1
|
Chen K, Sui C, Wang Z, Liu Z, Qi L, Li X. Habitat radiomics based on CT images to predict survival and immune status in hepatocellular carcinoma, a multi-cohort validation study. Transl Oncol 2025; 52:102260. [PMID: 39752907 PMCID: PMC11754828 DOI: 10.1016/j.tranon.2024.102260] [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: 08/13/2024] [Revised: 11/25/2024] [Accepted: 12/23/2024] [Indexed: 01/25/2025] Open
Abstract
BACKGROUND AND OBJECTIVE Though several clinicopathological features are identified as prognostic indicators, potentially prognostic radiomic models are expected to preoperatively and noninvasively predict survival for HCC. Traditional radiomic models are lacking in a consideration for intratumoral regional heterogeneity. The study aimed to establish and validate the predictive power of multiple habitat radiomic models in predicting prognosis of hepatocellular carcinoma (HCC). METHODS A total of 232 HCC patients were retrospectively included, including a training/validation cohort and two external testing cohorts from 4 centers. For habitat radiomics, intratumoral habitat partitioning based on CT images was first performed by using Otsu thresholding method. Second, a total of 350 habitat radiomic models were constructed to select the optimal model. Then, both ROC curve analyses and Kaplan-Meier survival curve analyses were applied to assess the predictive performances. Ultimately, an immune status profiling was conducted based on bioinformatic analyses and multiplex immunohistochemistry (mIHC) assays to reveal the potential mechanisms. RESULTS A total of 4 habitats were segmented, and the corresponding habitat radiomic models were constructed based on each habitat and an integration of all the four habitats. Generally, habitat radiomic models outperformed traditional radiomic models in stratifying prognosis for HCC. The habitat radiomic model based on the segmented habitat 4 involving decision tree (DT) screening and random forest (RF) classifier was identified as the optimal model with an AUCmean of 0.806. Distinct resting natural killer (NK) cell infiltrations significantly contributed to the prognosis stratification of HCC by the optimal habitat radiomic model. CONCLUSIONS The habitat radiomic model based on CT images was potentially predictive of overall survival for HCC, with a superiority over the traditional radiomic model. The prognostic power of the habitat radiomic model was partly attributed to the distinct immune status captured in the CT images.
Collapse
Affiliation(s)
- Kun Chen
- Department of Nuclear Medicine, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang 310022, China; Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China
| | - Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Ziyang Wang
- Department of Nuclear medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin 300304, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China
| | - Lisha Qi
- Department of Pathology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin 300060, China; Tianjin's Clinical Research Center for Cancer, Tianjin 300060, China.
| |
Collapse
|
2
|
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
|
3
|
Sui C, Su Q, Chen K, Tan R, Wang Z, Liu Z, Xu W, Li X. 18F-FDG PET/CT-based habitat radiomics combining stacking ensemble learning for predicting prognosis in hepatocellular carcinoma: a multi-center study. BMC Cancer 2024; 24:1457. [PMID: 39604895 PMCID: PMC11600565 DOI: 10.1186/s12885-024-13206-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2024] [Accepted: 11/15/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND This study aims to develop habitat radiomic models to predict overall survival (OS) for hepatocellular carcinoma (HCC), based on the characterization of the intratumoral heterogeneity reflected in 18F-FDG PET/CT images. METHODS A total of 137 HCC patients from two institutions were retrospectively included. First, intratumoral habitats were achieved by a two-step unsupervised clustering process based on k-means clustering. Second, a total of 4032 radiomic features were extracted based on each habitat, including 2016 PET-based and 2016 CT-based radiomic features. Then, after feature selection, the stacking ensemble learning approach which combined six machine learning classifiers as the first-level learners with Cox proportional hazards regression as the second-level learner, was employed to build multiple radiomic models. Finally, the optimal model was selected based on the calculation of the C-index, and a combined model integrating with a clinical model was also constructed to identify the potentially complementary effect. RESULTS Three spatially distinct habitats were identified in the two cohorts. Among a total of 30 stacking ensemble learning models established based on different combinations of 5 types of segmented volumes of interest (VOIs) with 6 types of classifiers, the MLP-Cox-habitat-2 model was selected as the optimal radiomic model with a C-index of 0.702 in the external validation cohort. Furthermore, the combined model integrating the optimal radiomic model with the clinical model achieved an improved C-index of 0.747. Consistently, the combined model outperformed the other models for OS prediction, with a time-dependent AUC of 0.835, 0.828, and 0.800 in the 1-year, 2-year, and 3-year OS, respectively. CONCLUSION 18F-FDG PET/CT-based habitat radiomics outperformed traditional radiomics in OS prediction for HCC, with a further improved predictive power by integrating with the clinical model. The optimal combined habitat model was potentially promising in guiding individualized treatment for HCC. TRIAL REGISTRATION This study was a retrospective study, so it was free from registration.
Collapse
Affiliation(s)
- Chunxiao Sui
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Qian Su
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Kun Chen
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, Zhejiang, 310022, China
| | - Rui Tan
- Department of Neurosurgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China
| | - Ziyang Wang
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Cancer Hospital Airport Hospital, Tianjin, 300060, China
| | - Zifan Liu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China
| | - Wengui Xu
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| | - Xiaofeng Li
- Department of Molecular Imaging and Nuclear Medicine, Tianjin Medical University Cancer Institute and Hospital, Tianjin, 300060, China.
- National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin, 300060, China.
| |
Collapse
|
4
|
Yuan J, Wu M, Qiu L, Xu W, Fei Y, Zhu Y, Shi K, Li Y, Luo J, Ding Z, Sun X, Zhou S. Tumor habitat-based MRI features assessing early response in locally advanced nasopharyngeal carcinoma. Oral Oncol 2024; 158:106980. [PMID: 39151333 DOI: 10.1016/j.oraloncology.2024.106980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 07/08/2024] [Accepted: 08/02/2024] [Indexed: 08/19/2024]
Abstract
OBJECTIVE The early response to concurrent chemoradiotherapy in patients with locally advanced nasopharyngeal carcinoma (LA-NPC) is closely correlated with prognosis. In this study, we aimed to predict early response using a combined model that combines sub-regional radiomics features from multi-sequence MRI with clinically relevant factors. METHODS A total of 104 patients with LA-NPC were randomly divided into training and test cohorts at a ratio of 3:1. Radiomic features were extracted from subregions within the tumor area using the K-means clustering method, and feature selection was performed using LASSO regression. Four models were established: a radiomics model, a clinical model, an Intratumor Heterogeneity (ITH) score-based model and a combined model that integrates the ITH score with clinical factors. The predictive performance of these models was evaluated using receiver operating characteristic (ROC) curves, calibration curves, and decision curve analysis (DCA). RESULTS Among the models, the combined model incorporating the ITH score and clinical factors exhibited the highest predictive performance in the test cohort (AUC=0.838). Additionally, the models based on ITH score showed superior prognostic value in both the training cohort (AUC=0.888) and the test cohort (AUC=0.833). CONCLUSION The combined model that integrates the ITH score with clinical factors exhibited superior performance in predicting early response following concurrent chemoradiotherapy in patients with LA-NPC.
Collapse
Affiliation(s)
- Jinling Yuan
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Mengxing Wu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Lei Qiu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Weilin Xu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yinjiao Fei
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yuchen Zhu
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Kexin Shi
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China; The First School of Clinical Medicine, Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Yurong Li
- Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, Zhejiang, China
| | - Jinyan Luo
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China
| | - Zhou Ding
- Department of Radiation Oncology, Lianshui County People's Hospital, Huai'an 223400, Jiangsu, China.
| | - Xinchen Sun
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| | - Shu Zhou
- Department of Radiation Oncology, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, Jiangsu, China.
| |
Collapse
|
5
|
Li Y, Li C, Wei Y, Price S, Schönlieb CB, Chen X. Multi-objective Bayesian optimization with enhanced features for adaptively improved glioblastoma partitioning and survival prediction. Comput Med Imaging Graph 2024; 116:102420. [PMID: 39079409 DOI: 10.1016/j.compmedimag.2024.102420] [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: 12/01/2023] [Revised: 03/30/2024] [Accepted: 07/17/2024] [Indexed: 09/02/2024]
Abstract
Glioblastoma, an aggressive brain tumor prevalent in adults, exhibits heterogeneity in its microstructures and vascular patterns. The delineation of its subregions could facilitate the development of region-targeted therapies. However, current unsupervised learning techniques for this task face challenges in reliability due to fluctuations of clustering algorithms, particularly when processing data from diverse patient cohorts. Furthermore, stable clustering results do not guarantee clinical meaningfulness. To establish the clinical relevance of these subregions, we will perform survival predictions using radiomic features extracted from them. Following this, achieving a balance between outcome stability and clinical relevance presents a significant challenge, further exacerbated by the extensive time required for hyper-parameter tuning. In this study, we introduce a multi-objective Bayesian optimization (MOBO) framework, which leverages a Feature-enhanced Auto-Encoder (FAE) and customized losses to assess both the reproducibility of clustering algorithms and the clinical relevance of their outcomes. Specifically, we embed the entirety of these processes within the MOBO framework, modeling both using distinct Gaussian Processes (GPs). The proposed MOBO framework can automatically balance the trade-off between the two criteria by employing bespoke stability and clinical significance losses. Our approach efficiently optimizes all hyper-parameters, including the FAE architecture and clustering parameters, within a few steps. This not only accelerates the process but also consistently yields robust MRI subregion delineations and provides survival predictions with strong statistical validation.
Collapse
Affiliation(s)
- Yifan Li
- Department of Computer Science, University of Bath, Bath, UK.
| | - Chao Li
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Yiran Wei
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Stephen Price
- Division of Neurosurgery, Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK.
| | - Carola-Bibiane Schönlieb
- Department of Applied Mathematics and Theoretical Physics, University of Cambridge, Cambridge, UK.
| | - Xi Chen
- Department of Computer Science, University of Bath, Bath, UK.
| |
Collapse
|
6
|
Yang P, Shan J, Ge X, Zhou Q, Ding M, Niu T, Du J. Prediction of SBRT response in liver cancer by combining original and delta cone-beam CT radiomics: a pilot study. Phys Eng Sci Med 2024; 47:295-307. [PMID: 38165634 DOI: 10.1007/s13246-023-01366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2023] [Accepted: 12/06/2023] [Indexed: 01/04/2024]
Abstract
This study aims to explore the feasibility of utilizing a combination of original and delta cone-beam CT (CBCT) radiomics for predicting treatment response in liver tumors undergoing stereotactic body radiation therapy (SBRT). A total of 49 patients are included in this study, with 36 receiving 5-fraction SBRT, 3 receiving 4-fraction SBRT, and 10 receiving 3-fraction SBRT. The CBCT and planning CT images from liver cancer patients who underwent SBRT are collected to extract overall 547 radiomics features. The CBCT features which are reproducible and interchangeable with pCT are selected for modeling analysis. The delta features between fractions are calculated to depict tumor change. The patients with 4-fraction SBRT are only used for screening robust features. In patients receiving 5-fraction SBRT, the predictive ability of both original and delta CBCT features for two-level treatment response (local efficacy vs. local non-efficacy; complete response (CR) vs. partial response (PR)) is assessed by utilizing multivariable logistic regression with leave-one-out cross-validation. Additionally, univariate analysis is conducted to validate the capability of CBCT features in identifying local efficacy in patients receiving 3-fraction SBRT. In patients receiving 5-fraction SBRT, the combined models incorporating original and delta CBCT radiomics features demonstrate higher area under the curve (AUC) values compared to models using either original or delta features alone for both classification tasks. The AUC values for predicting local efficacy vs. local non-efficacy are 0.58 for original features, 0.82 for delta features, and 0.90 for combined features. For distinguishing PR from CR, the respective AUC values for original, delta and combined features are 0.79, 0.80, and 0.89. In patients receiving 3-fraction SBRT, eight valuable CBCT radiomics features are identified for predicting local efficacy. The combination of original and delta radiomics derived from fractionated CBCT images in liver cancer patients undergoing SBRT shows promise in providing comprehensive information for predicting treatment response.
Collapse
Affiliation(s)
- Pengfei Yang
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, 100049, China
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China
| | - Jingjing Shan
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Xin Ge
- School of Science, Shenzhen Campus of Sun Yat-sen University, Shenzhen, Guangdong, China
| | - Qinxuan Zhou
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Mingchao Ding
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, 100049, China
| | - Tianye Niu
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, 100049, China.
