5, P = 0.001) were the independent predictors of RFS after PA-TACE. A combined model was established by integrating Rad-score, NLR, and tumor size in the training cohort (C-index 0.822; 95% CI 0.805-0.861), internal validation cohort (0.823; 95% CI 0.771-0.876) and external validation cohort (0.846; 95% CI 0.768-0.924). The calibration curve exhibited a satisfactory correspondence. CONCLUSION A multiparametric MRI-based radiomics model can predict RFS of HCC patients receiving PA-TACE and a nomogram can be served as an individualized tool for prognosis.
Collapse
Affiliation(s)
- Xinyu Guo
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Jingjing Song
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
| | - Lingyi Zhu
- Second Clinical Medical School, Zhejiang Chinese Medicine University, Hangzhou, 310003, Zhejiang, China
| | - Shuang Liu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Chaoming Huang
- Second Clinical Medical School, Zhejiang Chinese Medicine University, Hangzhou, 310003, Zhejiang, China
| | - Lingling Zhou
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Weiyue Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Guihan Lin
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Zhongwei Zhao
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Jianfei Tu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Minjiang Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China
| | - Feng Chen
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, Zhejiang, China
| | - Liyun Zheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Zhejiang Key Laboratory of Imaging and Interventional Medicine, Lishui Hospital of Zhejiang University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, 323000, China.
| |
Collapse
|
11
|
Azbergenov NK, Akhmetova SZ, Nurulla TA, Kaliev AR, Ramankulova AB, Tulyayeva AB, Kereeva NM. Biomarkers used in the diagnosis and prognosis of gastric cancer in young patients: a scientometric analysis. Front Med (Lausanne) 2025; 12:1586742. [PMID: 40270496 PMCID: PMC12014542 DOI: 10.3389/fmed.2025.1586742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Introduction Gastric cancer in young people is a global health burden, although it is less common than in other age groups. The use of biomarkers is developing in the diagnosis, treatment selection and prognosis of gastric cancer in young patients. In this bibliometric analysis we aim to evaluate the progress of this knowledge, trend topic development and scientific teams and countries involvements in the topic of biomarkers role in gastric cancer in young patients. Methods The data were obtained from Scopus (536 publications) for the period 1993-2024, all relevant metadata were analyzed using RStudio and Biblioshiny package to perform global trends and hotspots analysis. Results Publication trends show a constant increase in interest in gastric cancer biomarkers used in the diagnosis and prognosis of gastric cancer in young patients (7.71% per year). The leading countries were China, USA, and Japan, between which there is strong and sustained collaborations. International co-authorship is relatively low (19.4%). The most prolific research centers were Sungkyunkwan University, Sun Yat-sen University, and Fudan University. The most productive researchers were Zhang X., Wang Y., and Li Y. Keywords analysis showed an increase in mentions of topics related to diagnostics (biomarkers, immunohistochemistry), personalized medicine and prognosis. Conclusion Bibliometric analysis of more than three decades research articles on gastric cancer biomarkers in young patients showed a steady increase, with strong contributions from leading countries and institutions, highlighting the growing focus on diagnostics, personalized medicine, and prognosis.
Collapse
Affiliation(s)
- Nurbek Kozhakhmetuly Azbergenov
- Department of Pathological Anatomy and Forensic Medicine, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Saule Zhumabaevna Akhmetova
- Department of Pathological Anatomy and Forensic Medicine, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Talshyn Amirkhanovna Nurulla
- Department of Pathological Anatomy and Forensic Medicine, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Abdiraman Rsalievich Kaliev
- Department of Pathological Anatomy and Forensic Medicine, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Aigul Bulatovna Ramankulova
- Department of Pathological Anatomy and Forensic Medicine, West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Anar Balkashevna Tulyayeva
- Department of Oncology, Medical Center of West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| | - Nurgul Meirimovna Kereeva
- Department of Oncology, Medical Center of West Kazakhstan Marat Ospanov Medical University, Aktobe, Kazakhstan
| |
Collapse
|
12
|
Jiang J, Fan Z, Jiang S, Chen X, Guo H, Dong S, Jiang T. Interpretable multimodal deep learning model for predicting post-surgical international society of urological pathology grade in primary prostate cancer. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07248-5. [PMID: 40183953 DOI: 10.1007/s00259-025-07248-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 03/21/2025] [Indexed: 04/05/2025]
Abstract
PURPOSE To address heterogeneity in prostate cancer (PCa) pathological grading, we developed an interpretable multimodal fusion model integrating 18F prostate-specific membrane antigen (18F-PSMA)-targeted positron emission tomography/computed tomography (18F-PSMA-PET/CT) imaging features with clinical variables for predicting post-surgical ISUP grade (psISUP ≥ 4 vs. < 4). METHODS This retrospective study analyzed 222 patients with PCa (2020-2024) undergoing 18F-PSMA PET/CT. We constructed a deep transfer learning framework incorporating radiomic features from PET/CT and clinical parameters. Model performance was validated against three established methods and preoperative biopsy Gleason scores. Additionally, SHapley Additive exPlanations (SHAP) values elucidated feature contributions, and a radiomic nomogram was developed for clinical translation. RESULTS The fusion model achieved superior discrimination in psISUP grading (test set area under the curve (AUC) = 0.850, 95% confidence interval [CI] 0.769-0.932; validation set AUC = 0.833, 95% CI 0.657-1.000), significantly outperforming preoperative Gleason scores. SHAP analysis identified PSMA uptake heterogeneity and PSA density as key predictive features. The nomogram demonstrated clinical interpretability through visualised risk stratification. CONCLUSION Our deep learning-based multimodal fusion model enables accurate preoperative prediction of aggressive PCa pathology (ISUP ≥ 4), potentially optimising surgical planning and personalised therapeutic strategies. The interpretable framework enhances clinical trustworthiness in artificial intelligence-assisted decision-making.
Collapse
Affiliation(s)
- Jiamei Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Zhiyan Fan
- Department of Emergency, Hangzhou First People's Hospital of West Lake University, Hangzhou, Zhejiang, 310006, China
| | - Shen Jiang
- Department of Urology, Jilin Cancer Hospital, Changchun, Jilin, 130021, China
| | - Xia Chen
- Department of Nuclear Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Hongyu Guo
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Shuangyong Dong
- Department of Emergency, Hangzhou First People's Hospital of West Lake University, Hangzhou, Zhejiang, 310006, China.
| | - Tianan Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
13
|
Zhou Y, Duan Y, Zhu Q, Li S, Liu X, Cheng T, Cheng D, Shi Y, Zhang J, Yang J, Zheng Y, Gao C, Wang J, Cao Y, Zhang C. Integrative deep learning and radiomics analysis for ovarian tumor classification and diagnosis: a multicenter large-sample comparative study. LA RADIOLOGIA MEDICA 2025:10.1007/s11547-025-02006-x. [PMID: 40167932 DOI: 10.1007/s11547-025-02006-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 03/14/2025] [Indexed: 04/02/2025]
Abstract
PURPOSE This study aims to evaluate the effectiveness of combining transvaginal ultrasound (US)-based radiomics and deep learning model for the accurate differentiation between benign and malignant ovarian tumors in large-scale studies. MATERIALS AND METHODS A multicenter retrospective study collected grayscale and color US images of ovarian tumors. Patients were divided into training, internal, and external validation groups. Models including a convolutional neural networks (CNN), optimal radiomics, and a combined model were constructed and evaluated for predictive performance using area under curve (AUC), sensitivity, and specificity. The DeLong test compared model AUCs with O-RADS and expert assessments. RESULTS 3193 images from 2078 patients were analyzed. The CNN achieved AUCs of 0.970 (internal) and 0.959 (external), respectively. Optimal radiomic model achieved AUCs of 0.949 (internal) and 0.954 (external), respectively. The combined CNN-radiomics model attained the highest AUC of 0.977 (internal) and 0.972 (external), respectively, outperforming individual models, O-RADS, and expert methods (p < 0.05). CONCLUSIONS The combined CNN-radiomics model using transvaginal US images provides more accurate and reliable ovarian tumor diagnosis, enhancing malignancy prediction and offering clinicians a more precise diagnostic tool.
Collapse
Affiliation(s)
- Yi Zhou
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Yayang Duan
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Qiwei Zhu
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Siyao Li
- Department of Ultrasound, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, Anhui Province, China
| | - Xiaoling Liu
- Department of Ultrasound, Nanchong Central Hospital, Nanchong, 637003, Sichuan, China
| | - Ting Cheng
- Department of Ultrasound, Lu'an Second Hospital, Lu'an, 237000, Anhui Province, China
| | - Dongliang Cheng
- Hebin Intelligent Robots Co., LTD, Hefei, 230022, Anhui Province, China
| | - Yuanyin Shi
- Hebin Intelligent Robots Co., LTD, Hefei, 230022, Anhui Province, China
| | - Jingshu Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Jinyan Yang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Yanyan Zheng
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Chuanfen Gao
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China
| | - Junli Wang
- Department of Ultrasound, Second People's Hospital of Wuhu, Jinghu District, NO.231 Jiuhuazhong 24 Road, Wuhu, 241000, Anhui Province, China.
| | - Yunxia Cao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China.
| | - Chaoxue Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Shushan District, NO.218 Jixi Road, Hefei, 230022, Anhui Province, China.
| |
Collapse
|
14
|
Wang Y, Chi S, Tian Y, Li X, Zhang H, Xu Y, Huang C, Gao Y, Jin G, Fu Q, Cao W, Chen C, Ding H, Zhang Y, Hong Y, Li J, Sun X, Li E, Zhang Y, Yao W, Liu R, Hua Y, Huang H, Xu M, Zhang B, Tao W, Yang T, Gao Y, Wang X, Lin C, Li J, Zhang Q, Liang T. Construction of an artificially intelligent model for accurate detection of HCC by integrating clinical, radiological, and peripheral immunological features. Int J Surg 2025; 111:2942-2952. [PMID: 39878177 DOI: 10.1097/js9.0000000000002281] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Integrating comprehensive information on hepatocellular carcinoma (HCC) is essential to improve its early detection. We aimed to develop a model with multimodal features (MMF) using artificial intelligence (AI) approaches to enhance the performance of HCC detection. MATERIALS AND METHODS A total of 1092 participants were enrolled from 16 centers. These participants were allocated into the training, internal validation, and external validation cohorts. Peripheral blood specimens were collected prospectively and subjected to mass cytometry analysis. Clinical and radiological data were obtained from electrical medical records. Various AI methods were employed to identify pertinent features and construct single-modal models with optimal performance. The XGBoost algorithm was utilized to amalgamate these models, integrating multimodal information and facilitating the development of a fusion model. Model evaluation and interpretability were demonstrated using the SHapley Additive exPlanations method. RESULTS We constructed the electronic health record, BioScore, RadiomicScore, and DLScore models based on clinical, radiological, and peripheral immunological features, respectively. Subsequently, these single-modal models were amalgamated to develop an all-in-one MMF model. The MMF model exhibited enhanced performance compared to models comprising only single-modal features in detecting HCC. This superiority in performance was confirmed through the internal and external validation cohorts, yielding area under the curve (AUC) values of 0.985 and 0.915, respectively. Additionally, the MMF model improved the detection ability in subpopulations of HCCs that were negative for alpha-fetoprotein and those with small size, with AUC values of 0.974 and 0.996, respectively. CONCLUSIONS Integrating MMF improved the performance of the model for HCC detection.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengqiang Chi
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yu Tian
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xueyao Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Hang Zhang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiting Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Huang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiwei Gao
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Gaowei Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qihan Fu
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanyue Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cao Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haonan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yupeng Hong
- Department of Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Junjian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xu Sun
- Department of General Surgery, Huzhou Central Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Enliang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuhua Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | - Weiyun Yao
- Department of Surgery, Changxing People's Hospital, Huzhou, China
| | - Runtian Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongfei Hua
- Department of General Surgery, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, China
| | - Haifeng Huang
- Department of General Surgery, Shengzhou People's Hospital, Shengzhou, China
| | - Minghui Xu
- Department of General Surgery, Haining People's Hospital, Haining, China
| | - Bo Zhang
- Department of General Surgery, Shenzhen University Luohu People's Hospital, Shenzhen, China
| | - Weifeng Tao
- Department of General Surgery, Shangyu People's Hospital of Shaoxing, Shangyu, China
| | - Tianxing Yang
- Department of Medical Oncology, Sanmen People's Hospital, Taizhou, China
| | - Yuming Gao
- Department of General Surgery, Jixi County People's Hospital, Jixi, China
| | - Xiaoguang Wang
- Department of General Surgery, Jiaxing Second People's Hospital, Jiaxing, China
| | - Cheng Lin
- Zhejiang Puluoting Health Technology Co Ltd, Hangzhou, China
| | - Jingsong Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
15
|
Zhou S, Xie Y, Feng X, Li Y, Shen L, Chen Y. Artificial intelligence in gastrointestinal cancer research: Image learning advances and applications. Cancer Lett 2025; 614:217555. [PMID: 39952597 DOI: 10.1016/j.canlet.2025.217555] [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: 12/04/2024] [Revised: 01/31/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025]
Abstract
With the rapid advancement of artificial intelligence (AI) technologies, including deep learning, large language models, and neural networks, these methodologies are increasingly being developed and integrated into cancer research. Gastrointestinal tumors are characterized by complexity and heterogeneity, posing significant challenges for early detection, diagnostic accuracy, and the development of personalized treatment strategies. The application of AI in digestive oncology has demonstrated its transformative potential. AI not only alleviates the diagnostic burden on clinicians, but it improves tumor screening sensitivity, specificity, and accuracy. Additionally, AI aids the detection of biomarkers such as microsatellite instability and mismatch repair, supports intraoperative assessments of tumor invasion depth, predicts treatment responses, and facilitates the design of personalized treatment plans to potentially significantly enhance patient outcomes. Moreover, the integration of AI with multiomics analyses and imaging technologies has led to substantial advancements in foundational research on the tumor microenvironment. This review highlights the progress of AI in gastrointestinal oncology over the past 5 years with focus on early tumor screening, diagnosis, molecular marker identification, treatment planning, and prognosis predictions. We also explored the potential of AI to enhance medical imaging analyses to aid tumor detection and characterization as well as its role in automating and refining histopathological assessments.
