1
|
Luo J, Lin Y, Zeng Z, Deng H, Li T. Research of Ultrasonic Shear Wave Elastography in Evaluating the Efficiency of Neoadjuvant Chemotherapy for Breast Cancer. JOURNAL OF CLINICAL ULTRASOUND : JCU 2025; 53:1026-1034. [PMID: 40125599 DOI: 10.1002/jcu.23951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/27/2024] [Accepted: 01/04/2025] [Indexed: 03/25/2025]
Abstract
OBJECTIVE This study aimed to identify reliable quantitative parameters of ultrasonic shear wave elastography (SWE) that demonstrate advantages in assessing the efficacy of neoadjuvant chemotherapy (NAC) for breast cancer by comparison to conventional ultrasound (US). This research also analyzed the associations between SWE parameters and tumor collagen fibers from post-NAC breast cancer lesions to explore the histological micro-mechanisms of stiffness change for breast cancer after NAC. METHODS Forty-Seven breast cancer lesions examined with US and SWE were eligible for enrollment from January 2021 to July 2023. The ultrasonic maximum diameter (Dmax), mean elastic value (Emean), maximum elastic value (Emax), and minimum elastic value (Emin) before and after NAC were determined. Receiver operating characteristic (ROC) curves were drawn to compare the diagnostic efficacy regarding the change rates of the above parameters. Additionally, correlation analyses were performed to examine the relationship between effective SWE quantitative parameters and collagen fibers after NAC. RESULTS Among the 47 breast cancer lesions, 24 lesions showed significant pathological responses, while the other 23 lesions exhibited that the pathological responses were nonsignificant. The area under the curve (AUC) of ΔEmin was the lowest (ΔEmin:0.466, ΔDmax: 0.679, ΔEmax: 0.803, and ΔEmean: 0.813). The collagen fiber category showed a significant positive correlation with Emean (r = 0.711) and Emax (r = 0.669) after NAC. However, the collagen fiber content demonstrated no significant correlation with Emean or Emax (p > 0.05). CONCLUSION Dmax, Emean, and Emax are valuable parameters for evaluating NAC efficacy, while Emin is not reliable. Emean and Emax demonstrate similar efficacy, both superior to Dmax. The stiffness of post-NAC breast cancer lesions is predominantly related to the category of collagen fibers rather than their content.
Collapse
Affiliation(s)
- Jingjing Luo
- Department of Ultrasound, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yunen Lin
- Department of Pathology, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ziyou Zeng
- Department of Ultrasound, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Haiyun Deng
- Department of Ultrasound, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| | - Tao Li
- Department of Ultrasound, Guangzhou Institute of Cancer Research, the Affiliated Cancer Hospital, Guangzhou Medical University, Guangzhou, China
| |
Collapse
|
2
|
Jiang J, Wang S, Xiao F, Gu H, Wang M, Tian H, Guan B, Sheng K, Xiong Y, Zhao H, Li M, Xu L, Sun Z, Du H, Du W, Li Y. Dual-Energy CT-Based Assessment of Thrombotic Heterogeneity for Predicting Stroke Source and Response to Machine Thrombectomy: A Step Toward Visualization Thrombus Treatment. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e17295. [PMID: 40432561 DOI: 10.1002/advs.202417295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 04/27/2025] [Indexed: 05/29/2025]
Abstract
The viability of using thrombus heterogeneity (TH) data derived from dual-energy CT (DECT) as a visual thrombotic biomarker is unclear. The first aim of this study is to develop a quantitative measure of TH on DECT and test its performance for predicting the stroke source (cardiogenic vs. non-cardiogenic) and clinical outcomes (functional status assessed by the modified Rankin Scale score at 90 days) following machine thrombectomy (MT). The second aim is to associate thrombus subregions with the thrombus composition to facilitate visualization of thrombus constituents. Radiomics data are extracted from the whole thrombus and subregions in CT/DECT to construct predictive models. The performances of all models are evaluated and compared in the validation and comparative cohorts. Histopathologic analysis is performed to correlate the subregion data with the actual thrombus composition. This study included 221 and 255 participants who underwent DECT and CT examinations, respectively. DECT outperformed CT in predicting stroke source and clinical outcomes, with the TH-related models showing the highest performance in the validation and comparative cohorts. Thrombus composition is correlated with the different CT/DECT-based subregions, with DECT-habitat_c showing the strongest association. Thrombus subregion analyses may help visualize the related constituents.
Collapse
Affiliation(s)
- Jingxuan Jiang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
- Department of Radiology, Affiliated Hospital of Nantong University, Nanton, 226001, China
| | - Sijia Wang
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Fan Xiao
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Hongmei Gu
- Department of Radiology, Affiliated Hospital of Nantong University, Nanton, 226001, China
| | - Mingkang Wang
- Wuhan United Imaging Life Science Instruments Ltd., Wuhan, 430206, China
| | - Hao Tian
- Department of Radiology, Affiliated Hospital of Nantong University, Nanton, 226001, China
| | - Baohui Guan
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Kai Sheng
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yijia Xiong
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Huilin Zhao
- Department of Radiology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, 200001, China
| | - Minda Li
- Department of Radiology, Affiliated Hospital of Nantong University, Nanton, 226001, China
| | - Li Xu
- Jiangsu Key Laboratory of Integrated Traditional Chinese and Western Medicine for Prevention and Treatment of Senile Diseases, Institute of Translational Medicine, Medical College, Yangzhou University, Yangzhou, 225001, China
| | - Zheng Sun
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Haiyan Du
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Wenxian Du
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yuehua Li
- Institute of Diagnostic and Interventional Radiology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
3
|
Wu Q, Qiang W, Pan L, Cha T, Li Q, Gao Y, Qiu K, Xing W. Performance of MRI-based radiomics for prediction of residual disease status in patients with nasopharyngeal carcinoma after radical radiotherapy. Sci Rep 2025; 15:16758. [PMID: 40368928 PMCID: PMC12078595 DOI: 10.1038/s41598-025-00186-0] [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: 10/09/2024] [Accepted: 04/25/2025] [Indexed: 05/16/2025] Open
Abstract
The purpose of this study was to determine if habitat radiomic features extracted from pretherapy multi-sequence MRI predict residual status in patients with Nasopharyngeal Carcinoma (NPC) after radical radiotherapy. The retrospective study enrolled 179 primary NPC patients, divided into training and validation cohorts at a 7:3 ratio. K-means clustering was employed to segment T2WI, CE-T1WI and FSCE-T1WI images, creating habitats within the volume of interest. Identify relevant features that can recognize NPC residuals. In the training cohort, support vector machine (SVM) models were developed utilizing the radiomic features extracted from each habitat and the entire tumor, selecting the most predictive features for each sequence. SVM models were constructed by combining the optimal radiomic features from each sequences with clinical data. Model performance was compared and validated using receiver operating characteristic (ROC) curves, calibration curves and decision curve analysis (DCA), and differences between models were assessed using the DeLong test. The optimal clustering results revealed 4 habitats in FSCE-T1WI, while 2 habitats in both CE-T1WI and T2WI sequences. In the training cohort, we compared the predictive accuracy of SVM models based on different habitats and total tumor characteristics from three sequences, and found that the features from T2 Hab2, CE-T1 Hab1, and FSCE-T1 Hab4 images showed higher performance. Incorporation of habitat-based radiomic features and clinical variables significantly enhanced the predictive performance. The integrated model exhibits the optimal predictive performance, with the area under the curve (AUC) values of 0.921 (SEN = 0.821, SPE = 0.830) in the training cohort and 0.811 (SEN = 0.778, SPE = 0.722) in the validation cohort. Compared to conventional radiomics, habitat imaging features that distinguish intratumoral heterogeneity have higher predictive value, making them potential non-invasive biomarkers for assessing NPC residual after radiotherapy. Integration of multi-sequence MRI habitat radiomic with clinical parameters further improved predictive accuracy.
Collapse
Affiliation(s)
- Qinqin Wu
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China
- Department of Radiology, Changzhou Xinbei District Sanjing People's Hospital, Changzhou, 213200, Jiangsu, China
| | - Weiguang Qiang
- Department of Oncology, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Liang Pan
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Tingting Cha
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Qilin Li
- Department of Radiotherapy, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China
| | - Yang Gao
- Department of Radiology, Changzhou Xinbei District Sanjing People's Hospital, Changzhou, 213200, Jiangsu, China
| | - Kaiyang Qiu
- Department of Radiology, Changzhou Xinbei District Sanjing People's Hospital, Changzhou, 213200, Jiangsu, China
| | - Wei Xing
- Department of Radiology, The Third Affiliated Hospital of Soochow University, Changzhou First People's Hospital, Changzhou, 213003, Jiangsu, China.
| |
Collapse
|
4
|
Zhuo L, Chen W, Xing L, Li X, Song Z, Dong J, Zhang Y, Li H, Cui J, Han Y, Hao J, Wang J, Yin X, Li C. MRI-based quantification of intratumoral heterogeneity for intrahepatic mass-forming cholangiocarcinoma grading: a multicenter study. Insights Imaging 2025; 16:101. [PMID: 40369381 PMCID: PMC12078897 DOI: 10.1186/s13244-025-01985-9] [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: 12/18/2024] [Accepted: 04/27/2025] [Indexed: 05/16/2025] Open
Abstract
OBJECTIVE This study aimed to develop a quantitative approach to measure intratumor heterogeneity (ITH) using MRI scans and predict the pathological grading of intrahepatic mass-forming cholangiocarcinoma (IMCC). METHODS Preoperative MRI scans from IMCC patients were retrospectively obtained from five academic medical centers, covering the period from March 2018 to April 2024. Radiomic features were extracted from the whole tumor and its subregions, which were segmented using K-means clustering. An ITH index was derived from a habitat model integrating output probabilities of the subregions-based models. Significant variables from clinical laboratory-imaging features, radiomics, and the habitat model were integrated into a predictive model, and its performance was evaluated using the area under the receiver operating characteristic curve (AUC). RESULTS The final training and internal validation datasets included 197 patients (median age, 59 years [IQR, 52-65 years]); the external validation dataset included 43 patients (median age, 58.5 years [IQR, 52.25-69.75 years]). The habitat model achieved AUCs of 0.847 (95% CI: 0.783, 0.911) in the training set and 0.753 (95% CI: 0.595, 0.911) in the internal validation set. Furthermore, the combined model, integrating imaging variables, the habitat model, and radiomics model, demonstrated improved predictive performance, with AUCs of 0.895 (95% CI: 0.845, 0.944) in the training dataset, 0.790 (95% CI: 0.65, 0.931) in the internal validation dataset, and 0.815 (95% CI: 0.68, 0.951) in the external validation dataset. CONCLUSION The combined model based on MRI-derived quantification of ITH, along with clinical, laboratory, radiological, and radiomic features, showed good performance in predicting IMCC grading. CRITICAL RELEVANCE STATEMENT This model, integrating MRI-derived intrahepatic mass-forming cholangiocarcinoma (IMCC) classification metrics with quantitative radiomic analysis of intratumor heterogeneity (ITH), demonstrates enhanced accuracy in tumor grade prediction, advancing risk stratification for clinical decision-making in IMCC management. KEY POINTS Grading of intrahepatic mass-forming cholangiocarcinoma (IMCC) is important for risk stratification, clinical decision-making, and personalized therapeutic optimization. Quantitative intratumor heterogeneity can accurately predict the pathological grading of IMCC. This combined model provides higher diagnostic accuracy.
Collapse
Affiliation(s)
- Liyong Zhuo
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Wenjing Chen
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd., Beijing, People's Republic of China
| | - Lihong Xing
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Xiaomeng Li
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Zijun Song
- Department of Critical Care Medicine, Baoding First Central Hospital, Baoding, People's Republic of China
| | - Jinghui Dong
- Department of Radiology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Yanyan Zhang
- Department of Radiology, Beijing You'an Hospital, Beijing, People's Republic of China
| | - Hongjun Li
- Department of Radiology, Beijing You'an Hospital, Beijing, People's Republic of China
| | - Jingjing Cui
- Department of Research and Development, United Imaging Intelligence (Beijing) Co., Ltd., Beijing, People's Republic of China
| | - Yuxiao Han
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Jiawei Hao
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Jianing Wang
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China
| | - Xiaoping Yin
- Department of Radiology, Affiliated Hospital of Hebei University, Baoding, People's Republic of China.
| | - Caiying Li
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang, People's Republic of China.
| |
Collapse
|
5
|
Xu J, Miao JG, Wang CX, Zhu YP, Liu K, Qin SY, Chen HS, Lang N. CT-based quantification of intratumoral heterogeneity for predicting distant metastasis in retroperitoneal sarcoma. Insights Imaging 2025; 16:99. [PMID: 40346399 PMCID: PMC12064543 DOI: 10.1186/s13244-025-01977-9] [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: 02/05/2025] [Accepted: 04/23/2025] [Indexed: 05/11/2025] Open
Abstract
OBJECTIVES Retroperitoneal sarcoma (RPS) is highly heterogeneous, leading to different risks of distant metastasis (DM) among patients with the same clinical stage. This study aims to develop a quantitative method for assessing intratumoral heterogeneity (ITH) using preoperative contrast-enhanced CT (CECT) scans and evaluate its ability to predict DM risk. METHODS We conducted a retrospective analysis of 274 PRS patients who underwent complete surgical resection and were monitored for ≥ 36 months at two centers. Conventional radiomics (C-radiomics), ITH radiomics, and deep-learning (DL) features were extracted from the preoperative CECT scans and developed single-modality models. Clinical indicators and high-throughput CECT features were integrated to develop a combined model for predicting DM. The performance of the models was evaluated by measuring the receiver operating characteristic curve and Harrell's concordance index (C-index). Distant metastasis-free survival (DMFS) was also predicted to further assess survival benefits. RESULTS The ITH model demonstrated satisfactory predictive capability for DM in internal and external validation cohorts (AUC: 0.735, 0.765; C-index: 0.691, 0.729). The combined model that combined clinicoradiological variables, ITH-score, and DL-score achieved the best predictive performance in internal and external validation cohorts (AUC: 0.864, 0.801; C-index: 0.770, 0.752), successfully stratified patients into high- and low-risk groups for DM (p < 0.05). CONCLUSIONS The combined model demonstrated promising potential for accurately predicting the DM risk and stratifying the DMFS risk in RPS patients undergoing complete surgical resection, providing a valuable tool for guiding treatment decisions and follow-up strategies. CRITICAL RELEVANCE STATEMENT The intratumoral heterogeneity analysis facilitates the identification of high-risk retroperitoneal sarcoma patients prone to distant metastasis and poor prognoses, enabling the selection of candidates for more aggressive surgical and post-surgical interventions. KEY POINTS Preoperative identification of retroperitoneal sarcoma (RPS) with a high potential for distant metastasis (DM) is crucial for targeted interventional strategies. Quantitative assessment of intratumoral heterogeneity achieved reasonable performance for predicting DM. The integrated model combining clinicoradiological variables, ITH radiomics, and deep-learning features effectively predicted distant metastasis-free survival.
Collapse
Affiliation(s)
- Jun Xu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Jian-Guo Miao
- The College of Computer Science & Technology, Qingdao University, No. 308, Ning Xia Road, Shinan District, Qingdao, Shandong, China
| | - Chen-Xi Wang
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Yu-Peng Zhu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Ke Liu
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Si-Yuan Qin
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China
| | - Hai-Song Chen
- Department of Radiology, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, Shandong, China.
| | - Ning Lang
- Department of Radiology, Peking University Third Hospital, No. 49, North Garden Road, Haidian District, Beijing, China.
| |
Collapse
|
6
|
Wang X, Huang Y, Shi J, Cao Y, Chen H, Li L, Wang L, Tang S, Gong X, Huang H, Yin T, Zhang J. Biomechanical parameters quantified by MR elastography for predicting response to neoadjuvant chemotherapy and disease-free survival in breast cancer: a prospective longitudinal study. Breast Cancer Res 2025; 27:72. [PMID: 40329402 PMCID: PMC12057177 DOI: 10.1186/s13058-025-02035-4] [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: 09/25/2024] [Accepted: 04/24/2025] [Indexed: 05/08/2025] Open
Abstract
BACKGROUND Little is known regarding biomechanical properties derived from multifrequency MR elastography temporal changes during neoadjuvant chemotherapy (NAC) and associated with pathologic complete response (pCR) and disease-free survival (DFS) in breast cancer. We aimed to investigate temporal changes in NAC-associated biomechanical parameters and assess biomechanical parameters as a predictor of pCR and DFS in breast cancer. METHODS In this prospective longitudinal study, participants with breast cancer who received NAC were enrolled from February 2021 to May 2023. All participants underwent multifrequency MR-elastography at four timepoints: before NAC (T1) and after 2 (T2), 4 (T3), and 6 (T4) cycles. Tomoelastography postprocessing provided biomechanical maps of shear-wave-speed (c) and loss-angle (φ) as proxies of stiffness and viscosity. The biomechanical parameters were validated by means of correlation with histopathologic measurements. Generalized estimating equations were used to compare temporal changes in biomechanical parameters at four time points. Logistic regression was used for pCR analysis and Cox proportional hazards regression was used for survival analysis. Predictive performance was assessed with area under the receiver operating characteristic curve (AUC) analysis. RESULTS A total of 235 women (50.6 ± 7.9 years) with 964 scans were enrolled. Biomechanical parameters were supported by positive correlations with pathologic examination-based stroma fraction (c: r =.76, P <.001; φ: r =.49, P =.008) and cellularity (c: r =.58, P =.001; φ: r =.40, P =.035). Progesterone receptor, human epidermal growth factor receptor-2 (HER2), T2-c, and T2-φ were independently associated with pCR (all P <.05). Estrogen receptor, HER2, clinical stage, and change in φ at the early stage of NAC were associated with PFS (all P <.05). The predictive model, which incorporated biomechanical parameters and clinicopathologic characteristics significantly outperformed the clinicopathologic model in predicting pCR (AUC: 0.95, 95% confidence interval [CI]: 0.92, 0.98 vs. 0.79, 95%CI: 0.73, 0.84; P <.001). The predictive model also showed good discrimination ability for DFS (C-index = 0.82, 95%CI: 0.72, 0.90) and stratified prognosis into low-risk and high-risk groups (log-rank, P <.001). CONCLUSIONS During NAC, patients with higher tumor stiffness and viscosity are less likely to achieve DFS and pCR. The biomechanical parameters exhibit excellent biological interpretability and serve as valuable biomarkers for predicting pCR and DFS in patients with breast cancer.
