1
|
Castro-Oropeza R, Velazquez-Velazquez C, Vazquez-Santillan K, Mantilla-Morales A, Ruiz Tachiquin ME, Torres J, Rios-Sarabia N, Mayani H, Piña-Sanchez P. Landscape of lncRNAs expressed in Mexican patients with triple‑negative breast cancer. Mol Med Rep 2025; 31:163. [PMID: 40211710 PMCID: PMC12015155 DOI: 10.3892/mmr.2025.13528] [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: 07/31/2024] [Accepted: 02/24/2025] [Indexed: 04/25/2025] Open
Abstract
Long non‑coding RNAs (lncRNAs) are key regulators of gene expression, that can regulate a range of carcinogenic processes. Moreover, they exhibit stability in biological fluids, with some displaying tissue specificity. As their expression depends on specific conditions or is linked to the regulation of particular signaling pathways, lncRNAs are promising candidates for providing insights into the likely progression of the disease. This allows for the stratification of patients based on their risk of progression, making them potential prognostic biomarkers in various types of cancer. In addition, the tissue‑specific expression profile of lncRNAs renders them ideal candidates for detection, prognosis and monitoring of cancer progression. The present study aims to provide an overview of differentially expressed lncRNAs in Mexican patients with triple‑negative breast cancer (TNBC), a subtype of breast cancer. The aim was to identify potential prognostic biomarkers that can be applied to improve the clinical management of Mexican patients with TNBC. Human Transcriptome Array 2.0 microarrays were used to analyze the transcriptome of TNBC and luminal tumors, which are reported to have a good prognosis amongst aggressive tumor types. Subsequently, results from these microarrays were validated in a cohort from The Cancer Genome Atlas, an independent cohort of Mexican patients and in breast cancer cell lines (MCF7, ZR75, T47D, MDA‑MB‑231, MDA‑MB‑468 and BT20). A total of 746 differentially expressed transcripts were identified, including 102 lncRNAs in TNBC compared with luminal tumors. Among the lncRNAs with the most significant changes in expression levels, SOX9‑AS was highly expressed in TNBC, whereas the expression of Lnc‑peroxidasin‑3:1 (Lnc‑PXDN‑3:1), Lnc‑RNA Synapse Defective Rho GTPase Homolog (Lnc‑SYDE) and long intergenic non‑coding RNA (LINC)01087 were decreased. In addition, the low expression of lncRNA LINC01087, LINC02568, ACO22196, and lncRNA eosinophil granule ontogeny transcript (Lnc‑EGOT) was associated with poor overall survival (OS). Further analysis revealed that the high expression levels of Lnc‑PXDN‑3:1, Lnc RNA fibrous sheath interacting protein 1‑6:3 and (LINC)00182 were associated with reduced survival in patients with the luminal subtype of breast cancer. Similarly, low expression levels of lncRNAs such as GATA binding protein 3‑1 (Lnc‑GATA‑3‑1), LINC01087, and BX679671.1 in luminal subtypes of breast cancer, as well as LINC00504 and LncRNA rho guanine nucleotide exchange factor 38 intronic transcript 1 (Lnc‑ARHGEF38‑IT1) in basal subtypes have been linked to poorer survival. The interactions and functions of LINC01087 were then investigated, revealing the interaction of LINC01087 with RNAs and transcription factors, highlighting their potential involvement in the estrogen receptor pathway. The present study provided a detailed analysis of the expression of lncRNAs in TNBC, which highlights the role of lncRNAs as a biomarker in the survival outcomes of patients with breast cancer to improve the understanding of transcriptional regulation in TNBC.
Collapse
Affiliation(s)
- Rosario Castro-Oropeza
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Cindy Velazquez-Velazquez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Karla Vazquez-Santillan
- Laboratory of Innovation in Precision Medicine, National Institute of Genomic Medicine, Mexico City 14610, Mexico
| | - Alejandra Mantilla-Morales
- Department of Pathology, High Specialty Medical Unit Oncology Hospital, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Martha-Eugenia Ruiz Tachiquin
- Molecular Biology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Javier Torres
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Nora Rios-Sarabia
- Infectious and Parasitic Diseases Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Hector Mayani
- Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| | - Patricia Piña-Sanchez
- Molecular Oncology Laboratory, Oncology Research Unit, XXI Century National Medical Center, The Mexican Institute of Social Security, Mexico City 06720, Mexico
| |
Collapse
|
2
|
Yang Y, Han K, Xu Z, Cai Z, Zhao H, Hong J, Pan J, Guo L, Huang W, Hu Q, Xu Z. Development and Validation of Multiparametric MRI-based Interpretable Deep Learning Radiomics Fusion Model for Predicting Lymph Node Metastasis and Prognosis in Rectal Cancer: A Two-center Study. Acad Radiol 2025; 32:2642-2654. [PMID: 39638641 DOI: 10.1016/j.acra.2024.11.045] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/07/2024] [Accepted: 11/16/2024] [Indexed: 12/07/2024]
Abstract
RATIONALE AND OBJECTIVES To develop interpretable machine learning models that utilize deep learning (DL) and radiomics based on multiparametric Magnetic resonance imaging (MRI) to predict preoperative lymph node (LN) metastasis in rectal cancer. MATERIALS AND METHODS This retrospective study involved 286 cancer patients confirmed by histopathology from center 1 (Training set) and 66 patients from center 2 (External test set). Radiomics features were extracted from T2-weighted imaging (T2WI) and diffusion-weighted imaging (DWI) sequences, whereas DL features were obtained using four models: MobileNet-V3-large, Inception-V3, ResNet50, and VGG16. These DL radiomics (DLR) features were then combined to construct a machine learning model. The Shapley additive interpretation (SHAP) tool was utilized to investigate the interpretability of the model. We evaluated and compared the diagnostic performance of senior and junior radiologists, with and without the aid of the optimal DLR model. Kaplan-Meier survival curve was used to analyze the prognosis of patients. RESULTS The DLR model outperforms individual DL models and the radiomics model. The MobileNet-V3-large combination radiomics signature demonstrated the best performance, achieving an AUC of 0.878 on the Training set and 0.752 on the External test set. Compared to the traditional radiomics model, the AUC for the Training set increased by 0.094 and by 0.051 for the External test set. This model facilitated improved diagnostic performance among both junior and senior radiologists. Specifically, the AUC values for junior and senior radiologists increased by 0.162 and 0.232, respectively, on the Training set; and by 0.096 and 0.113, respectively, on the External test set. The DLR model demonstrated strong performance in risk stratification for disease-free survival. CONCLUSION The DLR model developed from multiparametric MRI can effectively distinguish cancer LN metastasis and enhance radiologists' diagnostic performance.
Collapse
Affiliation(s)
- Yunjun Yang
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.)
| | - Kaiting Han
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.)
| | - Zhenyu Xu
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.)
| | - Zhiping Cai
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China (Z.C., Q.H.)
| | - Hai Zhao
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.)
| | - Julu Hong
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.)
| | - Jiawei Pan
- Department of information system, The First People's Hospital of Foshan, Foshan, China (J.P.)
| | - Li Guo
- Department of Institute of Translational Medicine, The First People's Hospital of Foshan, Foshan, China (L.G.)
| | - Weijun Huang
- Department of Ultrasound, The First People's Hospital of Foshan, Foshan, China (W.H.)
| | - Qiugen Hu
- Department of Radiology, Shunde Hospital, Southern Medical University (The First People's Hospital of Shunde), Foshan, China (Z.C., Q.H.)
| | - Zhifeng Xu
- Department of Radiology, The First People's Hospital of Foshan, Foshan, China (Y.Y., K.H., Z.X., H.Z., J.H., Z.X.).
| |
Collapse
|
3
|
Yang T, Wang X, Jin Y, Yao X, Sun Z, Chen P, Zhou S, Zhu W, Chen W. Deep learning radiopathomics predicts targeted therapy sensitivity in EGFR-mutant lung adenocarcinoma. J Transl Med 2025; 23:482. [PMID: 40301933 PMCID: PMC12039126 DOI: 10.1186/s12967-025-06480-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Accepted: 04/11/2025] [Indexed: 05/01/2025] Open
Abstract
BACKGROUND Ttyrosine kinase inhibitors (TKIs) represent the standard first-line treatment for patients with epidermal growth factor receptor (EGFR)-mutant lung adenocarcinoma. However, not all patients with EGFR mutations respond to TKIs. This study aims to develop a deep learning radiological-pathological-clinical (DLRPC) model that integrates computed tomography (CT) images, hematoxylin and eosin (H&E)-stained aspiration biopsy samples, and clinical data to predict the response in EGFR-mutant lung adenocarcinoma patients undergoing TKIs treatment. METHODS We retrospectively analyzed data from 214 lung adenocarcinoma patients who received TKIs treatment from two medical centers between September 2013 and June 2023. The DLRPC model leverages paired CT, pathological images and clinical data, incorporating a clinical-based attention mask to further explore the cross-modality associations. To evaluate its diagnostic performance, we compared the DLRPC model against single-modality models and a decision level fusion model based on Dempster-Shafer theory. Model performances metrics, including area under the curve (AUC), accuracy, sensitivity, specificity, positive predictive value (PPV), and negative predictive value (NPV), were used for evaluation. The Delong test assessed statistically significantly differences in AUC among models. RESULTS The DLRPC model demonstrated strong performance, achieving an AUC value of 0.8424. It outperformed the single-modality models (AUC = 0.6894, 0.7753, 0.8052 for CT model, pathology model and clinical model, respectively. P < 0.05). Additionally, the DLRPC model surpassed the decision level fusion model (AUC = 0.8132, P < 0.05). CONCLUSION The DLRPC model effectively predicts the response of EGFR-mutant lung adenocarcinoma patients to TKIs, providing a promising tool for personalized treatment decisions in lung cancer management.
Collapse
Affiliation(s)
- Taotao Yang
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
| | - Xianqi Wang
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China
| | - Yuan Jin
- Zhejiang Lab, Hangzhou, 311121, China
| | - Xiaohong Yao
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Army Medical University, (Third Military Medical University), Chongqing, 400038, China
| | - Zhiyuan Sun
- Department of Radiology, Jinling Hospital, Medical School of Nanjing University, Nanjing, 210002, China
| | - Pinzhen Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | - Suyi Zhou
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China
| | | | - Wei Chen
- Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, 400038, China.