Collapse
Affiliation(s)
- Shengyuan Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yi Xie
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Xujiao Feng
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yanyan Li
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Lin Shen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yang Chen
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education, Beijing), Peking University Cancer Hospital and Institute, Beijing, China; Department of Gastrointestinal Cancer, Beijing GoBroad Hospital, Beijing, 102200, China.
| |
Collapse
|
16
|
Li D, Hu W, Ma L, Yang W, Liu Y, Zou J, Ge X, Han Y, Gan T, Cheng D, Ai K, Liu G, Zhang J. Deep learning radiomics nomograms predict Isocitrate dehydrogenase (IDH) genotypes in brain glioma: A multicenter study. Magn Reson Imaging 2025; 117:110314. [PMID: 39708927 DOI: 10.1016/j.mri.2024.110314] [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: 09/27/2024] [Revised: 12/16/2024] [Accepted: 12/16/2024] [Indexed: 12/23/2024]
Abstract
PURPOSE To explore the feasibility of Deep learning radiomics nomograms (DLRN) in predicting IDH genotype. METHODS A total of 402 glioma patients from two independent centers were retrospectively included, and the data from center I was randomly divided into a training cohort (n = 239) and an internal validation cohort (n = 103) on a 7:3 basis. Center II served as an independent external validation cohort (n = 60). We developed a DLRN for IDH classification of gliomas based on T2 images. This hybrid model integrates deep learning features, radiomics features, and clinical features most relevant to IDH genotypes and finally classifies them using multivariate logistic regression analysis. We used the area under the curve (AUC) of the receiver operating characteristic (ROC) to evaluate the performance of the model and applied the DLRN score to the survival analysis of some of the follow-up glioma patients. RESULTS The proposed model had an area under the curve (AUC) of 0.98 in an externally validated cohort, and DLRN scores were significantly associated with the overall survival of glioma patients. CONCLUSIONS Deep learning radiomics nomograms performed well in non-invasively predicting IDH mutation status in gliomas, assisting stratified management and targeted therapy for glioma patients.
Collapse
Affiliation(s)
- Darui Li
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Wanjun Hu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Laiyang Ma
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Wenxia Yang
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Yang Liu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Jie Zou
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Xin Ge
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Yuping Han
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Tiejun Gan
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Dan Cheng
- Department of Radiology, Beijing Tiantan Hospital, Capital Medical University, Beijing, China
| | - Kai Ai
- Philips Healthcare, Xi'an, China
| | - Guangyao Liu
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China
| | - Jing Zhang
- Department of Nuclear Magnetic Resonance, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China; Gansu Province Clinical Research Center for Functional and Molecular Imaging, Lanzhou 730030, China.
| |
Collapse
|
17
|
Huang X, Qin M, Fang M, Wang Z, Hu C, Zhao T, Qin Z, Zhu H, Wu L, Yu G, De Cobelli F, Xie X, Palumbo D, Tian J, Dong D. The application of artificial intelligence in upper gastrointestinal cancers. JOURNAL OF THE NATIONAL CANCER CENTER 2025; 5:113-131. [PMID: 40265096 PMCID: PMC12010392 DOI: 10.1016/j.jncc.2024.12.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Revised: 09/17/2024] [Accepted: 12/20/2024] [Indexed: 04/24/2025] Open
Abstract
Upper gastrointestinal cancers, mainly comprising esophageal and gastric cancers, are among the most prevalent cancers worldwide. There are many new cases of upper gastrointestinal cancers annually, and the survival rate tends to be low. Therefore, timely screening, precise diagnosis, appropriate treatment strategies, and effective prognosis are crucial for patients with upper gastrointestinal cancers. In recent years, an increasing number of studies suggest that artificial intelligence (AI) technology can effectively address clinical tasks related to upper gastrointestinal cancers. These studies mainly focus on four aspects: screening, diagnosis, treatment, and prognosis. In this review, we focus on the application of AI technology in clinical tasks related to upper gastrointestinal cancers. Firstly, the basic application pipelines of radiomics and deep learning in medical image analysis were introduced. Furthermore, we separately reviewed the application of AI technology in the aforementioned aspects for both esophageal and gastric cancers. Finally, the current limitations and challenges faced in the field of upper gastrointestinal cancers were summarized, and explorations were conducted on the selection of AI algorithms in various scenarios, the popularization of early screening, the clinical applications of AI, and large multimodal models.
Collapse
Affiliation(s)
- Xiaoying Huang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Minghao Qin
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology Beijing, Beijing, China
| | - Mengjie Fang
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Zipei Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Chaoen Hu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Tongyu Zhao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
- University of Science and Technology of China, Hefei, China
| | - Zhuyuan Qin
- Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, China
- Beijing University of Chinese Medicine, Beijing, China
| | | | - Ling Wu
- KiangWu Hospital, Macau, China
| | | | | | | | - Diego Palumbo
- Department of Radiology, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China
- Key Laboratory of Big Data-Based Precision Medicine, Beihang University, Ministry of Industry and Information Technology, Beijing, China
| | - Di Dong
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
18
|
Li R, Li J, Wang Y, Liu X, Xu W, Sun R, Xue B, Zhang X, Ai Y, Du Y, Jiang J. The artificial intelligence revolution in gastric cancer management: clinical applications. Cancer Cell Int 2025; 25:111. [PMID: 40119433 PMCID: PMC11929235 DOI: 10.1186/s12935-025-03756-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 03/18/2025] [Indexed: 03/24/2025] Open
Abstract
Nowadays, gastric cancer has become a significant issue in the global cancer burden, and its impact cannot be ignored. The rapid development of artificial intelligence technology is attempting to address this situation, aiming to change the clinical management landscape of gastric cancer fundamentally. In this transformative change, machine learning and deep learning, as two core technologies, play a pivotal role, bringing unprecedented innovations and breakthroughs in the diagnosis, treatment, and prognosis evaluation of gastric cancer. This article comprehensively reviews the latest research status and application of artificial intelligence algorithms in gastric cancer, covering multiple dimensions such as image recognition, pathological analysis, personalized treatment, and prognosis risk assessment. These applications not only significantly improve the sensitivity of gastric cancer risk monitoring, the accuracy of diagnosis, and the precision of survival prognosis but also provide robust data support and a scientific basis for clinical decision-making. The integration of artificial intelligence, from optimizing the diagnosis process and enhancing diagnostic efficiency to promoting the practice of precision medicine, demonstrates its promising prospects for reshaping the treatment model of gastric cancer. Although most of the current AI-based models have not been widely used in clinical practice, with the continuous deepening and expansion of precision medicine, we have reason to believe that a new era of AI-driven gastric cancer care is approaching.
Collapse
Affiliation(s)
- Runze Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Jingfan Li
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yuman Wang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xiaoyu Liu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Weichao Xu
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Runxue Sun
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China
| | - Binqing Xue
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Xinqian Zhang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China
| | - Yikun Ai
- North China University of Science and Technology, Tanshan 063000, China
| | - Yanru Du
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Provincial Key Laboratory of Integrated Traditional and Western Medicine Research on Gastroenterology, Hebei, 050011, China.
- Hebei Key Laboratory of Turbidity and Toxicology, Hebei, 050011, China.
| | - Jianming Jiang
- Hebei University of Traditional Chinese Medicine, Hebei, 050011, China.
- Hebei Hospital of Traditional Chinese Medicine, Hebei, 050011, China.
| |
Collapse
|
19
|
Kang D, Jeon HJ, Kim JH, Oh SI, Seong YS, Jang JY, Kim JW, Kim JS, Nam SJ, Bang CS, Choi HS. Enhancing Lymph Node Metastasis Risk Prediction in Early Gastric Cancer Through the Integration of Endoscopic Images and Real-World Data in a Multimodal AI Model. Cancers (Basel) 2025; 17:869. [PMID: 40075715 PMCID: PMC11898873 DOI: 10.3390/cancers17050869] [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: 10/29/2024] [Revised: 02/14/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
Objectives: The accurate prediction of lymph node metastasis (LNM) and lymphovascular invasion (LVI) is crucial for determining treatment strategies for early gastric cancer (EGC). This study aimed to develop and validate a deep learning-based clinical decision support system (CDSS) to predict LNM including LVI in EGC using real-world data. Methods: A deep learning-based CDSS was developed by integrating endoscopic images, demographic data, biopsy pathology, and CT findings from the data of 2927 patients with EGC across five institutions. We compared a transformer-based model to an image-only (basic convolutional neural network (CNN)) model and a multimodal classification (CNN with random forest) model. Internal testing was conducted on 449 patients from the five institutions, and external validation was performed on 766 patients from two other institutions. Model performance was assessed using the area under the receiver operating characteristic curve (AUC), probability density function, and clinical utility curve. Results: In the training, internal, and external validation cohorts, LNM/LVI was observed in 379 (12.95%), 49 (10.91%), 15 (9.09%), and 41 (6.82%) patients, respectively. The transformer-based model achieved an AUC of 0.9083, sensitivity of 85.71%, and specificity of 90.75%, outperforming the CNN (AUC 0.5937) and CNN with random forest (AUC 0.7548). High sensitivity and specificity were maintained in internal and external validations. The transformer model distinguished 91.8% of patients with LNM in the internal validation dataset, and 94.0% and 89.1% in the two different external datasets. Conclusions: We propose a deep learning-based CDSS for predicting LNM/LVI in EGC by integrating real-world data, potentially guiding treatment strategies in clinical settings.
Collapse
Affiliation(s)
- Donghoon Kang
- Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Seoul 06591, Republic of Korea;
| | - Han Jo Jeon
- Department of Internal Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.)
| | - Jie-Hyun Kim
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Sang-Il Oh
- Waycen Inc., Seoul 06167, Republic of Korea;
| | - Ye Seul Seong
- Department of Internal Medicine, Gangnam Severance Hospital, Yonsei University College of Medicine, Seoul 06273, Republic of Korea;
| | - Jae Young Jang
- Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (J.Y.J.); (J.-W.K.)
| | - Jung-Wook Kim
- Department of Internal Medicine, Kyung Hee University Medical Center, Kyung Hee University College of Medicine, Seoul 05278, Republic of Korea; (J.Y.J.); (J.-W.K.)
| | - Joon Sung Kim
- Department of Internal Medicine, Incheon St. Mary’s Hospital, The Catholic University of Korea College of Medicine, Incheon 21431, Republic of Korea;
| | - Seung-Joo Nam
- Department of Internal Medicine, Kangwon National University Hospital, Kangwon National University School of Medicine, Chuncheon 24289, Republic of Korea;
| | - Chang Seok Bang
- Department of Internal Medicine, Chuncheon Sacred Heart Hospital, Hallym University College of Medicine, Chuncheon 24253, Republic of Korea;
| | - Hyuk Soon Choi
- Department of Internal Medicine, Anam Hospital, Korea University College of Medicine, Seoul 02841, Republic of Korea; (H.J.J.); (H.S.C.)
| |
Collapse
|
20
|
Cen X, He J, Tong Y, Yang H, Lu Y, Li Y, Dong W, Hu C. A Deep Radiomics Model for Lymph Node Metastasis Prediction of Early-Stage Gastric Cancer Based on CT Images. Acad Radiol 2025; 32:1771-1772. [PMID: 39904665 DOI: 10.1016/j.acra.2024.12.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 12/15/2024] [Accepted: 12/16/2024] [Indexed: 02/06/2025]
Affiliation(s)
- Xiaoping Cen
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China (X.C., H.Y.); HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China (X.C., H.Y., W.D.); BGI Research, Shenzhen 518083, China (X.C.); Guangzhou National Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, China (X.C., Y.L.)
| | - Jingyang He
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China (J.H., C.H.); Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang, Hangzhou 310022, China (J.H., Y.T., C.H.)
| | - Yahan Tong
- Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang, Hangzhou 310022, China (J.H., Y.T., C.H.); Department of Radiology, Zhejiang Cancer Hospital, Hangzhou 310022, China (Y.T.)
| | - Huanming Yang
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China (X.C., H.Y.); HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China (X.C., H.Y., W.D.); BGI, Shenzhen 518083, China (H.Y.); James D. Watson Institute of Genome Sciences, Hangzhou 310029, China (H.Y.)
| | - Youyong Lu
- Laboratory of Molecular Oncology, Peking University Cancer Hospital and Institute, Beijing 100142, China (Y.L.)
