1
|
Wen G, Niu S, Mei S, Wang S. The cancer stem cells characteristics analysis of LGR5 + cells that influence lung cancer risk by using single cell RNA-seq analysis. Sci Rep 2025; 15:16085. [PMID: 40341189 PMCID: PMC12062498 DOI: 10.1038/s41598-025-00585-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 04/29/2025] [Indexed: 05/10/2025] Open
Abstract
Lung adenocarcinoma (LUAD) is the most popular lung cancer type with highly mortality. We performed a single cell RNA-seq analysis to explore characteristic of cancer stem cells in LUAD. We downloaded the single cell RNA-seq data (GSE149655) from the GEO database, the scRNA-seq analysis was performed by using the "Seurat" and "harmony" R package. The FindMarkers function and "ClusterProlifer" package was used for differentially expressed genes (DEGs) and function enrichment analysis. The protein-protein interaction and transcriptional regulatory network were performed by STRING and ChIPBase database. Immunohistochemistry tests to be used to observe differences in the expression of specific genes in LUAD and paracancerous tissue samples. BEAS-2B and A549 cells was used for vitro assay and the qRT-PCR, western blotting, wound healing, trans-well assays, EdU tests, and flow cytometry were performed. A total of 9 cell clusters were obtained after scRNA-seq analysis, in which the cancer stem cells had higher proportion in LUAD samples. Subsequently, function enrichment analysis revealed that the amino sugar and nucleotide sugar metabolism and DNA replication pathways were activated in cancer stem cells (CSCs), which were further sub-divided into 3 subtypes, the LGR5 + stem cell is a major contributor to cancer progression, its hub genes, such as HLA-DPB1, CD74, CTSH and HLA-DRB5 mediated the unique transcriptional state. In addition, the marker genes of three CSCs were also overexpressed in LUAD cells and the CXCL3 played an important role in mediating cell proliferation, apoptosis, migration and invasion of tumor. We performed a scRNA-seq analysis and identified the LGR5 + stem cell as a major contributor in LUAD progression, our findings are expected to provide new insights into the pathogenesis of LUAD.
Collapse
Affiliation(s)
- Ge Wen
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
- Department of Radiation Oncology, Guangdong Provincial Key Laboratory of Major Obstetric Diseases, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital, Guangzhou Medical University, Guangzhou, 510150, China
| | - Shaoqing Niu
- Department of Radiation Oncology, First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510080, China
| | - Shiqi Mei
- Department of Oncology, The Second School of Clinical Medicine, Southern Medical University, Guangzhou, 510280, China
| | - Senming Wang
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China.
| |
Collapse
|
2
|
Su R, Zhang Y, Li X, Li X, Zhang H, Huang X, Liu X, Li P. CT-based Machine Learning Radiomics Modeling: Survival Prediction and Mechanism Exploration in Ovarian Cancer Patients. Acad Radiol 2025; 32:2763-2775. [PMID: 39827000 DOI: 10.1016/j.acra.2024.12.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 12/18/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
RATIONALE AND OBJECTIVES To create a radiomics model based on computed tomography (CT) to predict overall survival in ovarian cancer patients. To combine Rad-score with genomic data to explore the association between gene expression and Rad-score. MATERIALS AND METHODS Imaging and clinical data from 455 patients with ovarian cancer were retrospectively analyzed. Patients were categorized into training cohort, validation cohort and test cohort. Cox regression analysis and the least absolute shrinkage and selection operator (LASSO) methods were utilized to identify characteristics and develop the Rad-score. Radiomics models were developed and evaluated for predictive efficacy and clinical incremental value. Application of genomic data from the cancer genome atlas (TCGA) to reveal differential genes in different Rad-score groups. Screening hub genes and exploring the functions of hub genes through bioinformatics analysis and machine learning. RESULTS Prognostic models based on FIGO, tumor residual disease and Rad-score were developed. The receiver operating characteristic (ROC) curves showed that the 1, 3, and 5 year area under curves (AUCs) of the model were in the training group (0.816, 0.865 and 0.862, respectively), validation group (0.845, 0.877, 0.869, respectively) and test group (0.899, 0.906 and 0.869, respectively) had good predictive accuracy. Calibration curves showed good agreement between observations and predictions. Decision curve analysis revealed a high net benefit of the clinical-radiomics model. The clinical impact curve (CIC) showed good clinical applicability of the clinical-radiomics model. Analysis of sequencing data from the TCGA database revealed EMP1 as a hub gene for radiomics modeling. It revealed that its biological function may be associated with extracellular matrix organization and focal adhesion. CONCLUSION Prognostic models based on FIGO, Tumor residual disease, and Rad-score can effectively predict the overall survival (OS) of ovarian cancer patients. Rad-score may enable prognostic prediction of ovarian cancer patients by revealing the expression level of EMP1 and its biological function.
Collapse
Affiliation(s)
- Rixin Su
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Yu Zhang
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Xueya Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Xiaoqin Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Huihui Zhang
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Xiaoyu Huang
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.)
| | - Xudong Liu
- Department of Medical Oncology, The First Affiliated Hospital of University of Science and Technology of China (USTC), Division of Life Sciences and Medicine, University of Science and Technology of China, Anhui Provincial Cancer Hospital, Hefei 230031, China (X.L.)
| | - Ping Li
- Department of Chinese Integrative Medicine Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei 230022, China (R.S., Y.Z., X.L., X.L., H.Z., X.H., P.L.); Department of Integrated Traditional Chinese and Western Medicine, Anhui Medical University, Hefei 230022, China (P.L.).
| |
Collapse
|
3
|
Guan J, Fan M, Li L. MVNMF: Multiview nonnegative matrix factorization for radio-multigenomic analysis in breast cancer prognosis. Med Image Anal 2025; 103:103566. [PMID: 40288334 DOI: 10.1016/j.media.2025.103566] [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: 08/07/2024] [Revised: 02/16/2025] [Accepted: 03/25/2025] [Indexed: 04/29/2025]
Abstract
Radiogenomic research provides a deeper understanding of breast cancer biology by investigating the correlations between imaging phenotypes and genetic data. However, current radiogenomic research primarily focuses on the correlation between imaging phenotypes and single-genomic data (e.g., gene expression data), overlooking the potential of multi-genomics data to unveil more nuances in cancer characterization. To this end, we propose a multiview nonnegative matrix factorization (MVNMF) method for the radio-multigenomic analysis that identifies dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) features associated with multi-genomics data, including DNA copy number alterations, mutations, and mRNAs, each of which is independently predictive of cancer outcomes. MVNMF incorporates subspace learning and multiview regularization into a unified model to simultaneously select features and explore correlations. Subspace learning is utilized to identify representative radiomic features crucial for tumor analysis, while multiview regularization enables the learning of the correlation between the identified radiomic features and multi-genomics data. Experimental results showed that, for overall survival prediction in breast cancer, MVNMF classified patients into two distinct groups characterized by significant differences in survival (p = 0.0012). Furthermore, it achieved better performance with a C-index of 0.698 compared to the method without considering any genomics data (C-index = 0.528). MVNMF is an effective framework for identifying radiomic features linked to multi-genomics data, which improves its predictive power and provides a better understanding of the biological mechanisms underlying observed phenotypes. MVNMF offers a novel framework for prognostic prediction in breast cancer, with the potential to catalyze further radiogenomic/radio-multigenomic studies.
Collapse
Affiliation(s)
- Jian Guan
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; College of Mathematics and Data Science, Minjiang University, Fuzhou 350121, China
| | - Ming Fan
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou 310018, China; Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou 310018, China.
| |
Collapse
|
4
|
Deng M, Lin Y, Yan L, Chen C, Fei Z, Ding J. A bibliometric analysis of nasopharyngeal carcinoma radiomics: trends and insights. Front Oncol 2025; 15:1506778. [PMID: 40201350 PMCID: PMC11975905 DOI: 10.3389/fonc.2025.1506778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Background Nasopharyngeal carcinoma (NPC) is a malignant tumor characterized by distinct geographic and pathological features. Enhancing diagnostic accuracy and timeliness in NPC is crucial for clinical implications. Radiomics has demonstrated significant potential in the clinical management of NPC. Nonetheless, a paucity of bibliometric studies has systematically examined the existing literature in th is domain. The objective of this study was to assess the current landscape and project future trends in NPC research. Methods This study conducted a search on English-language literature concerning the application of radiomics within the field of nasopharyngeal carcinoma (NPC) research from January 2015 to July 1, 2024, utilizing the Web of Science Core Collection (WoSCC) database. Bibliometric and visual analyses were performed using VOSviewer and CiteSpace software on publications related to countries/regions, authors, journals, references, and keywords. Results A total of 311 documents were retrieved, yielding 229 eligible documents after screening, comprising 209 articles and 20 reviews. Annual publications showed an upward trend, while citations revealed a generally declining trend. Notably, China contributed the most publications (n=175). Tian Jie and Dong Di each published 13 papers, and Zhang B was the most frequently co-cited author. Frontiers in Oncology published the most articles (n=25), and the International Journal of Radiation Oncology Biology Physics had the highest citation count (n=331). Sun Yat-sen University led institutional publications (n=39). The radiomics research in NPC focuses on survival prediction, texture analysis, and distant metastasis, and may guide future research directions. Conclusion The application of radiomics in NRC is growing annually, as indicated by bibliometric analysis. Radiomics has enhanced the precision of preoperative diagnosis, prediction, and prognosis in NRC. Bibliometric findings offer insights into radiomics research trends. However, creating extensive NPC datasets and bridging the research-to-clinical gap pose significant challenges. Future research should focus on these areas to advance the development.
Collapse
Affiliation(s)
| | | | | | | | - Zhaodong Fei
- Department of Radiation Oncology, School of Oncology Clinical Medicine, Fujian Medical
University, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, China
| | - Jianming Ding
- Department of Radiation Oncology, School of Oncology Clinical Medicine, Fujian Medical
University, Fujian Provincial Cancer Hospital, Fuzhou, Fujian, China
| |
Collapse
|
5
|
Zhang X, Zhang X, Zhu J, Yi Z, Cao H, Tang H, Zhang H, Huang G. An MRI Radiogenomic Signature to Characterize the Transcriptional Heterogeneity Associated with Prognosis and Biological Functions in Glioblastoma. FRONT BIOSCI-LANDMRK 2025; 30:36348. [PMID: 40152396 DOI: 10.31083/fbl36348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Revised: 02/05/2025] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND The study sought to establish a radiogenomic signature to evaluate the transcriptional heterogeneity that reflects the prognosis and tumour-related biological functions of patients with glioblastoma. METHODS Transcriptional subclones were identified via fully unsupervised deconvolution of RNA sequencing. A genomic prognostic risk score was developed from transcriptional subclone proportions in the development dataset (n = 532) and independently verified in the testing dataset (n = 225). Multimodal magnetic resonance imaging (MRI) analysis involved feature extraction from three distinct anatomical regions across four imaging sequences. Key features were selected to construct a radiogenomic signature predictive of the genomic risk score in the radiogenomic dataset (n = 99), with subsequent survival analysis conducted in the image testing dataset (n = 233). RESULTS A total of 8 transcriptional subclones were identified, of which the metabolic pathway subclone and spinocerebellar ataxia subclone were independent risk factors for overall survival. The genomic risk score effectively differentiated patient subgroups with divergent survival outcomes in both development (p < 0.001) and testing datasets (p = 0.0003). Nineteen radiomic features were selected to construct a radiogenomic signature, with these features being linked to hallmark cancer pathways and the malignant behaviours of cancer cells. The radiogenomic signature predicted overall survival in the image testing dataset (hazard ratios (HR) = 1.67, p = 0.011). CONCLUSIONS A prognostic radiogenomic signature was established and verified to characterize transcriptional subclones with underlying biological functions in glioblastoma.
Collapse
Affiliation(s)
- Xiaoqing Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Xiaoyu Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Jie Zhu
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, 510620 Guangzhou, Guangdong, China
| | - Zhuoya Yi
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, 510620 Guangzhou, Guangdong, China
| | - Huijiao Cao
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Hailin Tang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Huan Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| | - Guoxian Huang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, 510060 Guangzhou, Guangdong, China
| |
Collapse
|
6
|
Khanyile R, Chipiti T, Hull R, Dlamini Z. Radiogenomic Landscape of Metastatic Endocrine-Positive Breast Cancer Resistant to Aromatase Inhibitors. Cancers (Basel) 2025; 17:808. [PMID: 40075655 PMCID: PMC11899325 DOI: 10.3390/cancers17050808] [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: 12/07/2024] [Revised: 02/11/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
Breast cancer poses a significant global health challenge and includes various subtypes, such as endocrine-positive, HER2-positive, and triple-negative. Endocrine-positive breast cancer, characterized by estrogen and progesterone receptors, is commonly treated with aromatase inhibitors. However, resistance to these inhibitors can hinder patient outcomes due to genetic and epigenetic alterations, mutations in the estrogen receptor 1 gene, and changes in signaling pathways. Radiogenomics combines imaging techniques like MRI and CT scans with genomic profiling methods to identify radiographic biomarkers associated with resistance. This approach enhances our understanding of resistance mechanisms and metastasis patterns, linking them to specific genomic profiles and common metastasis sites like the bone and brain. By integrating radiogenomic data, personalized treatment strategies can be developed, improving predictive and prognostic capabilities. Advancements in imaging and genomic technologies offer promising avenues for enhancing radiogenomic research. A thorough understanding of resistance mechanisms is crucial for developing effective treatment strategies, making radiogenomics a valuable integrative approach in personalized medicine that aims to improve clinical outcomes for patients with metastatic endocrine-positive breast cancer.
Collapse
Affiliation(s)
- Richard Khanyile
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.K.); (T.C.); (R.H.)
