1
|
Zhang D, Zhou W, Lu WW, Qin XC, Zhang XY, Luo YH, Wu J, Wang JL, Zhao JJ, Zhang CX. Ultrasound-based deep learning radiomics for enhanced axillary lymph node metastasis assessment: a multicenter study. Oncologist 2025; 30:oyaf090. [PMID: 40349137 PMCID: PMC12065944 DOI: 10.1093/oncolo/oyaf090] [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: 08/25/2024] [Accepted: 03/26/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Accurate preoperative assessment of axillary lymph node metastasis (ALNM) in breast cancer is crucial for guiding treatment decisions. This study aimed to develop a deep-learning radiomics model for assessing ALNM and to evaluate its impact on radiologists' diagnostic accuracy. METHODS This multicenter study included 866 breast cancer patients from 6 hospitals. The data were categorized into training, internal test, external test, and prospective test sets. Deep learning and handcrafted radiomics features were extracted from ultrasound images of primary tumors and lymph nodes. The tumor score and LN score were calculated following feature selection, and a clinical-radiomics model was constructed based on these scores along with clinical-ultrasonic risk factors. The model's performance was validated across the 3 test sets. Additionally, the diagnostic performance of radiologists, with and without model assistance, was evaluated. RESULTS The clinical-radiomics model demonstrated robust discrimination with AUCs of 0.94, 0.92, 0.91, and 0.95 in the training, internal test, external test, and prospective test sets, respectively. It surpassed the clinical model and single score in all sets (P < .05). Decision curve analysis and clinical impact curves validated the clinical utility of the clinical-radiomics model. Moreover, the model significantly improved radiologists' diagnostic accuracy, with AUCs increasing from 0.71 to 0.82 for the junior radiologist and from 0.75 to 0.85 for the senior radiologist. CONCLUSIONS The clinical-radiomics model effectively predicts ALNM in breast cancer patients using noninvasive ultrasound features. Additionally, it enhances radiologists' diagnostic accuracy, potentially optimizing resource allocation in breast cancer management.
Collapse
Affiliation(s)
- Di Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wang Zhou
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Wen-Wu Lu
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Xia-Chuan Qin
- Department of Medical Ultrasound, Chengdu Second People’s Hospital, Chengdu, China
| | - Xian-Ya Zhang
- Department of Medical Ultrasound, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yan-Hong Luo
- Department of Ultrasound, The Third Affiliated Hospital of Anhui Medical University, Hefei First People’s Hospital, Hefei, China
| | - Jun Wu
- Department of Ultrasound, The Second Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Jun-Li Wang
- Department of Ultrasound, WuHu Hospital, East China Normal University (The Second People’s Hospital, WuHu), Wuhu, China
| | - Jun-Jie Zhao
- Department of Medical Ultrasound, Fuyang Cancer Hospital, Fuyang, China
| | - Chao-Xue Zhang
- Department of Ultrasound, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Dou T, Chen Y, Liu L, Zhang Y, Pei W, Li J, Lei Y, Wang Y, Jia H. Radiogenomic analysis of clinical and ultrasonic characteristics in correlation to immune-related genes in breast cancer. Sci Rep 2025; 15:15918. [PMID: 40335526 PMCID: PMC12058982 DOI: 10.1038/s41598-025-00891-w] [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: 10/23/2024] [Accepted: 05/02/2025] [Indexed: 05/09/2025] Open
Abstract
Breast ultrasound plays a significant role in the non-invasive screening and diagnosis of breast cancer. The application of immunotherapy for breast cancer can significantly prolong the overall survival of advanced patients, which is an important research area of breast cancer treatment. The combination of ultrasound and immunotherapy helps patients diagnose and predict survival and develop a personalized treatment plan. This study analyzed the correlation between the clinical and ultrasonic characteristics of breast cancer and immune-related genes. First, the differential expression of immune-related genes was obtained using the GEO and IMMPORT database. Then, differentially expressed immune-related genes related to the overall survival of breast cancer were obtained using the GEPIA and Kaplan-Meier plotter platforms. Additionally, clinical, ultrasonic characteristics and pathological specimens of breast cancer patients' tumors were collected. Whole transcriptome sequencing and immunohistochemical staining were performed on the tumor specimens to obtain gene expression. CXCL2, MIA, NR3C2, PTX3, S100B, SAA1, SAA2, and CXCL9 genes were correlated with each other and with clinical and ultrasonic characteristics. The high expression of MIA was related to the positive expression of PR in breast cancer. The low expression of NR3C2 was correlated with the clinical characteristics of tumor size ≥ 20 mm, later stage, Her-2 positive, Ki-67 ≥ 20%. NR3C2 was negatively correlated with the value of PKI and AUC in contrast-enhanced ultrasound parameters, and positively correlated with the value of AT and TTP. The expression of the PTX3 gene was also negatively correlated with the value of PKI and Emax of shear wave elastography. SAA2 was related to the presence or absence of edge burrs characterized by ultrasound. The expression of the CXCL9 gene was associated with the age of onset and tumor stage. In this study, 8 differentially expressed immune-related genes related to the overall survival of breast cancer were screened, which had been proved to be associated with some characteristics of cancer in previous studies, and could be further studied in the subsequent immunotherapy of breast cancer. Some clinical and ultrasonic characteristics of breast cancer were significantly correlated with immune-related genes, such as NR3C2, SAA2, and CXCL9. Further analysis of these genes provides new ideas for the diagnosis and treatment of breast cancer.
Collapse
Affiliation(s)
- Tingyao Dou
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaodong Chen
- Department of Ultrasonic Imaging, First Hospital of Shanxi Medical University, Taiyuan, 030001, Shanxi, China.
| | - Lunhang Liu
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yaochen Zhang
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Wanru Pei
- Department of First Clinical Medicine, Shanxi Medical University, Taiyuan, Shanxi, China
| | - Jing Li
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yan Lei
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China
| | - Yanhong Wang
- Department of Microbiology and Immunology, School of Basic Medical Sciences, ShanxiMedical University, Taiyuan, Shanxi, China.
- key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China.
- Department of Microbiology and Immunology, Shanxi Medical University, Taiyuan, Shanxi, China.
| | - Hongyan Jia
- Department of Breast Surgery, First Hospital of Shanxi Medical University, Taiyuan, Shanxi, China.
- key Laboratory of Cellular Physiology, Shanxi Medical University, Ministry of Education, Taiyuan, Shanxi, China.
| |
Collapse
|
3
|
Pan G, Pan Z, Chen W, Wu Y, Di X, Zhou F, Liao Y. Integration of MRI radiomics and clinical data for preoperative prediction of vascular invasion in breast cancer: A deep learning approach. Magn Reson Imaging 2025; 118:110339. [PMID: 39880177 DOI: 10.1016/j.mri.2025.110339] [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: 10/26/2024] [Revised: 01/24/2025] [Accepted: 01/26/2025] [Indexed: 01/31/2025]
Abstract
BACKGROUND Accurate preoperative prediction of vascular invasion in breast cancer is crucial for surgical planning and patient management. MRI radiomics has shown promise in enhancing diagnostic precision. This study aims to evaluate the effectiveness of integrating MRI radiomic features with clinical data using a deep learning approach to predict vascular invasion in breast cancer patients. METHODS A retrospective analysis was conducted on 102 patients with invasive breast cancer confirmed by surgical pathology. Using the MR750 3.0 T as the examination device, the subject underwent the examination in standard breast positions and sequences. Diffusion-weighted imaging (DWI) was performed with two selected b-values, specifically 0 and 1000 s/mm2. Following the injection of the contrast agent, dynamic scans were conducted across six phases, and delayed phase sagittal images were acquired using the VIBRANT sequence. Texture features were extracted from MRI images, and key radiomic and clinical features were selected using variance thresholding, correlation filtering, and logistic regression. A predictive model was developed combining these features, and its performance was evaluated through sensitivity, specificity, and area under the curve (AUC) metrics. RESULTS The univariate models based on individual MRI sequences or clinical data demonstrated variable diagnostic performance. In contrast, the multifactorial model that combined radiomic features with clinical data achieved significantly higher accuracy, with an AUC of 0.829, sensitivity of 76.9 %, and specificity of 83.3 %. CONCLUSION Integrating MRI radiomics and clinical data enhances the preoperative prediction of vascular invasion in breast cancer. This approach can improve diagnostic accuracy, providing valuable insights for clinical decision-making and personalized treatment strategies.
Collapse
Affiliation(s)
- Guihai Pan
- Department of Radiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Zejun Pan
- Department of Radiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Wubiao Chen
- Department of Radiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Yongjun Wu
- Department of Radiology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Xiaoqing Di
- Department of Pathology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China
| | - Fei Zhou
- Department of Endocrinology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, China.
| | | |
Collapse
|
4
|
Meng F, Zhang T, Pan Y, Kan X, Xia Y, Xu M, Cai J, Liu F, Ge Y. A deep learning algorithm for automated adrenal gland segmentation on non-contrast CT images. BMC Med Imaging 2025; 25:142. [PMID: 40312690 PMCID: PMC12046700 DOI: 10.1186/s12880-025-01682-5] [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: 11/07/2024] [Accepted: 04/18/2025] [Indexed: 05/03/2025] Open
Abstract
BACKGROUND The adrenal glands are small retroperitoneal organs, few reference standards exist for adrenal CT measurements in clinical practice. This study aims to develop a deep learning (DL) model for automated adrenal gland segmentation on non-contrast CT images, and to conduct a preliminary large-scale study on age-related volume changes in normal adrenal glands using the model output values. METHODS The model was trained and evaluated on a development dataset of annotated non-contrast CT scans of bilateral adrenal glands, utilizing nnU-Net for segmentation task. The ground truth was manually established by two experienced radiologists, and the model performance was assessed using the Dice similarity coefficient (DSC). Additionally, five radiologists provided annotations on a subset of 20 randomly selected cases to measure inter-observer variability. Following validation, the model was applied to a large-scale normal adrenal glands dataset to segment adrenal glands. RESULTS The DL model development dataset contained 1301 CT examinations. In the test set, the median DSC scores for the segmentation model of left and right adrenal glands were 0.899 and 0.904 respectively, and in the independent test set were 0.900 and 0.896. Inter-observer DSC for radiologist manual segmentation did not differ from automated machine segmentation (P = 0.541). The large-scale normal adrenal glands dataset contained 2000 CT examinations, the graph shows that adrenal gland volume increases first and then decreases with age. CONCLUSION The developed DL model demonstrates accurate adrenal gland segmentation, and enables a comprehensive study of age-related adrenal gland volume variations.
Collapse
Affiliation(s)
- Fanxing Meng
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Tuo Zhang
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Yukun Pan
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Xiaojing Kan
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Yuwei Xia
- Shanghai United Imaging Intelligence Co. Ltd, 701 Yunjin Road, Xuhui District, Shanghai, 200030, China
| | - Mengyuan Xu
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Jin Cai
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Fangbin Liu
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China
| | - Yinghui Ge
- Department of Radiology, Central China Subcenter of National Center for Cardiovascular Diseases, Fuwai Central-China Cardiovascular Hospital, Central China Fuwai Hospital of Zhengzhou University, Zhengzhou, 450046, China.
| |
Collapse
|
5
|
Chen Y, Shao X, Shi K, Rominger A, Caobelli F. AI in Breast Cancer Imaging: An Update and Future Trends. Semin Nucl Med 2025; 55:358-370. [PMID: 40011118 DOI: 10.1053/j.semnuclmed.2025.01.008] [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: 01/27/2025] [Revised: 01/30/2025] [Accepted: 01/30/2025] [Indexed: 02/28/2025]
Abstract
Breast cancer is one of the most common types of cancer affecting women worldwide. Artificial intelligence (AI) is transforming breast cancer imaging by enhancing diagnostic capabilities across multiple imaging modalities including mammography, digital breast tomosynthesis, ultrasound, magnetic resonance imaging, and nuclear medicines techniques. AI is being applied to diverse tasks such as breast lesion detection and classification, risk stratification, molecular subtyping, gene mutation status prediction, and treatment response assessment, with emerging research demonstrating performance levels comparable to or potentially exceeding those of radiologists. The large foundation models are showing remarkable potential in different breast cancer imaging tasks. Self-supervised learning gives an insight into data inherent correlation, and federated learning is an alternative way to maintain data privacy. While promising results have been obtained so far, data standardization from source, large-scale annotated multimodal datasets, and extensive prospective clinical trials are still needed to fully explore and validate deep learning's clinical utility and address the legal and ethical considerations, which will ultimately determine its widespread adoption in breast cancer care. We hereby provide a review of the most up-to-date knowledge on AI in breast cancer imaging.
Collapse
Affiliation(s)
- Yizhou Chen
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Xiaoliang Shao
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland; Department of Nuclear Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Kuangyu Shi
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Axel Rominger
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Federico Caobelli
- Department of Nuclear Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
6
|
Nair A, Ong W, Lee A, Leow NW, Makmur A, Ting YH, Lee YJ, Ong SJ, Tan JJH, Kumar N, Hallinan JTPD. Enhancing Radiologist Productivity with Artificial Intelligence in Magnetic Resonance Imaging (MRI): A Narrative Review. Diagnostics (Basel) 2025; 15:1146. [PMID: 40361962 PMCID: PMC12071790 DOI: 10.3390/diagnostics15091146] [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: 02/04/2025] [Revised: 04/06/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Artificial intelligence (AI) shows promise in streamlining MRI workflows by reducing radiologists' workload and improving diagnostic accuracy. Despite MRI's extensive clinical use, systematic evaluation of AI-driven productivity gains in MRI remains limited. This review addresses that gap by synthesizing evidence on how AI can shorten scanning and reading times, optimize worklist triage, and automate segmentation. On 15 November 2024, we searched PubMed, EMBASE, MEDLINE, Web of Science, Google Scholar, and Cochrane Library for English-language studies published between 2000 and 15 November 2024, focusing on AI applications in MRI. Additional searches of grey literature were conducted. After screening for relevance and full-text review, 67 studies met inclusion criteria. Extracted data included study design, AI techniques, and productivity-related outcomes such as time savings and diagnostic accuracy. The included studies were categorized into five themes: reducing scan times, automating segmentation, optimizing workflow, decreasing reading times, and general time-saving or workload reduction. Convolutional neural networks (CNNs), especially architectures like ResNet and U-Net, were commonly used for tasks ranging from segmentation to automated reporting. A few studies also explored machine learning-based automation software and, more recently, large language models. Although most demonstrated gains in efficiency and accuracy, limited external validation and dataset heterogeneity could reduce broader adoption. AI applications in MRI offer potential to enhance radiologist productivity, mainly through accelerated scans, automated segmentation, and streamlined workflows. Further research, including prospective validation and standardized metrics, is needed to enable safe, efficient, and equitable deployment of AI tools in clinical MRI practice.
Collapse
Affiliation(s)
- Arun Nair
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
| | - Wilson Ong
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
| | - Aric Lee
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
| | - Naomi Wenxin Leow
- AIO Innovation Office, National University Health System, 3 Research Link, #02-04 Innovation 4.0, Singapore 117602, Singapore;
| | - Andrew Makmur
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
| | - Yong Han Ting
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
| | - You Jun Lee
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
| | - Shao Jin Ong
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
| | - Jonathan Jiong Hao Tan
- National University Spine Institute, Department of Orthopaedic Surgery, National University Health System, 1E Lower Kent Ridge Road, Singapore 119228, Singapore; (J.J.H.T.); (N.K.)
| | - Naresh Kumar
- National University Spine Institute, Department of Orthopaedic Surgery, National University Health System, 1E Lower Kent Ridge Road, Singapore 119228, Singapore; (J.J.H.T.); (N.K.)
| | - James Thomas Patrick Decourcy Hallinan
- Department of Diagnostic Imaging, National University Hospital, 5 Lower Kent Ridge Rd, Singapore 119074, Singapore; (A.N.); (W.O.); (A.L.); (A.M.); (Y.H.T.); (Y.J.L.); (S.J.O.)
- Department of Diagnostic Radiology, Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Drive, Singapore 117597, Singapore
| |
Collapse
|
7
|
Agyekum EA, Kong W, Ren YZ, Issaka E, Baffoe J, Xian W, Tan G, Xiong C, Wang Z, Qian X, Shen X. A comparative analysis of three graph neural network models for predicting axillary lymph node metastasis in early-stage breast cancer. Sci Rep 2025; 15:13918. [PMID: 40263507 PMCID: PMC12015543 DOI: 10.1038/s41598-025-97257-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: 08/25/2024] [Accepted: 04/03/2025] [Indexed: 04/24/2025] Open
Abstract
The presence of axillary lymph node metastasis (ALNM) in breast cancer patients is an important factor in deciding whether to have axillary surgery or pursue alternative treatments. Based on axillary ultrasound (US) and histopathologic data, three graph neural network models were compared to predict ALNM in early-stage breast cancer. The patients were randomly divided into two data sets: training (80%) and testing (20%). Predictive performance was measured using accuracy, sensitivity, specificity, positive predictive value, negative predictive value, F1 score, and area under the curve (AUC). In the test cohort, the graph convolutional network (GCN) performed the best in predicting ALNM, with an AUC of 0.77 (95% confidence interval [CI]: 0.69-0.84). In conclusion, the GCN model has the potential to provide a noninvasive tool for detecting ALNM and can aid in clinical decision-making. Prospective studies are expected to provide high-level evidence for clinical usage in future investigations.