- Institute of Biomedical Engineering, Shenzhen Bay Laboratory, Shenzhen, China.
| | - Jichen Du
- Peking University Aerospace School of Clinical Medicine, Aerospace Center Hospital, Beijing, 100049, China.
| |
Collapse
|
7
|
Dai Y, Liu D, Xin Y, Li Y, Wang D, He B, Zeng X, Li J, Jia F, Jiang H. Efficacy and Interpretability Analysis of Noninvasive Imaging Based on Computed Tomography in Patients with Hepatocellular Carcinoma After Initial Transarterial Chemoembolization. Acad Radiol 2023; 30 Suppl 1:S61-S72. [PMID: 37393179 DOI: 10.1016/j.acra.2023.05.027] [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: 04/25/2023] [Revised: 05/19/2023] [Accepted: 05/24/2023] [Indexed: 07/03/2023]
Abstract
RATIONALE AND OBJECTIVES The objective of this study is to accurately and timely assess the efficacy of patients with hepatocellular carcinoma (HCC) after the initial transarterial chemoembolization (TACE). MATERIALS AND METHODS This retrospective study consisted of 279 patients with HCC in Center 1, who were split into training and validation cohorts in the ratio of 4:1, and 72 patients in Center 2 as an external testing cohort. Radiomics signatures both in the arterial phase and venous phase of contrast-enhanced computed tomography images were selected by univariate analysis, correlation analysis, and least absolute shrinkage and selection operator regression to build the predicting models. The clinical model and combined model were constructed by independent risk factors after univariate and multivariate logistic regression analysis. The biological interpretability of radiomics signatures correlating transcriptome sequencing data was explored using publicly available data sets. RESULTS A total of 31 radiomics signatures in the arterial phase and 13 radiomics signatures in the venous phase were selected to construct Radscore_arterial and Radscore_venous, respectively, which were independent risk factors. After constructing the combined model, the area under the receiver operating characteristic curve in three cohorts was 0.865, 0.800, and 0.745, respectively. Through correlation analysis, 11 radiomics signatures in the arterial phase and 4 radiomics signatures in the venous phase were associated with 8 and 5 gene modules, respectively (All P < .05), which enriched some pathways closely related to tumor development and proliferation. CONCLUSION Noninvasive imaging has considerable value in predicting the efficacy of patients with HCC after initial TACE. The biological interpretability of the radiological signatures can be mapped at the micro level.
Collapse
Affiliation(s)
- Yanmei Dai
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.)
| | - Dongmin Liu
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.)
| | - Yanjie Xin
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.)
| | - Yuchong Li
- Research Center for Medical Artificial Intelligence, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (Y.L., B.H., F.J.)
| | - Dandan Wang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.)
| | - Baochun He
- Research Center for Medical Artificial Intelligence, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (Y.L., B.H., F.J.)
| | - Xu Zeng
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.)
| | - Jiaping Li
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China (J.L.)
| | - Fucang Jia
- Research Center for Medical Artificial Intelligence, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China (Y.L., B.H., F.J.); Pazhou Lab, Guangzhou, China (F.J.)
| | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, Heilongjiang 150086, China (Y.D., D.L., Y.X., D.W., X.Z., H.J.).
| |
Collapse
|
8
|
Nie P, Zhang J, Miao W, Duan S, Wang T, Zhang J, Gu J, Wang N, Zhang R, Wang X, Yang G, Rao W, Wang Z. Incremental value of radiomics-based heterogeneity to the existing risk criteria in predicting recurrence of hepatocellular carcinoma after liver transplantation. Eur Radiol 2023; 33:6608-6618. [PMID: 37012548 DOI: 10.1007/s00330-023-09591-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 01/02/2023] [Accepted: 02/17/2023] [Indexed: 04/05/2023]
Abstract
OBJECTIVES The aim of the study was to evaluate the association between the radiomics-based intratumoral heterogeneity (ITH) and the recurrence risk in hepatocellular carcinoma (HCC) patients after liver transplantation (LT), and to assess its incremental to the Milan, University of California San Francisco (UCSF), Metro-Ticket 2.0, and Hangzhou criteria. METHODS A multicenter cohort of 196 HCC patients were investigated. The endpoint was recurrence-free survival (RFS) after LT. A CT-based radiomics signature (RS) was constructed and assessed in the whole cohort and in the subgroups stratified by the Milan, UCSF, Metro-Ticket 2.0, and Hangzhou criteria. The R-Milan, R-UCSF, R-Metro-Ticket 2.0, and R-Hangzhou nomograms which combined RS and the four existing risk criteria were developed respectively. The incremental value of RS to the four existing risk criteria in RFS prediction was evaluated. RESULTS RS was significantly associated with RFS in the training and test cohorts as well as in the subgroups stratified by the existing risk criteria. The four combined nomograms showed better predictive capability than the existing risk criteria did with higher C-indices (R-Milan [training/test] vs. Milan, 0.745/0.765 vs. 0.677; R-USCF vs. USCF, 0.748/0.767 vs. 0.675; R-Metro-Ticket 2.0 vs. Metro-Ticket 2.0, 0.756/0.783 vs. 0.670; R-Hangzhou vs. Hangzhou, 0.751/0.760 vs. 0.691) and higher clinical net benefit. CONCLUSIONS The radiomics-based ITH can predict outcomes and provide incremental value to the existing risk criteria in HCC patients after LT. Incorporating radiomics-based ITH in HCC risk criteria may facilitate candidate selection, surveillance, and adjuvant trial design. KEY POINTS • Milan, USCF, Metro-Ticket 2.0, and Hangzhou criteria may be insufficient for outcome prediction in HCC after LT. • Radiomics allows for the characterization of tumor heterogeneity. • Radiomics adds incremental value to the existing criteria in outcome prediction.
Collapse
Affiliation(s)
- Pei Nie
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Juntao Zhang
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Wenjie Miao
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, Shandong, 266061, China
| | - Shaofeng Duan
- GE Healthcare, Precision Health Institution, Shanghai, China
| | - Tongyu Wang
- Department of Radiology, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ju Zhang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, Shandong, 266061, China
| | - Jinyang Gu
- Department of Transplantation, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Ning Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China
| | - Ran Zhang
- Huiying Medical Technology Co. Ltd, Beijing, China
| | - Ximing Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, No. 324, Jingwu Road, Jinan, Shandong, 250021, China.
| | - Guangjie Yang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, Shandong, 266061, China.
| | - Wei Rao
- Division of Hepatology, Liver Disease Center, Organ Transplantation Center, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, Shandong, 266061, China.
| | - Zhenguang Wang
- Department of Nuclear Medicine, The Affiliated Hospital of Qingdao University, No. 59, Haier Road, Qingdao, Shandong, 266061, China.
| |
Collapse
|
9
|
McMahon B, Cohen C, Brown Jr RS, El-Serag H, Ioannou GN, Lok AS, Roberts LR, Singal AG, Block T. Opportunities to address gaps in early detection and improve outcomes of liver cancer. JNCI Cancer Spectr 2023; 7:pkad034. [PMID: 37144952 PMCID: PMC10212536 DOI: 10.1093/jncics/pkad034] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2023] [Accepted: 04/10/2023] [Indexed: 05/06/2023] Open
Abstract
Death rates from primary liver cancer (hepatocellular carcinoma [HCC]) have continued to rise in the United States over the recent decades despite the availability of an increasing range of treatment modalities, including new systemic therapies. Prognosis is strongly associated with tumor stage at diagnosis; however, most cases of HCC are diagnosed beyond an early stage. This lack of early detection has contributed to low survival rates. Professional society guidelines recommend semiannual ultrasound-based HCC screening for at-risk populations, yet HCC surveillance continues to be underused in clinical practice. On April 28, 2022, the Hepatitis B Foundation convened a workshop to discuss the most pressing challenges and barriers to early HCC detection and the need to better leverage existing and emerging tools and technologies that could improve HCC screening and early detection. In this commentary, we summarize technical, patient-level, provider-level, and system-level challenges and opportunities to improve processes and outcomes across the HCC screening continuum. We highlight promising approaches to HCC risk stratification and screening, including new biomarkers, advanced imaging incorporating artificial intelligence, and algorithms for risk stratification. Workshop participants emphasized that action to improve early detection and reduce HCC mortality is urgently needed, noting concern that many of the challenges we face today are the same or similar to those faced a decade ago and that HCC mortality rates have not meaningfully improved. Increasing the uptake of HCC screening was identified as a short-term priority while developing and validating better screening tests and risk-appropriate surveillance strategies.
Collapse
Affiliation(s)
- Brian McMahon
- Liver Disease and Hepatitis Program, Alaska Native Tribal Health Consortium, Anchorage, AK, USA
| | | | - Robert S Brown Jr
- Department of Medicine, Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Hashem El-Serag
- Department of Medicine, Baylor College of Medicine, Houston, TX, USA
| | - George N Ioannou
- Department of Medicine, Division of Gastroenterology, VA Puget Sound Health Care System, Seattle, WA, USA
| | - Anna S Lok
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Lewis R Roberts
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Amit G Singal
- Department of Internal Medicine, Division of Digestive and Liver Diseases, UT Southwestern, Dallas, TX, USA
| | - Timothy Block
- Baruch S. Blumberg Institute and Hepatitis B Foundation, Doylestown, PA, USA
| |
Collapse
|
10
|
Ren Y, Bo L, Shen B, Yang J, Xu S, Shen W, Chen H, Wang X, Chen H, Cai X. Development and validation of a clinical-radiomics model to predict recurrence for patients with hepatocellular carcinoma after curative resection. Med Phys 2023; 50:778-790. [PMID: 36269204 DOI: 10.1002/mp.16061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 09/15/2022] [Accepted: 09/23/2022] [Indexed: 11/08/2022] Open
Abstract
PURPOSE Recurrence is the leading cause of death in hepatocellular carcinoma (HCC) patients with curative resection. In this study, we aimed to develop a preoperative predictive model based on high-throughput radiomics features and clinical factors for prediction of long- and short-term recurrence for these patients. METHODS A total of 270 patients with HCC who were followed up for at least 5 years after curative hepatectomy between June 2014 and December 2017 were enrolled in this retrospective study. Regions of interest were manually delineated in preoperative T2-weighted images using ITK-SNAP software on each HCC tumor slice. A total of 1197 radiomics features were extracted, and the recursive feature elimination method based on logistic regression was used for radiomics signature building. Tenfold cross-validation was applied for model development. Nomograms were constructed and assessed by calibration plot, which compares nomogram-predicated probability with observed outcome. Receiver-operating characteristic was then generated to evaluate the predictive performance of the model in the development and test cohorts. RESULTS The 10 most recurrence-free survival-related radiomics features were selected for the radiomics signatures. A multiparametric clinical-radiomics model combining albumin and radiomics score for recurrence prediction was further established. The integrated model demonstrated good calibration and satisfactory discrimination, with the area under the curve (AUC) of 0.864, 95% CI 0.842-0.903, sensitivity of 0.889, and specificity of 0.644 in the test set. Calibration curve showed good agreement concerning 5-year recurrence risk predicted by the nomogram. In addition, the AUC of 1-, 2-, 3-, and 4-year recurrence was 0.935 (95% CI 0.836-1.000), 0.861 (95% CI 0.723-0.999), 0.878 (95% CI 0.762-0.994), and 0.878 (95% CI 0.762-0.994) in the test set, respectively. CONCLUSIONS The clinical-radiomics model integrating radiomics features and clinical factors can improve recurrence predictions beyond predictions made using clinical factors or radiomics features alone. Our clinical-radiomics model is a valid method to predict recurrence that should improve preoperative prognostic performance and allow more individualized treatment decisions.
Collapse
Affiliation(s)
- Yiyue Ren
- Department of General Surgery, Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Linlin Bo
- Shandong Key Laboratory of Medical Physics and Image Processing, Shandong Institute of Industrial Technology for Health Sciences and Precision Medicine, School of Physics and Electronics, Shandong Normal University, Jinan, Shandong, China
| | - Bo Shen
- Department of Radiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Jing Yang
- Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shufeng Xu
- Department of Radiology, People's Hospital of Quzhou, Quzhou Hospital Affiliated to Wenzhou Medical University, Quzhou, Zhejiang, China
| | - Weiqiang Shen
- Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Hao Chen
- Department of Radiology, Huzhou Central Hospital, Affiliated Central Hospital of Huzhou University; Affiliated Huzhou Hospital, Zhejiang University School of Medicine, Huzhou, Zhejiang, China
| | - Xiaoyan Wang
- Department of Medical Imaging, Bengbu Medical College, Bengbu, Anhui, China
| | - Haipeng Chen
- Deepwise Artificial Intelligence Laboratory, Beijing, China
| | - Xiujun Cai
- Department of General Surgery, Department of Head and Neck Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|
11
|
Miranda J, Horvat N, Fonseca GM, Araujo-Filho JDAB, Fernandes MC, Charbel C, Chakraborty J, Coelho FF, Nomura CH, Herman P. Current status and future perspectives of radiomics in hepatocellular carcinoma. World J Gastroenterol 2023; 29:43-60. [PMID: 36683711 PMCID: PMC9850949 DOI: 10.3748/wjg.v29.i1.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Given the frequent co-existence of an aggressive tumor and underlying chronic liver disease, the management of hepatocellular carcinoma (HCC) patients requires experienced multidisciplinary team discussion. Moreover, imaging plays a key role in the diagnosis, staging, restaging, and surveillance of HCC. Currently, imaging assessment of HCC entails the assessment of qualitative characteristics which are prone to inter-reader variability. Radiomics is an emerging field that extracts high-dimensional mineable quantitative features that cannot be assessed visually with the naked eye from medical imaging. The main potential applications of radiomic models in HCC are to predict histology, response to treatment, genetic signature, recurrence, and survival. Despite the encouraging results to date, there are challenges and limitations that need to be overcome before radiomics implementation in clinical practice. The purpose of this article is to review the main concepts and challenges pertaining to radiomics, and to review recent studies and potential applications of radiomics in HCC.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-010, Brazil
| | - Natally Horvat
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | | | - Maria Clara Fernandes
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Charlotte Charbel
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Jayasree Chakraborty
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | - Cesar Higa Nomura
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Paulo Herman
- Department of Gastroenterology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| |
Collapse
|
12
|
Piao S, Luo X, Bao Y, Hu B, Liu X, Zhu Y, Yang L, Geng D, Li Y. An MRI-based joint model of radiomics and spatial distribution differentiates autoimmune encephalitis from low-grade diffuse astrocytoma. Front Neurol 2022; 13:998279. [PMID: 36408523 PMCID: PMC9669344 DOI: 10.3389/fneur.2022.998279] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2022] [Accepted: 10/12/2022] [Indexed: 04/17/2024] Open
Abstract
BACKGROUND The differential diagnosis between autoimmune encephalitis and low-grade diffuse astrocytoma remains challenging. We aim to develop a quantitative model integrating radiomics and spatial distribution features derived from MRI for discriminating these two conditions. METHODS In our study, we included 188 patients with confirmed autoimmune encephalitis (n = 81) and WHO grade II diffuse astrocytoma (n = 107). Patients with autoimmune encephalitis (AE, n = 59) and WHO grade II diffuse astrocytoma (AS, n = 79) were divided into training and test sets, using stratified sampling according to MRI scanners. We further included an independent validation set (22 patients with AE and 28 patients with AS). Hyperintensity fluid-attenuated inversion recovery (FLAIR) lesions were segmented for each subject. Ten radiomics and eight spatial distribution features were selected via the least absolute shrinkage and selection operator (LASSO), and joint models were constructed by logistic regression for disease classification. Model performance was measured in the test set using the area under the receiver operating characteristic (ROC) curve (AUC). The discrimination performance of the joint model was compared with neuroradiologists. RESULTS The joint model achieved better performance (AUC 0.957/0.908, accuracy 0.914/0.840 for test and independent validation sets, respectively) than the radiomics and spatial distribution models. The joint model achieved lower performance than a senior neuroradiologist (AUC 0.917/0.875) but higher performance than a junior neuroradiologist (AUC 0.692/0.745) in the test and independent validation sets. CONCLUSION The joint model of radiomics and spatial distribution from a single FLAIR could effectively classify AE and AS, providing clinical decision support for the differential diagnosis between the two conditions.