Collapse
Affiliation(s)
- Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yao Huang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Jinfang Shi
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ying Cao
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Huifang Chen
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lan Li
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Lu Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Sun Tang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Xueqin Gong
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Haiping Huang
- Department of Pathology, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Ting Yin
- MR Collaborations, Siemens Healthineers Ltd, Chengdu, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing, 400030, China.
- Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), Chongqing University Cancer Hospital, No.181 Hanyu road, Shapingba district, Chongqing, 400030, China.
| |
Collapse
|
7
|
Xie Y, Wang F, Wei J, Shen Z, Song X, Wang Y, Chen H, Tao L, Zheng J, Lin L, Niu Z, Guan X, Zhou T, Xu Z, Liu Y, Du D, Pan H, Li S, Ji W, Zhou W, Yang Y, Tian J, Xu J, Hu H, Liang X. Noninvasive prognostic classification of ITH in HCC with multi-omics insights and therapeutic implications. SCIENCE ADVANCES 2025; 11:eads8323. [PMID: 40315307 PMCID: PMC12047409 DOI: 10.1126/sciadv.ads8323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Accepted: 03/31/2025] [Indexed: 05/04/2025]
Abstract
Intratumoral heterogeneity (ITH) is a critical factor associated with treatment failure and disease relapse in hepatocellular carcinoma (HCC). However, decoding ITH in a noninvasive and comprehensive manner remains a notable challenge. In this study involving 851 patients from five centers, we developed a noninvasive prognostic classification for ITH using radiomics based on multisequence MRI, termed radiomics ITH (RITH) phenotypes. The RITH phenotypes highly correlated with prognosis and pathological ITH. In addition, through an integrated multi-omics analysis, we uncovered the molecular mechanisms underlying RITH, notably enhancing its biological interpretability. Specifically, high-RITH tumors demonstrated an enrichment of cancer-associated fibroblasts and activation of extracellular matrix remodeling. Our approach facilitates the noninvasive refined classification of ITH using radiomics and multi-omics, paving the way for tailored treatment strategies in HCC. Extracellular matrix-receptor interaction could be a potential therapeutic target in patients with high-RITH tumors. Given the routine use of radiologic imaging in oncology, our methodology ignites versatile framework for broader application to other solid tumors.
Collapse
Affiliation(s)
- Yangyang Xie
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Fang Wang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030 Wuhan, China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, 430022 Wuhan, China
- Hubei Key Laboratory of Molecular Imaging, 430022 Wuhan, China
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Jingwei Wei
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
| | - Zefeng Shen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Xue Song
- Department of Respiratory and Critical Care Medicine, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Yali Wang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjun Chen
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Liye Tao
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Junhao Zheng
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Lanfen Lin
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Ziwei Niu
- The College of Computer Science and Technology, Zhejiang University, 310027 Hangzhou, China
| | - Xiaojun Guan
- Department of Radiology, The Second Affiliated Hospital, Zhejiang University School of Medicine, 310009 Hangzhou, China
| | - Tianhan Zhou
- Department of General Surgery, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310007 Hangzhou, China
| | - Zhengao Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Yang Liu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Danwei Du
- Department of Anorectal, Hangzhou TCM Hospital Affiliated to Zhejiang Chinese Medical University, 310000 Hangzhou, China
| | - Haoyu Pan
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Shihao Li
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Wenbin Ji
- Department of Radiology, Taizhou Hospital of Zhejiang Province Affiliated to Wenzhou Medical University, 317000 Taizhou, China
| | - Wei Zhou
- Department of Radiology, Huzhou Central Hospital, Fifth School of Clinical Medicine of Zhejiang Chinese Medical University, 313000 Huzhou, China
| | - Yunjun Yang
- Department of Radiology, The First Affiliated Hospital, Wenzhou Medical University, 325006 Wenzhou, China
| | - Jie Tian
- Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, 100190 Beijing, China
- Beijing Key Laboratory of Molecular Imaging, 100190 Beijing, China
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, 100191 Beijing, China
- Engineering Research Center of Molecular and Neuro Imaging of Ministry of Education, School of Life Science and Technology, Xidian University, 710126 Xi’an, China
| | - Junjie Xu
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- Zhejiang Minimal Invasive Diagnosis and Treatment Technology Research Center of Severe Hepatobiliary Disease, Zhejiang Research and Development Engineering Laboratory of Minimally Invasive Technology and Equipment, 310016 Hangzhou, China
- Zhejiang University Cancer Center, 310058 Hangzhou, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 311121 Hangzhou, China
| | - Hongjie Hu
- Department of Radiology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
| | - Xiao Liang
- Zhejiang Key Laboratory of Multi-omics Precision Diagnosis and Treatment of Liver Diseases, Department of General Surgery, Sir Run-Run Shaw Hospital, Zhejiang University School of Medicine, 310016 Hangzhou, China
- School of Medicine, Shaoxing University, 312000 Shaoxing, China
- School of Basic Medical Sciences and Forensic Medicine, Hangzhou Medical College, 310000 Hangzhou, China
| |
Collapse
|
8
|
Samari M, Alamzadeh Z, Irajirad R, Sarikhani A, Mahabadi VP, Ghaznavi H, Khoei S. FROP-1 peptide-conjugated ultrasmall superparamagnetic nanoparticles as a targeted T1-weighted MR contrast agent for breast cancer: in vitro study. BMC Biomed Eng 2025; 7:5. [PMID: 40307948 PMCID: PMC12044754 DOI: 10.1186/s42490-025-00091-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Accepted: 04/02/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND The aim of this study was to produce ultrasmall superparamagnetic iron oxide (USPIO) nanoparticles (NPs) conjugated to the FROP-1 peptide for targeted magnetic resonance imaging (MRI) of breast cancer cell lines and to evaluate its application as a specific and targeted T1-weighted MR imaging contrast agent in vitro. Sodium citrate-stabilized Fe3O4 NPs were conjugated with the FROP-1 peptide by 1-ethyl-3-(3-dimethylaminopropyl) carbide diamide hydrochloride (EDC) to form a novel Fe3O4@FROP-1 specific target contrast agent. The specificity and targeting of Fe3O4@FROP-1 to bind FROP-1 receptors were investigated in vitro by cellular uptake and cellular MR imaging. RESULTS In this study, the synthesis of water-soluble ultrasmall Fe3O4 NPs was performed by the co-precipitation method. XRD, TEM, and VSM analyses showed the formation of the Fe3O4 NPs with an average size of about 3.78 ± 0.2 nm. FT-IR spectroscopy approved the conjugation of the FROP-1 peptide with the Fe3O4 NPs. The synthesized Fe3O4@FROP-1 NPs showed good biocompatibility, and the high r1 relaxivity and r2/r1, respectively, were 2.608 mM- 1S- 1 and 1.18. The biocompatibility of the Fe3O4 and Fe3O4@FROP-1 NPs on the MCF-7, SKBR-3, MDA-MB-231, and MCF-10 cell lines was determined using cytotoxicity analysis. The specific targeting effect on the cells was verified by in vitro cellular uptake and cell MR imaging. CONCLUSION It was found that the contrast intensity of the Fe3O4@FROP-1 nanoprobe increases as Fe concentration increases. Cellular uptake of the Fe3O4 and Fe3O4@FROP-1 NPs was quantified using ICP-MS. The synthesized NPs had better imaging performance than Dotarem (gadoterate meglumine). The findings showed that Fe3O4@FROP-1 NPs have potential utility as a specific and targeted T1-weighted contrast agent in breast cancer MR imaging.
Collapse
Affiliation(s)
- Melika Samari
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | - Zahra Alamzadeh
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Rasoul Irajirad
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Abolfazl Sarikhani
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran
| | | | - Habib Ghaznavi
- Pharmacology Research Center, Zahedan University of Medical Sciences, Zahedan, Iran.
| | - Samideh Khoei
- Finetech in Medicine Research Center, Iran University of Medical Sciences, Tehran, Iran.
- Medical Physics Department, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
9
|
Chen S, Zhang Y, Su Y, Tian J, Chen Y, Tang W, Fan Y, Jin C, He Y, Xu Y, Hu H, Guo Y, Li J. Habitat Radiomics Based on Dynamic Contrast-Enhanced Magnetic Resonance Imaging for Assessing Axillary Lymph Node Burden in Clinical T1-T2 Stage Breast Cancer: A Multicenter and Interpretable Study. J Magn Reson Imaging 2025. [PMID: 40256826 DOI: 10.1002/jmri.29796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 04/02/2025] [Accepted: 04/03/2025] [Indexed: 04/22/2025] Open
Abstract
BACKGROUND Axillary lymph node burden(ALNB) is a critical factor in determining treatment strategies for clinical T1-T2 (cT1-T2) stage breast cancer. However, as ALNB assessment relies on invasive procedures, exploring non-invasive methods is essential. PURPOSE To develop and validate a habitat radiomics model for assessing ALNB in cT1-T2 breast cancer, incorporating radiogenomic data to improve interpretability. STUDY TYPE Retrospective. POPULATION 468 patients with cT1-T2 stage breast cancer from two institutions and The Cancer Imaging Archive (TCIA) and The Cancer Genome Atlas (TCGA)-Breast Invasive Carcinoma (BRCA) were included. The cohort was divided into training (n = 173), internal validation (n = 58), external validation (n = 130), and TCGA-BRCA sets (n = 107). Patients were categorized into high nodal burden (HNB; > 3 positive lymph nodes) and non-HNB (≤ 3 positive lymph nodes) groups. FIELD STRENGTH/SEQUENCE 1.5-T MRI and 3.0-T MRI, and three-dimensional dynamic contrast-enhanced T1-weighted gradient-echo sequences. ASSESSMENT Two logistic regression models were developed using habitat-based and clinical features. Model performance was evaluated using the AUC. SHapley Additive exPlanations (SHAP) analysis was employed to identify key features. Radiogenomic analysis, including gene set enrichment and drug sensitivity assessments, was conducted using transcriptomic data from the TCGA-BRCA set. STATISTICAL TESTS Pearson correlation, Mann-Whitney U, genetic algorithm, logistic regression, AUC analysis, delong test, and SHAP analysis. A p-value < 0.05 was considered statistically significant. RESULTS The Habitat model outperformed the Clinical model (AUCs: 0.840-0.932 vs. 0.558-0.673). The SHAP analysis was used to rank feature importance, with subregion 3 showing the highest average SHAP value. Radiogenomic analysis indicated upregulation of the KEGG ribosome pathway in the HNB group and identified differential drug sensitivity profiles among risk groups. DATA CONCLUSION The Habitat model has the potential to assess ALNB in cT1-T2 breast cancer and assist radiologists in axillary diagnosis, which may help reduce the need for unnecessary ALN dissection. EVIDENCE LEVEL 3. Technical Efficacy: Stage 2.
Collapse
Affiliation(s)
- Siyi Chen
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yue Zhang
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yuqi Su
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Jie Tian
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yongxin Chen
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Wenjie Tang
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Yaheng Fan
- Shukun Technology Co., Ltd, Beijing, China
| | - Chen Jin
- College of Computer Science and Technology, Zhejiang University of Technology, Zhejiang, China
| | - Yangcheng He
- Department of Ultrasound, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | | | - Hong Hu
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| | - Yuan Guo
- Department of Radiology, Guangzhou First People's Hospital, South China University of Technology, Guangzhou, China
| | - Junping Li
- Department of Radiology, Xiangyang No. 1 People's Hospital, Hubei University of Medicine, Xiangyang, China
| |
Collapse
|
10
|
Wang H, Qi Y, Liu X, Song LJ, Yang WB, Li MA, Bai XY, Xu MS, Zhu HN, Cai SQ, Wang Y, Yang ZH, Li YZ, Wang ZC, Guo YF. Habitat analysis of iron deposition in the basal ganglia for diagnosing cognitive impairment in chronic kidney disease: evidence from a case-control study. BMC Med Imaging 2025; 25:113. [PMID: 40200199 PMCID: PMC11980340 DOI: 10.1186/s12880-025-01656-7] [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: 11/21/2024] [Accepted: 04/01/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Chronic kidney disease induces alterations in the heterogeneity of iron deposition within the basal ganglia. Quantitative analysis of the heterogeneity of iron deposition within the basal ganglia may be valuable for diagnosing chronic kidney disease-related cognitive impairment. METHODS In this prospective observational cohort study, quantitative susceptibility mapping (QSM) was performed in chronic kidney disease patients. Susceptibility values of each nucleus within the basal ganglia were measured. Radiomic features were extracted from habitats of the basal ganglia on QSM images. Habitat-based models for diagnosing cognitive impairment were constructed using the random forest algorithm. Logistic regression was employed to build the clinical model and the combined model. The performance of each model was evaluated by the receiver operating characteristic (ROC) analysis. RESULTS A total of 146 patients (mean age, 51 ± 13 years; 92 male) were included, of which 79 had cognitive impairment. The two habitats-based model achieved an area under the curve of 0.926 (95% CI 0.842-1.000) on the test set, the highest among all prediction models. The two-habitat maps indicated that chronic kidney disease had two distinct patterns of impact on iron deposition in the basal ganglia region. The capability of the two habitats-based model to identify chronic kidney disease-related cognitive impairment was significantly superior to that of the susceptibility values measured in various nuclei (all p < 0.05). CONCLUSIONS This study innovatively applied a habitat-based quantitative analysis technique to QSM, successfully constructing a model that accurately diagnoses chronic kidney disease-related cognitive impairment. TRIAL REGISTRATION This study was approved by the Beijing Friendship Hospital Ethics Board (ClinicalTrials.gov Identifier: NCTO5137470) and conducted in accordance with the Declaration of Helsinki ethical standards.
Collapse
Affiliation(s)
- Hao Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Yu Qi
- Department of Radiology, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Xu Liu
- Department of Nephrology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li-Jun Song
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Wen-Bo Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Ming-An Li
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Xiao-Yan Bai
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Mao-Sheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Hao-Nan Zhu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Si-Qing Cai
- Center of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
- School of Medical Imaging, Fujian Medical University, Fuzhou, Fujian, China
| | - Yi Wang
- Center of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China
| | - Zheng-Han Yang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China
| | - Yuan-Zhe Li
- Center of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, Fujian, China.
- School of Medical Imaging, Fujian Medical University, Fuzhou, Fujian, China.
| | - Zhen-Chang Wang
- Department of Radiology, Beijing Friendship Hospital, Capital Medical University, No. 95 Yong An Road, Xicheng District, Beijing, 100050, China.
| | - Yi-Fan Guo
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China.
| |
Collapse
|
11
|
Du S, Xie W, Gao S, Zhao R, Wang H, Tian J, Liu J, Liu Z, Zhang L. Early prediction of neoadjuvant therapy response in breast cancer using MRI-based neural networks: data from the ACRIN 6698 trial and a prospective Chinese cohort. Breast Cancer Res 2025; 27:52. [PMID: 40181457 PMCID: PMC11969705 DOI: 10.1186/s13058-025-02009-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Early prediction of treatment response to neoadjuvant therapy (NAT) in breast cancer patients can facilitate timely adjustment of treatment regimens. We aimed to develop and validate a MRI-based enhanced self-attention network (MESN) for predicting pathological complete response (pCR) based on longitudinal images at the early stage of NAT. METHODS Two imaging datasets were utilized: a subset from the ACRIN 6698 trial (dataset A, n = 227) and a prospective collection from a Chinese hospital (dataset B, n = 245). These datasets were divided into three cohorts: an ACRIN 6698 training cohort (n = 153) from dataset A, an ACRIN 6698 test cohort (n = 74) from dataset A, and an external test cohort (n = 245) from dataset B. The proposed MESN allowed for the integration of multiple timepoint features and extraction of dynamic information from longitudinal MR images before and after early-NAT. We also constructed the Pre model based on pre-NAT MRI features. Clinicopathological characteristics were added to these image-based models to create integrated models (MESN-C and Pre-C), and their performance was evaluated and compared. RESULTS The MESN-C yielded area under the receiver operating characteristic curve (AUC) values of 0.944 (95% CI: 0.906 - 0.973), 0.903 (95%CI: 0.815 - 0.965), and 0.861 (95%CI: 0.811 - 0.906) in the ACRIN 6698 training, ACRIN 6698 test and external test cohorts, respectively, which were significantly higher than those of the clinical model (AUC: 0.720 [95%CI: 0.587 - 0.842], 0.738 [95%CI: 0.669 - 0.796] for the two test cohorts, respectively; p < 0.05) and Pre-C (AUC: 0.697 [95%CI: 0.554 - 0.819], 0.726 [95%CI: 0.666 - 0.797] for the two test cohorts, respectively; p < 0.05). High AUCs of the MESN-C maintained in the ACRIN 6698 standard (AUC = 0.853 [95%CI: 0.676 - 1.000]) and experimental (AUC = 0.905 [95%CI: 0.817 - 0.993]) subcohorts, and the interracial and external subcohort (AUC = 0.861 [95%CI: 0.811 - 0.906]). Moreover, the MESN-C increased the positive predictive value from 48.6 to 71.3% compared with Pre-C model, and maintained a high negative predictive value (80.4-86.7%). CONCLUSION The MESN-C using longitudinal multiparametric MRI after a short-term therapy achieved favorable performance for predicting pCR, which could facilitate timely adjustment of treatment regimens, increasing the rates of pCR and avoiding toxic effects. TRIAL REGISTRATION Trial registration at https://www.chictr.org.cn/ . REGISTRATION NUMBER ChiCTR2000038578, registered September 24, 2020.
Collapse
Affiliation(s)
- Siyao Du
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Wanfang Xie
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China
| | - Si Gao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Ruimeng Zhao
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China
| | - Huidong Wang
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110165, Liaoning Province, China
| | - Jie Tian
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
| | - Jiangang Liu
- School of Engineering Medicine, Beihang University, Beijing, 100191, People's Republic of China.
- Key Laboratory of Big Data-Based Precision Medicine (Beihang University), Ministry of Industry and Information Technology of the People's Republic of China, Beijing, 100191, People's Republic of China.
- Beijing Engineering Research Center of Cardiovascular Wisdom Diagnosis and Treatment, Beijing, 100029, People's Republic of China.
| | - Zhenyu Liu
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Beijing, 100190, People's Republic of China.
- University of Chinese Academy of Sciences, Beijing, 100080, People's Republic of China.
| | - Lina Zhang
- Department of Radiology, The First Hospital of China Medical University, Shenyang, 110001, Liaoning Province, China.