Collapse
Affiliation(s)
- Enock Adjei Agyekum
- Department of Ultrasound Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, Jiangsu, China
| | - Wentao Kong
- Department of Ultrasound Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yong-Zhen Ren
- Department of Ultrasound Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Eliasu Issaka
- College of Engineering, Birmingham City University, Birmingham, B4 7XG, UK
| | - Josephine Baffoe
- School of Automotive and Traffic Engineering, Jiangsu University, Zhenjiang, 212013, P.R. China
| | - Wang Xian
- Department of Ultrasound Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Gongxun Tan
- Department of Ultrasound Medicine, Affiliated People's Hospital of Jiangsu University, Zhenjiang, Jiangsu, China
| | - Chunjing Xiong
- Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou Clinical Medical College of Jiangsu University, Yangzhou, Jiangsu, China
| | - Zhangye Wang
- Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou Clinical Medical College of Jiangsu University, Yangzhou, Jiangsu, China.
| | - Xiaoqin Qian
- Northern Jiangsu People's Hospital, Northern Jiangsu People's Hospital Affiliated to Yangzhou University, The Yangzhou Clinical Medical College of Xuzhou Medical University, The Yangzhou Clinical Medical College of Jiangsu University, Yangzhou, Jiangsu, China.
| | - Xiangjun Shen
- School of Computer Science and Communication Engineering, Jiangsu University, Zhenjiang, Jiangsu, China.
| |
Collapse
|
8
|
Liang X, Luo S, Liu Z, Liu Y, Luo S, Zhang K, Li L. Unsupervised machine learning analysis of optical coherence tomography radiomics features for predicting treatment outcomes in diabetic macular edema. Sci Rep 2025; 15:13389. [PMID: 40251316 PMCID: PMC12008428 DOI: 10.1038/s41598-025-96988-3] [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: 01/16/2025] [Accepted: 04/01/2025] [Indexed: 04/20/2025] Open
Abstract
This study aimed to identify distinct clusters of diabetic macular edema (DME) patients with differential anti-vascular endothelial growth factor (VEGF) treatment outcomes using an unsupervised machine learning (ML) approach based on radiomic features extracted from pre-treatment optical coherence tomography (OCT) images. Retrospective data from 234 eyes with DME treated with three anti-VEGF therapies between January 2020 and March 2024 were collected from two clinical centers. Radiomic analysis was conducted on pre-treatment OCT images. Following principal component analysis (PCA) for dimensionality reduction, two unsupervised clustering methods (K-means and hierarchical clustering) were applied. Baseline characteristics and treatment outcomes were compared across clusters to assess clustering efficacy. Feature selection employed a three-stage pipeline: exclusion of collinear features (Pearson's r > 0.8); sequential filtering through ANOVA (P < 0.05) and Boruta algorithm (500 iterations); multivariate stepwise regression (entry criteria: univariate P < 0.1) to identify outcome-associated predictors. From 1165 extracted radiomic features, four distinct DME clusters were identified. Cluster 4 exhibited a significantly lower incidence of residual/recurrent DME (RDME) (34.29%) compared to Clusters 1-3 (P = 0.003, P = 0.005 and P = 0.002, respectively). This cluster also demonstrated the highest proportion of eyes (71.43%) with best-corrected visual acuity (BCVA) exceeding 20/63 (P = 0.003, P = 0.005 and P = 0.002, respectively). Multivariate analysis identified logarithm_gldm_DependenceVariance as an independent risk factor for RDME (OR 1.75, 95% CI 1.28-2.40; P < 0.001), while Wavelet-LH_Firstorder_Mean correlated with worse visual outcomes (OR 8.76, 95% CI 1.22-62.84; P = 0.031). Unsupervised ML leveraging pre-treatment OCT radiomics successfully stratifies DME eyes into clinically distinct subgroups with divergent therapeutic responses. These quantitative features may serve as non-invasive biomarkers for personalized outcome prediction and retinal pathology assessment.
Collapse
Affiliation(s)
- Xuemei Liang
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shaozhao Luo
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
| | - Zhigao Liu
- Department of Ophthalmology, Jinan Aier Eye Hospital, No. 1916, Erhuan East Road, Licheng District, Jinan City, Shandong Province, People's Republic of China
| | - Yunsheng Liu
- Department of Ophthalmology, Cenxi Aier Eye Hospital, No. 101, Yuwu Avenue, Cenxi City, Wuzhou City, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Shinan Luo
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Kaiqing Zhang
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China
| | - Li Li
- Department of Ophthalmology, Aier Eye Hospital, Jinan University, No, 191, Huanshi Middle Road, Yuexiu District, Guangzhou, 510071, Guangdong, People's Republic of China.
- Department of Ophthalmology, Nanning Aier Eye Hospital, No, 63, Chaoyang Road, Xingning District, Nanning, 530012, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
9
|
Li Z, Miao H, Bao W, Zhang L. Development and validation of a nomogram model of lung metastasis in breast cancer based on machine learning algorithm and cytokines. BMC Cancer 2025; 25:692. [PMID: 40229760 PMCID: PMC11998148 DOI: 10.1186/s12885-025-14101-3] [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: 01/16/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND The relationship between cytokines and lung metastasis (LM) in breast cancer (BC) remains unclear and current clinical methods for identifying breast cancer lung metastasis (BCLM) lack precision, thus underscoring the need for an accurate risk prediction model. This study aimed to apply machine learning algorithms for identifying the key risk factors for BCLM before developing a reliable prediction model centered on cytokines. METHODS This population-based retrospective study included 326 BC patients admitted to the Second Affiliated Hospital of Xuzhou Medical University between September 2018 and September 2023. After randomly assigning the patients to a training cohort (70%; n = 228) or a validation cohort (30%; n = 98) the risk factors for BCLM were identified using Least Absolute Shrinkage and Selection Operator (LASSO), Extreme Gradient Boosting (XGBoost) and Random Forest (RF) models. Significant risk factors were visualized with a Venn diagram and incorporated into a nomogram model, the performance of which was then evaluated according to three criteria, namely discrimination, calibration and clinical utility using calibration plots, receiver operating characteristic (ROC) curves and decision curve analysis (DCA). RESULTS Among the cohort, 70 patients developed LM. A nomogram was then developed to predict the 5-year and 10-year BCLM risk by incorporating five key variables, namely endocrine therapy, hsCRP, IL6, IFN-ɑ and TNF-ɑ. For the 5-year prediction model, the training and validation cohorts had AUC values of 0.786 (95% CI: 0.691-0.881) and 0.627 (95% CI: 0.441-0.813), respectively, while for the 10-year prediction model, the corresponding AUC values were 0.687 (95% CI: 0.528-0.847) and 0.797 (95% CI: 0.605-0.988), respectively. ROC analysis further confirmed the model's strong discriminative ability, while calibration plots indicated that the predicted and observed outcomes were in good agreement in both cohorts. Finally, DCA demonstrated the model's effectiveness in clinical practice. CONCLUSION Using machine learning algorithms, this study developed aa nomogram that could effectively identify BC patients who were at a higher risk of developing LM, thus providing a valuable tool for decision-making in clinical settings.
Collapse
Affiliation(s)
- Zhaoyi Li
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Meijian Road 32, Xuzhou, 221000, China
| | - Hao Miao
- Xuzhou Medical University, Tongshan Road 209, Xuzhou, 221000, China
| | - Wei Bao
- Tongji University, Yangpu District, Siping Road 1239, Shanghai, 310000, China
| | - Lansheng Zhang
- Department of Radiotherapy, The Second Affiliated Hospital of Xuzhou Medical University, Meijian Road 32, Xuzhou, 221000, China.
| |
Collapse
|
10
|
Yao Q, Du Y, Liu W, Liu X, Zhang M, Zha H, Du L, Zha X, Wang J, Li C. Improving Prediction Accuracy of Residual Axillary Lymph Node Metastases in Node-Positive Triple-Negative Breast Cancer: A Radiomics Analysis of Ultrasound-Guided Clip Locations Using the SHAP Method. Acad Radiol 2025; 32:1827-1837. [PMID: 39523140 DOI: 10.1016/j.acra.2024.10.039] [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: 09/19/2024] [Revised: 10/20/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
RATIONALE AND OBJECTIVES To construct a radiomics nomogram derived from multiparametric ultrasound (US) imaging using the SHapley Additive exPlanations (SHAP) method for the accurate identification of residual axillary lymph node metastases post-neoadjuvant chemotherapy (NAC) among patients with triple-negative breast cancer (TNBC). METHODS A total of 405 consecutive patients with pathologically confirmed TNBC between 2016 and 2023 were recruited in the study and were divided into training (n = 284) and validation cohorts (n = 121). Radiomics features capturing detailed tumor characteristics were extracted from pre-NAC gray-scale US images at the locations of US-guided clip placement. The least absolute shrinkage and selection operator and the maximum relevance minimum redundancy algorithm were employed to identify key features and formulate the radiomics signature (RS). A nomogram based on US radiomics was then constructed using multivariable logistic regression analysis. The predictive efficacy of this model was evaluated through receiver operating characteristic curve analysis, calibration assessment, and decision curve analysis. SHAP summary plots were used to visualize the distribution of SHAP values across all features. RESULTS The nomogram integrates clinical and US characteristics with RS, yielded optimal AUC of 0.922 (95% CI, 0.890-0.954) in the training cohort, 0.904 (95% CI, 0.853-0.955) in the validation cohort. The calibration and decision curves confirmed favorable calibration and clinical value of the nomogram. SHAP provided further insight into the contributions of each feature to the model's outcomes. CONCLUSION The combined multiparametric US based radiomics nomogram plays a potential role in predicting residual axillary lymph node metastases after NAC in TNBCs.
Collapse
Affiliation(s)
- Qing Yao
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| | - Yu Du
- Department of Ultrasound, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China (Y.D.).
| | - Wei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| | - Xinpei Liu
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| | - Manqi Zhang
- Department of Ultrasound, Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China (M.Z.)
| | - Hailing Zha
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| | - Liwen Du
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| | - Xiaoming Zha
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (X.Z., J.W.)
| | - Jue Wang
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (X.Z., J.W.)
| | - Cuiying Li
- Department of Ultrasound, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China (Q.Y., W.L., X.L., H.Z., L.D., C.L.)
| |
Collapse
|
11
|
Wang Y, Han Q, Wen B, Yang B, Zhang C, Song Y, Zhang L, Xian J. Development and validation of a prediction model for malignant sinonasal tumors based on MR radiomics and machine learning. Eur Radiol 2025; 35:2074-2083. [PMID: 39210161 DOI: 10.1007/s00330-024-11033-7] [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: 01/09/2024] [Revised: 06/23/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024]
Abstract
OBJECTIVES This study aimed to utilize MR radiomics-based machine learning classifiers on a large-sample, multicenter dataset to develop an optimal model for predicting malignant sinonasal tumors and tumor-like lesions. METHODS This study included 1711 adult patients (875 benign and 836 malignant) with sinonasal tumors or tumor-like lesions from three institutions. Patients from institution 1 (n = 1367) constituted both the training and validation cohorts, while those from institution 2 and 3 (n = 158/186) made up the test cohorts. Manual segmentation of the region of interest of the tumor was performed on T1WI, T2WI, and contrast-enhanced T1WI (CE-T1WI). Data normalization, dimensional reductions, feature selection, and classifications were performed using ten machine-learning classifiers. Four fusion models, namely T1WI + T2WI, T1WI + CE-T1WI, T2WI + CE-T1WI, and T1WI + T2WI + CE-T1WI, were constructed using the top ten features with the highest contribution in feature selection in the optimal models of T1WI, T2WI, and CE-T1WI. The Delong test compared areas under the curve (AUC) between models. RESULTS The AUCs of training/validation/test1/test2 datasets for T1WI, T2WI, and CE-T1WI were 0.900/0.842/0.872/0.839, 0.876/0.789/0.842/0.863, and 0.899/0.824/0.831/0.707, respectively. The fusion model from T1WI + T2WI + CE-T1WI had the highest AUC. The AUCs of training/validation/test1/test2 datasets were 0.947/0.849/0.871/0.887. The T1WI + T2WI + CE-T1WI model demonstrated a significantly higher AUC than the T2WI + CE-T1WI model in both cohorts (p < 0.05) and outperformed the T2WI model in test 1 (p = 0.008) and the T1WI model in test 2 (p = 0.006). CONCLUSIONS This fusion model based on radiomics from T1WI + T2WI + CE-T1WI images and machine learning can improve the power in predicting malignant sinonasal tumors with high accuracy, resilience, and robustness. CLINICAL RELEVANCE STATEMENT Our study proposes a radiomics-based machine learning fusion model from T1- and T2-weighted images and contrast-enhanced T1-weighted images, which can non-invasively identify the nature of sinonasal tumors and improve the performance in predicting malignant sinonasal tumors. KEY POINTS Differentiating benign and malignant sinonasal tumors is difficult due to similar clinical presentations. A radiomics model from T1 + T2 + contrast-enhanced T1 images can identify the nature of sinonasal tumors. This model can help distinguish benign and malignant sinonasal tumors.
Collapse
Affiliation(s)
- Yuchen Wang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Qinghe Han
- Department of Radiology, The Second Hospital of Jilin University, Changchun, China
| | - Baohong Wen
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bingbing Yang
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China
| | - Chen Zhang
- MR Research Collaboration Team, Siemens Healthcare, Beijing, China
| | - Yang Song
- MR Research Collaboration Team, Siemens Healthcare, Beijing, China
| | - Luo Zhang
- Department of Otolaryngology-Head and Neck Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
- Beijing Laboratory of Allergic Diseases and Beijing Key Laboratory of Nasal Diseases, Beijing Institute of Otorhinolaryngology, Beijing, China.
- Research Unit of Diagnosis and Treatment of Chronic Nasal Diseases, Chinese Academy of Medical Sciences, Beijing, China.
