1
|
Horne A, Abravan A, Fornacon-Wood I, O'Connor JPB, Price G, McWilliam A, Faivre-Finn C. Mastering CT-based radiomic research in lung cancer: a practical guide from study design to critical appraisal. Br J Radiol 2025; 98:653-668. [PMID: 40100283 DOI: 10.1093/bjr/tqaf051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/18/2024] [Accepted: 02/25/2025] [Indexed: 03/20/2025] Open
Abstract
Radiomics is a health technology that has the potential to extract clinically meaningful biomarkers from standard of care imaging. Despite a wealth of exploratory analysis performed on scans acquired from patients with lung cancer and existing guidelines describing some of the key steps, no radiomic-based biomarker has been widely accepted. This is primarily due to limitations with methodology, data analysis, and interpretation of the available studies. There is currently a lack of guidance relating to the entire radiomic workflow from study design to critical appraisal. This guide, written with early career lung cancer researchers, describes a more complete radiomic workflow. Lung cancer image analysis is the focus due to some of the unique challenges encountered such as patient movement from breathing. The guide will focus on CT imaging as these are the most common scans performed on patients with lung cancer. The aim of this article is to support the production of high-quality research that has the potential to positively impact outcome of patients with lung cancer.
Collapse
Affiliation(s)
- Ashley Horne
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| | - Azadeh Abravan
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Isabella Fornacon-Wood
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - James P B O'Connor
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Division of Radiotherapy and Imaging, Institute of Cancer Research, London, SW7 3RP, United Kingdom
| | - Gareth Price
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Alan McWilliam
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
| | - Corinne Faivre-Finn
- Division of Cancer Sciences, The University of Manchester, Manchester, M13 9NT, United Kingdom
- Department of Thoracic Oncology, The Christie NHS Foundation Trust, Manchester, M20 4BX, United Kingdom
| |
Collapse
|
2
|
Chen Z, Zhu Y, Wang L, Cong R, Feng B, Cai W, Liang M, Li D, Wang S, Hu M, Mi Y, Wang S, Ma X, Zhao X. Virtual MR Elastography and Multi-b-value DWI Models for Predicting Microvascular Invasion in Solitary BCLC Stage A Hepatocellular Carcinoma. Acad Radiol 2025; 32:2569-2584. [PMID: 39643466 DOI: 10.1016/j.acra.2024.11.027] [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: 06/12/2024] [Revised: 11/07/2024] [Accepted: 11/11/2024] [Indexed: 12/09/2024]
Abstract
RATIONALE AND OBJECTIVES To evaluate the performance of virtual MR elastography (vMRE) for predicting microvascular invasion (MVI) in Barcelona Clinic Liver Cancer (BCLC) stage A (≤ 5.0 cm) hepatocellular carcinoma (HCC) and to construct a combined nomogram based on vMRE, multi-b-value DWI models, and clinical-radiological (CR) features. METHODS Consecutive patients with suspected HCC who underwent multi-b-value DWI examinations were prospectively collected. Quantitative parameters from vMRE, mono-exponential, intravoxel incoherent motion, and diffusion kurtosis imaging models were obtained. Multivariate logistic regression was used to identify independent MVI predictors and build prediction models. A combined MRI_Score was constructed using independent quantitative parameters. A visualized nomogram was built based on significant CR features and MRI_Score. The predictive performance of quantitative parameters and models was evaluated. RESULTS The study included 103 patients (median age: 56 years; range: 35-70 years; 87 males and 16 females). Diffusion-based shear modulus (μDiff) exhibited a predictive performance for MVI with area under the curve (AUC) of 0.735. The MRI_Score was developed employing true diffusion coefficient (D), mean kurtosis (MK), and μDiff. CR model and MRI_Score achieved AUCs of 0.787 and 0.840, respectively. The combined nomogram based on AFP, corona enhancement, tumor capsule, TTPVI, and MRI_Score significantly improved the predictive performance to an AUC of 0.931 (Delong test p < 0.05). CONCLUSION vMRE exhibited great potential for predicting MVI in BCLC stage A HCC. The combined nomogram integrating CR features, vMRE, and quantitative diffusion parameters significantly improved the predictive accuracy and could potentially assist clinicians in identifying appropriate treatment options.
Collapse
Affiliation(s)
- Zhaowei Chen
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Leyao Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Rong Cong
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Bing Feng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Meng Liang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Dengfeng Li
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Shuang Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Mancang Hu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Yongtao Mi
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Sicong Wang
- Magnetic Resonance Imaging Research, General Electric Healthcare (China), Beijing 100176, China (S.W.).
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100021, China (Z.C., Y.Z., L.W., R.C., B.F., W.C., M.L., D.L., S.W., M.H., Y.M., X.M., X.Z.).
| |
Collapse
|
3
|
Tixier F, Lopez-Ramirez F, Blanco A, Javed AA, Chu LC, Hruban RH, Yasrab M, Fouladi DF, Shayesteh S, Ghandili S, Fishman EK, Kawamoto S. Diagnostic Performance of Combined Conventional CT Imaging Features and Radiomics Signature in Differentiating Grade 1 Tumors from Higher-Grade Pancreatic Neuroendocrine Neoplasms. Cancers (Basel) 2025; 17:1047. [PMID: 40149380 PMCID: PMC11941307 DOI: 10.3390/cancers17061047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/16/2025] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND/OBJECTIVES Accurate identification of grade 1 (G1) pancreatic neuroendocrine tumors (PanNETs) is crucial due to their rising incidence and emerging nonsurgical management strategies. This study evaluated whether combining conventional CT imaging features, CT radiomics features, and clinical data improves differentiation of G1 PanNETs from higher-grade tumors (G2/G3 PanNETs and pancreatic neuroendocrine carcinomas [PanNECs]) compared to using these features individually. METHODS A retrospective analysis included 133 patients with pathologically confirmed PanNETs or PanNECs (70 males, 63 females; mean age, 58.5 years) who underwent pancreas protocol CT. A total of 28 conventional imaging features, 4892 radiomics features, and clinical data (age, gender, and tumor location) were analyzed using a support vector machine (SVM) model. Data were divided into 70% training and 30% testing sets. RESULTS The SVM model using the top 10 conventional imaging features (e.g., suspicious lymph nodes and hypoattenuating tumors) achieved 75% sensitivity, 81% specificity, and 79% accuracy for identifying higher-grade tumors (G2/G3 PanNETs and PanNECs). The top 10 radiomics features yielded 94% sensitivity, 46% specificity, and 69% accuracy. Combining all features (imaging, radiomics, and clinical data) improved performance, with 94% sensitivity, 69% specificity, 79% accuracy, and an F1-score of 0.77. The radiomics score demonstrated an AUC of 0.85 in the training and 0.83 in the testing set. CONCLUSIONS Conventional imaging features provided higher specificity, while radiomics offered greater sensitivity for identifying higher-grade tumors. Integrating all three features improved diagnostic accuracy, highlighting their complementary roles. This combined model may serve as a valuable tool for distinguishing higher-grade tumors from G1 PanNETs and potentially guiding patient management.
Collapse
Affiliation(s)
- Florent Tixier
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Ammar A. Javed
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA;
| | - Linda C. Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Ralph H. Hruban
- The Sol Goldman Pancreatic Cancer Research Center, Department of Pathology, School of Medicine, Hopkins University, Baltimore, MD 21205, USA;
- Sidney Kimmel Comprehensive Cancer Center, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Daniel Fadaei Fouladi
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Shahab Shayesteh
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Saeed Ghandili
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Elliot K. Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, School of Medicine, Johns Hopkins University, Baltimore, MD 21205, USA; (F.T.); (F.L.-R.); (A.B.); (L.C.C.); (M.Y.); (D.F.F.); (S.S.); (S.G.); (E.K.F.)
| |
Collapse
|
4
|
Qiu C, Ma Y, Xiao M, Wang Z, Wu S, Han K, Wang H. Nomogram to Predict Tumor Remnant of Small Hepatocellular Carcinoma after Microwave Ablation. Acad Radiol 2025; 32:1419-1430. [PMID: 39448339 DOI: 10.1016/j.acra.2024.09.066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 09/11/2024] [Accepted: 09/29/2024] [Indexed: 10/26/2024]
Abstract
RATIONALE AND OBJECTIVES This investigation sought to create a nomogram to predict the ablation effect after microwave ablation in patients with hepatocellular carcinoma, which can guide the selection of microwave ablation for small hepatocellular carcinomas. METHODS In this two-center retrospective study, 233 patients with hepatocellular carcinoma treated with microwave ablation (MWA) between January 2016 and December 2023 were enrolled and analyzed for their clinical baseline data, laboratory parameters, and MR imaging characteristics. Logistic regression analysis was used to screen the features, and clinical and imaging feature models were developed separately. Finally, a nomogram was established. All models were evaluated using the area under the curve (AUC), accuracy, sensitivity, specificity, and decision curve analysis (DCA). RESULTS Two models and a nomogram were developed to predict ablation outcomes after MWA based on a training set (n = 182, including complete ablation: 136, incomplete ablation: 46) and an external validation set (n = 51, complete ablation: 36, incomplete ablation: 15). The clinical models and nomogram performed well in the external validation cohort. The AUC of the nomogram was 0.966 (95% CI: 0.944- 0.989), with a sensitivity of 0.935, a specificity of 0.882, and an accuracy of 0.896. CONCLUSIONS Combining clinical data and imaging features, a nomogram was constructed that could effectively predict the postoperative ablation outcome in hepatocellular carcinoma patients undergoing MWA, which could help clinicians provide treatment options for hepatocellular carcinoma patients.
Collapse
Affiliation(s)
- Chenyang Qiu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Yinchao Ma
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Mengjun Xiao
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Zhipeng Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Shuzhen Wu
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Kun Han
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| | - Haiyan Wang
- Department of Radiology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, China (C.Q., Y.M., M.X., Z.W., S.W., K.H., H.W.).
| |
Collapse
|
5
|
Tanaka T, Motegi T, Sumikawa N, Mori M, Kurokawa S, Akiyoshi H. Early Enhancement in Contrast-Enhanced Computed Tomography Is an Index of DUSP9, SLPI, ALDH1L2, and SLC1A1 Expression in Canine Hepatocellular Carcinoma: A Preliminary Study. Vet Sci 2025; 12:137. [PMID: 40005897 PMCID: PMC11860268 DOI: 10.3390/vetsci12020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
Canine hepatocellular carcinoma (HCC) is characterized by distinct computed tomography (CT) findings. HCC exhibits tumor heterogeneity, with different genomic information and histopathological features within the same tumor. In human HCC, genetic alterations affect the prognosis and treatment, and research has begun to assess genetic alterations using minimally invasive and reproducible CT. However, the relationship between CT findings and the genomic information of canine HCC is unknown. Early contrast of HCC indicates increased intratumoral neovascular growth. In this study, we aimed to investigate the relationship between enhancement patterns in the arterial phase of CT imaging and gene expression in canine HCC using RNA sequencing. Based on the CT findings, three of the eight dogs studied were classified as having enhancement HCC and five as having non-enhancement HCC. RNA sequencing was performed using the mRNA extracted from the specimens. Eight differentially expressed genes met the cutoff criteria. Among these, DUSP9, SLPI, and ALDH1L2 were the most upregulated genes in enhancement HCC, whereas SLC1A1 was the most downregulated in non-enhancement HCC. Canine HCC may involve different angiogenesis mechanisms. CT findings can be used to assess the gene expression status in canine HCC and may add new value to CT imaging.
Collapse
Affiliation(s)
- Toshiyuki Tanaka
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| | - Tomoki Motegi
- Section of Computational Biomedicine, Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA;
| | - Nanami Sumikawa
- Laboratory of Veterinary Surgery, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (N.S.); (M.M.)
| | - Misaki Mori
- Laboratory of Veterinary Surgery, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (N.S.); (M.M.)
| | - Shohei Kurokawa
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| | - Hideo Akiyoshi
- Laboratory of Veterinary Advanced Diagnosis and Treatment, School of Veterinary Science, Osaka Metropolitan University, Osaka 5988531, Japan; (T.T.); (S.K.)
| |
Collapse
|
6
|
Yang G, Chen Y, Wang M, Wang H, Chen Y. Impact of microvascular invasion risk on tumor progression of hepatocellular carcinoma after conventional transarterial chemoembolization. Oncologist 2025; 30:oyae286. [PMID: 39475355 PMCID: PMC11884753 DOI: 10.1093/oncolo/oyae286] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Accepted: 09/11/2024] [Indexed: 03/08/2025] Open
Abstract
OBJECTIVE To assess tumor progression in patients with hepatocellular carcinoma (HCC) without macrovascular invasion who underwent treatment with conventional transarterial chemoembolization (cTACE) based on microvascular invasion (MVI) risk within 2 years. METHODS This retrospective investigation comprised adult patients with HCC who had either liver resection or cTACE as their first treatment from January 2016 to December 2021. A predictive model for MVI was developed and validated using preoperative clinical and MRI data from patients with HCC treated with liver resection. The MVI predictive model was applied to patients with HCC receiving cTACE, and differences in tumor progression between the MVI high- and low-risk groups were examined throughout 2 years. RESULTS The MVI prediction model incorporated nonsmooth margin, intratumoral artery, incomplete or absent tumor capsule, and tumor DWI/T2WI mismatch. The area under the receiver operating characteristic curve (AUC) for the prediction model, in the training cohort, was determined to be 0.904 (95% CI, 0.862-0.946), while in the validation cohort, it was 0.888 (0.782-0.994). Among patients with HCC undergoing cTACE, those classified as high risk for MVI possessed a lower rate of achieving a complete response after the first tumor therapy and a higher risk of tumor progression within 2 years. CONCLUSIONS The MVI prediction model developed in this study demonstrates a considerable degree of accuracy. Patients at high risk for MVI who underwent cTACE treatment exhibited a higher risk of tumor progression within 2 years.
Collapse
Affiliation(s)
- Guanhua Yang
- The First School of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Yuxin Chen
- Department of Paediatrics, Division of Respiratory Medicine and Allergology, Sophia Children’s Hospital, Erasmus MC, Rotterdam, The Netherlands
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Minglei Wang
- The First School of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Hongfang Wang
- The First School of Clinical Medicine, General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| | - Yong Chen
- Department of Interventional Radiology, General Hospital of Ningxia Medical University, Yinchuan, People’s Republic of China
| |
Collapse
|
7
|
Ghosh S, Zhao X, Alim M, Brudno M, Bhat M. Artificial intelligence applied to 'omics data in liver disease: towards a personalised approach for diagnosis, prognosis and treatment. Gut 2025; 74:295-311. [PMID: 39174307 PMCID: PMC11874365 DOI: 10.1136/gutjnl-2023-331740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 07/24/2024] [Indexed: 08/24/2024]
Abstract
Advancements in omics technologies and artificial intelligence (AI) methodologies are fuelling our progress towards personalised diagnosis, prognosis and treatment strategies in hepatology. This review provides a comprehensive overview of the current landscape of AI methods used for analysis of omics data in liver diseases. We present an overview of the prevalence of different omics levels across various liver diseases, as well as categorise the AI methodology used across the studies. Specifically, we highlight the predominance of transcriptomic and genomic profiling and the relatively sparse exploration of other levels such as the proteome and methylome, which represent untapped potential for novel insights. Publicly available database initiatives such as The Cancer Genome Atlas and The International Cancer Genome Consortium have paved the way for advancements in the diagnosis and treatment of hepatocellular carcinoma. However, the same availability of large omics datasets remains limited for other liver diseases. Furthermore, the application of sophisticated AI methods to handle the complexities of multiomics datasets requires substantial data to train and validate the models and faces challenges in achieving bias-free results with clinical utility. Strategies to address the paucity of data and capitalise on opportunities are discussed. Given the substantial global burden of chronic liver diseases, it is imperative that multicentre collaborations be established to generate large-scale omics data for early disease recognition and intervention. Exploring advanced AI methods is also necessary to maximise the potential of these datasets and improve early detection and personalised treatment strategies.