| | - Yixue Li
- Guangzhou National Laboratory, No.9 XingDaoHuanBei Road, Guangzhou International Bio Island, Guangzhou 510005, China (X.C., Y.L.); GZMU-GIBH Joint School of Life Sciences, The Guangdong-Hong Kong-Macau Joint Laboratory for Cell Fate Regulation and Diseases, Guangzhou Medical University, Guangzhou 511436, China (Y.L.)
| | - Wei Dong
- HIM-BGI Omics Center, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China (X.C., H.Y., W.D.); Clin Lab, BGI Genomics, Beijing 100000, China (W.D.)
| | - Can Hu
- Department of Gastric Surgery, Zhejiang Cancer Hospital, Hangzhou 310022, China (J.H., C.H.); Key Laboratory of Prevention, Diagnosis and Therapy of Upper Gastrointestinal Cancer of Zhejiang, Hangzhou 310022, China (J.H., Y.T., C.H.).
| |
Collapse
|
21
|
Zeng Y, Liu Y, Li J, Feng B, Lu J. Value of Computed Tomography Scan for Detecting Lymph Node Metastasis in Early Esophageal Squamous Cell Carcinoma. Ann Surg Oncol 2025; 32:1635-1650. [PMID: 39586955 DOI: 10.1245/s10434-024-16568-z] [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: 08/02/2024] [Accepted: 11/10/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND The necessity of computed tomography (CT) scan for detecting potential lymph node metastasis (LNM) in early esophageal squamous cell carcinoma (ESCC) before endoscopic and surgical treatments is under debate. METHODS Patients with histologically proven ESCC limited to the mucosa or submucosa were examined retrospectively. Diagnostic performance of CT for detecting LNM was analyzed by comparing original CT reports with pathology reports. The sensitivity, specificity, accuracy, positive predictive value (PPV), and negative predictive value (NPV) were calculated. RESULTS A total of 625 patients from three tertiary referral hospitals were included. The rate of pathologically confirmed LNM was 12.5%. Based on original CT reports, the sensitivity, specificity, accuracy, PPV, and NPV of CT to determine LNM in T1 ESCC were 41.0%, 83.2%, 77.9%, 25.8%, and 90.8% respectively. For mucosal cancers (T1a), these parameters were 50.0%, 81.7%, 80.9%, 6.8%, and 98.4%, respectively. For submucosal cancers (T1b), they were 40.0%, 85.0%, 75.0%, 43.0%, and 83.3%, respectively. Additionally, the diagnostic performance of CT for LNM was relatively better for ESCC in the lower esophagus. Pathologically, 69.2% of patients with LNM did not exhibit lymphovascular invasion (LVI), and the sensitivity of CT for recognizing LNM in these patients (33.3%) was lower than those with LVI (58.3%). CONCLUSIONS Computed tomography can detect nearly half of the LNM cases in early ESCC with high specificity. The performance of CT further improved in LNM cases with LVI. Therefore, we conclude that routine preoperative CT for the assessment of potential LNM risk in patients with early ESCC is necessary.
Collapse
Affiliation(s)
- Yunqing Zeng
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Yaping Liu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
| | - Jinhou Li
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China
- Department of Gastroenterology, Taian City Central Hospital, Taian, Shandong, China
| | - Bingcheng Feng
- Department of Gastroenterology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Jiaoyang Lu
- Department of Gastroenterology, Qilu Hospital of Shandong University, Jinan, Shandong, China.
- Medical Integration and Practice Center, Shandong University, Jinan, China.
| |
Collapse
|
22
|
Zhu X, Sun H, Wang Y, Hu G, Shao L, Zhang S, Liu F, Chi C, He K, Tang J, An Y, Tian J, Liu Z. Prediction of Lymph Node Metastasis in Colorectal Cancer Using Intraoperative Fluorescence Multi-Modal Imaging. IEEE TRANSACTIONS ON MEDICAL IMAGING 2025; 44:1568-1580. [PMID: 40030456 DOI: 10.1109/tmi.2024.3510836] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The diagnosis of lymph node metastasis (LNM) is essential for colorectal cancer (CRC) treatment. The primary method of identifying LNM is to perform frozen sections and pathologic analysis, but this method is labor-intensive and time-consuming. Therefore, combining intraoperative fluorescence imaging with deep learning (DL) methods can improve efficiency. The majority of recent studies only analyze uni-modal fluorescence imaging, which provides less semantic information. In this work, we mainly established a multi-modal fluorescence imaging feature fusion prediction (MFI-FFP) model combining white light, fluorescence, and pseudo-color imaging of lymph nodes for LNM prediction. Firstly, based on the properties of various modal imaging, distinct feature extraction networks are chosen for feature extraction, which could significantly enhance the complementarity of various modal information. Secondly, the multi-modal feature fusion (MFF) module, which combines global and local information, is designed to fuse the extracted features. Furthermore, a novel loss function is formulated to tackle the issue of imbalanced samples, challenges in differentiating samples, and enhancing sample variety. Lastly, the experiments show that the model has a higher area under the receiver operating characteristic (ROC) curve (AUC), accuracy (ACC), and F1 score than the uni-modal and bi-modal models and has a better performance compared to other efficient image classification networks. Our study demonstrates that the MFI-FFP model has the potential to help doctors predict LNM and shows its promise in medical image analysis.
Collapse
|
23
|
Jiang C, Fang W, Wei N, Ma W, Dai C, Liu R, Cai A, Feng Q. Node Reporting and Data System Combined With Computed Tomography Radiomics Can Improve the Prediction of Nonenlarged Lymph Node Metastasis in Gastric Cancer. J Comput Assist Tomogr 2025; 49:215-224. [PMID: 39438281 DOI: 10.1097/rct.0000000000001673] [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: 10/25/2024]
Abstract
OBJECTIVES To investigate the diagnostic performance of Node Reporting and Data System (Node-RADS) combined with computed tomography (CT) radiomics for assessing nonenlargement regional lymph nodes in gastric cancer (GC). METHODS Preoperative CT images were retrospectively collected from 376 pathologically confirmed of gastric adenocarcinoma from January 2019 to December 2023, with 605 lymph nodes included for analysis. They were divided into training (n = 362) and validation (n = 243) sets. Radiomics features were extracted from venous-phase, and the radiomics score was obtained. Clinical information, CT parameters, and Node-RADS classification were collected. A combined model was built using machine-learning approach and tested in validation set using receiver operating characteristic curve analysis. Further validation was conducted in different subgroups of lymph node short-axis diameter (SD) range. RESULTS Node-RADS score, SD, maximum diameter of thickness of tumor, and radiomics were identified as the most predictive factors. The results demonstrated that the integrated model combining SD, maximum diameter of thickness of tumor, Node-RADS, and radiomics outperformed the model excluding radiomics, yielding an area under the receiver operating characteristic curve of 0.82 compared with 0.79, with a statistically significant difference ( P < 0.001). Subgroup analysis based on different SDs of lymph nodes also revealed enhanced diagnostic accuracy when incorporating the radiomics score for the 4- to 7.9-mm subgroups, all P < 0.05. However, for the 8- to 9.9-mm subgroup, the combination of the radiomics did not significantly improve the prediction, with an area under the receiver operating characteristic curve of 0.85 versus 0.85, P = 0.877. CONCLUSION The integration of radiomics scores with Node-RADS assessments significantly enhances the accuracy of lymph node metastasis evaluation for GC. This combined model is particularly effective for lymph nodes with smaller standard deviations, yielding a marked improvement in diagnostic precision. CLINICAL RELEVANCE STATEMENT The findings of this study indicate that a composite model, which incorporates Node-RADS, radiomics features, and conventional parameters, may serve as an effective method for the assessment of nonenlarged lymph nodes in GC.
Collapse
Affiliation(s)
| | - Wei Fang
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Na Wei
- Yidu Central Hospital of Shandong Second Medical University, Qingzhou
| | - Wenwen Ma
- Radiology Department, Affiliated Hospital of Shandong Second Medical University, Weifang
| | - Cong Dai
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Ruixue Liu
- Pathology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong Province, China
| | - Anzhen Cai
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| | - Qiang Feng
- Radiology Department, Yidu Central Hospital of Shandong Second Medical University, Qingzhou, Shandong
| |
Collapse
|
24
|
Zhou YH, Chen XL, Zhang X, Pu H, Li H. Dual-phase contrast-enhanced CT-based intratumoral and peritumoral radiomics for preoperative prediction of lymph node metastasis in gastric cancer. BMC Gastroenterol 2025; 25:123. [PMID: 40021977 PMCID: PMC11869644 DOI: 10.1186/s12876-025-03728-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/04/2025] [Accepted: 02/24/2025] [Indexed: 03/03/2025] Open
Abstract
OBJECTIVE To determine whether intratumoral and peritumoral radiomics derived from dual-phase contrast-enhanced CT imaging could predict lymph node metastasis (LNM) in gastric cancer. METHODS Patients with gastric cancer from January 2017 to January 2022 were retrospectively collected and were randomly divided into training cohort (n = 287) and test cohort (n = 121) with a ratio of 7: 3. Clinical features and traditional radiological features were analyzed to construct clinical model. Radiomics features based on intratumoral (ITV) and peritumoral volumetric (PTV) regions of the tumor were extracted and screened to construct radiomics models. Clinical-radiomics combined model was constructed by the most predictive radiomics features and clinical independent predictors. The correlation between LNM predicted by the best model and 2-year disease-free survival (DFS) was evaluated by the Kaplan-Meier analysis. RESULTS CT-LNM and CT-T stage were independent predictors of LNM. Compared with other radiomics models, ITV + PTV on atrial and venous phase (ITV + PTV-AP + VP) radiomics model presented moderate AUCs of 0.679 and 0.670 in the training cohort and validation cohort, respectively. Among the models, clinical-radiomics combined model achieved the highest AUC of 0.894 and 0.872 in the training and test cohorts, and 0.744 and 0.784 in the T1-2 and T3-4 subgroups, respectively. Clinical-radiomics combined model based LNM could stratify patients into high-risk and low-risk groups, and 2-year DFS of high-risk group was significantly lower than that of low-risk group (p < 0.001). CONCLUSION Clinical-radiomics combined model integrating CT-LNM, CT-T stage, and ITV-PTV-AP + VP radiomics features could predict LNM, and this combined model based LNM was associated with 2-year DFS.
Collapse
Affiliation(s)
- Yun-Hui Zhou
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610072, China
- Department of Radiology, The Third Affiliated Hospital of Chengdu Medical College•Chengdu pidu District People's Hospital, 666# Second Section of Deyuan North Road, Pidu District, Chengdu, Sichuan, 611730, China
| | - Xiao-Li Chen
- Department of Radiology, Affiliated Cancer Hospital of Medical School, University of Electronic Science and Technology of China, Sichuan Cancer Hospital, Chengdu, 610000, China
| | - Xin Zhang
- GE Healthcare (China), 1# Tongji South Road, Daxing District, Beijing, 100176, China
| | - Hong Pu
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610072, China
| | - Hang Li
- Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, 32# Second Section of First Ring Road, Qingyang District, Chengdu, Sichuan, 610072, China.
| |
Collapse
|
25
|
Shen Y, Wu S, Wu Y, Cui C, Li H, Yang S, Liu X, Chen X, Huang C, Wang X. Radiomics model building from multiparametric MRI to predict Ki-67 expression in patients with primary central nervous system lymphomas: a multicenter study. BMC Med Imaging 2025; 25:54. [PMID: 39962371 PMCID: PMC11834475 DOI: 10.1186/s12880-025-01585-5] [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: 12/18/2023] [Accepted: 02/10/2025] [Indexed: 02/20/2025] Open
Abstract
OBJECTIVES To examine the correlation of apparent diffusion coefficient (ADC), diffusion weighted imaging (DWI), and T1 contrast enhanced (T1-CE) with Ki-67 in primary central nervous system lymphomas (PCNSL). And to assess the diagnostic performance of MRI radiomics-based machine-learning algorithms in differentiating the high proliferation and low proliferation groups of PCNSL. METHODS 83 patients with PCNSL were included in this retrospective study. ADC, DWI and T1-CE sequences were collected and their correlation with Ki-67 was examined using Spearman's correlation analysis. The Kaplan-Meier method and log-rank test were used to compare the survival rates of the high proliferation and low proliferation groups. The radiomics features were extracted respectively, and the features were screened by machine learning algorithm and statistical method. Radiomics models of seven different sequence permutations were constructed. The area under the receiver operating characteristic curve (ROC AUC) was used to evaluate the predictive performance of all models. DeLong test was utilized to compare the differences of models. RESULTS Relative mean apparent diffusion coefficient (rADCmean) (ρ=-0.354, p = 0.019), relative mean diffusion weighted imaging (rDWImean) (b = 1000) (ρ = 0.273, p = 0.013) and relative mean T1 contrast enhancement (rT1-CEmean) (ρ = 0.385, p = 0.001) was significantly correlated with Ki-67. Interobserver agreements between the two radiologists were almost perfect for all parameters (rADCmean ICC = 0.978, 95%CI 0.966-0.986; rDWImean (b = 1000) ICC = 0.931, 95% CI 0.895-0.955; rT1-CEmean ICC = 0.969, 95% CI 0.953-0.980). The differences in PFS (p = 0.016) and OS (p = 0.014) between the low and high proliferation groups were statistically significant. The best prediction model in our study used a combination of ADC, DWI, and T1-CE achieving the highest AUC of 0.869, while the second ranked model used ADC and DWI, achieving an AUC of 0.828. CONCLUSION rDWImean, rADCmean and rT1-CEmean were correlated with Ki-67. The radiomics model based on MRI sequences combined is promising to distinguish low proliferation PCNSL from high proliferation PCNSL.