- Yu-Yue Pathology Scientific Research Center, Chongqing, 400039, China.
| |
Collapse
|
4
|
Xu H, Yang A, Kang M, Lai H, Zhou X, Chen Z, Lin L, Zhou P, Deng H. Intratumoral and peritumoral radiomics signature based on DCE-MRI can distinguish between luminal and non-luminal breast cancer molecular subtypes. Sci Rep 2025; 15:14720. [PMID: 40289183 PMCID: PMC12034752 DOI: 10.1038/s41598-025-98155-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2024] [Accepted: 04/09/2025] [Indexed: 04/30/2025] Open
Abstract
Distinguishing the luminal subtypes of breast cancer (BC) remaining challenging. Thus, the aim of this study was to investigate the feasibility of radiomic signature using intratumoral and peritumoral features obtained from dynamic contrast-enhanced MRI (DCE-MRI) in preoperatively discriminating the luminal from non-luminal type in patients with BC. A total of 305 patients with pathologically confirmed BC from three hospitals were retrospectively enrolled. The LASSO method was then used for selecting features, and the radiomic score (radscore) for each patient was calculated. Based on the radscore, Radiomic signature of intratumoral, peritumoral, and combined intratumoral and peritumoral were established, respectively. The performances of the radiomic signatures were validated with receiver operator characteristic (ROC) curve and decision curve analysis. For predicting molecular subtypes, the AUC for intratumoral radiomic signature was 0.817, 0.838, and 0.883 in the training set, internal validation set, and external validation set, respectively. AUC for the peritumoral radiomic signature was 0.863, 0.895, and 0.889 in the training set, internal validation set, and external validation set, respectively. The AUC for combined intratumoral and peritumoral radiomic signature was 0.956, 0.945, and 0.896 in the training set, internal validation set, and external validation set, respectively. Additional contributing value of combined intratumoral and peritumoral radiomic signature to the intratumoral radiomic signature was statistically significant [NRI, 0.300 (95% CI: 0.117-0.482), P = 0.001 in internal validation set; NRI, 0.224 (95% CI: 0.038-0.410), P = 0.018 in external validation set]. These results indicated that the radiomic signature combining intratumoral and peritumoral features showed good performance in predicting the luminal type of breast cancer.
Collapse
Affiliation(s)
- Hao Xu
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Ao Yang
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Min Kang
- Department of Radiology, Sichuan Provincial Maternity and Child Health Care Hospital, Chengdu, China
| | - Hua Lai
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Xinzhu Zhou
- Department of Radiology, Chengdu Women's and Children's Central Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Zhe Chen
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Libo Lin
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Peng Zhou
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China
| | - Heping Deng
- Department of Radiology, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Center, Sichuan Cancer Hospital & Institute, University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
5
|
Yao X, Deng S, Han X, Huang D, Cao Z, Ning X, Ao W. Deep Learning Algorithm‑Based MRI Radiomics and Pathomics for Predicting Microsatellite Instability Status in Rectal Cancer: A Multicenter Study. Acad Radiol 2025; 32:1934-1945. [PMID: 39289097 DOI: 10.1016/j.acra.2024.09.008] [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: 07/14/2024] [Revised: 08/27/2024] [Accepted: 09/01/2024] [Indexed: 09/19/2024]
Abstract
RATIONALE AND OBJECTIVES To develop and validate multimodal deep-learning models based on clinical variables, multiparametric MRI (mp-MRI) and hematoxylin and eosin (HE) stained pathology slides for predicting microsatellite instability (MSI) status in rectal cancer patients. MATERIALS AND METHODS A total of 467 surgically confirmed rectal cancer patients from three centers were included in this study. Patients from center 1 were randomly divided into a training set (242 patients) and an internal validation (invad) set (105 patients) in a 7:3 ratio. Patients from centers 2 and 3 (120 patients) were included in an external validation (exvad) set. HE and immunohistochemistry (IHC) staining were analyzed, and MSI status was confirmed by IHC staining. Independent predictive factors were identified through univariate and multivariate analyses based on clinical evaluations and were used to construct a clinical model. Deep learning with ResNet-101 was applied to preoperative MRI (T2WI, DWI, and contrast-enhanced T1WI sequences) and postoperative HE-stained images to calculate deep-learning radiomics score (DLRS) and deep-learning pathomics score (DLPS), respectively, and to DLRS and DLPS models. Receiver operating characteristic (ROC) curves were plotted, and the area under the curve (AUC) was used to evaluate and compare the predictive performance of each model. RESULTS Among all rectal cancer patients, 82 (17.6%) had MSI. Long diameter (LD) and pathological T stage (pT) were identified as independent predictors and were used to construct the clinical model. After undergoing deep learning and feature selection, a final set of 30 radiomics features and 30 pathomics features were selected to construct the DLRS and DLPS models. A nomogram combining the clinical model, DLRS, and DLPS was created through weighted linear combination. The AUC values of the clinical model for predicting MSI were 0.714, 0.639, and 0.697 in the training, invad, and exvad sets, respectively. The AUCs of DLPS and DLRS ranged from 0.896 to 0.961 across the training, invad, and exvad sets. The nomogram achieved AUC values of 0.987, 0.987, and 0.974, with sensitivities of 1.0, 0.963, and 1.0 and specificities of 0.919, 0.949, and 0.867 in the training, invad, and exvad sets, respectively. The nomogram outperformed the other three models in all sets, with DeLong test results indicating superior predictive performance in the training set. CONCLUSION The nomogram, incorporating clinical data, mp-MRI, and HE staining, effectively reflects tumor heterogeneity by integrating multimodal data. This model demonstrates high predictive accuracy and generalizability in predicting MSI status in rectal cancer patients.
Collapse
Affiliation(s)
- Xiuzhen Yao
- Department of Ultrasound, Putuo People's Hospital, School of Medicine, Tongji University, Shanghai, China (X.Y.)
| | - Shuitang Deng
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Xiaoyu Han
- Department of Pathology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (X.H.)
| | - Danjiang Huang
- Department of Radiology, Huangyan Hospital, Wenzhou Medical University, Taizhou First People's Hospital, Taizhou, Zhejiang Province, China (D.H.)
| | - Zhengyu Cao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Xiaoxiang Ning
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.)
| | - Weiqun Ao
- Department of Radiology, Tongde Hospital of Zhejiang Province, Hangzhou, Zhejiang Province, China (S.D., Z.C., X.N., W.A.).
| |
Collapse
|
6
|
Wan CF, Jiang ZY, Wang YQ, Wang L, Fang H, Jin Y, Dong Q, Zhang XQ, Jiang LX. Radiomics of Multimodal Ultrasound for Early Prediction of Pathologic Complete Response to Neoadjuvant Chemotherapy in Breast Cancer. Acad Radiol 2025; 32:1861-1873. [PMID: 39690072 DOI: 10.1016/j.acra.2024.11.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/03/2024] [Accepted: 11/04/2024] [Indexed: 12/19/2024]
Abstract
RATIONALE AND OBJECTIVES To construct and validate a clinical-radiomics model based on radiomics features extracted from two-stage multimodal ultrasound and clinicopathologic information for early predicting pathologic complete response (pCR) to neoadjuvant chemotherapy (NAC) in breast cancer patients treated with NAC. MATERIALS AND METHODS Consecutive women with biopsy-proven breast cancer undergoing multimodal US pretreatment and after two cycles of NAC and followed by surgery between January 2014 and November 2023 were retrospectively collected for clinical-radiomics model construction (n = 274) and retrospective test (n = 134). The predictive performance of it was further tested in a subsequent prospective internal test set recruited between January 2024 to July 2024 (n = 76). Finally, a total of 484 patients were enrolled. The clinical-radiomics model predictive performance was compared with radiomics model, clinical model and radiologists' visual assessment by area under the receiver operating characteristic curve (AUC) analysis and DeLong test. RESULTS The proposed clinical-radiomics model obtained the AUC values of 0.92 (95%CI: 0.88, 0.94) and 0.85 (95%CI: 0.79, 0.89) in retrospective and prospective test sets, respectively, which were significantly higher than that those of the radiomics model (AUCs: 0.75-0.85), clinical model (AUCs: 0.68-0.72) and radiologists' visual assessments (AUCs:0.59-0.68) (all p < 0.05). In addition, the predictive efficacy of the radiologists was improved under the assistance of the clinical-radiomics model significantly. CONCLUSION The clinical-radiomics model developed in this study, which integrated clinicopathologic information and two-stage multimodal ultrasound features, was able to early predict pCR to NAC in breast cancer patients with favorable predictive effectiveness.
Collapse
Affiliation(s)
- Cai-Feng Wan
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Zhuo-Yun Jiang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, PR China (Z-y.J.)
| | - Yu-Qun Wang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Lin Wang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Hua Fang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Ye Jin
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Qi Dong
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.)
| | - Xue-Qing Zhang
- Department of Pathology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (X-q.Z.)
| | - Li-Xin Jiang
- Department of Ultrasound, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, PR China (C-f.W., Y-q.W., L.W., H.F., Y.J., Q.D., L-x.J.).