Collapse
Affiliation(s)
- Soumita Ghosh
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Xun Zhao
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
| | - Mouaid Alim
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
| | - Michael Brudno
- Department of Computer Science, University of Toronto, Toronto, Ontario, Canada
- Princess Margaret Cancer Centre, University Health Network, Toronto, Ontario, Canada
- Vector Institute of Artificial Intelligence, Toronto, Ontario, Canada
| | - Mamatha Bhat
- Transplant AI Initiative, Ajmera Transplant Program, University Health Network, Toronto, Ontario, Canada
- Department of Medicine, University of Toronto, Toronto, Ontario, Canada
- Division of Gastroenterology, University of Toronto Faculty of Medicine, Toronto, Ontario, Canada
- Toronto General Hospital Research Institute, University Health Network, Toronto, Ontario, Canada
| |
Collapse
|
8
|
Tixier F, Lopez-Ramirez F, Blanco A, Yasrab M, Javed AA, Chu LC, Fishman EK, Kawamoto S. Can CT Image Reconstruction Parameters Impact the Predictive Value of Radiomics Features in Grading Pancreatic Neuroendocrine Neoplasms? Bioengineering (Basel) 2025; 12:80. [PMID: 39851354 PMCID: PMC11763079 DOI: 10.3390/bioengineering12010080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2024] [Revised: 01/08/2025] [Accepted: 01/13/2025] [Indexed: 01/26/2025] Open
Abstract
The WHO grading of pancreatic neuroendocrine neoplasms (PanNENs) is essential in patient management and an independent prognostic factor for patient survival. Radiomics features from CE-CT images hold promise for the outcome and tumor grade prediction. However, variations in reconstruction parameters can impact the predictive value of radiomics. 127 patients with histopathologically confirmed PanNENs underwent CT scans with filtered back projection (B20f) and iterative (I26f) reconstruction kernels. 3190 radiomic features were extracted from tumors and pancreatic volumes. Wilcoxon paired tests assessed the impact of reconstruction kernels and ComBat harmonization efficiency. SVM models were employed to predict tumor grade using the entire set of radiomics features or only those identified as harmonizable. The models' performance was assessed on an independent dataset of 36 patients. Significant differences, after correction for multiple testing, were observed in 69% of features in the pancreatic volume and 51% in the tumor volume with B20f and I26f kernels. SVM models demonstrated accuracy ranging from 0.67 (95%CI: 0.50-0.81) to 0.83 (95%CI: 0.69-0.94) in distinguishing grade 1 cases from higher grades. Reconstruction kernels alter radiomics features and iterative kernel models trended towards higher performance. ComBat harmonization mitigates kernel impacts but addressing this effect is crucial in studies involving data from different kernels.
Collapse
Affiliation(s)
- Florent Tixier
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Felipe Lopez-Ramirez
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Alejandra Blanco
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Mohammad Yasrab
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Ammar A. Javed
- Department of Surgery, The NYU Grossman School of Medicine and NYU Langone Health, New York, NY 10016, USA;
| | - Linda C. Chu
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Elliot K. Fishman
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| | - Satomi Kawamoto
- The Russell H. Morgan Department of Radiology and Radiological Science, Johns Hopkins Medical Institutions, Baltimore, MD 21287, USA; (F.L.-R.); (A.B.); (M.Y.); (L.C.C.); (E.K.F.); (S.K.)
| |
Collapse
|
9
|
Dong M, Li C, Zhang L, Zhou J, Xiao Y, Zhang T, Jin X, Fang Z, Zhang L, Han Y, Guan J, Weng Z, Cheng N, Wang J. Intertumoral Heterogeneity Based on MRI Radiomic Features Estimates Recurrence in Hepatocellular Carcinoma. J Magn Reson Imaging 2025; 61:168-181. [PMID: 38712652 DOI: 10.1002/jmri.29428] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2024] [Revised: 04/16/2024] [Accepted: 04/16/2024] [Indexed: 05/08/2024] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) heterogeneity impacts prognosis, and imaging is a potential indicator. PURPOSE To characterize HCC image subtypes in MRI and correlate subtypes with recurrence. STUDY TYPE Retrospective. POPULATION A total of 440 patients (training cohort = 213, internal test cohort = 140, external test cohort = 87) from three centers. FIELD STRENGTH/SEQUENCE 1.5-T/3.0-T, fast/turbo spin-echo T2-weighted, spin-echo echo-planar diffusion-weighted, contrast-enhanced three-dimensional gradient-recalled-echo T1-weighted with extracellular agents (Gd-DTPA, Gd-DTPA-BMA, and Gd-BOPTA). ASSESSMENT Three-dimensional volume-of-interest of HCC was contoured on portal venous phase, then coregistered with precontrast and late arterial phases. Subtypes were identified using non-negative matrix factorization by analyzing radiomics features from volume-of-interests, and correlated with recurrence. Clinical (demographic and laboratory data), pathological, and radiologic features were compared across subtypes. Among clinical, radiologic features and subtypes, variables with variance inflation factor above 10 were excluded. Variables (P < 0.10) in univariate Cox regression were included in stepwise multivariate analysis. Three recurrence estimation models were built: clinical-radiologic model, subtype model, hybrid model integrating clinical-radiologic characteristics, and subtypes. STATISTICAL TESTS Mann-Whitney U test, Kruskal-Wallis H test, chi-square test, Fisher's exact test, Kaplan-Meier curves, log-rank test, concordance index (C-index). Significance level: P < 0.05. RESULTS Two subtypes were identified across three cohorts (subtype 1:subtype 2 of 86:127, 60:80, and 36:51, respectively). Subtype 1 showed higher microvascular invasion (MVI)-positive rates (53%-57% vs. 26%-31%), and worse recurrence-free survival. Hazard ratio (HR) for the subtype is 6.10 in subtype model. Clinical-radiologic model included alpha-fetoprotein (HR: 3.01), macrovascular invasion (HR: 2.32), nonsmooth tumor margin (HR: 1.81), rim enhancement (HR: 3.13), and intratumoral artery (HR: 2.21). Hybrid model included alpha-fetoprotein (HR: 2.70), nonsmooth tumor margin (HR: 1.51), rim enhancement (HR: 3.25), and subtypes (HR: 5.34). Subtype model was comparable to clinical-radiologic model (C-index: 0.71-0.73 vs. 0.71-0.73), but hybrid model outperformed both (C-index: 0.77-0.79). CONCLUSION MRI radiomics-based clustering identified two HCC subtypes with distinct MVI status and recurrence-free survival. Hybrid model showed superior capability to estimate recurrence. LEVEL OF EVIDENCE 3 TECHNICAL EFFICACY STAGE: 2.
Collapse
Affiliation(s)
- Mengshi Dong
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Chao Li
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Lina Zhang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jinhui Zhou
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yuanqiang Xiao
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Tianhui Zhang
- Department of Radiology, Meizhou People's Hospital, Meizhou, China
| | - Xin Jin
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zebin Fang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Linqi Zhang
- Department of Radiology, Affiliated Cancer Hospital and Institute of Guangzhou Medical University, Guangzhou, China
| | - Yu Han
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jiexia Guan
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zijin Weng
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Na Cheng
- Department of Pathology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| |
Collapse
|
10
|
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
|
11
|
Pei J, Wang L, Li H. Development of a Better Nomogram for Prediction of Preoperative Microvascular Invasion and Postoperative Prognosis in Hepatocellular Carcinoma Patients: A Comparison Study. J Comput Assist Tomogr 2025; 49:9-22. [PMID: 38663025 PMCID: PMC11801467 DOI: 10.1097/rct.0000000000001618] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2024] [Accepted: 02/26/2024] [Indexed: 01/19/2025]
Abstract
OBJECTIVE Personalized precision medicine can be facilitated by clinically available preoperative microvascular invasion (MVI) prediction models that are reliable and postoperative MVI pathological grade-related recurrence prediction models that are accurate. In this study, we aimed to compare different mathematical models to derive the best preoperative prediction and postoperative recurrence prediction models for MVI. METHODS A total of 143 patients with hepatocellular carcinoma (HCC) whose clinical, laboratory, imaging, and pathological data were available were included in the analysis. Logistic regression, Cox proportional hazards regression, LASSO regression with 10-fold cross-validation, stepwise regression, and random forest methods were used for variable screening and predictive modeling. The accuracy and validity of seven preoperative MVI prediction models and five postoperative recurrence prediction models were compared in terms of C-index, net reclassification improvement, and integrated discrimination improvement. RESULTS Multivariate logistic regression analysis revealed that a preoperative nomogram model with the variables cirrhosis diagnosis, alpha-fetoprotein > 400, and diameter, shape, and number of lesions can predict MVI in patients with HCC reliably. Postoperatively, a nomogram model with MVI grade, number of lesions, capsule involvement status, macrovascular invasion, and shape as the variables was selected after LASSO regression and 10-fold cross-validation analysis to accurately predict the prognosis for different MVI grades. The number and shape of the lesions were the most common predictors of MVI preoperatively and recurrence postoperatively. CONCLUSIONS Our study identified the best statistical approach for the prediction of preoperative MVI as well as postoperative recurrence in patients with HCC based on clinical, imaging, and laboratory tests results. This could expedite preoperative treatment decisions and facilitate postoperative management.
Collapse
|
12
|
Ma L, Zhang C, Wen Y, Xing K, Li S, Geng Z, Liao S, Yuan S, Li X, Zhong C, Hou J, Zhang J, Gao M, Xu B, Guo R, Wei W, Xie C, Lu L. Imaging-based surrogate classification for risk stratification of hepatocellular carcinoma with microvascular invasion to adjuvant hepatic arterial infusion chemotherapy: a multicenter retrospective study. Int J Surg 2025; 111:872-883. [PMID: 39051653 PMCID: PMC11745592 DOI: 10.1097/js9.0000000000001903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 06/24/2024] [Indexed: 07/27/2024]
Abstract
BACKGROUND Patients with microvascular invasion (MVI)-positive hepatocellular carcinoma (HCC) have shown promising results with adjuvant hepatic arterial infusion chemotherapy (HAIC) with FOLFOX after curative resection. The authors aim to develop an imaging-derived biomarker to depict MVI-positive HCC patients more precisely and promote individualized treatment strategies of adjuvant HAIC. MATERIALS AND METHODS Patients with MVI-positive HCC were identified from five academic centers and utilized for model development ( n =470). Validation cohorts were pooled from a previously reported prospective clinical study conducted [control cohort ( n =145), adjuvant HAIC cohort ( n =143)] (NCT03192618). The primary endpoint was recurrence-free survival (RFS). Imaging features were thoroughly reviewed, and multivariable logistic regression analysis was employed for model development. Transcriptomic sequencing was conducted to identify the associated biological processes. RESULTS Arterial phase peritumoral enhancement, boundary of the tumor enhancement, tumor necrosis stratification, and boundary of the necrotic area were selected and incorporated into the nomogram for RFS. The imaging-based model successfully stratified patients into two distinct prognostic subgroups in both the training, control, and adjuvant HAIC cohorts (median RFS, 6.00 vs. 66.00 months, 4.86 vs. 24.30 months, 11.46 vs. 39.40 months, all P <0.01). Furthermore, no significant statistical difference was observed between patients at high risk of adjuvant HAIC and those in the control group ( P =0.61). The area under the receiver operating characteristic curve at 2 years was found to be 0.83, 0.84, and 0.73 for the training, control, and adjuvant HAIC cohorts, respectively. Transcriptomic sequencing analyses revealed associations between the radiological features and immune-regulating signal transduction pathways. CONCLUSION The utilization of this imaging-based model could help to better characterize MVI-positive HCC patients and facilitate the precise subtyping of patients who genuinely benefit from adjuvant HAIC treatment.
Collapse
Affiliation(s)
- Lidi Ma
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China
| | - Cheng Zhang
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China
| | - Yuhua Wen
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Liver Surgery, Sun Yat-sen University Cancer Center
| | - Kaili Xing
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Anesthesiology, Sun Yat-Sen University Cancer Center
| | - Shaohua Li
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Liver Surgery, Sun Yat-sen University Cancer Center
| | - Zhijun Geng
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China
| | - Shuting Liao
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China
| | - Shasha Yuan
- Department of Pathology, The First Affiliated Hospital, Sun Yat-sen University
| | - Xinming Li
- Department of Radiology, Zhujiang Hospital, Southern Medical University
| | - Chong Zhong
- Department of Hepatobilliary Surgery, The First Affiliated Hospital of Guangzhou University of Chinese Medicine
| | - Jing Hou
- Department of Radiology, Hunan Cancer Hospital; Changsha
| | - Jie Zhang
- Department of Radiology, Zhuhai People ‘s Hospital (Zhuhai hospital affiliated with Jinan University), Zhuhai, P.R China
| | - Mingyong Gao
- Department of Radiology, The First People’s Hospital of Foshan, Foshan, Guangdong
| | - Baojun Xu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou
| | - Rongping Guo
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Liver Surgery, Sun Yat-sen University Cancer Center
| | - Wei Wei
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Liver Surgery, Sun Yat-sen University Cancer Center
| | - Chuanmiao Xie
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Radiology, Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China
| | - Lianghe Lu
- Collaborative Innovation Center for Cancer Medicine, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center
- Department of Liver Surgery, Sun Yat-sen University Cancer Center
| |
Collapse
|
13
|
Famularo S, Penzo C, Maino C, Milana F, Oliva R, Marescaux J, Diana M, Romano F, Giuliante F, Ardito F, Grazi GL, Donadon M, Torzilli G. Preoperative detection of hepatocellular carcinoma's microvascular invasion on CT-scan by machine learning and radiomics: A preliminary analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:108274. [PMID: 38538504 DOI: 10.1016/j.ejso.2024.108274] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/20/2024] [Accepted: 03/16/2024] [Indexed: 08/22/2024]
Abstract
INTRODUCTION Microvascular invasion (MVI) is the main risk factor for overall mortality and recurrence after surgery for hepatocellular carcinoma (HCC).The aim was to train machine-learning models to predict MVI on preoperative CT scan. METHODS 3-phases CT scans were retrospectively collected among 4 Italian centers. DICOM files were manually segmented to detect the liver and the tumor(s). Radiomics features were extracted from the tumoral, peritumoral and healthy liver areas in each phase. Principal component analysis (PCA) was performed to reduce the dimensions of the dataset. Data were divided between training (70%) and test (30%) sets. Random-Forest (RF), fully connected MLP Artificial neural network (neuralnet) and extreme gradient boosting (XGB) models were fitted to predict MVI. Prediction accuracy was estimated in the test set. RESULTS Between 2008 and 2022, 218 preoperative CT scans were collected. At the histological specimen, 72(33.02%) patients had MVI. First and second order radiomics features were extracted, obtaining 672 variables. PCA selected 58 dimensions explaining >95% of the variance.In the test set, the XGB model obtained Accuracy = 68.7% (Sens: 38.1%, Spec: 83.7%, PPV: 53.3% and NPV: 73.4%). The neuralnet showed an Accuracy = 50% (Sens: 52.3%, Spec: 48.8%, PPV: 33.3%, NPV: 67.7%). RF was the best performer (Acc = 96.8%, 95%CI: 0.91-0.99, Sens: 95.2%, Spec: 97.6%, PPV: 95.2% and NPV: 97.6%). CONCLUSION Our model allowed a high prediction accuracy of the presence of MVI at the time of HCC diagnosis. This could lead to change the treatment allocation, the surgical extension and the follow-up strategy for those patients.