- Department of Radiology, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110165, Liaoning Province, China.
| |
Collapse
|
12
|
Zhang K, Ru J, Wang W, Xu M, Mu L, Pan J, Gu J, Zhang H, Tian J, Yang W, Jiang T, Wang K. ViT-based quantification of intratumoral heterogeneity for predicting the early recurrence in HCC following multiple ablation. Liver Int 2025; 45:e16051. [PMID: 39526488 DOI: 10.1111/liv.16051] [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: 04/03/2024] [Revised: 07/03/2024] [Accepted: 07/11/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This study aimed to develop a quantitative intratumoral heterogeneity (ITH) model for assessing the risk of early recurrence (ER) in pre-treatment multimodal imaging for hepatocellular carcinoma (HCC) patients undergoing ablation treatments. METHODS This multi-centre study enrolled 633 HCC patients who underwent ultrasound-guided local ablation between January 2015 and September 2022. Among them, 422, 85, 57 and 69 patients underwent radiofrequency ablation (RFA), microwave ablation (MWA), laser ablation (LA) and irreversible electroporation (IRE) ablation, respectively. Vision-Transformer-based quantitative ITH (ViT-Q-ITH) features were extracted from the US and MRI sequences. Multivariable logistic regression analysis was used to identify variables associated with ER. A combined model integrated clinic-radiologic and ViT-Q-ITH scores. The prediction performance was evaluated concerning calibration, clinical usefulness and discrimination. RESULTS The final training cohort and internal validation cohort included 318 patients and 83 patients, respectively, who underwent RFA and MWA. The three external testing cohorts comprised of 106 patients treated with RFA, 57 patients treated with LA and 69 patients who underwent IRE ablation. The combined model showed excellent predictive performance for ER in the training (AUC: .99, 95% CI: .99-1.00), internal validation (AUC: .86, 95% CI: .78-.94), external testing (AUC: .83, 95% CI: .73-.92), LA (AUC: .84, 95% CI: .73-.95) and IRE (AUC: .82, 95% CI: .72-.93) cohorts, respectively. Decision curve analysis further affirmed the clinical utility of the combined model. CONCLUSIONS The multimodal-based model, incorporating clinic-radiologic factors and ITH features, demonstrated superior performance in predicting ER among early-stage HCC patients undergoing different ablation modalities.
Collapse
Affiliation(s)
- Ke Zhang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinyu Ru
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Wenbo Wang
- Department of Ultrasound, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Min Xu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Lei Mu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jinhua Pan
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jionghui Gu
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haoyan Zhang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| | - Jie Tian
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Engineering Medicine, Beihang University, Beijing, China
| | - Wei Yang
- Department of Ultrasound, Key laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Peking University Cancer Hospital & Institute, Beijing, China
| | - Tianan Jiang
- Department of Ultrasound Medicine, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Kun Wang
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
13
|
Feng K, Yi Z, Xu B. Artificial Intelligence and Breast Cancer Management: From Data to the Clinic. CANCER INNOVATION 2025; 4:e159. [PMID: 39981497 PMCID: PMC11840326 DOI: 10.1002/cai2.159] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2024] [Revised: 10/10/2024] [Accepted: 10/22/2024] [Indexed: 02/22/2025]
Abstract
Breast cancer (BC) remains a significant threat to women's health worldwide. The oncology field had an exponential growth in the abundance of medical images, clinical information, and genomic data. With its continuous advancement and refinement, artificial intelligence (AI) has demonstrated exceptional capabilities in processing intricate multidimensional BC-related data. AI has proven advantageous in various facets of BC management, encompassing efficient screening and diagnosis, precise prognosis assessment, and personalized treatment planning. However, the implementation of AI into precision medicine and clinical practice presents ongoing challenges that necessitate enhanced regulation, transparency, fairness, and integration of multiple clinical pathways. In this review, we provide a comprehensive overview of the current research related to AI in BC, highlighting its extensive applications throughout the whole BC cycle management and its potential for innovative impact. Furthermore, this article emphasizes the significance of constructing patient-oriented AI algorithms. Additionally, we explore the opportunities and potential research directions within this burgeoning field.
Collapse
Affiliation(s)
- Kaixiang Feng
- Department of Breast and Thyroid Surgery, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Zongbi Yi
- Department of Radiation and Medical Oncology, Hubei Key Laboratory of Tumor Biological Behaviors, Hubei Cancer Clinical Study CenterZhongnan Hospital of Wuhan UniversityWuhanHubeiChina
| | - Binghe Xu
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
14
|
Lin Y, Cheng M, Wu C, Huang Y, Zhu T, Li J, Gao H, Wang K. MRI-based artificial intelligence models for post-neoadjuvant surgery personalization in breast cancer: a narrative review of evidence from Western Pacific. THE LANCET REGIONAL HEALTH. WESTERN PACIFIC 2025; 57:101254. [PMID: 40443543 PMCID: PMC12121432 DOI: 10.1016/j.lanwpc.2024.101254] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 11/06/2024] [Accepted: 11/19/2024] [Indexed: 06/02/2025]
Abstract
Breast magnetic resonance imaging (MRI) is the most sensitive imaging method for diagnosing breast cancer and assessing treatment response. Artificial intelligence (AI) and radiomics offer new opportunities to identify patterns in imaging data, supporting personalized post-neoadjuvant surgical decisions. This paper reviewed breast MRI-based AI models for predicting outcomes after neoadjuvant therapy, with a focus on evidence from the Western Pacific region, to evaluate the quality of existing models, discuss their inherent limitations, and outline potential future directions. A literature search in MEDLINE, EMBASE, and Web of Science identified 51 relevant studies in the region, with the majority conducted in China, followed by South Korea and Japan. Most studies focused on predicting pathologic complete response (pCR), with a median sample size of 152 and largely retrospective single-center designs. Model performance was commonly assessed using validation sets, with pooled sensitivity and specificity for pCR prediction showing promising results. Models incorporating multitemporal MRI features were associated with improved accuracy. While MRI-based AI models show potential for guiding surgical planning, improved methodological quality and algorithmic explainability are needed to facilitate clinical translation.
Collapse
Affiliation(s)
- Yingyi Lin
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Minyi Cheng
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Cangui Wu
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
- Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Yuhong Huang
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Teng Zhu
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Jieqing Li
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Hongfei Gao
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| | - Kun Wang
- School of Medicine, South China University of Technology, Guangzhou, Guangdong 510006, China
- Department of Breast Cancer, Cancer Centre, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong 510080, China
| |
Collapse
|
15
|
Huang Y, Wang X, Cao Y, Lan X, Hu X, Mou F, Chen H, Gong X, Li L, Tang S, Wang L, Zhang J. Nomogram for Predicting Neoadjuvant Chemotherapy Response in Breast Cancer Using MRI-based Intratumoral Heterogeneity Quantification. Radiology 2025; 315:e241805. [PMID: 40232145 DOI: 10.1148/radiol.241805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025]
Abstract
Background Intratumoral heterogeneity (ITH) in breast cancer contributes to treatment failure and relapse. Noninvasive methods to quantify ITH are currently limited. Purpose To quantify ITH in breast cancer using pretreatment MRI, develop a nomogram to predict pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) and recurrence-free survival (RFS), and investigate biologic pathways associated with nomogram scores. Materials and Methods This retrospective study included patients with breast cancer who underwent NAC at nine centers between April 1988 and December 2023. Tumor regions on MRI scans were clustered and integrated with global pixel distribution patterns to calculate ITH scores. A nomogram for predicting pCR was developed using multivariable logistic regression. A survival dataset was used to evaluate the association between nomogram score and RFS, and a genomics dataset was used to explore the relationship between nomogram score and biologic pathways. Results The study included 1448 women (median age, 49 years [IQR, 43-54 years]). To predict pCR to NAC, the 505 patients from center A served as the training set, and the patients from center B, centers C-F, and center G served as three external validation sets (n = 331, 107, and 384, respectively). The survival set included patients from centers A and H (n = 179), and the genomics set included patients from center I (n = 74). The ITH score was an independent predictor of pCR (odds ratio, 0.12 [95% CI: 0.03, 0.43]; P < .001). The nomogram model achieved area under the receiver operating characteristic curve values of 0.82, 0.81, and 0.79, respectively, in the three external validation sets. A lower nomogram score was correlated with poorer RFS (hazard ratio, 4.04 [95% CI: 1.90, 8.60]; P < .001) and was associated with upregulation of biologic pathways related to tumor proliferation. Conclusion A nomogram model combining ITH score and clinicopathologic variables showed good performance in predicting pCR to NAC and RFS. Published under a CC BY 4.0 license. Supplemental material is available for this article.
Collapse
Affiliation(s)
- Yao Huang
- School of Medicine, Chongqing University, Chongqing, China
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Xiaoxia Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Ying Cao
- School of Medicine, Chongqing University, Chongqing, China
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Xiaosong Lan
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Xiaofei Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University, Chongqing, China
| | - Fangsheng Mou
- Chongqing Three Gorges University Hospital, Chongqing, China
| | - Huifang Chen
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Xueqin Gong
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Lan Li
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Sun Tang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Lu Wang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| | - Jiuquan Zhang
- Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer, No. 181 Hanyu Rd, Shapingba District, Chongqing 400030, China
| |
Collapse
|
16
|
Shen J, Li Q, Li L, Lu T, Han J, Xie Z, Wang P, Cao Z, Zeng M, Zhou J, Yu T, Xu Y, Sun H. Contrast-enhanced MRI-based intratumoral heterogeneity assessment for predicting lymph node metastasis in resectable pancreatic ductal adenocarcinoma. Insights Imaging 2025; 16:76. [PMID: 40159327 PMCID: PMC11955437 DOI: 10.1186/s13244-025-01956-0] [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: 01/08/2025] [Accepted: 03/17/2025] [Indexed: 04/02/2025] Open
Abstract
OBJECTIVES To develop and validate a contrast-enhanced MRI-based intratumoral heterogeneity (ITH) model for predicting lymph node (LN) metastasis in resectable pancreatic ductal adenocarcinoma (PDAC). METHODS Lesions were encoded into different habitats based on enhancement ratios at arterial, venous, and delayed phases of contrast-enhanced MRI. Habitat models on enhanced ratio mapping and single sequences, radiomic models, and clinical models were developed for evaluating LN metastasis. The performance of the models was evaluated via different metrics. Additionally, patients were stratified into high-risk and low-risk groups based on an ensembled model to assess prognosis after adjuvant therapy. RESULTS We developed an ensembled radiomics-habitat-clinical (RHC) model that integrates radiomics, habitat, and clinical data for precise prediction of LN metastasis in PDAC. The RHC model showed strong predictive performance, with area under the curve (AUC) values of 0.805, 0.779, and 0.615 in the derivation, internal validation, and external validation cohorts, respectively. Using an optimal threshold of 0.46, the model effectively stratified patients, revealing significant differences in recurrence-free survival and overall survival (OS) (p = 0.004 and p < 0.001). Adjuvant therapy improved OS in the high-risk group (p = 0.004), but no significant benefit was observed in the low-risk group (p = 0.069). CONCLUSION We developed an MRI-based ITH model that provides reliable estimates of LN metastasis for resectable PDAC and may offer additional value in guiding clinical decision-making. CRITICAL RELEVANCE STATEMENT This ensemble RHC model facilitates preoperative prediction of LN metastasis in resectable PDAC using contrast-enhanced MRI. This offers a foundation for enhanced prognostic assessment and supports the management of personalized adjuvant treatment strategies. KEY POINTS MRI-based habitat models can predict LN metastasis in PDAC. Both the radiomics model and clinical characteristics were useful for predicting LN metastasis in PDAC. The RHC models have the potential to enhance predictive accuracy and inform personalized therapeutic decisions.
Collapse
Affiliation(s)
- Junjian Shen
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Qing Li
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Lei Li
- Department of Radiology, Fengyang County People's Hospital, Chuzhou, China
| | - Tianyu Lu
- Department of Radiology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Jun Han
- Department of Radiology, The First Hospital of Jiaxing, Affiliated Hospital of Jiaxing University, Jiaxing, China
| | - Zongyu Xie
- Department of Radiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Peng Wang
- Department of Radiology, Affiliated Hospital of Jiangnan University, Wuxi, P.R. China
| | - Zirui Cao
- Department of Research Center, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jianjun Zhou
- Department of Radiology, Zhongshan Hospital (Xiamen), Fudan University, Xiamen Municipal Clinical Research Center for Medical Imaging, Fujian Province Key Clinical Specialty for Medical Imaging, Xiamen Key Laboratory of Clinical Transformation of Imaging Big Data and Artificial Intelligence, Xiamen, China
| | - Tianzhu Yu
- Department of Interventional Radiology, Zhongshan Hospital, Shanghai, China
| | - Yaolin Xu
- Department of Pancreatic Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Haitao Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China.
| |
Collapse
|
17
|
Song S, Zhang G, Yao Z, Chen R, Liu K, Zhang T, Zeng G, Wang Z, Liu R. Deep learning based on intratumoral heterogeneity predicts histopathologic grade of hepatocellular carcinoma. BMC Cancer 2025; 25:497. [PMID: 40102774 PMCID: PMC11917083 DOI: 10.1186/s12885-025-13781-1] [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: 10/22/2024] [Accepted: 02/20/2025] [Indexed: 03/20/2025] Open
Abstract
OBJECTIVES The potential of medical imaging to non-invasively assess intratumoral heterogeneity (ITH) is increasingly being recognized. This study aimed to investigate the value of the ITH-based deep learning model for preoperative prediction of histopathologic grade in hepatocellular carcinoma (HCC). MATERIALS AND METHODS A total of 858 patients from primary cohort and two external cohorts were included. 3.0T or 1.5T axial portal venous phase MRI images were collected. We conducted radiomics feature-driven K-means clustering for automatic partition to reveal ITH. 2.5D and 3D deep learning models based on ResNet architecture were trained to extract deep learning hidden features of each subregion. The selected features were used to train Random Forest classifier, which constructed the feature-fusion model. RESULTS The extracted voxel-level radiomics features were unsupervised clustered by K-means to generate three subregions. In the 2.5D deep learning, the feature-fusion model based on ITH had superior predictive efficacy than the whole-tumor model (AUC: 0.82 vs. 0.72; p = 0.004). Even in the validation and external test sets, this model maintained a high AUC of 0.78-0.83, and net reclassification indices indicated that it could improve prediction by 25-28%. Regarding the prognostic value, overall survival (OS) and recurrence-free survival (RFS) could be significantly stratified by the 2.5D feature-fusion model, and multivariable Cox regressions indicated its signature was identified as a risk predictor for OS and RFS (p < 0.05). CONCLUSION The ITH-based feature-fusion model provided a non-invasive method for classifying tumor differentiation in HCC, which may serve as a promising strategy for stratification management.
Collapse
Affiliation(s)
- Shaoming Song
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Gong Zhang
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Key Laboratory of Digital Hepatobiliary Surgery, Chinese PLA General Hospital, Beijing, 100853, China
| | - Zhiyuan Yao
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Department of Hepatobiliary Surgery, The Sixth Medical Center of Chinese PLA General Hospital, Beijing, 100048, China
| | - Ruiqiu Chen
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Kai Liu
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
| | - Tianchen Zhang
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China
| | - Guineng Zeng
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China
- Nankai University School of Medicine, Nankai University, Tianjin, 300071, China
| | - Zizheng Wang
- Department of Hepatobiliary Surgery, Senior Department of Hepatology, The Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Rong Liu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou, 730000, China.
- Faculty of Hepatopancreatobiliary Surgery, The First Medical Center of Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
18
|
Zeng Y, Wu H, Zhu Y, Li C, Du D, Song Y, Su S, Qin J, Jiang G. MRI-based intra-tumoral ecological diversity features and temporal characteristics for predicting microvascular invasion in hepatocellular carcinoma. Front Oncol 2025; 15:1510071. [PMID: 40098699 PMCID: PMC11911209 DOI: 10.3389/fonc.2025.1510071] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 02/10/2025] [Indexed: 03/19/2025] Open
Abstract
Objective To investigate the predictive value of radiomics models based on intra-tumoral ecological diversity (iTED) and temporal characteristics for assessing microvascular invasion (MVI) in patients with hepatocellular carcinoma (HCC). Material and Methods We retrospectively analyzed the data of 398 HCC patients who underwent dynamic contrast-enhanced MRI with Gd-EOB-DTPA (training set: 318; testing set: 80). The tumors were segmented into five distinct habitats using case-level clustering and a Gaussian mixture model was used to determine the optimal clusters based on the Bayesian information criterion to produce an iTED feature vector for each patient, which was used to assess intra-tumoral heterogeneity. Radiomics models were developed using iTED features from the arterial phase (AP), portal venous phase (PVP), and hepatobiliary phase (HBP), referred to as MiTED-AP, MiTED-PVP, and MiTED-HBP, respectively. Additionally, temporal features were derived by subtracting the PVP features from the AP features, creating a delta-radiomics model (MDelta). Conventional radiomics features were also extracted from the AP, PVP, and HBP images, resulting in three models: MCVT-AP, MCVT-PVP, and MCVT-HBP. A clinical-radiological model (CR model) was constructed, and two fusion models were generated by combining the radiomics or/and CR models using a stacking algorithm (fusion_R and fusion_CR). Model performance was evaluated using AUC, accuracy, sensitivity, and specificity. Results The MDelta model demonstrated higher sensitivity compared to the MCVT-AP and MCVT-PVP models. No significant differences in performance were observed across different imaging phases for either conventional radiomics (p = 0.096-0.420) or iTED features (p = 0.106-0.744). Similarly, for images from the same phase, we found no significant differences between the performance of conventional radiomics and iTED features (AP: p = 0.158; PVP: p = 0.844; HBP: p = 0.157). The fusion_R and fusion_CR models enhanced MVI discrimination, achieving AUCs of 0.823 (95% CI: 0.816-0.831) and 0.830 (95% CI: 0.824-0.835), respectively. Conclusion Delta radiomics features are temporal and predictive of MVI, providing additional predictive information for MVI beyond conventional AP and PVP features. The iTED features provide an alternative perspective in interpreting tumor characteristics and hold the potential to replace conventional radiomics features to some extent for MVI prediction.