| |
Collapse
|
7
|
Li P, Yin M, Guerrini S, Gao W. Roles of artificial intelligence and high frame-rate contrast-enhanced ultrasound in the differential diagnosis of Breast Imaging Reporting and Data System 4 breast nodules. Gland Surg 2025; 14:462-478. [PMID: 40256461 PMCID: PMC12004330 DOI: 10.21037/gs-24-187] [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: 05/23/2024] [Accepted: 03/07/2025] [Indexed: 04/22/2025]
Abstract
Background Breast cancer prevalence and mortality are rising, emphasizing the need for early, accurate diagnosis. Contrast-enhanced ultrasound (CEUS) and artificial intelligence (AI) show promise in distinguishing benign from malignant breast nodules. We compared the diagnostic values of AI, high frame-rate CEUS (HiFR-CEUS), and their combination in Breast Imaging Reporting and Data System (BI-RADS) 4 nodules, using pathology as the gold standard. Methods Patients with BI-RADS 4 breast nodules who were hospitalized at the Department of Thyroid and Breast Surgery, Taizhou People's Hospital from December 2021 to June 2022 were enrolled in the study.80 female patients (80 lesions) underwent preoperative AI and/or HiFR-CEUS. We assessed diagnostic outcomes of AI, HiFR-CEUS, and their combination, calculating sensitivity (SE), specificity (SP), accuracy (ACC), positive/negative predictive values (PPV/NPV). Reliability was compared using Kappa statistics, and AI-HiFR-CEUS correlation was analyzed with Pearson's test. Receiver operating characteristic curves were plotted to compare diagnostic accuracy of AI, HiFR-CEUS, and their combined approach in differentiating BI-RADS 4 lesions. Results Of the 80 lesions, 18 were pathologically confirmed to be benign, while the remaining 62 were malignant. The SE, SP, ACC, PPV, and NPV were 75.81%, 94.44%, 80.00%, 97.92%, and 53.13% in the AI group, 74.20%, 94.44%, 78.75%, 97.91%, and 51.51% in the HiFR-CEUS group, and 98.39%, 88.89%, 96.25%, 96.83%, and 94.12% in the combination group, respectively. Thus, the SE, ACC, and NPV of the combination group were significantly higher than those of the AI and HiFR-CEUS groups, and the SP of the combination group was lower (all P<0.05); however, no significant difference was found between the groups in terms of the PPV (P>0.05). No statistically significant difference was observed in the diagnostic performance of the AI and HiFR-CEUS groups (all P>0.05). The AI and HiFR-CEUS groups had moderate agreement with the "gold standard" (Kappa =0.551, Kappa =0.530, respectively), while the combination group had high agreement (Kappa =0.890). AI was positively correlated with HiFR-CEUS (r=0.249, P<0.05). The area under the curves (AUCs) of AI, HiFR-CEUS, and both in combination were 0.851±0.039, 0.815±0.047, and 0.936±0.039, respectively. Thus, the AUC of the combination group was significantly higher than those of the AI and HiFR-CEUS groups (Z1=2.207, Z2=2.477, respectively, both P<0.05). The AI group had a higher AUC than the HiFR-CEUS group, but the difference was not statistically significant (Z3=0.554, P>0.05). Conclusions Compared with AI alone or HiFR-CEUS alone, the combined use of these two methods had higher diagnostic performance in distinguishing between benign and malignant BI-RADS 4 breast nodules. Thus, our combination method could further improve the diagnostic accuracy and guide clinical decision making.
Collapse
Affiliation(s)
- Ping Li
- Ultrasound Medicine Department, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Ming Yin
- Ultrasound Medicine Department, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| | - Susanna Guerrini
- Unit of Diagnostic Imaging, Department of Medical Sciences, Azienda Ospedaliero-Universitaria Senese, University of Siena, Siena, Italy
| | - Wenxiang Gao
- Ultrasound Medicine Department, The Affiliated Taizhou People’s Hospital of Nanjing Medical University, Taizhou, China
| |
Collapse
|
8
|
Mao HM, Zhang JJ, Zhu B, Guo WL. A novel deep learning radiopathomics model for predicting carcinogenesis promotor cyclooxygenase-2 expression in common bile duct in children with pancreaticobiliary maljunction: a multicenter study. Insights Imaging 2025; 16:74. [PMID: 40146354 PMCID: PMC11950503 DOI: 10.1186/s13244-025-01951-5] [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: 01/23/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025] Open
Abstract
OBJECTIVES To develop and validate a deep learning radiopathomics model (DLRPM) integrating radiological and pathological imaging data to predict biliary cyclooxygenase-2 (COX-2) expression in children with pancreaticobiliary maljunction (PBM), and to compare its performance with single-modality radiomics, deep learning radiomics (DLR), and pathomics models. METHODS This retrospective study included 219 PBM patients, divided into a training set (n = 104; median age, 2.8 years, 75.0% females) and internal test set (n = 71; median age, 2.2 years, 83.1% females) from center I, and an external test set (n = 44; median age, 3.4 years, 65.9% females) from center II. Biliary COX-2 expression was detected using immunohistochemistry. Radiomics, DLR, and pathomics features were extracted from portal venous-phase CT images and H&E-stained histopathological slides, respectively, to build individual single-modality models. These were then integrated to develop the DLRPM, combining three predictive signatures. Model performance was evaluated using AUC, net reclassification index (NRI, for assessing improvement in correct classification) and integrated discrimination improvement (IDI). RESULTS The DLRPM demonstrated the highest performance, with AUCs of 0.851 (95% CI, 0.759-0.942) in internal test set and 0.841 (95% CI, 0.721-0.960) in external test set. In comparison, AUCs for the radiomics, DLR, and pathomics models were 0.532-0.602, 0.658-0.660, and 0.787-0.805, respectively. The DLRPM significantly outperformed three single-modality models, as demonstrated by the NRI and IDI tests (all p < 0.05). CONCLUSION The multimodal DLRPM could accurately and robustly predict COX-2 expression, facilitating risk stratification and personalized postoperative management in PBM. However, prospective multicenter studies with larger cohorts are needed to further validate its generalizability. CRITICAL RELEVANCE STATEMENT Our proposed deep learning radiopathomics model, integrating CT and histopathological images, provides a novel and cost-effective approach to accurately predict biliary cyclooxygenase-2 expression, potentially advancing individualized risk stratification and improving long-term outcomes for pediatric patients with pancreaticobiliary maljunction. KEY POINTS Predicting biliary COX-2 expression in pancreaticobiliary maljunction (PBM) is critical but challenging. A deep learning radiopathomics model achieved high predictive accuracy for COX-2. The model supports patient stratification and personalized postoperative management in PBM.
Collapse
Affiliation(s)
- Hui-Min Mao
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China
| | - Jian-Jun Zhang
- Department of Neonatal Surgery, Xuzhou Children's Hospital, Xuzhou, China
| | - Bin Zhu
- Department of Interventional Therapy, Xuzhou Children's Hospital, Xuzhou, China
| | - Wan-Liang Guo
- Department of Radiology, Children's Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
9
|
Xu K, Wang X, Zhou C, Zuo J, Zeng C, Zhou P, Zhang L, Gao X, Wang X. Synergic value of 3D CT-derived body composition and triglyceride glucose body mass for survival prognostic modeling in unresectable pancreatic cancer. Front Nutr 2025; 12:1499188. [PMID: 40177184 PMCID: PMC11961436 DOI: 10.3389/fnut.2025.1499188] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 03/03/2025] [Indexed: 04/05/2025] Open
Abstract
Background Personalized and accurate survival risk prognostication remains a significant challenge in advanced pancreatic ductal adenocarcinoma (PDAC), despite extensive research on prognostic and predictive markers. Patients with PDAC are prone to muscle loss, fat consumption, and malnutrition, which is associated with inferior outcomes. This study investigated the use of three-dimensional (3D) anthropometric parameters derived from computed tomography (CT) scans and triglyceride glucose-body mass index (TyG-BMI) in relation to overall survival (OS) outcomes in advanced PDAC patients. Additionally, a predictive model for 1 year OS was developed based on body components and hematological indicators. Methods A retrospective analysis was conducted on 303 patients with locally advanced PDAC or synchronous metastases undergoing first-line chemotherapy, all of whom had undergone pretreatment abdomen-pelvis CT scans. Automatic 3D measurements of subcutaneous and visceral fat volume, skeletal muscle volume, and skeletal muscle density (SMD) were assessed at the L3 vertebral level by an artificial intelligence assisted diagnosis system (HY Medical). Various indicators including TyG-BMI, nutritional indicators [geriatric nutritional risk index (GNRI) and prealbumin], and inflammation indicators [(C-reactive protein (CRP) and neutrophil to lymphocyte ratio (NLR)] were also recorded. All patients underwent follow-up for at least 1 year and a dynamic nomogram for personalized survival prediction was constructed. Results We included 211 advanced PDAC patients [mean (standard deviation) age, 63.4 ± 11.2 years; 89 women (42.2) %)]. Factors such as low skeletal muscle index (SMI) (P = 0.011), high visceral to subcutaneous adipose tissue area ratio (VSR) (P < 0.001), high visceral fat index (VFI) (P < 0.001), low TyG-BMI (P = 0.004), and low prealbumin (P = 0.001) were identified as independent risk factors associated with 1 year OS. The area under the curve of the established dynamic nomogram was 0.846 and the calibration curve showed good consistency. High-risk patients (> 211.9 points calculated using the nomogram) had significantly reduced survival rates. Conclusion In this study, the proposed nomogram model (with web-based tool) enabled individualized prognostication of OS and could help to guide risk-adapted nutritional treatment for patients with unresectable PDAC or synchronous metastases.
Collapse
Affiliation(s)
- Kangjing Xu
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinbo Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Changsheng Zhou
- Department of Radiology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Junbo Zuo
- Department of General Surgery, The Affiliated People’s Hospital of Jiangsu University, Zhenjiang, China
| | - Chenghao Zeng
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Pinwen Zhou
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Zhang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xuejin Gao
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Xinying Wang
- Department of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
10
|
Zeng Q, Liu L, He C, Zeng X, Wei P, Xu D, Mao N, Yu T. Early and noninvasive prediction of response to neoadjuvant therapy for breast cancer via longitudinal ultrasound and MR deep learning: A multicentre study. Acad Radiol 2025; 32:1264-1273. [PMID: 39542804 DOI: 10.1016/j.acra.2024.10.033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/18/2024] [Accepted: 10/21/2024] [Indexed: 11/17/2024]
Abstract
RATIONALE AND OBJECTIVES The early prediction of response to neoadjuvant chemotherapy (NAC) will aid in the development of personalized treatments for patients with breast cancer. This study investigated the value of longitudinal multimodal deep learning (DL) based on breast MR and ultrasound (US) in predicting pathological complete response (pCR) after NAC. MATERIALS AND METHODS We retrospectively reviewed the pre-NAC and post-2nd-NAC MR and/or US images of 448 patients enrolled from three centers and extracted DL features from the largest section of the breast tumour using ResNet50. T test, Pearson correlation analysis and least absolute shrinkage and selection operator regression were used to select the most significant DL features for the pre-NAC and post-2nd-NAC MR and US DL models. The stacking model integrates different single-modality DL models and meaningful clinical data. The diagnostic performance of the models was evaluated. RESULTS In all the patients, the pCR rate was 36.65%. There was no significant difference in diagnostic performance between the different single-modality DL models (DeLong test, p > 0.05). The stacking model integrating the above four DL models with HER2 status yielded areas under the curves of 0.951-0.979, accuracies of 91.55%-92.65%, sensitivities of 90.63%-93.94%, and specificities of 89.47%-94.44% in the cohorts. CONCLUSION Longitudinal multimodal DL can be useful in predicting pCR. The stacking model can be used as a new tool for the early noninvasive prediction of the response to NAC, as evidenced by its excellent performance, and therefore aid the development of personalized treatment strategies for patients with breast cancer.