Collapse
Affiliation(s)
- Simone Famularo
- Hepatobiliary Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Catholic University of the Sacred Heart, Rome, Italy; Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; IRCAD, Research Institute Against Cancer of the Digestive System, 1 Place de l'Hôpital, Strasbourg, 67091, France.
| | - Camilla Penzo
- Pole d'Expertise de la Regulation Numérique (PEReN), Paris, France
| | - Cesare Maino
- Department of Radiology, San Gerardo Hospital, Monza, Italy
| | - Flavio Milana
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - Riccardo Oliva
- IRCAD, Research Institute Against Cancer of the Digestive System, 1 Place de l'Hôpital, Strasbourg, 67091, France
| | - Jacques Marescaux
- IRCAD, Research Institute Against Cancer of the Digestive System, 1 Place de l'Hôpital, Strasbourg, 67091, France
| | - Michele Diana
- IRCAD, Research Institute Against Cancer of the Digestive System, 1 Place de l'Hôpital, Strasbourg, 67091, France; Department of General, Digestive and Endocrine Surgery, University Hospital of Strasbourg, France; ICube Lab, Photonics for Health, Strasbourg, France
| | - Fabrizio Romano
- School of Medicine and Surgery, University of Milan-Bicocca, Department of Surgery, San Gerardo Hospital, Monza, Italy
| | - Felice Giuliante
- Hepatobiliary Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Francesco Ardito
- Hepatobiliary Surgery Unit, Fondazione Policlinico Universitario A. Gemelli, IRCCS, Catholic University of the Sacred Heart, Rome, Italy
| | - Gian Luca Grazi
- Division of Hepatobiliarypancreatic Unit, IRCCS - Regina Elena National Cancer Institute, Rome, Italy
| | - Matteo Donadon
- Department of Health Sciences, Università del Piemonte Orientale, Novara, Italy; Department of General Surgery, University Maggiore Hospital Della Carità, Novara, Italy
| | - Guido Torzilli
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Department of Hepatobiliary and General Surgery, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| |
Collapse
|
14
|
Jiang H, Li B, Zheng T, Qin Y, Wu Y, Wu Z, Ronot M, Chernyak V, Fowler KJ, Bashir MR, Chen W, Wang YC, Ju S, Song B. MRI-based prediction of microvascular invasion/high tumor grade and adjuvant therapy benefit for solitary HCC ≤ 5 cm: a multicenter cohort study. Eur Radiol 2024:10.1007/s00330-024-11295-1. [PMID: 39702639 DOI: 10.1007/s00330-024-11295-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 10/25/2024] [Accepted: 11/16/2024] [Indexed: 12/21/2024]
Abstract
OBJECTIVES To develop and externally validate an MRI-based diagnostic model for microvascular invasion (MVI) or Edmondson-Steiner G3/4 (i.e., high-risk histopathology) in solitary BCLC 0/A hepatocellular carcinoma (HCC) ≤ 5 cm and to assess its performance in predicting adjuvant therapy benefits. MATERIALS AND METHODS This multicenter retrospective cohort study included 577 consecutive adult patients who underwent contrast-enhanced MRI and subsequent curative resection or ablation for solitary BCLC 0/A HCC ≤ 5 cm (December 2011 to January 2024) from four hospitals. For resection-treated patients, a diagnostic model integrating clinical and 50 semantic MRI features was developed against pathology with logistic regression analyses on the training set (center 1) and externally validated on the testing dataset (centers 2-4), with its utilities in predicting posttreatment recurrence-free survival (RFS) and adjuvant therapy benefit evaluated by Cox regression analyses. RESULTS Serum α-fetoprotein > 100 ng/mL (odds ratio (OR), 1.94; p = 0.006), non-simple nodular growth subtype (OR, 1.69; p = 0.03), and the VICT2 trait (OR, 4.49; p < 0.001) were included in the MVI or high-grade (MHG) trait, with testing set AUC, sensitivity, and specificity of 0.832, 74.0%, and 82.5%, respectively. In the multivariable Cox analysis, the MHG-positive status was associated with worse RFS (resection testing set HR, 3.55, p = 0.02; ablation HR, 3.45, p < 0.001), and adjuvant therapy was associated with improved RFS only for the MHG-positive patients (resection HR, 0.39, p < 0.001; ablation HR, 0.30, p = 0.005). CONCLUSION The MHG trait effectively predicted high-risk histopathology, RFS and adjuvant therapy benefit among patients receiving curative resection or ablation for solitary BCLC 0/A HCC ≤ 5 cm. KEY POINTS Question Despite being associated with increased recurrence and potential benefit from adjuvancy in HCC, microvascular invasion or Edmondson-Steiner grade 3/4 are hardly assessable noninvasively. Findings We developed and externally validated an MRI-based model for predicting high-risk histopathology, post-resection/ablation recurrence-free survival, and adjuvant therapy benefit in solitary HCC ≤ 5 cm. Clinical relevance Among patients receiving curative-intent resection or ablation for solitary HCC ≤ 5 cm, noninvasive identification of high-risk histopathology (MVI or high-grade) using our proposed MRI model may help improve individualized prognostication and patient selection for adjuvant therapies.
Collapse
Affiliation(s)
- Hanyu Jiang
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Binrong Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China
| | - Tianying Zheng
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yun Qin
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuanan Wu
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhenru Wu
- Laboratory of Pathology, Key Laboratory of Transplant Engineering and Immunology, NHC, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Maxime Ronot
- Université Paris Cité, UMR 1149, CRI, Paris & Service de Radiologie, Hôpital Beaujon, APHP.Nord, Clichy, France
| | - Victoria Chernyak
- Department of Radiology, Memorial Sloan Kettering Cancer Center, NYC, New York, NY, USA
| | - Kathryn J Fowler
- Department of Radiology, University of California San Diego, San Diego, CA, USA
| | - Mustafa R Bashir
- Department of Radiology, Center for Advanced Magnetic Resonance in Medicine, and Division of Gastroenterology, Department of Medicine, Duke University Medical Center, Durham, NC, USA
| | - Weixia Chen
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan-Cheng Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, Jiangsu, China.
| | - Bin Song
- Department of Radiology, Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
- Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China.
| |
Collapse
|
15
|
Zeng S, Wang XL, Yang H. Radiomics and radiogenomics: extracting more information from medical images for the diagnosis and prognostic prediction of ovarian cancer. Mil Med Res 2024; 11:77. [PMID: 39673071 PMCID: PMC11645790 DOI: 10.1186/s40779-024-00580-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 11/07/2024] [Indexed: 12/15/2024] Open
Abstract
Ovarian cancer (OC) remains one of the most lethal gynecological malignancies globally. Despite the implementation of various medical imaging approaches for OC screening, achieving accurate differential diagnosis of ovarian tumors continues to pose significant challenges due to variability in image performance, resulting in a lack of objectivity that relies heavily on the expertise of medical professionals. This challenge can be addressed through the emergence and advancement of radiomics, which enables high-throughput extraction of valuable information from conventional medical images. Furthermore, radiomics can integrate with genomics, a novel approach termed radiogenomics, which allows for a more comprehensive, precise, and personalized assessment of tumor biological features. In this review, we present an extensive overview of the application of radiomics and radiogenomics in diagnosing and predicting ovarian tumors. The findings indicate that artificial intelligence methods based on imaging can accurately differentiate between benign and malignant ovarian tumors, as well as classify their subtypes. Moreover, these methods are effective in forecasting survival rates, treatment outcomes, metastasis risk, and recurrence for patients with OC. It is anticipated that these advancements will function as decision-support tools for managing OC while contributing to the advancement of precision medicine.
Collapse
Affiliation(s)
- Song Zeng
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Xin-Lu Wang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China
| | - Hua Yang
- Department of Ultrasound, Shengjing Hospital of China Medical University, Shenyang, 110004, China.
| |
Collapse
|
16
|
Xu ZL, Qian GX, Li YH, Lu JL, Wei MT, Bu XY, Ge YS, Cheng Y, Jia WD. Evaluating microvascular invasion in hepatitis B virus-related hepatocellular carcinoma based on contrast-enhanced computed tomography radiomics and clinicoradiological factors. World J Gastroenterol 2024; 30:4801-4816. [PMID: 39649551 PMCID: PMC11606376 DOI: 10.3748/wjg.v30.i45.4801] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 08/28/2024] [Accepted: 09/23/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND Microvascular invasion (MVI) is a significant indicator of the aggressive behavior of hepatocellular carcinoma (HCC). Expanding the surgical resection margin and performing anatomical liver resection may improve outcomes in patients with MVI. However, no reliable preoperative method currently exists to predict MVI status or to identify patients at high-risk group (M2). AIM To develop and validate models based on contrast-enhanced computed tomography (CECT) radiomics and clinicoradiological factors to predict MVI and identify M2 among patients with hepatitis B virus-related HCC (HBV-HCC). The ultimate goal of the study was to guide surgical decision-making. METHODS A total of 270 patients who underwent surgical resection were retrospectively analyzed. The cohort was divided into a training dataset (189 patients) and a validation dataset (81) with a 7:3 ratio. Radiomics features were selected using intra-class correlation coefficient analysis, Pearson or Spearman's correlation analysis, and the least absolute shrinkage and selection operator algorithm, leading to the construction of radscores from CECT images. Univariate and multivariate analyses identified significant clinicoradiological factors and radscores associated with MVI and M2, which were subsequently incorporated into predictive models. The models' performance was evaluated using calibration, discrimination, and clinical utility analysis. RESULTS Independent risk factors for MVI included non-smooth tumor margins, absence of a peritumoral hypointensity ring, and a high radscore based on delayed-phase CECT images. The MVI prediction model incorporating these factors achieved an area under the curve (AUC) of 0.841 in the training dataset and 0.768 in the validation dataset. The M2 prediction model, which integrated the radscore from the 5 mm peritumoral area in the CECT arterial phase, α-fetoprotein level, enhancing capsule, and aspartate aminotransferase level achieved an AUC of 0.865 in the training dataset and 0.798 in the validation dataset. Calibration and decision curve analyses confirmed the models' good fit and clinical utility. CONCLUSION Multivariable models were constructed by combining clinicoradiological risk factors and radscores to preoperatively predict MVI and identify M2 among patients with HBV-HCC. Further studies are needed to evaluate the practical application of these models in clinical settings.
Collapse
Affiliation(s)
- Zi-Ling Xu
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Gui-Xiang Qian
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yong-Hai Li
- Department of Anorectal Surgery, The First People's Hospital of Hefei, Hefei 230001, Anhui Province, China
| | - Jian-Lin Lu
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Ming-Tong Wei
- Department of General Surgery, Anhui Provincial Hospital Affiliated to Anhui Medical University, Hefei 230001, Anhui Province, China
| | - Xiang-Yi Bu
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yong-Sheng Ge
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Yuan Cheng
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| | - Wei-Dong Jia
- Department of General Surgery, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Division of Life Science and Medicine, University of Science and Technology of China, Hefei 230001, Anhui Province, China
| |
Collapse
|
17
|
Gu P, Mendonca O, Carter D, Dube S, Wang P, Huang X, Li D, Moore JH, McGovern DPB. AI-luminating Artificial Intelligence in Inflammatory Bowel Diseases: A Narrative Review on the Role of AI in Endoscopy, Histology, and Imaging for IBD. Inflamm Bowel Dis 2024; 30:2467-2485. [PMID: 38452040 DOI: 10.1093/ibd/izae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Indexed: 03/09/2024]
Abstract
Endoscopy, histology, and cross-sectional imaging serve as fundamental pillars in the detection, monitoring, and prognostication of inflammatory bowel disease (IBD). However, interpretation of these studies often relies on subjective human judgment, which can lead to delays, intra- and interobserver variability, and potential diagnostic discrepancies. With the rising incidence of IBD globally coupled with the exponential digitization of these data, there is a growing demand for innovative approaches to streamline diagnosis and elevate clinical decision-making. In this context, artificial intelligence (AI) technologies emerge as a timely solution to address the evolving challenges in IBD. Early studies using deep learning and radiomics approaches for endoscopy, histology, and imaging in IBD have demonstrated promising results for using AI to detect, diagnose, characterize, phenotype, and prognosticate IBD. Nonetheless, the available literature has inherent limitations and knowledge gaps that need to be addressed before AI can transition into a mainstream clinical tool for IBD. To better understand the potential value of integrating AI in IBD, we review the available literature to summarize our current understanding and identify gaps in knowledge to inform future investigations.
Collapse
Affiliation(s)
- Phillip Gu
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | | | - Dan Carter
- Department of Gastroenterology, Sheba Medical Center, Tel Aviv, Israel
| | - Shishir Dube
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Paul Wang
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Xiuzhen Huang
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Debiao Li
- Biomedical Research Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Jason H Moore
- Department of Computational Biomedicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Dermot P B McGovern
- F. Widjaja Inflammatory Bowel Disease Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
18
|
Elias-Neto A, Gonzaga APFC, Braga FA, Gomes NBN, Torres US, D'Ippolito G. Imaging Prognostic Biomarkers in Hepatocellular Carcinoma: A Comprehensive Review. Semin Ultrasound CT MR 2024; 45:454-463. [PMID: 39067621 DOI: 10.1053/j.sult.2024.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide with its incidence on the rise globally. This paper provides a comprehensive review of prognostic imaging markers in HCC, emphasizing their role in risk stratification and clinical decision-making. We explore quantitative and qualitative criteria derived from imaging studies, such as computed tomography (CT) and magnetic resonance imaging (MRI), which can offer valuable insights into the biological behavior of the tumor. While many of these markers are not yet widely integrated into current clinical guidelines, they represent a promising future direction for approaching this highly heterogeneous cancer. However, standardization and validation of these markers remain important challenges. We conclude by emphasizing the importance of ongoing research to enhance clinical practices and improve outcomes for patients with HCC.
Collapse
Affiliation(s)
- Abrahão Elias-Neto
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Ana Paula F C Gonzaga
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Fernanda A Braga
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Natália B N Gomes
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil
| | - Ulysses S Torres
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil; Department of Radiology, Grupo Fleury, São Paulo, São Paulo, Brazil.
| | - Giuseppe D'Ippolito
- Department of Diagnostic Imaging, Escola Paulista de Medicina, Universidade Federal de São Paulo (UNIFESP), São Paulo, São Paulo, Brazil; Department of Radiology, Grupo Fleury, São Paulo, São Paulo, Brazil
| |
Collapse
|
19
|
Nguyen T, Vennatt J, Downs L, Surabhi V, Stanietzky N. Advanced Imaging of Hepatocellular Carcinoma: A Review of Current and Novel Techniques. J Gastrointest Cancer 2024; 55:1469-1484. [PMID: 39158837 DOI: 10.1007/s12029-024-01094-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/16/2024] [Indexed: 08/20/2024]
Abstract
Hepatocellular carcinoma (HCC) is the most common primary carcinoma arising from the liver. Although HCC can arise de novo, the vast majority of cases develop in the setting of chronic liver disease. Hepatocarcinogenesis follows a well-studied process during which chronic inflammation and cellular damage precipitate cellular and genetic aberrations, with subsequent propagation of precancerous and cancerous lesions. Surveillance of individuals at high risk of HCC, early diagnosis, and individualized treatment are keys to reducing the mortality associated with this disease. Radiological imaging plays a critical role in the diagnosis and management of these patients. HCC is a unique cancer in that it can be diagnosed with confidence by imaging that meets all radiologic criteria, obviating the risks associated with tissue sampling. This article discusses conventional and emerging imaging techniques for the evaluation of HCC.
Collapse
Affiliation(s)
- Trinh Nguyen
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Jaijo Vennatt
- Department of Diagnostic Radiology, Baylor College of Medicine, 1 Baylor Plaza, Houston, TX, 77030, USA
| | - Lincoln Downs
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Venkateswar Surabhi
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | - Nir Stanietzky
- Department of Abdominal Imaging, The University of Texas MD Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA.
| |
Collapse
|
20
|
Rai R. Deep learning in image segmentation for cancer. J Med Radiat Sci 2024; 71:505-508. [PMID: 39503190 PMCID: PMC11638342 DOI: 10.1002/jmrs.839] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Accepted: 10/22/2024] [Indexed: 12/14/2024] Open
Abstract
This article discusses the role of deep learning (DL) in cancer imaging, focusing on its applications for automatic image segmentation. It highlights two studies that demonstrate how U-Net- and convolutional neural networks-based architectures have improved the speed and accuracy of body composition analysis in CT scans and rectal tumour segmentation in MRI images. While the results are promising, the article stresses the need for further research to address issues like image quality variability across different imaging systems.