- Department of Allergy, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| | - Junfang Xian
- Department of Radiology, Beijing Tongren Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
12
|
Yang Z, Wang S, Yin W, Wang Y, Liu F, Xu J, Han L, Liu C. Radiomics-clinical nomogram for preoperative tumor-node-metastasis staging prediction in breast cancer patients using dynamic enhanced magnetic resonance imaging. Transl Cancer Res 2025; 14:1836-1848. [PMID: 40225004 PMCID: PMC11985186 DOI: 10.21037/tcr-24-1559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 01/09/2025] [Indexed: 04/15/2025]
Abstract
Background Breast cancer is one of the most commonly diagnosed malignancies in women worldwide, and the disease burden continues to aggravate. The tumor-node-metastasis (TNM) staging information is crucial for oncology physicians to develop appropriate clinical strategies. This study aimed to investigate the value of a radiomics-clinical model for predicting TNM stage in breast cancer patients using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). Methods DCE-MRI images from 166 patients with pathologically confirmed breast cancer were retrospectively collected, including early stage (TNM0-TNM2) and locally advanced or advanced stage (TNM3-TNM4). Included patients were divided into a training cohort (n=116) and a test cohort (n=50). The radiomics, clinical and integrated models were constructed and a nomogram was established to distinguish the TNM0-TNM2 stage from the TNM3-TNM4 stage. Receiver operating characteristic (ROC) curves, calibration curves and decision curve analysis (DCA) were employed to assess the predictability of the models. Results Eighty-five patients were at the early stages, while 81 patients were at the other stages. In the training and test cohorts, the area under the curve (AUC) values for distinguishing early and advanced breast cancer were 0.870 and 0.818 for the nomogram, respectively. The nomogram calibration curves showed good agreement between the predicted and observed TNM stages in the training and test cohorts. The Hosmer-Lemeshow test showed that the nomogram fit perfectly in the two cohorts. DCA indicated that the nomogram displayed clear superiority in forecasting TNM staging over clinical and radiomic signatures. Conclusions Compared to traditional imaging methods, the clinical-radiomics nomogram acquired by DCE-MRI could potentially be utilized to preoperatively evaluate the TNM stage of breast cancer with relatively high accuracy. It can be an effective method to guide clinical decisions.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Shouen Wang
- Department of Pathology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Wei Yin
- Department of Radiology, Beijing Friendship Hospital of Capital Medical University, Beijing, China
| | - Ying Wang
- Department of Radiology, the First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Fanghua Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Jianshu Xu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Long Han
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| | - Chenglong Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, China
| |
Collapse
|
13
|
Li X, Fan F, Zhang T. Efficacy and influencing factors of immunotherapy crossover combined with targeted therapy in advanced esophageal cancer patients following first-line chemotherapy combined with immunotherapy failure. Am J Cancer Res 2025; 15:1321-1334. [PMID: 40226448 PMCID: PMC11982723 DOI: 10.62347/gboq6704] [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: 10/14/2024] [Accepted: 02/28/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Advanced esophageal cancer presents significant treatment challenges, especially after immunochemotherapy failure. This study evaluates the efficacy of further treatment with combination chemotherapy versus combination immunotherapy crossover in terms of tumor regression, quality of life, and identifies factors influencing treatment outcomes. METHODS In a retrospective case-control study, clinical data from 293 patients with advanced esophageal cancer treated at Shanxi Province Cancer Hospital between February 2021 and February 2023 were analyzed. Patients excluded from radical resection due to failure of first-line immunotherapy were divided into two groups: 95 received combination chemotherapy with Irinotecan and Tigio (S-1, Tegafur/Gimeracil/Oteracil Potassium), and 198 underwent Anlotinib targeted therapy combined with immunotherapy crossover. Treatment efficacy was assessed using tumor regression grading (TRG), and quality of life was evaluated using EORTC QLQ-C30 and QLQ-OES18 scales. Potential factors affecting treatment efficacy were examined using multivariate logistic regression analysis. RESULTS Baseline characteristics, including age, gender, body mass index (BMI), and history of smoking and alcohol consumption, were comparable between the two groups. TRG showed no significant differences in distribution, with objective response rates of 40% in the Irinotecan/S-1 group and 44.44% in the combined immunotherapy crossover group (P = 0.472). However, quality of life measures indicated superior outcomes from immunotherapy crossover in physical (P = 0.024), emotional (P = 0.002), and general health scores (P = 0.003). Factors negatively impacting treatment success included male gender, smoking, alcohol consumption history, and certain tumor locations. Elevated CEA levels positively correlated with treatment efficacy. Logistic regression analysis identified male gender (OR, 2.109; P = 0.021), smoking (OR, 2.575; P = 0.003), alcohol consumption (OR, 1.995; P = 0.043), and CEA levels (OR, 0.742; P = 0.017) as significant predictors of treatment efficacy. CONCLUSION Immunotherapy combined with targeted therapy and chemotherapy alone showed comparable efficacy in tumor regression. However, immunotherapy combined with targeted therapy improved certain aspects of quality of life. Factors such as gender, lifestyle habits, and CEA levels can significantly influence treatment outcomes.
Collapse
Affiliation(s)
- Xiuxiu Li
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive CancerTianjin 300060, China
- Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Fan Fan
- Department of Gastroenterology, Shanxi Province Cancer Hospital, Shanxi Hospital Affiliated to Cancer Hospital, Chinese Academy of Medical Sciences, Cancer Hospital Affiliated to Shanxi Medical UniversityTaiyuan 030001, Shanxi, China
| | - Ti Zhang
- Department of Hepatobiliary Surgery, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin’s Clinical Research Center for Cancer, Tianjin Key Laboratory of Digestive CancerTianjin 300060, China
- Department of Hepatobiliary Surgery, Fudan University Shanghai Cancer CenterShanghai 200032, China
- Department of Oncology, Shanghai Medical College, Fudan UniversityShanghai 200032, China
| |
Collapse
|
14
|
Liao J, Xu Z, Xie Y, Liang Y, Hu Q, Liu C, Yan L, Diao W, Liu Z, Wu L, Liang C. Assessing Axillary Lymph Node Burden and Prognosis in cT1-T2 Stage Breast Cancer Using Machine Learning Methods: A Retrospective Dual-Institutional MRI Study. J Magn Reson Imaging 2025; 61:1221-1231. [PMID: 39175033 DOI: 10.1002/jmri.29554] [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: 05/11/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/24/2024] Open
Abstract
BACKGROUND Pathological axillary lymph node (pALN) burden is an important factor for treatment decision-making in clinical T1-T2 (cT1-T2) stage breast cancer. Preoperative assessment of the pALN burden and prognosis aids in the individualized selection of therapeutic approaches. PURPOSE To develop and validate a machine learning (ML) model based on clinicopathological and MRI characteristics for assessing pALN burden and survival in patients with cT1-T2 stage breast cancer. STUDY TYPE Retrospective. POPULATION A total of 506 females (range: 24-83 years) with cT1-T2 stage breast cancer from two institutions, forming the training (N = 340), internal validation (N = 85), and external validation cohorts (N = 81), respectively. FIELD STRENGTH/SEQUENCE This study used 1.5-T, axial fat-suppressed T2-weighted turbo spin-echo sequence and axial three-dimensional dynamic contrast-enhanced fat-suppressed T1-weighted gradient echo sequence. ASSESSMENT Four ML methods (eXtreme Gradient Boosting [XGBoost], Support Vector Machine, k-Nearest Neighbor, Classification and Regression Tree) were employed to develop models based on clinicopathological and MRI characteristics. The performance of these models was evaluated by their discriminative ability. The best-performing model was further analyzed to establish interpretability and used to calculate the pALN score. The relationships between the pALN score and disease-free survival (DFS) were examined. STATISTICAL TESTS Chi-squared test, Fisher's exact test, univariable logistic regression, area under the curve (AUC), Delong test, net reclassification improvement, integrated discrimination improvement, Hosmer-Lemeshow test, log-rank, Cox regression analyses, and intraclass correlation coefficient were performed. A P-value <0.05 was considered statistically significant. RESULTS The XGB II model, developed based on the XGBoost algorithm, outperformed the other models with AUCs of 0.805, 0.803, and 0.818 in the three cohorts. The Shapley additive explanation plot indicated that the top variable in the XGB II model was the Node Reporting and Data System score. In multivariable Cox regression analysis, the pALN score was significantly associated with DFS (hazard ratio: 4.013, 95% confidence interval: 1.059-15.207). DATA CONCLUSION The XGB II model may allow to evaluate pALN burden and could provide prognostic information in cT1-T2 stage breast cancer patients. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Jiayi Liao
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zeyan Xu
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Yu Xie
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University, Peking University Cancer Hospital Yunnan, Kunming, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Qingru Hu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Chunling Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Lifen Yan
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Wenjun Diao
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Lei Wu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Changhong Liang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
15
|
D'Amiano AJ, Cheunkarndee T, Azoba C, Chen KY, Mak RH, Perni S. Transparency and Representation in Clinical Research Utilizing Artificial Intelligence in Oncology: A Scoping Review. Cancer Med 2025; 14:e70728. [PMID: 40059400 PMCID: PMC11891267 DOI: 10.1002/cam4.70728] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 05/13/2025] Open
Abstract
INTRODUCTION Artificial intelligence (AI) has significant potential to improve health outcomes in oncology. However, as AI utility increases, it is imperative to ensure that these models do not systematize racial and ethnic bias and further perpetuate disparities in health. This scoping review evaluates the transparency of demographic data reporting and diversity of participants included in published clinical studies utilizing AI in oncology. METHODS We utilized PubMed to search for peer-reviewed research articles published between 2016 and 2021 with the query type "("deep learning" or "machine learning" or "neural network" or "artificial intelligence") and ("neoplas$" or "cancer$" or "tumor$" or "tumour$")." We included clinical trials and original research studies and excluded reviews and meta-analyses. Oncology-related studies that described data sets used in training or validation of the AI models were eligible. Data regarding public reporting of patient demographics were collected, including age, sex at birth, and race. We used descriptive statistics to analyze these data across studies. RESULTS Out of 220 total studies, 118 were eligible and 47 (40%) had at least one described training or validation data set publicly available. 69 studies (58%) reported age data for patients included in training or validation sets, 60 studies (51%) reported sex, and six studies (5%) reported race. Of the studies that reported race, a range of 70.7%-93.4% of individuals were White. Only three studies reported racial demographic data with greater than two categories (i.e. "White" vs. "non-White" or "White" vs. "Black"). CONCLUSIONS We found that a minority of studies (5%) analyzed reported racial and ethnic demographic data. Furthermore, studies that did report racial demographic data had few non-White patients. Increased transparency regarding reporting of demographics and greater representation in data sets is essential to ensure fair and unbiased clinical integration of AI in oncology.
Collapse
Affiliation(s)
| | | | - Chinenye Azoba
- Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Krista Y. Chen
- Johns Hopkins University School of MedicineBaltimoreMarylandUSA
| | - Raymond H. Mak
- Brigham and Women's Hospital/Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
| | - Subha Perni
- Brigham and Women's Hospital/Dana‐Farber Cancer InstituteHarvard Medical SchoolBostonMassachusettsUSA
- The University of Texas MD Anderson Cancer CenterHoustonTexasUSA
| |
Collapse
|
16
|
Zeng Q, Deng Y, Nan J, Zou Z, Yu T, Liu L. Delta dual‑region DCE-MRI radiomics from breast masses predicts axillary lymph node response after neoadjuvant therapy for breast cancer. BMC Cancer 2025; 25:264. [PMID: 39953506 PMCID: PMC11827315 DOI: 10.1186/s12885-025-13678-z] [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: 10/23/2024] [Accepted: 02/06/2025] [Indexed: 02/17/2025] Open
Abstract
OBJECTIVES This study was designed to develop and validate models based on delta intratumoral and peritumoral radiomics features from breast masses on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for the prediction of axillary lymph node (ALN) pathological complete response (pCR) after neoadjuvant therapy (NAT) in patients with breast cancer (BC). METHODS We retrospectively collected data from 187 BC patients with ALN metastases. Radiomics features were extracted from the intratumoral and 3 mm-peritumoral regions on DCE-MRI at baseline and after the 2nd course of NAT to calculate delta intratumoral and peritumoral radiomics features, respectively. After feature selection, the delta intratumoral radiomics (DIR) model and delta peritumoral radiomics (DPR) model were built using the retained features. An ultrasound model was constructed on the basis of preoperative axillary ultrasound results. All variables were screened by univariate and multivariate logistic regression to construct the combined model. The above models were evaluated and compared. RESULTS In the validation set, the ultrasound model had the lowest AUC, which was lower than those of the DIR, DPR and combined models (0.627 vs 0.825, 0.687, 0.846, respectively). The combined model constructed by delta dual-region radiomics and ultrasound dianogsis was significantly better than the ultrasound model in terms of the Delong test and integrated discrimination improvement (all p < 0.05). CONCLUSIONS Delta intratumoral and peritumoral radiomics based on DCE-MRI have the potential to predict ALN status after NAT. The combined model based on delta dual-region radiomics of breast mass can accurately diagnose ALN-pCR and provide assistance in the selection of axillary surgical approaches for patients.
Collapse
Affiliation(s)
- Qiao Zeng
- Department of Radiology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Yiwen Deng
- Department of Ultrasound, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Jiayu Nan
- Department of Radiology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China
| | - Zhennan Zou
- Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital & Institute, Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang, China.
| |
Collapse
|
17
|
Duenweg SR, Bobholz SA, Lowman A, Winiarz A, Nath B, Barrett MJ, Kyereme F, Vincent-Sheldon S, LaViolette P. Comparison of intensity normalization methods in prostate, brain, and breast cancer multi-parametric magnetic resonance imaging. Front Oncol 2025; 15:1433444. [PMID: 39990680 PMCID: PMC11842255 DOI: 10.3389/fonc.2025.1433444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 01/20/2025] [Indexed: 02/25/2025] Open
Abstract
Objectives Intensity variation in multi-parametric magnetic resonance imaging (MP-MRI) is a confounding factor in MRI analyses. Previous studies have employed several normalization methods, but there is a lack of consensus on which method results in the most comparable images across vendors and acquisitions. This study used MP-MRI collected from patients with confirmed prostate, brain, or breast cancer to examine common intensity normalization methods to identify which best harmonizes intensity values across cofounds. Materials and methods Multiple normalization methods were deployed for intensity comparison between three unique sites, MR vendors, and magnetic field strength. Additionally, we calculated radiomic features before and after intensity normalization to determine how downstream analyses may be affected. Specifically, in the prostate cancer cohort, we tested these methods on T2-weighted imaging (T2WI) and additionally looked at a subset of patients who were scanned with and without the use of an endorectal coil (ERC). In a cohort of glioblastoma (GBM) patients, we tested these methods in T1 pre- and post-contrast enhancement (T1, T1C), fluid attenuated inversion recovery (FLAIR), and apparent diffusion coefficient (ADC) maps. Finally, in the breast cancer cohort, we tested methods on T1-weighted nonfat-suppressed images. All methods were compared using a two one-sided test (TOST) to test for equivalence of mean and standard deviation of intensity distributions. Results While each organ had unique results, across every tested comparison, using the Z-score of intensity within a mask of the organ consistently provided an equivalent distribution (all p < 0.001). Conclusions Our results suggest that intensity normalization using the Z-score of intensity within prostate, breast, and brain MR images produces the most comparable intensities between sites, MR vendors, magnetic field strength, and prostate endorectal coil usage. Likewise, Z-score normalization provided the highest percentage of radiomic features that were statistically equal across the three organs.
Collapse
Affiliation(s)
- Savannah R. Duenweg
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Samuel A. Bobholz
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Allison K. Lowman
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Aleksandra Winiarz
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Biprojit Nath
- Department of Biophysics, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Michael J. Barrett
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | - Fitzgerald Kyereme
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| | | | - Peter LaViolette
- Department of Radiology, Medical College of Wisconsin, Milwaukee, WI, United States
| |
Collapse
|
18
|
Wang J, Dong Z, He H, Gao Z, Huang Y, Yuan G, Jiang L, Zhao M. Integrating manual annotation with deep transfer learning and radiomics for vertebral fracture analysis. BMC Med Imaging 2025; 25:41. [PMID: 39915711 PMCID: PMC11800457 DOI: 10.1186/s12880-025-01573-9] [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: 08/25/2024] [Accepted: 01/28/2025] [Indexed: 02/11/2025] Open
Abstract
BACKGROUND Vertebral compression fractures (VCFs) are prevalent in the elderly, often caused by osteoporosis or trauma. Differentiating acute from chronic VCFs is vital for treatment planning, but MRI, the gold standard, is inaccessible for some. However, CT, a more accessible alternative, lacks precision. This study aimed to enhance CT's diagnostic accuracy for VCFs using deep transfer learning (DTL) and radiomics. METHODS We retrospectively analyzed 218 VCF patients scanned with CT and MRI within 3 days from Oct 2022 to Feb 2024. MRI categorized VCFs. 3D regions of interest (ROIs) from CT scans underwent feature extraction and DTL modeling. Receiver operating characteristic (ROC) analysis evaluated models, with the best fused with radiomic features via LASSO. AUCs compared via Delong test, and clinical utility assessed by decision curve analysis (DCA). RESULTS Patients were split into training (175) and test (43) sets. Traditional radiomics with LR yielded AUCs of 0.973 (training) and 0.869 (test). Optimal DTL modeling improved to 0.992 (training) and 0.941 (test). Feature fusion further boosted AUCs to 1.000 (training) and 0.964 (test). DCA validated its clinical significance. CONCLUSION The feature fusion model enhances the differential diagnosis of acute and chronic VCFs, outperforming single-model approaches and offering a valuable decision-support tool for patients unable to undergo spinal MRI.
Collapse
Affiliation(s)
- Jing Wang
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhirui Dong
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Huanxin He
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Zhiyang Gao
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Yukai Huang
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China
| | - Guangcheng Yuan
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Libo Jiang
- Department of Orthopaedic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China.
| | - Mingdong Zhao
- Department of Orthopaedic Surgery, Jinshan Hospital, Fudan University, Shanghai, China.
| |
Collapse
|
19
|
Wang H, Qiu J, Lu W, Xie J, Ma J. Radiomics based on multiple machine learning methods for diagnosing early bone metastases not visible on CT images. Skeletal Radiol 2025; 54:335-343. [PMID: 39028463 DOI: 10.1007/s00256-024-04752-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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 07/20/2024]
Abstract
OBJECTIVES This study utilizes [99mTc]-methylene diphosphate (MDP) single photon emission computed tomography (SPECT) images as a reference standard to evaluate whether the integration of radiomics features from computed tomography (CT) and machine learning algorithms can identify microscopic early bone metastases. Additionally, we also determine the optimal machine learning approach. MATERIALS AND METHODS We retrospectively studied 63 patients with early bone metastasis from July 2020 to March 2023. The ITK-SNAP software was used to delineate early bone metastases and normal bone tissue in SPECT images of each patient, which were then registered onto CT images to outline the volume of interest (VOI). The VOI includes 63 early bone metastasis volumes and 63 normal bone tissue volumes. 126 VOIs were randomly distributed in a 7:3 ratio between the training and testing groups, and 944 radiomics features were extracted from every VOI. We established 20 machine learning models using 5 feature selection algorithms and 4 classification methods. Evaluate the performance of the model using the area under the receiver operating characteristic curve (AUC). RESULTS Most machine learning models demonstrated outstanding discriminative capacity, with AUCs higher than 0.70. Notably, the K-Nearest Neighbors (KNN) classifier exhibited significant performance improvement compared to the other four classifiers. Specifically, the model constructed utilizing eXtreme Gradient Boosting (XGBoost) feature selection method integrated with KNN classifier achieved the maximum AUC, which is 0.989 in the training set and 0.975 in the testing set. CONCLUSIONS Radiomics features integrated with machine learning methods can identify early bone metastases that are not visible on CT images. In our analysis, KNN is considered the optimal classification method.