Collapse
Affiliation(s)
- Yelong Shen
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China
| | - Siyu Wu
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China
| | - Yanan Wu
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China
| | - Chao Cui
- Qilu Hospital of Shandong University Dezhou Hospital, Dezhou, 253000, Shandong, China
| | - Haiou Li
- Cheeloo College of Medicine, Qilu Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Shuang Yang
- Department of Radiology, The First Affiliated Hospital of Shandong First Medical University& Shandong Provincial Qianfoshan Hospital, Jinan, 250021, Shandong, China
| | - Xuejun Liu
- Department of Radiology, the Affiliated Hospital of Qingdao University, Qingdao, 266000, Shandong, China
| | - Xingzhi Chen
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, 100080, Beijing, China
| | - Chencui Huang
- Department of Research Collaboration, R&D center, Beijing Deepwise & League of PHD Technology Co., Ltd, 100080, Beijing, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital, No. 324, Jingwu Road, Jinan, 250021, Shandong, China.
- Department of Radiology, Shandong University, No. 44, West Wenhua Road, Jinan, 250021, Shandong, China.
| |
Collapse
|
26
|
Ishizu K, Takahashi S, Kouno N, Takasawa K, Takeda K, Matsui K, Nishino M, Hayashi T, Yamagata Y, Matsui S, Yoshikawa T, Hamamoto R. Establishment of a machine learning model for predicting splenic hilar lymph node metastasis. NPJ Digit Med 2025; 8:93. [PMID: 39934302 DOI: 10.1038/s41746-025-01480-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Accepted: 01/25/2025] [Indexed: 02/13/2025] Open
Abstract
Upper gastrointestinal cancer (UGC) sometimes metastasizes to the splenic hilum lymph node (SHLN). However, surgical removal of SHLN is technically difficult, and the risk of postoperative complications is high. Although there are models that predict SHLN metastasis, they usually only provide point estimates of risk, and there is a lack of sufficient information. To address this issue, we aimed to develop a Bayesian logistic regression model called Bayes-SHLNM. The performance of the models was compared with that of the frequentist logistic regression (FLR) model as a benchmark, and the posterior probability distribution (PPD) was shown individually. The performance of Bayes-SHLNM was equivalent to that of the FLR model, and the PPD for each case was visualized as the uncertainty. These results indicate that the Bayes-SHLNM model has the potential to be used as a decision support system in clinical settings where uncertainty is high.
Collapse
Affiliation(s)
- Kenichi Ishizu
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Satoshi Takahashi
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan.
| | - Nobuji Kouno
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Ken Takasawa
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Katsuji Takeda
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan
| | - Kota Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Masashi Nishino
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Tsutomu Hayashi
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Yukinori Yamagata
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Shigeyuki Matsui
- Department of Biostatistics, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, Japan
| | - Takaki Yoshikawa
- Department of Gastric Surgery, National Cancer Center Hospital, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan
| | - Ryuji Hamamoto
- Division of Medical AI Research and Development, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo, 104-0045, Japan.
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, 1-4-1 Nihonbashi, Chuo-ku, Tokyo, 103-0027, Japan.
| |
Collapse
|
27
|
Naemi A, Tashk A, Sorayaie Azar A, Samimi T, Tavassoli G, Bagherzadeh Mohasefi A, Nasiri Khanshan E, Heshmat Najafabad M, Tarighi V, Wiil UK, Bagherzadeh Mohasefi J, Pirnejad H, Niazkhani Z. Applications of Artificial Intelligence for Metastatic Gastrointestinal Cancer: A Systematic Literature Review. Cancers (Basel) 2025; 17:558. [PMID: 39941923 PMCID: PMC11817159 DOI: 10.3390/cancers17030558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/18/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND/OBJECTIVES This systematic literature review examines the application of Artificial Intelligence (AI) in the diagnosis, treatment, and follow-up of metastatic gastrointestinal cancers. METHODS The databases PubMed, Scopus, Embase (Ovid), and Google Scholar were searched for published articles in English from January 2010 to January 2022, focusing on AI models in metastatic gastrointestinal cancers. RESULTS forty-six studies were included in the final set of reviewed papers. The critical appraisal and data extraction followed the checklist for systematic reviews of prediction modeling studies. The risk of bias in the included papers was assessed using the prediction risk of bias assessment tool. CONCLUSIONS AI techniques, including machine learning and deep learning models, have shown promise in improving diagnostic accuracy, predicting treatment outcomes, and identifying prognostic biomarkers. Despite these advancements, challenges persist, such as reliance on retrospective data, variability in imaging protocols, small sample sizes, and data preprocessing and model interpretability issues. These challenges limit the generalizability, clinical application, and integration of AI models.
Collapse
Affiliation(s)
- Amin Naemi
- Nordcee, Department of Biology, University of Southern Denmark, 5230 Odense, Denmark
| | - Ashkan Tashk
- Cognitive Systems, DTU Compute, The Technical University of Denmark (DTU), 2800 Copenhagen, Denmark;
| | - Amir Sorayaie Azar
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Tahereh Samimi
- Student Research Committee, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Department of Medical Informatics, Urmia University of Medical Sciences, Urmia 1138, Iran
| | - Ghanbar Tavassoli
- Department of Computer Engineering, Urmia Branch, Islamic Azad University, Urmia 969, Iran;
| | - Anita Bagherzadeh Mohasefi
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Elaheh Nasiri Khanshan
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Mehrdad Heshmat Najafabad
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Vafa Tarighi
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Uffe Kock Wiil
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
| | - Jamshid Bagherzadeh Mohasefi
- SDU Health Informatics and Technology, The Maersk Mc-Kinney Moller Institute, University of Southern Denmark, 5230 Odense, Denmark; (A.S.A.); (U.K.W.); (J.B.M.)
- Department of Computer Engineering, Urmia University, Urmia 165, Iran; (A.B.M.); (E.N.K.); (M.H.N.); (V.T.)
| | - Habibollah Pirnejad
- Patient Safety Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Department of Family Medicine, Amsterdam University Medical Center, 7057 Amsterdam, The Netherlands
| | - Zahra Niazkhani
- Nephrology and Kidney Transplant Research Center, Clinical Research Institute, Urmia University of Medical Sciences, Urmia 1138, Iran;
- Erasmus School of Health Policy and Management (ESHPM), Erasmus University Rotterdam, 3000 Rotterdam, The Netherlands
| |
Collapse
|
28
|
Yang L, Ding Y, Zhang D, Yang G, Dong X, Zhang Z, Zhang C, Zhang W, Dai Y, Li Z. Predictive value of enhanced CT and pathological indicators in lymph node metastasis in patients with gastric cancer based on GEE model. BMC Med Imaging 2025; 25:36. [PMID: 39894837 PMCID: PMC11789337 DOI: 10.1186/s12880-025-01577-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2023] [Accepted: 01/28/2025] [Indexed: 02/04/2025] Open
Abstract
OBJECTIVES A predictive model was developed based on enhanced computed tomography (CT), laboratory test results, and pathological indicators to achieve the convenient and effective prediction of single lymph node metastasis (LNM) in gastric cancer. METHODS Sixty-six consecutive patients (235 regional lymph nodes) with pathologically confirmed gastric cancer who underwent surgery at our hospital between December 2020 and November 2021 were retrospectively reviewed. They were randomly allocated to training (n = 38, number of lymph nodes = 119) and validation (n = 28, number of lymph nodes = 116) datasets. The clinical data, laboratory test results, enhanced CT characteristics, and pathological indicators from gastroscopy-guided needle biopsies were obtained. Multivariable logistic regression with generalised estimation equations (GEEs) was used to develop a predictive model for LNM in gastric cancer. The predictive performance of the model developed using the training and validation datasets was validated using receiver operating characteristic curves. RESULTS Lymph node enhancement pattern, Ki67 level, and lymph node long-axis diameter were independent predictors of LNM in gastric cancer (p < 0.01). The GEE-logistic model was associated with LNM (p = 0.001). The area under the curve and accuracy of the model, with 95% confidence intervals, were 0.944 (0.890-0.998) and 0.897 (0.813-0.952), respectively, in the training dataset and 0.836 (0.751-0.921) and 0.798 (0.699-0.876), respectively, in the validation dataset. CONCLUSION The predictive model constructed based on lymph node enhancement pattern, Ki67 level, and lymph node long-axis diameter exhibited good performance in predicting LNM in gastric cancer and should aid the lymph node staging of gastric cancer and clinical decision-making.
Collapse
Affiliation(s)
- Ling Yang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Yingying Ding
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Dafu Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Guangjun Yang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Xingxiang Dong
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Zhiping Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Caixia Zhang
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China
| | - Wenjie Zhang
- Department of Gastrointestinal Oncology, Harbin Medical University Cancer Hospital, Harbin, 150086, China.
| | - Youguo Dai
- Department of Gastrointestinal Surgery, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China.
| | - Zhenhui Li
- Department of Radiology, the Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, 650118, China.
| |
Collapse
|
29
|
Chen J, Wei L, Deng CM, Xiong J, Chen SM, Lu D, Li ZH, Chen Y, Xiao J, Chen TW. A liver CT based nomogram to preoperatively predict lung metastasis secondary to hepatic alveolar echinococcosis. Eur J Radiol 2025; 183:111865. [PMID: 39644597 DOI: 10.1016/j.ejrad.2024.111865] [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: 09/29/2024] [Revised: 11/24/2024] [Accepted: 11/28/2024] [Indexed: 12/09/2024]
Abstract
PURPOSE To develop a nomogram based on liver CT and clinical features to preoperatively predict lung metastasis (LM) secondary to hepatic alveolar echinococcosis (HAE). METHODS A total of 291 consecutive HAE patients from Institution A undergoing preoperative abdominal contrast-enhanced CT and chest unenhanced CT were retrospectively reviewed, and were randomly divided into the training and internal validation sets at the 7:3 ratio. 82 consecutive patients from Institution B were enrolled as an external validation set. A nomogram was constructed based on the significant CT and clinical features of HAE from the training set selected by univariable and multivariable analyses to predict LM, and its predictive accuracy was assessed by area under the receiver operating characteristic curve (AUC) and Brier score. Decision-curve analysis was applied to evaluate the clinical effectiveness. This nomogram was verified in two independent validation sets. RESULTS Eosinophil (odds ratio [OR] = 9.60; 95 % confidence interval [CI]: 1.80-51.11), lesion size (OR = 1.02; 95 %CI: 1.01-1.04), and moderate-severe invasion of inferior vena cava (IVC) (OR = 5.57; 95 %CI: 1.82-17.10) were independently associated with LM (all P-values < 0.05). The nomogram based on the three independent predictors displayed AUCs of 0.875 (95 %CI, 0.824-0.927), 0.872 (95 %CI, 0.787-0.957) and 0.836 (95 %CI, 0.729-0.943), and Brier score of 0.105, 0.1 and 0.118 in the training, internal validation and external validation sets, respectively. Decision-curve analysis showed good clinical utility. CONCLUSION A nomogram based on eosinophil, lesion size and moderate-severe invasion of IVC showed good ability and accuracy for preoperative prediction of LM due to HAE.
Collapse
Affiliation(s)
- Jing Chen
- The First Clinical College of Jinan University, Guangzhou 510630, Guangdong, China; Department of Radiology, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, Sichuan, China.
| | - Li Wei
- Department of Radiology, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Chun-Mei Deng
- Department of Radiology, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Jing Xiong
- Department of Radiology, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Song-Mei Chen
- Department of Radiology, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Ding Lu
- Sichuan Provincial Center for Disease Control and Prevention, Chengdu 610044, Sichuan, China.
| | - Zhi-Hong Li
- Department of Hepato-biliary Surgery, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Yao Chen
- Department of Digestive Medical, Ganzi Hospital, West China Hospital of Sichuan University (Ganzi Tibetan Autonomous Prefecture People's Hospital), Ganzi 626000, Sichuan, China.
| | - Jun Xiao
- Department of Pulmonary and Critical Care Medicine, West China Hospital of Sichuan University, Chengdu 610041, Sichuan, China.
| | - Tian-Wu Chen
- The First Clinical College of Jinan University, Guangzhou 510630, Guangdong, China; Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
30
|
Guo X, Chen M, Zhou L, Zhu L, Liu S, Zheng L, Chen Y, Li Q, Xia S, Lu C, Chen M, Chen F, Ji J. Predicting early recurrence in locally advanced gastric cancer after gastrectomy using CT-based deep learning model: a multicenter study. Int J Surg 2025; 111:2089-2100. [PMID: 39715142 DOI: 10.1097/js9.0000000000002184] [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: 07/22/2024] [Accepted: 11/20/2024] [Indexed: 12/25/2024]
Abstract
BACKGROUND Early recurrence in patients with locally advanced gastric cancer (LAGC) portends aggressive biological characteristics and a dismal prognosis. Predicting early recurrence may help determine treatment strategies for LAGC. The goal is to develop a deep learning model for early recurrence prediction (DLER) based on preoperative multiphase computed tomography (CT) images and to further explore the underlying biological basis of the proposed model. MATERIALS AND METHODS In this retrospective study, 620 LAGC patients from January 2015 to March 2023 were included in three medical centers and The Cancer Image Archive (TCIA). The DLER model was developed using DenseNet169 and multiphase 2.5D CT images, and then crucial clinical factors of early recurrence were integrated into the multilayer perceptron (MLP) classifier model (DLER MLP ). The area under the receiver operating characteristic curve (AUC), accuracy, sensitivity, and specificity were applied to measure the performance of different models. The log-rank test was used to analyze survival outcomes. The genetic analysis was performed using RNA-sequencing data from TCIA. RESULTS Using the MLP classifier combined with clinical factors, DLER MLP showed higher performance than DLER and clinical models in predicting early recurrence in the internal validation set (AUC: 0.891 vs. 0.797, 0.752) and two external test sets: test set 1 (0.814 vs. 0.666, 0.808) and test set 2 (0.834 vs. 0.756, 0.766). Early recurrence-free survival, disease-free survival, and overall survival can be stratified using the DLER MLP (all P < 0.001). High DLER MLP score is associated with upregulated tumor proliferation pathways (WNT, MYC, and KRAS signaling) and immune cell infiltration in the tumor microenvironment. CONCLUSION The DLER MLP based on CT images was able to predict early recurrence of patients with LAGC and served as a useful tool for optimizing treatment strategies and monitoring.