- Department of Medical Oncology, Steve Biko Academic Hospital and University of Pretoria, Pretoria 0001, South Africa
| | - Talent Chipiti
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.K.); (T.C.); (R.H.)
| | - Rodney Hull
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.K.); (T.C.); (R.H.)
| | - Zodwa Dlamini
- SAMRC Precision Oncology Research Unit (PORU), DSI/NRF SARChI Chair in Precision Oncology and Cancer Prevention (POCP), Pan African Cancer Research Institute (PACRI), University of Pretoria, Hatfield 0028, South Africa; (R.K.); (T.C.); (R.H.)
| |
Collapse
|
7
|
Wang X, Lv L, Tang Q, Wang G, Shang E, Zheng H, Zhang L. A feature fusion method based on radiomic features and revised deep features for improving tumor prediction in ultrasound images. Comput Biol Med 2025; 185:109605. [PMID: 39721417 DOI: 10.1016/j.compbiomed.2024.109605] [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: 05/18/2024] [Revised: 12/01/2024] [Accepted: 12/19/2024] [Indexed: 12/28/2024]
Abstract
BACKGROUND Radiomic features and deep features are both vitally helpful for the accurate prediction of tumor information in breast ultrasound. However, whether integrating radiomic features and deep features can improve the prediction performance of tumor information is unclear. METHODS A feature fusion method based on radiomic features and revised deep features was proposed to predict tumor information. Radiomic features were extracted from the tumor region on ultrasound images, and the optimal radiomic features were subsequently selected based on Gini score. Revised deep features, which were extracted using the revised CNN models integrating prior information, were combined with radiomic features to build a logistic regression classifier for tumor prediction. The performance was evaluated using area under the receiver operating characteristic (ROC) curve (AUC). RESULTS The results showed that the proposed feature fusion method (AUC = 0.9845) obtained better prediction performance than that based on radiomic features (AUC = 0.9796) or deep features (AUC = 0.9342). CONCLUSIONS Our results demonstrate that the proposed feature fusion framework integrating the radiomic features and revised deep features is an efficient method to improve the prediction performance of tumor information.
Collapse
Affiliation(s)
- Xianyang Wang
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Linlin Lv
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Qingfeng Tang
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Guangjun Wang
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Enci Shang
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Hang Zheng
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Liangliang Zhang
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China.
| |
Collapse
|
8
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
9
|
Lee J, Yoon JH, Lee E, Lee HY, Jeong S, Park S, Jo YS, Kwak JY. Immune response and mesenchymal transition of papillary thyroid carcinoma reflected in ultrasonography features assessed by radiologists and deep learning. J Adv Res 2024; 62:219-228. [PMID: 37783270 PMCID: PMC11331164 DOI: 10.1016/j.jare.2023.09.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2023] [Revised: 09/07/2023] [Accepted: 09/29/2023] [Indexed: 10/04/2023] Open
Abstract
INTRODUCTION Ultrasonography (US) features of papillary thyroid cancers (PTCs) are used to select nodules for biopsy due to their association with tumor behavior. However, the molecular biological mechanisms that lead to the characteristic US features of PTCs are largely unknown. OBJECTIVES This study aimed to investigate the molecular biological mechanisms behind US features assessed by radiologists and three convolutional neural networks (CNN) through transcriptome analysis. METHODS Transcriptome data from 273 PTC tissue samples were generated and differentially expressed genes (DEGs) were identified according to US feature. Pathway enrichment analyses were also conducted by gene set enrichment analysis (GSEA) and ClusterProfiler according to assessments made by radiologists and three CNNs - CNN1 (ResNet50), CNN2 (ResNet101) and CNN3 (VGG16). Signature gene scores for PTCs were calculated by single-sample GSEA (ssGSEA). RESULTS Individual suspicious US features consistently suggested an upregulation of genes related to immune response and epithelial-mesenchymal transition (EMT). Likewise, PTCs assessed as positive by radiologists and three CNNs showed the coordinate enrichment of similar gene sets with abundant immune and stromal components. However, PTCs assessed as positive by radiologists had the highest number of DEGs, and those assessed as positive by CNN3 had more diverse DEGs and gene sets compared to CNN1 or CNN2. The percentage of PTCs assessed as positive or negative concordantly by radiologists and three CNNs was 85.6% (231/273) and 7.1% (3/273), respectively. CONCLUSION US features assessed by radiologists and CNNs revealed molecular biologic features and tumor microenvironment in PTCs.
Collapse
Affiliation(s)
- Jandee Lee
- Department of Surgery, Open NBI Convergence Technology Research Laboratory, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Jung Hyun Yoon
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University, College of Medicine, Seoul 03722, South Korea
| | - Eunjung Lee
- School of Mathematics and Computing (Computational Science and Engineering), Yonsei University, Seoul 03722, South Korea
| | - Hwa Young Lee
- Department of Surgery, Open NBI Convergence Technology Research Laboratory, Yonsei Cancer Center, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Seonhyang Jeong
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Sunmi Park
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Young Suk Jo
- Department of Internal Medicine, Open NBI Convergence Technology Research Laboratory, Yonsei University College of Medicine, Seoul 03722, South Korea.
| | - Jin Young Kwak
- Department of Radiology, Severance Hospital, Research Institute of Radiological Science, Yonsei University, College of Medicine, Seoul 03722, South Korea.
| |
Collapse
|
10
|
Lauer D, Magnin CY, Kolly LR, Wang H, Brunner M, Chabria M, Cereghetti GM, Gabryś HS, Tanadini-Lang S, Uldry AC, Heller M, Verleden SE, Klein K, Sarbu AC, Funke-Chambour M, Ebner L, Distler O, Maurer B, Gote-Schniering J. Radioproteomics stratifies molecular response to antifibrotic treatment in pulmonary fibrosis. JCI Insight 2024; 9:e181757. [PMID: 39012714 PMCID: PMC11383602 DOI: 10.1172/jci.insight.181757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2024] [Accepted: 06/26/2024] [Indexed: 07/18/2024] Open
Abstract
Antifibrotic therapy with nintedanib is the clinical mainstay in the treatment of progressive fibrosing interstitial lung disease (ILD). High-dimensional medical image analysis, known as radiomics, provides quantitative insights into organ-scale pathophysiology, generating digital disease fingerprints. Here, we performed an integrative analysis of radiomic and proteomic profiles (radioproteomics) to assess whether changes in radiomic signatures can stratify the degree of antifibrotic response to nintedanib in (experimental) fibrosing ILD. Unsupervised clustering of delta radiomic profiles revealed 2 distinct imaging phenotypes in mice treated with nintedanib, contrary to conventional densitometry readouts, which showed a more uniform response. Integrative analysis of delta radiomics and proteomics demonstrated that these phenotypes reflected different treatment response states, as further evidenced on transcriptional and cellular levels. Importantly, radioproteomics signatures paralleled disease- and drug-related biological pathway activity with high specificity, including extracellular matrix (ECM) remodeling, cell cycle activity, wound healing, and metabolic activity. Evaluation of the preclinical molecular response-defining features, particularly those linked to ECM remodeling, in a cohort of nintedanib-treated fibrosing patients with ILD, accurately stratified patients based on their extent of lung function decline. In conclusion, delta radiomics has great potential to serve as a noninvasive and readily accessible surrogate of molecular response phenotypes in fibrosing ILD. This could pave the way for personalized treatment strategies and improved patient outcomes.
Collapse
Affiliation(s)
- David Lauer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Cheryl Y Magnin
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Luca R Kolly
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Huijuan Wang
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Switzerland
| | - Matthias Brunner
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Mamta Chabria
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
| | - Grazia M Cereghetti
- Department of Diagnostic, Interventional, and Pediatric Radiology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Hubert S Gabryś
- Department of Radiation Oncology, University Hospital Zurich, Zurich, Switzerland
| | | | - Anne-Christine Uldry
- Proteomics & Mass Spectrometry Core Facility (PMSCF), DBMR, University of Bern, Bern, Switzerland
| | - Manfred Heller
- Proteomics & Mass Spectrometry Core Facility (PMSCF), DBMR, University of Bern, Bern, Switzerland
| | - Stijn E Verleden
- Department of ASTARC, University of Antwerp, Antwerp, Wilrijk, Belgium
| | - Kerstin Klein
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Adela-Cristina Sarbu
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
| | - Manuela Funke-Chambour
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Lukas Ebner
- Department of Health Sciences and Technology, ETH Zurich, Zurich, Switzerland
- Department of Radiology, Cantonal Hospital Lucerne, Luzern, Switzerland
- Institute for Radiology, Hirslanden Bern Klinik Beau-Site, Bern, Switzerland
| | - Oliver Distler
- Department of Rheumatology, Center of Experimental Rheumatology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Britta Maurer
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
| | - Janine Gote-Schniering
- Department of Rheumatology and Immunology, Inselspital, Bern University Hospital, and
- Lung Precision Medicine (LPM), Department for BioMedical Research (DBMR), University of Bern, Bern, Switzerland
- Department of Pulmonary Medicine, Allergology and Clinical Immunology, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| |
Collapse
|
11
|
Wang M, Peng Y, Wang Y, Luo D. Research Trends and Evolution in Radiogenomics (2005-2023): Bibliometric Analysis. Interact J Med Res 2024; 13:e51347. [PMID: 38980713 PMCID: PMC11267093 DOI: 10.2196/51347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 03/10/2024] [Accepted: 05/20/2024] [Indexed: 07/10/2024] Open
Abstract
BACKGROUND Radiogenomics is an emerging technology that integrates genomics and medical image-based radiomics, which is considered a promising approach toward achieving precision medicine. OBJECTIVE The aim of this study was to quantitatively analyze the research status, dynamic trends, and evolutionary trajectory in the radiogenomics field using bibliometric methods. METHODS The relevant literature published up to 2023 was retrieved from the Web of Science Core Collection. Excel was used to analyze the annual publication trend. VOSviewer was used for constructing the keywords co-occurrence network and the collaboration networks among countries and institutions. CiteSpace was used for citation keywords burst analysis and visualizing the references timeline. RESULTS A total of 3237 papers were included and exported in plain-text format. The annual number of publications showed an increasing annual trend. China and the United States have published the most papers in this field, with the highest number of citations in the United States and the highest average number per item in the Netherlands. Keywords burst analysis revealed that several keywords, including "big data," "magnetic resonance spectroscopy," "renal cell carcinoma," "stage," and "temozolomide," experienced a citation burst in recent years. The timeline views demonstrated that the references can be categorized into 8 clusters: lower-grade glioma, lung cancer histology, lung adenocarcinoma, breast cancer, radiation-induced lung injury, epidermal growth factor receptor mutation, late radiotherapy toxicity, and artificial intelligence. CONCLUSIONS The field of radiogenomics is attracting increasing attention from researchers worldwide, with the United States and the Netherlands being the most influential countries. Exploration of artificial intelligence methods based on big data to predict the response of tumors to various treatment methods represents a hot spot research topic in this field at present.
Collapse
Affiliation(s)
- Meng Wang
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Yun Peng
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Ya Wang
- Department of Radiology, The Second Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
| | - Dehong Luo
- Department of Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| |
Collapse
|
12
|
Wu CT, Du D, Chen L, Dai R, Liu C, Yu G, Bhardwaj S, Parker SJ, Zhang Z, Clarke R, Herrington DM, Wang Y. CAM3.0: determining cell type composition and expression from bulk tissues with fully unsupervised deconvolution. Bioinformatics 2024; 40:btae107. [PMID: 38407991 PMCID: PMC10924278 DOI: 10.1093/bioinformatics/btae107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 01/13/2024] [Accepted: 02/25/2024] [Indexed: 02/28/2024] Open
Abstract
MOTIVATION Complex tissues are dynamic ecosystems consisting of molecularly distinct yet interacting cell types. Computational deconvolution aims to dissect bulk tissue data into cell type compositions and cell-specific expressions. With few exceptions, most existing deconvolution tools exploit supervised approaches requiring various types of references that may be unreliable or even unavailable for specific tissue microenvironments. RESULTS We previously developed a fully unsupervised deconvolution method-Convex Analysis of Mixtures (CAM), that enables estimation of cell type composition and expression from bulk tissues. We now introduce CAM3.0 tool that improves this framework with three new and highly efficient algorithms, namely, radius-fixed clustering to identify reliable markers, linear programming to detect an initial scatter simplex, and a smart floating search for the optimum latent variable model. The comparative experimental results obtained from both realistic simulations and case studies show that the CAM3.0 tool can help biologists more accurately identify known or novel cell markers, determine cell proportions, and estimate cell-specific expressions, complementing the existing tools particularly when study- or datatype-specific references are unreliable or unavailable. AVAILABILITY AND IMPLEMENTATION The open-source R Scripts of CAM3.0 is freely available at https://github.com/ChiungTingWu/CAM3/(https://github.com/Bioconductor/Contributions/issues/3205). A user's guide and a vignette are provided.
Collapse
Affiliation(s)
- Chiung-Ting Wu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Dongping Du
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Lulu Chen
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, United States
| | - Guoqiang Yu
- Department of Automation, Tsinghua University, Beijing 100084, P. R. China
| | - Saurabh Bhardwaj
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
- Department of Electrical and Instrumentation Engineering, Thapar Institute of Engineering & Technology, Punjab 147004, India
| | - Sarah J Parker
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, United States
| | - Zhen Zhang
- Department of Pathology, Johns Hopkins University, Baltimore, MD 21231, United States
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, United States
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University, Winston-Salem, NC 27157, United States
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, United States
| |
Collapse
|
13
|
Zhang L, Fan M, Li L. Deconvolution-Based Pharmacokinetic Analysis to Improve the Prediction of Pathological Information of Breast Cancer. JOURNAL OF IMAGING INFORMATICS IN MEDICINE 2024; 37:13-24. [PMID: 38343210 PMCID: PMC10976965 DOI: 10.1007/s10278-023-00915-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 08/30/2023] [Accepted: 09/01/2023] [Indexed: 03/02/2024]
Abstract
Pharmacokinetic (PK) parameters, revealing changes in the tumor microenvironment, are related to the pathological information of breast cancer. Tracer kinetic models (e.g., Tofts-Kety model) with a nonlinear least square solver are commonly used to estimate PK parameters. However, the method is sensitive to noise in images. To relieve the effects of noise, a deconvolution (DEC) method, which was validated on synthetic concentration-time series, was proposed to accurately calculate PK parameters from breast dynamic contrast-enhanced magnetic resonance imaging. A time-to-peak-based tumor partitioning method was used to divide the whole tumor into three tumor subregions with different kinetic patterns. Radiomic features were calculated from the tumor subregion and whole tumor-based PK parameter maps. The optimal features determined by the fivefold cross-validation method were used to build random forest classifiers to predict molecular subtypes, Ki-67, and tumor grade. The diagnostic performance evaluated by the area under the receiver operating characteristic curve (AUC) was compared between the subregion and whole tumor-based PK parameters. The results showed that the DEC method obtained more accurate PK parameters than the Tofts method. Moreover, the results showed that the subregion-based Ktrans (best AUCs = 0.8319, 0.7032, 0.7132, 0.7490, 0.8074, and 0.6950) achieved a better diagnostic performance than the whole tumor-based Ktrans (AUCs = 0.8222, 0.6970, 0.6511, 0.7109, 0.7620, and 0.5894) for molecular subtypes, Ki-67, and tumor grade. These findings indicate that DEC-based Ktrans in the subregion has the potential to accurately predict molecular subtypes, Ki-67, and tumor grade.