Collapse
Affiliation(s)
- Yuli Zeng
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Huiqin Wu
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
| | - Yanqiu Zhu
- Department of Radiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Chao Li
- Department of Radiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Dongyang Du
- School of Computer Science, Inner Mongolia University, Inner Mongolia, China
| | - Yang Song
- Magnetic Resonance (MR) Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China
| | - Sulian Su
- Department of Radiology, Xiamen Humanity Hospital of Fujian Medical University, Xiamen, Fujian, China
| | - Jie Qin
- Department of Radiology, Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, Guangdong, China
| | - Guihua Jiang
- Department of Medical Imaging, The Affiliated Guangdong Second Provincial General Hospital of Jinan University, Guangzhou, Guangdong, China
- Department of Radiology, Xiamen Humanity Hospital of Fujian Medical University, Xiamen, Fujian, China
- Guangzhou Key Laboratory of Molecular Functional Imaging and Artificial Intelligence for Major Brain Diseases, Guangdong Second Provincial General Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
19
|
Wang C, Wu F, Wang F, Chong HH, Sun H, Huang P, Xiao Y, Yang C, Zeng M. The Association Between Tumor Radiomic Analysis and Peritumor Habitat-Derived Radiomic Analysis on Gadoxetate Disodium-Enhanced MRI With Microvascular Invasion in Hepatocellular Carcinoma. J Magn Reson Imaging 2025; 61:1428-1439. [PMID: 38997242 DOI: 10.1002/jmri.29523] [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: 04/17/2024] [Revised: 06/14/2024] [Accepted: 06/17/2024] [Indexed: 07/14/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has a poor prognosis, often characterized by microvascular invasion (MVI). Radiomics and habitat imaging offer potential for preoperative MVI assessment. PURPOSE To identify MVI in HCC by habitat imaging, tumor radiomic analysis, and peritumor habitat-derived radiomic analysis. STUDY TYPE Retrospective. SUBJECTS Three hundred eighteen patients (53 ± 11.42 years old; male = 276) with pathologically confirmed HCC (training:testing = 224:94). FIELD STRENGTH/SEQUENCE 1.5 T, T2WI (spin echo), and precontrast and dynamic T1WI using three-dimensional gradient echo sequence. ASSESSMENT Clinical model, habitat model, single sequence radiomic models, the peritumor habitat-derived radiomic model, and the combined models were constructed for evaluating MVI. Follow-up clinical data were obtained by a review of medical records or telephone interviews. STATISTICAL TESTS Univariable and multivariable logistic regression, receiver operating characteristic (ROC) curve, calibration, decision curve, Delong test, K-M curves, log rank test. A P-value less than 0.05 (two sides) was considered to indicate statistical significance. RESULTS Habitat imaging revealed a positive correlation between the number of subregions and MVI probability. The Radiomic-Pre model demonstrated AUCs of 0.815 (95% CI: 0.752-0.878) and 0.708 (95% CI: 0.599-0.817) for detecting MVI in the training and testing cohorts, respectively. Similarly, the AUCs for MVI detection using Radiomic-HBP were 0.790 (95% CI: 0.724-0.855) for the training cohort and 0.712 (95% CI: 0.604-0.820) for the test cohort. Combination models exhibited improved performance, with the Radiomics + Habitat + Dilation + Habitat 2 + Clinical Model (Model 7) achieving the higher AUC than Model 1-4 and 6 (0.825 vs. 0.688, 0.726, 0.785, 0.757, 0.804, P = 0.013, 0.048, 0.035, 0.041, 0.039, respectively) in the testing cohort. High-risk patients (cutoff value >0.11) identified by this model showed shorter recurrence-free survival. DATA CONCLUSION The combined model including tumor size, habitat imaging, radiomic analysis exhibited the best performance in predicting MVI, while also assessing prognostic risk. EVIDENCE LEVEL 3 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Cheng Wang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fei Wu
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fang Wang
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Huan-Huan Chong
- Department of Radiology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai, China
| | - Haitao Sun
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peng Huang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yuyao Xiao
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chun Yang
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Mengsu Zeng
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
20
|
Zhong J, Liu X, Lu J, Yang J, Zhang G, Mao S, Chen H, Yin Q, Cen Q, Jiang R, Song Y, Lu M, Chu J, Xing Y, Hu Y, Ding D, Ge X, Zhang H, Yao W. Overlooked and underpowered: a meta-research addressing sample size in radiomics prediction models for binary outcomes. Eur Radiol 2025; 35:1146-1156. [PMID: 39789271 PMCID: PMC11835977 DOI: 10.1007/s00330-024-11331-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Revised: 11/10/2024] [Accepted: 11/30/2024] [Indexed: 01/12/2025]
Abstract
OBJECTIVES To investigate how studies determine the sample size when developing radiomics prediction models for binary outcomes, and whether the sample size meets the estimates obtained by using established criteria. METHODS We identified radiomics studies that were published from 01 January 2023 to 31 December 2023 in seven leading peer-reviewed radiological journals. We reviewed the sample size justification methods, and actual sample size used. We calculated and compared the actual sample size used to the estimates obtained by using three established criteria proposed by Riley et al. We investigated which characteristics factors were associated with the sufficient sample size that meets the estimates obtained by using established criteria proposed by Riley et al. RESULTS: We included 116 studies. Eleven out of one hundred sixteen studies justified the sample size, in which 6/11 performed a priori sample size calculation. The median (first and third quartile, Q1, Q3) of the total sample size is 223 (130, 463), and those of sample size for training are 150 (90, 288). The median (Q1, Q3) difference between total sample size and minimum sample size according to established criteria are -100 (-216, 183), and those differences between total sample size and a more restrictive approach based on established criteria are -268 (-427, -157). The presence of external testing and the specialty of the topic were associated with sufficient sample size. CONCLUSION Radiomics studies are often designed without sample size justification, whose sample size may be too small to avoid overfitting. Sample size justification is encouraged when developing a radiomics model. KEY POINTS Question Sample size justification is critical to help minimize overfitting in developing a radiomics model, but is overlooked and underpowered in radiomics research. Findings Few of the radiomics models justified, calculated, or reported their sample size, and most of them did not meet the recent formal sample size criteria. Clinical relevance Radiomics models are often designed without sample size justification. Consequently, many models are too small to avoid overfitting. It should be encouraged to justify, perform, and report the considerations on sample size when developing radiomics models.
Collapse
Affiliation(s)
- Jingyu Zhong
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China.
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Xianwei Liu
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Junjie Lu
- Department of Epidemiology and Population Health, Stanford University School of Medicine, Stanford, CA, USA
| | - Jiarui Yang
- Department of Biomedical Engineering, Boston University, Boston, MA, USA
| | - Guangcheng Zhang
- Department of Orthopedics, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Shiqi Mao
- Department of Medical Oncology, Shanghai Pulmonary Hospital, Tongji University School of Medicine, Shanghai, China
| | - Haoda Chen
- Department of General Surgery, Pancreatic Disease Center, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qian Yin
- Department of Pathology, Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qingqing Cen
- Department of Dermatology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Run Jiang
- Department of Pharmacovigilance, SciClone Pharmaceuticals (Holdings) Ltd., Shanghai, China
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China
| | - Minda Lu
- MR Application, Siemens Healthineers Ltd., Shanghai, China
| | - Jingshen Chu
- Editorial Office of Journal of Diagnostics Concepts & Practice, Department of Science and Technology Development, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yue Xing
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yangfan Hu
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Defang Ding
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Xiang Ge
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Huan Zhang
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Weiwu Yao
- Laboratory of Key Technology and Materials in Minimally Invasive Spine Surgery, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
- Center for Spinal Minimally Invasive Research, Shanghai Jiao Tong University, Shanghai, China.
- Department of Imaging, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
21
|
Huang Y, Shi Z, Zhu T, Zhou T, Li Y, Li W, Qiu H, Wang S, He L, Wu Z, Lin Y, Wang Q, Gu W, Gu C, Song X, Zhou Y, Guan D, Wang K. Longitudinal MRI-Driven Multi-Modality Approach for Predicting Pathological Complete Response and B Cell Infiltration in Breast Cancer. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413702. [PMID: 39921294 PMCID: PMC11948082 DOI: 10.1002/advs.202413702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/25/2024] [Revised: 12/30/2024] [Indexed: 02/10/2025]
Abstract
Accurately predicting pathological complete response (pCR) to neoadjuvant treatment (NAT) in breast cancer remains challenging due to tumor heterogeneity. This study enrolled 2279 patients across 12 centers and develops a novel multi-modality model integrating longitudinal magnetic resonance imaging (MRI) spatial habitat radiomics, transcriptomics, and single-cell RNA sequencing for predicting pCR. By analyzing tumor subregions on multi-timepoint MRI, the model captures dynamic intra-tumoral heterogeneity during NAT. It shows superior performance over traditional radiomics, with areas under the curve of 0.863, 0.813, and 0.888 in the external validation, immunotherapy, and multi-omics cohorts, respectively. Subgroup analysis shows its robustness across varying molecular subtypes and clinical stages. Transcriptomic and single-cell RNA sequencing analysis reveals that high model scores correlate with increased immune activity, notably elevated B cell infiltration, indicating the biological basis of the imaging model. The integration of imaging and molecular data demonstrates promise in spatial habitat radiomics to monitor dynamic changes in tumor heterogeneity during NAT. In clinical practice, this study provides a noninvasive tool to accurately predict pCR, with the potential to guide treatment planning and improve breast-conserving surgery rates. Despite promising results, the model requires prospective validation to confirm its utility across diverse patient populations and clinical settings.
Collapse
Affiliation(s)
- Yu‐Hong Huang
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityNo. 106 Zhongshan Second Road, Yuexiu DistrictGuangzhouGuangdong Province510080China
| | - Zhen‐Yi Shi
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong Province510515China
- Guangdong Key Laboratory of Single Cell Technology and ApplicationSouthern Medical University, GuangzhouGuangdong Province510515China
| | - Teng Zhu
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityNo. 106 Zhongshan Second Road, Yuexiu DistrictGuangzhouGuangdong Province510080China
| | - Tian‐Han Zhou
- The Department of General SurgeryHangzhou TCM HospitalAffiliated to Zhejiang Chinese Medical UniversityXihu DistrictHangzhouZhejiang Province310000China
| | - Yi Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong Province510515China
- Guangdong Key Laboratory of Single Cell Technology and ApplicationSouthern Medical University, GuangzhouGuangdong Province510515China
| | - Wei Li
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityNo. 106 Zhongshan Second Road, Yuexiu DistrictGuangzhouGuangdong Province510080China
| | - Han Qiu
- Galactophore DepartmentJingzhou Hospital Affiliated to Yangtze UniversityShashi DistrictJingzhou434000China
| | - Si‐Qi Wang
- Department of BiostatisticsHarvard T.H. Chan School of Public HealthBostonMA02115USA
| | - Li‐Fang He
- Breast CenterCancer Hospital of Shantou University Medical CollegeJinping DistrictShantouGuangdong Province515000China
| | - Zhi‐Yong Wu
- Clinical research center & Breast disease diagnosis and treatment centerShantou Central HospitalNo. 114 Waima Road, Jinping DistrictShantouGuangdong Province515000China
| | - Ying Lin
- Breast Disease Center, The First Affiliated HospitalSun Yat‐sen UniversityNo. 58 Zhongshan Second Road, Yuexiu DistrictGuangzhouGuangdong Province510080China
| | - Qian Wang
- Department of RadiologyThe Affiliated Huaian No.1 People's Hospital of Nanjing Medical UniversityHuaiyin DistrictHuaianJiangsu Province223001China
| | - Wen‐Chao Gu
- Department of Artificial Intelligence MedicineGraduate School of MedicineChiba UniversityChiba263‐8522Japan
| | - Chang‐Cong Gu
- Department of Medical ImagingThe First Hospital of QinhuangdaoHaigang DistrictQinhuangdaoHebei Province066000China
| | - Xin‐Yang Song
- Department of RadiologyThe First Affiliated Hospital of Jinan UniversityNo. 613 Huangpu West Road, Tianhe DistrictGuangzhouGuangdong510627China
| | - Yang Zhou
- Department of PathologyThe Second People's Hospital of Changzhou, The Third Affiliated Hospital of Nanjing Medical UniversityNo. 29 Xinglong LaneChangzhouJiangsu Province213164China
| | - Dao‐Gang Guan
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical UniversityGuangzhouGuangdong Province510515China
- Guangdong Key Laboratory of Single Cell Technology and ApplicationSouthern Medical University, GuangzhouGuangdong Province510515China
| | - Kun Wang
- Department of Breast CancerCancer CenterGuangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences)Southern Medical UniversityNo. 106 Zhongshan Second Road, Yuexiu DistrictGuangzhouGuangdong Province510080China
| |
Collapse
|
22
|
Shi L, Li C, Bai Y, Cao Y, Zhao S, Chen X, Cheng Z, Zhang Y, Li H. CT radiomics to predict pathologic complete response after neoadjuvant immunotherapy plus chemoradiotherapy in locally advanced esophageal squamous cell carcinoma. Eur Radiol 2025; 35:1594-1604. [PMID: 39470794 DOI: 10.1007/s00330-024-11141-4] [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: 03/10/2024] [Revised: 08/26/2024] [Accepted: 09/19/2024] [Indexed: 11/01/2024]
Abstract
OBJECTIVE To develop and validate a CT-based radiomics model to predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC). METHODS A total of 105 patients with locally advanced ESCC receiving NICRT from February 2019 to December 2023 were enrolled. Patients were randomly divided into the training cohort and the test cohort at a 3:1 ratio. Enhanced CT scans were obtained before NICRT treatment. The 2D and 3D regions of interest were segmented, and features were extracted, followed by feature selection. Six algorithms were applied to construct the radiomics and clinical models. These models were evaluated by area under curve (AUC), accuracy, sensitivity, and specificity, and their respective optimal algorithms were further compared. RESULTS Forty-eight patients (45.75%) achieved pCR after NICRT. The AUC values of three algorithms in 2D radiomics models were higher than those in the 3D radiomics model and clinical model. Among these, the 2D radiomics model based on eXtreme Gradient Boosting (XGBoost) exhibited the best performance, with an AUC of 0.89 (95% CI, 0.81-0.97), accuracy of 0.85, sensitivity of 0.86, and specificity of 0.84 in the training cohort, and an AUC of 0.80 (95% CI, 0.64-0.97), accuracy of 0.77, sensitivity of 0.84, and specificity of 0.69 in the test cohort. Calibration curves also showed good agreement between predicted and actual response, and the decision curve analysis further confirmed its clinical applicability. CONCLUSION The 2D radiomics model can effectively predict pCR to NICRT in locally advanced ESCC. KEY POINTS Question Can CT-based radiomics predict pathologic complete response (pCR) after neoadjuvant immunotherapy plus chemoradiotherapy (NICRT) in locally advanced esophageal squamous cell carcinoma (ESCC)? Findings The model based on eXtreme Gradient Boosting (XGBoost) performed best, with an AUC of 0.89 in the training and 0.80 in the test cohort. Clinical relevance This CT-based radiomics model exhibits promising performance for predicting pCR to NICRT in locally advanced ESCC, which may be valuable in personalized treatment plan optimization.
Collapse
Affiliation(s)
- Liqiang Shi
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Chengqiang Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yaya Bai
- Department of Nuclear Medicine, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yuqin Cao
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Shengguang Zhao
- Department of Radiotherapy, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Xiaoyan Chen
- Department of Pathology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Zenghui Cheng
- Department of Radiology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yajie Zhang
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Hecheng Li
- Department of Thoracic Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
23
|
Feng X, Shi Y, Wu M, Cui G, Du Y, Yang J, Xu Y, Wang W, Liu F. Predicting the efficacy of neoadjuvant chemotherapy in breast cancer patients based on ultrasound longitudinal temporal depth network fusion model. Breast Cancer Res 2025; 27:30. [PMID: 40016785 PMCID: PMC11869678 DOI: 10.1186/s13058-025-01971-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 01/30/2025] [Indexed: 03/01/2025] Open
Abstract
OBJECTIVE The aim of this study was to develop and validate a deep learning radiomics (DLR) model based on longitudinal ultrasound data and clinical features to predict pathologic complete response (pCR) after neoadjuvant chemotherapy (NAC) in breast cancer patients. METHODS Between January 2018 and June 2023, 312 patients with histologically confirmed breast cancer were enrolled and randomly assigned to a training cohort (n = 219) and a test cohort (n = 93) in a 7:3 ratio. Next, pre-NAC and post-treatment 2-cycle ultrasound images were collected, and radiomics and deep learning features were extracted from NAC pre-treatment (Pre), post-treatment 2 cycle (Post), and Delta (pre-NAC-NAC 2 cycle) images. In the training cohort, to filter features, the intraclass correlation coefficient test, the Boruta algorithm, and the least absolute shrinkage and selection operator (LASSO) logistic regression were used. Single-modality models (Pre, Post, and Delta) were constructed based on five machine-learning classifiers. Finally, based on the classifier with the optimal predictive performance, the DLR model was constructed by combining Pre, Post, and Delta ultrasound features and was subsequently combined with clinical features to develop a combined model (Integrated). The discriminative power, predictive performance, and clinical utility of the models were further evaluated in the test cohort. Furthermore, patients were assigned into three subgroups, including the HR+/HER2-, HER2+, and TNBC subgroups, according to molecular typing to validate the predictability of the model across the different subgroups. RESULTS After feature screening, 16, 13, and 10 features were selected to construct the Pre model, Post model, and Delta model based on the five machine learning classifiers, respectively. The three single-modality models based on the XGBoost classifier displayed optimal predictive performance. Meanwhile, the DLR model (AUC of 0.827) was superior to the single-modality model (Pre, Post, and Delta AUCs of 0.726, 0.776, and 0.710, respectively) in terms of prediction performance. Moreover, multivariate logistic regression analysis identified Her-2 status and histological grade as independent risk factors for NAC response in breast cancer. In both the training and test cohorts, the Integrated model, which included Pre, Post, and Delta ultrasound features and clinical features, exhibited the highest predictive ability, with AUC values of 0.924 and 0.875, respectively. Likewise, the Integrated model displayed the highest predictive performance across the different subgroups. CONCLUSION The Integrated model, which incorporated pre-NAC treatment and early treatment ultrasound data and clinical features, accurately predicted pCR after NAC in breast cancer patients and provided valuable insights for personalized treatment strategies, allowing for timely adjustment of chemotherapy regimens.