Collapse
Affiliation(s)
- Xinyu Guo
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
| | - Mingzhen Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
| | - Lingling Zhou
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
| | - Lingyi Zhu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Shuang Liu
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Liyun Zheng
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Yongjun Chen
- Department of Radiology, the Sixth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Qiang Li
- Department of Radiology, the Affiliated People's Hospital of Ningbo University, Ningbo, China
| | - Shuiwei Xia
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Chenying Lu
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Minjiang Chen
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| | - Feng Chen
- Department of Radiology, the First Affiliated Hospital of Zhejiang University, School of Medicine, Hangzhou, China
| | - Jiansong Ji
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Lishui Hospital, School of Medicine, Zhejiang University, Lishui, China
- Department of Radiology, the Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, China
| |
Collapse
|
31
|
Wang Y, Chen F, Ouyang Z, He S, Qin X, Liang X, Huang W, Wang R, Hu K. MRI-based deep learning and radiomics for predicting the efficacy of PD-1 inhibitor combined with induction chemotherapy in advanced nasopharyngeal carcinoma: A prospective cohort study. Transl Oncol 2025; 52:102245. [PMID: 39662448 PMCID: PMC11697067 DOI: 10.1016/j.tranon.2024.102245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/20/2024] [Accepted: 12/07/2024] [Indexed: 12/13/2024] Open
Abstract
BACKGROUND An increasing number of nasopharyngeal carcinoma (NPC) patients benefit from immunotherapy with chemotherapy as an induction treatment. Currently, there isn't a reliable method to assess the efficacy of this regimen, which hinders informed decision-making for follow-up care. AIM To establish and evaluate a model for predicting the efficacy of programmed death-1 (PD-1) inhibitor combined with GP (gemcitabine and cisplatin) induction chemotherapy based on deep learning features (DLFs) and radiomic features. METHODS Ninety-nine patients diagnosed with advanced NPC were enrolled and randomly divided into training set and test set in a 7:3 ratio. From MRI scans, DLFs and conventional radiomic characteristics were recovered. The random forest algorithm was employed to identify the most valuable features. A prediction model was then created using these radiomic characteristics and DLFs to determine the effectiveness of PD-1 inhibitor combined with GP chemotherapy. The model's performance was assessed using Receiver Operating Characteristic (ROC) curve analysis, area under the curve (AUC), accuracy (ACC), and negative predictive value (NPV). RESULTS Twenty-one prediction models were constructed. The Tf_Radiomics+Resnet101 model, which combines radiomic features and DLFs, demonstrated the best performance. The model's AUC, ACC, and NPV values in the training and test sets were 0.936 (95%CI: 0.827-1.0), 0.9, and 0.923, respectively. CONCLUSION The Tf_Radiomics+Resnet101 model, based on MRI and Resnet101 deep learning, shows a high ability to predict the clinically complete response (cCR) efficacy of PD-1 inhibitor combined with GP in advanced NPC. This model can significantly enhance the treatment management of patients with advanced NPC.
Collapse
Affiliation(s)
- Yiru Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Fuli Chen
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Zhechen Ouyang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Siyi He
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xinling Qin
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Xian Liang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Weimei Huang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China
| | - Rensheng Wang
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China.
| | - Kai Hu
- Department of Radiation Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning 530021, Guangxi, China; Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor (Guangxi Medical University), Ministry of Education, Nanning 530021, Guangxi, China; Guangxi Key Laboratory of Immunology and Metabolism for Liver Diseases, Nanning 530021, Guangxi, China; State Key Laboratory of Targeting Oncology, Guangxi Medical University, Nanning 530021, Guangxi, China.
| |
Collapse
|
32
|
Zhang HY, Aimaiti M, Bai L, Yuan MQ, Zhu CC, Yan JJ, Cai JH, Dong ZY, Zhang ZZ. Bi-phase CT radiomics nomogram for the preoperative prediction of pylorus lymph node metastasis in non-pyloric gastric cancer patients. Abdom Radiol (NY) 2025; 50:608-618. [PMID: 39225717 DOI: 10.1007/s00261-024-04537-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND The expansion of function-preserving surgery became possible due to a more profound understanding of gastric cancer (GC), and T1N + or T2N + gastric cancer patients might be potential beneficiaries. However, ways to evaluate the possibility of function-preserving pylorus surgery are still unknown. METHODS A total of 288 patients at Renji Hospital and 58 patients at Huadong Hospital, pathologically diagnosed with gastric cancer staging at T1 and T2 with tumors located in the upper two-thirds of the stomach, were retrospectively enrolled from March 2015 to October 2022. Tumor regions of interest (ROIs) were manually delineated on bi-phase CT images, and a nomogram was built and evaluated. RESULTS The radiomic features distributed differently between positive and negative pLNm groups. Two radiomic signatures (RS1 and RS2) and one clinical signature were constructed. The radiomic signatures exhibited good performance for discriminating pLNm status in the test set. The three signatures were then combined into an integrated nomogram (IN). The IN showed good discrimination of pLNm in the Renji cohort (AUC 0.918) and the Huadong cohort (AUC 0.649). The verification models showed high values. CONCLUSION For GC patients with T1 and T2 tumors located in the upper two-thirds of the stomach, a nomogram was successfully built for predicting pylorus lymph node metastasis, which would guide the surgical indication extension of conservative gastrectomies.
Collapse
Affiliation(s)
- Hao-Yu Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Muerzhate Aimaiti
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Long Bai
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Meng-Qing Yuan
- The Hongkong University of Science and Technology, Hongkong, China
| | - Chun-Chao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jia-Jun Yan
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China
| | - Jian-Hua Cai
- Department of General Surgery, Huadong Hospital Affiliated to Fudan University, Shanghai, China.
| | - Zhong-Yi Dong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| | - Zi-Zhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, China.
| |
Collapse
|
33
|
Heydari S, Peymani M, Hashemi M, Ghaedi K, Entezari M. Potential prognostic and predictive biomarkers: METTL5, METTL7A, and METTL7B expression in gastrointestinal cancers. Mol Biol Rep 2025; 52:151. [PMID: 39847131 DOI: 10.1007/s11033-024-10207-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 12/29/2024] [Indexed: 01/24/2025]
Abstract
BACKGROUND The methyltransferase gene family is known for its diverse biological functions and critical role in tumorigenesis. This study aimed to identify these family genes in common gastrointestinal (GI) cancers using comprehensive methodologies. METHODS Gene identification involved analysis of scientific literature and insights from The Cancer Genome Atlas (TCGA) database. RNA sequencing (RNA-seq) data for colon, gastric, pancreatic, esophageal, and liver cancers were collected, processed, and normalized. Differential expression analysis was conducted using R software with the Limma package. Additionally, real-time PCR analysis was performed on 30 tumor and 30 normal tissue samples from patients with colon and gastric cancer. Pathway analysis was conducted via the EnrichR web tool, while survival analysis used Cox regression methods, and biomarker potential was assessed with the pROC package. Prognostic significance was evaluated by examining associations between gene expression, patient survival, and recurrence rates. The study also investigated diagnostic potential through receiver operating characteristic (ROC) analysis, and assessed how small molecules affect gene expression, with implications for drug resistance and sensitivity, analyzed via CCLE and GDSC datasets. RESULTS Findings revealed METTL5 overexpression in colon, liver, esophagus, and pancreas cancers, while METTL7A was underexpressed in gastric, esophagus, liver, and colon cancers. METTL7B expression varied, being higher in gastric and esophagus cancers but lower in liver and colon cancers. Enrichment analysis identified pathways related to these genes, and survival analysis associated altered METTL7A and METTL5 expressions with poor prognosis and higher recurrence rates. CONCLUSIONS These findings suggest that METTL genes could serve as predictive biomarkers in GI cancers, offering potential implications for patient prognosis and treatment response.
Collapse
Affiliation(s)
- Soraya Heydari
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran
| | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Shahrekord, Iran.
| | - Mehrdad Hashemi
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Kamran Ghaedi
- Division of Cellular and Molecular Biology, Department of Biology, Faculty of Sciences, University of Isfahan, Isfahan, Iran
| | - Maliheh Entezari
- Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| |
Collapse
|
34
|
Cai ZR, Zheng YQ, Hu Y, Ma MY, Wu YJ, Liu J, Yang LP, Zheng JB, Tian T, Hu PS, Liu ZX, Zhang L, Xu RH, Ju HQ. Construction of exosome non-coding RNA feature for non-invasive, early detection of gastric cancer patients by machine learning: a multi-cohort study. Gut 2025:gutjnl-2024-333522. [PMID: 39753334 DOI: 10.1136/gutjnl-2024-333522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 12/08/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND AND OBJECTIVE Gastric cancer (GC) remains a prevalent and preventable disease, yet accurate early diagnostic methods are lacking. Exosome non-coding RNAs (ncRNAs), a type of liquid biopsy, have emerged as promising diagnostic biomarkers for various tumours. This study aimed to identify a serum exosome ncRNA feature for enhancing GC diagnosis. DESIGNS Serum exosomes from patients with GC (n=37) and healthy donors (n=20) were characterised using RNA sequencing, and potential biomarkers for GC were validated through quantitative reverse transcription PCR (qRT-PCR) in both serum exosomes and tissues. A combined diagnostic model was developed using LASSO-logistic regression based on a cohort of 518 GC patients and 460 healthy donors, and its diagnostic performance was evaluated via receiver operating characteristic curves. RESULTS RNA sequencing identified 182 candidate biomarkers for GC, of which 31 were validated as potential biomarkers by qRT-PCR. The combined diagnostic score (cd-score), derived from the expression levels of four long ncRNAs (RP11.443C10.1, CTD-2339L15.3, LINC00567 and DiGeorge syndrome critical region gene (DGCR9)), was found to surpass commonly used biomarkers, such as carcinoembryonic antigen, carbohydrate antigen 19-9 (CA19-9) and CA72-4, in distinguishing GC patients from healthy donors across training, testing and external validation cohorts, with AUC values of 0.959, 0.942 and 0.949, respectively. Additionally, the cd-score could effectively identify GC patients with negative gastrointestinal tumour biomarkers and those in early-stage. Furthermore, molecular biological assays revealed that knockdown of DGCR9 inhibited GC tumour growth. CONCLUSIONS Our proposed serum exosome ncRNA feature provides a promising liquid biopsy approach for enhancing the early diagnosis of GC.
Collapse
Affiliation(s)
- Ze-Rong Cai
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yong-Qiang Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Yan Hu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Meng-Yao Ma
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, People's Republic of China
| | - Yi-Jin Wu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lu-Ping Yang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Jia-Bo Zheng
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Tian Tian
- Department of Medical Biochemistry and Molecular Biology, School of Medicine, Jinan University, Guangzhou, People's Republic of China
| | - Pei-Shan Hu
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, People's Republic of China
| | - Ze-Xian Liu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Lin Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Rui-Hua Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
| | - Huai-Qiang Ju
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, People's Republic of China
- Department of Clinical Oncology, Shenzhen Key Laboratory for Cancer Metastasis and Personalized Therapy, The University of Hong Kong-Shenzhen Hospital, Shenzhen, People's Republic of China
| |
Collapse
|
35
|
Shi B, Wang W, Fang S, Wu S, Zhu L, Chen Y, Dong H, Yan F, Yuan F, Ye J, Zhang H, Lin LL. Raman spectroscopy analysis combined with computed tomography imaging to identify microsatellite instability in gastric cancers. SPECTROCHIMICA ACTA. PART A, MOLECULAR AND BIOMOLECULAR SPECTROSCOPY 2025; 325:125062. [PMID: 39226670 DOI: 10.1016/j.saa.2024.125062] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/05/2024] [Accepted: 08/25/2024] [Indexed: 09/05/2024]
Abstract
Accurate determination of microsatellite instability (MSI) status is critical for tailoring treatment approaches for gastric cancer patients. Existing clinical techniques for MSI diagnosis are plagued by problems of suboptimal time efficiency, high cost, and burdensome experimental requirements. Here, we for the first time establish the classification model of gastric cancer MSI status based on Raman spectroscopy. To begin with, we reveal that tumor heterogeneity-induced signal variations pose a prominent impact on MSI classification. To eliminate this issue, we develop Euclidean distance-based Raman Spectroscopy (EDRS) algorithm, which establishes a standard spectrum to represent the "most microsatellite stable" status. The similarity between each spectrum of tissues with the standard spectrum is calculated to provide a direct assessment on the MSI status. Compared to machine learning-algorithms including k-Nearest Neighbors, Random Forest, and Extreme Learning Machine, the EDRS method shows the highest accuracy of 94.6 %. Finally, we integrate the EDRS method with the clinical diagnostic modality, computed tomography, to construct an innovative joint classification model with good classification performance (AUC = 0.914, Accuracy = 94.6 %). Our work demonstrates a robust, rapid, non-invasive, and convenient tool in identifying the MSI status, and opens new avenues for Raman techniques to fit into existing clinical workflow.