Collapse
Affiliation(s)
- Sirong Piao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Xiao Luo
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yifang Bao
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Bin Hu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Xueling Liu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Yuqi Zhu
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
| | - Liqin Yang
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| | - Yuxin Li
- Department of Radiology, Huashan Hospital, Fudan University, Shanghai, China
- Institute of Functional and Molecular Medical Imaging, Fudan University, Shanghai, China
- Academy for Engineering and Technology, Fudan University, Shanghai, China
| |
Collapse
|
13
|
Li Y, Song Y, Zhang Y, Liu S. Progress in gallbladder cancer with lymph node metastasis. Front Oncol 2022; 12:966835. [PMID: 36072797 PMCID: PMC9441950 DOI: 10.3389/fonc.2022.966835] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Accepted: 08/04/2022] [Indexed: 11/13/2022] Open
Abstract
Gallbladder cancer (GBC) is a malignant tumor that originates from the mucosal lining of the gallbladder. It is distinctly regional and is common in certain geographic regions of developing countries. GBC has a high degree of insidiousness as well as a high propensity for metastatic spread, resulting in the majority of patients being diagnosed at an advanced stage. Lymph node metastasis (LNM) is fairly common in GBC patients and is an independent risk factor for a poor prognosis. This article is focused on the lymph node pathways and metastatic directions of GBC. Furthermore, it summarizes the different lymph node groupings, disease stages and treatments. In the future, it is of great significance to develop individualized treatment and predict the outcomes of GBC patients with different lymph node conditions.
Collapse
Affiliation(s)
- Yuhang Li
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yinghui Song
- Central Laboratory of Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Yujing Zhang
- Key Laboratory of Molecular Epidemiology of Hunan Province, School of Medicine, Hunan Normal University, Changsha, China
| | - Sulai Liu
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Hunan Normal University, Changsha, China
- Central Laboratory of Hunan Provincial People's Hospital/The First Affiliated Hospital of Hunan Normal University, Changsha, China
- *Correspondence: Sulai Liu,
| |
Collapse
|
14
|
Liu H, Wang Y, Liu Y, Lin D, Zhang C, Zhao Y, Chen L, Li Y, Yuan J, Chen Z, Yu J, Kong W, Chen T. Contrast-Enhanced Computed Tomography–Based Radiogenomics Analysis for Predicting Prognosis in Gastric Cancer. Front Oncol 2022; 12:882786. [PMID: 35814414 PMCID: PMC9257248 DOI: 10.3389/fonc.2022.882786] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 05/16/2022] [Indexed: 12/12/2022] Open
Abstract
Objective The aim of this study is to identify prognostic imaging biomarkers and create a radiogenomics nomogram to predict overall survival (OS) in gastric cancer (GC). Material RNA sequencing data from 407 patients with GC and contrast-enhanced computed tomography (CECT) imaging data from 46 patients obtained from The Cancer Genome Atlas (TCGA) and The Cancer Imaging Archive (TCIA) were utilized to identify radiogenomics biomarkers. A total of 392 patients with CECT images from the Nanfang Hospital database were obtained to create and validate a radiogenomics nomogram based on the biomarkers. Methods The prognostic imaging features that correlated with the prognostic gene modules (selected by weighted gene coexpression network analysis) were identified as imaging biomarkers. A nomogram that integrated the radiomics score and clinicopathological factors was created and validated in the Nanfang Hospital database. Nomogram discrimination, calibration, and clinical usefulness were evaluated. Results Three prognostic imaging biomarkers were identified and had a strong correlation with four prognostic gene modules (P < 0.05, FDR < 0.05). The radiogenomics nomogram (AUC = 0.838) resulted in better performance of the survival prediction than that of the TNM staging system (AUC = 0.765, P = 0.011; Delong et al.). In addition, the radiogenomics nomogram exhibited good discrimination, calibration, and clinical usefulness in both the training and validation cohorts. Conclusions The novel prognostic radiogenomics nomogram that was constructed achieved excellent correlation with prognosis in both the training and validation cohort of Nanfang Hospital patients with GC. It is anticipated that this work may assist in clinical preferential treatment decisions and promote the process of precision theranostics in the future.
Collapse
Affiliation(s)
- Han Liu
- Department of Ultrasound, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
| | - Yiyun Wang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Yingqiao Liu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Dingyi Lin
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Cangui Zhang
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Yuyun Zhao
- School of Biomedical Engineering, Southern Medical University, Guangzhou, China
| | - Li Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Yi Li
- Department of Radiology, Southern Medical University, Guangzhou, China
| | - Jianyu Yuan
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Zhao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Jiang Yu
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
| | - Wentao Kong
- Department of Ultrasound, The Affiliated Drum Tower Hospital of Nanjing University Medical School, Nanjing, China
- *Correspondence: Tao Chen, ; Wentao Kong,
| | - Tao Chen
- Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangdong Provincial Engineering Technology Research Center of Minimally Invasive Surgery, Guangzhou, China
- *Correspondence: Tao Chen, ; Wentao Kong,
| |
Collapse
|
15
|
Sheng C, Yang K, He B, Li T, Wang X, Du W, Hu X, Jiang J, Jiang X, Jessen F, Han Y. Cross-Cultural Longitudinal Study on Cognitive Decline (CLoCODE) for Subjective Cognitive Decline in China and Germany: A Protocol for Study Design. J Alzheimers Dis 2022; 87:1319-1333. [PMID: 35431240 DOI: 10.3233/jad-215452] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Background: Subjective cognitive decline (SCD) is considered as the first symptomatic manifestation of Alzheimer’s disease (AD), which is also affected by different cultural backgrounds. Establishing cross-cultural prediction models of SCD is challenging. Objective: To establish prediction models of SCD available for both the Chinese and European populations. Methods: In this project, 330 SCD from China and 380 SCD from Germany are intended to be recruited. For all participants, standardized assessments, including clinical, neuropsychological, apolipoprotein E (APOE) genotype, blood, and multi-parameter magnetic resonance imaging (MRI) at baseline will be conducted. Participants will voluntarily undergo amyloid positron emission tomography (PET) and are classified into amyloid-β (Aβ) positive SCD (SCD+) and Aβ negative SCD (SCD-). First, baseline data of all SCD individuals between the two cohorts will be compared. Then, key features associated with brain amyloidosis will be extracted in SCD+ individuals, and the diagnosis model will be established using the radiomics method. Finally, the follow-up visits will be conducted every 12 months and the primary outcome is the conversion to mild cognitive impairment or dementia. After a 4-year follow-up, we will extract factors associated with the conversion risk of SCD using Cox regression analysis. Results: At present, 141 SCD from China and 338 SCD from Germany have been recruited. Initial analysis showed significant differences in demographic information, neuropsychological tests, and regional brain atrophy in SCD compared with controls in both cohorts. Conclusion: This project may be of great value for future implications of SCD studies in different cultural backgrounds. Trial registration: ClinicalTrials.gov, NCT04696315. Registered 3 January 2021.
Collapse
Affiliation(s)
- Can Sheng
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Kun Yang
- Evidence-Based Medicine Center, Xuanwu Hospital of Capital Medical University, Beijing, China
- Department of Epidemiology and Biostatistics, School of Public Health, Capital Medical University, Beijing, China
| | - Beiqi He
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
| | - Taoran Li
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiaoqi Wang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Wenying Du
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
| | - Xiaochen Hu
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
| | - Jiehui Jiang
- Institute of Biomedical Engineering, School of Information and Communication Engineering, Shanghai University, Shanghai, China
| | - Xueyan Jiang
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- German Center for Neurodegenerative Disease, Clinical Research Group, Bonn, Germany
| | - Frank Jessen
- Department of Psychiatry and Psychotherapy, Medical Faculty, University of Cologne, Cologne, Germany
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
- Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Ying Han
- Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing, China
- Key Laboratory of Biomedical Engineering of Hainan Province, School of Biomedical Engineering, Hainan University, Haikou, China
- Center of Alzheimer’s Disease, Beijing Institute for Brain Disorders, Beijing, China
- National Clinical Research Center for Geriatric Disorders, Beijing, China
| |
Collapse
|
16
|
Li X, Cheng L, Li C, Hu X, Hu X, Tan L, Li Q, Liu C, Wang J. Associating Preoperative MRI Features and Gene Expression Signatures of Early-stage Hepatocellular Carcinoma Patients using Machine Learning. J Clin Transl Hepatol 2022; 10:63-71. [PMID: 35233374 PMCID: PMC8845145 DOI: 10.14218/jcth.2021.00023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 05/07/2021] [Accepted: 05/18/2021] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND AND AIMS The relationship between quantitative magnetic resonance imaging (MRI) imaging features and gene-expression signatures associated with the recurrence of hepatocellular carcinoma (HCC) is not well studied. METHODS In this study, we generated multivariable regression models to explore the correlation between the preoperative MRI features and Golgi membrane protein 1 (GOLM1), SET domain containing 7 (SETD7), and Rho family GTPase 1 (RND1) gene expression levels in a cohort study including 92 early-stage HCC patients. A total of 307 imaging features of tumor texture and shape were computed from T2-weighted MRI. The key MRI features were identified by performing a multi-step feature selection procedure including the correlation analysis and the application of RELIEFF algorithm. Afterward, regression models were generated using kernel-based support vector machines with 5-fold cross-validation. RESULTS The features computed from higher specificity MRI better described GOLM1 and RND1 gene-expression levels, while imaging features computed from lower specificity MRI data were more descriptive for the SETD7 gene. The GOLM1 regression model generated with three features demonstrated a moderate positive correlation (p<0.001), and the RND1 model developed with five variables was positively associated (p<0.001) with gene expression levels. Moreover, RND1 regression model integrating four features was moderately correlated with expressed RND1 levels (p<0.001). CONCLUSIONS The results demonstrated that MRI radiomics features could help quantify GOLM1, SETD7, and RND1 expression levels noninvasively and predict the recurrence risk for early-stage HCC patients.