Collapse
Affiliation(s)
- Xiaodan Feng
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yan Shi
- Department of Ultrasonography, Weihai Municipal Hospital, Cheeloo College of Medicine, Shandong University, Weihai, Shandong, 264200, China
| | - Meng Wu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Guanghe Cui
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yao Du
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Jie Yang
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Yuyuan Xu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Wenjuan Wang
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China
| | - Feifei Liu
- Department of Ultrasound, Binzhou Medical University Hospital, Binzhou, Shandong, 256603, China.
| |
Collapse
|
24
|
Li J, Li Z, Wang Y, Li Y, Zhang J, Li Z, Tang L. CT radiomics-based intratumoral and intertumoral heterogeneity indicators for prognosis prediction in gastric cancer patients receiving neoadjuvant chemotherapy. Eur Radiol 2025:10.1007/s00330-025-11430-6. [PMID: 39953151 DOI: 10.1007/s00330-025-11430-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Revised: 11/30/2024] [Accepted: 01/13/2025] [Indexed: 02/17/2025]
Abstract
OBJECTIVES CT-based intratumoral and intertumoral heterogeneity indicators were integrated to develop a prognostic model for locally advanced gastric cancer (LAGC) patients undergoing neoadjuvant chemotherapy (NACT). METHODS This retrospective study included 568 LAGC patients treated with NACT from two hospitals. The intratumor heterogeneity score (ITHscore) was developed to quantify the intratumoral heterogeneity of LAGCs on CT; intertumoral heterogeneity was characterized by combining the primary tumor (PT) and lymph node (LN) sizes on CT. CT indicators were measured on baseline and posttreatment CT scans; the reduction rates (%Δ) were calculated. The overall survival (OS) of all patients was recorded. Cox regression analysis was used to construct a preoperative survival prediction model (Pre-SPM) based on the baseline indicators and %Δ indicators. The predictive performance of Pre-SPM for OS was assessed. The clinicopathological data, including the ypTNM stage, were also collected to evaluate their impact on OS. RESULTS Patients with lower baseline ITHscore had better prognoses (p < 0.001). Approximately 13.01% of patients exhibited contradictory changes in PT and LN sizes. Cox regression analysis selected the baseline ITHscore, baseline PT area, %ΔPT, and %ΔLN to establish the Pre-SPM. In the external validation cohort, the c-index of Pre-SPM for predicting OS was 0.72, while the AUC for predicting 5-year OS was 0.73. After adjusting for the influence of clinicopathological features, including the ypTNM stage, Pre-SPM remained an independent prognostic factor. CONCLUSION The Pre-SPM model, combining intratumoral heterogeneity and intertumoral heterogeneity, has the potential to predict the OS of LAGC patients receiving NACT. KEY POINTS Question Increased tumor heterogeneity in LAGC affects prognosis, but effective non-invasive CT methods for assessing intratumoral and intertumoral heterogeneity are lacking. Findings ITHscore indicates intratumoral heterogeneity, while changes in PT and LN sizes reflect intertumoral heterogeneity. The Pre-SPM model, integrating both, independently predicts patient outcomes. Clinical relevance Pre-SPM enhances prognosis prediction by quantifying intratumoral and intertumoral heterogeneity, potentially guiding more personalized and effective treatment strategies for patients with LAGC.
Collapse
Affiliation(s)
- Jiazheng Li
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhenhui Li
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan Cancer Center, Kunming, China
| | - Yinkui Wang
- Department of Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China
| | - Yuzhuo Li
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing, China
| | - Jing Zhang
- MOE Key Laboratory of Bioinformatics and Bioinformatics Division of BNRIST, Department of Automation, Tsinghua University, Beijing, China.
| | - Ziyu Li
- Department of Gastrointestinal Cancer Center, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| | - Lei Tang
- Department of Radiology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
25
|
Xie H, Tan T, Li Q, Li T. Revolutionizing HER-2 assessment: multidimensional radiomics in breast cancer diagnosis. BMC Cancer 2025; 25:265. [PMID: 39953417 PMCID: PMC11829378 DOI: 10.1186/s12885-025-13549-7] [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: 11/06/2024] [Accepted: 01/17/2025] [Indexed: 02/17/2025] Open
Abstract
OBJECTIVE To explore the application value of multidimensional radiomics based on ultrasound imaging in assessing the HER-2 status of breast cancer. METHODS We retrospectively analyzed the ultrasound imaging, clinical, and laboratory data of 850 breast cancer patients from two centers. During the study, we first utilized automation technology to accurately delineate the tumor region of interest (ROI) in breast ultrasound imaging. Subsequently, the intra-tumoral ROI was automatically expanded by 1 cm and 2 cm to obtain larger areas including the peritumoral tissues, and further generated three-dimensional volumes of interest (VOI) within and around the tumor. Through the K-means clustering method, we identified the sub-regions of interest within the ROI and extracted corresponding radiomic features using the pyradiomics toolkit. Additionally, we employed an advanced Vision Transformer (VIT) model to perform deep radiomic feature extraction on the ROI. Based on feature selection, we utilized various machine learning algorithms for modeling and analysis to assess the HER-2 status of breast cancer. RESULTS After comprehensive comparison and evaluation of multiple models, we found that the diagnostic model based on multidimensional feature fusion exhibited excellent diagnostic performance in assessing the HER-2 status of breast cancer. In the training set, the model achieved an accuracy of 0.949 and an AUC value of 0.990 (95% CI: 0.986-0.995), with outstanding key performance indicators such as sensitivity, specificity, positive predictive value, negative predictive value, and F1 score. The model showed good generalization in the test set, with accuracy 0.747, AUC 0.848 (95% CI: 0.791-0.904), and sensitivity 0.911. Specificity was slightly lower, but other indicators remained high, and the F1 score was 0.703. Calibration and clinical decision curves further confirmed the model's effectiveness and reliability. CONCLUSION This study fully demonstrates that multidimensional breast ultrasonography-based radiomic features can effectively assess the HER-2 status of breast cancer. This finding not only provides new evidence for early diagnosis of breast cancer but also offers new ideas and methods for personalized treatment planning and prognosis assessment.
Collapse
Affiliation(s)
- Hui Xie
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, P. R. China
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, P. R. China
| | - Tao Tan
- Faulty of Applied Sciences, Macao Polytechnic University, Macao, 999078, P. R. China
| | - Qing Li
- Department of Radiation Oncology, Affiliated Hospital (Clinical College) of Xiangnan University, Chenzhou, 423000, P. R. China
- Key Experimental Project of Higher Education Institutes in Hunan Province (Key Laboratory of Tumor Precision Medicine), Chenzhou, 423000, P. R. China
- College of Medical Imaging, Laboratory Diagnostics, and Rehabilitation, Xiangnan University, Chenzhou, 423000, P. R. China
| | - Tao Li
- College of Medical Imaging, Laboratory Diagnostics, and Rehabilitation, Xiangnan University, Chenzhou, 423000, P. R. China.
- Department of Medical, Xiangnan University, Chenzhou, 423000, P. R. China.
| |
Collapse
|
26
|
Park JH, Kwon LM, Lee HK, Koo T, Suh YJ, Kwon MJ, Kim HY. Radiomic Analysis of Magnetic Resonance Imaging for Breast Cancer with TP53 Mutation: A Single Center Study. Diagnostics (Basel) 2025; 15:428. [PMID: 40002579 PMCID: PMC11854707 DOI: 10.3390/diagnostics15040428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 01/29/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Radiomics is a non-invasive and cost-effective method for predicting the biological characteristics of tumors. In this study, we explored the association between radiomic features derived from magnetic resonance imaging (MRI) and genetic alterations in patients with breast cancer. Methods: We reviewed electronic medical records of patients with breast cancer patients with available targeted next-generation sequencing data available between August 2018 and May 2021. Substraction imaging of T1-weighted sequences was utilized. The tumor area on MRI was segmented semi-automatically, based on a seeded region growing algorithm. Radiomic features were extracted using the open-source software 3D slicer (version 5.6.1) with PyRadiomics extension. The association between genetic alterations and radiomic features was examined. Results: In total, 166 patients were included in this study. Among the 50 panel genes analyzed, only TP53 mutations were significantly associated with radiomic features. Compared with TP53 wild-type tumors, TP53 mutations were associated with larger tumor size, advanced stage, negative hormonal receptor status, and HER2 positivity. Tumors with TP53 mutations exhibited higher values for Gray Level Non-Uniformity, Dependence Non-Uniformity, and Run Length Non-Uniformity, and lower values for Sphericity, Low Gray Level Emphasis, and Small Dependence Low Gray Level emphasis compared to TP53 wild-type tumors. Six radiomic features were selected to develop a composite radiomics score. Receiver operating characteristic curve analysis showed an area under the curve of 0.786 (95% confidence interval, 0.719-0.854; p < 0.001). Conclusions: TP53 mutations in breast cancer can be predicted using MRI-derived radiomic analysis. Further research is needed to assess whether radiomics can help guide treatment decisions in clinical practice.
Collapse
Affiliation(s)
- Jung Ho Park
- Division of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea (Y.J.S.)
| | - Lyo Min Kwon
- Department of Radiology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea;
| | - Hong Kyu Lee
- Department of Thoracic and Cardiovascular Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Taeryool Koo
- Department of Radiation Oncology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Yong Joon Suh
- Division of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea (Y.J.S.)
| | - Mi Jung Kwon
- Department of Pathology, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| | - Ho Young Kim
- Department of Internal Medicine, Hallym University Sacred Heart Hospital, Anyang 14068, Republic of Korea
| |
Collapse
|
27
|
Zhu HN, Guo YF, Lin Y, Sun ZC, Zhu X, Li Y. Radiomics analysis of thoracic vertebral bone marrow microenvironment changes before bone metastasis of breast cancer based on chest CT. J Bone Oncol 2025; 50:100653. [PMID: 39712652 PMCID: PMC11655691 DOI: 10.1016/j.jbo.2024.100653] [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: 08/30/2024] [Revised: 11/13/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
Bone metastasis from breast cancer significantly elevates patient morbidity and mortality, making early detection crucial for improving outcomes. This study utilizes radiomics to analyze changes in the thoracic vertebral bone marrow microenvironment from chest computerized tomography (CT) images prior to bone metastasis in breast cancer, and constructs a model to predict metastasis. METHODS This study retrospectively gathered data from breast cancer patients who were diagnosed and continuously monitored for five years from January 2013 to September 2023. Radiomic features were extracted from the bone marrow of thoracic vertebrae on non-contrast chest CT scans. Multiple machine learning algorithms were utilized to construct various radiomics models for predicting the risk of bone metastasis, and the model with optimal performance was integrated with clinical features to develop a nomogram. The effectiveness of this combined model was assessed through receiver operating characteristic (ROC) analysis as well as decision curve analysis (DCA). RESULTS The study included a total of 106 patients diagnosed with breast cancer, among whom 37 developed bone metastases within five years. The radiomics model's area under the curve (AUC) for the test set, calculated using logistic regression, is 0.929, demonstrating superior predictive performance compared to alternative machine learning models. Furthermore, DCA demonstrated the potential of radiomics models in clinical application, with a greater clinical benefit in predicting bone metastasis than clinical model and nomogram. CONCLUSION CT-based radiomics can capture subtle changes in the thoracic vertebral bone marrow before breast cancer bone metastasis, offering a predictive tool for early detection of bone metastasis in breast cancer.
Collapse
Affiliation(s)
- Hao-Nan Zhu
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Yi-Fan Guo
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - YingMin Lin
- The Department of Thyroid and Breast Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| | - Zhi-Chao Sun
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University, (Zhejiang Provincial Hospital of Traditional Chinese Medicine), Hangzhou, Zhejiang, China
| | - Xi Zhu
- Department of Radiology, Northern Jiangsu People’s Hospital Affiliated to Yangzhou, University, Yangzhou, Jiangsu, China
| | - YuanZhe Li
- Center of Radiology, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China
| |
Collapse
|
28
|
Wu X, Ling Y, Zhang S, Zhang B. Advancing radiomics from contrast-enhanced mammography in breast cancer. Br J Cancer 2025; 132:154-155. [PMID: 39668161 PMCID: PMC11756395 DOI: 10.1038/s41416-024-02932-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Revised: 11/27/2024] [Accepted: 12/05/2024] [Indexed: 12/14/2024] Open
Affiliation(s)
- Xuewei Wu
- Department of Radiology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Ye Ling
- Department of Radiology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Shuixing Zhang
- Department of Radiology, the First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Bin Zhang
- Department of Radiology, the First Affiliated Hospital of Jinan University, Guangzhou, China.
| |
Collapse
|
29
|
Li C, Tan J, Li H, Lei Y, Yang G, Zhang C, Song Y, Wu Y, Bi G, Bi Q. The value of multiparametric MRI-based habitat imaging for differentiating uterine sarcomas from atypical leiomyomas: a multicentre study. Abdom Radiol (NY) 2025; 50:995-1008. [PMID: 39183205 DOI: 10.1007/s00261-024-04539-7] [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: 05/10/2024] [Revised: 08/14/2024] [Accepted: 08/15/2024] [Indexed: 08/27/2024]
Abstract
PURPOSE To explore the feasibility of multiparametric MRI-based habitat imaging for distinguishing uterine sarcoma (US) from atypical leiomyoma (ALM). METHODS This retrospective study included the clinical and preoperative MRI data of 69 patients with US and 225 patients with ALM from three hospitals. At both the individual and cohort levels, the K-means and Gaussian mixture model (GMM) algorithms were utilized to perform habitat imaging on MR images, respectively. Specifically, T2-weighted images (T2WI) and contrast-enhanced T1-weighted images (CE-T1WI) were clustered to generate structural habitats, while apparent diffusion coefficient (ADC) maps and CE-T1WI were clustered to create functional habitats. Parameters of each habitat subregion were extracted to construct distinct habitat models. The integrated models were constructed by combining habitat and clinical independent predictors. Model performance was assessed using the area under the curve (AUC). RESULTS Abnormal vaginal bleeding, lactate dehydrogenase (LDH), and white blood cell (WBC) counts can serve as clinical independent predictors of US. The GMM-based functional habitat model at the cohort level had the highest mean AUC (0.766) in both the training and validation cohorts, followed by the GMM-based structural habitat model at the cohort level (AUC = 0.760). Within the integrated models, the K-means functional habitat model based on the cohort level achieved the highest mean AUC (0.905) in both the training and validation cohorts. CONCLUSION Habitat imaging based on multiparametric MRI has the potential to distinguish US from ALM. The combination of clinical independent predictors with the habitat models can effectively improve the performance.
Collapse
Affiliation(s)
- Chenrong Li
- Medical school, Kunming University of Science and Technology, The First People's Hospital of Yunnan Province, Kunming, 650500, Yunnan, China
| | - Jing Tan
- Department of Radiology, Yunnan Cancer Hospital, The Third Affiliated Hospital of Kunming Medical University,Peking University Cancer Hospital Yunnan, Kunming, 650118, Yunnan, China
| | - Haiyan Li
- Department of MRI, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, 650032, Yunnan, China
| | - Ying Lei
- Department of Radiology, School of Medicine, Zhongda Hospital, Southeast University, Nanjing, 210009, Jiangsu, China
| | - Guang Yang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200241, China
| | - Chengxiu Zhang
- Shanghai Key Laboratory of Magnetic Resonance, East China Normal University, Shanghai, 200241, China
| | - Yang Song
- MR Research Collaboration, Siemens Healthineers, Shanghai, 201318, China
| | - Yunzhu Wu
- School of Artificial Intelligence, Institute for AI in Medicine, Nanjing University of Information Science and Technology, Nanjing, 210044, China
| | - Guoli Bi
- Department of MRI, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, 650032, Yunnan, China
| | - Qiu Bi
- Department of MRI, The First People's Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, No. 157 Jinbi Road, Kunming, 650032, Yunnan, China.