Collapse
Affiliation(s)
- Huili Wang
- College of Preventive Medicine & Institute of Radiation Medicine, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250012, China
| | - Jianfeng Qiu
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China
| | - Weizhao Lu
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China
| | - Jindong Xie
- College of Preventive Medicine & Institute of Radiation Medicine, Shandong First Medical University (Shandong Academy of Medical Sciences), Jinan, 250012, China.
| | - Junchi Ma
- School of Radiology, Shandong First Medical University (Shandong Academy of Medical Sciences), Taian, 271016, China.
| |
Collapse
|
20
|
Yan T, Yan Z, Chen G, Xu S, Wu C, Zhou Q, Wang G, Li Y, Jia M, Zhuang X, Yang J, Liu L, Wang L, Wu Q, Wang B, Yan T. Survival outcome prediction of esophageal squamous cell carcinoma patients based on radiomics and mutation signature. Cancer Imaging 2025; 25:9. [PMID: 39891186 PMCID: PMC11783911 DOI: 10.1186/s40644-024-00821-5] [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: 12/26/2023] [Accepted: 12/29/2024] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The present study aimed to develop a nomogram model for predicting overall survival (OS) in esophageal squamous cell carcinoma (ESCC) patients. METHODS A total of 205 patients with ESCC were enrolled and randomly divided into a training cohort (n = 153) and a test cohort (n = 52) at a ratio of 7:3. Multivariate Cox regression was used to construct the radiomics model based on CT data. The mutation signature was constructed based on whole genome sequencing data and found to be significantly associated with the prognosis of patients with ESCC. A nomogram model combining the Rad-score and mutation signature was constructed. An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors was constructed. RESULTS A total of 8 CT features were selected for multivariate Cox regression analysis to determine whether the Rad-score was significantly correlated with OS. The area under the curve (AUC) of the radiomics model was 0.834 (95% CI, 0.767-0.900) for the training cohort and 0.733 (95% CI, 0.574-0.892) for the test cohort. The Rad-score, S3, and S6 were used to construct an integrated RM nomogram. The predictive performance of the RM nomogram model was better than that of the radiomics model, with an AUC of 0. 830 (95% CI, 0.761-0.899) in the training cohort and 0.793 (95% CI, 0.653-0.934) in the test cohort. The Rad-score, TNM stage, lymph node metastasis status, S3, and S6 were used to construct an integrated RMC nomogram. The predictive performance of the RMC nomogram model was better than that of the radiomics model and RM nomogram model, with an AUC of 0. 862 (95% CI, 0.795-0.928) in the training cohort and 0. 837 (95% CI, 0.705-0.969) in the test cohort. CONCLUSION An integrated nomogram model combining the Rad-score, mutation signature, and clinical factors can better predict the prognosis of patients with ESCC.
Collapse
Affiliation(s)
- Ting Yan
- Second Clinical Medical College, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Zhenpeng Yan
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guohui Chen
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Songrui Xu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Chenxuan Wu
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China
| | - Qichao Zhou
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Guolan Wang
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Ying Li
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China
| | - Mengjiu Jia
- School of Computer Information Engineering, Shanxi Technology and Business University, Taiyuan, Shanxi, 030006, People's Republic of China
| | - Xiaofei Zhuang
- Department of Thoracic Surgery, Shanxi Cancer Hospital, Taiyuan, Shanxi, 030013, People's Republic of China
| | - Jie Yang
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lili Liu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Lu Wang
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Qinglu Wu
- Translational Medicine Research Center, Shanxi Medical University, Taiyuan, Shanxi, 030001, People's Republic of China
| | - Bin Wang
- College of Information and Computer, Taiyuan University of Technology, Taiyuan, Shanxi, 030024, People's Republic of China.
| | - Tianyi Yan
- School of Life Science, Beijing Institute of Technology, Beijing, People's Republic of China.
| |
Collapse
|
21
|
Liu Y, Xu L, Dou Y, He Y. AXL: shapers of tumor progression and immunosuppressive microenvironments. Mol Cancer 2025; 24:11. [PMID: 39799359 PMCID: PMC11724481 DOI: 10.1186/s12943-024-02210-9] [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: 09/26/2024] [Accepted: 12/24/2024] [Indexed: 01/15/2025] Open
Abstract
As research progresses, our understanding of the tumor microenvironment (TME) has undergone profound changes. The TME evolves with the developmental stages of cancer and the implementation of therapeutic interventions, transitioning from an immune-promoting to an immunosuppressive microenvironment. Consequently, we focus intently on the significant role of the TME in tumor proliferation, metastasis, and the development of drug resistance. AXL is highly associated with tumor progression; however, previous studies on AXL have been limited to its impact on the biological behavior of cancer cells. An increasing body of research now demonstrates that AXL can influence the function and differentiation of immune cells, mediating immune suppression and thereby fostering tumor growth. A comprehensive analysis to identify and overcome the causes of immunosuppressive microenvironments represents a novel approach to conquering cancer. In this review, we focus on elucidating the role of AXL within the immunosuppressive microenvironments, discussing and analyzing the effects of AXL on tumor cells, T cells, macrophages, natural killer (NK) cells, fibroblasts, and other immune-stromal cells. We aim to clarify the contributions of AXL to the progression and drug resistance of cancer from its functional role in the immune microenvironment.
Collapse
Affiliation(s)
- Yihui Liu
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
| | - Lei Xu
- Department of Otolaryngology, Southwest Hospital, Army Medical University, Chongqing, 400000, China
| | - Yuanyao Dou
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yong He
- Department of Respiratory Disease, Daping Hospital, Army Medical University, Chongqing, 400042, China.
| |
Collapse
|
22
|
Dai M, Yan Y, Li Z, Xiao J. Machine-learning models for differentiating benign and malignant breast masses: Integrating automated breast volume scanning intra-tumoral, peri-tumoral features, and clinical information. Digit Health 2025; 11:20552076251332738. [PMID: 40177119 PMCID: PMC11963789 DOI: 10.1177/20552076251332738] [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: 11/07/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Differentiating between benign and malignant breast masses is critical for clinical decision-making. Automated breast volume scanning (ABVS) provides high-resolution three-dimensional imaging, addressing the limitations of conventional ultrasound. However, the impact of peritumoral region size on predictive performance has not been systematically studied. This study aims to optimize diagnostic performance by integrating radiomics features and clinical data using multiple machine-learning models. Methods This retrospective study included ABVS images and clinical data from 250 patients with breast masses. Radiomics features were extracted from both intratumoral and peritumoral regions (5, 10, and 20 mm). These features, combined with clinical data, were used to develop models based on four algorithms: Support vector machine, random forest, extreme gradient boosting, and light gradient boosting machine (LGBM). Model performance was evaluated using area under the receiver operating characteristic curve (AUC), calibration curves, and decision curves, with SHapley Additive exPlanations (SHAP) analysis employed for interpretability. Results The inclusion of peritumoral features improved the diagnostic performance to varying degrees, with the model incorporating a 10 mm peritumoral region achieving the highest overall accuracy. Combining radiomics with clinical features further enhanced predictive performance. The LGBM model outperformed the other algorithms across subgroups, achieving a maximum AUC of 0.909, an accuracy of 0.878, and an F1-score of 0.971. SHAP analysis revealed the contribution of key features, improving model interpretability. Conclusion This study demonstrates the value of integrating radiomics and clinical features for breast mass diagnosis, with optimized peritumoral regions enhancing model performance. The LGBM model emerged as the preferred algorithm due to its superior performance. These findings provide strong support for the clinical application of ABVS imaging and future multicenter studies, highlighting the importance of microenvironmental features in diagnosis.
Collapse
Affiliation(s)
- Meixue Dai
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Yueqiong Yan
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha, China
| | - Zhong Li
- Department of Orthodontics, Hunan Xiangya Stomatological Hospital, Central South University, Changsha, China
| | - Jidong Xiao
- Department of Ultrasound, The Third Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
23
|
Zhong H, Lai Y, Ouyang W, Yu Y, Wu Y, He X, Zeng L, Qiu X, Chen P, Li L, Zhou J, Luo T, Huang H. Integrative analysis of cuproptosis-related lncRNAs: Unveiling prognostic significance, immune microenvironment, and copper-induced mechanisms in prostate cancer. CANCER PATHOGENESIS AND THERAPY 2025; 3:48-59. [PMID: 39872368 PMCID: PMC11764251 DOI: 10.1016/j.cpt.2024.03.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 03/17/2024] [Accepted: 03/26/2024] [Indexed: 10/31/2024]
Abstract
BACKGROUND Long non-coding ribonucleic acids (lncRNAs) regulate messenger RNA (mRNA) expression and influence cancer development and progression. Cuproptosis, a newly discovered form of cell death, plays an important role in cancer. Nonetheless, additional research investigating the association between cuproptosis-related lncRNAs and prostate cancer (PCa) prognosis is required. METHODS Sequencing data and copy number variant data were obtained from 492 patients with PCa from The Cancer Genome Atlas (TCGA) Program. Prognostic models of PCa based on cuproptosis-related lncRNAs were constructed using a multi-level attention graph neural network (MLA-GNN) deep learning algorithm. Immune escape scoring was performed using Tumor Immune Dysfunction and Exclusion. Cellular experiments were conducted to explore the correlation between key lncRNAs and cuproptosis. RESULTS Data from 492 patients with PCa were randomized into two groups at a 1:1 ratio. Prognostic modeling was successfully established using MLA-GNN. Survival analysis suggested that patients could be divided into high- and low-risk groups according to model scores and that there was a significant difference in disease-free survival (DFS) (P < 0.01). The area under the receiver operating characteristic (ROC) curve (AUC) indicated a strong predictive performance for the model, with AUCs of 0.913, 0.847, and 0.863 for the training group and 0.815, 0.907, and 0.866 for the test group at 12, 36, and 60 months, respectively. The immune escape score and immune microenvironment analysis suggested that the high-risk group corresponded to a stronger immune escape and a poorer immune microenvironment (P < 0.05). Cellular experiments revealed that the expression of all six key lncRNAs was upregulated in the presence of copper ion carriers (P < 0.05). CONCLUSIONS This study identified cuproptosis-related lncRNAs that were strongly associated with PCa prognosis. Key lncRNAs could affect copper metabolism and may serve as new therapeutic targets.
Collapse
Affiliation(s)
- Haitao Zhong
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Yiming Lai
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, The Fifth Affiliated Hospital of Xinjiang Medical University, Urumqi, Xinjiang 830011, China
| | - Wenhao Ouyang
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Yunfang Yu
- Department of Medical Oncology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Faculty of Medicine, Macau University of Science and Technology, Avenida WaiLong, Taipa, Macao 999078, China
| | - Yongxin Wu
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Xinxin He
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Lexiang Zeng
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Xueen Qiu
- Department of Emergency, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Peixian Chen
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Lingfeng Li
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Jie Zhou
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Tianlong Luo
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
| | - Hai Huang
- Department of Urology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Guangdong Provincial Clinical Research Center for Urological Diseases, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong 510120, China
- Department of Urology, The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan, Guangdong 511518, China
| |
Collapse
|
24
|
Feng FW, Jiang FY, Liu YQ, Sun Q, Hong R, Hu CH, Hu S. Radiomics analysis of dual-layer spectral-detector CT-derived iodine maps for predicting tumor deposits in colorectal cancer. Eur Radiol 2025; 35:105-116. [PMID: 38987399 DOI: 10.1007/s00330-024-10918-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: 12/06/2023] [Revised: 04/24/2024] [Accepted: 05/25/2024] [Indexed: 07/12/2024]
Abstract
OBJECTIVE To investigate the value of radiomics analysis of dual-layer spectral-detector computed tomography (DLSCT)-derived iodine maps for predicting tumor deposits (TDs) preoperatively in patients with colorectal cancer (CRC). MATERIALS AND METHODS A total of 264 pathologically confirmed CRC patients (TDs + (n = 80); TDs - (n = 184)) who underwent preoperative DLSCT from two hospitals were retrospectively enrolled, and divided into training (n = 124), testing (n = 54), and external validation cohort (n = 86). Conventional CT features and iodine concentration (IC) were analyzed and measured. Radiomics features were derived from venous phase iodine maps from DLSCT. The least absolute shrinkage and selection operator (LASSO) was performed for feature selection. Finally, a support vector machine (SVM) algorithm was employed to develop clinical, radiomics, and combined models based on the most valuable clinical parameters and radiomics features. Area under receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis were used to evaluate the model's efficacy. RESULTS The combined model incorporating the valuable clinical parameters and radiomics features demonstrated excellent performance in predicting TDs in CRC (AUCs of 0.926, 0.881, and 0.887 in the training, testing, and external validation cohorts, respectively), which outperformed the clinical model in the training cohort and external validation cohorts (AUC: 0.839 and 0.695; p: 0.003 and 0.014) and the radiomics model in two cohorts (AUC: 0.922 and 0.792; p: 0.014 and 0.035). CONCLUSION Radiomics analysis of DLSCT-derived iodine maps showed excellent predictive efficiency for preoperatively diagnosing TDs in CRC, and could guide clinicians in making individualized treatment strategies. CLINICAL RELEVANCE STATEMENT The radiomics model based on DLSCT iodine maps has the potential to aid in the accurate preoperative prediction of TDs in CRC patients, offering valuable guidance for clinical decision-making. KEY POINTS Accurately predicting TDs in CRC patients preoperatively based on conventional CT features poses a challenge. The Radiomics model based on DLSCT iodine maps outperformed conventional CT in predicting TDs. The model combing DLSCT iodine maps radiomics features and conventional CT features performed excellently in predicting TDs.
Collapse
Affiliation(s)
- Fei-Wen Feng
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Fei-Yu Jiang
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yuan-Qing Liu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Medical Imaging, Soochow University, Suzhou, China
| | - Qi Sun
- Department of Radiology, Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Rong Hong
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Chun-Hong Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Medical Imaging, Soochow University, Suzhou, China.
| | - Su Hu
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China.
- Institute of Medical Imaging, Soochow University, Suzhou, China.
| |
Collapse
|
25
|
Guo F, Sun S, Deng X, Wang Y, Yao W, Yue P, Wu S, Yan J, Zhang X, Zhang Y. Predicting axillary lymph node metastasis in breast cancer using a multimodal radiomics and deep learning model. Front Immunol 2024; 15:1482020. [PMID: 39735531 PMCID: PMC11671510 DOI: 10.3389/fimmu.2024.1482020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2024] [Accepted: 11/27/2024] [Indexed: 12/31/2024] Open
Abstract
Objective To explore the value of combined radiomics and deep learning models using different machine learning algorithms based on mammography (MG) and magnetic resonance imaging (MRI) for predicting axillary lymph node metastasis (ALNM) in breast cancer (BC). The objective is to provide guidance for developing scientifically individualized treatment plans, assessing prognosis, and planning preoperative interventions. Methods A retrospective analysis was conducted on clinical and imaging data from 270 patients with BC confirmed by surgical pathology at the Third Hospital of Shanxi Medical University between November 2022 and April 2024. Multiple sequence images from MG and MRI were selected, and regions of interest in the lesions were delineated. Radiomics and deep learning (3D-Resnet18) features were extracted and fused. The samples were randomly divided into training and test sets in a 7:3 ratio. Dimensionality reduction and feature selection were performed using the least absolute shrinkage and selection operator (LASSO) regression model, and other methods. Various machine learning algorithms were used to construct radiomics, deep learning, and combined models. These models were visualized and evaluated for performance using receiver operating characteristic curves, area under the curve (AUC), calibration curves, and decision curves. Results The highest AUCs in the test set were achieved using radiomics-logistic regression (AUC = 0.759), deep learning-multilayer perceptron (MLP) (AUC = 0.712), and combined-MLP models (AUC = 0.846). The MLP model demonstrated strong classification performance, with the combined model (AUC = 0.846) outperforming both the radiomics (AUC = 0.756) and deep learning (AUC = 0.712) models. Conclusion The multimodal radiomics and deep learning models developed in this study, incorporating various machine learning algorithms, offer significant value for the preoperative prediction of ALNM in BC.