Collapse
Affiliation(s)
- Xiaoming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Lin Cheng
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Chuanming Li
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xianling Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Xiaofei Hu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
| | - Liang Tan
- Department of Neurosurgery, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
- Department of Electrical and Computer Engineering, Faculty of Science and Technology, University of Macau, Macau, China
| | - Qing Li
- MR Collaborations, Siemens Healthcare Ltd., Shanghai, China
| | - Chen Liu
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
- Correspondence to: Chen Liu and Jian Wang, Department of Radiology, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, China. ORCID: https://orcid.org/0000-0001-5149-2496 (CL), https://orcid.org/0000-0003-1210-0837 (JW). Tel: +86-13108968808 (CL), +86-13883785811 (JW), Fax: +86-23-6546-3026, E-mail: (CL), (JW)
| | - Jian Wang
- Department of Radiology, Southwest Hospital, Third Military Medical University (Army Military Medical University), Chongqing, China
- Correspondence to: Chen Liu and Jian Wang, Department of Radiology, Southwest Hospital, Third Military Medical University, Shapingba District, Chongqing 400038, China. ORCID: https://orcid.org/0000-0001-5149-2496 (CL), https://orcid.org/0000-0003-1210-0837 (JW). Tel: +86-13108968808 (CL), +86-13883785811 (JW), Fax: +86-23-6546-3026, E-mail: (CL), (JW)
| |
Collapse
|
17
|
Shi J, Cui L, Wang H, Dong Y, Yu T, Yang H, Wang X, Liu G, Jiang W, Luo Y, Yang Z, Jiang X. MRI-based intratumoral and peritumoral radiomics on prediction of lymph-vascular space invasion in cervical cancer: A multi-center study. Biomed Signal Process Control 2022. [DOI: 10.1016/j.bspc.2021.103373] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
18
|
Zheng RR, Cai MT, Lan L, Huang XW, Yang YJ, Powell M, Lin F. An MRI-based radiomics signature and clinical characteristics for survival prediction in early-stage cervical cancer. Br J Radiol 2022; 95:20210838. [PMID: 34797703 PMCID: PMC8722251 DOI: 10.1259/bjr.20210838] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
OBJECTIVES To investigate the prognostic role of magnetic resonance imaging (MRI)-based radiomics signature and clinical characteristics for overall survival (OS) and disease-free survival (DFS) in the early-stage cervical cancer. METHODS A total of 207 cervical cancer patients (training cohort: n = 144; validation cohort: n = 63) were enrolled. 792 radiomics features were extracted from T2W and diffusion-weighted imaging (DWI). 19 clinicopathological parameters were collected from the electronic medical record system. Least absolute shrinkage and selection operator (LASSO) regression analysis was used to select significant features to construct prognostic model for OS and DFS. Kaplan-Meier (KM) analysis and log-rank test were applied to identify the association between the radiomics score (Rad-score) and survival time. Nomogram discrimination and calibration were evaluated as well. Associations between radiomics features and clinical parameters were investigated by heatmaps. RESULTS A radiomics signature derived from joint T2W and DWI images showed better prognostic performance than that from either T2W or DWI image alone. Higher Rad-score was associated with worse OS (p < 0.05) and DFS (p < 0.05) in the training and validation set. The joint models outperformed both radiomics model and clinicopathological model alone for 3-year OS and DFS estimation. The calibration curves reached an agreement. Heatmap analysis demonstrated significant associations between radiomics features and clinical characteristics. CONCLUSIONS The MRI-based radiomics nomogram showed a good performance on survival prediction for the OS and DFS in the early-stage cervical cancer. The prediction of the prognostic models could be improved by combining with clinical characteristics, suggesting its potential for clinical application. ADVANCES IN KNOWLEDGE This is the first study to build the radiomics-derived models based on T2W and DWI images for the prediction of survival outcomes on the early-stage cervical cancer patients, and further construct a combined risk scoring system incorporating the clinical features.
Collapse
Affiliation(s)
- Ru-ru Zheng
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Meng-ting Cai
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Li Lan
- Department of Ultrasound Imaging, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Xiao Wan Huang
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Yun Jun Yang
- Department of Radiology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| | - Martin Powell
- Nottingham University Affiliated Hospital, Nottingham Treatment Centre, Nottingham, UK
| | - Feng Lin
- Department of Gynecology, First Affiliated Hospital of Wenzhou Medical University, Wenzhou, PR China
| |
Collapse
|
19
|
Pfaehler E, Zhovannik I, Wei L, Boellaard R, Dekker A, Monshouwer R, El Naqa I, Bussink J, Gillies R, Wee L, Traverso A. A systematic review and quality of reporting checklist for repeatability and reproducibility of radiomic features. Phys Imaging Radiat Oncol 2021; 20:69-75. [PMID: 34816024 PMCID: PMC8591412 DOI: 10.1016/j.phro.2021.10.007] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 10/28/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Main factors impacting feature stability: Image acquisition, reconstruction, tumor segmentation, and interpolation. Textural features are less robust than morphological or statistical features. A checklist is provided including items that should be reported in a radiomic study.
Purpose Although quantitative image biomarkers (radiomics) show promising value for cancer diagnosis, prognosis, and treatment assessment, these biomarkers still lack reproducibility. In this systematic review, we aimed to assess the progress in radiomics reproducibility and repeatability in the recent years. Methods and materials Four hundred fifty-one abstracts were retrieved according to the original PubMed search pattern with the publication dates ranging from 2017/05/01 to 2020/12/01. Each abstract including the keywords was independently screened by four observers. Forty-two full-text articles were selected for further analysis. Patient population data, radiomic feature classes, feature extraction software, image preprocessing, and reproducibility results were extracted from each article. To support the community with a standardized reporting strategy, we propose a specific reporting checklist to evaluate the feasibility to reproduce each study. Results Many studies continue to under-report essential reproducibility information: all but one clinical and all but two phantom studies missed to report at least one important item reporting image acquisition. The studies included in this review indicate that all radiomic features are sensitive to image acquisition, reconstruction, tumor segmentation, and interpolation. However, the amount of sensitivity is feature dependent, for instance, textural features were, in general, less robust than statistical features. Conclusions Radiomics repeatability, reproducibility, and reporting quality can substantially be improved regarding feature extraction software and settings, image preprocessing and acquisition, cutoff values for stable feature selection. Our proposed radiomics reporting checklist can serve to simplify and improve the reporting and, eventually, guarantee the possibility to fully replicate and validate radiomic studies.
Collapse
Affiliation(s)
- Elisabeth Pfaehler
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Ivan Zhovannik
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Lise Wei
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Ronald Boellaard
- Department of Nuclear Medicine and Molecular Imaging, Medical Imaging Center, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Radiology & Nuclear Medicine, VU University Medical Center, Amsterdam, The Netherlands
| | - Andre Dekker
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - René Monshouwer
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Issam El Naqa
- Department of Radiation Oncology, University of Michigan, Ann Arbor, MI, USA
| | - Jan Bussink
- Department of Radiation Oncology, Radboud Institute for Health Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert Gillies
- Department of Radiology, Moffitt Cancer Center, Tampa, FL, USA
| | - Leonard Wee
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| | - Alberto Traverso
- Department of Radiation Oncology (MAASTRO), GROW School for Oncology, Maastricht University Medical Centre+, Maastricht, The Netherlands
| |
Collapse
|
20
|
Fan Y, Dong Y, Yang H, Chen H, Yu Y, Wang X, Wang X, Yu T, Luo Y, Jiang X. Subregional radiomics analysis for the detection of the EGFR mutation on thoracic spinal metastases from lung cancer. Phys Med Biol 2021; 66. [PMID: 34633298 DOI: 10.1088/1361-6560/ac2ea7] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Accepted: 10/11/2021] [Indexed: 01/20/2023]
Abstract
The present study intended to use radiomic analysis of spinal metastasis subregions to detect epidermal growth factor receptor (EGFR) mutation. In total, 94 patients with thoracic spinal metastasis originated from primary lung adenocarcinoma (2017-2020) were studied. All patients underwent T1-weighted (T1W) and T2 fat-suppressed (T2FS) MRI scans. The spinal metastases (tumor region) were subdivided into phenotypically consistent subregions based on patient- and population-level clustering: Three subregions, S1, S2 and S3, and the total tumor region. Radiomics features were extracted from each subregion and from the whole tumor region as well. Least shrinkage and selection operator (LASSO) regression were used for feature selection and radiomics signature definition. Detection performance of S3 was better than all other regions using T1W (AUCs, S1 versus S2 versus S3 versus whole tumor, 0.720 versus 0.764 versus 0.786 versus 0.758) and T2FS (AUCs, S1 versus S2 versus S3 versus whole tumor, 0.791 versus 0.708 versus 0.838 versus 0.797) MRI. The multi-regional radiomics signature derived from the joint of inner subregion S3 from T1W and T2FS MRI achieved the best detection capabilities with AUCs of 0.879 (ACC = 0.774, SEN = 0.838, SPE = 0.840) and 0.777 (ACC = 0.688, SEN = 0.947, SPE = 0.615) in the training and test sets, respectively. Our study revealed that MRI-based radiomic analysis of spinal metastasis subregions has the potential to detect the EGFR mutation in patients with primary lung adenocarcinoma.
Collapse
Affiliation(s)
- Ying Fan
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Yue Dong
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Huazhe Yang
- Department of Biophysics, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| | - Huanhuan Chen
- Department of Oncology, Shengjing Hospital of China Medical University, Shenyang, 110004, People's Republic of China
| | - Yalian Yu
- Department of Otorhinolaryngology, the First Affiliated Hospital of China Medical University, Shenyang, 110122, People's Republic of China
| | - Xiaoyu Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xinling Wang
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Tao Yu
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Yahong Luo
- Department of Radiology, Cancer Hospital of China Medical University, Liaoning Cancer Hospital and Institute, Shenyang, 110042, People's Republic of China
| | - Xiran Jiang
- Department of Biomedical Engineering, School of Intelligent Medicine, China Medical University, Shenyang, 110122, People's Republic of China
| |
Collapse
|
21
|
Liu X, Liang X, Ruan L, Yan S. A Clinical-Radiomics Nomogram for Preoperative Prediction of Lymph Node Metastasis in Gallbladder Cancer. Front Oncol 2021; 11:633852. [PMID: 34631512 PMCID: PMC8493033 DOI: 10.3389/fonc.2021.633852] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2020] [Accepted: 08/31/2021] [Indexed: 12/24/2022] Open
Abstract
Objectives The aim of the current study was to develop and validate a nomogram based on CT radiomics features and clinical variables for predicting lymph node metastasis (LNM) in gallbladder cancer (GBC). Methods A total of 353 GBC patients from two hospitals were enrolled in this study. A Radscore was developed using least absolute shrinkage and selection operator (LASSO) logistic model based on the radiomics features extracted from the portal venous-phase computed tomography (CT). Four prediction models were constructed based on the training cohort and were validated using internal and external validation cohorts. The most effective model was then selected to build a nomogram. Results The clinical-radiomics nomogram, which comprised Radscore and three clinical variables, showed the best diagnostic efficiency in the training cohort (AUC = 0.851), internal validation cohort (AUC = 0.819), and external validation cohort (AUC = 0.824). Calibration curves showed good discrimination ability of the nomogram using the validation cohorts. Decision curve analysis (DCA) showed that the nomogram had a high clinical utility. Conclusion The findings showed that the clinical-radiomics nomogram based on radiomics features and clinical parameters is a promising tool for preoperative prediction of LN status in patients with GBC.
Collapse
Affiliation(s)
- Xingyu Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaoyuan Liang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Lingxiang Ruan
- Department of Radiology, The First Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| | - Sheng Yan
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
22
|
Harding-Theobald E, Louissaint J, Maraj B, Cuaresma E, Townsend W, Mendiratta-Lala M, Singal AG, Su GL, Lok AS, Parikh ND. Systematic review: radiomics for the diagnosis and prognosis of hepatocellular carcinoma. Aliment Pharmacol Ther 2021; 54:890-901. [PMID: 34390014 PMCID: PMC8435007 DOI: 10.1111/apt.16563] [Citation(s) in RCA: 74] [Impact Index Per Article: 18.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/08/2021] [Accepted: 07/25/2021] [Indexed: 12/12/2022]
Abstract
BACKGROUND Advances in imaging technology have the potential to transform the early diagnosis and treatment of hepatocellular carcinoma (HCC) through quantitative image analysis. Computational "radiomic" techniques extract biomarker information from images which can be used to improve diagnosis and predict tumour biology. AIMS To perform a systematic review on radiomic features in HCC diagnosis and prognosis, with a focus on reporting metrics and methodologic standardisation. METHODS We performed a systematic review of all full-text articles published from inception through December 1, 2019. Standardised data extraction and quality assessment metrics were applied to all studies. RESULTS A total of 54 studies were included for analysis. Radiomic features demonstrated good discriminatory performance to differentiate HCC from other solid lesions (c-statistics 0.66-0.95), and to predict microvascular invasion (c-statistic 0.76-0.92), early recurrence after hepatectomy (c-statistics 0.71-0.86), and prognosis after locoregional or systemic therapies (c-statistics 0.74-0.81). Common stratifying features for diagnostic and prognostic radiomic tools included analyses of imaging skewness, analysis of the peritumoural region, and feature extraction from the arterial imaging phase. The overall quality of the included studies was low, with common deficiencies in both internal and external validation, standardised imaging segmentation, and lack of comparison to a gold standard. CONCLUSIONS Quantitative image analysis demonstrates promise as a non-invasive biomarker to improve HCC diagnosis and management. However, standardisation of protocols and outcome measurement, sharing of algorithms and analytic methods, and external validation are necessary prior to widespread application of radiomics to HCC diagnosis and prognosis in clinical practice.