Collapse
Affiliation(s)
- Qiao Zeng
- Department of Radiology, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (Q.Z., L.L., P.W.)
| | - Lan Liu
- Department of Radiology, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (Q.Z., L.L., P.W.)
| | - Chongwu He
- Department of Breast Surgery, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (C.H., X.Z., T.Y.)
| | - Xiaoqiang Zeng
- Department of Breast Surgery, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (C.H., X.Z., T.Y.)
| | - Pengfei Wei
- Department of Radiology, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (Q.Z., L.L., P.W.)
| | - Dong Xu
- Department of Ultrasound, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Cancer and Basic Medicine (IBMC), Chinese Academy of Sciences, Hangzhou 310022, China (D.X.)
| | - Ning Mao
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China (N.M.)
| | - Tenghua Yu
- Department of Breast Surgery, Jiangxi Cancer Hospital&Institute,Jiangxi Clinical Research Center for Cancer, The Second Affiliated Hospital of Nanchang Medical College, Nanchang 330029, Jiangxi Province, China (C.H., X.Z., T.Y.).
| |
Collapse
|
11
|
Dai X, Lu H, Wang X, Liu Y, Zang J, Liu Z, Sun T, Gao F, Sui X. Ultrasound-based artificial intelligence model for prediction of Ki-67 proliferation index in soft tissue tumors. Acad Radiol 2025; 32:1178-1188. [PMID: 39406581 DOI: 10.1016/j.acra.2024.09.067] [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: 07/30/2024] [Revised: 09/13/2024] [Accepted: 09/30/2024] [Indexed: 03/03/2025]
Abstract
RATIONALE AND OBJECTIVES To investigate the value of deep learning (DL) combined with radiomics and clinical and imaging features in predicting the Ki-67 proliferation index of soft tissue tumors (STTs). MATERIALS AND METHODS In this retrospective study, a total of 394 patients with STTs admitted from January 2021 to December 2023 in two separate hospitals were collected. Hospital-1 was the training cohort (323 cases, of which 89 and 234 were high and low Ki-67, respectively) and Hospital-2 was the external validation cohort (71 cases, of which 23 and 48 were high and low Ki-67, respectively). Clinical and ultrasound characteristics including age, sex, tumor size, morphology, margins, internal echoes and blood flow were assessed. Risk factors with significant correlations were screened by univariate and multivariate logistic regression analyses. After extracting the radiomics and DL features, the feature fusion model is constructed by Support Vector Machine. The prediction results obtained from separate clinical features, radiomics features and DL features were combined to construct decision fusion models. Finally, the DeLong test was used to compare whether the AUCs between the models were significantly different. RESULTS The three feature fusion models and three decision fusion models constructed demonstrated excellent diagnostic performance in predicting Ki-67 expression levels in STTs. Among them, the feature fusion model based on clinical, radiomics, and DL performed the best with an AUC of 0.911 (95% CI: 0.886-0.935) in the training cohort and 0.923 (95% CI: 0.873-0.972) in the validation cohort, and proved to be well-calibrated and clinically useful. The DeLong test showed that the decision fusion models based on clinical, radiomics and DL performed significantly worse than the three feature fusion models on the validation set. There was no statistical difference in diagnostic performance between the other models. CONCLUSION The ultrasound-based fusion model of clinical, radiomics, and DL features showed good performance in predicting Ki-67 expression levels in STTs.
Collapse
Affiliation(s)
- Xinpeng Dai
- Department of Ultrasound, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (X.D., X.W., Y.L., Z.L., X.S.).
| | - Haiyong Lu
- Department of Ultrasound, The First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China (H.L.).
| | - Xinying Wang
- Department of Ultrasound, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (X.D., X.W., Y.L., Z.L., X.S.).
| | - Yujia Liu
- Department of Ultrasound, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (X.D., X.W., Y.L., Z.L., X.S.).
| | - Jiangnan Zang
- Hebei Medical University, Shijiazhuang, Hebei province, China (J.Z.).
| | - Zongjie Liu
- Department of Ultrasound, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (X.D., X.W., Y.L., Z.L., X.S.).
| | - Tao Sun
- Department of Orthopaedic Oncology, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (T.S.).
| | - Feng Gao
- Department of Pathology, The Thrid Hospital of Hebei Medical University, Shijiazhuang, Hebei province, China (G.F.).
| | - Xin Sui
- Department of Ultrasound, Hebei Medical University Third Hospital, Shijiazhuang, Hebei province, China (X.D., X.W., Y.L., Z.L., X.S.).
| |
Collapse
|
12
|
Du H, Wang X, Wang K, Ai Q, Shen J, Zhu R, Wu J. Identifying invasiveness to aid lung adenocarcinoma diagnosis using deep learning and pathomics. Sci Rep 2025; 15:4913. [PMID: 39929860 PMCID: PMC11810995 DOI: 10.1038/s41598-025-87094-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 01/16/2025] [Indexed: 02/13/2025] Open
Abstract
Most classification efforts for primary subtypes of lung adenocarcinoma (LUAD) have not yet been integrated into clinical practice. This study explores the feasibility of combining deep learning and pathomics to identify tumor invasiveness in LUAD patients, highlighting its potential clinical value in assisting junior and intermediate pathologists. We retrospectively analyzed whole slide image (WSI) data from 289 patients with surgically resected ground-glass nodules (GGNs). First, three ResNet deep learning models were used to identify tumor regions. Second, features from the best-performing model were extracted to build pathomics using machine learning classifiers. Third, the accuracy of pathomics in predicting tumor invasiveness was compared with junior and intermediate pathologists' diagnoses. Performance was evaluated using the area under receiver operator characteristic curve (AUC). On the test cohort, ResNet18 achieved the highest AUC (0.956) and sensitivity (0.832) in identifying tumor areas, with an accuracy of 0.904; Random Forest provided high accuracy and AUC values of 0.814 and 0.807 in assessing tumor invasiveness. Pathology assistance improved diagnostic accuracy for junior and intermediate pathologists, with AUC values increasing from 0.547 to 0.759 and 0.656 to 0.769. This study suggests that deep learning and pathomics can enhance diagnostic accuracy, offering valuable support to pathologists.
Collapse
Affiliation(s)
- Hai Du
- Department of Radiology, Ordos Central Hospital, Ordos Inner Mongolia, China
| | - Xiulin Wang
- Stem Cell Clinical Research Center, The First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning, China
- Dalian Innovation Institute of Stem Cell and Precision Medicine, Dalian, Liaoning, China
| | - Kaifeng Wang
- Fujian Medical University, Fuzhou, Fujian, China
| | - Qi Ai
- Department of Radiology, Affiliated Xinhua Hospital of Dalian University, Dalian, Liaoning, China
| | - Jing Shen
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Ruiping Zhu
- Department of Pathology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China
| | - Jianlin Wu
- Department of Radiology, Affiliated Zhongshan Hospital of Dalian University, Dalian, Liaoning, China.
| |
Collapse
|
13
|
Liu H, Ying L, Song X, Xiang X, Wei S. Development of metastasis and survival prediction model of luminal and non-luminal breast cancer with weakly supervised learning based on pathomics. PeerJ 2025; 13:e18780. [PMID: 39866573 PMCID: PMC11759606 DOI: 10.7717/peerj.18780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 12/09/2024] [Indexed: 01/28/2025] Open
Abstract
Objective Breast cancer stands as the most prevalent form of cancer among women globally. This heterogeneous disease exhibits varying clinical behaviors. The stratification of breast cancer patients into risk groups, determined by their metastasis and survival outcomes, is pivotal for tailoring personalized treatments and therapeutic interventions. The pathological sections of radical specimens encompass a diverse range of histological information pertinent to the metastasis and survival of patients. In this study, our objective is to develop a deep learning model utilizing pathological images to predict the metastasis and survival outcomes for breast cancer patients. Methods This study utilized pathological sections from 204 radical mastectomy specimens obtained between January 2013 and December 2014 at the Second Affiliated Hospital of the Medical College of Zhejiang University. The 204 pathological slices were scanned and transformed into whole slide imaging (WSI), with manual labeling of all tumor areas. The WSI was then partitioned into smaller tiles measuring 512 × 512 pixels. Three networks, namely Densely Connected Convolutional Network 121 (DenseNet121), Residual Network (ResNet50), and Inception_v3, were assessed. Subsequently, we combined patch-level predictions, probability histograms, and Term Frequency-Inverse Document Frequency (TF-IDF) features to create comprehensive participants representations. These features served as the foundational input for developing a machine learning algorithm for metastasis analysis and a Cox regression model for survival analysis. Result Our results show that the Inception_v3 model shows a particularly robust patch recognition ability for estrogen receptor (ER) recognition. Our pathological model shows high accuracy in predicting tumor regions. The train area under the curve (AUC) of the Inception_v3 model based on supervised learning is 0.975, which is higher than the model established by weakly supervised learning. But the AUC of the metastasis prediction in training and testing sets is higher than value based on supervised learning. Furthermore, the C-index of the survival prediction model is 0.710 in the testing sets, which is also better than the value by supervised learning. Conclusion Our study demonstrates the significant potential of deep learning models in predicting breast cancer metastasis and prognosis, with the pathomic model showing high accuracy in identifying tumor areas and ER status. The integration of clinical features and pathomics signature into a nomogram further provides a valuable tool for clinicians to make individualized treatment decisions.