Collapse
Affiliation(s)
- Liangliang Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China
- School of Computer and Information, Anqing Normal University, Anqing, 246133, China
| | - Ming Fan
- Institute of Intelligent Biomedicine, School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, China.
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, China.
- Institute of Intelligent Biomedicine, School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, China.
| |
Collapse
|
14
|
Yang Y, Xiang T, Lv X, Li L, Lui LM, Zeng T. Double Transformer Super-Resolution for Breast Cancer ADC Images. IEEE J Biomed Health Inform 2024; 28:917-928. [PMID: 38079366 DOI: 10.1109/jbhi.2023.3341250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Diffusion-weighted imaging (DWI) has been extensively explored in guiding the clinic management of patients with breast cancer. However, due to the limited resolution, accurately characterizing tumors using DWI and the corresponding apparent diffusion coefficient (ADC) is still a challenging problem. In this paper, we aim to address the issue of super-resolution (SR) of ADC images and evaluate the clinical utility of SR-ADC images through radiomics analysis. To this end, we propose a novel double transformer-based network (DTformer) to enhance the resolution of ADC images. More specifically, we propose a symmetric U-shaped encoder-decoder network with two different types of transformer blocks, named as UTNet, to extract deep features for super-resolution. The basic backbone of UTNet is composed of a locally-enhanced Swin transformer block (LeSwin-T) and a convolutional transformer block (Conv-T), which are responsible for capturing long-range dependencies and local spatial information, respectively. Additionally, we introduce a residual upsampling network (RUpNet) to expand image resolution by leveraging initial residual information from the original low-resolution (LR) images. Extensive experiments show that DTformer achieves superior SR performance. Moreover, radiomics analysis reveals that improving the resolution of ADC images is beneficial for tumor characteristic prediction, such as histological grade and human epidermal growth factor receptor 2 (HER2) status.
Collapse
|
15
|
Lv T, Hong X, Liu Y, Miao K, Sun H, Li L, Deng C, Jiang C, Pan X. AI-powered interpretable imaging phenotypes noninvasively characterize tumor microenvironment associated with diverse molecular signatures and survival in breast cancer. COMPUTER METHODS AND PROGRAMS IN BIOMEDICINE 2024; 243:107857. [PMID: 37865058 DOI: 10.1016/j.cmpb.2023.107857] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 08/23/2023] [Accepted: 10/08/2023] [Indexed: 10/23/2023]
Abstract
BACKGROUND AND OBJECTIVES Tumor microenvironment (TME) is a determining factor in decision-making and personalized treatment for breast cancer, which is highly intra-tumor heterogeneous (ITH). However, the noninvasive imaging phenotypes of TME are poorly understood, even invasive genotypes have been largely known in breast cancer. METHODS Here, we develop an artificial intelligence (AI)-driven approach for noninvasively characterizing TME by integrating the predictive power of deep learning with the explainability of human-interpretable imaging phenotypes (IMPs) derived from 4D dynamic imaging (DCE-MRI) of 342 breast tumors linked to genomic and clinical data, which connect cancer phenotypes to genotypes. An unsupervised dual-attention deep graph clustering model (DGCLM) is developed to divide bulk tumor into multiple spatially segregated and phenotypically consistent subclusters. The IMPs ranging from spatial heterogeneity to kinetic heterogeneity are leveraged to capture architecture, interaction, and proximity between intratumoral subclusters. RESULTS We demonstrate that our IMPs correlate with well-known markers of TME and also can predict distinct molecular signatures, including expression of hormone receptor, epithelial growth factor receptor and immune checkpoint proteins, with the performance of accuracy, reliability and transparency superior to recent state-of-the-art radiomics and 'black-box' deep learning methods. Moreover, prognostic value is confirmed by survival analysis accounting for IMPs. CONCLUSIONS Our approach provides an interpretable, quantitative, and comprehensive perspective to characterize TME in a noninvasive and clinically relevant manner.
Collapse
Affiliation(s)
- Tianxu Lv
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, China.
| | - Xiaoyan Hong
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, China.
| | - Yuan Liu
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, China.
| | - Kai Miao
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China
| | - Heng Sun
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China.
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, China.
| | - Chuxia Deng
- Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China.
| | - Chunjuan Jiang
- Department of Nuclear Medicine, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China.
| | - Xiang Pan
- School of Artificial Intelligence and Computer Science, Jiangnan University, Wuxi 214122, China; Cancer Center, Faculty of Health Sciences, University of Macau, Macau SAR, China; MOE Frontier Science Centre for Precision Oncology, University of Macau, Macau SAR, China; Shanghai Key Laboratory of Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai 201318, China.
| |
Collapse
|
16
|
Zhang L, Fan M, Li L. Efficient estimation of pharmacokinetic parameters from breast dynamic contrast-enhanced MRI based on a convolutional neural network for predicting molecular subtypes. Phys Med Biol 2023; 68:245001. [PMID: 37983902 DOI: 10.1088/1361-6560/ad0e39] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2023] [Accepted: 11/20/2023] [Indexed: 11/22/2023]
Abstract
Objective. Tracer kinetic models allow for estimating pharmacokinetic (PK) parameters, which are related to pathological characteristics, from breast dynamic contrast-enhanced magnetic resonance imaging. However, existing tracer kinetic models subject to inaccuracy are time-consuming for PK parameters estimation. This study aimed to accurately and efficiently estimate PK parameters for predicting molecular subtypes based on convolutional neural network (CNN).Approach. A CNN integrating global and local features (GL-CNN) was trained using synthetic data where known PK parameters map was used as the ground truth, and subsequently used to directly estimate PK parameters (volume transfer constantKtransand flux rate constantKep) map. The accuracy assessed by the peak signal-to-noise ratio (PSNR), structural similarity (SSIM), and concordance correlation coefficient (CCC) was compared between the GL-CNN and Tofts-based PK parameters in synthetic data. Radiomic features were calculated from the PK parameters map in 208 breast tumors. A random forest classifier was constructed to predict molecular subtypes using a discovery cohort (n= 144). The diagnostic performance evaluated on a validation cohort (n= 64) using the area under the receiver operating characteristic curve (AUC) was compared between the GL-CNN and Tofts-based PK parameters.Main results. The average PSNR (48.8884), SSIM (0.9995), and CCC (0.9995) between the GL-CNN-basedKtransmap and ground truth were significantly higher than those between the Tofts-basedKtransmap and ground truth. The GL-CNN-basedKtransobtained significantly better diagnostic performance (AUCs = 0.7658 and 0.8528) than the Tofts-basedKtransfor luminal B and HER2 tumors. The GL-CNN method accelerated the computation by speed approximately 79 times compared to the Tofts method for the whole breast of all patients.Significance. Our results indicate that the GL-CNN method can be used to accurately and efficiently estimate PK parameters for predicting molecular subtypes.
Collapse
Affiliation(s)
- Liangliang Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
- School of Computer and Information, Anqing Normal University, Anqing, 246133, People's Republic of China
| | - Ming Fan
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| |
Collapse
|
17
|
Lin L, Li H, Wang X, Wang Z, Su G, Zhou J, Sun S, Ma X, Chen Y, You C, Gu Y. Components of the tumor immune microenvironment based on m-IHC correlate with prognosis and subtype of triple-negative breast cancer. Cancer Med 2023; 12:21639-21650. [PMID: 38059408 PMCID: PMC10757132 DOI: 10.1002/cam4.6718] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 08/26/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
BACKGROUND AND AIM The spatial distribution and interactions of cells in the tumor immune microenvironment (TIME) might be related to the different responses of triple-negative breast cancer (TNBC) to immunomodulators. The potential of multiplex IHC (m-IHC) in evaluating the TIME has been reported, but the efficacy is insufficient. We aimed to research whether m-IHC results could be used to reflect the TIME, and thus to predict prognosis and complement the TNBC subtyping system. METHODS The clinical, imaging, and prognosis data for 86 TNBC patients were retrospectively reviewed. CD3, CD4, CD8, Foxp3, PD-L1, and Pan-CK markers were stained by m-IHC. Particular cell spatial distributions and interactions in the TIME were evaluated with the HALO multispectral analysis platform. Then, we calculated the prognostic value of components of the TIME and their correlations with TNBC transcriptomic subtypes and MRI radiomic features reflecting TNBC subtypes. RESULTS The components of the TIME score were established by m-IHC and demonstrated positive prognostic value for TNBC (p = 0.0047, 0.039, <0.0001 for DMFS, RFS, and OS). The score was calculated from several indicators, including Treg% in the tumor core (TC) or stromal area (SA), PD-L1+ cell% in the SA, CD3 + cell% in the TC, and PD-L1+ /CD8+ cells in the invasive margin and SA. According to the TNBC subtyping system, a few TIME indicators were significantly different in different subtypes and significantly correlated with MRI radiomic features reflecting TNBC subtypes. CONCLUSION We demonstrated that the m-IHC-based quantitative score and indicators related to the spatial distribution and interactions of cells in the TIME can aid in the accurate diagnosis of TNBC in terms of prognosis and classification.
Collapse
Affiliation(s)
- Luyi Lin
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Haiming Li
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Xin Wang
- Department of PathologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Zezhou Wang
- Department of Cancer PreventionFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Shanghai Municipal Hospital Oncological Specialist AllianceShanghaiChina
| | - Guanhua Su
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
- Department of Breast Surgery, Key Laboratory of Breast Cancer in ShanghaiFudan University Shanghai Cancer CenterShanghaiChina
| | - Jiayin Zhou
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Shiyun Sun
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Xiaowen Ma
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Yan Chen
- Division of Cancer and Stem CellSchool of Medicine at University of NottinghamNottinghamUK
| | - Chao You
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| | - Yajia Gu
- Department of RadiologyFudan University Shanghai Cancer CenterShanghaiChina
- Department of OncologyShanghai Medical College, Fudan UniversityShanghaiChina
| |
Collapse
|
18
|
Fan M, Wang K, Zhang Y, Ge Y, Lü Z, Li L. Radiogenomic analysis of cellular tumor-stroma heterogeneity as a prognostic predictor in breast cancer. J Transl Med 2023; 21:851. [PMID: 38007511 PMCID: PMC10675940 DOI: 10.1186/s12967-023-04748-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Accepted: 11/22/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND The tumor microenvironment and intercellular communication between solid tumors and the surrounding stroma play crucial roles in cancer initiation, progression, and prognosis. Radiomics provides clinically relevant information from radiological images; however, its biological implications in uncovering tumor pathophysiology driven by cellular heterogeneity between the tumor and stroma are largely unknown. We aimed to identify radiogenomic signatures of cellular tumor-stroma heterogeneity (TSH) to improve breast cancer management and prognosis analysis. METHODS This retrospective multicohort study included five datasets. Cell subpopulations were estimated using bulk gene expression data, and the relative difference in cell subpopulations between the tumor and stroma was used as a biomarker to categorize patients into good- and poor-survival groups. A radiogenomic signature-based model utilizing dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) was developed to target TSH, and its clinical significance in relation to survival outcomes was independently validated. RESULTS The final cohorts of 1330 women were included for cellular TSH biomarker identification (n = 112, mean age, 57.3 years ± 14.6) and validation (n = 886, mean age, 58.9 years ± 13.1), radiogenomic signature of TSH identification (n = 91, mean age, 55.5 years ± 11.4), and prognostic (n = 241) assessments. The cytotoxic lymphocyte biomarker differentiated patients into good- and poor-survival groups (p < 0.0001) and was independently validated (p = 0.014). The good survival group exhibited denser cell interconnections. The radiogenomic signature of TSH was identified and showed a positive association with overall survival (p = 0.038) and recurrence-free survival (p = 3 × 10-4). CONCLUSION Radiogenomic signatures provide insights into prognostic factors that reflect the imbalanced tumor-stroma environment, thereby presenting breast cancer-specific biological implications and prognostic significance.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Kailang Wang
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - You Zhang
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Yuanyuan Ge
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, China
| | - Zhong Lü
- Affiliated Dongyang Hospital of Wenzhou Medical University, Dongyang, 322100, China.
| | - Lihua Li
- Institute of Intelligent Biomedicine, Hangzhou Dianzi University, Hangzhou, 310018, China.
| |
Collapse
|
19
|
Yu X, Xiang J, Zhang Q, Chen S, Tang W, Li X, Sui Y, Liu W, Kong Q, Guo Y. Triple-negative breast cancer: predictive model of early recurrence based on MRI features. Clin Radiol 2023; 78:e798-e807. [PMID: 37596179 DOI: 10.1016/j.crad.2023.07.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/13/2023] [Accepted: 07/18/2023] [Indexed: 08/20/2023]
Abstract
AIM To develop an integrated model based on preoperative magnetic resonance imaging (MRI) features for predicting early recurrence in patients with triple-negative breast cancer (TNBC). MATERIALS AND METHODS Women with TNBC who underwent breast MRI and surgery between 2009 and 2019 were evaluated retrospectively. Two breast radiologists reviewed MRI images independently based on the Breast Imaging Reporting and Data System Lexicon (BI-RADS), and classified the breast oedema scores on T2-weighted imaging (WI) as no oedema, peritumoural oedema, prepectoral oedema, or subcutaneous oedema. The relationship between disease-free survival (DFS) and MRI features was analysed by Cox regression, and a nomogram model was generated based on the results. RESULTS 150 patients with TNBC were included and divided into a training cohort (n=78) and validation cohort (n=72). MRI features including subcutaneous oedema and rim enhancement showed a tendency to worsen DFS in univariate analysis. Multivariate analysis showed that subcutaneous oedema (p=0.049, HR [95% confidence interval {CI} = 8.24 [1.01-67.52]) and rim enhancement (p=0.016, HR [95% CI] = 4.38 [1.32-14.54]) were independent predictors for DFS. In the nomogram, the areas under the curves (AUCs) of the training cohort was 0.808, and that of the validation cohort was 0.875. CONCLUSION The presence of subcutaneous oedema or rim enhancement on preoperative breast MRI was shown to be a good predictor of poor survival outcomes in patients with TNBC.