Collapse
Affiliation(s)
- Robba Rai
- South Western Sydney Clinical SchoolUniversity of New South WalesLiverpoolNew South WalesAustralia
- Liverpool and Macarthur Cancer Therapy CentreLiverpool HospitalLiverpoolNew South WalesAustralia
- Ingham Institute for Applied Medical ResearchLiverpoolNew South WalesAustralia
| |
Collapse
|
21
|
Ren L, Chen DB, Yan X, She S, Yang Y, Zhang X, Liao W, Chen H. Bridging the Gap Between Imaging and Molecular Characterization: Current Understanding of Radiomics and Radiogenomics in Hepatocellular Carcinoma. J Hepatocell Carcinoma 2024; 11:2359-2372. [PMID: 39619602 PMCID: PMC11608547 DOI: 10.2147/jhc.s423549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy worldwide and the third leading cause of cancer-related deaths. Imaging plays a crucial role in the screening, diagnosis, and monitoring of HCC; however, the potential mechanism regarding phenotypes or molecular subtyping remains underexplored. Radiomics significantly expands the selection of features available by extracting quantitative features from imaging data. Radiogenomics bridges the gap between imaging and genetic/transcriptomic information by associating imaging features with critical genes and pathways, thereby providing biological annotations to these features. Despite challenges in interpreting these connections, assessing their universality, and considering the diversity in HCC etiology and genetic information across different populations, radiomics and radiogenomics offer new perspectives for precision treatment in HCC. This article provides an up-to-date summary of the advancements in radiomics and radiogenomics throughout the HCC care continuum, focusing on the clinical applications, advantages, and limitations of current techniques and offering prospects. Future research should aim to overcome these challenges to improve the prognosis of HCC patients and leverage imaging information for patient benefit.
Collapse
Affiliation(s)
- Liying Ren
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Dong Bo Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xuanzhi Yan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Shaoping She
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Yao Yang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Xue Zhang
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, Guilin, Guangxi, 541001, People’s Republic of China
| | - Hongsong Chen
- Peking University People’s Hospital, Peking University Hepatology Institute, Infectious Disease and Hepatology Center of Peking University People’s Hospital, Beijing Key Laboratory of Hepatitis C and Immunotherapy for Liver Diseases, Beijing International Cooperation Base for Science and Technology on NAFLD Diagnosis, Beijing, 100044, People’s Republic of China
| |
Collapse
|
22
|
Yoon JK, Han DH, Lee S, Choi JY, Choi GH, Kim DY, Kim MJ. Intraindividual comparison of prognostic imaging features of HCCs between MRIs with extracellular and hepatobiliary contrast agents. Liver Int 2024; 44:2847-2857. [PMID: 39105495 DOI: 10.1111/liv.16059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Revised: 07/18/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
BACKGROUND & AIMS Accumulating evidence suggests that certain imaging features of hepatocellular carcinoma (HCC) may have prognostic implications. This study aimed to intraindividually compare MRIs with extracellular contrast agent (ECA-MRI) and hepatobiliary agent (HBA-MRI) for prognostic imaging features of HCC and to compare the prediction of microvascular invasion (MVI) and early recurrence between the two MRIs. METHODS The present study included 102 prospectively enrolled at-risk patients (median age, 61.0 years; 83 men) with surgically resected single HCC with both preoperative ECA-MRI and HBA-MRI between July 2019 and June 2023. The McNemar test was used to compare each prognostic imaging feature between the two MRIs. Significant imaging features associated with MVI were identified by multivariable logistic regression analysis, and early recurrence rates (<2 years) were compared between the two MRIs. RESULTS The frequencies of prognostic imaging features were not significantly different between the two MRIs (p = .07 to >.99). Non-smooth tumour margin (ECA-MRI, odds ratio [OR] = 5.30; HBA-MRI, OR = 7.07) and peritumoral arterial phase hyperenhancement (ECA-MRI, OR = 4.26; HBA-MRI, OR = 4.43) were independent factors significantly associated with MVI on both MRIs. Two-trait predictor of venous invasion (presence of internal arteries and absence of hypoattenuating halo) on ECA-MRI (OR = 11.24) and peritumoral HBP hypointensity on HBA-MRI (OR = 20.42) were other predictors of MVI. Early recurrence rates of any two or more significant imaging features (49.8% on ECA-MRI vs 51.3% on HBA-MRI, p = .75) were not significantly different between the two MRIs. CONCLUSION Prognostic imaging features of HCC may be comparable between ECA-MRI and HBA-MRI.
Collapse
Affiliation(s)
- Ja Kyung Yoon
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dai Hoon Han
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sunyoung Lee
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jin-Young Choi
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Gi Hong Choi
- Department of Surgery, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Do Young Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Myeong-Jin Kim
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
23
|
Ou J, Zhou HY, Qin HL, Wang YS, Gou YQ, Luo H, Zhang XM, Chen TW. Baseline CT radiomics features to predict pathological complete response of advanced esophageal squamous cell carcinoma treated with neoadjuvant chemotherapy using paclitaxel and cisplatin. Eur J Radiol 2024; 181:111763. [PMID: 39341168 DOI: 10.1016/j.ejrad.2024.111763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 09/20/2024] [Accepted: 09/25/2024] [Indexed: 09/30/2024]
Abstract
PURPOSE To develop a CT radiomics model to predict pathological complete response (pCR) of advanced esophageal squamous cell carcinoma (ESCC) toneoadjuvant chemotherapy using paclitaxel and cisplatin. MATERIALS AND METHODS 326 consecutive patients with advanced ESCC from two hospitals undergoing baseline contrast-enhanced CT followed by neoadjuvant chemotherapy using paclitaxel and cisplatin were enrolled, including 115 patients achieving pCR and 211 patients without pCR. Of the 271 cases from 1st hospital, 188 and 83 cases were randomly allocated to the training and test cohorts, respectively. The 55 patients from a second hospital were assigned as an external validation cohort. Region of interest was segmented on the baseline thoracic contrast-enhanced CT. Useful radiomics features were generated by dimension reduction using least absolute shrinkage and selection operator. The optimal radiomics features were chosen using support vector machine (SVM). Discriminating performance was assessed with area under the receiver operating characteristic curve (ROC) and F-1score. The calibration curves and Brier score were used to evaluate the predictive accuracy. RESULTS Eight radiomics features were selected to create radiomics models related to pCR of advanced ESCC (P-values < 0.01 for both the training and test cohorts). SVM model showed the best performance (AUCs = 0.929, 0.868 and 0.866, F-1scores = 0.857, 0.847 and 0.737 in the training, test and external validation cohorts, respectively). The calibration curves and Brier scores indicated goodness-of-fit and its great predictive accuracy. CONCLUSION CT radiomics models could well help predict pCR of advanced ESCC, and SVM model could be a suitable predictive model.
Collapse
Affiliation(s)
- Jing Ou
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hai-Ying Zhou
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hui-Lin Qin
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yue-Su Wang
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Yue-Qin Gou
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Hui Luo
- Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China
| | - Xiao-Ming Zhang
- The First Clinical College of Jinan University, and Jinan University First Affiliated Hospital, Guangzhou, Guangdong 510630, China; Medical Imaging Key Laboratory of Sichuan Province, and Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan 637000, China.
| | - Tian-Wu Chen
- Department of Radiology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400010, China.
| |
Collapse
|
24
|
Birgin E, Nebelung H, Abdelhadi S, Rink JS, Froelich MF, Hetjens S, Rahbari M, Téoule P, Rasbach E, Reissfelder C, Weitz J, Schoenberg SO, Riediger C, Plodeck V, Rahbari NN. Development and validation of a digital biopsy model to predict microvascular invasion in hepatocellular carcinoma. Front Oncol 2024; 14:1360936. [PMID: 39376989 PMCID: PMC11457731 DOI: 10.3389/fonc.2024.1360936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2023] [Accepted: 08/30/2024] [Indexed: 10/09/2024] Open
Abstract
Background Microvascular invasion is a major histopathological risk factor of postoperative recurrence in patients with hepatocellular carcinoma. This study aimed to develop and validate a digital biopsy model using imaging features to predict microvascular invasion before hepatectomy. Methods A total of 217 consecutive patients who underwent hepatectomy for resectable hepatocellular carcinoma were enrolled at two tertiary-care reference centers. An imaging-based digital biopsy model was developed and internally validated using logistic regression analysis with adjustments for age, sex, etiology of disease, size and number of lesions. Results Three imaging features, i.e., non-smoothness of lesion margin (OR = 16.40), ill-defined pseudocapsula (OR = 4.93), and persistence of intratumoral internal artery (OR = 10.50), were independently associated with microvascular invasion and incorporated into a prediction model. A scoring system with 0 - 3 points was established for the prediction model. Internal validation confirmed an excellent calibration of the model. A cutoff of 2 points indicates a high risk of microvascular invasion (area under the curve 0.87). The overall survival and recurrence-free survival stratified by the risk model was significantly shorter in patients with high risk features of microvascular invasion compared to those patients with low risk of microvascular invasion (overall survival: median 35 vs. 75 months, P = 0.027; recurrence-free survival: median 17 vs. 38 months, P < 0.001)). Conclusion A preoperative assessment of microvascular invasion by digital biopsy is reliable, easily applicable, and might facilitate personalized treatment strategies.
Collapse
Affiliation(s)
- Emrullah Birgin
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany
| | - Heiner Nebelung
- Department of Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Schaima Abdelhadi
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Johann S. Rink
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Matthias F. Froelich
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Svetlana Hetjens
- Department of Medical Statistics and Biomathematics, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Mohammad Rahbari
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Patrick Téoule
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Erik Rasbach
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Reissfelder
- Department of Surgery, Universitätsmedizin Mannheim, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ Hector Cancer Institute at the University Medical Center Mannheim, Mannheim, Germany
| | - Jürgen Weitz
- Department of Visceral-, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Stefan O. Schoenberg
- Department of Radiology and Nuclear Medicine, University Medical Centre Mannheim, University of Heidelberg, Mannheim, Germany
| | - Carina Riediger
- Department of Visceral-, Thoracic and Vascular Surgery, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Verena Plodeck
- Department of Radiology, Faculty of Medicine and University Hospital Carl Gustav Carus, Technische Universität Dresden, Dresden, Germany
| | - Nuh N. Rahbari
- Department of General and Visceral Surgery, University Hospital Ulm, Ulm, Germany
| |
Collapse
|
25
|
Che F, Wei Y, Xu Q, Li Q, Zhang T, Wang LY, Li M, Yuan F, Song B. Noninvasive identification of SOX9 status using radiomics signatures may help construct personalized treatment strategy in hepatocellular carcinoma. Abdom Radiol (NY) 2024; 49:3024-3035. [PMID: 38446180 DOI: 10.1007/s00261-024-04190-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 12/31/2023] [Accepted: 01/16/2024] [Indexed: 03/07/2024]
Abstract
OBJECTIVES To develop and validate a radiomics-based model for predicting SOX9-positive hepatocellular carcinoma (HCC) using preoperative contrast-enhanced computed tomography (CT) images. METHODS From January 2013 to April 2017, patients with histologically proven HCC who received systemic sorafenib treatment after curative resection were retrospectively enrolled. Radiomic features were extracted from portal venous phase CT images and selected to build a radiomics score using logistic regression analysis. The factors associated with SOX9 expression were selected and combined by univariate and multivariate analyses to establish clinico-liver imaging (CL) model and clinico-liver imaging-radiomics (CLR) model. Diagnostic performance was measured by area under curve (AUC). Overall survival (OS) and recurrence-free survival (RFS) rates were compared using Kaplan-Meier method. RESULTS A total of 108 patients (training cohort: n = 80; validation cohort: n = 28) were enrolled. Multivariate analyses revealed that the albumin-bilirubin grade and tumor size were significant independent factors for predicting SOX9-positive HCCs and were included in the CL model. The CLR model integrating the radiomics score with albumin-bilirubin grade and tumor size showed better discriminative performance than the CL model with AUCs of 0.912 and 0.790 in the training and validation cohorts. Survival curves for RFS and OS showed that SOX9 expression was closely related to the prognosis of HCC patients. RFS and OS rates were significantly lower in patients with SOX9-positive than SOX9-negative (51.02% vs. 75.00% at 1-year RFS rates; 76.92% vs. 94.94% at 2-year OS rates). CONCLUSION Radiomics signatures may serve as noninvasive predictors for SOX9 status evaluation in patients with HCC and may aid in constructing individualized treatment strategies.
Collapse
Affiliation(s)
- Feng Che
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Qing Xu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Tong Zhang
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China
| | - Li-Ye Wang
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Man Li
- Department of Research and Development, Shanghai United Imaging Intelligence Co., Ltd., Shanghai, China
| | - Fang Yuan
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, No 37, Guoxue Alley, Chengdu, 610041, Sichuan, China.
- Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China.
| |
Collapse
|
26
|
Li H, Zhang D, Pei J, Hu J, Li X, Liu B, Wang L. Dual-energy computed tomography iodine quantification combined with laboratory data for predicting microvascular invasion in hepatocellular carcinoma: a two-centre study. Br J Radiol 2024; 97:1467-1475. [PMID: 38870535 PMCID: PMC11256957 DOI: 10.1093/bjr/tqae116] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2023] [Revised: 05/16/2024] [Accepted: 06/05/2024] [Indexed: 06/15/2024] Open
Abstract
OBJECTIVES Microvascular invasion (MVI) is a recognized biomarker associated with poorer prognosis in patients with hepatocellular carcinoma. Dual-energy computed tomography (DECT) is a highly sensitive technique that can determine the iodine concentration (IC) in tumour and provide an indirect evaluation of internal microcirculatory perfusion. This study aimed to assess whether the combination of DECT with laboratory data can improve preoperative MVI prediction. METHODS This retrospective study enrolled 119 patients who underwent DECT liver angiography at 2 medical centres preoperatively. To compare DECT parameters and laboratory findings between MVI-negative and MVI-positive groups, Mann-Whitney U test was used. Additionally, principal component analysis (PCA) was conducted to determine fundamental components. Mann-Whitney U test was applied to determine whether the principal component (PC) scores varied across MVI groups. Finally, a general linear classifier was used to assess the classification ability of each PC score. RESULTS Significant differences were noted (P < .05) in alpha-fetoprotein (AFP) level, normalized arterial phase IC, and normalized portal phase IC between the MVI groups in the primary and validation datasets. The PC1-PC4 accounted for 67.9% of the variance in the primary dataset, with loadings of 24.1%, 16%, 15.4%, and 12.4%, respectively. In both primary and validation datasets, PC3 and PC4 were significantly different across MVI groups, with area under the curve values of 0.8410 and 0.8373, respectively. CONCLUSIONS The recombination of DECT IC and laboratory features based on varying factor loadings can well predict MVI preoperatively. ADVANCES IN KNOWLEDGE Utilizing PCA, the amalgamation of DECT IC and laboratory features, considering diverse factor loadings, showed substantial promise in accurately classifying MVI. There have been limited endeavours to establish such a combination, offering a novel paradigm for comprehending data in related research endeavours.
Collapse
Affiliation(s)
- Huan Li
- Department of Radiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imaging Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Dai Zhang
- Department of Radiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imaging Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Jinxia Pei
- Department of Radiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imaging Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Jingmei Hu
- Department of Radiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imaging Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| | - Xiaohu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Bin Liu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
| | - Longsheng Wang
- Department of Radiology, The Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui 230601, China
- Medical Imaging Research Center, Anhui Medical University, Hefei, Anhui 230601, China
| |
Collapse
|
27
|
Li C, Hu J, Zhang Z, Wei C, Chen T, Wang X, Dai Y, Shen J. Biparametric MRI of the prostate radiomics model for prediction of pelvic lymph node metastasis in prostate cancers : a two-centre study. BMC Med Imaging 2024; 24:185. [PMID: 39054441 PMCID: PMC11271060 DOI: 10.1186/s12880-024-01372-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 07/18/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVES Exploring the value of adding correlation analysis (radiomic features (RFs) of pelvic metastatic lymph nodes and primary lesions) to screen RFs of primary lesions in the feature selection process of establishing prediction model. METHODS A total of 394 prostate cancer (PCa) patients (263 in the training group, 74 in the internal validation group and 57 in the external validation group) from two tertiary hospitals were included in the study. The cases with pelvic lymph node metastasis (PLNM) positive in the training group were diagnosed by biopsy or MRI with a short-axis diameter ≥ 1.5 cm, PLNM-negative cases in the training group and all cases in validation group were underwent both radical prostatectomy (RP) and extended pelvic lymph node dissection (ePLND). The RFs of PLNM-negative lesion and PLNM-positive tissues including primary lesions and their metastatic lymph nodes (MLNs) in the training group were extracted from T2WI and apparent diffusion coefficient (ADC) map to build the following two models by fivefold cross-validation: the lesion model, established according to the primary lesion RFs selected by t tests and absolute shrinkage and selection operator (LASSO); the lesion-correlation model, established according to the primary lesion RFs selected by Pearson correlation analysis (RFs of primary lesions and their MLNs, correlation coefficient > 0.9), t test and LASSO. Finally, we compared the performance of these two models in predicting PLNM. RESULTS The AUC and the DeLong test of AUC in the lesion model and lesion-correlation model were as follows: training groups (0.8053, 0.8466, p = 0.0002), internal validation group (0.7321, 0.8268, p = 0.0429), and external validation group (0.6445, 0.7874, p = 0.0431), respectively. CONCLUSION The lesion-correlation model established by features of primary tumors correlated with MLNs has more advantages than the lesion model in predicting PLNM.