Collapse
Affiliation(s)
- Fuyu Guo
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Shiwei Sun
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xiaoqian Deng
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Yue Wang
- Department of Urology, Xinzhou People’s Hospital, Xinzhou, China
| | - Wei Yao
- Department of Urology, Datong Fifth People’s Hospital, Datong, China
| | - Peng Yue
- Department of Urology, Handan First Hospital, Handan, China
| | - Shaoduo Wu
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
| | - Junrong Yan
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiaojun Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yangang Zhang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, China
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Dong F, Li J, Wang J, Yang X. Diagnostic performance of DCE-MRI radiomics in predicting axillary lymph node metastasis in breast cancer patients: A meta-analysis. PLoS One 2024; 19:e0314653. [PMID: 39625963 PMCID: PMC11614294 DOI: 10.1371/journal.pone.0314653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Accepted: 11/13/2024] [Indexed: 12/06/2024] Open
Abstract
Radiomics offers a novel strategy for the differential diagnosis, prognosis evaluation, and prediction of treatment responses in breast cancer. Studies have explored radiomic signatures from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for predicting axillary lymph node metastasis (ALNM) and sentinel lymph node metastasis (SLNM), but the diagnostic accuracy varies widely. To evaluate this performance, we conducted a meta-analysis performing a comprehensive literature search across databases including PubMed, EMBASE, SCOPUS, Web of Science (WOS), Cochrane Library, China National Knowledge Infrastructure (CNKI), Wanfang Data, and the Chinese BioMedical Literature Database (CBM) until March 31, 2024. The pooled sensitivity, specificity, positive likelihood ratio (PLR), negative likelihood ratio (NLR), diagnostic odds ratio (DOR), and the area under the receiver operating characteristic curve (AUC) were calculated. Twenty-four eligible studies encompassing 5588 breast cancer patients were included in the meta-analysis. The meta-analysis yielded a pooled sensitivity of 0.81 (95% confidence interval [CI]: 0.77-0.84), specificity of 0.85 (95%CI: 0.81-0.87), PLR of 5.24 (95%CI: 4.32-6.34), NLR of 0.23 (95%CI: 0.19-0.27), DOR of 23.16 (95%CI: 17.20-31.19), and AUC of 0.90 (95%CI: 0.87-0.92), indicating good diagnostic performance. Significant heterogeneity was observed in analyses of sensitivity (I2 = 74.64%) and specificity (I2 = 83.18%). Spearman's correlation coefficient suggested no significant threshold effect (P = 0.538). Meta-regression and subgroup analyses identified several potential heterogeneity sources, including data source, integration of clinical factors and peritumor features, MRI equipment, magnetic field strength, lesion segmentation, and modeling methods. In conclusion, DCE-MRI radiomic models exhibit good diagnostic performance in predicting ALNM and SLNM in breast cancer. This non-invasive and effective tool holds potential for the preoperative diagnosis of lymph node metastasis in breast cancer patients.
Collapse
Affiliation(s)
- Fei Dong
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Jie Li
- Department of Anesthesiology, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Junbo Wang
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| | - Xiaohui Yang
- Department of Medical Imaging, Yuncheng Central Hospital Affiliated to Shanxi Medical University, Yuncheng, Shanxi Province, China
| |
Collapse
|
27
|
Yu Y, Cai G, Lin R, Wang Z, Chen Y, Tan Y, He Z, Sun Z, Ouyang W, Yao H, Zhang K. Multimodal data fusion AI model uncovers tumor microenvironment immunotyping heterogeneity and enhanced risk stratification of breast cancer. MedComm (Beijing) 2024; 5:e70023. [PMID: 39669975 PMCID: PMC11635117 DOI: 10.1002/mco2.70023] [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: 02/18/2024] [Revised: 08/06/2024] [Accepted: 08/15/2024] [Indexed: 12/14/2024] Open
Abstract
Breast cancer is the leading cancer among women, with a significant number experiencing recurrence and metastasis, thereby reducing survival rates. This study focuses on the role of long noncoding RNAs (lncRNAs) in breast cancer immunotherapy response. We conducted an analysis involving 1027 patients from Sun Yat-sen Memorial Hospital, Sun Yat-sen University, and The Cancer Genome Atlas, utilizing RNA sequencing and pathology whole-slide images. We employed unsupervised clustering to identify distinct lncRNA expression patterns and developed an AI-based pathology model using convolutional neural networks to predict immune-metabolic subtypes. Additionally, we created a multimodal model integrating lncRNA data, immune-cell scores, clinical information, and pathology images for prognostic prediction. Our findings revealed four unique immune-metabolic subtypes, and the AI model demonstrated high predictive accuracy, highlighting the significant impact of lncRNAs on antitumor immunity and metabolic states within the tumor microenvironment. The AI-based pathology model, DeepClinMed-IM, exhibited high accuracy in predicting these subtypes. Additionally, the multimodal model, DeepClinMed-PGM, integrating pathology images, lncRNA data, immune-cell scores, and clinical information, showed superior prognostic performance. In conclusion, these AI models provide a robust foundation for precise prognostication and the identification of potential candidates for immunotherapy, advancing breast cancer research and treatment strategies.
Collapse
Affiliation(s)
- Yunfang Yu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
- Faculty of MedicineMacau University of Science and TechnologyTaipaMacaoChina
| | - Gengyi Cai
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Ruichong Lin
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
| | - Zehua Wang
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
| | - Yongjian Chen
- Dermatology and Venereology Division, Department of Medicine Solna, Center for Molecular MedicineKarolinska InstituteStockholmSweden
| | - Yujie Tan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zifan He
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Zhuo Sun
- Institute for Advanced Study on Eye Health and DiseasesWenzhou Medical UniversityWenzhouChina
| | - Wenhao Ouyang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Herui Yao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Department of Medical OncologyBreast Tumor Centre, Phase I Clinical Trial Centre, Sun Yat‐sen Memorial Hospital, Sun Yat‐sen UniversityGuangzhouChina
| | - Kang Zhang
- Faculty of MedicineMacau University of Science and TechnologyTaipaMacaoChina
- Faculty of Innovation EngineeringMacau University of Science and TechnologyTaipaMacauChina
- Institute for Advanced Study on Eye Health and DiseasesWenzhou Medical UniversityWenzhouChina
- Guangzhou National LaboratoryGuangzhouChina
- Zhuhai International Eve CenterZhuhai People's Hospital and the First Affiliated Hospital of Faculty of Medicine, Macau University of Science and Technology and University HospitalZhuhaiChina
| |
Collapse
|
28
|
Rong RY, Shen YK, Wu SN, Xu SH, Hu JY, Zou J, He L, Chen C, Kang M, Ying P, Wei H, Ling Q, Ge QM, Lou Y, Shao Y. Prediction model for ocular metastasis of breast cancer: machine learning model development and interpretation study. BMC Cancer 2024; 24:1472. [PMID: 39614215 DOI: 10.1186/s12885-024-12928-w] [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: 05/11/2024] [Accepted: 09/10/2024] [Indexed: 12/01/2024] Open
Abstract
BACKGROUND Breast cancer (BC) is caused by the uncontrolled proliferation of breast epithelial cells followed by malignant transformation, and it has the highest incidence among female malignant tumors. The metastasis of BC occurs through direct and lymphatic spread. Although ocular metastasis is relatively rare, it is a good indicator of a worse prognosis. We used machine learning (ML) to establish a model to analyze the risk factors of BC eye metastasis. METHODS The clinical data of 2225 patients with BC from 2003 to 2019 were collected and randomly classified into the training and test sets using a ratio of 7:3. Based on the presence or absence of eye metastasis, the patients with BC were classified into the ocular metastasis (OM) and non-ocular metastasis (NOM) groups. Univariate and multivariate logistic regression analyses and least absolute shrinkage and selection operator (LASSO) were conducted. We used six ML algorithms to establish a predictive BC model and used 10-fold cross-validation for internal verification. The area under the receiver operating characteristic (ROC) curve was used to evaluate the predictive ability of the model. In addition, we established a web hazard calculator depending on the best-performing model to facilitate its clinical application. Shapley additive interpretation (SHAP) was used to determine the risk factors and the interpretability of the black box model. RESULTS Univariate logistic regression analysis showed that histopathology (other types), axillary lymph node metastasis (ALNM) (> 4), Ca2+, total cholesterol (TC), low-density lipoprotein (LDL), apolipoprotein A (ApoA), carcinoembryonic antigen (CEA), carbohydrate antigen (CA) 125, CA153, CA199, alkaline phosphatase (ALP), and hemoglobin (Hb) were risk factors for BC eye metastasis. Multivariate logistic regression analysis showed that CA153, ApoA, and LDL were hazardous components for BC eye metastasis. LASSO showed that ALNM, LDL, CA125, Hb, ALP, and CA199 were the first six key variables that were useful for the diagnosis of ocular metastasis in breast cancer. Bootstrapped aggregation (BAG) demonstrated the discriminative ability (area under ROC curve [AUC] = 0.992, accuracy = 0.953, sensitivity = 0.987). Based on this, we applied the BAG machine learning model to build an online web computing system to help clinicians assist in determining the risk of BC eye metastasis. In addition, two typical cases are analyzed to determine the interpretability of the model. CONCLUSION We used ML to establish a risk prediction model for BC ocular metastasis, and BAG showed the greatest performance. The model can predict the risk of OM in patients with BC, facilitate early and timely diagnosis and treatment, and reduce the burden on society.
Collapse
Affiliation(s)
- Ru-Yi Rong
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
| | - Yan-Kun Shen
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of General Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200030, China
| | - Shi-Nan Wu
- School of Medicine, Eye Institute of Xiamen University, Xiamen University, Xiamen, Fujian, China
| | - San-Hua Xu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jin-Yu Hu
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Jie Zou
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Liangqi He
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Cheng Chen
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Min Kang
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Ping Ying
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Hong Wei
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qian Ling
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Qian-Ming Ge
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China
| | - Yan Lou
- School of Medical Information and Engineering, Southwest Medical University, Luzhou, Sichuan, 646000, China.
| | - Yi Shao
- Department of Ophthalmology, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi Province, 330006, China.
- Department of Ophthalmology, Shanghai General Hospital, National Clinical Research Center for Eye Diseases, Shanghai Key Laboratory of Ocular Fundus Diseases, Shanghai Engineering Center for Visual Science and Photomedicine, Shanghai Engineering Center for Precise Diagnosis and Treatment of Eye Diseases, Shanghai Jiao Tong University School of Medicine, Shanghai, 200080, China.
| |
Collapse
|
29
|
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
|
30
|
Xia Y, Yang M, Qian T, Zhou J, Bai M, Luo S, Lu C, Zhu Y, Wang L, Qiao Z. Prediction of feeding difficulties in neonates with hypoxic-ischemic encephalopathy using magnetic resonance imaging-derived radiomics features. Pediatr Radiol 2024; 54:2036-2045. [PMID: 39349660 DOI: 10.1007/s00247-024-06065-6] [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: 03/25/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND The mechanisms behind brain and spinal cord injuries in hypoxic-ischemic encephalopathy (HIE) and associated feeding difficulties are unclear, with previous magnetic resonance imaging (MRI) attempts yielding inconclusive results. OBJECTIVE We aim to evaluate an MRI radiomics model for predicting feeding difficulties in HIE infants. Additionally, we investigate changes in predictive capability after incorporating the duration of mechanical ventilation and the timing of MRI examination. MATERIALS AND METHODS Retrospective study with 151 HIE infants (January 2013 to December 2021), randomly divided into training and validation sets. Radiomics features extracted from basal ganglia-thalamus and brainstem in T1-weighted and T2-weighted MRI. Established single-modality, single-site, and multimodality/multisite models. Receiver operating characteristic analysis and area under the curve evaluated models. Decision curve analysis assessed changes in predictive capability. RESULTS The combined radiomics model of the basal ganglia-thalamus and brainstem regions on the T2-weighted imaging demonstrated superior performance (area under the curve: 0.958 and 0.875 for training and validation, respectively). Combining scores with duration of mechanical ventilation and MRI examination time in a calibration plot model improved and stabilized performance, showing high fitting and clinical utility. Decision curve analysis favored the combined calibration plot model. CONCLUSION The MRI-based radiomics model predicts feeding difficulties in HIE infants, with basal ganglia-thalamus and brainstem as relevant factors. The combined calibration plot model exhibits the highest clinical predictive efficacy.
Collapse
Affiliation(s)
- Yaqin Xia
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Mingshu Yang
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Tianyang Qian
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Jiayu Zhou
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Mei Bai
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Siqi Luo
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Chaogang Lu
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China
| | - Yinghao Zhu
- Institute of Artificial Intelligence, Beihang University, Beijing, China
| | - Laishuan Wang
- National Health Commission Key Laboratory of Neonatal Diseases, Department of Neonatology, Children's Hospital of Fudan University, Shanghai, China
| | - Zhongwei Qiao
- Department of Radiology, Children's Hospital of Fudan University, No. 399 Wanyuan Road, Minhang District, Shanghai, China.
| |
Collapse
|
31
|
Yan L, Chen Y, He J. Leveraging MRI radiomics signature for predicting the diagnosis of CXCL9 in breast cancer. Heliyon 2024; 10:e38640. [PMID: 39430466 PMCID: PMC11490775 DOI: 10.1016/j.heliyon.2024.e38640] [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: 02/24/2024] [Revised: 09/26/2024] [Accepted: 09/26/2024] [Indexed: 10/22/2024] Open
Abstract
Objective A non-invasive predictive model was developed using radiomic features to forecast CXCL9 expression level in breast cancer patients. Methods CXCL9 expression data and MRI images of breast cancer patients were obtained from The Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA) databases, respectively. Local tissue samples from 20 breast cancer patients were collected to measure CXCL9 expression levels. Radiomic features were extracted from MRI images using 3DSlicer, and the minimum Redundancy Maximum Relevance and Recursive Feature Elimination (mRMR_RFE) method was employed to select the most pertinent radiomic features associated with CXCL9 expression levels. Support vector machine (SVM) and Logistic Regression (LR) models were utilized to construct the predictive model, and the area under the receiver operating characteristic curve (AUC) was calculated for performance evaluation. Results CXCL9 was found to be upregulated in breast cancer patients and linked to breast cancer prognosis. Nine radiomic features were ultimately selected using the mRMR_RFE method, and SVM and LR models were trained and validated. The SVM model achieved AUC values of 0.748 and 0.711 in the training and validation sets, respectively. The LR model obtained AUC values of 0.771 and 0.724 in the training and validation sets, respectively. Conclusion The utilization of MRI radiomic features for predicting CXCL9 expression level provides a novel non-invasive approach for breast cancer Prognostic research.
Collapse
Affiliation(s)
- Liping Yan
- Department of Breast Surgery, Maternal and Child Health Hospital of Jiangxi Province, Nanchang, China
- Department of Surgery, the First Affiliated Hospital of Guangxi Medical University, China
| | - Yuexia Chen
- Department of Pathology, The Third Hospital of Nanchang, Nanchang, China
| | - Jianxin He
- Department of Ultrasound Medicine, The First Affiliated Hospital of Nanchang University, China
| |
Collapse
|
32
|
Yu Y, Chen R, Yi J, Huang K, Yu X, Zhang J, Song C. Non-invasive prediction of axillary lymph node dissection exemption in breast cancer patients post-neoadjuvant therapy: A radiomics and deep learning analysis on longitudinal DCE-MRI data. Breast 2024; 77:103786. [PMID: 39137488 PMCID: PMC11369401 DOI: 10.1016/j.breast.2024.103786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 07/15/2024] [Accepted: 08/08/2024] [Indexed: 08/15/2024] Open
Abstract
PURPOSE In breast cancer (BC) patients with clinical axillary lymph node metastasis (cN+) undergoing neoadjuvant therapy (NAT), precise axillary lymph node (ALN) assessment dictates therapeutic strategy. There is a critical demand for a precise method to assess the axillary lymph node (ALN) status in these patients. MATERIALS AND METHODS A retrospective analysis was conducted on 160 BC patients undergoing NAT at Fujian Medical University Union Hospital. We analyzed baseline and two-cycle reassessment dynamic contrast-enhanced MRI (DCE-MRI) images, extracting 3668 radiomic and 4096 deep learning features, and computing 1834 delta-radiomic and 2048 delta-deep learning features. Light Gradient Boosting Machine (LightGBM), Support Vector Machine (SVM), RandomForest, and Multilayer Perceptron (MLP) algorithms were employed to develop risk models and were evaluated using 10-fold cross-validation. RESULTS Of the patients, 61 (38.13 %) achieved ypN0 status post-NAT. Univariate and multivariable logistic regression analyses revealed molecular subtypes and Ki67 as pivotal predictors of achieving ypN0 post-NAT. The SVM-based "Data Amalgamation" model that integrates radiomic, deep learning features, and clinical data, exhibited an outstanding AUC of 0.986 (95 % CI: 0.954-1.000), surpassing other models. CONCLUSION Our study illuminates the challenges and opportunities inherent in breast cancer management post-NAT. By introducing a sophisticated, SVM-based "Data Amalgamation" model, we propose a way towards accurate, dynamic ALN assessments, offering potential for personalized therapeutic strategies in BC.