| |
Collapse
|
30
|
Li L, Xu X, Wang W, Huang P, Yu L, Ren Z, Fan J, Zhou J, Zhang L, Wang Z. Safety and efficacy of PD-1 inhibitor (sintilimab) combined with transarterial chemoembolization as the initial treatment in patients with intermediate-stage hepatocellular carcinoma beyond up-to-seven criteria. J Immunother Cancer 2025; 13:e010035. [PMID: 39824532 PMCID: PMC11749212 DOI: 10.1136/jitc-2024-010035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 01/06/2025] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND Numerous studies have demonstrated limited survival benefits of transarterial chemoembolization (TACE) alone in the treatment of intermediate-stage hepatocellular carcinoma (HCC) beyond up-to-seven criteria. The advent of immunotherapy, particularly immune checkpoint inhibitors (ICIs), has opened new avenues for HCC treatment. However, TACE combined with ICIs has not been investigated for patients with intermediate-stage HCC beyond the up-to-seven criteria. The study aims to evaluate the efficacy and safety of this treatment strategy for such patients. METHODS In this single-arm, prospective, phase II study, we enrolled eligible patients with HCC who were treated with TACE plus programmed cell death protein 1 (PD-1) inhibitors (sintilimab) from April 2021 to February 2023. The study's primary objectives were to assess progression-free survival (PFS) and safety. Secondary objectives included measuring the objective response rate (ORR) and disease control rate (DCR) as per both Response Evaluation Criteria in Solid Tumors (RECIST) V.1.1 and modified RECIST (mRECIST) criteria, as well as overall survival (OS). Additionally, we conducted correlation analyses to identify predictors influencing the efficacy of tumor treatment. RESULT 20 patients participated in this study, with a median follow-up duration of 22.0 months. Median PFS was 8.4 months (95% CI: 4.7 to 19.7) according to both RECIST V.1.1 and mRECIST. The ORR was 30.0% (95% CI: 14.6% to 51.9%) per RECIST 1.1% and 60% (95% CI: 38.7% to 78.1%) per mRECIST. DCR was 95.0% (95% CI: 76.4% to 99.1%) according to both RECIST V.1.1 and mRECIST. Median OS was not yet reached. Notably, 20% (4/20) of patients underwent successful conversion to curative surgical resection. Treatment-related adverse events (TRAEs) mainly included elevated aspartate aminotransferase levels (19/20, 95.0%), elevated alanine aminotransferase levels (18/20, 90.0%), hypothyroidism (18/20, 90.0%), and reduced appetite (10/20, 50.0%). Among all participants, only one experienced grade 3 TRAE (myocarditis). We employed the Elastic Net regression model to analyze radiomic features from tumor and peritumoral areas to predict the efficacy of this treatment strategy. CONCLUSION TACE plus PD-1 inhibitors demonstrated promising efficacy and an acceptable safety profile, suggesting it as a potential treatment option for patients with intermediate-stage HCC beyond up-to-seven criteria. Furthermore, our study indicates that specific image-based features may serve as predictors for patients likely to benefit from this treatment approach. TRIAL REGISTRATION NUMBER NCT04842565.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/mortality
- Male
- Liver Neoplasms/therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/drug therapy
- Liver Neoplasms/mortality
- Female
- Chemoembolization, Therapeutic/methods
- Middle Aged
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/pharmacology
- Antibodies, Monoclonal, Humanized/adverse effects
- Aged
- Immune Checkpoint Inhibitors/therapeutic use
- Immune Checkpoint Inhibitors/adverse effects
- Immune Checkpoint Inhibitors/pharmacology
- Prospective Studies
- Programmed Cell Death 1 Receptor/antagonists & inhibitors
- Neoplasm Staging
- Adult
- Treatment Outcome
Collapse
Affiliation(s)
- Lixing Li
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xin Xu
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wentao Wang
- Department of Radiology, Shanghai Institute of Medical Imaging, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Peiran Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lei Yu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zhenggang Ren
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
- Institute of Biomedical Sciences, Fudan University, Shanghai, China
- State Key Laboratory of Genetic Engineering, Fudan University, Shanghai, China
| | - Lan Zhang
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Hepatic Oncology, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Zheng Wang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Shanghai, China
- Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Zhongshan Hospital, Fudan University, Shanghai, China
- Shanghai Key Laboratory of Organ Transplantation, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
31
|
Li B, Yu Y, Xia T. Editorial for "Intertumoral Heterogeneity Based on MRI Radiomic Features Estimates Recurrence in Hepatocellular Carcinoma". J Magn Reson Imaging 2025; 61:182-183. [PMID: 38712658 DOI: 10.1002/jmri.29433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Accepted: 04/01/2024] [Indexed: 05/08/2024] Open
Affiliation(s)
- Binrong Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yaoyao Yu
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tianyi Xia
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
32
|
Liu J, Li X, Wang G, Zeng W, Zeng H, Wen C, Xu W, He Z, Qin G, Chen W. Time-Series MR Images Identifying Complete Response to Neoadjuvant Chemotherapy in Breast Cancer Using a Deep Learning Approach. J Magn Reson Imaging 2025; 61:184-197. [PMID: 38850180 DOI: 10.1002/jmri.29405] [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: 01/04/2024] [Revised: 04/09/2024] [Accepted: 04/10/2024] [Indexed: 06/10/2024] Open
Abstract
BACKGROUND Pathological complete response (pCR) is an essential criterion for adjusting follow-up treatment plans for patients with breast cancer (BC). The value of the visual geometry group and long short-term memory (VGG-LSTM) network using time-series dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) for pCR identification in BC is unclear. PURPOSE To identify pCR to neoadjuvant chemotherapy (NAC) using deep learning (DL) models based on the VGG-LSTM network. STUDY TYPE Retrospective. POPULATION Center A: 235 patients (47.7 ± 10.0 years) were divided 7:3 into training (n = 164) and validation set (n = 71). Center B: 150 patients (48.5 ± 10.4 years) were used as test set. FIELD STRENGTH/SEQUENCE 3-T, T2-weighted spin-echo sequence imaging, and gradient echo DCE sequence imaging. ASSESSMENT Patients underwent MRI examinations at three sequential time points: pretreatment, after three cycles of treatment, and prior to surgery, with tumor regions of interest manually delineated. Histopathology was the gold standard. We used VGG-LSTM network to establish seven DL models using time-series DCE-MR images: pre-NAC images (t0 model), early NAC images (t1 model), post-NAC images (t2 model), pre-NAC and early NAC images (t0 + t1 model), pre-NAC and post-NAC images (t0 + t2 model), pre-NAC, early NAC and post-NAC images (t0 + t1 + t2 model), and the optimal model combined with the clinical features and imaging features (combined model). The models were trained and optimized on the training and validation set, and tested on the test set. STATISTICAL TESTS The DeLong, Student's t-test, Mann-Whitney U, Chi-squared, Fisher's exact, Hosmer-Lemeshow tests, decision curve analysis, and receiver operating characteristics analysis were performed. P < 0.05 was considered significant. RESULTS Compared with the other six models, the combined model achieved the best performance in the test set yielding an AUC of 0.927. DATA CONCLUSION The combined model that used time-series DCE-MR images, clinical features and imaging features shows promise for identifying pCR in BC. LEVEL OF EVIDENCE: 4 TECHNICAL EFFICACY Stage 4.
Collapse
Affiliation(s)
- Jialing Liu
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Xu Li
- Department of Radiotherapy, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, Guangdong Province, China
| | - Gang Wang
- Department of Radiology, The Tenth Affiliated Hospital, Southern Medical University (Dongguan People's Hospital), Dongguan, Guangdong Province, China
| | - Weixiong Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Hui Zeng
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Chanjuan Wen
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Weimin Xu
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Zilong He
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Genggeng Qin
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| | - Weiguo Chen
- Department of Radiology, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong Province, China
| |
Collapse
|
33
|
Luo T, Yan M, Zhou M, Dekker A, Appelt AL, Ji Y, Zhu J, de Ruysscher D, Wee L, Zhao L, Zhang Z. Improved prognostication of overall survival after radiotherapy in lung cancer patients by an interpretable machine learning model integrating lung and tumor radiomics and clinical parameters. LA RADIOLOGIA MEDICA 2025; 130:96-109. [PMID: 39542968 DOI: 10.1007/s11547-024-01919-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2024] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Accurate prognostication of overall survival (OS) for non-small cell lung cancer (NSCLC) patients receiving definitive radiotherapy (RT) is crucial for developing personalized treatment strategies. This study aims to construct an interpretable prognostic model that combines radiomic features extracted from normal lung and from primary tumor with clinical parameters. Our model aimed to clarify the complex, nonlinear interactions between these variables and enhance prognostic accuracy. METHODS We included 661 stage III NSCLC patients from three multi-national datasets: a training set (N = 349), test-set-1 (N = 229), and test-set-2 (N = 83), all undergoing definitive RT. A total of 104 distinct radiomic features were separately extracted from the regions of interest in the lung and the tumor. We developed four predictive models using eXtreme gradient boosting and selected the top 10 features based on the Shapley additive explanations (SHAP) values. These models were the tumor radiomic model (Model-T), lung radiomic model (Model-L), a combined radiomic model (Model-LT), and an integrated model incorporating clinical parameters (Model-LTC). Model performance was evaluated through Harrell's concordance index, Kaplan-Meier survival curves, time-dependent area under the receiver operating characteristic curve (AUC), calibration curves, and decision curve analysis. Interpretability was assessed using the SHAP framework. RESULTS Model-LTC exhibited superior performance, with notable predictive accuracy (C-index: training set, 0.87; test-set-2, 0.76) and time-dependent AUC above 0.75. Complex nonlinear relationships and interactions were evident among the model's variables. CONCLUSION The integration of radiomic and clinical factors within an interpretable framework significantly improved OS prediction. The SHAP analysis provided insightful interpretability, enhancing the model's clinical applicability and potential for aiding personalized treatment decisions.
Collapse
Affiliation(s)
- Tianchen Luo
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
- Institute of System Science, National University of Singapore, Singapore, 119260, Singapore
| | - Meng Yan
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Meng Zhou
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Ane L Appelt
- Leeds Institute of Medical Research at St James's, University of Leeds, and Leeds Cancer Centre, St James's University Hospital, Leeds, UK
| | - Yongling Ji
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Ji Zhu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Dirk de Ruysscher
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Leonard Wee
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lujun Zhao
- Department of Radiation Oncology, Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Tianjin, 300060, China.
| | - Zhen Zhang
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China.
- Department of Radiation Oncology (Maastro), GROW Research Institute for Oncology and Reproduction, Maastricht University Medical Centre+, Maastricht, The Netherlands.
| |
Collapse
|
34
|
Li Y, Li W, Xiao H, Chen W, Lu J, Huang N, Li Q, Zhou K, Kojima I, Liu Y, Ou Y. Automated classification of pathological differentiation in head and neck squamous cell carcinoma using combined radiomics models from CET1WI and T2WI. Clin Oral Investig 2024; 29:25. [PMID: 39708187 DOI: 10.1007/s00784-024-06110-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 12/12/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVES This study aims to develop an automated radiomics-based model to grade the pathological differentiation of head and neck squamous cell carcinoma (HNSCC) and to assess the influence of various magnetic resonance imaging (MRI) sequences on the model's performance. MATERIALS AND METHODS We retrospectively analyzed MRI data from 256 patients across two medical centers, including both contrast-enhanced T1-weighted images (CET1WI) and T2-weighted images (T2WI). Regions of interest were delineated for radiomics feature extraction, followed by dimensionality reduction. An XGBoost classifier was then employed to build the predictive model, with its classification efficiency assessed using receiver operating characteristic curves and the area under the curve (AUC). RESULTS In validation cohort, the AUC (macro/micro) values for models utilizing CET1WI, T2WI, and the combination of CET1WI and T2WI were 0.801/0.814, 0.741/0.798, and 0.885/0.895, respectively. The AUC for the three differentiations, ranging from well-differentiated to poorly differentiated, were 0.867, 0.909, and 0.837, respectively. The macro/micro precision, recall, and F1 scores of 0.688/0.736, 0.744/0.828, and 0.685/0.779 for the CET1WI + T2WI model. CONCLUSION This study demonstrates that constructing a radiomics model based on CET1WI and T2WI sequences can be used to predict the pathological differentiation grading of HNSCC patients. CLINICAL RELEVANCE This study suggests that a radiomics model integrating CET1WI and T2WI MRI sequences can effectively predict the pathological differentiation of HNSCC, providing an alternative diagnostic approach through non-invasive preoperative methods.
Collapse
Affiliation(s)
- Yang Li
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Wen Li
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Haotian Xiao
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Weizhong Chen
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Jie Lu
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Nengwen Huang
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China
| | - Qingling Li
- Department of Periodontology and Endodontology, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Kangwei Zhou
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Ikuho Kojima
- Department of Oral Diagnosis, Tohoku University Graduate School of Dentistry, Sendai, Japan
| | - Yiming Liu
- Department of Oral and Maxillofacial Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yanjing Ou
- Institute of Stomatology & Research Center of Dental and Craniofacial Implants, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, China.
| |
Collapse
|
35
|
Wu Y, Li Y, Chen B, Zhang Y, Xing W, Guo B, Wang W. 18F-FDG PET/CT for early prediction of pathological complete response in breast cancer neoadjuvant therapy: a retrospective analysis. Oncologist 2024; 29:e1646-e1655. [PMID: 39045652 PMCID: PMC11630790 DOI: 10.1093/oncolo/oyae185] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 05/23/2024] [Indexed: 07/25/2024] Open
Abstract
BACKGROUND Neoadjuvant treatment has been developed as a systematic approach for patients with early breast cancer and has resulted in improved breast-conserving rate and survival. However, identifying treatment-sensitive patients at the early phase of therapy remains a problem, hampering disease management and raising the possibility of disease progression during treatment. METHODS In this retrospective analysis, we collected 2-deoxy-2-[F-18] fluoro-d-glucose (18F-FDG) positron emission tomography (PET)/computed tomography (CT) images of primary tumor sites and axillary areas and reciprocal clinical pathological data from 121 patients who underwent neoadjuvant treatment and surgery in our center. The univariate and multivariate logistic regression analyses were performed to investigate features associated with pathological complete response (pCR). An 18F-FDG PET/CT-based prediction model was trained, and the performance was evaluated by receiver operating characteristic curves (ROC). RESULTS The maximum standard uptake values (SUVmax) of 18F-FDG PET/CT were a powerful indicator of tumor status. The SUVmax values of axillary areas were closely related to metastatic lymph node counts (R = 0.62). Moreover, the early SUVmax reduction rates (between baseline and second cycle of neoadjuvant treatment) were statistically different between pCR and non-pCR patients. The early SUVmax reduction rates-based model showed great ability to predict pCR (AUC = 0.89), with all molecular subtypes (HR+HER2-, HR+HER2+, HR-HER2+, and HR-HER2-) considered. CONCLUSION Our research proved that the SUVmax reduction rate of 18F-FDG PET/CT contributed to the early prediction of pCR, providing rationales for utilizing PET/CT in NAT in the future.
Collapse
Affiliation(s)
- Yilin Wu
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People’s Republic of China
| | - Yanling Li
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Bin Chen
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People’s Republic of China
| | - Ying Zhang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People’s Republic of China
| | - Wanying Xing
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People’s Republic of China
| | - Baoliang Guo
- Department of General Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin 150001, People’s Republic of China
| | - Wan Wang
- Department of Breast Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, People’s Republic of China
| |
Collapse
|
36
|
Zhu Y, Zheng D, Xu S, Chen J, Wen L, Zhang Z, Ruan H. Intratumoral habitat radiomics based on magnetic resonance imaging for preoperative prediction treatment response to neoadjuvant chemotherapy in nasopharyngeal carcinoma. Jpn J Radiol 2024; 42:1413-1424. [PMID: 39162780 DOI: 10.1007/s11604-024-01639-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 07/27/2024] [Indexed: 08/21/2024]
Abstract
PURPOSE The aim of this study is to determine intratumoral habitat regions from multi-sequences magnetic resonance imaging (MRI) and to assess the value of those regions for prediction of patient response to neoadjuvant chemotherapy (NAC) in nasopharyngeal carcinoma (NPC). MATERIALS AND METHODS Two hundred and ninety seven patients with NPC were enrolled. Multi-sequences MRI data were used to outline three-dimensional volumes of interest (VOI) of the whole tumor. The original imaging data were divided into two groups, which were resampled to an isotropic resolution of 1 × 1 × 1 mm3 (group_1mm) and 3 × 3 × 3 mm3 (group_3mm). Nineteen radiomics features were computed for each voxel of three sequences in group_3mm, within the tumor region to extract local information. Then, k-means clustering was implemented to segment the whole tumor regions in two groups. After radiomics features were extracted and dimension reduction, habitat models were built using Multi-Layer Perceptron (MLP) algorithm. RESULTS Only T stage was included as the clinical model. The habitat3mm model, which included 10 radiomics features, achieved AUCs of 0.752 and 0.724 in the training and validation cohorts, respectively. Given the slightly better outcome of habitat3mm model, nomogram was developed in combination with habitat3mm model and T stage with the AUC of 0.749 and 0.738 in the training and validation cohorts. The decision curve analysis provides further evidence of the nomogram's clinical practicality. CONCLUSIONS A nomogram based on intratumoral habitat predicts the efficacy of NAC in NPC patients, offering the potential to improve both the treatment plan and patient outcomes.
Collapse
Affiliation(s)
- Yuemin Zhu
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| | - Dechun Zheng
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China.
| | - Shugui Xu
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| | - Jianwei Chen
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| | - Liting Wen
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| | - Zhichao Zhang
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| | - Huiping Ruan
- Department of Radiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, No. 420, Fuma Road, Jin'an District, Fuzhou, 350014, Fujian, People's Republic of China
| |
Collapse
|
37
|
Chen H, Liu Y, Zhao J, Jia X, Chai F, Peng Y, Hong N, Wang S, Wang Y. Quantification of intratumoral heterogeneity using habitat-based MRI radiomics to identify HER2-positive, -low and -zero breast cancers: a multicenter study. Breast Cancer Res 2024; 26:160. [PMID: 39578913 PMCID: PMC11583526 DOI: 10.1186/s13058-024-01921-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2-targeted (HER2) therapy with antibody-drug conjugates has proven effective for patients with HER2-low breast cancer. However, intratumoral heterogeneity (ITH) poses a great challenge in identifying HER2-low tumors. ITH signatures were developed by quantifying ITH to differentiate HER2-positive, -low and -zero breast cancers. METHODS This retrospective study included 614 patients from two institutions. The study was structured into two primary tasks: task 1 was to differentiate between HER2-positive and -negative tumors, followed by task 2 to differentiate HER2-low and -zero tumors. Whole-tumor radiomics features and habitat radiomics features were extracted from MRI to construct the radiomics and ITH signatures. Multivariable logistic regression analysis was used to determine significant independent predictors. A combined model integrating significant clinicopathologic variables, radiomics signature, and ITH signature was developed for task (1) Subsequently, the better-performing model was established using the same approach for task (2) The area under the receiver operating characteristic curve (AUC) was used to assess the performance of each model. RESULTS Task 1 comprised 614 patients (training, n = 348; validation, n = 149; and test cohorts, n = 117). Task 2 encompassed 501 patients (training, n = 283; validation, n = 122; and test cohorts, n = 96). For task1, the ITH signature showed outstanding performance, achieving AUCs of 0.81, 0.81, and 0.81 in the training, validation and test cohorts, respectively. The combined model achieved improved performance, with AUCs of 0.83, 0.84 and 0.83 across the three cohorts, respectively. For task2, the ITH signature maintained superior performance, with AUCs of 0.94, 0.93 and 0.84 across the training, validation and test cohorts, respectively. Multivariable logistic regression analysis indicated that none of the clinicopathologic characteristics were retained as predictors associated with odds of HER2-low tumors. CONCLUSIONS Our study developed ITH signatures that quantified ITH using habitat-based MRI radiomics, achieving outstanding performance in differentiating HER2-postive and -negative tumors, and further differentiating HER2-low and -zero breast cancers.
Collapse
Affiliation(s)
- Haoquan Chen
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
| | - Yulu Liu
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
- Department of Radiology, The Affiliated Hospital of Southwest Jiaotong University/ The Third People's Hospital of Chengdu, Chengdu, 610031, China
| | - Jiaqi Zhao
- Department of Radiology, Jiangmen Central Hospital, Jiangmen, 529030, China
| | - Xiaoxuan Jia
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
| | - Fan Chai
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
| | - Yuan Peng
- Department of Breast Surgery, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
| | - Nan Hong
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China
| | - Shu Wang
- Department of Breast Surgery, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China.
| | - Yi Wang
- Department of Radiology, Peking University People's Hospital, No. 11, Xizhimen South St, Beijing, 100044, China.
| |
Collapse
|
38
|
Wang S, Wang T, Guo S, Zhu S, Chen R, Zheng J, Jiang T, Li R, Li J, Li J, Shen X, Qian M, Yang M, Yu S, You C, Gu Y. Whole tumour- and subregion-based radiomics of contrast-enhanced mammography in differentiating HER2 expression status of invasive breast cancers: A double-centre pilot study. Br J Cancer 2024; 131:1613-1622. [PMID: 39379571 PMCID: PMC11554679 DOI: 10.1038/s41416-024-02871-9] [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: 04/21/2024] [Revised: 09/23/2024] [Accepted: 09/27/2024] [Indexed: 10/10/2024] Open
Abstract
OBJECTIVES To explore the value of whole tumour- and subregion-based radiomics of contrast-enhanced mammography (CEM) in differentiating the HER2 expression status of breast cancers. METHODS 352 patients underwent preoperative CEM from two centres were consecutively enroled and divided into the training, internal validation, and external validation cohorts. The lesions were divided into HER2-positive and HER2-negative groups. Besides the radiological features, radiomics features capturing the whole tumour-based (wITH) and subregion-based intratumoral heterogeneity (sITH) were extracted from the craniocaudal view of CEM recombined images. The XGBoost classifier was applied to develop the radiological, sITH, and wITH models. A combined model was constructed by fusing the prediction results of the three models. RESULTS The mean age of the patients was 51.1 ± 10.7 years. Two radiological features, four wITH features, and three sITH features were selected to establish the models. The combined model significantly improved the AUC to 0.80 ± 0.03 (95% CI: 0.73-0.86), 0.79 ± 0.06 (95% CI: 0.67-0.90), and 0.79 ± 0.05 (95% CI: 0.69-0.89) in the training, internal validation, and external validation cohorts, respectively (All P < 0.05). The combined model showed good agreement between the predicted and observed probabilities and favourable net clinical benefit in the validation cohorts. CONCLUSIONS Both whole tumour- and subregion-based ITH radiomics features of CEM exhibited potential for differentiating the HER2 expression status. Combining conventional radiological features and ITH features can improve the model's performance.