Collapse
Affiliation(s)
- Chunxing Li
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
- Department of MRI Room, Yancheng First Hospital Affiliated Hospital of NanJing University Medical School, Yancheng, China
| | - Jisu Hu
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Zhiyuan Zhang
- School of Medical Imaging, Biomedical Engineering, Xuzhou Medical University, Xuzhou, China
| | - Chaogang Wei
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Chen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Ximing Wang
- Department of Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Yakang Dai
- Suzhou Institute of Biomedical Engineering and Technology, Chinese Academy of Sciences, Suzhou, China
| | - Junkang Shen
- Department of Radiology, The Second Affiliated Hospital of Soochow University, Suzhou, China.
| |
Collapse
|
28
|
Schmidt R, Hamm CA, Rueger C, Xu H, He Y, Gottwald LA, Gebauer B, Savic LJ. Decision-Tree Models Indicative of Microvascular Invasion on MRI Predict Survival in Patients with Hepatocellular Carcinoma Following Tumor Ablation. J Hepatocell Carcinoma 2024; 11:1279-1293. [PMID: 38974016 PMCID: PMC11227855 DOI: 10.2147/jhc.s454487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2023] [Accepted: 04/18/2024] [Indexed: 07/09/2024] Open
Abstract
Purpose Histological microvascular invasion (MVI) is a risk factor for poor survival and early recurrence in hepatocellular carcinoma (HCC) after surgery. Its prognostic value in the setting of locoregional therapies (LRT), where no tissue samples are obtained, remains unknown. This study aims to establish CT-derived indices indicative of MVI on liver MRI with superior soft tissue contrast and evaluate their association with patient survival after ablation via interstitial brachytherapy (iBT) versus iBT combined with prior conventional transarterial chemoembolization (cTACE). Patients and Methods Ninety-five consecutive patients, who underwent ablation via iBT alone (n = 47) or combined with cTACE (n = 48), were retrospectively included between 01/2016 and 12/2017. All patients received contrast-enhanced MRI prior to LRT. Overall (OS), progression-free survival (PFS), and time-to-progression (TTP) were assessed. Decision-tree models to determine Radiogenomic Venous Invasion (RVI) and Two-Trait Predictor of Venous Invasion (TTPVI) on baseline MRI were established, validated on an external test set (TCGA-LIHC), and applied in the study cohorts to investigate their prognostic value for patient survival. Statistics included Fisher's exact and t-test, Kaplan-Meier and cox-regression analysis, area under the receiver operating characteristic curve (AUC-ROC) and Pearson's correlation. Results OS, PFS, and TTP were similar in both treatment groups. In the external dataset, RVI showed low sensitivity but relatively high specificity (AUC-ROC = 0.53), and TTPVI high sensitivity but only low specificity (AUC-ROC = 0.61) for histological MVI. In patients following iBT alone, positive RVI and TTPVI traits were associated with poorer OS (RVI: p < 0.01; TTPVI: p = 0.08), PFS (p = 0.04; p = 0.04), and TTP (p = 0.14; p = 0.03), respectively. However, when patients with combined cTACE and iBT were stratified by RVI or TTPVI, no differences in OS (p = 0.75; p = 0.55), PFS (p = 0.70; p = 0.43), or TTP (p = 0.33; p = 0.27) were observed. Conclusion The study underscores the role of non-invasive imaging biomarkers indicative of MVI to identify patients, who would potentially benefit from embolotherapy via cTACE prior to ablation rather than ablation alone.
Collapse
Affiliation(s)
- Robin Schmidt
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
- Experimental Clinical Research Center (ECRC) at Charité - Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, 13125, Germany
| | - Charlie Alexander Hamm
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, 10117, Germany
| | - Christopher Rueger
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
| | - Han Xu
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
| | - Yubei He
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
- Experimental Clinical Research Center (ECRC) at Charité - Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, 13125, Germany
| | | | - Bernhard Gebauer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
| | - Lynn Jeanette Savic
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Department of Radiology, Berlin, 13353, Germany
- Experimental Clinical Research Center (ECRC) at Charité - Universitätsmedizin Berlin and Max-Delbrück-Centrum für Molekulare Medizin (MDC), Berlin, 13125, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, Berlin, 10117, Germany
| |
Collapse
|
29
|
Arefan D, D'Ardenne NM, Iranpour N, Catania R, Yousef J, Chupetlovska K, Moghe A, Sholosh B, Thangasamy S, Borhani AA, Singhi AD, Monga SP, Furlan A, Wu S. Quantitative radiomics and qualitative LI-RADS imaging descriptors for non-invasive assessment of β-catenin mutation status in hepatocellular carcinoma. Abdom Radiol (NY) 2024; 49:2220-2230. [PMID: 38782785 DOI: 10.1007/s00261-024-04344-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/09/2024] [Accepted: 04/16/2024] [Indexed: 05/25/2024]
Abstract
PURPOSE Gain-of-function mutations in CTNNB1, gene encoding for β-catenin, are observed in 25-30% of hepatocellular carcinomas (HCCs). Recent studies have shown β-catenin activation to have distinct roles in HCC susceptibility to mTOR inhibitors and resistance to immunotherapy. Our goal was to develop and test a computational imaging-based model to non-invasively assess β-catenin activation in HCC, since liver biopsies are often not done due to risk of complications. METHODS This IRB-approved retrospective study included 134 subjects with pathologically proven HCC and available β-catenin activation status, who also had either CT or MR imaging of the liver performed within 1 year of histological assessment. For qualitative descriptors, experienced radiologists assessed the presence of imaging features listed in LI-RADS v2018. For quantitative analysis, a single biopsy proven tumor underwent a 3D segmentation and radiomics features were extracted. We developed prediction models to assess the β-catenin activation in HCC using both qualitative and quantitative descriptors. RESULTS There were 41 cases (31%) with β-catenin mutation and 93 cases (69%) without. The model's AUC was 0.70 (95% CI 0.60, 0.79) using radiomics features and 0.64 (0.52, 0.74; p = 0.468) using qualitative descriptors. However, when combined, the AUC increased to 0.88 (0.80, 0.92; p = 0.009). Among the LI-RADS descriptors, the presence of a nodule-in-nodule showed a significant association with β-catenin mutations (p = 0.015). Additionally, 88 radiomics features exhibited a significant association (p < 0.05) with β-catenin mutations. CONCLUSION Combination of LI-RADS descriptors and CT/MRI-derived radiomics determine β-catenin activation status in HCC with high confidence, making precision medicine a possibility.
Collapse
Affiliation(s)
- Dooman Arefan
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nicholas M D'Ardenne
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
| | - Negaur Iranpour
- Department of Radiology, Stanford University School of Medicine, Stanford, CA, USA
| | - Roberta Catania
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. Saint Clair Street, Suite 800, Chicago, IL, 60611, USA
| | - Jacob Yousef
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
| | - Kalina Chupetlovska
- Diagnostic Imaging Department, University Hospital "Saint Ivan Rilski", Sofia, Bulgaria
| | - Akshata Moghe
- Division of Gastroenterology and Hepatology, Department of Medicine, University of Texas Medical Branch, Galveston, TX, USA
| | - Biatta Sholosh
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
| | - Senthur Thangasamy
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
| | - Amir A Borhani
- Department of Radiology, Northwestern University Feinberg School of Medicine, 676 N. Saint Clair Street, Suite 800, Chicago, IL, 60611, USA
| | - Aatur D Singhi
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh Medical Center, S405A-BST, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
| | - Satdarshan P Monga
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh Medical Center, S405A-BST, 200 Lothrop Street, Pittsburgh, PA, 15261, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Department of Medicine, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Alessandro Furlan
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Shandong Wu
- Department of Radiology, University of Pittsburgh Medical Center, 200 Lothrop Street, Suite 200, Pittsburgh, PA, 15213, USA.
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| |
Collapse
|
30
|
Wang Q, Zhou Y, Yang H, Zhang J, Zeng X, Tan Y. MRI-based clinical-radiomics nomogram model for predicting microvascular invasion in hepatocellular carcinoma. Med Phys 2024; 51:4673-4686. [PMID: 38642400 DOI: 10.1002/mp.17087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/22/2024] Open
Abstract
BACKGROUND Preoperative microvascular invasion (MVI) of liver cancer is an effective method to reduce the recurrence rate of liver cancer. Hepatectomy with extended resection and additional adjuvant or targeted therapy can significantly improve the survival rate of MVI+ patients by eradicating micrometastasis. Preoperative prediction of MVI status is of great clinical significance for surgical decision-making and the selection of other adjuvant therapy strategies to improve the prognosis of patients. PURPOSE Established a radiomics machine learning model based on multimodal MRI and clinical data, and analyzed the preoperative prediction value of this model for microvascular invasion (MVI) of hepatocellular carcinoma (HCC). METHOD The preoperative liver MRI data and clinical information of 130 HCC patients who were pathologically confirmed to be pathologically confirmed were retrospectively studied. These patients were divided into MVI-positive group (MVI+) and MVI-negative group (MVI-) based on postoperative pathology. After a series of dimensionality reduction analysis, six radiomic features were finally selected. Then, linear support vector machine (linear SVM), support vector machine with rbf kernel function (rbf-SVM), logistic regression (LR), Random forest (RF) and XGBoost (XGB) algorithms were used to establish the MVI prediction model for preoperative HCC patients. Then, rbf-SVM with the best predictive performance was selected to construct the radiomics score (R-score). Finally, we combined R-score and clinical-pathology-image independent predictors to establish a combined nomogram model and corresponding individual models. The predictive performance of individual models and combined nomogram was evaluated and compared by receiver operating characteristic curve (ROC). RESULT Alpha-fetoprotein concentration, peritumor enhancement, maximum tumor diameter, smooth tumor margins, tumor growth pattern, presence of intratumor hemorrhage, and RVI were independent predictors of MVI. Compared with individual models, the final combined nomogram model (AUC: 0.968, 95% CI: 0.920-1.000) constructed by radiometry score (R-score) combined with clinicopathological parameters and apparent imaging features showed the optimal predictive performance. CONCLUSION This multi-parameter combined nomogram model had a good performance in predicting MVI of HCC, and had certain auxiliary value for the formulation of surgical plan and evaluation of prognosis.
Collapse
Affiliation(s)
- Qinghua Wang
- Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center For Medical Imaging In Jiangxi Province, Nanchang, China
| | - Yongjie Zhou
- Department of Radiology, Jiangxi Cancer Hospital, Nanchang, China
| | - Hongan Yang
- Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center For Medical Imaging In Jiangxi Province, Nanchang, China
| | - Jingrun Zhang
- Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center For Medical Imaging In Jiangxi Province, Nanchang, China
| | - Xianjun Zeng
- Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center For Medical Imaging In Jiangxi Province, Nanchang, China
| | - Yongming Tan
- Department of Radiology, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China
- Clinical Research Center For Medical Imaging In Jiangxi Province, Nanchang, China
| |
Collapse
|
31
|
Zhu Y, Feng B, Wang P, Wang B, Cai W, Wang S, Meng X, Wang S, Zhao X, Ma X. Bi-regional dynamic contrast-enhanced MRI for prediction of microvascular invasion in solitary BCLC stage A hepatocellular carcinoma. Insights Imaging 2024; 15:149. [PMID: 38886267 PMCID: PMC11183021 DOI: 10.1186/s13244-024-01720-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 05/23/2024] [Indexed: 06/20/2024] Open
Abstract
OBJECTIVES To construct a combined model based on bi-regional quantitative dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI), as well as clinical-radiological (CR) features for predicting microvascular invasion (MVI) in solitary Barcelona Clinic Liver Cancer (BCLC) stage A hepatocellular carcinoma (HCC), and to assess its ability for stratifying the risk of recurrence after hepatectomy. METHODS Patients with solitary BCLC stage A HCC were prospective collected and randomly divided into training and validation sets. DCE perfusion parameters were obtained both in intra-tumoral region (ITR) and peritumoral region (PTR). Combined DCE perfusion parameters (CDCE) were constructed to predict MVI. The combined model incorporating CDCE and CR features was developed and evaluated. Kaplan-Meier method was used to investigate the prognostic significance of the model and the survival benefits of different hepatectomy approaches. RESULTS A total of 133 patients were included. Total blood flow in ITR and arterial fraction in PTR exhibited the best predictive performance for MVI with areas under the curve (AUCs) of 0.790 and 0.792, respectively. CDCE achieved AUCs of 0.868 (training set) and 0.857 (validation set). A combined model integrated with the α-fetoprotein, corona enhancement, two-trait predictor of venous invasion, and CDCE could improve the discrimination ability to AUCs of 0.966 (training set) and 0.937 (validation set). The combined model could stratify the prognosis of HCC patients. Anatomical resection was associated with a better prognosis in the high-risk group (p < 0.05). CONCLUSION The combined model integrating DCE perfusion parameters and CR features could be used for MVI prediction in HCC patients and assist clinical decision-making. CRITICAL RELEVANCE STATEMENT The combined model incorporating bi-regional DCE-MRI perfusion parameters and CR features predicted MVI preoperatively, which could stratify the risk of recurrence and aid in optimizing treatment strategies. KEY POINTS Microvascular invasion (MVI) is a significant predictor of prognosis for hepatocellular carcinoma (HCC). Quantitative DCE-MRI could predict MVI in solitary BCLC stage A HCC; the combined model improved performance. The combined model could help stratify the risk of recurrence and aid treatment planning.
Collapse
Affiliation(s)
- Yongjian Zhu
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bing Feng
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Peng Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Bingzhi Wang
- Department of Pathology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Wei Cai
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Shuang Wang
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xuan Meng
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Sicong Wang
- Magnetic Resonance Imaging Research, General Electric Healthcare (China), Beijing, 100176, China
| | - Xinming Zhao
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China
| | - Xiaohong Ma
- Department of Diagnostic Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, China.
| |
Collapse
|
32
|
Yan X, Li Y, Qin W, Liao J, Fan J, Xie Y, Wang Z, Li S, Liao W. Radiomics model based on contrast-enhanced computed tomography imaging for early recurrence monitoring after radical resection of AFP-negative hepatocellular carcinoma. BMC Cancer 2024; 24:700. [PMID: 38849749 PMCID: PMC11157869 DOI: 10.1186/s12885-024-12436-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
BACKGROUND Although radical surgical resection is the most effective treatment for hepatocellular carcinoma (HCC), the high rate of postoperative recurrence remains a major challenge, especially in patients with alpha-fetoprotein (AFP)-negative HCC who lack effective biomarkers for postoperative recurrence surveillance. Emerging radiomics can reveal subtle structural changes in tumors by analyzing preoperative contrast-enhanced computer tomography (CECT) imaging data and may provide new ways to predict early recurrence (recurrence within 2 years) in AFP-negative HCC. In this study, we propose to develop a radiomics model based on preoperative CECT to predict the risk of early recurrence after surgery in AFP-negative HCC. PATIENTS AND METHODS Patients with AFP-negative HCC who underwent radical resection were included in this study. A computerized tool was used to extract radiomic features from the tumor region of interest (ROI), select the best radiographic features associated with patient's postoperative recurrence, and use them to construct the radiomics score (RadScore), which was then combined with clinical and follow-up information to comprehensively evaluate the reliability of the model. RESULTS A total of 148 patients with AFP-negative HCC were enrolled in this study, and 1,977 radiographic features were extracted from CECT, 2 of which were the features most associated with recurrence in AFP-negative HCC. They had good predictive ability in both the training and validation cohorts, with an area under the ROC curve (AUC) of 0.709 and 0.764, respectively. Tumor number, microvascular invasion (MVI), AGPR and radiomic features were independent risk factors for early postoperative recurrence in patients with AFP-negative HCC. The AUCs of the integrated model in the training and validation cohorts were 0.793 and 0.791, respectively. The integrated model possessed the clinical value of predicting early postoperative recurrence in patients with AFP-negative HCC according to decision curve analysis, which allowed the classification of patients into subgroups of high-risk and low-risk for early recurrence. CONCLUSION The nomogram constructed by combining clinical and imaging features has favorable performance in predicting the probability of early postoperative recurrence in AFP-negative HCC patients, which can help optimize the therapeutic decision-making and prognostic assessment of AFP-negative HCC patients.