Collapse
Affiliation(s)
- Emily Harding-Theobald
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Jeremy Louissaint
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Bharat Maraj
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Edward Cuaresma
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Whitney Townsend
- Division of Library Sciences, University of Michigan, Ann Arbor, MI, USA
| | | | - Amit G Singal
- Division of Digestive and Liver Diseases, University of Texas Southwestern, Dallas, TX, USA
| | - Grace L Su
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Anna S Lok
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, University of Michigan Health System, Ann Arbor, MI, USA
| |
Collapse
|
23
|
Xu H, Lv W, Feng H, Du D, Yuan Q, Wang Q, Dai Z, Yang W, Feng Q, Ma J, Lu L. Subregional Radiomics Analysis of PET/CT Imaging with Intratumor Partitioning: Application to Prognosis for Nasopharyngeal Carcinoma. Mol Imaging Biol 2021; 22:1414-1426. [PMID: 31659574 DOI: 10.1007/s11307-019-01439-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
PURPOSE This work aims to identify intratumoral habitats with distinct heterogeneity based on 2-deoxy-2-[18F]fluro-D-glucose positron emission tomography (PET)/X-ray computed tomography (CT) imaging, and to develop a subregional radiomics approach to predict progression-free survival (PFS) in patients with nasopharyngeal carcinoma (NPC). PROCEDURES In total, 128 NPC patients (85 vs. 43 for primary vs. validation cohorts) who underwent pre-treatment PET/CT scan were enrolled retrospectively. Each tumor was partitioned into several phenotypically consistent subregions based on individual- and population-level clustering. For each subregion, 202 radiomics features were extracted to construct imaging biomarker for prognosis via Cox's proportional hazard model combined with forward stepwise feature selection. Relevance of imaging biomarkers and clinicopathological factors were assessed by multivariate Cox regression analysis and Spearman's correlation analysis. To investigate whether imaging biomarkers could provide complementary prognosis information beyond existing predictors, a scoring system was further developed for risk stratification and compared with AJCC staging system. RESULTS Three subregions (denoted as S1, S2, and S3) were discovered with distinct PET/CT imaging characteristics in the two cohorts. The prognostic performance of imaging biomarker S3 outperformed the whole tumor (C-index, 0.69 vs. 0.58; log-rank test, p < 0.001 vs. p = 0.552). Imaging biomarker S3 and AJCC stage were identified as independent predictors (p = 0.011 and 0.042, respectively) after adjusting for clinicopathological factors. The scoring system outperformed the traditional AJCC staging system (log-rank test, p < 0.0001 vs. p = 0.0002 in primary cohort and p = 0.0021 vs. p = 0.0277 in validation cohort, respectively). CONCLUSIONS Subregional radiomics analysis of PET/CT imaging has the potential to predict PFS in patients with NPC, which also provides complementary prognostic information for traditional predictors.
Collapse
Affiliation(s)
- Hui Xu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Wenbing Lv
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Hui Feng
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Dongyang Du
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qingyu Yuan
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Quanshi Wang
- Nanfang PET Center, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Zhenhui Dai
- Department of Radiotherapy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510120, China
| | - Wei Yang
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Qianjin Feng
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Jianhua Ma
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China
| | - Lijun Lu
- School of Biomedical Engineering and Guangdong Provincial Key Laboratory of Medical Image Processing, Southern Medical University, Guangzhou, Guangdong, 510515, China.
| |
Collapse
|
24
|
Borhani AA, Catania R, Velichko YS, Hectors S, Taouli B, Lewis S. Radiomics of hepatocellular carcinoma: promising roles in patient selection, prediction, and assessment of treatment response. Abdom Radiol (NY) 2021; 46:3674-3685. [PMID: 33891149 DOI: 10.1007/s00261-021-03085-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Revised: 04/02/2021] [Accepted: 04/09/2021] [Indexed: 12/13/2022]
Abstract
Radiomics refers to the process of conversion of conventional medical images into quantifiable data ("features") which can be further mined to reveal complex patterns and relationships between the voxels in the image. These high throughput features can potentially reflect the histology of biologic tissues at macroscopic and microscopic levels. Several studies have investigated radiomics of hepatocellular carcinoma (HCC) before and after treatment. HCC is a heterogeneous disease with diverse phenotypical and genotypical landscape. Due to this inherent heterogeneity, HCC lesions can manifest variable aggressiveness with different response to treatment options, including the newer targeted therapies. Hence, radiomics can be used as a potential tool to enable patient selection for therapies and to predict response to treatments and outcome. Additionally, radiomics may serve as a tool for earlier and more efficient assessment of response to treatment. Radiomics, radiogenomics, and radio-immunoprofiling and their potential roles in management of patients with HCC will be discussed and critically reviewed in this article.
Collapse
Affiliation(s)
- Amir A Borhani
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. Saint Clair Street, Suite 800, Chicago, IL, 60611, USA.
| | - Roberta Catania
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. Saint Clair Street, Suite 800, Chicago, IL, 60611, USA
| | - Yuri S Velichko
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. Saint Clair Street, Suite 800, Chicago, IL, 60611, USA
| | - Stefanie Hectors
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine At Mount Sinai, 1Gustave L. Levy Place, New York, NY, 1470, USA
| | - Bachir Taouli
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine At Mount Sinai, 1Gustave L. Levy Place, New York, NY, 1470, USA
| | - Sara Lewis
- BioMedical Engineering and Imaging Institute, Icahn School of Medicine At Mount Sinai, New York, NY, USA
- Department of Diagnostic, Molecular and Interventional Radiology, Icahn School of Medicine At Mount Sinai, 1Gustave L. Levy Place, New York, NY, 1470, USA
| |
Collapse
|
25
|
Li Z, Zhong Q, Zhang L, Wang M, Xiao W, Cui F, Yu F, Huang C, Feng Z. Computed Tomography-Based Radiomics Model to Preoperatively Predict Microsatellite Instability Status in Colorectal Cancer: A Multicenter Study. Front Oncol 2021; 11:666786. [PMID: 34277413 PMCID: PMC8281816 DOI: 10.3389/fonc.2021.666786] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 06/16/2021] [Indexed: 12/11/2022] Open
Abstract
Objectives To establish and validate a combined radiomics model based on radiomics features and clinical characteristics, and to predict microsatellite instability (MSI) status in colorectal cancer (CRC) patients preoperatively. Methods A total of 368 patients from four hospitals, who underwent preoperative contrast-enhanced CT examination, were included in this study. The data of 226 patients from a single hospital were used as the training dataset. The data of 142 patients from the other three hospitals were used as an independent validation dataset. The regions of interest were drawn on the portal venous phase of contrast-enhanced CT images. The filtered radiomics features and clinical characteristics were combined. A total of 15 different discrimination models were constructed based on a feature selection strategy from a pool of 3 feature selection methods and a classifier from a pool of 5 classification algorithms. The generalization capability of each model was evaluated in an external validation set. The model with high area under the curve (AUC) value from the training set and without a significant decrease in the external validation set was final selected. The Brier score (BS) was used to quantify overall performance of the selected model. Results The logistic regression model using the mutual information (MI) dimensionality reduction method was final selected with an AUC value of 0.79 for the training set and 0.73 for the external validation set to predicting MSI. The BS value of the model was 0.12 in the training set and 0.19 in the validation set. Conclusion The established combined radiomics model has the potential to predict MSI status in CRC patients preoperatively.
Collapse
Affiliation(s)
- Zhi Li
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qi Zhong
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Liang Zhang
- Department of Radiology, Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Hangzhou, China
| | - Minhong Wang
- Department of Radiology, First Affiliated Hospital of Wannan Medical College, Wuhu, China
| | - Wenbo Xiao
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Feng Cui
- Department of Radiology, Hangzhou Hospital of Traditional Chinese Medicine, Hangzhou, China
| | - Fang Yu
- Department of Pathology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chencui Huang
- Department of Research Collaboration, R&D Center, Beijing Deepwise & League of PHD Technology Co, Ltd, Beijing, China
| | - Zhan Feng
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
26
|
Tong Y, Sun P, Yong J, Zhang H, Huang Y, Guo Y, Yu J, Zhou S, Wang Y, Wang Y, Ji Q, Wang Y, Chang C. Radiogenomic Analysis of Papillary Thyroid Carcinoma for Prediction of Cervical Lymph Node Metastasis: A Preliminary Study. Front Oncol 2021; 11:682998. [PMID: 34268116 PMCID: PMC8276635 DOI: 10.3389/fonc.2021.682998] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 06/09/2021] [Indexed: 12/19/2022] Open
Abstract
Background Papillary thyroid carcinoma (PTC) is characterized by frequent metastases to cervical lymph nodes (CLNs), and the presence of lymph node metastasis at diagnosis has a significant impact on the surgical approach. Therefore, we established a radiomic signature to predict the CLN status of PTC patients using preoperative thyroid ultrasound, and investigated the association between the radiomic features and underlying molecular characteristics of PTC tumors. Methods In total, 270 patients were enrolled in this prospective study, and radiomic features were extracted according to multiple guidelines. A radiomic signature was built with selected features in the training cohort and validated in the validation cohort. The total protein extracted from tumor samples was analyzed with LC/MS and iTRAQ technology. Gene modules acquired by clustering were chosen for their diagnostic significance. A radiogenomic map linking radiomic features to gene modules was constructed with the Spearman correlation matrix. Genes in modules related to metastasis were extracted for Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) pathway enrichment analyses, and a protein-protein interaction (PPI) network was built to identify the hub genes in the modules. Finally, the screened hub genes were validated by immunohistochemistry analysis. Results The radiomic signature showed good performance for predicting CLN status in training and validation cohorts, with area under curve of 0.873 and 0.831 respectively. A radiogenomic map was created with nine significant correlations between radiomic features and gene modules, and two of them had higher correlation coefficient. Among these, MEmeganta representing the upregulation of telomere maintenance via telomerase and cell-cell adhesion was correlated with ‘Rectlike’ and ‘deviation ratio of tumor tissue and normal thyroid gland’ which reflect the margin and the internal echogenicity of the tumor, respectively. MEblue capturing cell-cell adhesion and glycolysis was associated with feature ‘minimum calcification area’ which measures the punctate calcification. The hub genes of the two modules were identified by protein-protein interaction network. Immunohistochemistry validated that LAMC1 and THBS1 were differently expressed in metastatic and non-metastatic tissues (p=0.003; p=0.002). And LAMC1 was associated with feature ‘Rectlike’ and ‘deviation ratio of tumor and normal thyroid gland’ (p<0.001; p<0.001); THBS1 was correlated with ‘minimum calcification area’ (p<0.001). Conclusions The radiomic signature proposed here has the potential to noninvasively predict the CLN status in PTC patients. Merging imaging phenotypes with genomic data could allow noninvasive identification of the molecular properties of PTC tumors, which might support clinical decision making and personalized management.
Collapse
Affiliation(s)
- Yuyang Tong
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China.,Department of Surgical Oncology, The Ohio State University, Columbus, OH, United States
| | - Peixuan Sun
- Diagnostic Imaging Center, Shanghai Children's Medical Center, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Juanjuan Yong
- Department of Pathology, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hongbo Zhang
- Pharmaceutical Sciences Laboratory, Åbo Akademi University, Turku, Finland.,Turku Biosciences Center, University of Turku and Åbo Akademi University, Turku, Finland
| | - Yunxia Huang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yi Guo
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Jinhua Yu
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Shichong Zhou
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yulong Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yu Wang
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qinghai Ji
- Department of Head and Neck Surgery, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yuanyuan Wang
- Department of Electronic Engineering, Fudan University and Key Laboratory of Medical Imaging Computing and Computer Assisted Intervention of Shanghai, Shanghai, China
| | - Cai Chang
- Department of Ultrasound, Fudan University Shanghai Cancer Center, Shanghai, China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| |
Collapse
|
27
|
Jin ZC, Zhong BY. Application of radiomics in hepatocellular carcinoma: A review. Artif Intell Med Imaging 2021; 2:64-72. [DOI: 10.35711/aimi.v2.i3.64] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2021] [Revised: 06/19/2021] [Accepted: 06/30/2021] [Indexed: 02/06/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer with low 5-year survival rate. The high molecular heterogeneity in HCC poses huge challenges for clinical practice or trial design and has become a major barrier to improving the management of HCC. However, current clinical practice based on single bioptic or archived tumor tissue has been deficient in identifying useful biomarkers. The concept of radiomics was first proposed in 2012 and is different from the traditional imaging analysis based on the qualitative or semi-quantitative analysis by radiologists. Radiomics refers to high-throughput extraction of large amounts number of high-dimensional quantitative features from medical images through machine learning or deep learning algorithms. Using the radiomics method could quantify tumoral phenotypes and heterogeneity, which may provide benefits in clinical decision-making at a lower cost. Here, we review the workflow and application of radiomics in HCC.
Collapse
Affiliation(s)
- Zhi-Cheng Jin
- Center of Interventional Radiology and Vascular Surgery, Department of Radiology, Zhongda Hospital, Medical School, Southeast University, Nanjing 210009, Jiangsu Province, China
| | - Bin-Yan Zhong
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou 215006, Jiangsu Province, China
| |
Collapse
|
28
|
The status of medical physics in radiotherapy in China. Phys Med 2021; 85:147-157. [PMID: 34010803 DOI: 10.1016/j.ejmp.2021.05.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 05/01/2021] [Accepted: 05/03/2021] [Indexed: 01/09/2023] Open
Abstract
PURPOSE To present an overview of the status of medical physics in radiotherapy in China, including facilities and devices, occupation, education, research, etc. MATERIALS AND METHODS: The information about medical physics in clinics was obtained from the 9-th nationwide survey conducted by the China Society for Radiation Oncology in 2019. The data of medical physics in education and research was collected from the publications of the official and professional organizations. RESULTS By 2019, there were 1463 hospitals or institutes registered to practice radiotherapy and the number of accelerators per million population was 1.5. There were 4172 medical physicists working in clinics of radiation oncology. The ratio between the numbers of radiation oncologists and medical physicists is 3.51. Approximately, 95% of medical physicists have an undergraduate or graduate degrees in nuclear physics and biomedical engineering. 86% of medical physicists have certificates issued by the Chinese Society of Medical Physics. There has been a fast growth of publications by authors from mainland of China in the top international medical physics and radiotherapy journals since 2018. CONCLUSIONS Demand for medical physicists in radiotherapy increased quickly in the past decade. The distribution of radiotherapy facilities in China became more balanced. High quality continuing education and training programs for medical physicists are deficient in most areas. The role of medical physicists in the clinic has not been clearly defined and their contributions have not been fully recognized by the community.