Collapse
Affiliation(s)
- X Yu
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - J Xiang
- Guangdong Women and Children Hospital, No. 13 West Guangyuan Road, Guangzhou, Guangdong, 510010, China
| | - Q Zhang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - S Chen
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - W Tang
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - X Li
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - Y Sui
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China
| | - W Liu
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| | - Q Kong
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Y Guo
- Department of Radiology, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, 510180, China.
| |
Collapse
|
20
|
Zhang J, Wu J, Zhou XS, Shi F, Shen D. Recent advancements in artificial intelligence for breast cancer: Image augmentation, segmentation, diagnosis, and prognosis approaches. Semin Cancer Biol 2023; 96:11-25. [PMID: 37704183 DOI: 10.1016/j.semcancer.2023.09.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2023] [Revised: 08/03/2023] [Accepted: 09/05/2023] [Indexed: 09/15/2023]
Abstract
Breast cancer is a significant global health burden, with increasing morbidity and mortality worldwide. Early screening and accurate diagnosis are crucial for improving prognosis. Radiographic imaging modalities such as digital mammography (DM), digital breast tomosynthesis (DBT), magnetic resonance imaging (MRI), ultrasound (US), and nuclear medicine techniques, are commonly used for breast cancer assessment. And histopathology (HP) serves as the gold standard for confirming malignancy. Artificial intelligence (AI) technologies show great potential for quantitative representation of medical images to effectively assist in segmentation, diagnosis, and prognosis of breast cancer. In this review, we overview the recent advancements of AI technologies for breast cancer, including 1) improving image quality by data augmentation, 2) fast detection and segmentation of breast lesions and diagnosis of malignancy, 3) biological characterization of the cancer such as staging and subtyping by AI-based classification technologies, 4) prediction of clinical outcomes such as metastasis, treatment response, and survival by integrating multi-omics data. Then, we then summarize large-scale databases available to help train robust, generalizable, and reproducible deep learning models. Furthermore, we conclude the challenges faced by AI in real-world applications, including data curating, model interpretability, and practice regulations. Besides, we expect that clinical implementation of AI will provide important guidance for the patient-tailored management.
Collapse
Affiliation(s)
- Jiadong Zhang
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China
| | - Jiaojiao Wu
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Xiang Sean Zhou
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Feng Shi
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China.
| | - Dinggang Shen
- School of Biomedical Engineering, ShanghaiTech University, Shanghai, China; Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China; Shanghai Clinical Research and Trial Center, Shanghai, China.
| |
Collapse
|
21
|
Su GH, Xiao Y, You C, Zheng RC, Zhao S, Sun SY, Zhou JY, Lin LY, Wang H, Shao ZM, Gu YJ, Jiang YZ. Radiogenomic-based multiomic analysis reveals imaging intratumor heterogeneity phenotypes and therapeutic targets. SCIENCE ADVANCES 2023; 9:eadf0837. [PMID: 37801493 PMCID: PMC10558123 DOI: 10.1126/sciadv.adf0837] [Citation(s) in RCA: 38] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Accepted: 09/06/2023] [Indexed: 10/08/2023]
Abstract
Intratumor heterogeneity (ITH) profoundly affects therapeutic responses and clinical outcomes. However, the widespread methods for assessing ITH based on genomic sequencing or pathological slides, which rely on limited tissue samples, may lead to inaccuracies due to potential sampling biases. Using a newly established multicenter breast cancer radio-multiomic dataset (n = 1474) encompassing radiomic features extracted from dynamic contrast-enhanced magnetic resonance images, we formulated a noninvasive radiomics methodology to effectively investigate ITH. Imaging ITH (IITH) was associated with genomic and pathological ITH, predicting poor prognosis independently in breast cancer. Through multiomic analysis, we identified activated oncogenic pathways and metabolic dysregulation in high-IITH tumors. Integrated metabolomic and transcriptomic analyses highlighted ferroptosis as a vulnerability and potential therapeutic target of high-IITH tumors. Collectively, this work emphasizes the superiority of radiomics in capturing ITH. Furthermore, we provide insights into the biological basis of IITH and propose therapeutic targets for breast cancers with elevated IITH.
Collapse
Affiliation(s)
- Guan-Hua Su
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and 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 and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Chao You
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ren-Cheng Zheng
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai 201203, China
| | - Shen Zhao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Shi-Yun Sun
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Jia-Yin Zhou
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Lu-Yi Lin
- Department of Radiology, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - He Wang
- Institute of Science and Technology for Brain-inspired Intelligence, Fudan University, Shanghai 201203, China
| | - Zhi-Ming Shao
- Key Laboratory of Breast Cancer in Shanghai, Department of Breast Surgery, Fudan University Shanghai Cancer Center and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| | - Ya-Jia Gu
- Department of Radiology, Fudan University Shanghai Cancer Center and 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 and Department of Oncology, Shanghai Medical College, Fudan University, Shanghai 200032, China
| |
Collapse
|
22
|
Liu Z, Duan T, Zhang Y, Weng S, Xu H, Ren Y, Zhang Z, Han X. Radiogenomics: a key component of precision cancer medicine. Br J Cancer 2023; 129:741-753. [PMID: 37414827 PMCID: PMC10449908 DOI: 10.1038/s41416-023-02317-8] [Citation(s) in RCA: 36] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Revised: 05/02/2023] [Accepted: 06/12/2023] [Indexed: 07/08/2023] Open
Abstract
Radiogenomics, focusing on the relationship between genomics and imaging phenotypes, has been widely applied to address tumour heterogeneity and predict immune responsiveness and progression. It is an inevitable consequence of current trends in precision medicine, as radiogenomics costs less than traditional genetic sequencing and provides access to whole-tumour information rather than limited biopsy specimens. By providing voxel-by-voxel genetic information, radiogenomics can allow tailored therapy targeting a complete, heterogeneous tumour or set of tumours. In addition to quantifying lesion characteristics, radiogenomics can also be used to distinguish benign from malignant entities, as well as patient characteristics, to better stratify patients according to disease risk, thereby enabling more precise imaging and screening. Here, we have characterised the radiogenomic application in precision medicine using a multi-omic approach. we outline the main applications of radiogenomics in diagnosis, treatment planning and evaluations in the field of oncology with the aim of developing quantitative and personalised medicine. Finally, we discuss the challenges in the field of radiogenomics and the scope and clinical applicability of these methods.
Collapse
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China
| | - Tian Duan
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuyuan Zhang
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Siyuan Weng
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Hui Xu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Yuqing Ren
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Institute of Zhengzhou University, 450052, Zhengzhou, Henan, China.
- Interventional Treatment and Clinical Research Center of Henan Province, 450052, Zhengzhou, Henan, China.
| |
Collapse
|
23
|
Zhong J, Frood R, McWilliam A, Davey A, Shortall J, Swinton M, Hulson O, West CM, Buckley D, Brown S, Choudhury A, Hoskin P, Henry A, Scarsbrook A. Prediction of prostate tumour hypoxia using pre-treatment MRI-derived radiomics: preliminary findings. LA RADIOLOGIA MEDICA 2023; 128:765-774. [PMID: 37198374 PMCID: PMC10264289 DOI: 10.1007/s11547-023-01644-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 04/26/2023] [Indexed: 05/19/2023]
Abstract
PURPOSE To develop a machine learning (ML) model based on radiomic features (RF) extracted from whole prostate gland magnetic resonance imaging (MRI) for prediction of tumour hypoxia pre-radiotherapy. MATERIAL AND METHODS Consecutive patients with high-grade prostate cancer and pre-treatment MRI treated with radiotherapy between 01/12/2007 and 1/08/2013 at two cancer centres were included. Cancers were dichotomised as normoxic or hypoxic using a biopsy-based 32-gene hypoxia signature (Ragnum signature). Prostate segmentation was performed on axial T2-weighted (T2w) sequences using RayStation (v9.1). Histogram standardisation was applied prior to RF extraction. PyRadiomics (v3.0.1) was used to extract RFs for analysis. The cohort was split 80:20 into training and test sets. Six different ML classifiers for distinguishing hypoxia were trained and tuned using five different feature selection models and fivefold cross-validation with 20 repeats. The model with the highest mean validation area under the curve (AUC) receiver operating characteristic (ROC) curve was tested on the unseen set, and AUCs were compared via DeLong test with 95% confidence interval (CI). RESULTS 195 patients were included with 97 (49.7%) having hypoxic tumours. The hypoxia prediction model with best performance was derived using ridge regression and had a test AUC of 0.69 (95% CI: 0.14). The test AUC for the clinical-only model was lower (0.57), but this was not statistically significant (p = 0.35). The five selected RFs included textural and wavelet-transformed features. CONCLUSION Whole prostate MRI-radiomics has the potential to non-invasively predict tumour hypoxia prior to radiotherapy which may be helpful for individualised treatment optimisation.
Collapse
Affiliation(s)
- Jim Zhong
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK.
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK.
| | - Russell Frood
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Alan McWilliam
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Angela Davey
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Jane Shortall
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Martin Swinton
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Oliver Hulson
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Catharine M West
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
| | - David Buckley
- Biomedical Imaging, Leeds Institute of Cardiovascular and Metabolic Medicine, University of Leeds, Leeds, UK
| | - Sarah Brown
- Leeds Cancer Research UK Clinical Trials Unit, Leeds Institute of Clinical Trials Research (LICTR), University of Leeds, Leeds, UK
| | - Ananya Choudhury
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Peter Hoskin
- Division of Cancer Sciences, School of Medical Sciences, Faculty of Biology, Medicine and Health, The University of Manchester, Manchester, UK
- Department of Radiotherapy Related Research, The Christie National Health Service (NHS) Foundation Trust, Manchester, UK
| | - Ann Henry
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Clinical Oncology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| | - Andrew Scarsbrook
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
- Department of Radiology, Leeds Cancer Centre, St James's University Hospital, Leeds Teaching Hospitals National Health Service (NHS) Trust, Beckett Street, Leeds, LS9 7TF, UK
| |
Collapse
|
24
|
Lother D, Robert M, Elwood E, Smith S, Tunariu N, Johnston SRD, Parton M, Bhaludin B, Millard T, Downey K, Sharma B. Imaging in metastatic breast cancer, CT, PET/CT, MRI, WB-DWI, CCA: review and new perspectives. Cancer Imaging 2023; 23:53. [PMID: 37254225 DOI: 10.1186/s40644-023-00557-8] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Accepted: 04/17/2023] [Indexed: 06/01/2023] Open
Abstract
BACKGROUND Breast cancer is the most frequent cancer in women and remains the second leading cause of death in Western countries. It represents a heterogeneous group of diseases with diverse tumoral behaviour, treatment responsiveness and prognosis. While major progress in diagnosis and treatment has resulted in a decline in breast cancer-related mortality, some patients will relapse and prognosis in this cohort of patients remains poor. Treatment is determined according to tumor subtype; primarily hormone receptor status and HER2 expression. Menopausal status and site of disease relapse are also important considerations in treatment protocols. MAIN BODY Staging and repeated evaluation of patients with metastatic breast cancer are central to the accurate assessment of disease extent at diagnosis and during treatment; guiding ongoing clinical management. Advances have been made in the diagnostic and therapeutic fields, particularly with new targeted therapies. In parallel, oncological imaging has evolved exponentially with the development of functional and anatomical imaging techniques. Consistent, reproducible and validated methods of assessing response to therapy is critical in effectively managing patients with metastatic breast cancer. CONCLUSION Major progress has been made in oncological imaging over the last few decades. Accurate disease assessment at diagnosis and during treatment is important in the management of metastatic breast cancer. CT (and BS if appropriate) is generally widely available, relatively cheap and sufficient in many cases. However, several additional imaging modalities are emerging and can be used as adjuncts, particularly in pregnancy or other diagnostically challenging cases. Nevertheless, no single imaging technique is without limitation. The authors have evaluated the vast array of imaging techniques - individual, combined parametric and multimodal - that are available or that are emerging in the management of metastatic breast cancer. This includes WB DW-MRI, CCA, novel PET breast cancer-epitope specific radiotracers and radiogenomics.
Collapse
Affiliation(s)
| | - Marie Robert
- Institut de Cancérologie de l'Ouest, St Herblain, France
| | | | - Sam Smith
- The Royal Marsden Hospital, London & Sutton, UK
| | - Nina Tunariu
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | - Stephen R D Johnston
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | | | | | | | - Kate Downey
- The Royal Marsden Hospital, London & Sutton, UK
- The Institute of Cancer Research (ICR), London & Sutton, UK
| | - Bhupinder Sharma
- The Royal Marsden Hospital, London & Sutton, UK.
- The Institute of Cancer Research (ICR), London & Sutton, UK.
| |
Collapse
|
25
|
Massafra R, Fanizzi A, Amoroso N, Bove S, Comes MC, Pomarico D, Didonna V, Diotaiuti S, Galati L, Giotta F, La Forgia D, Latorre A, Lombardi A, Nardone A, Pastena MI, Ressa CM, Rinaldi L, Tamborra P, Zito A, Paradiso AV, Bellotti R, Lorusso V. Analyzing breast cancer invasive disease event classification through explainable artificial intelligence. Front Med (Lausanne) 2023; 10:1116354. [PMID: 36817766 PMCID: PMC9932275 DOI: 10.3389/fmed.2023.1116354] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Accepted: 01/13/2023] [Indexed: 02/05/2023] Open
Abstract
Introduction Recently, accurate machine learning and deep learning approaches have been dedicated to the investigation of breast cancer invasive disease events (IDEs), such as recurrence, contralateral and second cancers. However, such approaches are poorly interpretable. Methods Thus, we designed an Explainable Artificial Intelligence (XAI) framework to investigate IDEs within a cohort of 486 breast cancer patients enrolled at IRCCS Istituto Tumori "Giovanni Paolo II" in Bari, Italy. Using Shapley values, we determined the IDE driving features according to two periods, often adopted in clinical practice, of 5 and 10 years from the first tumor diagnosis. Results Age, tumor diameter, surgery type, and multiplicity are predominant within the 5-year frame, while therapy-related features, including hormone, chemotherapy schemes and lymphovascular invasion, dominate the 10-year IDE prediction. Estrogen Receptor (ER), proliferation marker Ki67 and metastatic lymph nodes affect both frames. Discussion Thus, our framework aims at shortening the distance between AI and clinical practice.