Collapse
Affiliation(s)
- Hui Liu
- Departments of Clinical Pathology, The Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Ying
- Departments of Clinical Pathology, The Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xing Song
- Departments of Clinical Pathology, The Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xueping Xiang
- Departments of Clinical Pathology, The Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| | - Shumei Wei
- Departments of Clinical Pathology, The Second Affiliated Hospital of Medical College of Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
14
|
Wang X, Wang Z, Wang W, Liu Z, Ma Z, Guo Y, Su D, Sun Q, Pei D, Duan W, Qiu Y, Wang M, Yang Y, Li W, Liu H, Ma C, Yu M, Yu Y, Chen T, Fu J, Li S, Yu B, Ji Y, Li W, Yan D, Liu X, Li ZC, Zhang Z. IDH-mutant glioma risk stratification via whole slide images: Identifying pathological feature associations. iScience 2025; 28:111605. [PMID: 39845415 PMCID: PMC11751506 DOI: 10.1016/j.isci.2024.111605] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 08/12/2024] [Accepted: 12/11/2024] [Indexed: 01/24/2025] Open
Abstract
This article aims to develop and validate a pathological prognostic model for predicting prognosis in patients with isocitrate dehydrogenase (IDH)-mutant gliomas and reveal the biological underpinning of the prognostic pathological features. The pathomic model was constructed based on whole slide images (WSIs) from a training set (N = 486) and evaluated on internal validation set (N = 209), HPPH validation set (N = 54), and TCGA validation set (N = 352). Biological implications of PathScore and individual pathomic features were identified by pathogenomics set (N = 100). The WSI-based pathological signature was an independent prognostic factor. Incorporating the pathological features into a clinical model resulted in a pathological-clinical model that predicted survival better than either the pathological model or clinical model alone. Ten categories of pathways (metabolism, proliferation, immunity, DNA damage response, disease, migrate, protein modification, synapse, transcription and translation, and complex cellular functions) were significantly correlated with the WSI-based pathological features.
Collapse
Affiliation(s)
- Xiaotao Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zilong Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zaoqu Liu
- Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Zeyu Ma
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial People’s Hospital, Zhengzhou, Henan, China
| | - Dingyuan Su
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Minkai Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yongqiang Yang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wenyuan Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Haoran Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Caoyuan Ma
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Miaomiao Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yinhui Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Te Chen
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Jing Fu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Sen Li
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Bin Yu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Yuchen Ji
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Wencai Li
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dongming Yan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
15
|
Komatsu M, Teraya N, Natsume T, Harada N, Takeda K, Hamamoto R. Clinical Application of Artificial Intelligence in Ultrasound Imaging for Oncology. JMA J 2025; 8:18-25. [PMID: 39926099 PMCID: PMC11799696 DOI: 10.31662/jmaj.2024-0203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/17/2024] [Indexed: 02/11/2025] Open
Abstract
Ultrasound (US) imaging is a widely used tool in oncology because of its noninvasiveness and real-time performance. However, its diagnostic accuracy can be limited by the skills of the examiner when performing manual scanning and by the presence of acoustic shadows that degrade image quality. Artificial intelligence (AI) technologies can support examiners in cancer screening and diagnosis by addressing these limitations. Here, we examine recent advances in AI research and development for US imaging in oncology. Breast cancer has been the most extensively studied cancer, with research predominantly focusing on tumor detection, differentiation between benign and malignant lesions, and prediction of lymph node metastasis. The American College of Radiology developed a medical imaging reporting and data system for various cancers that is often used to evaluate the accuracy of AI models. We will also explore the application of AI in clinical settings for US imaging in oncology. Despite progress, the number of approved AI-equipped software as medical devices for US imaging remains limited in Japan, the United States, and Europe. Practical issues that need to be addressed for clinical application include domain shifts, black boxes, and acoustic shadows. To address these issues, advances in image quality control, AI explainability, and preprocessing of acoustic shadows are essential.
Collapse
Affiliation(s)
- Masaaki Komatsu
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
| | - Naoki Teraya
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Department of Obstetrics and Gynecology, Showa University School of Medicine, Tokyo, Japan
| | - Takashi Natsume
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Department of Gynecology, National Cancer Center Hospital, Tokyo, Japan
| | - Naoaki Harada
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- HLPF Data Analytics Department, Fujitsu Ltd., Kawasaki, Japan
- Department of NCC Cancer Science, Biomedical Science and Engineering Track, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| | - Katsuji Takeda
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
| | - Ryuji Hamamoto
- Cancer Translational Research Team, RIKEN Center for Advanced Intelligence Project, Tokyo, Japan
- Division of Medical AI Research and Development, National Cancer Center Research Institute, Tokyo, Japan
- Department of NCC Cancer Science, Biomedical Science and Engineering Track, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University, Tokyo, Japan
| |
Collapse
|
16
|
Chia JLL, He GS, Ngiam KY, Hartman M, Ng QX, Goh SSN. Harnessing Artificial Intelligence to Enhance Global Breast Cancer Care: A Scoping Review of Applications, Outcomes, and Challenges. Cancers (Basel) 2025; 17:197. [PMID: 39857979 PMCID: PMC11764353 DOI: 10.3390/cancers17020197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND In recent years, Artificial Intelligence (AI) has shown transformative potential in advancing breast cancer care globally. This scoping review seeks to provide a comprehensive overview of AI applications in breast cancer care, examining how they could reshape diagnosis, treatment, and management on a worldwide scale and discussing both the benefits and challenges associated with their adoption. METHODS In accordance with PRISMA-ScR and ensuing guidelines on scoping reviews, PubMed, Web of Science, Cochrane Library, and Embase were systematically searched from inception to end of May 2024. Keywords included "Artificial Intelligence" and "Breast Cancer". Original studies were included based on their focus on AI applications in breast cancer care and narrative synthesis was employed for data extraction and interpretation, with the findings organized into coherent themes. RESULTS Finally, 84 articles were included. The majority were conducted in developed countries (n = 54). The majority of publications were in the last 10 years (n = 83). The six main themes for AI applications were AI for breast cancer screening (n = 32), AI for image detection of nodal status (n = 7), AI-assisted histopathology (n = 8), AI in assessing post-neoadjuvant chemotherapy (NACT) response (n = 23), AI in breast cancer margin assessment (n = 5), and AI as a clinical decision support tool (n = 9). AI has been used as clinical decision support tools to augment treatment decisions for breast cancer and in multidisciplinary tumor board settings. Overall, AI applications demonstrated improved accuracy and efficiency; however, most articles did not report patient-centric clinical outcomes. CONCLUSIONS AI applications in breast cancer care show promise in enhancing diagnostic accuracy and treatment planning. However, persistent challenges in AI adoption, such as data quality, algorithm transparency, and resource disparities, must be addressed to advance the field.
Collapse
Affiliation(s)
- Jolene Li Ling Chia
- NUS Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr. S117597, Singapore 119077, Singapore (G.S.H.)
| | - George Shiyao He
- NUS Yong Loo Lin School of Medicine, National University of Singapore, 10 Medical Dr. S117597, Singapore 119077, Singapore (G.S.H.)
| | - Kee Yuen Ngiam
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| | - Mikael Hartman
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| | - Qin Xiang Ng
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
- SingHealth Duke-NUS Global Health Institute, Singapore 169857, Singapore
| | - Serene Si Ning Goh
- Department of Surgery, National University Hospital, Singapore 119074, Singapore; (K.Y.N.); (M.H.)
- Saw Swee Hock School of Public Health, National University of Singapore and National University Health System, 12 Science Drive 2, #10-01, Singapore 117549, Singapore
| |
Collapse
|
17
|
Yao Y, Zhao Y, Guo X, Xu X, Fu B, Cui H, Xue J, Tian J, Lu K, Zhang L. Deep Learning for Distinguishing Mucinous Breast Carcinoma From Fibroadenoma on Ultrasound. Clin Breast Cancer 2025; 25:75-84. [PMID: 39317636 DOI: 10.1016/j.clbc.2024.09.001] [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: 11/13/2023] [Revised: 08/21/2024] [Accepted: 09/01/2024] [Indexed: 09/26/2024]
Abstract
PURPOSE Mucinous breast carcinoma (MBC) tends to be misdiagnosed as fibroadenomas (FA) due to its benign imaging characteristics. We aimed to develop a deep learning (DL) model to differentiate MBC and FA based on ultrasound (US) images. The model could contribute to the diagnosis of MBC for radiologists. METHODS In this retrospective study, 884 eligible patients (700 FA patients and 184 MBC patients) with 2257 US images were enrolled. The images were randomly divided into a training set (n = 1805 images) and a test set (n = 452 images) in a ratio of 8:2. First, we used the training set to establish DL model, DL+ age-cutoff model and DL+ age-tree model. Then, we compared the diagnostic performance of three models to get the optimal model. Finally, we evaluated the diagnostic performance of radiologists (4 junior and 4 senior radiologists) with and without the assistance of the optimal model in the test set. RESULTS The DL+ age-tree model yielded higher areas under the receiver operating characteristic curve (AUC) than DL model and DL+ age-cutoff model (0.945 vs. 0.835, P < .001; 0.945 vs. 0.931, P < .001, respectively). With the assistance of DL+ age-tree model, both junior and senior radiologists' AUC had significant improvement (0.746-0.818, P = .010, 0.827-0.860, P = .005, respectively). CONCLUSIONS The DL+ age-tree model based on US images and age showed excellent performance in the differentiation of MBC and FA. Moreover, it can effectively improve the performance of radiologists with different degrees of experience that may contribute to reducing the misdiagnosis of MBC.