Collapse
Affiliation(s)
- Xuanzhi Yan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China
| | - Yicheng Li
- Department of Burns, Nanfang Hospital, Southern Medical University, Guangzhou, 510515, Guangdong, P.R. China
| | - Wanying Qin
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China
| | - Jiayi Liao
- School of medical, Youjiang Medical University for Nationalities, Baise, 533000, Guangxi, P.R. China
| | - Jiaxing Fan
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China
| | - Yujin Xie
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China
| | - Zewen Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Guilin Medical University, No. 212, Renmin Road, Lingui District, Guilin, 541100, Guangxi, P.R. China.
| | - Siming Li
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China.
| | - Weijia Liao
- Laboratory of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Guilin Medical University, No. 15, Lequn Road, Xiufeng District, Guilin, 541001, Guangxi, P.R. China.
| |
Collapse
|
33
|
Jiang J, Li L, Yin G, Luo H, Li J. A Molecular Typing Method for Invasive Breast Cancer by Serum Raman Spectroscopy. Clin Breast Cancer 2024; 24:376-383. [PMID: 38492997 DOI: 10.1016/j.clbc.2024.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2023] [Revised: 01/17/2024] [Accepted: 02/12/2024] [Indexed: 03/18/2024]
Abstract
BACKGROUND The incidence of breast cancer ranks highest among cancers and is exceedingly heterogeneous. Immunohistochemical staining is commonly used clinically to identify the molecular subtype for subsequent treatment and prognosis. PURPOSE Raman spectroscopy and support vector machine (SVM) learning algorithm were utilized to identify blood samples from breast cancer patients in order to investigate a novel molecular typing approach. METHOD Tumor tissue coarse needle aspiration biopsy samples, and peripheral venous blood samples were gathered from 459 invasive breast cancer patients admitted to the breast department of Sichuan Cancer Hospital between June 2021 and September 2022. Immunohistochemical staining and in situ hybridization were performed on the coarse needle aspiration biopsy tissues to obtain their molecular typing pathological labels, including: 70 cases of Luminal A, 167 cases of Luminal B (HER2-positive), 57 cases of Luminal B (HER2-negative), 84 cases of HER2-positive, and 81 cases of triple-negative. Blood samples were processed to obtained Raman spectra taken for SVM classification models establishment with machine algorithms (using 80% of the sample data as the training set), and then the performance of the SVM classification models was evaluated by the independent validation set (20% of the sample data). RESULTS The AUC values of SVM classification models remained above 0.85, demonstrating outstanding model performance and excellent subtype discrimination of breast cancer molecular subtypes. CONCLUSION Raman spectroscopy of serum samples can promptly and precisely detect the molecular subtype of invasive breast cancer, which has the potential for clinical value.
Collapse
Affiliation(s)
- Jun Jiang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, China; Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Lintao Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Gang Yin
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Huaichao Luo
- Department of Laboratory, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China
| | - Junjie Li
- Department of Breast Surgery, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| |
Collapse
|
34
|
Zhang J, Li Y, Xia J, Pan X, Lu L, Fu J, Jia N. Prediction of Microvascular Invasion and Recurrence After Curative Resection of LI-RADS Category 5 Hepatocellular Carcinoma on Gd-BOPTA Enhanced MRI. J Hepatocell Carcinoma 2024; 11:941-952. [PMID: 38813100 PMCID: PMC11135558 DOI: 10.2147/jhc.s459686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 05/17/2024] [Indexed: 05/31/2024] Open
Abstract
Objective This study aims to investigate the predictive value of Gadobenate dimeglumine (Gd-BOPTA) enhanced MRI features on microvascular invasion (MVI) and recurrence in patients with Liver Imaging Reporting and Data System (LI-RADS) category 5 hepatocellular carcinoma (HCC). Methods A total of 132 patients with LI-RADS category 5 HCC who underwent curative resection and Gd-BOPTA enhanced MRI at our hospital between January 2016 and December 2018 were retrospectively analyzed. Qualitative evaluation based on LI-RADS v2018 imaging features was performed. Logistic regression analyses were conducted to assess the predictive significance of these features for MVI, and the Cox proportional hazards model was used to identify postoperative risk factors of recurrence. The recurrence-free survival (RFS) was analyzed by using the Kaplan-Meier curve and Log rank test. Results Multivariate logistic regression analysis identified that corona enhancement (odds ratio [OR] = 3.217; p < 0.001), internal arteries (OR = 4.147; p = 0.004), and peritumoral hypointensity on hepatobiliary phase (HBP) (OR = 5.165; p < 0.001) were significantly associated with MVI. Among the 132 patients with LR-5 HCC, 62 patients experienced postoperative recurrence. Multivariate Cox regression analysis showed that mosaic architecture (hazard ratio [HR] = 1.982; p = 0.014), corona enhancement (HR = 1.783; p = 0.039), and peritumoral hypointensity on HBP (HR = 2.130; p = 0.009) were risk factors for poor RFS. Conclusion MRI features based on Gd-BOPTA can be noninvasively and effectively predict MVI and recurrence of LR-5 HCC patients.
Collapse
Affiliation(s)
- Juan Zhang
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Yinqiao Li
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, China
| | - Jinju Xia
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Xingpeng Pan
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Lun Lu
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Jiazhao Fu
- Department of Organ Transplantation, Changhai Hospital, First Affiliated Hospital of Naval Medical University, Shanghai, China
| | - Ningyang Jia
- Department of Radiology, Eastern Hepatobiliary Surgery Hospital, Third Affiliated Hospital of Naval Medical University, Shanghai, China
| |
Collapse
|
35
|
Li X, Liang X, Li Z, Liang J, Qi Z, Zhong L, Geng Z, Liang W, Quan X, Liang C, Liu Z. A novel stratification scheme combined with internal arteries in CT imaging for guiding postoperative adjuvant transarterial chemoembolization in hepatocellular carcinoma: a retrospective cohort study. Int J Surg 2024; 110:2556-2567. [PMID: 38377071 DOI: 10.1097/js9.0000000000001191] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Accepted: 01/31/2024] [Indexed: 02/22/2024]
Abstract
BACKGROUND Although postoperative adjuvant transarterial chemoembolization (PA-TACE) improves survival outcomes in a subset of patients with resected hepatocellular carcinoma (HCC), the lack of reliable biomarkers for patient selection remains a significant challenge. The present study aimed to evaluate whether computed tomography imaging can provide more value for predicting benefits from PA-TACE and to establish a new scheme for guiding PA-TACE benefits. METHODS In this retrospective study, patients with HCC who had undergone preoperative contrast-enhanced computed tomography and curative hepatectomy were evaluated. Inverse probability of treatment weight was performed to balance the difference of baseline characteristics. Cox models were used to test the interaction among PA-TACE, imaging features, and pathological indicators. An HCC imaging and pathological classification (HIPC) scheme incorporating these imaging and pathological indicators was established. RESULTS This study included 1488 patients [median age, 52 years (IQR, 45-61 years); 1309 male]. Microvascular invasion (MVI) positive, and diameter >5 cm tumors achieved a higher recurrence-free survival (RFS), and overall survival (OS) benefit, respectively, from PA-TACE than MVI negative, and diameter ≤5 cm tumors. Patients with internal arteries (IA) positive benefited more than those with IA-negative in terms of RFS ( P =0.016) and OS ( P =0.018). PA-TACE achieved significant RFS and OS improvements in HIPC3 (IA present and diameter >5 cm, or two or three tumors) patients but not in HIPC1 (diameter ≤5 cm, MVI negative) and HIPC2 (other single tumor) patients. Our scheme may decrease the number of patients receiving PA-TACE by ~36.5% compared to the previous suggestion. CONCLUSIONS IA can provide more value for predicting the benefit of PA-TACE treatment. The proposed HIPC scheme can be used to stratify patients with and without survival benefits from PA-TACE.
Collapse
Affiliation(s)
- Xinming Li
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Department of Radiology
| | - Xiangjing Liang
- Ultrasound Medical Center, Zhujiang Hospital Southern Medical University
| | - Zhipeng Li
- Department of Radiology, Sun Yat-sen University Cancer Center
| | - Jianye Liang
- Department of Radiology, Sun Yat-sen University Cancer Center
| | | | - Liming Zhong
- School of Biomedical Engineering, Southern Medical University
| | - Zhijun Geng
- Department of Radiology, Sun Yat-sen University Cancer Center
| | | | | | - Changhong Liang
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, People's Republic of China
| | - Zaiyi Liu
- Department of Radiology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University
- Guangdong Provincial Key Laboratory of Artificial Intelligence in Medical Image Analysis and Application, Guangzhou, People's Republic of China
| |
Collapse
|
36
|
Naghavi AO, Bryant JM, Kim Y, Weygand J, Redler G, Sim AJ, Miller J, Coucoules K, Michael LT, Gloria WE, Yang G, Rosenberg SA, Ahmed K, Bui MM, Henderson-Jackson EB, Lee A, Lee CD, Gonzalez RJ, Feygelman V, Eschrich SA, Scott JG, Torres-Roca J, Latifi K, Parikh N, Costello J. Habitat escalated adaptive therapy (HEAT): a phase 2 trial utilizing radiomic habitat-directed and genomic-adjusted radiation dose (GARD) optimization for high-grade soft tissue sarcoma. BMC Cancer 2024; 24:437. [PMID: 38594603 PMCID: PMC11003059 DOI: 10.1186/s12885-024-12151-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2023] [Accepted: 03/20/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND Soft tissue sarcomas (STS), have significant inter- and intra-tumoral heterogeneity, with poor response to standard neoadjuvant radiotherapy (RT). Achieving a favorable pathologic response (FPR ≥ 95%) from RT is associated with improved patient outcome. Genomic adjusted radiation dose (GARD), a radiation-specific metric that quantifies the expected RT treatment effect as a function of tumor dose and genomics, proposed that STS is significantly underdosed. STS have significant radiomic heterogeneity, where radiomic habitats can delineate regions of intra-tumoral hypoxia and radioresistance. We designed a novel clinical trial, Habitat Escalated Adaptive Therapy (HEAT), utilizing radiomic habitats to identify areas of radioresistance within the tumor and targeting them with GARD-optimized doses, to improve FPR in high-grade STS. METHODS Phase 2 non-randomized single-arm clinical trial includes non-metastatic, resectable high-grade STS patients. Pre-treatment multiparametric MRIs (mpMRI) delineate three distinct intra-tumoral habitats based on apparent diffusion coefficient (ADC) and dynamic contrast enhanced (DCE) sequences. GARD estimates that simultaneous integrated boost (SIB) doses of 70 and 60 Gy in 25 fractions to the highest and intermediate radioresistant habitats, while the remaining volume receives standard 50 Gy, would lead to a > 3 fold FPR increase to 24%. Pre-treatment CT guided biopsies of each habitat along with clip placement will be performed for pathologic evaluation, future genomic studies, and response assessment. An mpMRI taken between weeks two and three of treatment will be used for biological plan adaptation to account for tumor response, in addition to an mpMRI after the completion of radiotherapy in addition to pathologic response, toxicity, radiomic response, disease control, and survival will be evaluated as secondary endpoints. Furthermore, liquid biopsy will be performed with mpMRI for future ancillary studies. DISCUSSION This is the first clinical trial to test a novel genomic-based RT dose optimization (GARD) and to utilize radiomic habitats to identify and target radioresistance regions, as a strategy to improve the outcome of RT-treated STS patients. Its success could usher in a new phase in radiation oncology, integrating genomic and radiomic insights into clinical practice and trial designs, and may reveal new radiomic and genomic biomarkers, refining personalized treatment strategies for STS. TRIAL REGISTRATION NCT05301283. TRIAL STATUS The trial started recruitment on March 17, 2022.
Collapse
Affiliation(s)
- Arash O Naghavi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA.
| | - J M Bryant
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Youngchul Kim
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Joseph Weygand
- Department of Radiation Oncology and Applied Sciences, Geisel School of Medicine, Dartmouth College, Hanover, NH, USA
| | - Gage Redler
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Austin J Sim
- Department of Radiation Oncology, James Cancer Hospital, The Ohio State University Comprehensive Cancer Center, Columbus, OH, USA
| | - Justin Miller
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kaitlyn Coucoules
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Lauren Taylor Michael
- Clinical Trials Office, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Warren E Gloria
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - George Yang
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Stephen A Rosenberg
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kamran Ahmed
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Marilyn M Bui
- Department of Pathology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | | | - Andrew Lee
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Caitlin D Lee
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ricardo J Gonzalez
- Department of Sarcoma, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Vladimir Feygelman
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Steven A Eschrich
- Department of Bioinformatics and Biostatistics, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Jacob G Scott
- Translational Hematology and Oncology Research, Radiation Oncology Department, Cleveland Clinic, Cleveland, OH, USA
| | - Javier Torres-Roca
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Kujtim Latifi
- Department of Radiation Oncology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Nainesh Parikh
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - James Costello
- Department of Diagnostic Imaging and Interventional Radiology, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| |
Collapse
|
37
|
Corrias G, Lai E, Ziranu P, Mariani S, Donisi C, Liscia N, Saba G, Pretta A, Persano M, Fanni D, Spanu D, Balconi F, Loi F, Deidda S, Restivo A, Pusceddu V, Puzzoni M, Solinas C, Massa E, Madeddu C, Gerosa C, Zorcolo L, Faa G, Saba L, Scartozzi M. Prediction of Response to Anti-Angiogenic Treatment for Advanced Colorectal Cancer Patients: From Biological Factors to Functional Imaging. Cancers (Basel) 2024; 16:1364. [PMID: 38611042 PMCID: PMC11011199 DOI: 10.3390/cancers16071364] [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: 02/25/2024] [Revised: 03/24/2024] [Accepted: 03/27/2024] [Indexed: 04/14/2024] Open
Abstract
Colorectal cancer (CRC) is a leading tumor worldwide. In CRC, the angiogenic pathway plays a crucial role in cancer development and the process of metastasis. Thus, anti-angiogenic drugs represent a milestone for metastatic CRC (mCRC) treatment and lead to significant improvement of clinical outcomes. Nevertheless, not all patients respond to treatment and some develop resistance. Therefore, the identification of predictive factors able to predict response to angiogenesis pathway blockade is required in order to identify the best candidates to receive these agents. Unfortunately, no predictive biomarkers have been prospectively validated to date. Over the years, research has focused on biologic factors such as genetic polymorphisms, circulating biomarkers, circulating tumor cells (CTCs), circulating tumor DNA (ctDNA), and microRNA. Moreover, research efforts have evaluated the potential correlation of molecular biomarkers with imaging techniques used for tumor assessment as well as the application of imaging tools in clinical practice. In addition to functional imaging, radiomics, a relatively newer technique, shows real promise in the setting of correlating molecular medicine to radiological phenotypes.