Collapse
Affiliation(s)
| | | | - Nicola Amoroso
- INFN, Sezione di Bari, Bari, Italy
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Samantha Bove
- IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Domenico Pomarico
- INFN, Sezione di Bari, Bari, Italy
- Dipartimento di Fisica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | | | | | - Luisa Galati
- International Agency for Research on Cancer, Lyon, France
| | | | | | | | - Angela Lombardi
- Dipartimento di Ingegneria Elettrica e dell'Informazione, Politecnico di Bari, Bari, Italy
| | | | | | | | - Lucia Rinaldi
- IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Alfredo Zito
- IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| | | | - Roberto Bellotti
- INFN, Sezione di Bari, Bari, Italy
- Dipartimento di Fisica, Università degli Studi di Bari Aldo Moro, Bari, Italy
| | - Vito Lorusso
- IRCCS Istituto Tumori “Giovanni Paolo II”, Bari, Italy
| |
Collapse
|
26
|
Yan J, Sun Q, Tan X, Liang C, Bai H, Duan W, Mu T, Guo Y, Qiu Y, Wang W, Yao Q, Pei D, Zhao Y, Liu D, Duan J, Chen S, Sun C, Wang W, Liu Z, Hong X, Wang X, Guo Y, Xu Y, Liu X, Cheng J, Li ZC, Zhang Z. Image-based deep learning identifies glioblastoma risk groups with genomic and transcriptomic heterogeneity: a multi-center study. Eur Radiol 2023; 33:904-914. [PMID: 36001125 DOI: 10.1007/s00330-022-09066-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Revised: 07/20/2022] [Accepted: 07/25/2022] [Indexed: 02/03/2023]
Abstract
OBJECTIVES To develop and validate a deep learning imaging signature (DLIS) for risk stratification in patients with multiforme (GBM), and to investigate the biological pathways and genetic alterations underlying the DLIS. METHODS The DLIS was developed from multi-parametric MRI based on a training set (n = 600) and validated on an internal validation set (n = 164), an external test set 1 (n = 100), an external test set 2 (n = 161), and a public TCIA set (n = 88). A co-profiling framework based on a radiogenomics analysis dataset (n = 127) using multiscale high-dimensional data, including imaging, transcriptome, and genome, was established to uncover the biological pathways and genetic alterations underpinning the DLIS. RESULTS The DLIS was associated with survival (log-rank p < 0.001) and was an independent predictor (p < 0.001). The integrated nomogram incorporating the DLIS achieved improved C indices than the clinicomolecular nomogram (net reclassification improvement 0.39, p < 0.001). DLIS significantly correlated with core pathways of GBM (apoptosis and cell cycle-related P53 and RB pathways, and cell proliferation-related RTK pathway), as well as key genetic alterations (del_CDNK2A). The prognostic value of DLIS-correlated genes was externally confirmed on TCGA/CGGA sets (p < 0.01). CONCLUSIONS Our study offers a biologically interpretable deep learning predictor of survival outcomes in patients with GBM, which is crucial for better understanding GBM patient's prognosis and guiding individualized treatment. KEY POINTS • MRI-based deep learning imaging signature (DLIS) stratifies GBM into risk groups with distinct molecular characteristics. • DLIS is associated with P53, RB, and RTK pathways and del_CDNK2A mutation. • The prognostic value of DLIS-correlated pathway genes is externally demonstrated.
Collapse
Affiliation(s)
- Jing Yan
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Qiuchang Sun
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Xiangliang Tan
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Chaofeng Liang
- Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, China
| | - Hongmin Bai
- Department of Neurosurgery, Guangzhou General Hospital of Guangzhou Military Command, Guangzhou, 510010, China
| | - Wenchao Duan
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Tianhao Mu
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Yang Guo
- Department of Neurosurgery, Henan Provincial Hospital, Zhengzhou, 450052, Henan Province, China
| | - Yuning Qiu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Weiwei Wang
- Department of Pathology, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan Province, China
| | - Qiaoli Yao
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Dongling Pei
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yuanshen Zhao
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Danni Liu
- HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Jingxian Duan
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Shifu Chen
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.,HaploX Biotechnology, Shenzhen, Guangdong, China
| | - Chen Sun
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Wenqing Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Zhen Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xuanke Hong
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Xiangxiang Wang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yu Guo
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, China
| | - Xianzhi Liu
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China
| | - Jingliang Cheng
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| | - Zhi-Cheng Li
- Institute of Biomedical and Health Engineering, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China. .,University of Chinese Academy of Sciences, Beijing, China. .,Shenzhen United Imaging Research Institute of Innovative Medical Equipment, Shenzhen, 518045, China.
| | - Zhenyu Zhang
- Department of Neurosurgery, The First Affiliated Hospital of Zhengzhou University, Jian she Dong Road 1, Zhengzhou, 450052, Henan province, China.
| |
Collapse
|
27
|
Cui H, Sun Y, Zhao D, Zhang X, Kong H, Hu N, Wang P, Zuo X, Fan W, Yao Y, Fu B, Tian J, Wu M, Gao Y, Ning S, Zhang L. Radiogenomic analysis of prediction HER2 status in breast cancer by linking ultrasound radiomic feature module with biological functions. J Transl Med 2023; 21:44. [PMID: 36694240 PMCID: PMC9875533 DOI: 10.1186/s12967-022-03840-7] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Accepted: 12/19/2022] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND Human epidermal growth factor receptor 2 (HER2) overexpressed associated with poor prognosis in breast cancer and HER2 has been defined as a therapeutic target for breast cancer treatment. We aimed to explore the molecular biological information in ultrasound radiomic features (URFs) of HER2-positive breast cancer using radiogenomic analysis. Moreover, a radiomics model was developed to predict the status of HER2 in breast cancer. METHODS This retrospective study included 489 patients who were diagnosed with breast cancer. URFs were extracted from a radiomics analysis set using PyRadiomics. The correlations between differential URFs and HER2-related genes were calculated using Pearson correlation analysis. Functional enrichment of the identified URFs-correlated HER2 positive-specific genes was performed. Lastly, the radiomics model was developed based on the URF-module mined from auxiliary differential URFs to assess the HER2 status of breast cancer. RESULTS Eight differential URFs (p < 0.05) were identified among the 86 URFs extracted by Pyradiomics. 25 genes that were found to be the most closely associated with URFs. Then, the relevant biological functions of each differential URF were obtained through functional enrichment analysis. Among them, Zone Entropy is related to immune cell activity, which regulate the generation of calcification in breast cancer. The radiomics model based on the Logistic classifier and URF-module showed good discriminative ability (AUC = 0.80, 95% CI). CONCLUSION We searched for the URFs of HER2-positive breast cancer, and explored the underlying genes and biological functions of these URFs. Furthermore, the radiomics model based on the Logistic classifier and URF-module relatively accurately predicted the HER2 status in breast cancer.
Collapse
Affiliation(s)
- Hao Cui
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Yue Sun
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Dantong Zhao
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Xudong Zhang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Hanqing Kong
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Nana Hu
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Panting Wang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Xiaoxuan Zuo
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Wei Fan
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Yuan Yao
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Baiyang Fu
- grid.412463.60000 0004 1762 6325Department of Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Jiawei Tian
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| | - Meixin Wu
- grid.412463.60000 0004 1762 6325Department of Clinical Medicine, The Second Affiliated Hospital of Harbin Medical University, Heilongjiang, 150086 China
| | - Yue Gao
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Shangwei Ning
- grid.410736.70000 0001 2204 9268College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, 150081 China
| | - Lei Zhang
- grid.412463.60000 0004 1762 6325Department of Ultrasound Medicine, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086 Heilongjiang China
| |
Collapse
|
28
|
Liu Q, Hu P. Radiogenomic association of deep MR imaging features with genomic profiles and clinical characteristics in breast cancer. Biomark Res 2023; 11:9. [PMID: 36694221 PMCID: PMC9872423 DOI: 10.1186/s40364-023-00455-y] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2022] [Accepted: 01/13/2023] [Indexed: 01/26/2023] Open
Abstract
BACKGROUND It has been believed that traditional handcrafted radiomic features extracted from magnetic resonance imaging (MRI) of tumors are normally shallow and low-ordered. Recent advancement in deep learning technology shows that the high-order deep radiomic features extracted automatically from tumor images can capture tumor heterogeneity in a more efficient way. We hypothesize that MRI-based deep radiomic phenotypes have significant associations with molecular profiles of breast cancer tumors. We aim to identify deep radiomic features (DRFs) from MRI, evaluate their significance in predicting breast cancer (BC) clinical characteristics and explore their associations with multi-level genomic factors. METHODS A denoising autoencoder was built to retrospectively extract 4,096 DRFs from 110 BC patients' MRI. Visualization and clustering were applied to these DRFs. Linear Mixed Effect models were used to test their associations with multi-level genomic features (GFs) (risk genes, gene signatures, and biological pathway activities) extracted from the same patients' mRNA expression profile. A Least Absolute Shrinkage and Selection Operator model was used to identify the most predictive DRFs for each clinical characteristic (tumor size (T), lymph node metastasis (N), estrogen receptor (ER), progesterone receptor (PR), and human epidermal growth factor receptor 2 (HER2) status). RESULTS Thirty-six conventional radiomic features (CRFs) for 87 of the 110 BC patients provided by a previous study were used for comparison. More than 1,000 DRFs were associated with the risk genes, gene signatures, and biological pathways activities (adjusted P-value < 0.05). DRFs produced better performance in predicting T, N, ER, PR, and HER2 status (AUC > 0.9) using DRFs. These DRFs showed significant powers of stratifying patients, linking to relevant biological and clinical characteristics. As a contrast, only eight risk genes were associated with CRFs. The RFs performed worse in predicting clinical characteristics than DRFs. CONCLUSIONS The deep learning-based auto MRI features perform better in predicting BC clinical characteristics, which are more significantly associated with GFs than traditional semi-auto MRI features. Our radiogenomic approach for identifying MRI-based imaging signatures may pave potential pathways for the discovery of genetic mechanisms regulating specific tumor phenotypes and may enable a more rapid innovation of novel imaging modalities, hence accelerating their translation to personalized medicine.
Collapse
Affiliation(s)
- Qian Liu
- grid.21613.370000 0004 1936 9609Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9 Canada ,grid.21613.370000 0004 1936 9609Department of Computer Science, University of Manitoba, E2-445 EITC, Winnipeg, MB R3T 2N2 Canada ,grid.21613.370000 0004 1936 9609Department of Statistics, University of Manitoba, 318 Machray Hall, Winnipeg, MB R3T 2N2 Canada
| | - Pingzhao Hu
- grid.21613.370000 0004 1936 9609Department of Biochemistry and Medical Genetics, University of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB R3E 0J9 Canada ,grid.21613.370000 0004 1936 9609Department of Computer Science, University of Manitoba, E2-445 EITC, Winnipeg, MB R3T 2N2 Canada ,grid.419404.c0000 0001 0701 0170CancerCare Manitoba Research Institute, 675 McDermot Avenue, Winnipeg, MB R3E 0V9 Canada ,grid.39381.300000 0004 1936 8884Department of Biochemistry, Western University, Medical Sciences Building Rm. 342, London, ON N6A 5C1 Canada
| |
Collapse
|
29
|
Wang X, Yu G, Yan Z, Wan L, Wang W, Cui L. Lung Cancer Subtype Diagnosis by Fusing Image-Genomics Data and Hybrid Deep Networks. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2023; 20:512-523. [PMID: 34855599 DOI: 10.1109/tcbb.2021.3132292] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Accurate diagnosis of cancer subtypes is crucial for precise treatment, because different cancer subtypes are involved with different pathology and require different therapies. Although deep learning techniques have made great success in computer vision and other fields, they do not work well on Lung cancer subtype diagnosis, due to the distinction of slide images between different cancer subtypes is ambiguous. Furthermore, they often over-fit to high-dimensional genomics data with limited samples, and do not fuse the image and genomics data in a sensible way. In this paper, we propose a hybrid deep network based approach LungDIG for Lung cancer subtype Diagnosis by fusing Image-Genomics data. LungDIG first tiles the tissue slide image into small patches and extracts the patch-level features by fine-tuning an Inception-V3 model. Since the patches may contain some false positives in non-diagnostic regions, it further designs a patch-level feature combination strategy to integrate the extracted patch features and maintain the diversity between different cancer subtypes. At the same time, it extracts the genomics features from Copy Number Variation data by an attention based nonlinear extractor. Next, it fuses the image and genomics features by an attention based multilayer perceptron (MLP) to diagnose cancer subtype. Experiments on TCGA lung cancer data show that LungDIG can not only achieve higher accuracy for cancer subtype diagnosis than state-of-the-art methods, but also have a high authenticity and good interpretability.
Collapse
|
30
|
Li S, Zhou B. A review of radiomics and genomics applications in cancers: the way towards precision medicine. Radiat Oncol 2022; 17:217. [PMID: 36585716 PMCID: PMC9801589 DOI: 10.1186/s13014-022-02192-2] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 12/27/2022] [Indexed: 01/01/2023] Open
Abstract
The application of radiogenomics in oncology has great prospects in precision medicine. Radiogenomics combines large volumes of radiomic features from medical digital images, genetic data from high-throughput sequencing, and clinical-epidemiological data into mathematical modelling. The amalgamation of radiomics and genomics provides an approach to better study the molecular mechanism of tumour pathogenesis, as well as new evidence-supporting strategies to identify the characteristics of cancer patients, make clinical decisions by predicting prognosis, and improve the development of individualized treatment guidance. In this review, we summarized recent research on radiogenomics applications in solid cancers and presented the challenges impeding the adoption of radiomics in clinical practice. More standard guidelines are required to normalize radiomics into reproducible and convincible analyses and develop it as a mature field.
Collapse
Affiliation(s)
- Simin Li
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| | - Baosen Zhou
- grid.412636.40000 0004 1757 9485Department of Clinical Epidemiology and Center of Evidence-Based Medicine, The First Hospital of China Medical University, Shenyang, 110001 Liaoning People’s Republic of China
| |
Collapse
|
31
|
Identifying Associations between DCE-MRI Radiomic Features and Expression Heterogeneity of Hallmark Pathways in Breast Cancer: A Multi-Center Radiogenomic Study. Genes (Basel) 2022; 14:genes14010028. [PMID: 36672769 PMCID: PMC9858814 DOI: 10.3390/genes14010028] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 12/12/2022] [Accepted: 12/20/2022] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND To investigate the relationship between dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) radiomic features and the expression activity of hallmark pathways and to develop prediction models of pathway-level heterogeneity for breast cancer (BC) patients. METHODS Two radiogenomic cohorts were analyzed (n = 246). Tumor regions were segmented semiautomatically, and 174 imaging features were extracted. Gene set enrichment analysis (GSEA) and gene set variation analysis (GSVA) were performed to identify significant imaging-pathway associations. Random forest regression was used to predict pathway enrichment scores. Five-fold cross-validation and grid search were used to determine the optimal preprocessing operation and hyperparameters. RESULTS We identified 43 pathways, and 101 radiomic features were significantly related in the discovery cohort (p-value < 0.05). The imaging features of the tumor shape and mid-to-late post-contrast stages showed more transcriptional connections. Ten pathways relevant to functions such as cell cycle showed a high correlation with imaging in both cohorts. The prediction model for the mTORC1 signaling pathway achieved the best performance with the mean absolute errors (MAEs) of 27.29 and 28.61% in internal and external test sets, respectively. CONCLUSIONS The DCE-MRI features were associated with hallmark activities and may improve individualized medicine for BC by noninvasively predicting pathway-level heterogeneity.