Collapse
Affiliation(s)
- Yuan Yao
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Yang Zhao
- The School of Engineering Science, University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Xu Guo
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Xiangli Xu
- The second hospital of Harbin, Harbin, People's Republic of China
| | - Baiyang Fu
- Department of Breast Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Hao Cui
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China
| | - Jian Xue
- The School of Engineering Science, University of Chinese Academy of Science, Beijing, People's Republic of China
| | - Jiawei Tian
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China.
| | - Ke Lu
- The School of Engineering Science, University of Chinese Academy of Science, Beijing, People's Republic of China; Peng Cheng Laboratory, Shenzhen, People's Republic of China.
| | - Lei Zhang
- Department of Ultrasound, The Second Affiliated Hospital of Harbin Medical University, Harbin, People's Republic of China.
| |
Collapse
|
18
|
Tan X, Pan F, Zhan N, Wang S, Dong Z, Li Y, Yang G, Huang B, Duan Y, Xia H, Cao Y, Zhou M, Lv Z, Huang Q, Tian S, Zhang L, Zhou M, Yang L, Jin Y. Multimodal integration to identify the invasion status of lung adenocarcinoma intraoperatively. iScience 2024; 27:111421. [PMID: 39687006 PMCID: PMC11647133 DOI: 10.1016/j.isci.2024.111421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 08/30/2024] [Accepted: 11/15/2024] [Indexed: 12/18/2024] Open
Abstract
Evaluating the invasiveness of lung adenocarcinoma is crucial for determining the appropriate surgical strategy, impacting postoperative outcomes. This study developed a multimodality model combining radiomics, intraoperative frozen section (FS) pathology, and clinical indicators to predict invasion status. The study enrolled 1,424 patients from two hospitals, divided into multimodal training, radiology testing, and pathology testing cohorts. A prospective validation cohort of 114 patients was selected between March and May 2023. The radiomics + pathology + clinical indicators multimodality model (multi-RPC model) achieved an area under the curve (AUC) of 0.921 (95% confidence interval [CI] 0.899-0.939) in the multimodal training cohort and 0.939 (95% CI 0.878-0.975) in the validation cohort, outperforming single- and dual-modality models. The multi-RPC model's predictive accuracy of 0.860 (95% CI 0.782-0.918) suggests that it could significantly reduce inappropriate surgical procedures, enhancing precision oncology by integrating multimodal information to guide surgical decisions.
Collapse
Affiliation(s)
- Xueyun Tan
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Feng Pan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Na Zhan
- Department of Pathology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Sufei Wang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zegang Dong
- Sino-US Telemed (Wuhan) Co., Ltd, Wuhan 430064, China
| | - Yan Li
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Guanghai Yang
- Department of Thoracic Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Bo Huang
- Department of Pathology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yanran Duan
- Department of Epidemiology and Biostatistics, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Hui Xia
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yaqi Cao
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Min Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Zhilei Lv
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Qi Huang
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shan Tian
- Department of Infectious Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liang Zhang
- Department of Radiology, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Mengmeng Zhou
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yang Jin
- Department of Respiratory and Critical Care Medicine, Hubei Province Clinical Research Center for Major Respiratory Disease, NHC Key Laboratory of Pulmonary Diseases, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Key Laboratory of Biological Targeted Therapy, MOE Key Laboratory of Biological Targeted Therapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
- Hubei Province Engineering Research Center for Tumor-Targeted Biochemotherapy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| |
Collapse
|
19
|
Mo S, Luo H, Wang M, Li G, Kong Y, Tian H, Wu H, Tang S, Pan Y, Wang Y, Xu J, Huang Z, Dong F. Machine learning radiomics based on intra and peri tumor PA/US images distinguish between luminal and non-luminal tumors in breast cancers. PHOTOACOUSTICS 2024; 40:100653. [PMID: 39399393 PMCID: PMC11467668 DOI: 10.1016/j.pacs.2024.100653] [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: 07/14/2024] [Revised: 09/06/2024] [Accepted: 09/20/2024] [Indexed: 10/15/2024]
Abstract
PURPOSE This study aimed to evaluate a radiomics model using Photoacoustic/ultrasound (PA/US) imaging at intra and peri-tumoral area to differentiate Luminal and non-Luminal breast cancer (BC) and to determine the optimal peritumoral area for accurate classification. MATERIALS AND METHODS From February 2022 to April 2024, this study continuously collected 322 patients at Shenzhen People's Hospital, using standardized conditions for PA/US imaging of BC. Regions of interest were delineated using ITK-SNAP, with peritumoral regions of 2 mm, 4 mm, and 6 mm automatically expanded using code from the Pyradiomic package. Feature extraction was subsequently performed using Pyradiomics. The study employed Z-score normalization, Spearman correlation for feature correlation, and LASSO regression for feature selection, validated through 10-fold cross-validation. The radiomics model integrated intra and peri-tumoral area, evaluated by receiver operating characteristic curve(ROC), Calibration and Decision Curve Analysis(DCA). RESULTS We extracted and selected features from intratumoral and peritumoral PA/US images regions at 2 mm, 4 mm, and 6 mm. The comprehensive radiomics model, integrating these regions, demonstrated enhanced diagnostic performance, especially the 4 mm model which showed the highest area under the curve(AUC):0.898(0.78-1.00) and comparably high accuracy (0.900) and sensitivity (0.937). This model outperformed the standalone clinical model and combined clinical-radiomics model in distinguishing between Luminal and non-Luminal BC, as evidenced in the test set results. CONCLUSION This study developed a radiomics model integrating intratumoral and peritumoral at 4 mm region PA/US model, enhancing the differentiation of Luminal from non-Luminal BC. It demonstrated the diagnostic utility of peritumoral characteristics, reducing the need for invasive biopsies and aiding chemotherapy planning, while emphasizing the importance of optimizing tumor surrounding size for improved model accuracy.
Collapse
Affiliation(s)
- Sijie Mo
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Hui Luo
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Mengyun Wang
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Guoqiu Li
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Yao Kong
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Hongtian Tian
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Huaiyu Wu
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Shuzhen Tang
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Yinhao Pan
- Mindray Bio-Medical Electronics Co.,Ltd., ShenZhen 518057,China
| | - Youping Wang
- Department of Clinical and Research, Shenzhen Mindray Bio-medical Electronics Co., Ltd., Shenzhen, China
| | - Jinfeng Xu
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Zhibin Huang
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
| | - Fajin Dong
- Department of Ultrasound, The Second Clinical Medical College, Jinan University, Guangdong 518020, China
- Department of Ultrasound, Shenzhen People’s Hospital, Guangdong 518020, China
- Department of Ultrasound, The First Affiliated Hospital, Southern University of Science and Technology, Shenzhen, Guangdong 518020, China
| |
Collapse
|
20
|
Dai X, Lu H, Wang X, Zhao B, Liu Z, Sun T, Gao F, Xie P, Yu H, Sui X. Development of ultrasound-based clinical, radiomics and deep learning fusion models for the diagnosis of benign and malignant soft tissue tumors. Front Oncol 2024; 14:1443029. [PMID: 39600644 PMCID: PMC11588752 DOI: 10.3389/fonc.2024.1443029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 10/16/2024] [Indexed: 11/29/2024] Open
Abstract
Objectives The aim of this study is to develop an ultrasound-based fusion model of clinical, radiomics and deep learning (CRDL) for accurate diagnosis of benign and malignant soft tissue tumors (STTs). Methods In this retrospective study, ultrasound images and clinical data of patients with STTs from two hospitals were collected between January 2021 and December 2023. Radiomics features and deep learning features were extracted from the ultrasound images, and the optimal features were selected to construct fusion models using support vector machines. The predictive performance of the model was evaluated based on three aspects: discrimination, calibration and clinical usefulness. The DeLong test was used to compare whether there was a significant difference in AUC between the models. Finally, two radiologists who were unaware of the clinical information performed an independent diagnosis and a model-assisted diagnosis of the tumor to compare the performance of the two diagnoses. Results A training cohort of 516 patients from Hospital-1 and an external validation cohort of 78 patients from Hospital-2 were included in the study. The Pre-FM CRDL showed the best performance in predicting STTs, with area under the curve (AUC) of 0.911 (95%CI: 0.894-0.928) and 0.948 (95%CI: 0.906-0.990) for training cohort and external validation cohort, respectively. The DeLong test showed that the Pre-FM CRDL significantly outperformed the clinical models (P< 0.05). In addition, the Pre-FM CRDL can improve the diagnostic accuracy of radiologists. Conclusion This study demonstrates the high clinical applicability of the fusion model in the differential diagnosis of STTs.
Collapse
Affiliation(s)
- Xinpeng Dai
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Haiyong Lu
- First Affiliated Hospital of Hebei North University, Zhangjiakou, Hebei, China
| | - Xinying Wang
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Bingxin Zhao
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zongjie Liu
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Tao Sun
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Feng Gao
- Department of Pathology, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Peng Xie
- Department of Nuclear Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, China
| | - Hong Yu
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| | - Xin Sui
- Third Hospital of Hebei Medical University, Shijiazhuang, China
| |
Collapse
|
21
|
Abouelezz HM, El-Kashef DH, Abdеlaziz RR, Nader MA. Tiron enhances the anti-cancer activity of doxorubicin in DMBA-induced breast cancer: Role of Notch signaling/apoptosis/autophagy/oxidative stress. Food Chem Toxicol 2024; 193:114968. [PMID: 39214269 DOI: 10.1016/j.fct.2024.114968] [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: 06/06/2024] [Revised: 08/06/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Existing work intended to investigate the outcomes of the localized mitochondrial antioxidant tiron (TR) alone or in combination with doxorubicin (DOX) in 7,12-dimethylbenz[a]anthracene (DMBA)-induced mammary carcinogenesis in rats and the mechanistic pathways behind these effects. Also, to examine the preventive role of TR against DOX-related cardiotoxicity. 64 female Sprague-Dawley rats were randomly assigned into 8 groups: CTRL, DOX, TR, DMBA, DMBA + DOX, DMBA + TR100, DMBA + TR200, and DMBA + DOX + TR200. Rats received TR (100 and 200 mg/kg), DOX (2mg/kg), and DMBA (7.5 mg/kg) for four consecutive weeks. TR alone or combined with DOX not only inhibited oxidative status-related parameters and Notch pathway proteins but also attenuated proliferation markers, and enhanced apoptosis, and autophagy-related genes. Consistently, the histopathological analysis showed better scores in mammary tissues isolated from groups treated with TR only or combined with DOX. Additionally, TR dramatically decreased relative heart weight, myocardial injury biomarkers, and heart oxidative stress parameters while maintaining the myocardial histological integrity. Here we provided evidence that TR acts via modulating Notch signaling/apoptosis/autophagy/oxidative stress to elicit anti-tumor activity and combination with DOX revealed a higher efficacy as a novel anticancer strategy. Moreover, TR could be a potential cardio-protective candidate during DOX-chemotherapy, possibly via its antioxidant activity.