Collapse
|
29
|
Shang S, Sun J, Yue Z, Wang Y, Wang X, Luo Y, Zhao D, Yu T, Jiang X. Multi-parametric MRI based radiomics with tumor subregion partitioning for differentiating benign and malignant soft-tissue tumors. Biomed Signal Process Control 2021. [DOI: 10.1016/j.bspc.2021.102522] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
|
30
|
Cui S, Tang T, Su Q, Wang Y, Shu Z, Yang W, Gong X. Radiomic nomogram based on MRI to predict grade of branching type intraductal papillary mucinous neoplasms of the pancreas: a multicenter study. Cancer Imaging 2021; 21:26. [PMID: 33750453 PMCID: PMC7942000 DOI: 10.1186/s40644-021-00395-6] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 02/26/2021] [Indexed: 12/15/2022] Open
Abstract
Background Accurate diagnosis of high-grade branching type intraductal papillary mucinous neoplasms (BD-IPMNs) is challenging in clinical setting. We aimed to construct and validate a nomogram combining clinical characteristics and radiomic features for the preoperative prediction of low and high-grade in BD-IPMNs. Methods Two hundred and two patients from three medical centers were enrolled. The high-grade BD-IPMN group comprised patients with high-grade dysplasia and invasive carcinoma in BD-IPMN (n = 50). The training cohort comprised patients from the first medical center (n = 103), and the external independent validation cohorts comprised patients from the second and third medical centers (n = 48 and 51). Within 3 months prior to surgery, all patients were subjected to magnetic resonance examination. The volume of interest was delineated on T1-weighted (T1-w) imaging, T2-weighted (T2-w) imaging, and contrast-enhanced T1-weighted (CET1-w) imaging, respectively, on each tumor slice. Quantitative image features were extracted using MITK software (G.E.). The Mann-Whitney U test or independent-sample t-test, and LASSO regression, were applied for data dimension reduction, after which a radiomic signature was constructed for grade assessment. Based on the training cohort, we developed a combined nomogram model incorporating clinical variables and the radiomic signature. Decision curve analysis (DCA), a receiver operating characteristic curve (ROC), a calibration curve, and the area under the ROC curve (AUC) were used to evaluate the utility of the constructed model based on the external independent validation cohorts. Results To predict tumor grade, we developed a nine-feature-combined radiomic signature. For the radiomic signature, the AUC values of high-grade disease were 0.836 in the training cohort, 0.811 in external validation cohort 1, and 0.822 in external validation cohort 2. The CA19–9 level and main pancreatic duct size were identified as independent parameters of high-grade of BD-IPMNs using multivariate logistic regression analysis. The CA19–9 level and main pancreatic duct size were then used to construct the radiomic nomogram. Using the radiomic nomogram, the high-grade disease-associated AUC values were 0.903 (training cohort), 0.884 (external validation cohort 1), and 0.876 (external validation cohort 2). The clinical utility of the developed nomogram was verified using the calibration curve and DCA. Conclusions The developed radiomic nomogram model could effectively distinguish high-grade patients with BD-IPMNs preoperatively. This preoperative identification might improve treatment methods and promote personalized therapy in patients with BD-IPMNs. Supplementary Information The online version contains supplementary material available at 10.1186/s40644-021-00395-6.
Collapse
Affiliation(s)
- Sijia Cui
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310000, China.,The Second Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310053, China
| | - Tianyu Tang
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China.,Zhejiang Provincial Key Laboratory of Pancreatic Disease, Hangzhou, China
| | - Qiuming Su
- Department of General Surgery, The Second Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yajie Wang
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310000, China
| | - Zhenyu Shu
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310000, China
| | - Wei Yang
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310000, China.,Bengbu Medical College, Bengbu, 233000, China
| | - Xiangyang Gong
- Department of Radiology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital of Hangzhou Medical College, 158 Shangtang Road, Hangzhou, 310000, China. .,Institute of Artificial Intelligence and Remote Imaging, Hangzhou Medical College, Hangzhou, China.
| |
Collapse
|
31
|
Shui L, Ren H, Yang X, Li J, Chen Z, Yi C, Zhu H, Shui P. The Era of Radiogenomics in Precision Medicine: An Emerging Approach to Support Diagnosis, Treatment Decisions, and Prognostication in Oncology. Front Oncol 2021; 10:570465. [PMID: 33575207 PMCID: PMC7870863 DOI: 10.3389/fonc.2020.570465] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/08/2020] [Indexed: 02/05/2023] Open
Abstract
With the rapid development of new technologies, including artificial intelligence and genome sequencing, radiogenomics has emerged as a state-of-the-art science in the field of individualized medicine. Radiogenomics combines a large volume of quantitative data extracted from medical images with individual genomic phenotypes and constructs a prediction model through deep learning to stratify patients, guide therapeutic strategies, and evaluate clinical outcomes. Recent studies of various types of tumors demonstrate the predictive value of radiogenomics. And some of the issues in the radiogenomic analysis and the solutions from prior works are presented. Although the workflow criteria and international agreed guidelines for statistical methods need to be confirmed, radiogenomics represents a repeatable and cost-effective approach for the detection of continuous changes and is a promising surrogate for invasive interventions. Therefore, radiogenomics could facilitate computer-aided diagnosis, treatment, and prediction of the prognosis in patients with tumors in the routine clinical setting. Here, we summarize the integrated process of radiogenomics and introduce the crucial strategies and statistical algorithms involved in current studies.
Collapse
Affiliation(s)
- Lin Shui
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Haoyu Ren
- Department of General, Visceral and Transplantation Surgery, University Hospital, LMU Munich, Munich, Germany
| | - Xi Yang
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Jian Li
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, China
| | - Ziwei Chen
- Department of Nephrology, Chengdu Integrated TCM and Western Medicine Hospital, Chengdu, China
| | - Cheng Yi
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Pixian Shui
- School of Pharmacy, Southwest Medical University, Luzhou, China
| |
Collapse
|
32
|
Abstract
PURPOSE OF REVIEW Radiogenomics is a growing field that has garnered immense interest over the past decade, owing to its numerous applications in the field of oncology and its potential value in improving patient outcomes. Current applications have only begun to delve into the potential of radiogenomics, and particularly in interventional oncology, there is room for development and increased value of these applications. RECENT FINDINGS The field of interventional oncology (IO) has seen valuable radiogenomic applications, from prediction of response to locoregional therapies in hepatocellular carcinoma to identification of genetic mutations in non-small cell lung cancer. Future directions that can increase the value of radiogenomics include applications that address tumor heterogeneity, predict immune responsiveness of tumors, and differentiate between oligoprogression and early widespread progression, among others. Radiogenomics, whether in terms of methodologies or applications, is still in the early stages of development and far from maturation. Future applications, particularly in the field of interventional oncology, will allow realization of its full potential.
Collapse
|
33
|
Abstract
The diagnosis of hepatocellular carcinoma relies largely on non-invasive imaging, and is well suited for radiomics analysis. Radiomics is an emerging method for quantification of tumor heterogeneity by mathematically analyzing the spatial distribution and relationships of gray levels in medical images. The published studies on radiomics analysis of HCC provide encouraging data demonstrating potential utility for prediction of tumor biology, molecular profiles, post-therapy response, and outcome. The combination of radiomics data and clinical/laboratory information provides added value in many studies. Radiomics is a multi-step process that requires optimization and standardization, the development of semi-automated or automated segmentation methods, robust data quality control, and refinement of algorithms and modeling approaches for high-throughput data analysis. While radiomics remains largely in the research setting, the strong associations of predictive models and nomograms with certain pathologic, molecular, and immune markers with tumor aggressiveness and patient outcomes, provide great potential for clinical applications to inform optimized treatment strategies and patient prognosis.
Collapse
|
34
|
Liu Z, Wu K, Wu B, Tang X, Yuan H, Pang H, Huang Y, Zhu X, Luo H, Qi Y. Imaging genomics for accurate diagnosis and treatment of tumors: A cutting edge overview. Biomed Pharmacother 2020; 135:111173. [PMID: 33383370 DOI: 10.1016/j.biopha.2020.111173] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2020] [Revised: 11/30/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
Imaging genomics refers to the establishment of the connection between invasive gene expression features and non-invasive imaging features. Tumor imaging genomics can not only understand the macroscopic phenotype of tumor, but also can deeply analyze the cellular and molecular characteristics of tumor tissue. In recent years, tumor imaging genomics has been a key in the field of medicine. The incidence of cancer in China has increased significantly, which is the main reason of disease death of urban residents. With the rapid development of imaging medicine, depending on imaging genomics, many experts have made remarkable achievements in tumor screening and diagnosis, prognosis evaluation, new treatment targets and understanding of tumor biological mechanism. This review analyzes the relationship between tumor radiology and gene expression, which provides a favorable direction for clinical staging, prognosis evaluation and accurate treatment of tumors.
Collapse
Affiliation(s)
- Zhen Liu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Kefeng Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Binhua Wu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiaoning Tang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China
| | - Huiqing Yuan
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Hao Pang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China
| | - Yongmei Huang
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China
| | - Xiao Zhu
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Hui Luo
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| | - Yi Qi
- Southern Marine Science and Engineering Guangdong Laboratory (Zhanjiang), Zhanjiang, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, China; The Key Lab of Zhanjiang for R&D Marine Microbial Resources in the Beibu Gulf Rim, Guangdong Medical University, Zhanjiang, China; The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, China.
| |
Collapse
|
35
|
Cao X, Xia W, Tang Y, Zhang B, Yang J, Zeng Y, Geng D, Zhang J. Radiomic Model for Distinguishing Dissecting Aneurysms from Complicated Saccular Aneurysms on high-Resolution Magnetic Resonance Imaging. J Stroke Cerebrovasc Dis 2020; 29:105268. [PMID: 32992167 DOI: 10.1016/j.jstrokecerebrovasdis.2020.105268] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2020] [Revised: 08/07/2020] [Accepted: 08/20/2020] [Indexed: 02/08/2023] Open
Abstract
OBJECTIVE To build radiomic model in differentiating dissecting aneurysm (DA) from complicated saccular aneurysm (SA) based on high-resolution magnetic resonance imaging (HR-MRI) through machine-learning algorithm. METHODS Overall, 851 radiomic features from 77 cases were retrospectively analyzed, and the ElasticNet algorithm was used to build the radiomic model. A clinico-radiological model using clinical features and conventional MRI findings was also built. An integrated model was then built by incorporating the radiomic model and clinico-radiological model. The diagnostic abilities of these models were evaluated using leave one out cross validation and quantified using the receiver operating characteristic (ROC) analysis. The diagnostic performance of radiologists was also evaluated for comparison. RESULTS Five features were used to form the radiomic model, which yielded an area under the ROC curve (AUC) of 0.912 (95 % CI 0.846-0.976), sensitivity of 0.852, and specificity of 0.861. The radiomic model achieved a better diagnostic performance than the clinico-radiological model (AUC=0.743, 95 % CI 0.623-0.862), integrated model (AUC=0.888, 95 % CI 0.811-0.965), and even many radiologists. CONCLUSION Radiomic features derived from HR-MRI can reliably be used to build a radiomic model for effectively differentiating between DA and complicated SA, and it can provide an objective basis for the selection of clinical treatment plan.
Collapse
Affiliation(s)
- Xin Cao
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Institute of Functional and Molecular Medical Imaging, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Wei Xia
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Academy for Engineering and Technology, Fudan University, 20 Handan Road, Yangpu District, Shanghai 200433, China; Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, 88 Keling Road, Suzhou New District, Suzhou 215163, Jiangsu, China
| | - Ye Tang
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Bo Zhang
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Jinming Yang
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Yanwei Zeng
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Institute of Functional and Molecular Medical Imaging, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China
| | - Daoying Geng
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Institute of Functional and Molecular Medical Imaging, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| | - Jun Zhang
- Department of Radiology, Huashan Hospital, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China; Institute of Functional and Molecular Medical Imaging, Fudan University, 12 Wulumuqi Middle Road, Shanghai 200040, China.
| |
Collapse
|
36
|
Granata V, Fusco R, Risi C, Ottaiano A, Avallone A, De Stefano A, Grimm R, Grassi R, Brunese L, Izzo F, Petrillo A. Diffusion-Weighted MRI and Diffusion Kurtosis Imaging to Detect RAS Mutation in Colorectal Liver Metastasis. Cancers (Basel) 2020; 12:cancers12092420. [PMID: 32858990 PMCID: PMC7565693 DOI: 10.3390/cancers12092420] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2020] [Revised: 08/24/2020] [Accepted: 08/25/2020] [Indexed: 12/11/2022] Open
Abstract
Simple Summary Imaging derived parameters can provide data on tumor phenotype as well as cancer microenvironment. Radiomics has recently shown potential in realizing personalized medicine. The aim of the manuscript is to detect RAS mutation in colorectal liver metastasis by Diffusion-Weighted Magnetic Resonance Imaging (DWI-MRI) - and Diffusion Kurtosis imaging (DKI)-derived parameters. We demonstrated that DKI derived parameters allows to detect RAS mutation in liver metastasis. Abstract Objectives: To detect RAS mutation in colorectal liver metastasis by Diffusion-Weighted Magnetic Resonance Imaging (DWI-MRI) - and Diffusion Kurtosis imaging (DKI)-derived parameters. Methods: In total, 106 liver metastasis (60 metastases with RAS mutation) in 52 patients were included in this retrospective study. Diffusion and perfusion parameters were derived by DWI (apparent diffusion coefficient (ADC), basal signal (S0), pseudo-diffusion coefficient (DP), perfusion fraction (FP) and tissue diffusivity (DT)) and DKI data (mean of diffusion coefficient (MD) and mean of diffusional Kurtosis (MK)). Wilcoxon–Mann–Whitney U tests for non-parametric variables and receiver operating characteristic (ROC) analyses were calculated with area under ROC curve (AUC). Moreover, pattern recognition approaches (linear classifier, support vector machine, k-nearest neighbours, decision tree), with features selection methods and a leave-one-out cross validation approach, were considered. Results: A significant discrimination between the group with RAS mutation and the group without RAS mutation was obtained by the standard deviation value of MK (MK STD), by the mean value of MD, and by that of FP. The best results were reached by MK STD with an AUC of 0.80 (sensitivity of 72%, specificity of 85%, accuracy of 79%) using a cut-off of 203.90 × 10−3, and by the mean value of MD with AUC of 0.80 (sensitivity of 84%, specificity of 73%, accuracy of 77%) using a cut-off of 1694.30 mm2/s × 10−6. Considering all extracted features or the predictors obtained by the features selection method (the mean value of S0, the standard deviation value of MK, FP and of DT), the tested pattern recognition approaches did not determine an increase in diagnostic accuracy to detect RAS mutation (AUC of 0.73 and 0.69, respectively). Conclusions: Diffusion-Weighted imaging and Diffusion Kurtosis imaging could be used to detect the RAS mutation in liver metastasis. The standard deviation value of MK and the mean value of MD were the more accurate parameters in the RAS mutation detection, with an AUC of 0.80.