Collapse
|
32
|
Zhong ME, Duan X, Ni-jia-ti MYDL, Qi H, Xu D, Cai D, Li C, Huang Z, Zhu Q, Gao F, Wu X. CT-based radiogenomic analysis dissects intratumor heterogeneity and predicts prognosis of colorectal cancer: a multi-institutional retrospective study. J Transl Med 2022; 20:574. [PMID: 36482390 PMCID: PMC9730572 DOI: 10.1186/s12967-022-03788-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 11/23/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to develop a radiogenomic prognostic prediction model for colorectal cancer (CRC) by investigating the biological and clinical relevance of intratumoural heterogeneity. METHODS This retrospective multi-cohort study was conducted in three steps. First, we identified genomic subclones using unsupervised deconvolution analysis. Second, we established radiogenomic signatures to link radiomic features with prognostic subclone compositions in an independent radiogenomic dataset containing matched imaging and gene expression data. Finally, the prognostic value of the identified radiogenomic signatures was validated using two testing datasets containing imaging and survival information collected from separate medical centres. RESULTS This multi-institutional retrospective study included 1601 patients (714 females and 887 males; mean age, 65 years ± 14 [standard deviation]) with CRC from 5 datasets. Molecular heterogeneity was identified using unsupervised deconvolution analysis of gene expression data. The relative prevalence of the two subclones associated with cell cycle and extracellular matrix pathways identified patients with significantly different survival outcomes. A radiogenomic signature-based predictive model significantly stratified patients into high- and low-risk groups with disparate disease-free survival (HR = 1.74, P = 0.003). Radiogenomic signatures were revealed as an independent predictive factor for CRC by multivariable analysis (HR = 1.59, 95% CI:1.03-2.45, P = 0.034). Functional analysis demonstrated that the 11 radiogenomic signatures were predominantly associated with extracellular matrix and immune-related pathways. CONCLUSIONS The identified radiogenomic signatures might be a surrogate for genomic signatures and could complement the current prognostic strategies.
Collapse
Affiliation(s)
- Min-Er Zhong
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.413405.70000 0004 1808 0686Department of Gastrointestinal Surgery, Department of General Surgery, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Xin Duan
- grid.12981.330000 0001 2360 039XSchool of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen, China
| | - Ma-yi-di-li Ni-jia-ti
- Department of Radiology, The First People’s Hospital of Kashi Prefecture, Kashi, Xinjiang China
| | - Haoning Qi
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Dongwei Xu
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Du Cai
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chenghang Li
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zeping Huang
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Qiqi Zhu
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.507012.10000 0004 1798 304XDepartment of Colorectal Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo, China
| | - Feng Gao
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China ,Shanghai Artificial Intelligence Laboratory, Shanghai, China
| | - Xiaojian Wu
- grid.488525.6Department of Colorectal Surgery, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, 510655 China ,grid.488525.6Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Diseases, The Sixth Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
33
|
Wang L, Liu W, Liu K, Wang L, Yin X, Bo L, Xu H, Lin S, Feng K, Zhou X, Lin L, Fei M, Zhang C, Ning S, Zhao H. The dynamic dysregulated network identifies stage-specific markers during lung adenocarcinoma malignant progression and metastasis. MOLECULAR THERAPY. NUCLEIC ACIDS 2022; 30:633-647. [PMID: 36514354 PMCID: PMC9722404 DOI: 10.1016/j.omtn.2022.11.019] [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: 06/08/2022] [Accepted: 11/17/2022] [Indexed: 11/23/2022]
Abstract
Brain metastasis occurs in approximately 30% of patients with lung adenocarcinoma (LUAD) and is closely associated with poor prognosis, recurrence, and death. However, dynamic gene regulation and molecular mechanism driving LUAD progression remain poorly understood. In this study, we performed a comprehensive single-cell transcriptome analysis using data from normal, early stage, advanced stage, and brain metastasis LUAD. Our single-cell-level analysis reveals the cellular composition heterogeneity at different stages during LUAD progression. We identified stage-specific risk genes that could contribute to LUAD progression and metastasis by reprogramming immune-related and metabolic-related functions. We constructed an early advanced metastatic dysregulated network and revealed the dynamic changes in gene regulations during LUAD progression. We identified 6 early advanced (HLA-DRB1, HLA-DQB1, SFTPB, SFTPC, PLA2G1B, and FOLR1), 8 advanced metastasis (RPS15, RPS11, RPL13A, RPS24, HLA-DRB5, LYPLA1, KCNJ15, and PSMA3), and 2 common risk genes in different stages (SFTPD and HLA-DRA) as prognostic markers in LUAD. Particularly, decreased expression of HLA-DRA, HLA-DRB1, HLA-DQB1, and HLA-DRB5 refer poor prognosis in LUAD by controlling antigen processing and presentation and T cell activation. Increased expression of PSMA3 and LYPLA1 refer poor prognosis by reprogramming fatty acid metabolism and RNA catabolic process. Our findings will help further understanding the pathobiology of brain metastases in LUAD.
Collapse
Affiliation(s)
- Li Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China,Corresponding author Li Wang, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| | - Wangyang Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Kailai Liu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Lixia Wang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xiangzhe Yin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Lin Bo
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Haotian Xu
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shihua Lin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Ke Feng
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Xinyu Zhou
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Lin Lin
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Meiting Fei
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Caiyu Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China
| | - Shangwei Ning
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China,Corresponding author Shangwei Ning, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| | - Hongying Zhao
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China,Corresponding author Hongying Zhao, College of Bioinformatics Science and Technology, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
34
|
Unsupervised Analysis Based on DCE-MRI Radiomics Features Revealed Three Novel Breast Cancer Subtypes with Distinct Clinical Outcomes and Biological Characteristics. Cancers (Basel) 2022; 14:cancers14225507. [PMID: 36428600 PMCID: PMC9688868 DOI: 10.3390/cancers14225507] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2022] [Revised: 11/06/2022] [Accepted: 11/07/2022] [Indexed: 11/11/2022] Open
Abstract
Background: This study aimed to reveal the heterogeneity of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) of breast cancer (BC) and identify its prognosis values and molecular characteristics. Methods: Two radiogenomics cohorts (n = 246) were collected and tumor regions were segmented semi-automatically. A total of 174 radiomics features were extracted, and the imaging subtypes were identified and validated by unsupervised analysis. A gene-profile-based classifier was developed to predict the imaging subtypes. The prognostic differences and the biological and microenvironment characteristics of subtypes were uncovered by bioinformatics analysis. Results: Three imaging subtypes were identified and showed high reproducibility. The subtypes differed remarkably in tumor sizes and enhancement patterns, exhibiting significantly different disease-free survival (DFS) or overall survival (OS) in the discovery cohort (p = 0.024) and prognosis datasets (p ranged from <0.0001 to 0.0071). Large sizes and rapidly enhanced tumors usually had the worst outcomes. Associations were found between imaging subtypes and the established subtypes or clinical stages (p ranged from <0.001 to 0.011). Imaging subtypes were distinct in cell cycle and extracellular matrix (ECM)-receptor interaction pathways (false discovery rate, FDR < 0.25) and different in cellular fractions, such as cancer-associated fibroblasts (p < 0.05). Conclusions: The imaging subtypes had different clinical outcomes and biological characteristics, which may serve as potential biomarkers.
Collapse
|
35
|
Ming W, Li F, Zhu Y, Bai Y, Gu W, Liu Y, Sun X, Liu X, Liu H. Predicting hormone receptors and PAM50 subtypes of breast cancer from multi-scale lesion images of DCE-MRI with transfer learning technique. Comput Biol Med 2022; 150:106147. [PMID: 36201887 DOI: 10.1016/j.compbiomed.2022.106147] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2022] [Revised: 09/06/2022] [Accepted: 09/24/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND The recent development of artificial intelligence (AI) technologies coupled with medical imaging data has gained considerable attention, and offers a non-invasive approach for cancer diagnosis and prognosis. In this context, improved breast cancer (BC) molecular characteristics assessment models are foreseen to enable personalized strategies with better clinical outcomes compared to existing screening strategies. And it is a promising approach to developing models for hormone receptors (HR) and subtypes of BC patients from dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) data. METHODS In this institutional review board-approved study, 174 BC patients with both DCE-MRI and RNA-seq data in the local database were analyzed. Slice images from tumor lesions and multi-scale peri-tumor regions were used as model inputs, and five representative pre-trained transfer learning (TF) networks, such as Inception-v3 and Xception, were employed to establish prediction models. A comprehensive analysis was performed using five-fold cross-validation to avoid overfitting, and accuracy (ACC) and area under the receiver operating characteristic curve (AUROC) to evaluate model performance. RESULTS Xception achieved the superior results when using solely tumor regions, with highest AUROCs of 0.844 (95% CI: [0.841, 0.847]) and 0.784 (95% CI: [0.781, 0.788]) for estrogen receptor (ER) and progesterone receptor (PR), respectively, and best ACC of 0.467 (95% CI: [0.462, 0.470]) for PAM50 subtypes. A significant improvement in the model performance was observed when images of the peri-tumor region were included, with optimal results achieved using images of the tumor and the 10 mm peri-tumor regions. Xception-based TF models performed most effectively in predicting ER and PR statuses, with the AUROCs were 0.942 (95% CI: [0.940, 0.944]) and 0.920 (95% CI: [0.917, 0.922]), respectively, whereas for PAM50 subtypes, the Inception-v3-based network yielded the highest ACC as 0.742 (95% CI: [0.738, 0.746]). CONCLUSIONS Transfer learning analysis based on DCE-MRI data of tumor and peri-tumor regions was helpful to the non-invasive assessment of molecular characteristics of BC.
Collapse
Affiliation(s)
- Wenlong Ming
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Fuyu Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Yanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, PR China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Wanjun Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China; Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, 210023, PR China
| | - Yun Liu
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, PR China
| | - Xiao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, PR China.
| | - Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, PR China.
| |
Collapse
|
36
|
Joint model- and immunohistochemistry-driven few-shot learning scheme for breast cancer segmentation on 4D DCE-MRI. APPL INTELL 2022. [DOI: 10.1007/s10489-022-04272-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
37
|
Hou J, Jia X, Xie Y, Qin W. Integrative Histology-Genomic Analysis Predicts Hepatocellular Carcinoma Prognosis Using Deep Learning. Genes (Basel) 2022; 13:genes13101770. [PMID: 36292654 PMCID: PMC9601633 DOI: 10.3390/genes13101770] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2022] [Revised: 09/25/2022] [Accepted: 09/28/2022] [Indexed: 11/04/2022] Open
Abstract
Cancer prognosis analysis is of essential interest in clinical practice. In order to explore the prognostic power of computational histopathology and genomics, this paper constructs a multi-modality prognostic model for survival prediction. We collected 346 patients diagnosed with hepatocellular carcinoma (HCC) from The Cancer Genome Atlas (TCGA), each patient has 1-3 whole slide images (WSIs) and an mRNA expression file. WSIs were processed by a multi-instance deep learning model to obtain the patient-level survival risk scores; mRNA expression data were processed by weighted gene co-expression network analysis (WGCNA), and the top hub genes of each module were extracted as risk factors. Information from two modalities was integrated by Cox proportional hazard model to predict patient outcomes. The overall survival predictions of the multi-modality model (Concordance index (C-index): 0.746, 95% confidence interval (CI): ±0.077) outperformed these based on histopathology risk score or hub genes, respectively. Furthermore, in the prediction of 1-year and 3-year survival, the area under curve of the model achieved 0.816 and 0.810. In conclusion, this paper provides an effective workflow for multi-modality prognosis of HCC, the integration of histopathology and genomic information has the potential to assist clinical prognosis management.
Collapse
Affiliation(s)
- Jiaxin Hou
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Shenzhen College of Advanced Technology, University of Chinese Academy of Sciences, Shenzhen 518055, China
| | - Xiaoqi Jia
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- School of Computer Science and Engineering, Northeastern University, Shenyang 110819, China
| | - Yaoqin Xie
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
| | - Wenjian Qin
- Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen 518055, China
- Correspondence:
| |
Collapse
|
38
|
A hybrid hemodynamic knowledge-powered and feature reconstruction-guided scheme for breast cancer segmentation based on DCE-MRI. Med Image Anal 2022; 82:102572. [PMID: 36055051 DOI: 10.1016/j.media.2022.102572] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2021] [Revised: 07/08/2022] [Accepted: 08/11/2022] [Indexed: 11/24/2022]
Abstract
Automatically and accurately annotating tumor in dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), which provides a noninvasive in vivo method to evaluate tumor vasculature architectures based on contrast accumulation and washout, is a crucial step in computer-aided breast cancer diagnosis and treatment. However, it remains challenging due to the varying sizes, shapes, appearances and densities of tumors caused by the high heterogeneity of breast cancer, and the high dimensionality and ill-posed artifacts of DCE-MRI. In this paper, we propose a hybrid hemodynamic knowledge-powered and feature reconstruction-guided scheme that integrates pharmacokinetics prior and feature refinement to generate sufficiently adequate features in DCE-MRI for breast cancer segmentation. The pharmacokinetics prior expressed by time intensity curve (TIC) is incorporated into the scheme through objective function called dynamic contrast-enhanced prior (DCP) loss. It contains contrast agent kinetic heterogeneity prior knowledge, which is important to optimize our model parameters. Besides, we design a spatial fusion module (SFM) embedded in the scheme to exploit intra-slices spatial structural correlations, and deploy a spatial-kinetic fusion module (SKFM) to effectively leverage the complementary information extracted from spatial-kinetic space. Furthermore, considering that low spatial resolution often leads to poor image quality in DCE-MRI, we integrate a reconstruction autoencoder into the scheme to refine feature maps in an unsupervised manner. We conduct extensive experiments to validate the proposed method and show that our approach can outperform recent state-of-the-art segmentation methods on breast cancer DCE-MRI dataset. Moreover, to explore the generalization for other segmentation tasks on dynamic imaging, we also extend the proposed method to brain segmentation in DSC-MRI sequence. Our source code will be released on https://github.com/AI-medical-diagnosis-team-of-JNU/DCEDuDoFNet.