Collapse
Affiliation(s)
- Giuseppe Corrias
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Eleonora Lai
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Pina Ziranu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Stefano Mariani
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Donisi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Nicole Liscia
- Department of Medical Oncology, IRCCS San Raffaele Scientific Institute, Vita-Salute San Raffaele University, 20132 Milan, Italy;
| | - Giorgio Saba
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Andrea Pretta
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Mara Persano
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Daniela Fanni
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Dario Spanu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesca Balconi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Francesco Loi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Simona Deidda
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Angelo Restivo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Valeria Pusceddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Marco Puzzoni
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Cinzia Solinas
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Elena Massa
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clelia Madeddu
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| | - Clara Gerosa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luigi Zorcolo
- Colorectal Surgery Unit, A.O.U. Cagliari, Department of Surgical Science, University of Cagliari, 09042 Cagliari, Italy; (S.D.); (A.R.); (L.Z.)
| | - Gavino Faa
- Division of Pathology, Department of Medical Sciences and Public Health, AOU Cagliari, University of Cagliari, 09124 Cagliari, Italy; (D.F.); (C.G.); (G.F.)
| | - Luca Saba
- Department of Radiology, University of Cagliari, 09042 Cagliari, Italy;
| | - Mario Scartozzi
- Medical Oncology Unit, University Hospital and University of Cagliari, 09042 Cagliari, Italy; (E.L.); (P.Z.); (S.M.); (C.D.); (G.S.); (A.P.); (M.P.); (D.S.); (F.B.); (F.L.); (V.P.); (M.P.); (C.S.); (E.M.); (C.M.); (M.S.)
| |
Collapse
|
38
|
Liu L, Zhao L, Jing Y, Li D, Linghu H, Wang H, Zhou L, Fang Y, Li Y. Exploring a multiparameter MRI-based radiomics approach to predict tumor proliferation status of serous ovarian carcinoma. Insights Imaging 2024; 15:74. [PMID: 38499907 PMCID: PMC10948697 DOI: 10.1186/s13244-024-01634-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2023] [Accepted: 01/27/2024] [Indexed: 03/20/2024] Open
Abstract
OBJECTIVES To develop a multiparameter magnetic resonance imaging (MRI)-based radiomics approach that can accurately predict the tumor cell proliferation status of serous ovarian carcinoma (SOC). MATERIALS AND METHODS A total of 134 patients with SOC who met the inclusion and exclusion criteria were retrospectively screened from institution A, spanning from January 2016 to March 2022. Additionally, an external validation set comprising 42 SOC patients from institution B was also included. The region of interest was determined by drawing each ovarian mass boundaries manually slice-by-slice on T2-weighted imaging fat-suppressed fast spin-echo (T2FSE) and T1 with contrast enhancement (T1CE) images using ITK-SNAP software. The handcrafted radiomic features were extracted, and then were selected using variance threshold algorithm, SelectKBest algorithm, and least absolute shrinkage and selection operator. The optimal radiomic scores and the clinical/radiological independent predictors were integrated as a combined model. RESULTS Compared with the area under the curve (AUC) values of each radiomic signature of T2FSE and T1CE, respectively, the AUC value of the radiomic signature (T1CE-T2FSE) was the highest in the training set (0.999 vs. 0.965 and 0.860). The homogeneous solid component of the ovarian mass was considered the only independent predictor of tumor cell proliferation status among the clinical/radiological variables. The AUC of the radiomic-radiological model was 0.999. CONCLUSIONS The radiomic-radiological model combining radiomic scores and the homogeneous solid component of the ovarian mass can accurately predict tumor cell proliferation status of SOC which has high repeatability and may enable more targeted and effective treatment strategies. CRITICAL RELEVANCE STATEMENT The proposed radiomic-radiological model combining radiomic scores and the homogeneous solid component of the ovarian mass can predict tumor cell proliferation status of SOC which has high repeatability and may guide individualized treatment programs. KEY POINTS • The radiomic-radiological nomogram may guide individualized treatment programs of SOC. • This radiomic-radiological nomogram showed a favorable prediction ability. • Homogeneous slightly higher signal intensity on T2FSE is vital for Ki-67.
Collapse
Affiliation(s)
- Li Liu
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, Yuanjiagang, China
| | - Ling Zhao
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
| | - Yang Jing
- Huiying Medical Technology Co., Ltd, Dongsheng Science and Technology Park, Room A206, B2Haidian District, Beijing, 100192, China
| | - Dan Li
- Department of Pathology, Chongqing Medical University, No.1 Medical College Road, Yuzhong District, Chongqing, 400016, China
| | - Hua Linghu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road Yuzhong District, Chongqing, 400016, Yuanjiagang, China
| | - Haiyan Wang
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China
| | - Linyi Zhou
- Department of Radiology, Army Medical Center, Daping Hospital, Army Medical University, 10# Changjiangzhilu, Chongqing, 40024, China
| | - Yuan Fang
- Department of Radiology, The People's Hospital of Yubei District of Chongqing City, No. 23 ZhongyangGongyuanBei Road, Yubei District, Chongqing, 401120, China.
| | - Yongmei Li
- Department of Radiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Road, Yuzhong District, Chongqing, 400016, Yuanjiagang, China.
| |
Collapse
|
39
|
Mao B, Ren Y, Yu X, Liang X, Ding X. Preoperative prediction for early recurrence of hepatocellular carcinoma using machine learning-based radiomics. Front Oncol 2024; 14:1346124. [PMID: 38559563 PMCID: PMC10978579 DOI: 10.3389/fonc.2024.1346124] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2023] [Accepted: 03/04/2024] [Indexed: 04/04/2024] Open
Abstract
Objective To develop a contrast-enhanced computed tomography (CECT) based radiomics model using machine learning method and assess its ability of preoperative prediction for the early recurrence of hepatocellular carcinoma (HCC). Methods A total of 297 patients confirmed with HCC were assigned to the training dataset and test dataset based on the 8:2 ratio, and the follow-up period of the patients was from May 2012 to July 2017. The lesion sites were manually segmented using ITK-SNAP, and the pyradiomics platform was applied to extract radiomic features. We established the machine learning model to predict the early recurrence of HCC. The accuracy, AUC, standard deviation, specificity, and sensitivity were applied to evaluate the model performance. Results 1,688 features were extracted from the arterial phase and venous phase images, respectively. When arterial phase and venous phase images were employed correlated with clinical factors to train a prediction model, it achieved the best performance (AUC with 95% CI 0.8300(0.7560-0.9040), sensitivity 89.45%, specificity 79.07%, accuracy 82.67%, p value 0.0064). Conclusion The CECT-based radiomics may be helpful to non-invasively reveal the potential connection between CECT images and early recurrence of HCC. The combination of radiomics and clinical factors could boost model performance.
Collapse
Affiliation(s)
- Bing Mao
- Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Yajun Ren
- Department of Gastroenterology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Xuan Yu
- Department of Medical Imaging, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, China
| | - Xinliang Liang
- Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital; Henan University People’s Hospital, Zhengzhou, Henan, China
| | - Xiangming Ding
- Department of Gastroenterology, Henan Provincial People’s Hospital, People’s Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
40
|
Xia T, Zhao B, Li B, Lei Y, Song Y, Wang Y, Tang T, Ju S. MRI-Based Radiomics and Deep Learning in Biological Characteristics and Prognosis of Hepatocellular Carcinoma: Opportunities and Challenges. J Magn Reson Imaging 2024; 59:767-783. [PMID: 37647155 DOI: 10.1002/jmri.28982] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 08/15/2023] [Accepted: 08/16/2023] [Indexed: 09/01/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the fifth most common malignancy and the third leading cause of cancer-related death worldwide. HCC exhibits strong inter-tumor heterogeneity, with different biological characteristics closely associated with prognosis. In addition, patients with HCC often distribute at different stages and require diverse treatment options at each stage. Due to the variability in tumor sensitivity to different therapies, determining the optimal treatment approach can be challenging for clinicians prior to treatment. Artificial intelligence (AI) technology, including radiomics and deep learning approaches, has emerged as a unique opportunity to improve the spectrum of HCC clinical care by predicting biological characteristics and prognosis in the medical imaging field. The radiomics approach utilizes handcrafted features derived from specific mathematical formulas to construct various machine-learning models for medical applications. In terms of the deep learning approach, convolutional neural network models are developed to achieve high classification performance based on automatic feature extraction from images. Magnetic resonance imaging offers the advantage of superior tissue resolution and functional information. This comprehensive evaluation plays a vital role in the accurate assessment and effective treatment planning for HCC patients. Recent studies have applied radiomics and deep learning approaches to develop AI-enabled models to improve accuracy in predicting biological characteristics and prognosis, such as microvascular invasion and tumor recurrence. Although AI-enabled models have demonstrated promising potential in HCC with biological characteristics and prognosis prediction with high performance, one of the biggest challenges, interpretability, has hindered their implementation in clinical practice. In the future, continued research is needed to improve the interpretability of AI-enabled models, including aspects such as domain knowledge, novel algorithms, and multi-dimension data sources. Overcoming these challenges would allow AI-enabled models to significantly impact the care provided to HCC patients, ultimately leading to their deployment for clinical use. LEVEL OF EVIDENCE: 5 TECHNICAL EFFICACY: Stage 2.
Collapse
Affiliation(s)
- Tianyi Xia
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ben Zhao
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Binrong Li
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Ying Lei
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Yang Song
- MR Scientific Marketing, Siemens Healthineers Ltd., Shanghai, China
| | - Yuancheng Wang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Tianyu Tang
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| | - Shenghong Ju
- Jiangsu Key Laboratory of Molecular and Functional Imaging, Department of Radiology, Zhongda Hospital, School of Medicine, Southeast University, Nanjing, China
| |
Collapse
|
41
|
Sheng L, Wei H, Yang T, Yang J, Zhang L, Zhu X, Jiang H, Song B. Extracellular contrast agent-enhanced MRI is as effective as gadoxetate disodium-enhanced MRI for predicting microvascular invasion in HCC. Eur J Radiol 2024; 170:111200. [PMID: 37995512 DOI: 10.1016/j.ejrad.2023.111200] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 08/31/2023] [Accepted: 11/13/2023] [Indexed: 11/25/2023]
Abstract
PURPOSE To compare the performances of gadoxetate disodium-enhanced MRI (EOB-MRI) and extracellular contrast agent-enhanced MRI (ECA-MRI) for predicting microvascular invasion (MVI) in HCC. MATERIALS AND METHODS From November 2009 to December 2021, consecutive HCC patients who underwent preoperative contrast-enhanced MRI were retrospectively enrolled into either an ECA-MRI or EOB-MRI cohort. In the ECA-MRI cohort, a preoperative MVI score was constructed in the training dataset using a logistic regression model that evaluated pathological type. In a propensity score-matched testing dataset of the ECA-MRI cohort, the MVI score was validated and compared with a previously proposed EOB-MRI-based MVI score calculated in the EOB-MRI cohort. Time-to-early recurrence survival was evaluated by the Kaplan-Meier method with the log-rank test. RESULTS A total of 536 patients were included (478 men; 53 years, interquartile range, 46-62 years), 322 (60.1 %) with pathologically confirmed MVI. Based on the training dataset, independent variables associated with MVI included serum alpha-fetoprotein > 400 ng/ml (odds ratio [OR] = 2.3), infiltrative appearance (OR = 4.9), internal artery (OR = 2.5) and nodule-in-nodule architecture (OR = 2.4), which were incorporated into the ECA-MRI-based MVI score. The testing dataset AUC of the ECA-MRI score was 0.720, which was comparable to that of the EOB-MRI-based MVI score (AUC = 0.721; P =.99). Patients from either the ECA-MRI or the EOB-MRI cohort with model-predicted MVI had significantly shorter time-to-early recurrence than those without MVI (P <.001). CONCLUSION Based on the preoperative serum alpha-fetoprotein and three MRI features, ECA-MRI demonstrated comparable performance to EOB-MRI for predicting MVI in HCC.
Collapse
Affiliation(s)
- Liuji Sheng
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hong Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ting Yang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jie Yang
- Department of Ultrasound, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Lin Zhang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xiaomei Zhu
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Hanyu Jiang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China.
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan, China; Department of Radiology, Sanya People's Hospital, Sanya, Hainan, China.
| |
Collapse
|
42
|
Li YX, Li WJ, Xu YS, Jia LL, Wang MM, Qu MM, Wang LL, Lu XD, Lei JQ. Clinical application of dual-layer spectral CT multi-parameter feature to predict microvascular invasion in hepatocellular carcinoma. Clin Hemorheol Microcirc 2024; 88:97-113. [PMID: 38848171 DOI: 10.3233/ch-242175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/09/2024]
Abstract
OBJECTIVE This study aimed to investigate the feasibility of using dual-layer spectral CT multi-parameter feature to predict microvascular invasion of hepatocellular carcinoma. METHODS This retrospective study enrolled 50 HCC patients who underwent multiphase contrast-enhanced spectral CT studies preoperatively. Combined clinical data, radiological features with spectral CT quantitative parameter were constructed to predict MVI. ROC was applied to identify potential predictors of MVI. The CT values obtained by simulating the conventional CT scans with 70 keV images were compared with those obtained with 40 keV images. RESULTS 50 hepatocellular carcinomas were detected with 30 lesions (Group A) with microvascular invasion and 20 (Group B) without. There were significant differences in AFP,tumer size, IC, NIC,slope and effective atomic number in AP and ICrr in VP between Group A ((1000(10.875,1000),4.360±0.3105, 1.7750 (1.5350,1.8825) mg/ml, 0.1785 (0.1621,0.2124), 2.0362±0.2108,8.0960±0.1043,0.2830±0.0777) and Group B (4.750(3.325,20.425),3.190±0.2979,1.4700 (1.4500,1.5775) mg/ml, 0.1441 (0.1373,0.1490),1.8601±0.1595, 7.8105±0.7830 and 0.2228±0.0612) (all p < 0.05). Using 0.1586 as the threshold for NIC, one could obtain an area-under-curve (AUC) of 0.875 in ROC to differentiate between tumours with and without microvascular invasion. AUC was 0.625 with CT value at 70 keV and improved to 0.843 at 40 keV. CONCLUSION Dual-layer spectral CT provides additional quantitative parameters than conventional CT to enhance the differentiation between hepatocellular carcinoma with and without microvascular invasion. Especially, the normalized iodine concentration (NIC) in arterial phase has the greatest potential application value in determining whether microvascular invasion exists, and can offer an important reference for clinical treatment plan and prognosis assessment.
Collapse
Affiliation(s)
- Yi-Xiang Li
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Wen-Jing Li
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Yong-Sheng Xu
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Lu-Lu Jia
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Miao-Miao Wang
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Meng-Meng Qu
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Li-Li Wang
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Xian-de Lu
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| | - Jun-Qiang Lei
- The First Clinical Medical of Lanzhou University, Lanzhou, China
- Department of Radiology, The First Hospital of Lanzhou University, Lanzhou, China
- Gansu Intelligent Imaging Medical Engineering Research Center, Lanzhou, China
- Precision Image Collaborative Innovation Gansu International Science and Technology Cooperation Base, Lanzhou, China
| |
Collapse
|
43
|
Gu J, Bao S, Akemuhan R, Jia Z, Zhang Y, Huang C. Radiomics Based on Contrast-Enhanced CT for Recognizing c-Met-Positive Hepatocellular Carcinoma: a Noninvasive Approach to Predict the Outcome of Sorafenib Resistance. Mol Imaging Biol 2023; 25:1073-1083. [PMID: 37932610 DOI: 10.1007/s11307-023-01870-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 10/16/2023] [Accepted: 10/23/2023] [Indexed: 11/08/2023]
Abstract
OBJECTIVES The purpose of our project was to investigate the effectiveness of radiomic features based on contrast-enhanced computed tomography (CT) that can detect the expression of c-Met in hepatocellular carcinoma (HCC) and to validate its efficacy in predicting the outcome of sorafenib resistance. MATERIALS AND METHODS In total, 130 patients (median age, 60 years) with pathologically confirmed HCC who underwent contrast material-enhanced CT from October 2012 to July 2020 were randomly divided into a training set (n = 91) and a test set (n = 39). Radiomic features were extracted from arterial phase (AP), portal venous phase (VP) and delayed phase (DP) images of every participant's enhanced CT images. RESULTS The entire group comprised 39 Met-positive and 91 Met-negative patients. The combined model, which included the clinical factors and the radiomic features, performed well in the training (area under the curve [AUC] = 0.878) and validation (AUC = 0.851) cohorts. The nomogram, which relied on the combined model, fits well in the calibration curves. Decision curve analysis (DCA) further confirmed that the clinical valuation of the nomogram achieved comparable accuracy in c-Met prediction. Among another 20 patients with HCC who had received sorafenib, the predicted high-risk group had shorter overall survival (OS) than the predicted low-risk group (p < 0.05). CONCLUSION A multivariate model acquired from three phases (AP, VP and DP) of enhanced CT, HBV-DNA and γ glutamyl transpeptidase isoenzyme II (GGT-II) could be considered a satisfactory preoperative marker of the expression of c-Met in patients with HCC. This approach may help in overcoming sorafenib resistance in advanced HCC.