Collapse
Affiliation(s)
- Hadeer M Abouelezz
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt.
| | - Dalia H El-Kashef
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Rania R Abdеlaziz
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| | - Manar A Nader
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Mansoura University, Mansoura, Egypt
| |
Collapse
|
22
|
Yan Y, Liu Y, Yao J, Sui L, Chen C, Jiang T, Liu X, Wang Y, Ou D, Chen J, Wang H, Feng L, Pan Q, Su Y, Wang Y, Wang L, Zhou L, Xu D. Deep learning-assisted distinguishing breast phyllodes tumours from fibroadenomas based on ultrasound images: a diagnostic study. Br J Radiol 2024; 97:1816-1825. [PMID: 39288312 DOI: 10.1093/bjr/tqae147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 06/25/2024] [Accepted: 08/09/2024] [Indexed: 09/19/2024] Open
Abstract
OBJECTIVES To evaluate the performance of ultrasound-based deep learning (DL) models in distinguishing breast phyllodes tumours (PTs) from fibroadenomas (FAs) and their clinical utility in assisting radiologists with varying diagnostic experiences. METHODS We retrospectively collected 1180 ultrasound images from 539 patients (247 PTs and 292 FAs). Five DL network models with different structures were trained and validated using nodule regions annotated by radiologists on breast ultrasound images. DL models were trained using the methods of transfer learning and 3-fold cross-validation. The model demonstrated the best evaluation index in the 3-fold cross-validation was selected for comparison with radiologists' diagnostic decisions. Two-round reader studies were conducted to investigate the value of DL model in assisting 6 radiologists with different levels of experience. RESULTS Upon testing, Xception model demonstrated the best diagnostic performance (area under the receiver-operating characteristic curve: 0.87; 95% CI, 0.81-0.92), outperforming all radiologists (all P < .05). Additionally, the DL model enhanced the diagnostic performance of radiologists. Accuracy demonstrated improvements of 4%, 4%, and 3% for senior, intermediate, and junior radiologists, respectively. CONCLUSIONS The DL models showed superior predictive abilities compared to experienced radiologists in distinguishing breast PTs from FAs. Utilizing the model led to improved efficiency and diagnostic performance for radiologists with different levels of experience (6-25 years of work). ADVANCES IN KNOWLEDGE We developed and validated a DL model based on the largest available dataset to assist in diagnosing PTs. This model has the potential to allow radiologists to discriminate 2 types of breast tumours which are challenging to identify with precision and accuracy, and subsequently to make more informed decisions about surgical plans.
Collapse
Affiliation(s)
- Yuqi Yan
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
| | - Yuanzhen Liu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Jincao Yao
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Lin Sui
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
| | - Chen Chen
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Tian Jiang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Postgraduate Training Base Alliance of Wenzhou Medical University, Hangzhou, Zhejiang 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Xiaofang Liu
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Yifan Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Di Ou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Jing Chen
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Hui Wang
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Lina Feng
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Qianmeng Pan
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
| | - Ying Su
- Department of Pathology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
| | - Yukai Wang
- Zunyi Medical University, Zunyi 563000, China
| | - Liping Wang
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Lingyan Zhou
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| | - Dong Xu
- Department of Diagnostic Ultrasound Imaging & Interventional Therapy, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou 310022, China
- Wenling Big Data and Artificial Intelligence Institute in Medicine, TaiZhou 317502, China
- Key Laboratory of Head & Neck Cancer Translational Research of Zhejiang Province, Hangzhou 310022, China
- Taizhou Key Laboratory of Minimally Invasive Interventional Therapy & Artificial Intelligence, Taizhou Campus of Zhejiang Cancer Hospital (Taizhou Cancer Hospital), Taizhou 317502, China
- Zhejiang Provincial Research Center for Cancer Intelligent Diagnosis and Molecular Technology, Hangzhou 310022, China
| |
Collapse
|
23
|
Yu P, Wang C, Zhang H, Zheng G, Jia C, Liu Z, Wang Q, Mu Y, Yang X, Mao N, Song X. Deep learning-based automatic pipeline system for predicting lateral cervical lymph node metastasis in patients with papillary thyroid carcinoma using computed tomography: A multi-center study. Chin J Cancer Res 2024; 36:545-561. [PMID: 39539818 PMCID: PMC11555202 DOI: 10.21147/j.issn.1000-9604.2024.05.07] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/18/2024] [Indexed: 11/16/2024] Open
Abstract
Objective The assessment of lateral lymph node metastasis (LLNM) in patients with papillary thyroid carcinoma (PTC) holds great significance. This study aims to develop and evaluate a deep learning-based automatic pipeline system (DLAPS) for diagnosing LLNM in PTC using computed tomography (CT). Methods A total of 1,266 lateral lymph nodes (LLNs) from 519 PTC patients who underwent CT examinations from January 2019 to November 2022 were included and divided into training and validation set, internal test set, pooled external test set, and prospective test set. The DLAPS consists of an auto-segmentation network based on RefineNet model and a classification network based on ensemble model (ResNet, Xception, and DenseNet). The performance of the DLAPS was compared with that of manually segmented DL models, the clinical model, and Node Reporting and Data System (Node-RADS). The improvement of radiologists' diagnostic performance under the DLAPS-assisted strategy was explored. In addition, bulk RNA-sequencing was conducted based on 12 LLNs to reveal the underlying biological basis of the DLAPS. Results The DLAPS yielded good performance with area under the receiver operating characteristic curve (AUC) of 0.872, 0.910, and 0.822 in the internal, pooled external, and prospective test sets, respectively. The DLAPS significantly outperformed clinical models (AUC 0.731, P<0.001) and Node-RADS (AUC 0.602, P<0.001) in the internal test set. Moreover, the performance of the DLAPS was comparable to that of the manually segmented deep learning (DL) model with AUCs ranging 0.814-0.901 in three test sets. Furthermore, the DLAPS-assisted strategy improved the performance of radiologists and enhanced inter-observer consistency. In clinical situations, the rate of unnecessary LLN dissection decreased from 33.33% to 7.32%. Furthermore, the DLAPS was associated with the cell-cell conjunction in the microenvironment. Conclusions Using CT images from PTC patients, the DLAPS could effectively segment and classify LLNs non-invasively, and this system had a good generalization ability and clinical applicability.
Collapse
Affiliation(s)
- Pengyi Yu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| | - Cai Wang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
- Cardiff China Medical Research Collaborative, Cardiff University School of Medicine, Cardiff CF14 4XN, UK
| | - Haicheng Zhang
- Big data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Guibin Zheng
- Department of Thyroid Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Chuanliang Jia
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| | - Zhonglu Liu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| | - Qi Wang
- Big data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Yakui Mu
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| | - Xin Yang
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| | - Ning Mao
- Big data and Artificial Intelligence Laboratory, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Department of Radiology, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
| | - Xicheng Song
- Department of Otorhinolaryngology, Head and Neck Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai 264000, China
- Shandong Provincial Key Laboratory of Neuroimmune Interaction and Regulation, Yantai 264000, China
- Shandong Provincial Clinical Research Center for Otorhinolaryngologic Diseases, Yantai 264000, China
- Yantai Key Laboratory of Otorhinolaryngologic Diseases, Yantai 264000, China
| |
Collapse
|
24
|
Qi YJ, Su GH, You C, Zhang X, Xiao Y, Jiang YZ, Shao ZM. Radiomics in breast cancer: Current advances and future directions. Cell Rep Med 2024; 5:101719. [PMID: 39293402 PMCID: PMC11528234 DOI: 10.1016/j.xcrm.2024.101719] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2024] [Revised: 07/10/2024] [Accepted: 08/14/2024] [Indexed: 09/20/2024]
Abstract
Breast cancer is a common disease that causes great health concerns to women worldwide. During the diagnosis and treatment of breast cancer, medical imaging plays an essential role, but its interpretation relies on radiologists or clinical doctors. Radiomics can extract high-throughput quantitative imaging features from images of various modalities via traditional machine learning or deep learning methods following a series of standard processes. Hopefully, radiomic models may aid various processes in clinical practice. In this review, we summarize the current utilization of radiomics for predicting clinicopathological indices and clinical outcomes. We also focus on radio-multi-omics studies that bridge the gap between phenotypic and microscopic scale information. Acknowledging the deficiencies that currently hinder the clinical adoption of radiomic models, we discuss the underlying causes of this situation and propose future directions for advancing radiomics in breast cancer research.
Collapse
Affiliation(s)
- Ying-Jia Qi
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Xu Zhang
- Department of Radiology, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Yi Xiao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Yi-Zhou Jiang
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China.
| |
Collapse
|
25
|
Cai F, Wen J, He F, Xia Y, Xu W, Zhang Y, Jiang L, Li J. SC-Unext: A Lightweight Image Segmentation Model with Cellular Mechanism for Breast Ultrasound Tumor Diagnosis. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:1505-1515. [PMID: 38424276 PMCID: PMC11300774 DOI: 10.1007/s10278-024-01042-9] [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: 10/27/2023] [Revised: 01/13/2024] [Accepted: 02/05/2024] [Indexed: 03/02/2024]
Abstract
Automatic breast ultrasound image segmentation plays an important role in medical image processing. However, current methods for breast ultrasound segmentation suffer from high computational complexity and large model parameters, particularly when dealing with complex images. In this paper, we take the Unext network as a basis and utilize its encoder-decoder features. And taking inspiration from the mechanisms of cellular apoptosis and division, we design apoptosis and division algorithms to improve model performance. We propose a novel segmentation model which integrates the division and apoptosis algorithms and introduces spatial and channel convolution blocks into the model. Our proposed model not only improves the segmentation performance of breast ultrasound tumors, but also reduces the model parameters and computational resource consumption time. The model was evaluated on the breast ultrasound image dataset and our collected dataset. The experiments show that the SC-Unext model achieved Dice scores of 75.29% and accuracy of 97.09% on the BUSI dataset, and on the collected dataset, it reached Dice scores of 90.62% and accuracy of 98.37%. Meanwhile, we conducted a comparison of the model's inference speed on CPUs to verify its efficiency in resource-constrained environments. The results indicated that the SC-Unext model achieved an inference speed of 92.72 ms per instance on devices equipped only with CPUs. The model's number of parameters and computational resource consumption are 1.46M and 2.13 GFlops, respectively, which are lower compared to other network models. Due to its lightweight nature, the model holds significant value for various practical applications in the medical field.