Collapse
Affiliation(s)
- Yushuai Yu
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Ruiliang Chen
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Jialu Yi
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China
| | - Kaiyan Huang
- Department of Breast and Thyroid Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian Province, 362000, China
| | - Xin Yu
- Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, 350014, China
| | - Jie Zhang
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China.
| | - Chuangui Song
- Department of Breast Surgery, Fujian Medical University Union Hospital, Fuzhou, Fujian Province, 350001, China; Department of Breast Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, 350014, China.
| |
Collapse
|
33
|
Zhang H, Ma Z, Mi H, Jiao J, Dong W, Yang S, Liu L, Zhou S, Feng L, Zhao X, Yang X, Tu C, Song X, Zhang H. Diagnostic Value of Magnetocardiography to Detect Abnormal Myocardial Perfusion: A Pilot Study. Rev Cardiovasc Med 2024; 25:379. [PMID: 39484136 PMCID: PMC11522775 DOI: 10.31083/j.rcm2510379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/03/2024] [Accepted: 05/20/2024] [Indexed: 11/03/2024] Open
Abstract
Background Magnetocardiography (MCG) is a novel non-invasive technique that detects subtle magnetic fields generated by cardiomyocyte electrical activity, offering sensitive detection of myocardial ischemia. This study aimed to assess the ability of MCG to predict impaired myocardial perfusion using single-photon emission computed tomography (SPECT). Methods A total of 112 patients with chest pain underwent SPECT and MCG scans, from which 65 MCG output parameters were analyzed. Using least absolute shrinkage and selection operator (LASSO) regression to screen for significant MCG variables, three machine learning models were established to detect impaired myocardial perfusion: random forest (RF), decision tree (DT), and support vector machine (SVM). The diagnostic performance was evaluated based on the sensitivity, specificity, accuracy, positive predictive value (PPV), negative predictive value (NPV), and area under the receiver operating characteristic curve (AUC). Results Five variables, the ratio of magnetic field amplitude at R-peak and positive T-peak (RoART+), R and T-peak magnetic field angle (RTA), maximum magnetic field angle (MAmax), maximum change in current angle (CCAmax), and change positive pole point area between the T-wave beginning and peak (CPPPATbp), were selected from 65 automatic output parameters. RTA emerged as the most critical variable in the RF, DT, and SVM models. All three models exhibited excellent diagnostic performance, with AUCs of 0.796, 0.780, and 0.804, respectively. While all models showed high sensitivity (RF = 0.870, DT = 0.826, SVM = 0.913), their specificity was comparatively lower (RF = 0.500, DT = 0.300, SVM = 0.100). Conclusions Machine learning models utilizing five key MCG variables successfully predicted impaired myocardial perfusion, as confirmed by SPECT. These findings underscore the potential of MCG as a promising future screening tool for detecting impaired myocardial perfusion. Clinical Trial Registration ChiCTR2200066942, https://www.chictr.org.cn/showproj.html?proj=187904.
Collapse
Affiliation(s)
- Huan Zhang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Zhao Ma
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Hongzhi Mi
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Jian Jiao
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Wei Dong
- Department of Nuclear Medicine, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Shuwen Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Linqi Liu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Shu Zhou
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Lanxin Feng
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xin Zhao
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xueyao Yang
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Chenchen Tu
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Xiantao Song
- Department of Cardiology, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| | - Hongjia Zhang
- Department of Cardiac Surgery, Beijing Anzhen Hospital, Capital Medical University, 100029 Beijing, China
| |
Collapse
|
34
|
Grøvik E. Editorial for "MRI-Based Kinetic Heterogeneity Evaluation in the Accurate Access of Axillary Lymph Node Status in Breast Cancer Using a Hybrid CNN-RNN Model". J Magn Reson Imaging 2024; 60:1365-1366. [PMID: 38217385 DOI: 10.1002/jmri.29226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 01/15/2024] Open
Affiliation(s)
- Endre Grøvik
- Department of Radiology, Division of Diagnostics, Møre and Romsdal Hospital Trust, Ålesund, Norway
- Department of Physics, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim Ålesund Gjøvik, Norway
| |
Collapse
|
35
|
Guo YJ, Yin R, Zhang Q, Han JQ, Dou ZX, Wang PB, Lu H, Liu PF, Chen JJ, Ma WJ. MRI-Based Kinetic Heterogeneity Evaluation in the Accurate Access of Axillary Lymph Node Status in Breast Cancer Using a Hybrid CNN-RNN Model. J Magn Reson Imaging 2024; 60:1352-1364. [PMID: 38205712 DOI: 10.1002/jmri.29225] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/20/2023] [Accepted: 12/21/2023] [Indexed: 01/12/2024] Open
Abstract
BACKGROUND Accurate evaluation of the axillary lymph node (ALN) status is needed for determining the treatment protocol for breast cancer (BC). The value of magnetic resonance imaging (MRI)-based tumor heterogeneity in assessing ALN metastasis in BC is unclear. PURPOSE To assess the value of deep learning (DL)-derived kinetic heterogeneity parameters based on BC dynamic contrast-enhanced (DCE)-MRI to infer the ALN status. STUDY TYPE Retrospective. SUBJECTS 1256/539/153/115 patients in the training cohort, internal validation cohort, and external validation cohorts I and II, respectively. FIELD STRENGTH/SEQUENCE 1.5 T/3.0 T, non-contrast T1-weighted spin-echo sequence imaging (T1WI), DCE-T1WI, and diffusion-weighted imaging. ASSESSMENT Clinical pathological and MRI semantic features were obtained by reviewing histopathology and MRI reports. The segmentation of the tumor lesion on the first phase of T1WI DCE-MRI images was applied to other phases after registration. A DL architecture termed convolutional recurrent neural network (ConvRNN) was developed to generate the KHimage (kinetic heterogeneity of DCE-MRI image) score that indicated the ALN status in patients with BC. The model was trained and optimized on training and internal validation cohorts, tested on two external validation cohorts. We compared ConvRNN model with other 10 models and the subgroup analyses of tumor size, magnetic field strength, and molecular subtype were also evaluated. STATISTICAL TESTS Chi-squared, Fisher's exact, Student's t, Mann-Whitney U tests, and receiver operating characteristics (ROC) analysis were performed. P < 0.05 was considered significant. RESULTS The ConvRNN model achieved area under the curve (AUC) of 0.802 in the internal validation cohort and 0.785-0.806 in the external validation cohorts. The ConvRNN model could well evaluate the ALN status of the four molecular subtypes (AUC = 0.685-0.868). The patients with larger tumor sizes (>5 cm) were more susceptible to ALN metastasis with KHimage scores of 0.527-0.827. DATA CONCLUSION A ConvRNN model outperformed traditional models for determining the ALN status in patients with BC. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Yi-Jun Guo
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Rui Yin
- School of Biomedical Engineering & Technology, Tianjin Medical University, Tianjin, China
| | - Qian Zhang
- Department of Radiology, Baoding No. 1 Central Hospital, Baoding, China
| | - Jun-Qi Han
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Zhao-Xiang Dou
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Peng-Bo Wang
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Hong Lu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Pei-Fang Liu
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| | - Jing-Jing Chen
- Department of Breast Imaging, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Wen-Juan Ma
- Department of Breast Imaging, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Key Laboratory of Breast Cancer Prevention and Therapy, Tianjin Medical University, Ministry of Education, Tianjin's Clinical Research Center for Cancer, Tianjin, China
| |
Collapse
|
36
|
Wang Q, Lin Y, Ding C, Guan W, Zhang X, Jia J, Zhou W, Liu Z, Bai G. Multi-modality radiomics model predicts axillary lymph node metastasis of breast cancer using MRI and mammography. Eur Radiol 2024; 34:6121-6131. [PMID: 38337068 DOI: 10.1007/s00330-024-10638-2] [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: 05/22/2023] [Revised: 12/05/2023] [Accepted: 01/20/2024] [Indexed: 02/12/2024]
Abstract
OBJECTIVES We aimed to develop a multi-modality model to predict axillary lymph node (ALN) metastasis by combining clinical predictors with radiomic features from magnetic resonance imaging (MRI) and mammography (MMG) in breast cancer. This model might potentially eliminate unnecessary axillary surgery in cases without ALN metastasis, thereby minimizing surgery-related complications. METHODS We retrospectively enrolled 485 breast cancer patients from two hospitals and extracted radiomics features from tumor and lymph node regions on MRI and MMG images. After feature selection, three random forest models were built using the retained features, respectively. Significant clinical factors were integrated with these radiomics models to construct a multi-modality model. The multi-modality model was compared to radiologists' diagnoses on axillary ultrasound and MRI. It was also used to assist radiologists in making a secondary diagnosis on MRI. RESULTS The multi-modality model showed superior performance with AUCs of 0.964 in the training cohort, 0.916 in the internal validation cohort, and 0.892 in the external validation cohort. It surpassed single-modality models and radiologists' ALN diagnosis on MRI and axillary ultrasound in all validation cohorts. Additionally, the multi-modality model improved radiologists' MRI-based ALN diagnostic ability, increasing the average accuracy from 70.70 to 78.16% for radiologist A and from 75.42 to 81.38% for radiologist B. CONCLUSION The multi-modality model can predict ALN metastasis of breast cancer accurately. Moreover, the artificial intelligence (AI) model also assisted the radiologists to improve their diagnostic ability on MRI. CLINICAL RELEVANCE STATEMENT The multi-modality model based on both MRI and mammography images allows preoperative prediction of axillary lymph node metastasis in breast cancer patients. With the assistance of the model, the diagnostic efficacy of radiologists can be further improved. KEY POINTS • We developed a novel multi-modality model that combines MRI and mammography radiomics with clinical factors to accurately predict axillary lymph node (ALN) metastasis, which has not been previously reported. • Our multi-modality model outperformed both the radiologists' ALN diagnosis based on MRI and axillary ultrasound, as well as single-modality radiomics models based on MRI or mammography. • The multi-modality model can serve as a potential decision support tool to improve the radiologists' ALN diagnosis on MRI.
Collapse
Affiliation(s)
- Qian Wang
- Department of Radiology, The Affiliated Huaian Clinical College of Xuzhou Medical University, Huaian, Jiangsu, China
| | - Yingyu Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58th, The Second Zhongshan Road, Guangzhou, Guangdong, China
| | - Cong Ding
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Wenting Guan
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Xiaoling Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University, 58th, The Second Zhongshan Road, Guangzhou, Guangdong, China
| | - Jianye Jia
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Wei Zhou
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Ziyan Liu
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China
| | - Genji Bai
- Department of Radiology, The Affiliated Huaian Clinical College of Xuzhou Medical University, Huaian, Jiangsu, China.
- Department of Radiology, The Affiliated Huaian No. 1 People's Hospital of Nanjing Medical University, Huaian, Jiangsu, China.
| |
Collapse
|
37
|
Shu K, Wang K, Zhang R, Wang C, Cai Z, Liu K, Lin H, Zeng Y, Cao Z, Lai C, Yan Z, Lu Y. Pituitary MRI Radiomics Improves Diagnostic Performance of Growth Hormone Deficiency in Children Short Stature: A Multicenter Radiomics Study. Acad Radiol 2024; 31:3783-3792. [PMID: 38796401 DOI: 10.1016/j.acra.2024.05.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Revised: 05/06/2024] [Accepted: 05/07/2024] [Indexed: 05/28/2024]
Abstract
RATIONALE AND OBJECTIVES To develop an efficient machine-learning model using pituitary MRI radiomics and clinical data to differentiate growth hormone deficiency (GHD) from idiopathic short stature (ISS), making the diagnostic process more acceptable to patients and their families. MATERIALS AND METHODS A retrospective cohort of 297 GHD and 300 ISS children (4-12 years) were enrolled as training and validation cohorts (8:2 ratio). An external cohort from another institution (49 GHD and 51 ISS) was employed as the testing cohort. Radiomics features extracted from the anterior pituitary gland on sagittal T1-weighted image (1.5 T or 3.0 T) were used to develop a radiomics model after feature selection. Hematological biomarkers were selected to create a clinical model and combine with the optimal radiomics features to create a clinical-radiomics model. The area under the receive operating characteristic curve (AUC) and Delong test compared the diagnostic performance of the previously mentioned three models across different validation and testing cohorts. RESULTS 17 radiomics features were selected for the radiomics model, and total protein, total cholesterol, free triiodothyronine, and triglyceride were utilized for the clinical model. In the training and validation cohorts, the diagnostic performance of the clinical-radiomics model (AUC=0.820 and 0.801) was comparable to the radiomics model (AUC=0.812 and 0.779, both P >0.05), both outperforming the clinical model (AUC=0.575 and 0.593, P <0.001). In the testing cohort, the clinical-radiomics model exhibited the highest AUC of 0.762 than the clinical and radiomics model (AUC=0.604 and 0.741, respectively, P <0.05). In addition, the clinical and radiomics models demonstrated similar diagnostic performance in the testing cohort (P >0.05). CONCLUSION Integrating radiomics features from conventional pituitary MRI with clinical indicators offers a minimally invasive approach for identifying GHD and shows robustness in a multicenter setting.
Collapse
Affiliation(s)
- Kun Shu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Keren Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Ruifang Zhang
- Department of Radiology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Chenyan Wang
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Zheng Cai
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Kun Liu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China
| | - Hu Lin
- Department of Endocrinology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Yan Zeng
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd, China
| | - Zirui Cao
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd, China
| | - Can Lai
- Department of Radiology, Children's hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, China
| | - Zhihan Yan
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang Province, China; Wenzhou Key Laboratory of Structural and Functional Imaging, Wenzhou, Zhejiang Province, China
| | - Yi Lu
- Department of Radiology, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Key Laboratory of Structural Malformations in Children of Zhejiang Province, Wenzhou, Zhejiang Province, China; Wenzhou Key Laboratory of Structural and Functional Imaging, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
38
|
Zhu T, Huang YH, Li W, Wu CG, Zhang YM, Zheng XX, Zhang TF, Lin YY, Liu ZY, Ye GL, Lin Y, Wu ZY, Wang K. A non-invasive artificial intelligence model for identifying axillary pathological complete response to neoadjuvant chemotherapy in breast cancer: a secondary analysis to multicenter clinical trial. Br J Cancer 2024; 131:692-701. [PMID: 38918556 PMCID: PMC11333754 DOI: 10.1038/s41416-024-02726-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 05/02/2024] [Accepted: 05/14/2024] [Indexed: 06/27/2024] Open
Abstract
BACKGROUND This study aims to develop a stacking model for accurately predicting axillary lymph node (ALN) response to neoadjuvant chemotherapy (NAC) using longitudinal MRI in breast cancer. METHODS We included patients with node-positive breast cancer who received NAC following surgery from January 2012 to June 2022. We collected MRIs before and after NAC, and extracted radiomics features from the tumour, peritumour, and ALN regions. The Mann-Whitney U test, least absolute shrinkage and selection operator, and Boruta algorithm were used to select features. We utilised machine learning techniques to develop three single-modality models and a stacking model for predicting ALN response to NAC. RESULTS This study consisted of a training cohort (n = 277), three external validation cohorts (n = 313, 164, and 318), and a prospective cohort (n = 81). Among the 1153 patients, 60.62% achieved ypN0. The stacking model achieved excellent AUCs of 0.926, 0.874, and 0.862 in the training, external validation, and prospective cohort, respectively. It also showed lower false-negative rates (FNRs) compared to radiologists, with rates of 14.40%, 20.85%, and 18.18% (radiologists: 40.80%, 50.49%, and 63.64%) in three cohorts. Additionally, there was a significant difference in disease-free survival between high-risk and low-risk groups (p < 0.05). CONCLUSIONS The stacking model can accurately predict ALN status after NAC in breast cancer, showing a lower false-negative rate than radiologists. TRIAL REGISTRATION NUMBER The clinical trial numbers were NCT03154749 and NCT04858529.