Collapse
Affiliation(s)
- Jingxiao Gu
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, the, People's Republic of China
- Department of Radiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China
| | - Shanlei Bao
- Department of Nuclear Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | | | - Zhongzheng Jia
- Department of Radiology, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, China.
| | - Yu Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Nantong University, Nantong, China.
| | - Chen Huang
- Department of Vascular Surgery, Affiliated Hospital of Nantong University, Medical School of Nantong University, Nantong, 226001, the, People's Republic of China.
| |
Collapse
|
44
|
Mendes Serrão E, Klug M, Moloney BM, Jhaveri A, Lo Gullo R, Pinker K, Luker G, Haider MA, Shinagare AB, Liu X. Current Status of Cancer Genomics and Imaging Phenotypes: What Radiologists Need to Know. Radiol Imaging Cancer 2023; 5:e220153. [PMID: 37921555 DOI: 10.1148/rycan.220153] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2023]
Abstract
Ongoing discoveries in cancer genomics and epigenomics have revolutionized clinical oncology and precision health care. This knowledge provides unprecedented insights into tumor biology and heterogeneity within a single tumor, among primary and metastatic lesions, and among patients with the same histologic type of cancer. Large-scale genomic sequencing studies also sparked the development of new tumor classifications, biomarkers, and targeted therapies. Because of the central role of imaging in cancer diagnosis and therapy, radiologists need to be familiar with the basic concepts of genomics, which are now becoming the new norm in oncologic clinical practice. By incorporating these concepts into clinical practice, radiologists can make their imaging interpretations more meaningful and specific, facilitate multidisciplinary clinical dialogue and interventions, and provide better patient-centric care. This review article highlights basic concepts of genomics and epigenomics, reviews the most common genetic alterations in cancer, and discusses the implications of these concepts on imaging by organ system in a case-based manner. This information will help stimulate new innovations in imaging research, accelerate the development and validation of new imaging biomarkers, and motivate efforts to bring new molecular and functional imaging methods to clinical radiology. Keywords: Oncology, Cancer Genomics, Epignomics, Radiogenomics, Imaging Markers Supplemental material is available for this article. © RSNA, 2023.
Collapse
Affiliation(s)
- Eva Mendes Serrão
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Maximiliano Klug
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Brian M Moloney
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Aaditeya Jhaveri
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Roberto Lo Gullo
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Katja Pinker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Gary Luker
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Masoom A Haider
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Atul B Shinagare
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| | - Xiaoyang Liu
- From the Joint Department of Medical Imaging, University Medical Imaging Toronto, University Health Network, University of Toronto, 585 University Ave, Toronto, ON, Canada M5G 2N2 (E.M.S., A.J., M.A.H., X.L.); Division of Diagnostic Imaging, Sheba Medical Center, Tel Aviv University, Tel Aviv, Israel (M.K.); Department of Radiology, The Christie NHS Trust, Manchester, England (B.M.M.); Department of Radiology, Breast Imaging Service, Memorial Sloan-Kettering Cancer Center, Weill Medical College of Cornell University, New York, NY (R.L.G., K.P.); Center for Molecular Imaging, Department of Radiology, University of Michigan, Ann Arbor, Mich (G.L.); Lunenfeld Tanenbaum Research Institute, Sinai Health System, Mount Sinai Hospital, Toronto, Ontario, Canada (M.A.H.); and Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Mass (A.B.S.)
| |
Collapse
|
45
|
Tan XZ, Liu P, Xiang H. The Influence of Single Arterial Phase Imaging on Prediction of Microvascular Invasion of Hepatocellular Carcinoma. Radiology 2023; 309:e231141. [PMID: 37962505 DOI: 10.1148/radiol.231141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2023]
Affiliation(s)
- Xian-Zheng Tan
- Department of Radiology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Peng Liu
- Department of Radiology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, Changsha, China
| | - Hua Xiang
- Department of Interventional Radiology, Hunan Provincial People's Hospital, the First Affiliated Hospital of Hunan Normal University, No. 61 Jiefang West Road, Changsha 410005, Hunan, China
| |
Collapse
|
46
|
Ding Z, Wu Y, Fang G, Lin Z, Lin K, Fu J, Huang Q, Tang Y, You W, Liu J, Zeng Y. Development and validation a radiomics nomogram for predicting thymidylate synthase status in hepatocellular carcinoma based on Gd-DTPA contrast enhanced MRI. BMC Cancer 2023; 23:991. [PMID: 37848807 PMCID: PMC10580573 DOI: 10.1186/s12885-023-11096-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 06/21/2023] [Indexed: 10/19/2023] Open
Abstract
OBJECTIVES The purpose of this study was to develop and validate a radiomics nomogram for predicting thymidylate synthase (TYMS) status in hepatocellular carcinoma (HCC) by using Gd-DTPA contrast enhanced MRI. METHODS We retrospectively enrolled 147 consecutive patients with surgically confirmed HCC and randomly allocated to training and validation set (7:3). The TYMS status was immunohistochemical determined and classified into low TYMS (positive cells ≤ 25%) and high TYMS (positive cells > 25%) groups. Radiomics features were extracted from the arterial phases and portal venous phase of Gd-DTPA contrast enhanced MRI. Least absolute shrinkage and selection operator (LASSO) were applied for generating the Rad score. Clinical data and MRI findings were assessed to build a clinical model. Rad score combined with clinical features was used to construct radiomics nomogram. RESULTS A total of 2260 features were extracted and reduced to 7 features as the most important discriminators to build the Rad score. InAFP was identified as the only independent clinical factors for TYMS status. The radiomics nomogram showed good discrimination in training (AUC, 0.759; 95% CI 0.665-0.838) and validation set (AUC, 0.739; 95% CI 0.585-0.860), and showed better discrimination capability (P < 0.05) compared with clinical model in training (AUC, 0.656; 95% CI 0.555-0.746) and validation set (AUC, 0.622; 95% CI 0.463-0.764). CONCLUSIONS The radiomics nomogram shows favorable predictive efficacy for TYMS status in HCC, which might be helpful for the personalized treatment of HCC.
Collapse
Affiliation(s)
- Zongren Ding
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| | - Yijun Wu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| | - Guoxu Fang
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| | - Zhaowang Lin
- Department of Radiology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025 China
| | - Kongying Lin
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| | - Jun Fu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| | - Qizhen Huang
- Department of Radiotherapy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025 China
| | - Yanyan Tang
- Department of Radiology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025 China
| | - Wuyi You
- Department of Radiology, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, 350025 China
| | - Jingfeng Liu
- Fujian Provincial Cancer Hospital, Fuzhou, 350025 China
| | - Yongyi Zeng
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Xihong Road 312, Fuzhou, 350025 China
| |
Collapse
|
47
|
Wang D, Zhang L, Sun Z, Jiang H, Zhang J. A radiomics signature associated with underlying gene expression pattern for the prediction of prognosis and treatment response in hepatocellular carcinoma. Eur J Radiol 2023; 167:111086. [PMID: 37708675 DOI: 10.1016/j.ejrad.2023.111086] [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: 06/12/2023] [Revised: 08/13/2023] [Accepted: 09/04/2023] [Indexed: 09/16/2023]
Abstract
PURPOSE Identifying robust prognosis and treatment efficiency predictive biomarkers of hepatocellular carcinoma (HCC) is challenging. The purpose of this study is to develop a radiomics approach for predicting the overall survival (OS) based on pretreatment CT images and to explore the radiomic-associated key genes. METHODS Patients with pathologically or clinically proven HCC from three data sets were retrospectively included in this study. The institute internal data that received transarterial chemoembolization (TACE) treatment was used as the training set to construct the radiomics signature to predict OS by the least absolute shrinkage and selection operator COX (LASSO-COX) regression algorithms. The model was externally tested in 41 patients from The Cancer Genome Atlas (TCGA) with available CT images. Area under the receiver operating characteristics curve (AUC) and the log-rank test were used for survival analysis based on high versus low radiomics score. RNA sequencing data of TCGA and Gene Expression Omnibus (GEO) public database were used for gene expression analysis. RESULTS A total of 752 patients were divided into the Radiomics cohort (n = 267), the TCGA cohort (n = 338) and GEO cohort (n = 147). The rad-score divided patients into high and low risk groups, with significant survival differences (P < 0.0001 and P = 0.0055) in the training and external test set. The AUC for 5 years' OS were 0.730 and 0.695, respectively. Seven OS-related genes (SPP1, GJA5, GJA4, INMT, PDZD4, ALDOA and MAFG) were identified, all of which were related with TACE efficiency, except for MAFG (P greater than 0.05). CONCLUSIONS CT-radiomics signature could effectively predict the prognosis and treatment response of HCC, which were also associated with the tumor microenvironment heterogeneity.
Collapse
Affiliation(s)
- Dandan Wang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Linhan Zhang
- Department of PET/CT, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zhongqi Sun
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Huijie Jiang
- Department of Radiology, The Second Affiliated Hospital of Harbin Medical University, Harbin, China.
| | - Jinfeng Zhang
- Department of Breast Surgery, Harbin Medical University Cancer Hospital, Harbin, China.
| |
Collapse
|
48
|
Zhao YM, Xie SS, Wang J, Zhang YM, Li WC, Ye ZX, Shen W. Added value of CE-CT radiomics to predict high Ki-67 expression in hepatocellular carcinoma. BMC Med Imaging 2023; 23:138. [PMID: 37737166 PMCID: PMC10514983 DOI: 10.1186/s12880-023-01069-4] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2022] [Accepted: 08/02/2023] [Indexed: 09/23/2023] Open
Abstract
BACKGROUND This study aimed to develop a computed tomography (CT) model to predict Ki-67 expression in hepatocellular carcinoma (HCC) and to examine the added value of radiomics to clinico-radiological features. METHODS A total of 208 patients (training set, n = 120; internal test set, n = 51; external validation set, n = 37) with pathologically confirmed HCC who underwent contrast-enhanced CT (CE-CT) within 1 month before surgery were retrospectively included from January 2014 to September 2021. Radiomics features were extracted and selected from three phases of CE-CT images, least absolute shrinkage and selection operator regression (LASSO) was used to select features, and the rad-score was calculated. CE-CT imaging and clinical features were selected using univariate and multivariate analyses, respectively. Three prediction models, including clinic-radiologic (CR) model, rad-score (R) model, and clinic-radiologic-radiomic (CRR) model, were developed and validated using logistic regression analysis. The performance of different models for predicting Ki-67 expression was evaluated using the area under the receiver operating characteristic curve (AUROC) and decision curve analysis (DCA). RESULTS HCCs with high Ki-67 expression were more likely to have high serum α-fetoprotein levels (P = 0.041, odds ratio [OR] 2.54, 95% confidence interval [CI]: 1.04-6.21), non-rim arterial phase hyperenhancement (P = 0.001, OR 15.13, 95% CI 2.87-79.76), portal vein tumor thrombus (P = 0.035, OR 3.19, 95% CI: 1.08-9.37), and two-trait predictor of venous invasion (P = 0.026, OR 14.04, 95% CI: 1.39-144.32). The CR model achieved relatively good and stable performance compared with the R model (AUC, 0.805 [95% CI: 0.683-0.926] vs. 0.678 [95% CI: 0.536-0.839], P = 0.211; and 0.805 [95% CI: 0.657-0.953] vs. 0.667 [95% CI: 0.495-0.839], P = 0.135) in the internal and external validation sets. After combining the CR model with the R model, the AUC of the CRR model increased to 0.903 (95% CI: 0.849-0.956) in the training set, which was significantly higher than that of the CR model (P = 0.0148). However, no significant differences were found between the CRR and CR models in the internal and external validation sets (P = 0.264 and P = 0.084, respectively). CONCLUSIONS Preoperative models based on clinical and CE-CT imaging features can be used to predict HCC with high Ki-67 expression accurately. However, radiomics cannot provide added value.
Collapse
Affiliation(s)
- Yu-meng Zhao
- Medical School of Nankai University, No. 94, Weijin Road, Nankai District, Tianjin, China
| | - Shuang-shuang Xie
- Department of Radiology, Tianjin First Center Hospital, Tianjin Institute of imaging medicine, School of Medicine, Nankai University, Nankai District, No. 24 Fukang Road, Tianjin, China
| | - Jian Wang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Nankai District, No. 24 Fukang Road, Tianjin, China
| | - Ya-min Zhang
- Department of Hepatobiliary Surgery, Tianjin First Central Hospital, School of Medicine, Nankai University, Nankai District, No. 24 Fukang Road, Tianjin, China
| | - Wen-Cui Li
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Zhao-Xiang Ye
- Department of Radiology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin, 300060 China
| | - Wen Shen
- Department of Radiology, Tianjin First Center Hospital, Tianjin Institute of imaging medicine, School of Medicine, Nankai University, Nankai District, No. 24 Fukang Road, Tianjin, China
| |
Collapse
|
49
|
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
|
50
|
Jin N, Qiao B, Zhao M, Li L, Zhu L, Zang X, Gu B, Zhang H. Predicting cervical lymph node metastasis in OSCC based on computed tomography imaging genomics. Cancer Med 2023; 12:19260-19271. [PMID: 37635388 PMCID: PMC10557859 DOI: 10.1002/cam4.6474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/01/2023] [Accepted: 08/15/2023] [Indexed: 08/29/2023] Open
Abstract
BACKGROUND To investigate the correlation between computed tomography (CT) radiomic characteristics and key genes for cervical lymph node metastasis (LNM) in oral squamous cell carcinoma (OSCC). METHODS The region of interest was annotated at the edge of the primary tumor on enhanced CT images from 140 patients with OSCC and obtained radiomic features. Ribonucleic acid (RNA) sequencing was performed on pathological sections from 20 patients. the DESeq software package was used to compare differential gene expression between groups. Weighted gene co-expression network analysis was used to construct co-expressed gene modules, and the KEGG and GO databases were used for pathway enrichment analysis of key gene modules. Finally, Pearson correlation coefficients were calculated between key genes of enriched pathways and radiomic features. RESULTS Four hundred and eighty radiomic features were extracted from enhanced CT images of 140 patients; seven of these correlated significantly with cervical LNM in OSCC (p < 0.01). A total of 3527 differentially expressed RNAs were screened from RNA sequencing data of 20 cases. original_glrlm_RunVariance showed significant positive correlation with most long noncoding RNAs. CONCLUSIONS OSCC cervical LNM is related to the salivary hair bump signaling pathway and biological process. Original_glrlm_RunVariance correlated with LNM and most differentially expressed long noncoding RNAs.
Collapse
Affiliation(s)
- Nenghao Jin
- Medical School of Chinese PLABeijingChina
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Bo Qiao
- Medical School of Chinese PLABeijingChina
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Min Zhao
- Pharmaceutical Diagnostics, GE HealthcareBeijingChina
- Research Center of Medical Big Data, Chinese PLA General HospitalBeijingChina
| | - Liangbo Li
- Medical School of Chinese PLABeijingChina
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Liang Zhu
- Medical School of Chinese PLABeijingChina
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Xiaoyi Zang
- Medical School of Chinese PLABeijingChina
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Bin Gu
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| | - Haizhong Zhang
- Department of Stomatology, The First Medical CentreChinese PLA General HospitalBeijingChina
| |
Collapse
|