Collapse
Affiliation(s)
- Simin Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting Wang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Sailing Guo
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Shuangshuang Zhu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Ruchuan Chen
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Jinlong Zheng
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Tingting Jiang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ruimin Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jinhui Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Shanghai Institute of Medical Imaging, Fudan University, Shanghai, China
| | - Jiawei Li
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Xigang Shen
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Min Qian
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Meng Yang
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Shengnan Yu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou, Jiangsu, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.
| |
Collapse
|
39
|
Yu S, Yang Y, Wang Z, Zheng H, Cui J, Zhan Y, Liu J, Li P, Fan Y, Jia W, Wang M, Chen B, Tao J, Li Y, Zhang X. CT-based conventional radiomics and quantification of intratumoral heterogeneity for predicting benign and malignant renal lesions. Cancer Imaging 2024; 24:130. [PMID: 39358821 PMCID: PMC11446113 DOI: 10.1186/s40644-024-00775-8] [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: 11/10/2023] [Accepted: 09/16/2024] [Indexed: 10/04/2024] Open
Abstract
BACKGROUND With the increasing incidence of renal lesions, pretreatment differentiation between benign and malignant lesions is crucial for optimized management. This study aimed to develop a machine learning model utilizing radiomic features extracted from various regions of interest (ROIs), intratumoral ecological diversity features, and clinical factors to classify renal lesions. METHODS CT images (arterial phase) of 1,795 renal lesions with confirmed pathology from three hospital sites were split into development (1184, 66%) and test (611, 34%) cohorts by surgery date. Conventional radiomic features were extracted from eight ROIs of arterial phase images. Intratumoral ecological diversity features were derived from intratumoral subregions. The combined model incorporating these features with clinical factors was developed, and its performance was compared with radiologists' interpretation. RESULTS Combining intratumoral and peritumoral radiomic features, along with ecological diversity features yielded the highest AUC of 0.929 among all combinations of features extracted from CT scans. After incorporating clinical factors into the features extracted from CT images, our combined model outperformed the interpretation of radiologists in the whole (AUC = 0.946 vs 0.823, P < 0.001) and small renal lesion (AUC = 0.935 vs 0.745, P < 0.001) test cohorts. Furthermore, the combined model exhibited favorable concordance and provided the highest net benefit across threshold probabilities exceeding 60%. In the whole and small renal lesion test cohorts, the AUCs for subgroups with predicted risk below or above 95% sensitivity and specificity cutoffs were 0.974 and 0.978, respectively. CONCLUSIONS The combined model, incorporating intratumoral and peritumoral radiomic features, ecological diversity features, and clinical factors showed good performance for distinguishing benign from malignant renal lesions, surpassing radiologists' diagnoses in both whole and small renal lesions. It has the potential to save patients from unnecessary invasive biopsies/surgeries and to enhance clinical decision-making.
Collapse
Affiliation(s)
- Shuanbao Yu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yang Yang
- Department of Information Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zeyuan Wang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Haoke Zheng
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jinshan Cui
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yonghao Zhan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Junxiao Liu
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Peng Li
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yafeng Fan
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Wendong Jia
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Meng Wang
- Department of Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Bo Chen
- Department of Urology, Tongliao Clinical College, Inner Mongolia Medical University, Tongliao, China
| | - Jin Tao
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yuhong Li
- Department of Information Management, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuepei Zhang
- Department of Urology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| |
Collapse
|
40
|
Wang X, Ba R, Huang Y, Cao Y, Chen H, Xu H, Shen H, Liu D, Huang H, Yin T, Wu D, Zhang J. Time-Dependent Diffusion MRI Helps Predict Molecular Subtypes and Treatment Response to Neoadjuvant Chemotherapy in Breast Cancer. Radiology 2024; 313:e240288. [PMID: 39436292 DOI: 10.1148/radiol.240288] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2024]
Abstract
Background Time-dependent diffusion MRI has the potential to help characterize tumor cell properties; however, to the knowledge of the authors, its usefulness for breast cancer diagnosis and prognostic evaluation is unknown. Purpose To investigate the clinical value of time-dependent diffusion MRI-based microstructural mapping for noninvasive prediction of molecular subtypes and pathologic complete response (pCR) in participants with breast cancer. Materials and Methods Participants with invasive breast cancer who underwent pretreatment with time-dependent diffusion MRI between February 2021 and May 2023 were prospectively enrolled. Four microstructural parameters were estimated using the IMPULSED method (a form of time-dependent diffusion MRI), along with three apparent diffusion coefficient (ADC) measurements and a relative ADC diffusion-weighted imaging parameter. Multivariable logistic regression analysis was used to identify parameters associated with each molecular subtype and pCR. A predictive model based on associated parameters was constructed, and its performance was assessed using the area under the receiver operating characteristic curve (AUC) and compared by using the DeLong test. The time-dependent diffusion MRI parameters were validated based on correlation with pathologic measurements. Results The analysis included 408 participants with breast cancer (mean age, 51.9 years ± 9.1 [SD]). Of these, 221 participants were administered neoadjuvant chemotherapy and 54 (24.4%) achieved pCR. The time-dependent diffusion MRI parameters showed reasonable performance in helping to identify luminal A (AUC, 0.70), luminal B (AUC, 0.78), and triple-negative breast cancer (AUC, 0.72) subtypes and high performance for human epidermal growth factor receptor 2 (HER2)-enriched breast cancer (AUC, 0.85), outperforming ADC measurements (all P < .05). Progesterone receptor status (odds ratio [OR], 0.08; P = .02), HER2 status (OR, 3.36; P = .009), and the cellularity index (OR, 0.01; P = .02) were independently associated with the odds of achieving pCR. The combined model showed high performance for predicting pCR (AUC, 0.88), outperforming ADC measurements and the clinical-pathologic model (AUC, 0.73 and 0.79, respectively; P < .001). The time-dependent diffusion MRI-estimated parameters correlated well with the pathologic measurements (n = 100; r = 0.67-0.81; P < .001). Conclusion Time-dependent diffusion MRI-based microstructural mapping was an effective method for helping to predict molecular subtypes and pCR to neoadjuvant chemotherapy in participants with breast cancer. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Partridge and Xu in this issue.
Collapse
Affiliation(s)
- Xiaoxia Wang
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Ruicheng Ba
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Yao Huang
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Ying Cao
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Huifang Chen
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Hanshan Xu
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Hesong Shen
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Daihong Liu
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Haiping Huang
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Ting Yin
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Dan Wu
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| | - Jiuquan Zhang
- From the Department of Radiology, Chongqing University Cancer Hospital, Chongqing Key Laboratory for Intelligent Oncology in Breast Cancer (iCQBC), No. 181 Hanyu Road, Shapingba, Chongqing 400030, China (X.W., H.C., H.X., H.S., D.L., J.Z.); Department of Biomedical Engineering, Key Laboratory for Biomedical Engineering of Ministry of Education, Zhejiang University College of Biomedical Engineering and Instrument Science, Hangzhou China (R.B., D.W.); Chongqing University School of Medicine, Chongqing, China (Y.H., Y.C.); Department of Pathology, Chongqing University Cancer Hospital, Chongqing, China (H.H.); and MR Collaborations, Siemens Healthineers, Chengdu, China (T.Y.)
| |
Collapse
|
41
|
Shang Y, Zeng Y, Luo S, Wang Y, Yao J, Li M, Li X, Kui X, Wu H, Fan K, Li ZC, Zheng H, Li G, Liu J, Zhao W. Habitat Imaging With Tumoral and Peritumoral Radiomics for Prediction of Lung Adenocarcinoma Invasiveness on Preoperative Chest CT: A Multicenter Study. AJR Am J Roentgenol 2024; 223:e2431675. [PMID: 39140631 DOI: 10.2214/ajr.24.31675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2024]
Abstract
BACKGROUND. Tumor growth processes result in spatial heterogeneity, with the development of tumor subregions (i.e., habitats) having unique biologic characteristics. OBJECTIVE. The purpose of our study was to develop and validate a habitat model combining tumor and peritumoral radiomic features on chest CT for predicting invasiveness of lung adenocarcinoma. METHODS. This retrospective study included 1156 patients (mean age, 57.5 years; 464 men, 692 women), from three centers and a public dataset, who underwent chest CT before lung adenocarcinoma resection (variable date ranges across datasets). Patients from one center formed training (n = 500) and validation (n = 215) sets; patients from the other sources formed three external test sets (n = 249, 113, 79). For each patient, a single nodule was manually segmented on chest CT. The nodule segmentation was combined with an automatically generated 4-mm peritumoral region into a whole-volume volume of interest (VOI). A gaussian mixture model (GMM) identified voxel clusters with similar first-order energy across patients. GMM results were used to divide each patient's whole-volume VOI into multiple habitats, which were defined consistently across patients. Radiomic features were extracted from each habitat. After feature selection, a habitat model was developed for predicting invasiveness, with the use of pathologic assessment as a reference. An integrated model was constructed, combining features extracted from habitats and whole-volume VOIs. Model performance was evaluated, including in subgroups based on nodule density (pure ground-glass, part-solid, and solid). The code for habitat imaging and model construction is publicly available (https://github.com/Shangyoulan/Habitat/). RESULTS. Invasive cancer was diagnosed in 626 of 1156 patients. GMM identified four as the optimal number of voxel clusters and thus of per-patient tumor habitats. The habitat model had an AUC of 0.932 in the validation set and 0.881, 0.880, and 0.764 in the three external test sets. The integrated model had an AUC of 0.947 in the validation set and 0.936, 0.908, and 0.800 in the three external test sets. In the three external test sets combined, across nodule densities, AUCs for the habitat model were 0.836-0.869 and for the integrated model were 0.846-0.917. CONCLUSION. Habitat imaging combining tumoral and peritumoral radiomic features could help predict lung adenocarcinoma invasiveness. Prediction is improved when combining information on tumor subregions and the tumor overall. CLINICAL IMPACT. The findings may aid personalized preoperative assessments to guide clinical decision-making in lung adenocarcinoma.
Collapse
Affiliation(s)
- Youlan Shang
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| | - Ying Zeng
- Department of Radiology, Xiangtan Central Hospital, Xiangtan City, China
| | - Shiwei Luo
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| | - Yisong Wang
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| | - Jiaqi Yao
- Imaging Center, The Second Affiliated Hospital of Xinjiang Medical University, Urumuqi, China
| | - Ming Li
- Department of Radiology, Huadong Hospital Affiliated to Fudan University, Shanghai, China
| | - Xiaoying Li
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| | - Xiaoyan Kui
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Hao Wu
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Kangxu Fan
- School of Computer Science and Engineering, Central South University, Changsha, China
| | - Zhi-Cheng Li
- The Key Laboratory of Biomedical Imaging Science and System, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Hairong Zheng
- Paul C. Lauterbur Research Center for Biomedical Imaging, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Ge Li
- Department of Radiology, Xiangya Hospital, Central South University, Changsha, China
| | - Jun Liu
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| | - Wei Zhao
- Department of Radiology, The Second Xiangya Hospital, Central South University, N o. 139 Middle Remin Rd, Changsha 410011, China
| |
Collapse
|
42
|
Lee HJ, Lee JH, Lee JE, Na YM, Park MH, Lee JS, Lim HS. Prediction of early clinical response to neoadjuvant chemotherapy in Triple-negative breast cancer: Incorporating Radiomics through breast MRI. Sci Rep 2024; 14:21691. [PMID: 39289507 PMCID: PMC11408492 DOI: 10.1038/s41598-024-72581-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Accepted: 09/09/2024] [Indexed: 09/19/2024] Open
Abstract
This study assessed pretreatment breast MRI coupled with machine learning for predicting early clinical responses to neoadjuvant chemotherapy (NAC) in triple-negative breast cancer (TNBC), focusing on identifying non-responders. A retrospective analysis of 135 TNBC patients (107 responders, 28 non-responders) treated with NAC from January 2015 to October 2022 was conducted. Non-responders were defined according to RECIST guidelines. Data included clinicopathologic factors and clinical MRI findings, with radiomics features from contrast-enhanced T1-weighted images, to train a stacking ensemble of 13 machine learning models. For subgroup analysis, propensity score matching was conducted to adjust for clinical disparities in NAC response. The efficacy of the models was evaluated using the area under the receiver-operating-characteristic curve (AUROC) before and after matching. The model combining clinicopathologic factors and clinical MRI findings achieved an AUROC of 0.752 (95% CI 0.644-0.860) for predicting non-responders, while radiomics-based models showed 0.749 (95% CI 0.614-0.884). An integrated model of radiomics, clinicopathologic factors, and clinical MRI findings reached an AUROC of 0.802 (95% CI 0.699-0.905). After propensity score matching, the hierarchical order of key radiomics features remained consistent. Our study demonstrated the potential of using machine learning models based on pretreatment MRI to non-invasively predict TNBC non-responders to NAC.
Collapse
Affiliation(s)
- Hyo-Jae Lee
- Department of Radiology, Chonnam National University Hospital, Gwangju, Republic of Korea
| | - Jeong Hoon Lee
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Jong Eun Lee
- Department of Radiology and the Research Institute of Radiology, Asan Medical Center, Seoul, Republic of Korea
| | - Yong Min Na
- Department of Surgery, Chonnam National University Hwasun Hospital, Hwasun, Republic of Korea
| | - Min Ho Park
- Department of Surgery, Chonnam National University Hwasun Hospital, Hwasun, Republic of Korea
- Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Ji Shin Lee
- Department of Pathology, Chonnam National University Hwasun Hospital, Hwasun, Republic of Korea
- Chonnam National University Medical School, Gwangju, Republic of Korea
| | - Hyo Soon Lim
- Department of Radiology, Chonnam National University Hwasun Hospital, Hwasun, Republic of Korea.
- Chonnam National University Medical School, Gwangju, Republic of Korea.
| |
Collapse
|
43
|
Hong M, Fan S, Xu Z, Fang Z, Ling K, Lai P, Han C, Chen Z, Hou J, Liang Y, Zhou C, Wang J, Chen X, Huang Y, Xu M. MRI radiomics and biological correlations for predicting axillary lymph node burden in early-stage breast cancer. J Transl Med 2024; 22:826. [PMID: 39243024 PMCID: PMC11378375 DOI: 10.1186/s12967-024-05619-4] [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: 04/14/2024] [Accepted: 08/15/2024] [Indexed: 09/09/2024] Open
Abstract
BACKGROUND AND AIMS Preoperative prediction of axillary lymph node (ALN) burden in patients with early-stage breast cancer is pivotal for individualised treatment. This study aimed to develop a MRI radiomics model for evaluating the ALN burden in early-stage breast cancer and to provide biological interpretability to predictions by integrating radiogenomic data. METHODS This study retrospectively analyzed 1211 patients with early-stage breast cancer from four centers, supplemented by data from The Cancer Imaging Archive (TCIA) and Duke University (DUKE). MRI radiomic features were extracted from dynamic contrast-enhanced MRI images and an ALN burden-related radscore was constructed by the backpropagation neural network algorithm. Clinical and combined models were developed, integrating ALN-related clinical variables and radscore. The Kaplan-Meier curve and log-rank test were used to assess the prognostic differences between the predicted high- and low-ALN burden groups in both Center I and DUKE cohorts. Gene set enrichment and immune infiltration analyses based on transcriptomic TCIA and TCIA Breast Cancer dataset were used to investigate the biological significance of the ALN-related radscore. RESULTS The MRI radiomics model demonstrated an area under the curve of 0.781-0.809 in three validation cohorts. The predicted high-risk population demonstrated a poorer prognosis (log-rank P < .05 in both cohorts). Radiogenomic analysis revealed migration pathway upregulation and cell differentiation pathway downregulation in the high radscore groups. Immune infiltration analysis confirmed the ability of radiological features to reflect the heterogeneity of the tumor microenvironment. CONCLUSIONS The MRI radiomics model effectively predicted the ALN burden and prognosis of early-stage breast cancer. Moreover, radiogenomic analysis revealed key cellular and immune patterns associated with the radscore.
Collapse
Affiliation(s)
- Minping Hong
- Department of Radiology, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Jiaxing, China
| | - Sijia Fan
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zeyan Xu
- Department of Radiology, The Third Affiliated Hospital of Kunming Medical University, Yunnan Cancer Hospital, Yunnan, China
| | - Zhen Fang
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Keng Ling
- Department of Clinical Laboratory, Jiaxing Maternity and Children Health Care Hospital, Jiaxing, China
| | - Penghao Lai
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Chaokang Han
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhonghua Chen
- Department of Radiology, Haining Hospital, The First Affiliated Hospital of Zhejiang University, Haining, China
| | - Jie Hou
- Department of Radiology, Zhejiang Provincial People's Hospital, Hangzhou, China
| | - Yanting Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Changyu Zhou
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China
| | - Junyan Wang
- Department of Radiology, Zhejiang Chinese Medical University Affiliated Jiaxing TCM Hospital, Jiaxing, China
| | - Xiaobo Chen
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Yanqi Huang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China.
- Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.
| | - Maosheng Xu
- Department of Radiology, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, China.