Collapse
Affiliation(s)
- Fenglin Cai
- Department of Intelligent Technology and Engineering, Chongqing University of Science and Technology, Chongqing, 401331, People's Republic of China
| | - Jiaying Wen
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Fangzhou He
- Department of Intelligent Technology and Engineering, Chongqing University of Science and Technology, Chongqing, 401331, People's Republic of China
| | - Yulong Xia
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Weijun Xu
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Yong Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China
| | - Li Jiang
- Department of Neurosurgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, People's Republic of China.
| | - Jie Li
- Department of Intelligent Technology and Engineering, Chongqing University of Science and Technology, Chongqing, 401331, People's Republic of China.
| |
Collapse
|
26
|
Jiang C, Zhang X, Qu T, Yang X, Xiu Y, Yu X, Zhang S, Qiao K, Meng H, Li X, Huang Y. The prediction of pCR and chemosensitivity for breast cancer patients using DLG3, RADL and Pathomics signatures based on machine learning and deep learning. Transl Oncol 2024; 46:101985. [PMID: 38805774 PMCID: PMC11154003 DOI: 10.1016/j.tranon.2024.101985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2024] [Revised: 04/19/2024] [Accepted: 04/30/2024] [Indexed: 05/30/2024] Open
Abstract
BACKGROUND Limited studies have investigated the predictive value of multiomics signatures (radiomics, deep learning features, pathological features and DLG3) in breast cancer patients who underwent neoadjuvant chemotherapy (NAC). However, no study has explored the relationships among radiomic, pathomic signatures and chemosensitivity. This study aimed to predict pathological complete response (pCR) using multiomics signatures, and to evaluate the predictive utility of radiomic and pathomic signatures for guiding chemotherapy selection. METHODS The oncogenic function of DLG3 was explored in breast cancer cells via DLG3 knockdown. Immunohistochemistry (IHC) was used to evaluate the relationship between DLG3 expression and docetaxel/epirubin sensitivity. Machine learning (ML) and deep learning (DL) algorithms were used to develop multiomics signatures. Survival analysis was conducted by K-M curves and log-rank. Multivariate logistic regression analysis was used to develop nomograms. RESULTS A total of 311 patients with malignant breast tumours who underwent NAC were retrospectively included in this multicentre study. Multiomics (DLG3, RADL and PATHO) signatures could accurately predict pCR (AUC: training: 0.900; testing: 0.814; external validation: 0.792). Its performance is also superior to that of clinical TNM staging and the single RADL signature in different cohorts. Patients in the low DLG3 group more easily achieved pCR, and those in the high RADL Signature_pCR and PATHO_Signature_pCR (OR = 7.93, 95 % CI: 3.49-18, P < 0.001) groups more easily achieved pCR. In the TEC regimen NAC group, patients who achieved pCR had a lower DLG3 score (4.00 ± 2.33 vs. 6.43 ± 3.01, P < 0.05). Patients in the low RADL_Signature_DLG3 and PATHO_Signature_DLG3 groups had lower DLG3 IHC scores (P < 0.05). Patients in the high RADL signature, PATHO signature and DLG3 signature groups had worse DFS and OS. CONCLUSIONS Multiomics signatures (RADL, PATHO and DLG3) demonstrated great potential in predicting the pCR of breast cancer patients who underwent NAC. The RADL and PATHO signatures are associated with DLG3 status and could help doctors or patients choose proper neoadjuvant chemotherapy regimens (TEC regimens). This simple, structured, convenient and inexpensive multiomics model could help clinicians and patients make treatment decisions.
Collapse
Affiliation(s)
- Cong Jiang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - XueFang Zhang
- Department of Pathology, The first people's hospital of Xiangtan City, Xiangtan 411100, China
| | - Tong Qu
- Department of Oncology, The second cancer hospital of Heilongjiang province, Harbin 150086, China
| | - Xinxin Yang
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Yuting Xiu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Xiao Yu
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Shiyuan Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Kun Qiao
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Hongxue Meng
- Department of Pathology, Harbin Medical University Cancer Hospital, Harbin 150086, China
| | - Xuelian Li
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China; State Key Laboratory of Frigid Zone Cardiovascular Diseases (SKLFZCD), Department of Pharmacology (State Key Labratoray -Province Key Laboratories of Biomedicine-Pharmaceutics of China, Key Laboratory of Cardiovascular Research, Ministry of Education), College of Pharmacy, Harbin Medical University, Harbin 150081, China
| | - Yuanxi Huang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin 150086, China.
| |
Collapse
|
27
|
Zhang J, Liu R, Wang X, Zhang S, Shao L, Liu J, Zhao J, Wang Q, Tian J, Lu Y. Deep learning model based on endoscopic images predicting treatment response in locally advanced rectal cancer undergo neoadjuvant chemoradiotherapy: a multicenter study. J Cancer Res Clin Oncol 2024; 150:350. [PMID: 39001926 PMCID: PMC11246300 DOI: 10.1007/s00432-024-05876-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 06/29/2024] [Indexed: 07/15/2024]
Abstract
PURPOSE Neoadjuvant chemoradiotherapy has been the standard practice for patients with locally advanced rectal cancer. However, the treatment response varies greatly among individuals, how to select the optimal candidates for neoadjuvant chemoradiotherapy is crucial. This study aimed to develop an endoscopic image-based deep learning model for predicting the response to neoadjuvant chemoradiotherapy in locally advanced rectal cancer. METHODS In this multicenter observational study, pre-treatment endoscopic images of patients from two Chinese medical centers were retrospectively obtained and a deep learning-based tumor regression model was constructed. Treatment response was evaluated based on the tumor regression grade and was defined as good response and non-good response. The prediction performance of the deep learning model was evaluated in the internal and external test sets. The main outcome was the accuracy of the treatment prediction model, measured by the AUC and accuracy. RESULTS This deep learning model achieved favorable prediction performance. In the internal test set, the AUC and accuracy were 0.867 (95% CI: 0.847-0.941) and 0.836 (95% CI: 0.818-0.896), respectively. The prediction performance was fully validated in the external test set, and the model had an AUC of 0.758 (95% CI: 0.724-0.834) and an accuracy of 0.807 (95% CI: 0.774-0.843). CONCLUSION The deep learning model based on endoscopic images demonstrated exceptional predictive power for neoadjuvant treatment response, highlighting its potential for guiding personalized therapy.
Collapse
Affiliation(s)
- Junhao Zhang
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266003, China
| | - Ruiqing Liu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266003, China
| | - Xujian Wang
- Graduate School for Elite Engineers, Shandong University, Jinan, China
| | - Shiwei Zhang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Lizhi Shao
- CAS Key Laboratory of Molecular Imaging, Institute of Automation, Chinese Academy of Sciences, Beijing, China
| | - Junheng Liu
- Department of Gastroenterology, The Affiliated Hospital of Qingdao University, Qingdao, China
| | - Jiahui Zhao
- Department of Gastroenterology, Endoscopy Center, The First Hospital of Jilin University, Changchun, China
| | - Quan Wang
- Department of Gastric and Colorectal Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, 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, 100191, China.
| | - Yun Lu
- Department of Gastrointestinal Surgery, The Affiliated Hospital of Qingdao University, No. 16, Jiangsu Road, Shinan District, Qingdao, 266003, China.
| |
Collapse
|
28
|
Lu G, Tian R, Yang W, Liu R, Liu D, Xiang Z, Zhang G. Deep learning radiomics based on multimodal imaging for distinguishing benign and malignant breast tumours. Front Med (Lausanne) 2024; 11:1402967. [PMID: 39036101 PMCID: PMC11257849 DOI: 10.3389/fmed.2024.1402967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2024] [Accepted: 06/14/2024] [Indexed: 07/23/2024] Open
Abstract
Objectives This study aimed to develop a deep learning radiomic model using multimodal imaging to differentiate benign and malignant breast tumours. Methods Multimodality imaging data, including ultrasonography (US), mammography (MG), and magnetic resonance imaging (MRI), from 322 patients (112 with benign breast tumours and 210 with malignant breast tumours) with histopathologically confirmed breast tumours were retrospectively collected between December 2018 and May 2023. Based on multimodal imaging, the experiment was divided into three parts: traditional radiomics, deep learning radiomics, and feature fusion. We tested the performance of seven classifiers, namely, SVM, KNN, random forest, extra trees, XGBoost, LightGBM, and LR, on different feature models. Through feature fusion using ensemble and stacking strategies, we obtained the optimal classification model for benign and malignant breast tumours. Results In terms of traditional radiomics, the ensemble fusion strategy achieved the highest accuracy, AUC, and specificity, with values of 0.892, 0.942 [0.886-0.996], and 0.956 [0.873-1.000], respectively. The early fusion strategy with US, MG, and MRI achieved the highest sensitivity of 0.952 [0.887-1.000]. In terms of deep learning radiomics, the stacking fusion strategy achieved the highest accuracy, AUC, and sensitivity, with values of 0.937, 0.947 [0.887-1.000], and 1.000 [0.999-1.000], respectively. The early fusion strategies of US+MRI and US+MG achieved the highest specificity of 0.954 [0.867-1.000]. In terms of feature fusion, the ensemble and stacking approaches of the late fusion strategy achieved the highest accuracy of 0.968. In addition, stacking achieved the highest AUC and specificity, which were 0.997 [0.990-1.000] and 1.000 [0.999-1.000], respectively. The traditional radiomic and depth features of US+MG + MR achieved the highest sensitivity of 1.000 [0.999-1.000] under the early fusion strategy. Conclusion This study demonstrated the potential of integrating deep learning and radiomic features with multimodal images. As a single modality, MRI based on radiomic features achieved greater accuracy than US or MG. The US and MG models achieved higher accuracy with transfer learning than the single-mode or radiomic models. The traditional radiomic and depth features of US+MG + MR achieved the highest sensitivity under the early fusion strategy, showed higher diagnostic performance, and provided more valuable information for differentiation between benign and malignant breast tumours.
Collapse
Affiliation(s)
- Guoxiu Lu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| | - Ronghui Tian
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Wei Yang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, Liaoning, China
| | - Ruibo Liu
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, Liaoning, China
| | - Dongmei Liu
- Department of Ultrasound, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zijie Xiang
- Biomedical Engineering, Shenyang University of Technology, Shenyang, Liaoning, China
| | - Guoxu Zhang
- Department of Nuclear Medicine, General Hospital of Northern Theater Command, Shenyang, Liaoning, China
| |
Collapse
|