Collapse
|
39
|
Ming W, Zhu Y, Bai Y, Gu W, Li F, Hu Z, Xia T, Dai Z, Yu X, Li H, Gu Y, Yuan S, Zhang R, Li H, Zhu W, Ding J, Sun X, Liu Y, Liu H, Liu X. Radiogenomics analysis reveals the associations of dynamic contrast-enhanced-MRI features with gene expression characteristics, PAM50 subtypes, and prognosis of breast cancer. Front Oncol 2022; 12:943326. [PMID: 35965527 PMCID: PMC9366134 DOI: 10.3389/fonc.2022.943326] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Accepted: 06/29/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND To investigate reliable associations between dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) features and gene expression characteristics in breast cancer (BC) and to develop and validate classifiers for predicting PAM50 subtypes and prognosis from DCE-MRI non-invasively. METHODS Two radiogenomics cohorts with paired DCE-MRI and RNA-sequencing (RNA-seq) data were collected from local and public databases and divided into discovery (n = 174) and validation cohorts (n = 72). Six external datasets (n = 1,443) were used for prognostic validation. Spatial-temporal features of DCE-MRI were extracted, normalized properly, and associated with gene expression to identify the imaging features that can indicate subtypes and prognosis. RESULTS Expression of genes including RBP4, MYBL2, and LINC00993 correlated significantly with DCE-MRI features (q-value < 0.05). Importantly, genes in the cell cycle pathway exhibited a significant association with imaging features (p-value < 0.001). With eight imaging-associated genes (CHEK1, TTK, CDC45, BUB1B, PLK1, E2F1, CDC20, and CDC25A), we developed a radiogenomics prognostic signature that can distinguish BC outcomes in multiple datasets well. High expression of the signature indicated a poor prognosis (p-values < 0.01). Based on DCE-MRI features, we established classifiers to predict BC clinical receptors, PAM50 subtypes, and prognostic gene sets. The imaging-based machine learning classifiers performed well in the independent dataset (areas under the receiver operating characteristic curve (AUCs) of 0.8361, 0.809, 0.7742, and 0.7277 for estrogen receptor (ER), human epidermal growth factor receptor 2 (HER2)-enriched, basal-like, and obtained radiogenomics signature). Furthermore, we developed a prognostic model directly using DCE-MRI features (p-value < 0.0001). CONCLUSIONS Our results identified the DCE-MRI features that are robust and associated with the gene expression in BC and displayed the possibility of using the features to predict clinical receptors and PAM50 subtypes and to indicate BC prognosis.
Collapse
Affiliation(s)
- Wenlong Ming
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yanhui Zhu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Yunfei Bai
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wanjun Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
- Collaborative Innovation Center of Jiangsu Province of Cancer Prevention and Treatment of Chinese Medicine, School of Artificial Intelligence and Information Technology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Fuyu Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Zixi Hu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Tiansong Xia
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Zuolei Dai
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Xiafei Yu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Huamei Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yu Gu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Shaoxun Yuan
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Rongxin Zhang
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Haitao Li
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Wenyong Zhu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Jianing Ding
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiao Sun
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Yun Liu
- Department of Information, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Hongde Liu
- State Key Laboratory of Bioelectronics, School of Biological Science and Medical Engineering, Southeast University, Nanjing, China
| | - Xiaoan Liu
- Department of Breast Surgery, The First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| |
Collapse
|
40
|
Fan M, Yuan C, Huang G, Xu M, Wang S, Gao X, Li L. A framework for deep multitask learning with multiparametric magnetic resonance imaging for the joint prediction of histological characteristics in breast cancer. IEEE J Biomed Health Inform 2022; 26:3884-3895. [PMID: 35635826 DOI: 10.1109/jbhi.2022.3179014] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The clinical management and decision-making process related to breast cancer are based on multiple histological indicators. This study aims to jointly predict the Ki-67 expression level, luminal A subtype and histological grade molecular biomarkers using a new deep multitask learning method with multiparametric magnetic resonance imaging. A multitask learning network structure was proposed by introducing a common-task layer and task-specific layers to learn the high-level features that are common to all tasks and related to a specific task, respectively. A network pretrained with knowledge from the ImageNet dataset was used and fine-tuned with MRI data. Information from multiparametric MR images was fused using the strategy at the feature and decision levels. The area under the receiver operating characteristic curve (AUC) was used to measure model performance. For single-task learning using a single image series, the deep learning model generated AUCs of 0.752, 0.722, and 0.596 for the Ki-67, luminal A and histological grade prediction tasks, respectively. The performance was improved by freezing the first 5 convolutional layers, using 20% shared layers and fusing multiparametric series at the feature level, which achieved AUCs of 0.819, 0.799 and 0.747 for Ki-67, luminal A and histological grade prediction tasks, respectively. Our study showed advantages in jointly predicting correlated clinical biomarkers using a deep multitask learning framework with an appropriate number of fine-tuned convolutional layers by taking full advantage of common and complementary imaging features. Multiparametric image series-based multitask learning could be a promising approach for the multiple clinical indicator-based management of breast cancer.
Collapse
|
41
|
Liu Q, Hu P. A novel integrative computational framework for breast cancer radiogenomic biomarker discovery. Comput Struct Biotechnol J 2022; 20:2484-2494. [PMID: 35664228 PMCID: PMC9136270 DOI: 10.1016/j.csbj.2022.05.031] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 05/14/2022] [Accepted: 05/15/2022] [Indexed: 12/22/2022] Open
Abstract
Bayesian tensor factorization is used to integrate multiomics data for radiogenomics analysis. A regression framework is proposed to handle the unmatched data issue in radiogenomics analysis. Deep learning is used to identify prognostic meaningful radiogenomic biomarkers for cancer.
In precise medicine, it is with great value to develop computational frameworks for identifying prognostic biomarkers which can capture both multi-genomic and phenotypic heterogeneity of breast cancer (BC). Radiogenomics is a field where medical images and genomic measurements are integrated and mined to solve challenging clinical problems. Previous radiogenomic studies suffered from data incompleteness, feature subjectivity and low interpretability. For example, the majority of the radiogenomic studies miss one or two of medical imaging data, genomic data, and clinical outcome data, which results in the data incomplete issue. Feature subjectivity issue comes from the extraction of imaging features with significant human involvement. Thus, there is an urgent need to address above-mentioned limitations so that fully automatic and transparent radiogenomic prognostic biomarkers could be identified for BC. We proposed a novel framework for BC prognostic radiogenomic biomarker identification. This framework involves an explainable DL model for image feature extraction, a Bayesian tensor factorization (BTF) processing for multi-genomic feature extraction, a leverage strategy to utilize unpaired imaging, genomic, and survival outcome data, and a mediation analysis to provide further interpretation for identified biomarkers. This work provided a new perspective for conducting a comprehensive radiogenomic study when only limited resources are given. Compared with baseline traditional radiogenomic biomarkers, the 23 biomarkers identified by the proposed framework performed better in indicating patients’ survival outcome. And their interpretability is guaranteed by different levels of build-in and follow-up analyses.
Collapse
Affiliation(s)
- Qian Liu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0W3, Canada
- Department of Computer Science, University of Manitoba, Winnipeg, Manitoba R3E 0W3, Canada
- Department of Statistics, University of Manitoba, Winnipeg, Manitoba R3E 0W3, Canada
| | - Pingzhao Hu
- Department of Biochemistry and Medical Genetics, University of Manitoba, Winnipeg, Manitoba R3E 0W3, Canada
- Department of Computer Science, University of Manitoba, Winnipeg, Manitoba R3E 0W3, Canada
- Corresponding author at: Department of Biochemistry and Medical Genetics, Room 308 - Basic Medical Sciences Building, 745 Bannatyne Avenue, University of Manitoba, Winnipeg, Manitoba R3E 0J9, Canada.
| |
Collapse
|
42
|
Song F, Song L, Xing T, Feng Y, Song X, Zhang P, Zhang T, Zhu Z, Song W, Zhang G. A Multi-Classification Model for Predicting the Invasiveness of Lung Adenocarcinoma Presenting as Pure Ground-Glass Nodules. Front Oncol 2022; 12:800811. [PMID: 35574301 PMCID: PMC9096139 DOI: 10.3389/fonc.2022.800811] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2021] [Accepted: 04/04/2022] [Indexed: 11/28/2022] Open
Abstract
Objectives To establish a multi-classification model for precisely predicting the invasiveness (pre-invasive adenocarcinoma, PIA; minimally invasive adenocarcinoma, MIA; invasive adenocarcinoma, IAC) of lung adenocarcinoma manifesting as pure ground-glass nodules (pGGNs). Methods By the inclusion and exclusion criteria, this retrospective study enrolled 346 patients (female, 297, and male, 49; age, 55.79 ± 10.53 (24-83)) presenting as pGGNs from 1292 consecutive patients with pathologically confirmed lung adenocarcinoma. A total of 27 clinical were collected and 1409 radiomics features were extracted by PyRadiomics package on python. After feature selection with L2,1-norm minimization, logistic regression (LR), extra w(ET) and gradient boosting decision tree (GBDT) were used to construct the three-classification model. Then, an ensemble model of the three algorithms based on model ensemble strategy was established to further improve the classification performance. Results After feature selection, a hybrid of 166 features consisting of 1 clinical (short-axis diameter, ranked 27th) and 165 radiomics (4 shape, 71 intensity and 90 texture) features were selected. The three most important features are wavelet-HLL_firstorder_Minimum, wavelet-HLL_ngtdm_Busyness and square_firstorder_Kurtosis. The hybrid-ensemble model based on hybrid clinical-radiomics features and the ensemble strategy showed more accurate predictive performance than other models (hybrid-LR, hybrid-ET, hybrid-GBDT, clinical-ensemble and radiomics-ensemble). On the training set and test set, the model can obtain the accuracy values of 0.918 ± 0.022 and 0.841, and its F1-scores respectively were 0.917 ± 0.024 and 0.824. Conclusion The multi-classification of invasive pGGNs can be precisely predicted by our proposed hybrid-ensemble model to assist patients in the early diagnosis of lung adenocarcinoma and prognosis.
Collapse
Affiliation(s)
- Fan Song
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Lan Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Tongtong Xing
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Youdan Feng
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Xiao Song
- School of Medical Imaging, Shanxi Medical University, Taiyuan, China
| | - Peng Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Tianyi Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Zhenchen Zhu
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- 4 + 4 MD Program, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wei Song
- Department of Radiology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guanglei Zhang
- Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| |
Collapse
|
43
|
Deep learning signatures reveal multiscale intratumor heterogeneity associated with biological functions and survival in recurrent nasopharyngeal carcinoma. Eur J Nucl Med Mol Imaging 2022; 49:2972-2982. [PMID: 35471254 DOI: 10.1007/s00259-022-05793-x] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 04/03/2022] [Indexed: 12/24/2022]
Abstract
PURPOSE How to discriminate different risks of recurrent nasopharyngeal carcinoma (rNPC) patients and guide individual treatment has become of great importance. This study aimed to explore the associations between deep learning signatures and biological functions as well as survival in (rNPC) patients. METHODS A total of 420 rNPC patients with PET/CT imaging and follow-up of overall survival (OS) were retrospectively enrolled. All patients were randomly divided into a training set (n = 269) and test set (n = 151) with a 6:4 ratio. We constructed multi-modality deep learning signatures from PET and CT images with a light-weighted deep convolutional neural network EfficienetNet-lite0 and survival loss DeepSurvLoss. An integrated nomogram was constructed incorporating clinical factors and deep learning signatures from PET/CT. Clinical nomogram and single-modality deep learning nomograms were also built for comparison. Furthermore, the association between biological functions and survival risks generated from an integrated nomogram was analyzed by RNA sequencing (RNA-seq). RESULTS The C-index of the integrated nomogram incorporating age, rT-stage, and deep learning PET/CT signature was 0.741 (95% CI: 0.688-0.794) in the training set and 0.732 (95% CI: 0.679-0.785) in the test set. The nomogram stratified patients into two groups with high risk and low risk in both the training set and test set with hazard ratios (HR) of 4.56 (95% CI: 2.80-7.42, p < 0.001) and 4.05 (95% CI: 2.21-7.43, p < 0.001), respectively. The C-index of the integrated nomogram was significantly higher than the clinical nomogram and single-modality nomograms. When stratified by sex, N-stage, or EBV DNA, risk prediction of our integrated nomogram was valid in all patient subgroups. Further subgroup analysis showed that patients with a low-risk could benefit from surgery and re-irradiation, while there was no difference in survival rates between patients treated by chemotherapy in the high-risk and low-risk groups. RNA sequencing (RNA-seq) of data further explored the mechanism of high- and low-risk patients from the genetic and molecular level. CONCLUSION Our study demonstrated that PET/CT-based deep learning signatures showed satisfactory prognostic predictive performance in rNPC patients. The nomogram incorporating deep learning signatures successfully divided patients into different risks and had great potential to guide individual treatment: patients with a low-risk were supposed to be treated with surgery and re-irradiation, while for high-risk patients, the application of palliative chemotherapy may be sufficient.
Collapse
|
44
|
Wang X, Xie T, Luo J, Zhou Z, Yu X, Guo X. Radiomics predicts the prognosis of patients with locally advanced breast cancer by reflecting the heterogeneity of tumor cells and the tumor microenvironment. Breast Cancer Res 2022; 24:20. [PMID: 35292076 PMCID: PMC8922933 DOI: 10.1186/s13058-022-01516-0] [Citation(s) in RCA: 73] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2021] [Accepted: 03/02/2022] [Indexed: 12/13/2022] Open
Abstract
Background This study investigated the efficacy of radiomics to predict survival outcome for locally advanced breast cancer (LABC) patients and the association of radiomics with tumor heterogeneity and microenvironment. Methods Patients with LABC from 2010 to 2015 were retrospectively reviewed. Radiomics features were extracted from enhanced MRI. We constructed the radiomics score using lasso and assessed its prognostic value. An external validation cohort from The Cancer Imaging Archive was used to assess phenotype reproducibility. Sequencing data from TCGA and our center were applied to reveal genomic landscape of different radiomics score groups. Tumor infiltrating lymphocytes map and bioinformatics methods were applied to evaluate the heterogeneity of tumor microenvironment. Computational histopathology was also applied. Results A total of 278 patients were divided into training cohort and validation cohort. Radiomics score was constructed and significantly associated with disease-free survival (DFS) of the patients in training cohort, validation cohort and external validation cohort (p < 0.001, p = 0.014 and p = 0.041, respectively). The radiomics-based nomogram showed better predictive performance of DFS compared with TNM model. Distinct gene expression patterns were identified. Immunophenotype and immune cell composition was different in each radiomics score group. The link between radiomics and computational histopathology was revealed. Conclusions The radiomics score could effectively predict prognosis of LABC after neoadjuvant chemotherapy and radiotherapy. Radiomics revealed heterogeneity of tumor cell and tumor microenvironment and holds great potential to facilitate individualized DFS estimation and guide personalized care. Supplementary Information The online version contains supplementary material available at 10.1186/s13058-022-01516-0.