Collapse
Affiliation(s)
- Vincenza Granata
- Radiology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (V.G.); (A.P.)
| | - Roberta Fusco
- Radiology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (V.G.); (A.P.)
- Correspondence: ; Tel.: +39-0815-90714; Fax: 39-0815-903825
| | - Chiara Risi
- Radiology Division, Universita’ Degli Studi Di Napoli Federico II, 80131 Naples, Italy;
| | - Alessandro Ottaiano
- Abdominal Oncology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (A.O.); (A.A.); (A.D.S.)
| | - Antonio Avallone
- Abdominal Oncology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (A.O.); (A.A.); (A.D.S.)
| | - Alfonso De Stefano
- Abdominal Oncology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (A.O.); (A.A.); (A.D.S.)
| | - Robert Grimm
- Siemens Healthcare GmbH, 91052 Erlangen, Germany;
| | - Roberta Grassi
- Radiology Division, Universita’ Degli Studi Della Campania Luigi Vanvitelli, Piazza Miraglia, 80138 Naples, Italy;
| | - Luca Brunese
- Rector of the Universita’ Degli Studi Del Molise, 86100 Molise, Italy;
| | - Francesco Izzo
- Hepatobiliary Surgical Oncology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy;
| | - Antonella Petrillo
- Radiology Division, Istituto Nazionale Tumori–IRCCS-Fondazione G. Pascale, Napoli, Italia, Via Mariano Semmola, 80131 Naples, Italy; (V.G.); (A.P.)
| |
Collapse
|
37
|
Chen Y, Jiang J, Shi J, Chang W, Shi J, Chen M, Zhang Q. Dual-mode ultrasound radiomics and intrinsic imaging phenotypes for diagnosis of lymph node lesions. ANNALS OF TRANSLATIONAL MEDICINE 2020; 8:742. [PMID: 32647667 PMCID: PMC7333147 DOI: 10.21037/atm-19-4630] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Background The ultrasonic diagnosis of lymph node lesions is usually based on a small number of subjective visual features from a single ultrasonic modality, which limits diagnostic accuracy. Therefore, our study aimed to propose a computerized method for using dual-mode ultrasound radiomics and the intrinsic imaging phenotypes for accurately differentiating benign, lymphomatous, and metastatic lymph nodes. Methods A total of 543 lymph nodes from 538 patients were examined with both B-mode ultrasonography and elastography. The data set was randomly divided into a training set of 407 nodes and a validation set of 136 nodes. First, we extracted 430 radiomic features from dual-mode images. Then, we combined the least absolute shrinkage and selection operator with the analysis of variance to select several typical features. We retrieved the intrinsic imaging phenotypes by using a hierarchical clustering of all radiomics features, and we integrated the phenotypes with the selected features for the classification of benign, lymphomatous, and metastatic nodes. Results The areas under the receiver operating characteristic curves (AUCs) on the validation set were 0.960 for benign vs. lymphomatous, 0.716 for benign vs. metastatic, 0.933 for lymphomatous vs. metastatic, and 0.856 for benign vs. malignant. Conclusions The radiomics features and intrinsic imaging phenotypes derived from the dual-mode ultrasound can capture the distinctions between benign, lymphomatous, and metastatic nodes and are valuable in node differentiation.
Collapse
Affiliation(s)
- Ying Chen
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Institute of Biomedical Engineering, Shanghai University, Shanghai, China.,School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Jianwei Jiang
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jie Shi
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Institute of Biomedical Engineering, Shanghai University, Shanghai, China.,School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Wanying Chang
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Jun Shi
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,School of Communication and Information Engineering, Shanghai University, Shanghai, China
| | - Man Chen
- Department of Ultrasound Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Qi Zhang
- Shanghai Institute for Advanced Communication and Data Science, Shanghai University, Shanghai, China.,The SMART (Smart Medicine and AI-based Radiology Technology) Lab, Institute of Biomedical Engineering, Shanghai University, Shanghai, China.,School of Communication and Information Engineering, Shanghai University, Shanghai, China.,Hangzhou YITU Healthcare Technology, Hangzhou, China
| |
Collapse
|
38
|
Wang T, Sun H, Guo Y, Zou L. 18F-FDG PET/CT Quantitative Parameters and Texture Analysis Effectively Differentiate Endometrial Precancerous Lesion and Early-Stage Carcinoma. Mol Imaging 2020; 18:1536012119856965. [PMID: 31198089 PMCID: PMC6572902 DOI: 10.1177/1536012119856965] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Objective: This study evaluated the metabolic parameters and texture features of fluorodeoxyglucose positron emission tomography–computed tomography (PET/CT) for the diagnosis and differentiation of endometrial atypical hyperplasia (EAH), EAH with field cancerization (FC), and stage 1A endometrial carcinoma (EC 1a). Materials and Methods: We retrospectively analyzed the metabolic parameters of PET/CT in 170 patients with diagnoses confirmed by pathology, including 57 cases of EAH (57/170, 33.53%), 45 cases of FC (45/170, 26.47%), and 68 cases of EC 1a (68/170, 40.0%). Then, the texture features of each tumor were extracted and compared with the metabolic parameters and pathological results using nonparametric tests and linear regression analysis. The diagnostic performance was assessed by the area under the curve (AUC) values obtained from receiver operating characteristic analysis. Results: There were moderate positive correlations between the PET standardized uptake values (SUVpeak, SUVmax, and SUVmean) and postoperative pathological features with correlation coefficients (rs) of 0.663, 0.651, and 0.651, respectively (P < .001). Total lesion glycolysis showed relatively low correlation with pathological characteristics (rs = 0.476), whereas metabolic tumor volume and age showed the weakest correlations (rs = 0.186 and 0.232, respectively). To differentiate between the diagnosis of EAH and FC, SUVmax displayed the largest AUC of 0.857 (sensitivity, 82.2%; specificity, 84.2%). Five texture features were screened out as Percentile 40, Percentile 45, InverseDifferenceMoment_AllDirection_offset 1, InverseDifferenceMoment_angle 45_offset 4, and ClusterProminence_angle 135_offset 7 (P < .001) by linear model of texture analysis (AUC = 0.851; specificity = 0.692; sensitivity = 0.871). To differentiate between the diagnoses of FC and EC 1a, SUVpeak displayed the largest AUC of 0.715 (sensitivity, 67.6%; specificity, 77.8%), and 2 texture features were identified as Percentile 10 and CP_angle 135_offset 7 (AUC = 0.819; specificity = 0.871; sensitivity = 0.766; P < .001). Conclusions: SUVmax and SUVpeak had the highest diagnostic values for EAH, FC, and EC 1a compared with the other tested parameters. SUVmax, Percentile 40, Percentile 45, InverseDifferenceMoment_AllDirection_offset 1, InverseDifferenceMoment_angle 45_offset 4, and ClusterProminence_angle 135_offset 7 distinguished EAH from FC. SUVpeak, Percentile 10, and ClusterProminence_angle 135_offset 7 distinguished FC from EC 1a. This study showed that the addition of texture features provides valuable information for differentiating EAH, FC, and EC 1a diagnoses.
Collapse
Affiliation(s)
- Tong Wang
- 1 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Hongzan Sun
- 1 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yan Guo
- 2 GE Healthcare, Beijing, China
| | - Lue Zou
- 1 Department of Radiology, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
39
|
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, typically develops on the background of chronic liver disease and is an aggressive disease with dismal prognosis. Studies using next-generation sequencing of multiple regions of the same tumour nodule suggest different patterns of HCC evolution and confirm the high molecular heterogeneity in a subset of patients. Different hypotheses have been proposed to explain tumour evolution, including clonal selection or neutral and punctuated acquisition of genetic alterations. In parallel, data indicate a fundamental contribution of nonmalignant cells of the tumour microenvironment to cancer clonal evolution. Delineating these dynamics is crucial to improve the treatment of patients with HCC, and particularly to help understand how HCC evolution drives resistance to systemic therapies. A number of new minimally invasive techniques, such as liquid biopsies, could help track cancer evolution in HCC. These tools might improve our understanding of how systemic therapies affect tumour clonal composition and could facilitate implementation of real-time molecular monitoring of patients with HCC.
Collapse
|
40
|
Wang XH, Long LH, Cui Y, Jia AY, Zhu XG, Wang HZ, Wang Z, Zhan CM, Wang ZH, Wang WH. MRI-based radiomics model for preoperative prediction of 5-year survival in patients with hepatocellular carcinoma. Br J Cancer 2020; 122:978-985. [PMID: 31937925 PMCID: PMC7109104 DOI: 10.1038/s41416-019-0706-0] [Citation(s) in RCA: 80] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/10/2019] [Accepted: 10/08/2019] [Indexed: 12/17/2022] Open
Abstract
Background Recurrence is the major cause of mortality in patients with resected HCC. However, without a standard approach to evaluate prognosis, it is difficult to select candidates for additional therapy. Methods A total of 201 patients with HCC who were followed up for at least 5 years after curative hepatectomy were enrolled in this retrospective, multicentre study. A total of 3144 radiomics features were extracted from preoperative MRI. The random forest method was used for radiomics signature building, and five-fold cross-validation was applied. A radiomics model incorporating the radiomics signature and clinical risk factors was developed. Results Patients were divided into survivor (n = 97) and non-survivor (n = 104) groups based on the 5-year survival after surgery. The 30 most survival-related radiomics features were selected for the radiomics signature. Preoperative AFP and AST were integrated into the model as independent clinical risk factors. The model demonstrated good calibration and satisfactory discrimination, with a mean AUC of 0.9804 and 0.7578 in the training and validation sets, respectively. Conclusions This radiomics model is a valid method to predict 5-year survival in patients with HCC and may be used to identify patients for clinical trials of perioperative therapies and for additional surveillance.
Collapse
Affiliation(s)
- Xiao-Hang Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Liu-Hua Long
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Yong Cui
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Angela Y Jia
- Department of Radiation Oncology and Molecular Radiation Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Xiang-Gao Zhu
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Hong-Zhi Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Zhi Wang
- Blot Info & Tech (Beijing) Co. Ltd, Beijing, China
| | | | - Zhao-Hai Wang
- Department of Hepatobiliary Surgery, The Fifth Medical Center of Chinese PLA General Hospital, Beijing Institute of Infectious Diseases, Beijing, China.
| | - Wei-Hu Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education/Beijing), Department of Radiation Oncology, Peking University Cancer Hospital and Institute, Beijing, China.
| |
Collapse
|
41
|
Abstract
Radiomics is an emerging field which extracts quantitative radiology data from medical images and explores their correlation with clinical outcomes in a non-invasive manner. This review aims to assess whether radiomics is a useful and reproducible method for clinical management of hepatocellular carcinoma (HCC) by reviewing the strengths and weaknesses of current radiomics literature pertaining specifically to HCC. From an initial set of 48 articles recovered through database searches, 23 articles were retained to be included in this review after full screening. Among these 23 studies, 7 used a radiomics approach in magnetic resonance imaging (MRI). Only two studies applied radiomics to positron emission tomography-computed tomography (PET-CT). In the remaining 14 articles, a radiomics analysis was performed on computed tomography (CT). Eight studies dealt with the relationship between biological signatures and imaging findings, and can be classified as radiogenomic studies. For each study included in our review, we computed a Radiomics Quality Score (RQS) as proposed by Lambin et al. We found that the RQS (mean ± standard deviation) was 8.35 ± 5.38 (out of a possible maximum value of 36). Although these scores are fairly low, and radiomics has not yet reached clinical utility in HCC, it is important to underscore the fact that these early studies pave the way for the radiomics field with a focus on HCC. Radiomics is still a very young field, and is far from being mature, but it remains a very promising technology for the future for developing adequate personalized treatment as a non-invasive approach, for complementing or replacing tumor biopsies, as well as for developing novel prognostic biomarkers in HCC patients.