Collapse
Affiliation(s)
- Bowen Shi
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Wenfang Wang
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai 200040, PR China
| | - Shiyan Fang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Siyi Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Lan Zhu
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Yong Chen
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Haipeng Dong
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Fuhua Yan
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Fei Yuan
- Department of Pathology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China
| | - Jian Ye
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China; Institute of Medical Robotics, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200025, PR China.
| | - Linley Li Lin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| |
Collapse
|
36
|
Wang X, Quan T, Chu X, Gao M, Zhang Y, Chen Y, Bai G, Chen S, Wei M. Deep Learning Radiomics Nomogram Based on MRI for Differentiating between Borderline Ovarian Tumors and Stage I Ovarian Cancer: A Multicenter Study. Acad Radiol 2025:S1076-6332(24)01055-9. [PMID: 39814661 DOI: 10.1016/j.acra.2024.12.067] [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: 10/31/2024] [Revised: 12/24/2024] [Accepted: 12/28/2024] [Indexed: 01/18/2025]
Abstract
RATIONALE AND OBJECTIVES To develop and validate a deep learning radiomics nomogram (DLRN) based on T2-weighted MRI to distinguish between borderline ovarian tumors (BOTs) and stage I epithelial ovarian cancer (EOC) preoperatively. MATERIALS AND METHODS This retrospective multicenter study enrolled 279 patients from three centers, divided into a training set (n = 207) and an external test set (n = 72). The intra- and peritumoral radiomics analysis was employed to develop a combined radiomics model. A deep learning model was constructed based on the largest orthogonal slices of the tumor volume, and a clinical model was constructed using independent clinical predictors. The DLRN was then constructed by integrating deep learning, intra- and peritumoral radiomics, and clinical predictors. For comparison, an original radiomics model based solely on tumor volume (excluding the peritumoral area) was also constructed. All models were validated through 10-fold cross-validation and external testing, and their predictive performance was evaluated by the area under the receiver operating characteristic curve (AUC). RESULTS The DLRN demonstrated superior performance across the 10-fold cross-validation, with the highest AUC of 0.825±0.082. On the external test set, the DLRN significantly outperformed the clinical model and the original radiomics model (AUC = 0.819 vs. 0.708 and 0.670, P = 0.047 and 0.015, respectively). Furthermore, the combined radiomics model performed significantly better than the original radiomics model (AUC = 0.778 vs. 0.670, P = 0.043). CONCLUSION The DLRN exhibited promising performance in distinguishing BOTs from stage I EOC preoperatively, thus potentially assisting clinical decision-making.
Collapse
Affiliation(s)
- Xinyi Wang
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China (X.W., X.C., Y.C., S.C., M.W.)
| | - Tao Quan
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (T.Q.)
| | - Xiao Chu
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China (X.W., X.C., Y.C., S.C., M.W.)
| | - Max Gao
- Computer Science and Engineering, University of California, Davis, Sacramento, CA (M.G.)
| | - Yu Zhang
- Department of Radiology, The Fourth Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China (Y.Z.)
| | - Ying Chen
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China (X.W., X.C., Y.C., S.C., M.W.)
| | - Genji Bai
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China (G.B.)
| | - Shuangqing Chen
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China (X.W., X.C., Y.C., S.C., M.W.)
| | - Mingxiang Wei
- Department of Radiology, the Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou, Jiangsu, China (X.W., X.C., Y.C., S.C., M.W.).
| |
Collapse
|
37
|
Xiao Y, Zhu J, Xie H, Wang Z, Huang Z, Su M. Intratumoral and peritumoral radiomics for forecasting microsatellite status in gastric cancer: a multicenter study. BMC Cancer 2025; 25:66. [PMID: 39794732 PMCID: PMC11724602 DOI: 10.1186/s12885-025-13450-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025] Open
Abstract
OBJECTIVE This investigation attempted to examine the effectiveness of CT-derived peritumoral and intratumoral radiomics in forecasting microsatellite instability (MSI) status preoperatively among gastric cancer (GC) patients. METHODS A retrospective analysis was performed on GC patients from February 2019 to December 2023 across three healthcare institutions. 364 patients (including 41 microsatellite instability-high (MSI-H) and 323 microsatellite instability-low/stable (MSI-L/S)) were stratified into a training set (n = 202), an internal validation set (n = 84), and an external validation set (n = 78). Radiomics features were obtained from both the intratumoral region (IR) and the intratumoral plus 3-mm peritumoral region (IPR) on preoperative contrast-enhanced CT images. After standardizing and reducing the dimensionality of these features, six radiomic models were constructed utilizing three machine learning techniques: Support Vector Machine (SVM), Linear Support Vector Classification (LinearSVC), and Logistic Regression (LR). The optimal model was determined by evaluating the Receiver Operating Characteristic (ROC) curve's Area Under the Curve (AUC), and the radiomics score (Radscore) was computed. A clinical model was developed using clinical characteristics and CT semantic features, with the Radscore integrated to create a combined model. Used ROC curves, calibration plots, and Decision Curve Analysis (DCA) to assess the performance of radiomics, clinical, and combined models. RESULTS The LinearSVC model using the IPR achieved the highest AUC of 0.802 in the external validation set. The combined model yielded superior AUCs in internal and external validation sets (0.891 and 0.856) in comparison to clinical model [(0.724, P = 0.193) and (0.655, P = 0.072)] and radiomics model [(0.826, P = 0.160) and (0.802, P = 0.068)]. Furthermore, results from calibration and DCA underscored the model's suitability and clinical relevance. CONCLUSION The combined model, which integrates IPR radiomics with clinical characteristics, accurately predicts MSI status and supports the development of personalized treatment strategies.
Collapse
Affiliation(s)
- Yunzhou Xiao
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China
| | - Jianping Zhu
- Department of Radiology, Ningbo Yinzhou NO.2 Hospital, Ningbo, 315100, China
| | - Huanhuan Xie
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Zhongchu Wang
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China
| | - Zhaohai Huang
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China.
| | - Miaoguang Su
- Department of Radiology, The People's Hospital of PingYang, Wenzhou Medical University, Wenzhou, 325400, China.
| |
Collapse
|
38
|
Zhang Y, Zhao F, Guo J, Liu Y, Cai M, Ding X, Li B, Zhang L, Zhang R, Deng J. The clinical significance assessment of the transverse lymph node metastasis in gastric cancer: The establishment and validation of nomogram from a single clinical medical center. Dig Liver Dis 2025; 57:125-133. [PMID: 39034188 DOI: 10.1016/j.dld.2024.07.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Revised: 06/22/2024] [Accepted: 07/07/2024] [Indexed: 07/23/2024]
Abstract
BACKGROUND Lymph node metastasis is an important route for gastric cancer metastasis. The clinical significance of transverse lymph node metastasis (TLNM) is still unclear. AIMS This study investigates effects of TLNM on the prognosis of GC patients and establishes two nomograms for evaluating the prognosis of GC patients and for predicting the risk clinicopathological factors to TLNM based on a Chinese medical database. METHODS A total of 902 GC patients with lymph node metastasis (LNM) who underwent R0 gastrectomy was included in this study. According to results of Cox proportional hazards analyses and logistic regression analyses, the prognostic and the predictive nomograms were established and validated. RESULTS The overall survival of patients with TLNM was significantly worse than those without TLNM (P < 0.001) and similar to patients with extra-gastric LNM (P > 0.05). TLNM independently influenced prognosis of GC patients. Prognostic and predictive nomograms were established and validated. Both nomograms were proven that have high accuracy by calculating each AUC (Area Under Cure) value. Calibration curves aligned well with actual outcomes. DCA (Decision Curve Analyses) analyses indicated the high clinical utility. CONCLUSION These nomograms offer precise survival and TLNM occurrence predictions, which may aid clinical decisions.
Collapse
Affiliation(s)
- Yizhao Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Fucheng Zhao
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jiamei Guo
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Yong Liu
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Mingzhi Cai
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Xuewei Ding
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Bin Li
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Li Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Rupeng Zhang
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jingyu Deng
- Department of Gastric Surgery, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy at Tianjin, Tianjin's Clinical Research Center for Cancer, Tianjin, China.
| |
Collapse
|
39
|
Chen C, Luo Y, Hou Q, Qiu J, Yuan S, Deng K. A vision transformer-based deep transfer learning nomogram for predicting lymph node metastasis in lung adenocarcinoma. Med Phys 2025; 52:375-387. [PMID: 39341208 DOI: 10.1002/mp.17414] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2024] [Revised: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 09/30/2024] Open
Abstract
BACKGROUND Lymph node metastasis (LNM) plays a crucial role in the management of lung cancer; however, the ability of chest computed tomography (CT) imaging to detect LNM status is limited. PURPOSE This study aimed to develop and validate a vision transformer-based deep transfer learning nomogram for predicting LNM in lung adenocarcinoma patients using preoperative unenhanced chest CT imaging. METHODS This study included 528 patients with lung adenocarcinoma who were randomly divided into training and validation cohorts at a 7:3 ratio. The pretrained vision transformer (ViT) was utilized to extract deep transfer learning (DTL) feature, and logistic regression was employed to construct a ViT-based DTL model. Subsequently, the model was compared with six classical convolutional neural network (CNN) models. Finally, the ViT-based DTL signature was combined with independent clinical predictors to construct a ViT-based deep transfer learning nomogram (DTLN). RESULTS The ViT-based DTL model showed good performance, with an area under the curve (AUC) of 0.821 (95% CI, 0.775-0.867) in the training cohort and 0.825 (95% CI, 0.758-0.891) in the validation cohort. The ViT-based DTL model demonstrated comparable performance to classical CNN models in predicting LNM, and the ViT-based DTL signature was then used to construct ViT-based DTLN with independent clinical predictors such as tumor maximum diameter, location, and density. The DTLN achieved the best predictive performance, with AUCs of 0.865 (95% CI, 0.827-0.903) and 0.894 (95% CI, 0845-0942), respectively, surpassing both the clinical factor model and the ViT-based DTL model (p < 0.001). CONCLUSION This study developed a new DTL model based on ViT to predict LNM status in lung adenocarcinoma patients and revealed that the performance of the ViT-based DTL model was comparable to that of classical CNN models, confirming that ViT was viable for deep learning tasks involving medical images. The ViT-based DTLN performed exceptionally well and can assist clinicians and radiologists in making accurate judgments and formulating appropriate treatment plans.
Collapse
Affiliation(s)
- Chuanyu Chen
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Yi Luo
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Qiuyang Hou
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Jun Qiu
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Shuya Yuan
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| | - Kexue Deng
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
40
|
Xiao ML, Fu L, Qian T, Wei Y, Ma FH, Li YA, Cheng JJ, Qian ZX, Zhang GF, Qiang JW. The deep learning radiomics nomogram helps to evaluate the lymph node status in cervical adenocarcinoma/adenosquamous carcinoma. Front Oncol 2024; 14:1414609. [PMID: 39735600 PMCID: PMC11671353 DOI: 10.3389/fonc.2024.1414609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 11/20/2024] [Indexed: 12/31/2024] Open
Abstract
Objectives The accurate assessment of lymph node metastasis (LNM) can facilitate clinical decision-making on radiotherapy or radical hysterectomy (RH) in cervical adenocarcinoma (AC)/adenosquamous carcinoma (ASC). This study aims to develop a deep learning radiomics nomogram (DLRN) to preoperatively evaluate LNM in cervical AC/ASC. Materials and methods A total of 652 patients from a multicenter were enrolled and randomly allocated into primary, internal, and external validation cohorts. The radiomics features were extracted from axial T2-weighted imaging (T2WI), diffusion-weighted imaging (DWI), and contrast-enhanced T1-weighted imaging (CE-T1WI). The DL features from T2WI, DWI, and CE-T1WI were exported from Resnet 34, which was pretrained by 14 million natural images of the ImageNet dataset. The radscore (RS) and DL score (DLS) were independently obtained after repeatability test, Pearson correlation coefficient (PCC), minimum redundancy maximum relevance (MRMR), and least absolute shrinkage and selection operator (LASSO) algorithm performed on the radiomics and DL feature sets. The DLRN was then developed by integrating the RS, DLS, and independent clinicopathological factors for evaluating the LNM in cervical AC/ASC. Results The nomogram of DLRN-integrated FIGO stage, menopause, RS, and DLS achieved AUCs of 0.79 (95% CI, 0.74-0.83), 0.87 (95% CI, 0.81-0.92), and 0.86 (95% CI, 0.79-0.91) in the primary, internal, and external validation cohorts. Compared with the RS, DLS, and clinical models, DLRN had a significant higher AUC for evaluating LNM (all P < 0.005). Conclusions The nomogram of DLRN can accurately evaluate LNM in cervical AC/ASC.