Collapse
|
45
|
Chen L, Wu CT, Lin CH, Dai R, Liu C, Clarke R, Yu G, Van Eyk JE, Herrington DM, Wang Y. swCAM: estimation of subtype-specific expressions in individual samples with unsupervised sample-wise deconvolution. Bioinformatics 2022; 38:1403-1410. [PMID: 34904628 PMCID: PMC8826012 DOI: 10.1093/bioinformatics/btab839] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 10/30/2021] [Accepted: 12/10/2021] [Indexed: 02/04/2023] Open
Abstract
MOTIVATION Complex biological tissues are often a heterogeneous mixture of several molecularly distinct cell subtypes. Both subtype compositions and subtype-specific (STS) expressions can vary across biological conditions. Computational deconvolution aims to dissect patterns of bulk tissue data into subtype compositions and STS expressions. Existing deconvolution methods can only estimate averaged STS expressions in a population, while many downstream analyses such as inferring co-expression networks in particular subtypes require subtype expression estimates in individual samples. However, individual-level deconvolution is a mathematically underdetermined problem because there are more variables than observations. RESULTS We report a sample-wise Convex Analysis of Mixtures (swCAM) method that can estimate subtype proportions and STS expressions in individual samples from bulk tissue transcriptomes. We extend our previous CAM framework to include a new term accounting for between-sample variations and formulate swCAM as a nuclear-norm and ℓ2,1-norm regularized matrix factorization problem. We determine hyperparameter values using cross-validation with random entry exclusion and obtain a swCAM solution using an efficient alternating direction method of multipliers. Experimental results on realistic simulation data show that swCAM can accurately estimate STS expressions in individual samples and successfully extract co-expression networks in particular subtypes that are otherwise unobtainable using bulk data. In two real-world applications, swCAM analysis of bulk RNASeq data from brain tissue of cases and controls with bipolar disorder or Alzheimer's disease identified significant changes in cell proportion, expression pattern and co-expression module in patient neurons. Comparative evaluation of swCAM versus peer methods is also provided. AVAILABILITY AND IMPLEMENTATION The R Scripts of swCAM are freely available at https://github.com/Lululuella/swCAM. A user's guide and a vignette are provided. SUPPLEMENTARY INFORMATION Supplementary data are available at Bioinformatics online.
Collapse
Affiliation(s)
- Lulu Chen
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Chiung-Ting Wu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Chia-Hsiang Lin
- Department of Electrical Engineering, National Cheng Kung University, Tainan 70101, Taiwan
| | - Rujia Dai
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Chunyu Liu
- Department of Psychiatry, SUNY Upstate Medical University, Syracuse, NY 13210, USA
| | - Robert Clarke
- The Hormel Institute, University of Minnesota, Austin, MN 55912, USA
| | - Guoqiang Yu
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| | - Jennifer E Van Eyk
- Advanced Clinical Biosystems Research Institute, Cedars Sinai Medical Center, Los Angeles, CA 90048, USA
| | - David M Herrington
- Department of Internal Medicine, Wake Forest University, Winston-Salem, NC 27157, USA
| | - Yue Wang
- Department of Electrical and Computer Engineering, Virginia Polytechnic Institute and State University, Arlington, VA 22203, USA
| |
Collapse
|
46
|
Diniz de Paula W. Editorial for "Radiomic Analysis of Pharmacokinetic Heterogeneity Within Tumor Based on the Unsupervised Decomposition of DCE-MRI for Predicting Histological Characteristics of Breast Cancer". J Magn Reson Imaging 2021; 55:1648-1649. [PMID: 34957642 DOI: 10.1002/jmri.28042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Accepted: 12/15/2021] [Indexed: 11/10/2022] Open
Affiliation(s)
- Wagner Diniz de Paula
- Diagnostic Imaging Unit, Brasilia University Hospital, University of Brasilia, Brasília, Brazil
| |
Collapse
|
47
|
Fan M, Yuan W, Liu W, Gao X, Xu M, Wang S, Li L. A deep matrix factorization framework for identifying underlying tissue-specific patterns of DCE-MRI: applications for molecular subtype classification in breast cancer. Phys Med Biol 2021; 66. [PMID: 34787109 DOI: 10.1088/1361-6560/ac3a25] [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: 09/16/2021] [Accepted: 11/16/2021] [Indexed: 11/12/2022]
Abstract
Objective.Breast cancer is heterogeneous in that different angiogenesis and blood flow characteristics could be present within a tumor. The pixel kinetics of dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) can assume several distinct signal patterns related to specific tissue characteristics. Identification of the latent, tissue-specific dynamic patterns of intratumor heterogeneity can shed light on the biological mechanisms underlying the heterogeneity of tumors.Approach.To mine this information, we propose a deep matrix factorization-based dynamic decomposition (DMFDE) model specifically designed according to DCE-MRI characteristics. The time-series imaging data were decomposed into tissue-specific dynamic patterns and their corresponding proportion maps. The image pixel matrix and the reference matrix of population-level kinetics obtained by clustering the dynamic signals were used as the inputs. Two multilayer neural network branches were designed to collaboratively project the input matrix into a latent dynamic pattern and a dynamic proportion matrix, which was justified using simulated data. Clinical implications of DMFDE were assessed by radiomics analysis of proportion maps obtained from the tumor/parenchyma region for classifying the luminal A subtype.Main results.The decomposition performance of DMFDE was evaluated by the root mean square error and was shown to be better than that of the conventional convex analysis of mixtures (CAM) method. The predictive model withK = 3, 4, and 5 dynamic proportion maps generated AUC values of 0.780, 0.786 and 0.790, respectively, in distinguishing between luminal A and nonluminal A tumors, which are better than the CAM method (AUC = 0.726). The combination of statistical features from images with different proportion maps has the highest prediction value (AUC = 0.813), which is significantly higher than that based on CAM.Conclusion.This proposed method identified the latent dynamic patterns associated with different molecular subtypes, and radiomics analysis based on the pixel compositions of the uncovered dynamic patterns was able to determine molecular subtypes of breast cancer.
Collapse
Affiliation(s)
- Ming Fan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, People's Republic of China
| | - Wei Yuan
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, People's Republic of China
| | - Weifen Liu
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, People's Republic of China
| | - Xin Gao
- King Abdullah University of Science and Technology (KAUST), Computational Bioscience Research Center (CBRC), Computer, Electrical and Mathematical Sciences and Engineering Division (CEMSE), Thuwal, 23955-6900, Saudi Arabia
| | - Maosheng Xu
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Shiwei Wang
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, People's Republic of China
| | - Lihua Li
- Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, Hangzhou 310018, People's Republic of China
| |
Collapse
|
48
|
Hoivik EA, Hodneland E, Dybvik JA, Wagner-Larsen KS, Fasmer KE, Berg HF, Halle MK, Haldorsen IS, Krakstad C. A radiogenomics application for prognostic profiling of endometrial cancer. Commun Biol 2021; 4:1363. [PMID: 34873276 PMCID: PMC8648740 DOI: 10.1038/s42003-021-02894-5] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2021] [Accepted: 11/09/2021] [Indexed: 12/12/2022] Open
Abstract
Prognostication is critical for accurate diagnosis and tailored treatment in endometrial cancer (EC). We employed radiogenomics to integrate preoperative magnetic resonance imaging (MRI, n = 487 patients) with histologic-, transcriptomic- and molecular biomarkers (n = 550 patients) aiming to identify aggressive tumor features in a study including 866 EC patients. Whole-volume tumor radiomic profiling from manually (radiologists) segmented tumors (n = 138 patients) yielded clusters identifying patients with high-risk histological features and poor survival. Radiomic profiling by a fully automated machine learning (ML)-based tumor segmentation algorithm (n = 336 patients) reproduced the same radiomic prognostic groups. From these radiomic risk-groups, an 11-gene high-risk signature was defined, and its prognostic role was reproduced in orthologous validation cohorts (n = 554 patients) and aligned with The Cancer Genome Atlas (TCGA) molecular class with poor survival (copy-number-high/p53-altered). We conclude that MRI-based integrated radiogenomics profiling provides refined tumor characterization that may aid in prognostication and guide future treatment strategies in EC.
Collapse
Affiliation(s)
- Erling A Hoivik
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway.
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway.
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Erlend Hodneland
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Julie A Dybvik
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kari S Wagner-Larsen
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Kristine E Fasmer
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Hege F Berg
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Mari K Halle
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway
| | - Ingfrid S Haldorsen
- Mohn Medical Imaging and Visualization Centre (MMIV), Department of Radiology, Haukeland University Hospital, Bergen, Norway.
- Section of Radiology, Department of Clinical Medicine, University of Bergen, Bergen, Norway.
| | - Camilla Krakstad
- Centre for Cancer Biomarkers, Department of Clinical Science, University of Bergen, Bergen, Norway.
- Department of Obstetrics and Gynecology, Haukeland University Hospital, Bergen, Norway.
| |
Collapse
|
49
|
Fan M, Cui Y, You C, Liu L, Gu Y, Peng W, Bai Q, Gao X, Li L. Radiogenomic Signatures of Oncotype DX Recurrence Score Enable Prediction of Survival in Estrogen Receptor-Positive Breast Cancer: A Multicohort Study. Radiology 2021; 302:516-524. [PMID: 34846204 DOI: 10.1148/radiol.2021210738] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Background Radiogenomics explores the association between imaging features and genomic assays to uncover relevant prognostic features; however, the prognostic implications of the derived signatures remain unclear. Purpose To identify preoperative radiogenomic signatures of estrogen receptor-positive breast cancer associated with the Oncotype DX recurrence score (RS) and to evaluate whether they are biomarkers for survival and responses to neoadjuvant chemotherapy (NACT). Materials and Methods In this retrospective multicohort study, three data sets were analyzed. The radiogenomic development data set, with preoperative dynamic contrast-enhanced MRI and RS data obtained between January 2016 and October 2019 was used to identify radiogenomic signatures. Prognostic implications of the imaging signatures were assessed by measuring overall survival and recurrence-free survival in the prognostic assessment data set using a multivariable Cox proportional hazards model. The therapeutic implication of the radiogenomic signatures was evaluated by determining their ability to predict the response to NACT using the treatment assessment data set obtained between August 2015 and March 2019. Prediction performance was estimated by using the area under the receiver operating characteristic curve (AUC). Results The final cohorts included a radiogenomic development data set with 130 women (mean age, 52 years ± 10 [standard deviation]), a prognostic assessment data set with 116 women (mean age, 48 years ± 9), and a treatment assessment data set with 135 women (mean age, 50 years ± 11). Radiogenomic signatures (n = 11) of texture and morphologic and statistical features were identified to generate the predicted RS (R2 = 0.33, P < .001). A predicted RS greater than 29.9 was associated with poor overall and recurrence-free survival (P = .001 and P = .007, respectively); predicted RS was greater in women with a good NACT response (30.51 ± 6.92 vs 27.35 ± 4.04 [responders vs nonresponders], P = .001). By combining the predicted RS and complementary features, the model achieved improved performance in prediction of the NACT response (AUC, 0.85; P < .001). Conclusion Radiogenomic signatures associated with genomic assays provide markers of prognosis and treatment in estrogen receptor-positive breast cancer. © RSNA, 2021 Online supplemental material is available for this article.
Collapse
Affiliation(s)
- Ming Fan
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Yajing Cui
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Chao You
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Li Liu
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Yajia Gu
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Weijun Peng
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Qianming Bai
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Xin Gao
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| | - Lihua Li
- From the Institute of Biomedical Engineering and Instrumentation, Hangzhou Dianzi University, High Education Zone, Hangzhou 310018, Zhejiang, China (M.F., Y.C., L. Li); Department of Radiology, Fudan University Shanghai Cancer Center, Shanghai, China (C.Y., L. Liu, Y.G., W.P., Q.B.); and Computational Bioscience Research Center, Computer, Electrical and Mathematical Sciences and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia (X.G.)
| |
Collapse
|
50
|
Zhang L, Fan M, Wang S, Xu M, Li L. Radiomic Analysis of Pharmacokinetic Heterogeneity Within Tumor Based on the Unsupervised Decomposition of Dynamic Contrast-Enhanced MRI for Predicting Histological Characteristics of Breast Cancer. J Magn Reson Imaging 2021; 55:1636-1647. [PMID: 34773446 DOI: 10.1002/jmri.27993] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Revised: 10/31/2021] [Accepted: 11/02/2021] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Breast tumor heterogeneity is associated with histological characteristics. However, pharmacokinetic (PK) heterogeneity within tumor might merit further exploration. PURPOSE To enhance the predictive power of molecular subtypes, Ki-67, and tumor grade by analyzing PK heterogeneity within tumor based on dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI). STUDY TYPE Retrospective. POPULATION Two hundred and eight biopsy-proven breast cancer patients, randomly divided into a training cohort (N = 144) and a testing cohort (N = 64). FIELD STRENGTH/SEQUENCE T1 -weighted DCE-MRI at 3.0 T. ASSESSMENT A convex analysis of mixtures-compartmental modeling decomposition method was used to estimate the PK parameter (i.e., the volume transfer constant Ktrans ) in tumor subregions with distinct physiological kinetic patterns, including fast-flow kinetics, slow-flow kinetics, and plasma input. Radiomic features based on the PK parameter were calculated from each tumor subregion. STATISTICAL TESTS The training cohort was used to build random forest classifiers based on the optimal features determined by the 5-fold cross-validation method. The performance was assessed on the testing cohort using the area under the receiver operating characteristic curve (AUC). The AUCs derived from the tumor subregion-based PK parameter were compared with those of the original images of the entire tumor using the DeLong test. A P-value of <0.05 was considered statistically significant. RESULTS The tumor subregion-based PK parameter, which yielded the highest AUCs of 0.8782, 0.7568, 0.7019, 0.7963, 0.8080, and 0.7375 for luminal A, luminal B, basal-like, human epidermal growth factor receptor 2, Ki-67, and tumor grade, respectively, obtained better diagnostic performance than the original images in the entire tumor (highest AUCs = 0.8612, 0.6191, 0.5593, 0.7704, 0.7494, and 0.6261, respectively). In particular, statistically significant improvement in the diagnostic performance was obtained for luminal B. DATA CONCLUSION Radiomic analysis of PK heterogeneity within tumor can enhance the predictive performance of radiomic models compared with that of the entire tumor. LEVEL OF EVIDENCE 4 TECHNICAL EFFICACY STAGE: 3.
Collapse
Affiliation(s)
- Liangliang Zhang
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China
| | - Ming Fan
- Institute of Biomedical Engineering and Instrumentation, School of Automation, Hangzhou Dianzi University, Hangzhou, China
| | - Shiwei Wang
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Maosheng Xu
- Department of Radiology, First Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, China
| | - Lihua Li
- School of Computer Science and Technology, Hangzhou Dianzi University, Hangzhou, China.,Institute of Biomedical Engineering and Instrumentation, School of Automation, Hangzhou Dianzi University, Hangzhou, China
| |
Collapse
|