Collapse
Affiliation(s)
- Teng Zhu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yu-Hong Huang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Wei Li
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510515, China
| | - Can-Gui Wu
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Yi-Min Zhang
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China
| | - Xing-Xing Zheng
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Ting-Feng Zhang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
| | - Ying-Yi Lin
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China
- Shantou University Medical College, Shantou, 515041, Guangdong, China
| | - Zai-Yi Liu
- Department of Radiology, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080, Guangdong, China
| | - Guo-Lin Ye
- Department of Breast Cancer, The First People's Hospital of Foshan, Foshan, Guangdong, China
| | - Ying Lin
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zhi-Yong Wu
- Diagnosis and Treatment Center of Breast Diseases, Shantou Central Hospital, Shantou, China.
| | - Kun Wang
- Department of Breast Cancer, Cancer Center, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, Guangdong, China.
| |
Collapse
|
39
|
Qu L, Mei X, Yi Z, Zou Q, Zhou Q, Zhang D, Zhou M, Pei L, Long Q, Meng J, Zhang H, Chen Q, Yi W. An unsupervised learning model based on CT radiomics features accurately predicts axillary lymph node metastasis in breast cancer patients: diagnostic study. Int J Surg 2024; 110:5363-5373. [PMID: 38847776 PMCID: PMC11392119 DOI: 10.1097/js9.0000000000001778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 05/29/2024] [Indexed: 09/15/2024]
Abstract
BACKGROUND The accuracy of traditional clinical methods for assessing the metastatic status of axillary lymph nodes (ALNs) is unsatisfactory. In this study, the authors propose the use of radiomic technology and three-dimensional (3D) visualization technology to develop an unsupervised learning model for predicting axillary lymph node metastasis in patients with breast cancer (BC), aiming to provide a new method for clinical axillary lymph node assessment in patients with this disease. METHODS In this study, we retrospectively analyzed the data of 350 patients with invasive BC who underwent lung-enhanced computed tomography (CT) and axillary lymph node dissection surgery at the Department of Breast Surgery of the Second Xiangya Hospital of Central South University. The authors used 3D visualization technology to create a 3D atlas of ALNs and identified the region of interest for the lymph nodes. Radiomic features were subsequently extracted and selected, and a prediction model for ALNs was constructed using the K-means unsupervised algorithm. To validate the model, the authors prospectively collected data from 128 BC patients who were clinically evaluated as negative at our center. RESULTS Using 3D visualization technology, we extracted and selected a total of 36 CT radiomics features. The unsupervised learning model categorized 1737 unlabeled lymph nodes into two groups, and the analysis of the radiomic features between these groups indicated potential differences in lymph node status. Further validation with 1397 labeled lymph nodes demonstrated that the model had good predictive ability for axillary lymph node status, with an area under the curve of 0.847 (0.825-0.869). Additionally, the model's excellent predictive performance was confirmed in the 128 axillary clinical assessment negative cohort (cN0) and the 350 clinical assessment positive (cN+) cohort, for which the correct classification rates were 86.72 and 87.43%, respectively, which were significantly greater than those of clinical assessment methods. CONCLUSIONS The authors created an unsupervised learning model that accurately predicts the status of ALNs. This approach offers a novel solution for the precise assessment of ALNs in patients with BC.
Collapse
Affiliation(s)
- Limeng Qu
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Xilong Mei
- Department of Radiology, The Second Xiangya Hospital of Central South University
| | - Zixi Yi
- Central South University, Changsha, Hunan
| | - Qiongyan Zou
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Qin Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Danhua Zhang
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Meirong Zhou
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Lei Pei
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Qian Long
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Jiahao Meng
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Huashan Zhang
- Urinary Surgery, Changsha Central Hospital, Changsha, Hunan, China
| | - Qitong Chen
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| | - Wenjun Yi
- Department of General Surgery, The Second Xiangya Hospital, Central South University
- Clinical Research Center For Breast Disease In Hunan Province, Changsha
| |
Collapse
|
40
|
Yang Z, Liu C. Research on the application of radiomics in breast cancer: A bibliometrics and visualization analysis. Medicine (Baltimore) 2024; 103:e39463. [PMID: 39213225 PMCID: PMC11365679 DOI: 10.1097/md.0000000000039463] [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: 03/26/2024] [Revised: 04/24/2024] [Accepted: 08/06/2024] [Indexed: 09/04/2024] Open
Abstract
Breast cancer is the most prevalent form of cancer worldwide. Therefore, improved disease detection has emerged as a focal point in clinical studies. At the forefront of innovation, radiomics has the capability to extract comprehensive insights from medical images, ultimately enhancing the accuracy of diagnostic procedures. There has been rapid growth in the field of radiomics research on breast cancer in the past few years. We explored pertinent research articles in the Web of Science Core Collection database to gain a thorough understanding of breast cancer radiomics. We used CiteSpace to conduct a bibliometric analysis of the annual distribution of different nations, institutions, journals, authors, keywords, and references in the field of breast cancer radiomics. GraphPad Prism software was used to examine and graph yearly and country-specific trends and the proportions of publications. The tools utilized for the visualization of science mapping included CiteSpace and VOSviewer. Of the 891 publications, most were original articles (731, 91.09%) and a few were reviews (160, 8.91%). Most academic research has been published in China and the United States. The study centers predominantly consisted of major academic institutions, such as Fudan University and the Chinese Academy of Sciences, with some of their members being prominent figures in the field. Pinker, Katja has published the largest number of research papers. The majority of these studies have been published in medical journals focusing on radiology and oncology in recent years. In the realm of cutting-edge medical research, the top two keywords, magnetic resonance imaging and machine learning stand at the forefront as current areas of intense focus. Breast cancer radiomics is advancing rapidly, presenting numerous opportunities and obstacles. Our study of the literature in this academic area aimed to pinpoint the primary themes addressed in the studies and anticipate prospective avenues for research.
Collapse
Affiliation(s)
- Zhe Yang
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| | - Chenglong Liu
- Department of Radiology, the Second Affiliated Hospital of Shandong First Medical University, Tai’an, Shandong, China
| |
Collapse
|
41
|
Zhao H, Wang Y, Sun Y, Wang Y, Shi B, Liu J, Zhang S. Hematological indicator-based machine learning models for preoperative prediction of lymph node metastasis in cervical cancer. Front Oncol 2024; 14:1400109. [PMID: 39193382 PMCID: PMC11347340 DOI: 10.3389/fonc.2024.1400109] [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: 03/13/2024] [Accepted: 07/29/2024] [Indexed: 08/29/2024] Open
Abstract
Background Lymph node metastasis (LNM) is an important prognostic factor for cervical cancer (CC) and determines the treatment strategy. Hematological indicators have been reported as being useful biomarkers for the prognosis of a variety of cancers. This study aimed to evaluate the feasibility of machine learning models characterized by preoperative hematological indicators to predict the LNM status of CC patients before surgery. Methods The clinical data of 236 patients with pathologically confirmed CC were retrospectively analyzed at the Gynecology Oncology Department of the First Affiliated Hospital of Bengbu Medical University from November 2020 to August 2022. The least absolute shrinkage and selection operator (LASSO) was used to select 21 features from 35 hematological indicators and for the construction of 6 machine learning predictive models, including Adaptive Boosting (AdaBoost), Gaussian Naive Bayes (GNB), and Logistic Regression (LR), as well as Random Forest (RF), Support Vector Machines (SVM), and Extreme Gradient Boosting (XGBoost). Evaluation metrics of predictive models included the area under the receiver operating characteristic curve (AUC), accuracy, specificity, sensitivity, and F1-score. Results RF has the best overall predictive performance for ten-fold cross-validation in the training set. The specific performance indicators of RF were AUC (0.910, 95% confidence interval [CI]: 0.820-1.000), accuracy (0.831, 95% CI: 0.702-0.960), specificity (0.835, 95% CI: 0.708-0.962), sensitivity (0.831, 95% CI: 0.702-0.960), and F1-score (0.829, 95% CI: 0.696-0.962). RF had the highest AUC in the testing set (AUC = 0.854). Conclusion RF based on preoperative hematological indicators that are easily available in clinical practice showed superior performance in the preoperative prediction of CC LNM. However, investigations on larger external cohorts of patients are required for further validation of our findings.
Collapse
Affiliation(s)
- Huan Zhao
- School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China
| | - Yuling Wang
- Department of Gynecology and Oncology, First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Yilin Sun
- Department of Gynecology and Oncology, First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Yongqiang Wang
- School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China
| | - Bo Shi
- School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China
| | - Jian Liu
- Department of Gynecology and Oncology, First Affiliated Hospital, Bengbu Medical University, Bengbu, Anhui, China
| | - Sai Zhang
- School of Medical Imaging, Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
42
|
Wang Y, Zhao H, Fu P, Tian L, Su Y, Lyu Z, Gu W, Wang Y, Liu S, Wang X, Zheng H, Du J, Zhang R. Preoperative prediction of lymph node metastasis in colorectal cancer using 18F-FDG PET/CT peritumoral radiomics analysis. Med Phys 2024; 51:5214-5225. [PMID: 38801340 DOI: 10.1002/mp.17193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/29/2024] Open
Abstract
BACKGROUND Radiomics has been used in the diagnosis of tumor lymph node metastasis (LNM). However, to date, most studies have been based on intratumoral radiomics. Few studies have focused on the use of 18F-fluorodeoxyglucose positron emission computed tomography (18F-FDG PET/CT) peritumoral radiomics for the diagnosis of LNM in colorectal cancer (CRC). PURPOSE Determining the value of radiomics features extracted from 18F-FDG PET/CT images of the peritumoral region in predicting LNM in patients with CRC. METHODS The clinical data and preoperative 18F-FDG PET/CT images of 244 CRC patients were retrospectively analyzed. Intratumoral and peritumoral radiomics features were screened using the mutual information method, and least absolute shrinkage and selection operator regression. Based on the selected radiomics features, a radiomics score (Rad-score) was calculated, and independent risk factors obtained from univariate and multivariate logistic regression analyses were used to construct clinical and combined (Radiomics + Clinical) models. The performance of these models was evaluated using the DeLong test, while their clinical utility was assessed by decision curve analysis. Finally, a nomogram was constructed to visualize the predictive model. RESULTS The most optimal set of features retained by the feature filtering process were all peritumoral radiomic features. Carcinoembryonic antigen levels, PET/CT-reported lymph node status and Rad-score were found to be independent risk factors for LNM. All three LNM risk assessment models exhibited good predictive performance, with the combined model showing the best classification results, with areas under the curve of 0.85 and 0.76 in the training and validation groups, respectively. The DeLong test revealed that the performance of the combined model was superior to that of the clinical and radiomics models in both the training and validation groups, although this difference was only statistically significant in the training group. DCA indicated that the combined model displayed better clinical utility. CONCLUSIONS 18F-FDG PET/CT peritumoral radiomics is uniquely suited to predict the presence of LNM in patients with CRC. In particular, the predictive efficacy of LNM for precision therapy and individualized patient management can be improved by using a combination of clinical risk factors.
Collapse
Affiliation(s)
- Yan Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Hongyue Zhao
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Peng Fu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Lin Tian
- Department of Pathology, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Yexin Su
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Zhehao Lyu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Wenchao Gu
- Department of Diagnostic and Interventional Radiology, University of Tsukuba, Tsukuba, Ibaraki, Japan
| | - Yang Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Shan Liu
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Xi Wang
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Han Zheng
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Jingjing Du
- Department of Nuclear Medicine, The First Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang, China
| | - Rui Zhang
- Department of Magnetic Resonance, The First Hospital of Qiqihar, Qiqihar, Heilongjiang, China
| |
Collapse
|
43
|
Lin G, Chen W, Fan Y, Zhou Y, Li X, Hu X, Cheng X, Chen M, Kong C, Chen M, Xu M, Peng Z, Ji J. Machine Learning Radiomics-Based Prediction of Non-sentinel Lymph Node Metastasis in Chinese Breast Cancer Patients with 1-2 Positive Sentinel Lymph Nodes: A Multicenter Study. Acad Radiol 2024; 31:3081-3095. [PMID: 38490840 DOI: 10.1016/j.acra.2024.02.010] [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: 12/07/2023] [Revised: 02/04/2024] [Accepted: 02/07/2024] [Indexed: 03/17/2024]
Abstract
RATIONALE AND OBJECTIVES This study aimed to construct a machine learning radiomics-based model using dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) images to evaluate non-sentinel lymph node (NSLN) metastasis in Chinese breast cancer (BC) patients who underwent total mastectomy (TM) and had 1-2 positive sentinel lymph nodes (SLNs). MATERIALS AND METHODS In total, 494 patients were retrospectively enrolled from two hospitals, and were divided into the training (n = 286), internal validation (n = 122), and external validation (n = 86) cohorts. Features were extracted from DCE-MRI images for each patient and screened. Six ML classifies were trained and the best classifier was evaluated to calculate radiomics (Rad)-scores. A combined model was developed based on Rad-scores and clinical risk factors, then the calibration, discrimination, reclassification, and clinical usefulness were evaluated. RESULTS 14 radiomics features were ultimately selected. The random forest (RF) classifier showed the best performance, with the highest average area under the curve (AUC) of 0.833 in the validation cohorts. The combined model incorporating RF-based Rad-scores, tumor size, lymphovascular invasion, and proportion of positive SLNs resulted in the best discrimination ability, with AUCs of 0.903, 0.890, and 0.836 in the training, internal validation, and external validation cohorts, respectively. Furthermore, the combined model significantly improved the classification accuracy and clinical benefit for NSLN metastasis prediction. CONCLUSION A RF-based combined model using DCE-MRI images exhibited a promising performance for predicting NSLN metastasis in Chinese BC patients who underwent TM and had 1-2 positive SLNs, thereby aiding in individualized clinical treatment decisions.
Collapse
Affiliation(s)
- Guihan Lin
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Weiyue Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Yingying Fan
- Department of Stomatology, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Yi Zhou
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Xia Li
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Xin Hu
- School of Medicine, Shaoxing University, Shaoxing 312000, China
| | - Xue Cheng
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Mingzhen Chen
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Chunli Kong
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui 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, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China
| | - Min Xu
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China; School of Medicine, Shaoxing University, Shaoxing 312000, China
| | - Zhiyi Peng
- Department of Radiology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China
| | - Jiansong Ji
- Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, Key Laboratory of Precision Medicine of Lishui City, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China; Clinical College of The Affiliated Central Hospital, School of Medicine, Lishui University, Lishui 323000, China; School of Medicine, Shaoxing University, Shaoxing 312000, China.
| |
Collapse
|
44
|
Lin Y, Wang J, Li M, Zhou C, Hu Y, Wang M, Zhang X. Prediction of breast cancer and axillary positive-node response to neoadjuvant chemotherapy based on multi-parametric magnetic resonance imaging radiomics models. Breast 2024; 76:103737. [PMID: 38696854 PMCID: PMC11070644 DOI: 10.1016/j.breast.2024.103737] [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: 10/25/2023] [Revised: 04/05/2024] [Accepted: 04/22/2024] [Indexed: 05/04/2024] Open
Abstract
PURPOSE Accurate identification of primary breast cancer and axillary positive-node response to neoadjuvant chemotherapy (NAC) is important for determining appropriate surgery strategies. We aimed to develop combining models based on breast multi-parametric magnetic resonance imaging and clinicopathologic characteristics for predicting therapeutic response of primary tumor and axillary positive-node prior to treatment. MATERIALS AND METHODS A total of 268 breast cancer patients who completed NAC and underwent surgery were enrolled. Radiomics features and clinicopathologic characteristics were analyzed through the analysis of variance and the least absolute shrinkage and selection operator algorithm. Finally, 24 and 28 optimal features were selected to construct machine learning models based on 6 algorithms for predicting each clinical outcome, respectively. The diagnostic performances of models were evaluated in the testing set by the area under the curve (AUC), sensitivity, specificity, and accuracy. RESULTS Of the 268 patients, 94 (35.1 %) achieved breast cancer pathological complete response (bpCR) and of the 240 patients with clinical positive-node, 120 (50.0 %) achieved axillary lymph node pathological complete response (apCR). The multi-layer perception (MLP) algorithm yielded the best diagnostic performances in predicting apCR with an AUC of 0.825 (95 % CI, 0.764-0.886) and an accuracy of 77.1 %. And MLP also outperformed other models in predicting bpCR with an AUC of 0.852 (95 % CI, 0.798-0.906) and an accuracy of 81.3 %. CONCLUSIONS Our study established non-invasive combining models to predict the therapeutic response of primary breast cancer and axillary positive-node prior to NAC, which may help to modify preoperative treatment and determine post-NAC surgery strategy.