Collapse
Affiliation(s)
- Mei Ling Xiao
- Department of Nuclear Medicine and PET Center, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, China
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Le Fu
- Department of Radiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Ting Qian
- Department of Radiology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Yan Wei
- College of Information Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, China
| | - Feng Hua Ma
- Departments of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Yong Ai Li
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| | - Jie Jun Cheng
- Department of Radiology, Shanghai First Maternity and Infant Hospital, Tongji University School of Medicine, Shanghai, China
| | - Zhao Xia Qian
- Department of Radiology, International Peace Maternity and Child Health Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | - Guo Fu Zhang
- Departments of Radiology, Obstetrics and Gynecology Hospital, Fudan University, Shanghai, China
| | - Jin Wei Qiang
- Department of Radiology, Jinshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
41
|
Fan Z, Wu T, Wang Y, Jin Z, Wang T, Liu D. Deep-Learning-Based Radiomics to Predict Surgical Risk Factors for Lumbar Disc Herniation in Young Patients: A Multicenter Study. J Multidiscip Healthc 2024; 17:5831-5851. [PMID: 39664265 PMCID: PMC11633295 DOI: 10.2147/jmdh.s493302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/25/2024] [Indexed: 12/13/2024] Open
Abstract
Objective The aim of this study is to develop and validate a deep-learning radiomics model for predicting surgical risk factors for lumbar disc herniation (LDH) in young patients to assist clinicians in identifying surgical candidates, alleviating symptoms, and improving prognosis. Methods A retrospective analysis of patients from two medical centers was conducted. From sagittal and axial MR images, the regions of interest were handcrafted to extract radiomics features. Various machine-learning algorithms were employed and combined with clinical features, resulting in the development of a deep-learning radiomics nomogram (DLRN) to predict surgical risk factors for LDH in young adults. The efficacy of the different models and the clinical benefits of the model were compared. Results We derived six sets of features, including clinical features, radiomics features (Rad_SAG and Rad_AXI) and deep learning features (DL_SAG and DL_AXI) from sagittal and axial MR images, as well as fused deep-learning radiomics (DLR) features. The support vector machine(SVM) algorithm exhibited the best performance. The area under the curve (AUC) of DLR in the training and testing cohorts of 0.991 and 0.939, respectively, were significantly better than those of the models developed with radiomics(Rad_SAG=0.914 and 0.863, Rad_AXI=0.927 and 0.85) and deep-learning features(DL_SAG=0.959 and 0.818, DL_AXI=0.960 and 0.811). The AUC of DLRN coupled with clinical features(ODI, Pfirrmann grade, SLRT, MMFI, and MSU classification) were 0.994 and 0.941 in the training and testing cohorts, respectively. Analysis of the calibration and decision curves demonstrated good agreement between the predicted and observed outcomes, and the use of the DLRN to predict the need for surgical treatment of LDH demonstrated significant clinical benefits. Conclusion The DLRN established based on clinical and DLR features effectively predicts surgical risk factors for LDH in young adults, offering valuable insights for diagnosis and treatment.
Collapse
Affiliation(s)
- Zheng Fan
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Tong Wu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Yang Wang
- Department of Orthopedics, China Medical University Shenyang Fourth People’s Hospital, Shenyang, People’s Republic of China
| | - Zhuoru Jin
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Tong Wang
- Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| | - Da Liu
- Department of Orthopedics, Shengjing Hospital of China Medical University, Shenyang, People’s Republic of China
| |
Collapse
|
42
|
Zheng HD, Tian YC, Huang QY, Huang QM, Ke XT, Xu JH, Liang XY, Lin S, Ye K. Enhancing lymph node metastasis prediction in adenocarcinoma of the esophagogastric junction: A study combining radiomic with clinical features. Med Phys 2024; 51:9057-9070. [PMID: 39207288 DOI: 10.1002/mp.17374] [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: 01/09/2024] [Revised: 08/10/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024] Open
Abstract
BACKGROUND The incidence of adenocarcinoma of the esophagogastric junction (AEJ) is increasing, and with poor prognosis. Lymph node status (LNs) is particularly important for planning treatment and evaluating the prognosis of patients with AEJ. However, the use of radiomic based on enhanced computed tomography (CT) to predict the preoperative lymph node metastasis (PLNM) status of the AEJ has yet to be reported. PURPOSE We sought to investigate the value of radiomic features based on enhanced CT in the accurate prediction of PLNM in patients with AEJ. METHODS Clinical features and enhanced CT data of 235 patients with AEJ from October 2017 to May 2023 were retrospectively analyzed. The data were randomly assigned to the training cohort (n = 164) or the external testing cohort (n = 71) at a ratio of 7:3. A CT-report model, clinical model, radiomic model, and radiomic-clinical combined model were developed to predict PLNM in patients with AEJ. Univariate and multivariate logistic regression were used to screen for independent clinical risk factors. Least absolute shrinkage and selection operator (LASSO) regression was used to select the radiomic features. Finally, a nomogram for the preoperative prediction of PLNM in AEJ was constructed by combining Radiomics-score and clinical risk factors. The models were evaluated by area under the receiver operating characteristic curve (AUC-ROC), calibration curve, and decision curve analyses. RESULTS A total of 181 patients (181/235, 77.02%) had LNM. In the testing cohort, the AUC of the radiomic-clinical model was 0.863 [95% confidence interval (CI) = 0.738-0.957], and the radiomic model (0.816; 95% CI = 0.681-0.929), clinical model (0.792; 95% CI = 0.677-0.888), and CT-report model (0.755; 95% CI = 0.647-0.840). CONCLUSION The radiomic-clinical model is a feasible method for predicting PLNM in patients with AEJ, helping to guide clinical decision-making and personalized treatment planning.
Collapse
Affiliation(s)
- Hui-da Zheng
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Yu-Chi Tian
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shenyang, China
| | - Qiao-Yi Huang
- Department of Gynaecology and Obstetrics, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Qi-Ming Huang
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiao-Ting Ke
- Department of CT/MRI, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Jian-Hua Xu
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Xiao-Yun Liang
- Institute of Research and Clinical Innovations, Neusoft Medical Systems Co., Ltd, Shenyang, China
| | - Shu Lin
- Centre of Neurological and Metabolic Research, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Kai Ye
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| |
Collapse
|
43
|
Li S, Wei X, Wang L, Zhang G, Jiang L, Zhou X, Huang Q. Dual-source dual-energy CT and deep learning for equivocal lymph nodes on CT images for thyroid cancer. Eur Radiol 2024; 34:7567-7579. [PMID: 38904758 DOI: 10.1007/s00330-024-10854-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2023] [Revised: 04/08/2024] [Accepted: 04/23/2024] [Indexed: 06/22/2024]
Abstract
OBJECTIVES This study investigated the diagnostic performance of dual-energy computed tomography (CT) and deep learning for the preoperative classification of equivocal lymph nodes (LNs) on CT images in thyroid cancer patients. METHODS In this prospective study, from October 2020 to March 2021, 375 patients with thyroid disease underwent thin-section dual-energy thyroid CT at a small field of view (FOV) and thyroid surgery. The data of 183 patients with 281 LNs were analyzed. The targeted LNs were negative or equivocal on small FOV CT images. Six deep-learning models were used to classify the LNs on conventional CT images. The performance of all models was compared with pathology reports. RESULTS Of the 281 LNs, 65.5% had a short diameter of less than 4 mm. Multiple quantitative dual-energy CT parameters significantly differed between benign and malignant LNs. Multivariable logistic regression analyses showed that the best combination of parameters had an area under the curve (AUC) of 0.857, with excellent consistency and discrimination, and its diagnostic accuracy and sensitivity were 74.4% and 84.2%, respectively (p < 0.001). The visual geometry group 16 (VGG16) based model achieved the best accuracy (86%) and sensitivity (88%) in differentiating between benign and malignant LNs, with an AUC of 0.89. CONCLUSIONS The VGG16 model based on small FOV CT images showed better diagnostic accuracy and sensitivity than the spectral parameter model. Our study presents a noninvasive and convenient imaging biomarker to predict malignant LNs without suspicious CT features in thyroid cancer patients. CLINICAL RELEVANCE STATEMENT Our study presents a deep-learning-based model to predict malignant lymph nodes in thyroid cancer without suspicious features on conventional CT images, which shows better diagnostic accuracy and sensitivity than the regression model based on spectral parameters. KEY POINTS Many cervical lymph nodes (LNs) do not express suspicious features on conventional computed tomography (CT). Dual-energy CT parameters can distinguish between benign and malignant LNs. Visual geometry group 16 model shows superior diagnostic accuracy and sensitivity for malignant LNs.
Collapse
Affiliation(s)
- Sheng Li
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
- Guangdong Esophageal Cancer Institute, Guangzhou, 510060, China
| | - Xiaoting Wei
- Department of Radiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518036, China
| | - Li Wang
- School of Artificial Intelligence, Optics and Electronics (iOPEN), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Guizhi Zhang
- Department of Radiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518036, China
| | - Linling Jiang
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, 510060, China
| | - Xuhui Zhou
- Department of Radiology, The Eighth Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518036, China.
| | - Qinghua Huang
- School of Artificial Intelligence, Optics and Electronics (iOPEN), Northwestern Polytechnical University, Xi'an, 710072, China.
| |
Collapse
|
44
|
Sun J, Wang Z, Zhu H, Yang Q, Sun Y. Advanced Gastric Cancer: CT Radiomics Prediction of Lymph Modes Metastasis After Neoadjuvant Chemotherapy. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:2910-2919. [PMID: 38886288 PMCID: PMC11612076 DOI: 10.1007/s10278-024-01148-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 05/16/2024] [Accepted: 05/20/2024] [Indexed: 06/20/2024]
Abstract
This study aims to create and assess machine learning models for predicting lymph node metastases following neoadjuvant treatment in advanced gastric cancer (AGC) using baseline and restaging computed tomography (CT). We evaluated CT images and pathological data from 158 patients with resected stomach cancer from two institutions in this retrospective analysis. Patients were eligible for inclusion if they had histologically proven gastric cancer. They had received neoadjuvant chemotherapy, with at least 15 lymph nodes removed. All patients received baseline and preoperative abdominal CT and had complete clinicopathological reports. They were divided into two cohorts: (a) the primary cohort (n = 125) for model creation and (b) the testing cohort (n = 33) for evaluating models' capacity to predict the existence of lymph node metastases. The diagnostic ability of the radiomics-model for lymph node metastasis was compared to traditional CT morphological diagnosis by radiologist. The radiomics model based on the baseline and preoperative CT images produced encouraging results in the training group (AUC 0.846) and testing cohort (AUC 0.843). In the training cohort, the sensitivity and specificity were 81.3% and 77.8%, respectively, whereas in the testing cohort, they were 84% and 75%. The diagnostic sensitivity and specificity of the radiologist were 70% and 42.2% (using baseline CT) and 46.3% and 62.2% (using preoperative CT). In particular, the specificity of radiomics model was higher than that of conventional CT in diagnosing N0 cases (no lymph node metastasis). The CT-based radiomics model could assess lymph node metastasis more accurately than traditional CT imaging in AGC patients following neoadjuvant chemotherapy.
Collapse
Affiliation(s)
- Jia Sun
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 GongtiSouth Road, Chaoyang District, Beijing, Beijing, 100020, China
| | - Zhilong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Haitao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China
| | - Qi Yang
- Department of Radiology, Beijing Chao-Yang Hospital, Capital Medical University, No. 8 GongtiSouth Road, Chaoyang District, Beijing, Beijing, 100020, China.
| | - Yingshi Sun
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Radiology, Peking University Cancer Hospital & Institute, Beijing, 100142, China.
| |
Collapse
|
45
|
Zheng H, Zheng H, Wei L, Xue Z, Xu B, Hu M, Yu J, Xie R, Zhang L, Zheng Z, Xie J, Zheng C, Huang C, Lin J, Li P. Risk stratification models incorporating oxidative stress factors to predict survival and recurrence in patients with gastric cancer after radical gastrectomy: A real-world multicenter study. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2024; 50:108658. [PMID: 39244978 DOI: 10.1016/j.ejso.2024.108658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/13/2024] [Accepted: 09/02/2024] [Indexed: 09/10/2024]
Abstract
BACKGROUND Oxidative stress significantly influences the development and progression of gastric cancer (GC). It remains unreported whether incorporating oxidative stress factors into nomograms can improve the predictive accuracy for survival and recurrence risk in GC patients. METHODS 3498 GC patients who underwent radical gastrectomy between 2009 and 2017 were enrolled and randomly divided into training cohort (TC) and internal validation cohort (IVC). Cox regression analysis model was used to evaluate six preoperative oxidative stress indicators to formulate the Systemic oxidative stress Score (SOSS). Two nomograms based on SOSS was constructed by multivariate Cox regression and validated using 322 patients from another two hospitals. RESULTS A total of 3820 patients were included. The SOSS, composed of three preoperative indicators-fibrinogen, albumin, and cholesterol-was an independent prognostic factor for both overall survival (OS) and disease-free survival (DFS). The two nomograms based on SOSS showed a significantly higher AUC than the pTNM stage (OS: 0.830 vs. 0.778, DFS: 0.824 vs. 0.775, all P < 0.001) and were validated in the IVC and EVC (all P < 0.001). The local recurrence rate, peritoneal recurrence rate, distant recurrence rate and multiple recurrence rate in high-risk group were significantly higher than those in low-risk group (P < 0.05). CONCLUSIONS The two novel nomograms based on SOSS which was a combination score of three preoperative blood indicators, demonstrated outstanding predictive abilities for both survival and recurrence in GC patients with different risk groups, which may potentially improve survival through perioperatively active intervention strategies and individualized postoperatively close surveillance.