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China.
| |
Collapse
|
44
|
Rauch GM. Biomarkers for Personalized Neoadjuvant Therapy in Triple-Negative Breast Cancer: Moving Forward. Radiology 2024; 312:e242011. [PMID: 39225606 DOI: 10.1148/radiol.242011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Affiliation(s)
- Gaiane M Rauch
- From the University of Texas MD Anderson Cancer Center, 1515 Holcombe Blvd, Unit 1473, Houston, TX 77030
| |
Collapse
|
45
|
Ramtohul T, Lepagney V, Bonneau C, Jin M, Menet E, Sauge J, Laas E, Romano E, Bello-Roufai D, Mechta-Grigoriou F, Vincent Salomon A, Bidard FC, Langer A, Malhaire C, Cabel L, Brisse HJ, Tardivon A. Use of Pretreatment Perfusion MRI-based Intratumoral Heterogeneity to Predict Pathologic Response of Triple-Negative Breast Cancer to Neoadjuvant Chemoimmunotherapy. Radiology 2024; 312:e240575. [PMID: 39225608 DOI: 10.1148/radiol.240575] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/04/2024]
Abstract
Background Neoadjuvant chemoimmunotherapy (NACI) has significantly increased the rate of pathologic complete response (pCR) in patients with early-stage triple-negative breast cancer (TNBC), although predictors of response to this regimen have not been identified. Purpose To investigate pretreatment perfusion MRI-based radiomics as a predictive marker for pCR in patients with TNBC undergoing NACI. Materials and Methods This prospective study enrolled women with early-stage TNBC who underwent NACI at two different centers from August 2021 to July 2023. Pretreatment dynamic contrast-enhanced MRI scans obtained using scanners from multiple vendors were analyzed using the Tofts model to segment tumors and analyze pharmacokinetic parameters. Radiomics features were extracted from the rate constant for contrast agent plasma-to-interstitial transfer (or Ktrans), volume fraction of extravascular and extracellular space (Ve), and maximum contrast agent uptake rate (Slopemax) maps and analyzed using unsupervised correlation and least absolute shrinkage and selector operator, or LASSO, to develop a radiomics score. Score effectiveness was assessed using the area under the receiver operating characteristic curve (AUC), and multivariable logistic regression was used to develop a multimodal nomogram for enhanced prediction. The discrimination, calibration, and clinical utility of the nomogram were evaluated in an external test set. Results The training set included 112 female participants from center 1 (mean age, 52 years ± 11 [SD]), and the external test set included 83 female participants from center 2 (mean age, 47 years ± 11). The radiomics score demonstrated an AUC of 0.80 (95% CI: 0.70, 0.89) for predicting pCR. A nomogram incorporating the radiomics score, grade, and Ki-67 yielded an AUC of 0.86 (95% CI: 0.78, 0.94) in the test set. Associations were found between higher radiomics score (>0.25) and tumor size (P < .001), washout enhancement (P = .01), androgen receptor expression (P = .009), and programmed death ligand 1 expression (P = .01), demonstrating a correlation with tumor immune environment in participants with TNBC. Conclusion A radiomics score derived from pharmacokinetic parameters at pretreatment dynamic contrast-enhanced MRI exhibited good performance for predicting pCR in participants with TNBC undergoing NACI, and could potentially be used to enhance clinical decision making. © RSNA, 2024 Supplemental material is available for this article. See also the editorial by Rauch in this issue.
Collapse
Affiliation(s)
- Toulsie Ramtohul
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Victoire Lepagney
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Claire Bonneau
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Maxime Jin
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Emmanuelle Menet
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Juliette Sauge
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Enora Laas
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Emanuela Romano
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Diana Bello-Roufai
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Fatima Mechta-Grigoriou
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Anne Vincent Salomon
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - François-Clément Bidard
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Adriana Langer
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Caroline Malhaire
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Luc Cabel
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Hervé J Brisse
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| | - Anne Tardivon
- From the Department of Radiology (T.R., V.L., M.J., C.M., H.J.B., A.T.), Department of Diagnostic and Theranostic Medicine-Pathology (J.S., A.V.S.), Department of Surgical Oncology (E.L.), Department of Medical Oncology (E.R.), Stress and Cancer Laboratory (F.M.G.), and INSERM U830 (F.M.G.), Institut Curie, PSL University, 26 rue d'Ulm, 75005 Paris, France; Department of Surgical Oncology and INSERM U900, Statistical Methods for Precision Medicine, Institut Curie, University of Versailles Saint-Quentin-en-Yvelines, Saint-Cloud, France (C.B.); Departments of Diagnostic and Theranostic Medicine-Pathology (E.M.), Medical Oncology (D.B.R., F.C.B., L.C.), and Radiology (A.L.), Institut Curie, PSL University, Saint-Cloud, France; Department of Immunology, PSL University, Paris, France (E.R.); and Circulating Tumor Biomarkers Laboratory, Department of Translational Research, Institut Curie, Paris, France (F.C.B.)
| |
Collapse
|
46
|
Zhou L, Wu H, Zhou H. Correlation Between Cognitive Impairment and Lenticulostriate Arteries: A Clinical and Radiomics Analysis. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1261-1272. [PMID: 38429561 PMCID: PMC11300411 DOI: 10.1007/s10278-024-01060-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/19/2024] [Accepted: 02/19/2024] [Indexed: 03/03/2024]
Abstract
Lenticulostriate arteries (LSA) are potentially valuable for studying vascular cognitive impairment. This study aims to investigate correlations between cognitive impairment and LSA through clinical and radiomics features analysis. We retrospectively included 102 patients (mean age 62.5±10.3 years, 60 males), including 58 with mild cognitive impairment (MCI) and 44 with moderate or severe cognitive impairment (MSCI). The MRI images of these patients were subjected to z-score preprocessing, manual regions of interest (ROI) outlining, feature extraction (pyradiomics), feature selection [max-relevance and min-redundancy (mRMR), least absolute shrinkage and selection operator (LASSO), and univariate analysis], model construction (multivariate logistic regression), and evaluation [receiver operating characteristic curve (ROC), decision curve analysis (DCA), and calibration curves (CC)]. In the training dataset (71 patients, 44 MCI) and the test dataset (31 patients, 17 MCI), the area under curve (AUC) of the combined model (training 0.88 [95% CI 0.78, 0.97], test 0.76 [95% CI 0.6, 0.93]) was better than that of the clinical model and the radiomics model. The DCA results demonstrated the highest net yield of the combined model relative to the clinical and radiomics models. In addition, we found that LSA total vessel count (0.79 [95% CI 0.08, 1.59], P = 0.038) and wavelet.HLH_glcm_MCC (-1.2 [95% CI -2.2, -0.4], P = 0.008) were independent predictors of MCI. The model that combines clinical and radiomics features of LSA can predict MCI. Besides, LSA vascular parameters may serve as imaging biomarkers of cognitive impairment.
Collapse
Affiliation(s)
- Langtao Zhou
- Department of Radiology of the First Affiliated Hospital, University of South China, Hengyang, 421001, China
- School of Cyberspace Security, Guangzhou University, Guangzhou, 510006, China
| | - Huiting Wu
- Department of Radiology of the First Affiliated Hospital, University of South China, Hengyang, 421001, China.
| | - Hong Zhou
- Department of Radiology of the First Affiliated Hospital, University of South China, Hengyang, 421001, China.
| |
Collapse
|
47
|
Yang X, Niu W, Wu K, Li X, Hou H, Tan Y, Wang X, Yang G, Wang L, Zhang H. Diffusion kurtosis imaging-based habitat analysis identifies high-risk molecular subtypes and heterogeneity matching in diffuse gliomas. Ann Clin Transl Neurol 2024; 11:2073-2087. [PMID: 38887966 PMCID: PMC11330218 DOI: 10.1002/acn3.52128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 05/14/2024] [Accepted: 06/02/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVE High-risk types of diffuse gliomas in adults include isocitrate dehydrogenase (IDH) wild-type glioblastomas and grade 4 astrocytomas. Achieving noninvasive prediction of high-risk molecular subtypes of gliomas is important for personalized and precise diagnosis and treatment. METHODS We retrospectively collected data from 116 patients diagnosed with adult diffuse gliomas. Multiple high-risk molecular markers were tested, and various habitat models and whole-tumor models were constructed based on preoperative routine and diffusion kurtosis imaging (DKI) sequences to predict high-risk molecular subtypes of gliomas. Feature selection and model construction utilized Least absolute shrinkage and selection operator (LASSO) and support vector machine (SVM). Finally, the Wilcoxon rank-sum test was employed to explore the correlation between habitat quantitative features (intra-tumor heterogeneity score,ITH score) and heterogeneity, as well as high-risk molecular subtypes. RESULTS The results showed that the habitat analysis model based on DKI performed remarkably well (with AUC values reaching 0.977 and 0.902 in the training and test sets, respectively). The model's performance was further enhanced when combined with clinical variables. (The AUC values were 0.994 and 0.920, respectively.) Additionally, we found a close correlation between ITH score and heterogeneity, with statistically significant differences observed between high-risk and non-high-risk molecular subtypes. INTERPRETATION The habitat model based on DKI is an ideal means for preoperatively predicting high-risk molecular subtypes of gliomas, holding significant value for noninvasively alerting malignant gliomas and those with malignant transformation potential.
Collapse
Affiliation(s)
- Xiangli Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi HospitalTaiyuan030032China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Wenju Niu
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Kai Wu
- Department of Information ManagementFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiang Li
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
| | - Heng Hou
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Yan Tan
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Xiaochun Wang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Guoqiang Yang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| | - Lei Wang
- Beijing Tiantan HospitalCapital Medical UniversityBeijing100050China
| | - Hui Zhang
- Department of RadiologyFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- College of Medical Imaging, Shanxi Medical UniversityTaiyuan030001China
- Shanxi Key Laboratory of Intelligent Imaging and NanomedicineFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
- Intelligent Imaging Big Data and Functional Nano‐imaging Engineering Research Center of Shanxi ProvinceFirst Hospital of Shanxi Medical UniversityTaiyuan030001China
| |
Collapse
|
48
|
Zhou J, Bai Y, Zhang Y, Wang Z, Sun S, Lin L, Gu Y, You C. A preoperative radiogenomic model based on quantitative heterogeneity for predicting outcomes in triple-negative breast cancer patients who underwent neoadjuvant chemotherapy. Cancer Imaging 2024; 24:98. [PMID: 39080809 PMCID: PMC11289960 DOI: 10.1186/s40644-024-00746-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 07/24/2024] [Indexed: 08/02/2024] Open
Abstract
BACKGROUND Triple-negative breast cancer (TNBC) is highly heterogeneous, resulting in different responses to neoadjuvant chemotherapy (NAC) and prognoses among patients. This study sought to characterize the heterogeneity of TNBC on MRI and develop a radiogenomic model for predicting both pathological complete response (pCR) and prognosis. MATERIALS AND METHODS In this retrospective study, TNBC patients who underwent neoadjuvant chemotherapy at Fudan University Shanghai Cancer Center were enrolled as the radiomic development cohort (n = 315); among these patients, those whose genetic data were available were enrolled as the radiogenomic development cohort (n = 98). The study population of the two cohorts was randomly divided into a training set and a validation set at a ratio of 7:3. The external validation cohort (n = 77) included patients from the DUKE and I-SPY 1 databases. Spatial heterogeneity was characterized using features from the intratumoral subregions and peritumoral region. Hemodynamic heterogeneity was characterized by kinetic features from the tumor body. Three radiomics models were developed by logistic regression after selecting features. Model 1 included subregional and peritumoral features, Model 2 included kinetic features, and Model 3 integrated the features of Model 1 and Model 2. Two fusion models were developed by further integrating pathological and genomic features (PRM: pathology-radiomics model; GPRM: genomics-pathology-radiomics model). Model performance was assessed with the AUC and decision curve analysis. Prognostic implications were assessed with Kaplan‒Meier curves and multivariate Cox regression. RESULTS Among the radiomic models, the multiregional model representing multiscale heterogeneity (Model 3) exhibited better pCR prediction, with AUCs of 0.87, 0.79, and 0.78 in the training, internal validation, and external validation sets, respectively. The GPRM showed the best performance for predicting pCR in the training (AUC = 0.97, P = 0.015) and validation sets (AUC = 0.93, P = 0.019). Model 3, PRM and GPRM could stratify patients by disease-free survival, and a predicted nonpCR was associated with poor prognosis (P = 0.034, 0.001 and 0.019, respectively). CONCLUSION Multiscale heterogeneity characterized by DCE-MRI could effectively predict the pCR and prognosis of TNBC patients. The radiogenomic model could serve as a valuable biomarker to improve the prediction performance.
Collapse
Affiliation(s)
- Jiayin Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yansong Bai
- Institute of Science and Technology for Brain-Inspired Intelligence, Fudan University, Shanghai, 200000, China
| | - Ying Zhang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Breast Surgery, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
| | - Zezhou Wang
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
- Department of Cancer Prevention, Fudan University Shanghai Cancer Center, Shanghai, 200032, China
- Shanghai Municipal Hospital Oncological Specialist Alliance, Shanghai, 200000, China
| | - Shiyun Sun
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Luyi Lin
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China
| | - Yajia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, No. 270 Dong'an Road, Shanghai, 200032, China.
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
49
|
Cai Z, Xu Z, Chen Y, Zhang R, Guo B, Chen H, Ouyang F, Chen X, Chen X, Liu D, Luo C, Li X, Liu W, Zhou C, Guan X, Liu Z, Zhao H, Hu Q. Multiparametric MRI subregion radiomics for preoperative assessment of high-risk subregions in microsatellite instability of rectal cancer patients: a multicenter study. Int J Surg 2024; 110:4310-4319. [PMID: 38498392 PMCID: PMC11254239 DOI: 10.1097/js9.0000000000001335] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 03/04/2024] [Indexed: 03/20/2024]
Abstract
BACKGROUND Microsatellite instability (MSI) is associated with treatment response and prognosis in patients with rectal cancer (RC). However, intratumoral heterogeneity limits MSI testing in patients with RC. The authors developed a subregion radiomics model based on multiparametric MRI to preoperatively assess high-risk subregions with MSI and predict the MSI status of patients with RC. METHODS This retrospective study included 475 patients (training cohort, 382; external test cohort, 93) with RC from two participating hospitals between April 2017 and June 2023. In the training cohort, subregion radiomic features were extracted from multiparametric MRI, which included T2-weighted, T1-weighted, diffusion-weighted, and contrast-enhanced T1-weighted imaging. MSI-related subregion radiomic features, classical radiomic features, and clinicoradiological variables were gathered to build five predictive models using logistic regression. Kaplan-Meier survival analysis was conducted to explore the prognostic information. RESULTS Among the 475 patients [median age, 64 years (interquartile range, IQR: 55-70 years); 304 men and 171 women], the prevalence of MSI was 11.16% (53/475). The subregion radiomics model outperformed the classical radiomics and clinicoradiological models in both training [area under the curve (AUC)=0.86, 0.72, and 0.59, respectively] and external test cohorts (AUC=0.83, 0.73, and 0.62, respectively). The subregion-clinicoradiological model combining clinicoradiological variables and subregion radiomic features performed the optimal, with AUCs of 0.87 and 0.85 in the training and external test cohorts, respectively. The 3-year disease-free survival rate of MSI groups predicted based on the model was higher than that of the predicted microsatellite stability groups in both patient cohorts (training, P =0.032; external test, P =0.046). CONCLUSIONS The authors developed and validated a model based on subregion radiomic features of multiparametric MRI to evaluate high-risk subregions with MSI and predict the MSI status of RC preoperatively, which may assist in individualized treatment decisions and positioning for biopsy.
Collapse
Affiliation(s)
- Zhiping Cai
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Zhenyu Xu
- Department of Radiology, The First People’s Hospital of Foshan, Foshan
| | - Yifan Chen
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Rong Zhang
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Baoliang Guo
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Haixiong Chen
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Fusheng Ouyang
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Xinjie Chen
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Xiaobo Chen
- Department of Radiology, Guangdong Provincial People’s Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, People’s Republic of China
| | - Dechao Liu
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Chun Luo
- Department of Radiology, The First People’s Hospital of Foshan, Foshan
| | - Xiaohong Li
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Wei Liu
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Cuiru Zhou
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Xinqun Guan
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Ziwei Liu
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| | - Hai Zhao
- Department of Radiology, The First People’s Hospital of Foshan, Foshan
| | - Qiugen Hu
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People’s Hospital of Shunde)
| |
Collapse
|
50
|
Wu L, He C, Zhao T, Li T, Xu H, Wen J, Xu X, Gao L. Diagnosis and treatment status of inoperable locally advanced breast cancer and the application value of inorganic nanomaterials. J Nanobiotechnology 2024; 22:366. [PMID: 38918821 PMCID: PMC11197354 DOI: 10.1186/s12951-024-02644-9] [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: 03/26/2024] [Accepted: 06/16/2024] [Indexed: 06/27/2024] Open
Abstract
Locally advanced breast cancer (LABC) is a heterogeneous group of breast cancer that accounts for 10-30% of breast cancer cases. Despite the ongoing development of current treatment methods, LABC remains a severe and complex public health concern around the world, thus prompting the urgent requirement for innovative diagnosis and treatment strategies. The primary treatment challenges are inoperable clinical status and ineffective local control methods. With the rapid advancement of nanotechnology, inorganic nanoparticles (INPs) exhibit a potential application prospect in diagnosing and treating breast cancer. Due to the unique inherent characteristics of INPs, different functions can be performed via appropriate modifications and constructions, thus making them suitable for different imaging technology strategies and treatment schemes. INPs can improve the efficacy of conventional local radiotherapy treatment. In the face of inoperable LABC, INPs have proposed new local therapeutic methods and fostered the evolution of novel strategies such as photothermal and photodynamic therapy, magnetothermal therapy, sonodynamic therapy, and multifunctional inorganic nanoplatform. This article reviews the advances of INPs in local accurate imaging and breast cancer treatment and offers insights to overcome the existing clinical difficulties in LABC management.
Collapse
Affiliation(s)
- Linxuan Wu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Chuan He
- Department of Laboratory Medicine, The First Hospital of China Medical University, Shenyang, 110001, China
| | - Tingting Zhao
- Department of Breast Surgery, The First Affiliated Hospital of China Medical University, Shenyang, 110001, China
| | - Tianqi Li
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Hefeng Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China
| | - Jian Wen
- Department of Breast Surgery, The Fourth Affiliated Hospital of China Medical University, Shenyang, 110032, China.
| | - Xiaoqian Xu
- School of Intelligent Medicine, China Medical University, Shenyang, 110122, China.
| | - Lin Gao
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110022, China.
| |
Collapse
|