Collapse
|
42
|
Xie C, Yang P, Zhang X, Xu L, Wang X, Li X, Zhang L, Xie R, Yang L, Jing Z, Zhang H, Ding L, Kuang Y, Niu T, Wu S. Sub-region based radiomics analysis for survival prediction in oesophageal tumours treated by definitive concurrent chemoradiotherapy. EBioMedicine 2019; 44:289-297. [PMID: 31129097 PMCID: PMC6606893 DOI: 10.1016/j.ebiom.2019.05.023] [Citation(s) in RCA: 75] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2019] [Revised: 04/28/2019] [Accepted: 05/09/2019] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Evaluating clinical outcome prior to concurrent chemoradiotherapy remains challenging for oesophageal squamous cell carcinoma (OSCC) as traditional prognostic markers are assessed at the completion of treatment. Herein, we investigated the potential of using sub-region radiomics as a novel tumour biomarker in predicting overall survival of OSCC patients treated by concurrent chemoradiotherapy. METHODS Independent patient cohorts from two hospitals were included for training (n = 87) and validation (n = 46). Radiomics features were extracted from sub-regions clustered from patients' tumour regions using K-means method. The LASSO regression for 'Cox' method was used for feature selection. The survival prediction model was constructed based on the sub-region radiomics features using the Cox proportional hazards model. The clinical and biological significance of radiomics features were assessed by correlation analysis of clinical characteristics and copy number alterations(CNAs) in the validation dataset. FINDINGS The overall survival prediction model combining with seven sub-regional radiomics features was constructed. The C-indexes of the proposed model were 0.729 (0.656-0.801, 95% CI) and 0.705 (0.628-0.782, 95%CI) in the training and validation cohorts, respectively. The 3-year survival receiver operating characteristic (ROC) curve showed an area under the ROC curve of 0.811 (0.670-0.952, 95%CI) in training and 0.805 (0.638-0.973, 95%CI) in validation. The correlation analysis showed a significant correlation between radiomics features and CNAs. INTERPRETATION The proposed sub-regional radiomics model could predict the overall survival risk for patients with OSCC treated by definitive concurrent chemoradiotherapy. FUND: This work was supported by the Zhejiang Provincial Foundation for Natural Sciences, National Natural Science Foundation of China.
Collapse
Affiliation(s)
- Congying Xie
- Cancer Centre, First Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Pengfei Yang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, PR China
| | - Xuebang Zhang
- Cancer Centre, First Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, PR China
| | - Lei Xu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Xiaoju Wang
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Xiadong Li
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China; Department of Radiation Therapy, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Luhan Zhang
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China
| | - Ruifei Xie
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Ling Yang
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Zhao Jing
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Hongfang Zhang
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Lingyu Ding
- Cancer Research Institute, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, PR China
| | - Yu Kuang
- Department of Medical Physics, University of Nevada, Las Vegas, Las Vegas, NV, USA
| | - Tianye Niu
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang, PR China.
| | - Shixiu Wu
- National Cancer Center/National Clinical Research Center For Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, PR China.
| |
Collapse
|
43
|
Feasibility of multiparametric imaging with PET/MR in nasopharyngeal carcinoma: A pilot study. Oral Oncol 2019; 93:91-95. [PMID: 31109702 DOI: 10.1016/j.oraloncology.2019.04.021] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2019] [Revised: 04/06/2019] [Accepted: 04/27/2019] [Indexed: 01/11/2023]
Abstract
OBJECTIVE The aim of this pilot study was to explore the integrated positron emission tomography and magnetic resonance imaging scanner (PET/MR) for biological characterization of nasopharyngeal carcinoma (NPC) and potential therapeutic applications of dose painting (DP). PATIENTS AND METHODS Twenty-one NPC patients with PET/MR were included in this study. Overlap of tumor volumes was analyzed on T2-weighted images (volume of interest, VOIT2), diffusion-weighted magnetic resonance imaging (VOIDWI) and 18F-fluorodeoxyglucose positron emission tomography (VOIPET). The overlap percentages of low-metabolic sub-region (cluster 1) and high-metabolic sub-region (cluster 2) in VOIPET and VOIDWI were analyzed by cluster analysis. RESULTS Both the VOIDWI and VOIPET were encompassed in the VOIT2, respectively 99.6% and 97.5%. The median tumor overlap was 94.4% (VOIDWI within VOIPET). The median overlap of cluster 2 in VOIPET and VOIDWI was 43.61% (27.67-52.66%) and 21.86%(10.47-40.89%), respectively. The median overlap of cluster 1 in VOIPET and VOIDWI was 48.03% (23.91-63.15%) and 24.40% (7.44-51.44%), respectively. Separation between clusters appeared to be defined by a SUV value. CONCLUSION For NPC, the VOIs of DWI and FDG PET were not overlapped completely and the volume defined by cluster-analysis might be meaningful for DP.
Collapse
|
44
|
Radiogenomics and Radiomics in Liver Cancers. Diagnostics (Basel) 2018; 9:diagnostics9010004. [PMID: 30591628 PMCID: PMC6468592 DOI: 10.3390/diagnostics9010004] [Citation(s) in RCA: 49] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 12/20/2018] [Accepted: 12/23/2018] [Indexed: 12/24/2022] Open
Abstract
Radiogenomics is a computational discipline that identifies correlations between cross-sectional imaging features and tissue-based molecular data. These imaging phenotypic correlations can then potentially be used to longitudinally and non-invasively predict a tumor’s molecular profile. A different, but related field termed radiomics examines the extraction of quantitative data from imaging data and the subsequent combination of these data with clinical information in an attempt to provide prognostic information and guide clinical decision making. Together, these fields represent the evolution of biomedical imaging from a descriptive, qualitative specialty to a predictive, quantitative discipline. It is anticipated that radiomics and radiogenomics will not only identify pathologic processes, but also unveil their underlying pathophysiological mechanisms through clinical imaging alone. Here, we review recent studies on radiogenomics and radiomics in liver cancers, including hepatocellular carcinoma, intrahepatic cholangiocarcinoma, and metastases to the liver.
Collapse
|
45
|
Jeong WK, Jamshidi N, Felker ER, Raman SS, Lu DS. Radiomics and radiogenomics of primary liver cancers. Clin Mol Hepatol 2018; 25:21-29. [PMID: 30441889 PMCID: PMC6435966 DOI: 10.3350/cmh.2018.1007] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2018] [Accepted: 08/16/2018] [Indexed: 02/07/2023] Open
Abstract
Concurrent advancements in imaging and genomic biomarkers have created opportunities to identify non-invasive imaging surrogates of molecular phenotypes. In order to develop such imaging surrogates radiomics and radiogenomics/imaging genomics will be necessary; there has been consistent progress in these fields for primary liver cancers. In this article we evaluate the current status of the field specifically with regards to hepatocellular carcinoma and intrahepatic cholangiocarcinoma, highlighting some of the up and coming results that were presented at the annual Radiological Society of North America Conference in 2017. There are an increasing number of studies in this area with a bias towards quantitative feature measurement, which is expected to benefit reproducibility of the findings and portends well for the future development of biomarkers for diagnosis, prognosis, and treatment response assessment. We review some of the advancements and look forward to some of the exciting future applications that are anticipated as the field develops.
Collapse
Affiliation(s)
- Woo Kyoung Jeong
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.,Department of Radiology, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea.,Department of Center for Imaging Sciences, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | - Neema Jamshidi
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Ely Richard Felker
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Steven Satish Raman
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - David Shinkuo Lu
- Department of Radiological Science, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
46
|
Preoperative radiomic signature based on multiparametric magnetic resonance imaging for noninvasive evaluation of biological characteristics in rectal cancer. Eur Radiol 2018; 29:3200-3209. [PMID: 30413959 DOI: 10.1007/s00330-018-5763-x] [Citation(s) in RCA: 106] [Impact Index Per Article: 15.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 08/21/2018] [Accepted: 09/14/2018] [Indexed: 12/14/2022]
Abstract
OBJECTIVES To develop and validate radiomic models in evaluating biological characteristics of rectal cancer based on multiparametric magnetic resonance imaging (MP-MRI). METHODS This study consisted of 345 patients with rectal cancer who underwent MP-MRI. We focused on evaluating five postoperative confirmed characteristics: lymph node (LN) metastasis, tumor differentiation, fraction of Ki-67-positive tumor cells, human epidermal growth factor receptor 2 (HER-2), and KRAS-2 gene mutation status. Data from 197 patients were used to develop the biological characteristics evaluation models. Radiomic features were extracted from MP-MRI and then refined for reproducibility and redundancy. The refined features were investigated for usefulness in building radiomic signatures by using two feature-ranking methods (MRMR and WLCX) and three classifiers (RF, SVM, and LASSO). Multivariable logistic regression was used to build an integrated evaluation model combining radiomic signatures and clinical characteristics. The performance was evaluated using an independent validation dataset comprising 148 patients. RESULTS The MRMR and LASSO regression produced the best-performing radiomic signatures for evaluating HER-2, LN metastasis, tumor differentiation, and KRAS-2 gene status, with AUC values of 0.696 (95% CI, 0.610-0.782), 0.677 (95% CI, 0.591-0.763), 0.720 (95% CI, 0.621-0.819), and 0.651 (95% CI, 0.539-0.763), respectively. The best-performing signatures for evaluating Ki-67 produced an AUC value of 0.699 (95% CI, 0.611-0.786), and it was developed by WLCX and RF algorithm. The integrated evaluation model incorporating radiomic signature and MRI-reported LN status had improved AUC of 0.697 (95% CI, 0.612-0.781). CONCLUSION Radiomic signatures based on MP-MRI have potential to noninvasively evaluate the biological characteristics of rectal cancer. KEY POINTS • Radiomic features were extracted from MP-MRI images of the rectal tumor. • The proposed radiomic signatures demonstrated discrimination ability in identifying the histopathological, immunohistochemical, and genetic characteristics of rectal cancer. • All MRI sequences were important and could provide complementary information in radiomic analysis.
Collapse
|
47
|
Liang W, Yang P, Huang R, Xu L, Wang J, Liu W, Zhang L, Wan D, Huang Q, Lu Y, Kuang Y, Niu T. A Combined Nomogram Model to Preoperatively Predict Histologic Grade in Pancreatic Neuroendocrine Tumors. Clin Cancer Res 2018; 25:584-594. [PMID: 30397175 DOI: 10.1158/1078-0432.ccr-18-1305] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2018] [Revised: 09/28/2018] [Accepted: 10/31/2018] [Indexed: 01/09/2023]
Abstract
PURPOSE The purpose of this study is to develop and validate a nomogram model combing radiomics features and clinical characteristics to preoperatively differentiate grade 1 and grade 2/3 tumors in patients with pancreatic neuroendocrine tumors (pNET).Experimental Design: A total of 137 patients who underwent contrast-enhanced CT from two hospitals were included in this study. The patients from the second hospital (n = 51) were selected as an independent validation set. The arterial phase in contrast-enhanced CT was selected for radiomics feature extraction. The Mann-Whitney U test and least absolute shrinkage and selection operator regression were applied for feature selection and radiomics signature construction. A combined nomogram model was developed by incorporating the radiomics signature with clinical factors. The association between the nomogram model and the Ki-67 index and rate of nuclear mitosis were also investigated respectively. The utility of the proposed model was evaluated using the ROC, area under ROC curve (AUC), calibration curve, and decision curve analysis (DCA). The Kaplan-Meier (KM) analysis was used for survival analysis. RESULTS An eight-feature-combined radiomics signature was constructed as a tumor grade predictor. The nomogram model combining the radiomics signature with clinical stage showed the best performance (training set: AUC = 0.907; validation set: AUC = 0.891). The calibration curve and DCA demonstrated the clinical usefulness of the proposed nomogram. A significant correlation was observed between the developed nomogram and Ki-67 index and rate of nuclear mitosis, respectively. The KM analysis showed a significant difference between the survival of predicted grade 1 and grade 2/3 groups (P = 0.002). CONCLUSIONS The combined nomogram model developed could be useful in differentiating grade 1 and grade 2/3 tumor in patients with pNETs.
Collapse
Affiliation(s)
- Wenjie Liang
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Key Laboratory of Precision Diagnosis and Treatment for Hepatobiliary and Pancreatic Tumor of Zhejiang Province, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Pengfei Yang
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China.,College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Rui Huang
- College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, Zhejiang, China
| | - Lei Xu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Jiawei Wang
- Department of Radiology, the Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Weihai Liu
- Department of Radiology, Beilun Branch Hospital of the First Affiliated Hospital, Zhejiang University School of Medicine, the People's Hospital of Beilun District, Ningbo, Zhejiang, China
| | - Lele Zhang
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Key Lab of Combined Multi-Organ Transplantation, Ministry of Public Health, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.,Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Dalong Wan
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Qiang Huang
- Department of Radiology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yao Lu
- Department of Medical Physics, University of Nevada, Las Vegas, Las Vegas, Nevada
| | - Yu Kuang
- Department of Medical Physics, University of Nevada, Las Vegas, Las Vegas, Nevada.
| | - Tianye Niu
- Department of Radiation Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China. .,Institute of Translational Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| |
Collapse
|