Collapse
Affiliation(s)
- Yingyu Lin
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Jifei Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Meizhi Li
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Chunxiang Zhou
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Yangling Hu
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Mengyi Wang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China
| | - Xiaoling Zhang
- Department of Radiology, The First Affiliated Hospital, Sun Yat-Sen University. 58th, The Second Zhongshan Road, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
45
|
Wang J, Song P, Zhang M, Liu W, Zeng X, Chen N, Li Y, Wang M. A prediction model based on deep learning and radiomics features of DWI for the assessment of microsatellite instability in endometrial cancer. Cancer Med 2024; 13:e70046. [PMID: 39171859 PMCID: PMC11339853 DOI: 10.1002/cam4.70046] [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: 03/24/2024] [Revised: 07/06/2024] [Accepted: 07/12/2024] [Indexed: 08/23/2024] Open
Abstract
BACKGROUND To explore the efficacy of a prediction model based on diffusion-weighted imaging (DWI) features extracted from deep learning (DL) and radiomics combined with clinical parameters and apparent diffusion coefficient (ADC) values to identify microsatellite instability (MSI) in endometrial cancer (EC). METHODS This study included a cohort of 116 patients with EC, who were subsequently divided into training (n = 81) and test (n = 35) sets. From DWI, conventional radiomics features and convolutional neural network-based DL features were extracted. Random forest (RF) and logistic regression were adopted as classifiers. DL features, radiomics features, clinical variables, ADC values, and their combinations were applied to establish DL, radiomics, clinical, ADC, and combined models, respectively. The predictive performance was evaluated through the area under the receiver operating characteristic curve (AUC), total integrated discrimination index (IDI), net reclassification index (NRI), calibration curves, and decision curve analysis (DCA). RESULTS The optimal predictive model, based on an RF classifier, comprised four DL features, three radiomics features, two clinical variables, and an ADC value. In the training and test sets, this model exhibited AUC values of 0.989 (95% CI: 0.935-1.000) and 0.885 (95% CI: 0.731-0.967), respectively, demonstrating different degrees of improvement compared with the clinical, DL, radiomics, and ADC models (AUC-training = 0.671, 0.873, 0.833, and 0.814, AUC-test = 0.685, 0.783, 0.708, and 0.713, respectively). The NRI and IDI analyses revealed that the combined model resulted in improved risk reclassification of the MSI status compared to the clinical, radiomics, DL, and ADC models. The calibration curves and DCA indicated good consistency and clinical utility of this model, respectively. CONCLUSIONS The predictive model based on DWI features extracted from DL and radiomics combined with clinical parameters and ADC values could effectively assess the MSI status in EC.
Collapse
Affiliation(s)
- Jing Wang
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Pujiao Song
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Meng Zhang
- Department of Magnetic Resonance ImagingThe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Wei Liu
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Xi Zeng
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Nanshan Chen
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| | - Yuxia Li
- Department of Magnetic Resonance ImagingThe First Affiliated Hospital of Xinxiang Medical UniversityXinxiangChina
| | - Minghua Wang
- Department of Nuclear MedicineThe Affiliated Hospital of Guizhou Medical UniversityGuiyangChina
| |
Collapse
|
46
|
Liu Y, Ren H, Pei Y, Shen L, Guo J, Zhou J, Li C, Liu Y. Development of a CT-Based comprehensive model combining clinical, radiomics with deep learning for differentiating pulmonary metastases from noncalcified pulmonary hamartomas: a retrospective cohort study. Int J Surg 2024; 110:4900-4910. [PMID: 38759692 PMCID: PMC11326030 DOI: 10.1097/js9.0000000000001593] [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: 01/26/2024] [Accepted: 04/26/2024] [Indexed: 05/19/2024]
Abstract
BACKGROUND Clinical differentiation between pulmonary metastases and noncalcified pulmonary hamartomas (NCPH) often presents challenges, leading to potential misdiagnosis. However, the efficacy of a comprehensive model that integrates clinical features, radiomics, and deep learning (CRDL) for differential diagnosis of these two diseases remains uncertain. OBJECTIVE This study evaluated the diagnostic efficacy of a CRDL model in differentiating pulmonary metastases from NCPH. METHODS The authors retrospectively analyzed the clinical and imaging data of 256 patients from the First Medical Centre of the General Hospital of the People's Liberation Army (PLA) and 85 patients from Shanghai Changhai Hospital, who were pathologically confirmed pulmonary hamartomas or pulmonary metastases after thoracic surgery. Employing Python 3.7 software suites, the authors extracted radiomic features and deep learning (DL) attributes from patient datasets. The cohort was divided into training set, internal validation set, and external validation set. The diagnostic performance of the constructed models was evaluated using receiver operating characteristic (ROC) curve analysis to determine their effectiveness in differentiating between pulmonary metastases and NCPH. RESULTS Clinical features such as white blood cell count (WBC), platelet count (PLT), history of cancer, carcinoembryonic antigen (CEA) level, tumor marker status, lesion margin characteristics (smooth or blurred), and maximum diameter were found to have diagnostic value in differentiating between the two diseases. In the domains of radiomics and DL. Of the 1130 radiomics features and 512 DL features, 24 and 7, respectively, were selected for model development. The area under the ROC curve (AUC) values for the four groups were 0.980, 0.979, 0.999, and 0.985 in the training set, 0.947, 0.816, 0.934, and 0.952 in the internal validation set, and 0.890, 0.904, 0.923, and 0.938 in the external validation set. This demonstrated that the CRDL model showed the greatest efficacy. CONCLUSIONS The comprehensive model incorporating clinical features, radiomics, and DL shows promise for aiding in the differentiation between pulmonary metastases and hamartomas.
Collapse
Affiliation(s)
- Yunze Liu
- Medical School of Chinese General Hospital of PLA
| | - Hong Ren
- Medical School of Chinese General Hospital of PLA
| | - Yanbin Pei
- Medical School of Chinese General Hospital of PLA
| | - Leilei Shen
- Department of Thoracic Surgery, Hainan Hospital of Chinese General Hospital of PLA, Sanya
| | - Juntang Guo
- Department of Thoracic Surgery, First Medical Center, Chinese General Hospital of PLA, Beijing
| | - Jian Zhou
- Department of Imaging, Changhai Hospital, Shanghai, People's Republic of China
| | - Chengrun Li
- Department of Thoracic Surgery, First Medical Center, Chinese General Hospital of PLA, Beijing
| | - Yang Liu
- Department of Thoracic Surgery, First Medical Center, Chinese General Hospital of PLA, Beijing
| |
Collapse
|
47
|
Hu W, Wen F, Zhao M, Li X, Luo P, Jiang G, Yang H, Herth FJF, Zhang X, Zhang Q. Endobronchial Ultrasound-Based Support Vector Machine Model for Differentiating between Benign and Malignant Mediastinal and Hilar Lymph Nodes. Respiration 2024; 103:675-685. [PMID: 39038439 DOI: 10.1159/000540467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Accepted: 07/10/2024] [Indexed: 07/24/2024] Open
Abstract
INTRODUCTION The aim of the study was to establish an ultrasonographic radiomics machine learning model based on endobronchial ultrasound (EBUS) to assist in diagnosing benign and malignant mediastinal and hilar lymph nodes (LNs). METHODS The clinical and ultrasonographic image data of 197 patients were retrospectively analyzed. The radiomics features extracted by EBUS-based radiomics were analyzed by the least absolute shrinkage and selection operator. Then, we used a support vector machine (SVM) algorithm to establish an EBUS-based radiomics model. A total of 205 lesions were randomly divided into training (n = 143) and validation (n = 62) groups. The diagnostic efficiency was evaluated by receiver operating characteristic (ROC) curve analysis. RESULTS A total of 13 stable radiomics features with non-zero coefficients were selected. The SVM model exhibited promising performance in both groups. In the training group, the SVM model achieved an ROC area under the curve (AUC) of 0.892 (95% CI: 0.885-0.899), with an accuracy of 85.3%, sensitivity of 93.2%, and specificity of 79.8%. In the validation group, the SVM model had an ROC AUC of 0.906 (95% CI: 0.890-0.923), an accuracy of 74.2%, a sensitivity of 70.3%, and a specificity of 74.1%. CONCLUSION The EBUS-based radiomics model can be used to differentiate mediastinal and hilar benign and malignant LNs. The SVM model demonstrated excellent potential as a diagnostic tool in clinical practice.
Collapse
Affiliation(s)
- Wenjia Hu
- Department of Ultrasound, Zhengzhou University People's Hospital, Zhengzhou, China
| | - Feifei Wen
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China,
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China,
| | - Mengyu Zhao
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Xiangnan Li
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Peiyuan Luo
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Guancheng Jiang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Huizhen Yang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
| | - Felix J F Herth
- Department of Pneumology and Respiratory Care Medicine, Thoraxklinik and Translational Lung Research Center, University of Heidelberg, Heidelberg, Germany
| | - Xiaoju Zhang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
- Henan International Joint Laboratory of Diagnosis and Treatment for Pulmonary Nodules, Zhengzhou, China
| | - Quncheng Zhang
- Department of Respiratory and Critical Care Medicine, Zhengzhou University People's Hospital, Zhengzhou, China
- Department of Respiratory and Critical Care Medicine, Henan Provincial People's Hospital, Zhengzhou, China
- Henan International Joint Laboratory of Diagnosis and Treatment for Pulmonary Nodules, Zhengzhou, China
| |
Collapse
|
48
|
Sohrabei S, Moghaddasi H, Hosseini A, Ehsanzadeh SJ. Investigating the effects of artificial intelligence on the personalization of breast cancer management: a systematic study. BMC Cancer 2024; 24:852. [PMID: 39026174 PMCID: PMC11256548 DOI: 10.1186/s12885-024-12575-1] [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: 10/26/2023] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
BACKGROUND Providing appropriate specialized treatment to the right patient at the right time is considered necessary in cancer management. Targeted therapy tailored to the genetic changes of each breast cancer patient is a desirable feature of precision oncology, which can not only reduce disease progression but also potentially increase patient survival. The use of artificial intelligence alongside precision oncology can help physicians by identifying and selecting more effective treatment factors for patients. METHOD A systematic review was conducted using the PubMed, Embase, Scopus, and Web of Science databases in September 2023. We performed the search strategy with keywords, namely: Breast Cancer, Artificial intelligence, and precision Oncology along with their synonyms in the article titles. Descriptive, qualitative, review, and non-English studies were excluded. The quality assessment of the articles and evaluation of bias were determined based on the SJR journal and JBI indices, as well as the PRISMA2020 guideline. RESULTS Forty-six studies were selected that focused on personalized breast cancer management using artificial intelligence models. Seventeen studies using various deep learning methods achieved a satisfactory outcome in predicting treatment response and prognosis, contributing to personalized breast cancer management. Two studies utilizing neural networks and clustering provided acceptable indicators for predicting patient survival and categorizing breast tumors. One study employed transfer learning to predict treatment response. Twenty-six studies utilizing machine-learning methods demonstrated that these techniques can improve breast cancer classification, screening, diagnosis, and prognosis. The most frequent modeling techniques used were NB, SVM, RF, XGBoost, and Reinforcement Learning. The average area under the curve (AUC) for the models was 0.91. Moreover, the average values for accuracy, sensitivity, specificity, and precision were reported to be in the range of 90-96% for the models. CONCLUSION Artificial intelligence has proven to be effective in assisting physicians and researchers in managing breast cancer treatment by uncovering hidden patterns in complex omics and genetic data. Intelligent processing of omics data through protein and gene pattern classification and the utilization of deep neural patterns has the potential to significantly transform the field of complex disease management.
Collapse
Affiliation(s)
- Solmaz Sohrabei
- Department of Health Information Technology and Management, Medical Informatics, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Hamid Moghaddasi
- Department of Health Information Technology and Management, Medical Informatics, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Azamossadat Hosseini
- Department of Health Information Technology and Management, Health Information Management, School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Seyed Jafar Ehsanzadeh
- Department of English Language, School of Health Management and Information Sciences, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Li W, Li Y, Gao S, Huang N, Kojima I, Kusama T, Ou Y, Iikubo M, Niu X. Integrating lipid metabolite analysis with MRI-based transformer and radiomics for early and late stage prediction of oral squamous cell carcinoma. BMC Cancer 2024; 24:795. [PMID: 38961418 PMCID: PMC11221018 DOI: 10.1186/s12885-024-12533-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Accepted: 06/18/2024] [Indexed: 07/05/2024] Open
Abstract
BACKGROUND Oral Squamous Cell Carcinoma (OSCC) presents significant diagnostic challenges in its early and late stages. This study aims to utilize preoperative MRI and biochemical indicators of OSCC patients to predict the stage of tumors. METHODS This study involved 198 patients from two medical centers. A detailed analysis of contrast-enhanced T1-weighted (ceT1W) and T2-weighted (T2W) MRI were conducted, integrating these with biochemical indicators for a comprehensive evaluation. Initially, 42 clinical biochemical indicators were selected for consideration. Through univariate analysis and multivariate analysis, only those indicators with p-values less than 0.05 were retained for model development. To extract imaging features, machine learning algorithms in conjunction with Vision Transformer (ViT) techniques were utilized. These features were integrated with biochemical indicators for predictive modeling. The performance of model was evaluated using the Receiver Operating Characteristic (ROC) curve. RESULTS After rigorously screening biochemical indicators, four key markers were selected for the model: cholesterol, triglyceride, very low-density lipoprotein cholesterol and chloride. The model, developed using radiomics and deep learning for feature extraction from ceT1W and T2W images, showed a lower Area Under the Curve (AUC) of 0.85 in the validation cohort when using these imaging modalities alone. However, integrating these biochemical indicators improved the model's performance, increasing the validation cohort AUC to 0.87. CONCLUSION In this study, the performance of the model significantly improved following multimodal fusion, outperforming the single-modality approach. CLINICAL RELEVANCE STATEMENT This integration of radiomics, ViT models, and lipid metabolite analysis, presents a promising non-invasive technique for predicting the staging of OSCC.
Collapse
Affiliation(s)
- Wen Li
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yang Li
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Shiyu Gao
- School of Mathematics and Statistics, Huazhong University of Science and Technology, Wuhan, China
| | - Nengwen Huang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Ikuho Kojima
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Taro Kusama
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yanjing Ou
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Masahiro Iikubo
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan.
| | - Xuegang Niu
- Department of Neurosurgey, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, China.
- Department of Neurosurgey, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China.
| |
Collapse
|
50
|
Xin W, Rixin S, Linrui L, Zhihui Q, Long L, Yu Z. Machine learning-based radiomics for predicting outcomes in cervical cancer patients undergoing concurrent chemoradiotherapy. Comput Biol Med 2024; 177:108593. [PMID: 38801795 DOI: 10.1016/j.compbiomed.2024.108593] [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: 11/22/2023] [Revised: 05/09/2024] [Accepted: 05/10/2024] [Indexed: 05/29/2024]
Abstract
PURPOSES To investigate the value of machine learning-based radiomics for predicting disease-free survival (DFS) and overall survival (OS) undergoing concurrent chemoradiotherapy (CCRT) for patients with locally advanced cervical cancer (LACC). MATERIALS AND METHODS In this multicentre study, 700 patients with IB2-IVA cervical cancer who underwent CCRT with ongoing follow-up were retrospectively analyzed. Three-dimensional radiomics features of primary lesions and its surrounding 5 mm region in T2WI sequences were collected. Six machine learning methods were used to construct the optimal radiomics model for accurate prediction of DFS and OS after CCRT in LACC patients. Eventually, TCGA and GEO databases were used to explore the mechanisms of radiomics in predicting the progression and survival of cervical cancer. This study adhered CLEAR for reporting and its quality was assessed using RQS and METRICS. RESULTS In the prediction of DFS, the RSF model combined tumor and peritumor radiomics demonstrated the best predictive efficacy, with the AUC for predicting 1-year, 3-year, and 5-year DFS in the training, validation, and test sets of 0.986, 0.989, 0.990, and 0.884, 0.838, 0.823, and 0.829, 0.809, 0.841, respectively. In the prediction of OS, the GBM model best performer, with AUC of 0.999, 0.995, 0.978, and 0.981, 0.975, 0.837, and 0.904, 0.860, 0.905. Differential genes in TCGA and GEO suggest that the prediction of radiomics model may be associated with KDELR2 and HK2. CONCLUSION Machine learning-based radiomics models help to predict DFS and OS after CCRT in LACC patients, and the combination of tumor and peritumor information has higher predictive efficacy, which can provide a reliable basis for therapeutic decision-making in cervical cancer patients.
Collapse
Affiliation(s)
- Wang Xin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Su Rixin
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Li Linrui
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Qin Zhihui
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China
| | - Liu Long
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Zhejiang University, Hangzhou, 310000, Zhejiang, China.
| | - Zhang Yu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
| |
Collapse
|