Collapse
Affiliation(s)
- Honghong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Hualong Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Linghua Wei
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Zhen Xue
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Binbin Xu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Minggao Hu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, The PLA Navy Anqing Hospital, Anqing, 246000, China
| | - Junhua Yu
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou, 324000, China
| | - Rongzhen Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Department of General Surgery, First Affiliated Hospital of Gannan Medical University, Ganzhou, 321000, China
| | - Lingkang Zhang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Zhiwei Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Jianwei Xie
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - Chaohui Zheng
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China
| | - ChangMing Huang
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| | - Jianxian Lin
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| | - Ping Li
- Department of Gastric Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, China; Key Laboratory of Ministry of Education of Gastrointestinal Cancer, Fujian Medical University, Fuzhou, 350000, China; Fujian Key Laboratory of Tumor Microbiology, Fujian Medical University, Fuzhou, 350000, China; Fujian Province Minimally Invasive Medical Center, Fuzhou, 350000, China.
| |
Collapse
|
46
|
Liao NQ, Deng ZJ, Wei W, Lu JH, Li MJ, Ma L, Chen QF, Zhong JH. Deep learning of pretreatment multiphase CT images for predicting response to lenvatinib and immune checkpoint inhibitors in unresectable hepatocellular carcinoma. Comput Struct Biotechnol J 2024; 24:247-257. [PMID: 38617891 PMCID: PMC11015163 DOI: 10.1016/j.csbj.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2023] [Revised: 04/01/2024] [Accepted: 04/01/2024] [Indexed: 04/16/2024] Open
Abstract
OBJECTIVES Combination therapy of lenvatinib and immune checkpoint inhibitors (CLICI) has emerged as a promising approach for managing unresectable hepatocellular carcinoma (HCC). However, the response to such treatment is observed in only a subset of patients, underscoring the pressing need for reliable methods to identify potential responders. MATERIALS & METHODS This was a retrospective analysis involving 120 patients with unresectable HCC. They were divided into training (n = 72) and validation (n = 48) cohorts. We developed an interpretable deep learning model using multiphase computed tomography (CT) images to predict whether patients will respond or not to CLICI treatment, based on the Response Evaluation Criteria in Solid Tumors, version 1.1 (RECIST v1.1). We evaluated the models' performance and analyzed the impact of each CT phase. Critical regions influencing predictions were identified and visualized through heatmaps. RESULTS The multiphase model outperformed the best biphase and uniphase models, achieving an area under the curve (AUC) of 0.802 (95% CI = 0.780-0.824). The portal phase images were found to significantly enhance the model's predictive accuracy. Heatmaps identified six critical features influencing treatment response, offering valuable insights to clinicians. Additionally, we have made this model accessible via a web server at http://uhccnet.com/ for ease of use. CONCLUSIONS The integration of multiphase CT images with deep learning-generated heatmaps for predicting treatment response provides a robust and practical tool for guiding CLICI therapy in patients with unresectable HCC.
Collapse
Affiliation(s)
- Nan-Qing Liao
- School of Medical, Guangxi University, Nanning, China
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Zhu-Jian Deng
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Wei Wei
- Radiology Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jia-Hui Lu
- School of Computer, Electronics and Information, Guangxi University, Nanning, China
| | - Min-Jun Li
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Liang Ma
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Qing-Feng Chen
- School of Computer, Electronics and Information, Guangxi University, Nanning, China
| | - Jian-Hong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
47
|
Wang H, Ding Y, Zhao S, Li K, Li D. Establishment and validation of a nomogram model for early diagnosis of gastric cancer: a large-scale cohort study. Front Oncol 2024; 14:1463480. [PMID: 39678515 PMCID: PMC11638037 DOI: 10.3389/fonc.2024.1463480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2024] [Accepted: 11/12/2024] [Indexed: 12/17/2024] Open
Abstract
Purpose Identifying high-risk populations and diagnosing gastric cancer (GC) early remains challenging. This study aimed to establish and verify a nomogram model for the early diagnosis of GC based on conventional laboratory indicators. Methods We performed a retrospective analysis of the clinical data of 2,770 individuals with first diagnosis of GC and 1,513 patients with benign gastric disease from January 2018 to December 2022. The cases were divided into the training set and validation set randomly, with a ratio of 7:3. Variable screening was performed by least absolute shrinkage and selection operator (LASSO) and logistic regression analysis. A nomogram was constructed in the training set to assist in the early diagnosis of GC. Results There were 4283 patients included in the study, with 2998 patients assigned in the training set and 1285 patients in the validation set. Through LASSO regression and logistic regression analysis, independent variables associated with GC were identified, including CEA, CA199, LYM, HGB, MCH, MCHC, PLT, ALB, TG, HDL, and AFR. The nomogram model was constructed using the above 11 independent indicators. The AUC was 0.803 for the training set and 0.797 for the validation set, indicating that the model showed high clinical diagnostic efficacy. The calibration curves and decision curve analysis (DCA) of the nomogram presented good calibration and clinical application ability. Conclusion Based on the analysis of large sample size, we constructed a nomogram model with 11 routine laboratory indicators, which showed good discrimination ability and calibration.
Collapse
Affiliation(s)
- Haiyu Wang
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Yumin Ding
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Shujing Zhao
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Kaixu Li
- School of Public Health, Gansu University of Chinese Medicine, Lanzhou, Gansu, China
| | - Dehong Li
- Department of Clinical Laboratory, Gansu Provincial Hospital, Lanzhou, Gansu, China
| |
Collapse
|
48
|
Dong Z, Cai J, Geng H, Ni B, Yuan M, Zhang Y, Xia X, Zhang H, Zhang J, Zhu C, Wai Choi U, Regmi A, Chan CI, Yan CK, Gu Y, Cao H, Zhang Z. Image-based deep learning model to predict stoma-site incisional hernia in patients with temporary ileostomy: A retrospective study. iScience 2024; 27:111235. [PMID: 39563889 PMCID: PMC11574812 DOI: 10.1016/j.isci.2024.111235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 05/22/2024] [Accepted: 10/21/2024] [Indexed: 11/21/2024] Open
Abstract
The prophylactic implantation of biological mesh can effectively prevent the occurrence of stoma-site incisional hernia (SSIH) in patients undergoing stoma retraction. Therefore, our study prospectively established and validated a mixed model, which combined radiomics, stepwise regression, and deep learning for the prediction of SSIH in patients with temporary ileostomy. The mixed model showed good discrimination of the SSIH patients on all cohorts, which outperformed deep learning, radiomics, and clinical models alone (overall area under the curve [AUC]: 0.947 in the primary cohort, 0.876 in the external validation cohort 1, and 0.776 in the external validation cohort 2). Moreover, the sensitivity, specificity, and precision for predicting SSIH were improved in the mixed model. Thus, the mixed model can provide more information for SSIH precaution and clinical decision-making.
Collapse
Affiliation(s)
- Zhongyi Dong
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Jianhua Cai
- Department of General Surgery, Fudan University Affiliated Huadong Hospital, Shanghai 200040, P.R. China
| | - Haigang Geng
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Bo Ni
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Mengqing Yuan
- School of Science, The Hongkong University of Science and Technology, Hongkong 999077, P.R. China
| | - Yeqian Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Xiang Xia
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Haoyu Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Jie Zhang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Chunchao Zhu
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Un Wai Choi
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Aksara Regmi
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Cheok I. Chan
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Cara Kou Yan
- School of Medicine, Shanghai Jiaotong University, Shanghai 200025, P.R. China
| | - Yan Gu
- Department of General Surgery, Fudan University Affiliated Huadong Hospital, Shanghai 200040, P.R. China
| | - Hui Cao
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| | - Zizhen Zhang
- Department of Gastrointestinal Surgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai 200127, P.R. China
| |
Collapse
|
49
|
Lyu GW, Tong T, Yang GD, Zhao J, Xu ZF, Zheng N, Zhang ZF. Bibliometric and visual analysis of radiomics for evaluating lymph node status in oncology. Front Med (Lausanne) 2024; 11:1501652. [PMID: 39610679 PMCID: PMC11602298 DOI: 10.3389/fmed.2024.1501652] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Background Radiomics, which involves the conversion of digital images into high-dimensional data, has been used in oncological studies since 2012. We analyzed the publications that had been conducted on this subject using bibliometric and visual methods to expound the hotpots and future trends regarding radiomics in evaluating lymph node status in oncology. Methods Documents published between 2012 and 2023, updated to August 1, 2024, were searched using the Scopus database. VOSviewer, R Package, and Microsoft Excel were used for visualization. Results A total of 898 original articles and reviews written in English and be related to radiomics for evaluating lymph node status in oncology, published between 2015 and 2023, were retrieved. A significant increase in the number of publications was observed, with an annual growth rate of 100.77%. The publications predominantly originated from three countries, with China leading in the number of publications and citations. Fudan University was the most contributing affiliation, followed by Sun Yat-sen University and Southern Medical University, all of which were from China. Tian J. from the Chinese Academy of Sciences contributed the most within 5885 authors. In addition, Frontiers in Oncology had the most publications and transcended other journals in recent 4 years. Moreover, the keywords co-occurrence suggested that the interplay of "radiomics" and "lymph node metastasis," as well as "major clinical study" were the predominant topics, furthermore, the focused topics shifted from revealing the diagnosis of cancers to exploring the deep learning-based prediction of lymph node metastasis, suggesting the combination of artificial intelligence research would develop in the future. Conclusion The present bibliometric and visual analysis described an approximately continuous trend of increasing publications related to radiomics in evaluating lymph node status in oncology and revealed that it could serve as an efficient tool for personalized diagnosis and treatment guidance in clinical patients, and combined artificial intelligence should be further considered in the future.
Collapse
Affiliation(s)
- Gui-Wen Lyu
- Department of Radiology, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Tong Tong
- Department of Ultrasound, Shenzhen People’s Hospital, The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, China
| | - Gen-Dong Yang
- Department of Radiology, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jing Zhao
- Department of Radiology, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Zi-Fan Xu
- Department of Pathology, Shenzhen University Medical School, Shenzhen, China
| | - Na Zheng
- Department of Pathology, Shenzhen University Medical School, Shenzhen, China
| | - Zhi-Fang Zhang
- Department of Radiology, The Third People's Hospital of Shenzhen, The Second Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
50
|
Zhou Y, Zhou J, Cai X, Ge S, Sang S, Yang Y, Zhang B, Deng S. Integrating 18F-FDG PET/CT radiomics and body composition for enhanced prognostic assessment in patients with esophageal cancer. BMC Cancer 2024; 24:1402. [PMID: 39543534 PMCID: PMC11566154 DOI: 10.1186/s12885-024-13157-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: 07/30/2024] [Accepted: 11/06/2024] [Indexed: 11/17/2024] Open
Abstract
BACKGROUND This study aimed to develop a predictive model utilizing radiomics and body composition features derived from 18F-FDG PET/CT scans to forecast progression-free survival (PFS) and overall survival (OS) outcomes in patients with esophageal squamous cell carcinoma (ESCC). METHODS We analyzed data from 91 patients who underwent baseline 18F-FDG PET/CT imaging. Radiomic features extracted from PET and CT images and subsequent radiomics scores (Rad-scores) were calculated. Body composition metrics were also quantified, including muscle and fat distribution at the L3 level from CT scans. Multiparametric survival models were constructed using Cox regression analysis, and their performance was assessed using the area under the time-dependent receiver operating characteristic (ROC) curve (AUC) and concordance index (C-index). RESULTS Multivariate analysis identified Rad-scorePFS (P = 0.003), sarcopenia (P < 0.001), and visceral adipose tissue index (VATI) (P < 0.001) as independent predictors of PFS. For OS, Rad-scoreOS (P = 0.001), sarcopenia (P = 0.002), VATI (P = 0.037), stage (P = 0.042), and body mass index (BMI) (P = 0.008) were confirmed as independent prognostic factors. Integration of the Rad-score with clinical variables and body composition parameters enhanced predictive accuracy, yielding C-indices of 0.810 (95% CI: 0.737-0.884) for PFS and 0.806 (95% CI: 0.720-0.891) for OS. CONCLUSIONS This study underscored the potential of combining Rad-score with clinical and body composition data to refine prognostic assessment in ESCC patients.
Collapse
Affiliation(s)
- Yeye Zhou
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Jin Zhou
- Department of Nuclear Medicine, Shuyang Hospital Affiliated to Medical College of Yangzhou University, Suqian, China
| | - Xiaowei Cai
- Department of Nuclear Medicine, The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suqian, China
| | - Shushan Ge
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China
| | - Shibiao Sang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Yi Yang
- Department of Nuclear Medicine, Affiliated Hospital of Medical School, Suzhou Hospital, Nanjing University, Suzhou, China.
| | - Bin Zhang
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
| | - Shengming Deng
- Department of Nuclear Medicine, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China.
- Nuclear Medicine Laboratory of Mianyang Central Hospital, Mianyang, 621099, China.
| |
Collapse
|