1
|
Li Y, Li X, Xiao X, Cheng J, Li Q, Liu C, Cai P, Chen W, Zhang H, Li X. A novel hybrid model for predicting tertiary lymphoid structures and targeted immunotherapy outcomes in hepatocellular carcinoma: a multicenter retrospective study. Eur Radiol 2025; 35:3206-3222. [PMID: 39658681 DOI: 10.1007/s00330-024-11255-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 09/29/2024] [Accepted: 11/24/2024] [Indexed: 12/12/2024]
Abstract
OBJECTIVE To develop a novel hybrid model for preoperative prediction of tertiary lymphoid structures (TLSs) of hepatocellular carcinoma (HCC), and to identify patients who may benefit from postoperative targeted immunotherapy. METHODS Retrospective data were gathered from 332 patients with HCC who underwent surgical resection and gadoxetate disodium (Gd-EOB-DTPA) enhanced MRI at two tertiary hospitals (training cohort, n = 205; internal validation cohort, n = 90; and external validation cohort, n = 37) between March 2020 and January 2023. Radiomic features were extracted from Gd-EOB-DTPA-enhanced MRI sequences. These signatures were integrated with clinical-radiologic (CR) factors into a hybrid model and nomogram for clinical application. The performance of the model was assessed using the area under the curve (AUC) and 95% confidence intervals (CI). RESULTS The hybrid model outperformed the radiomics and CR models in the training cohort (AUC = 0.860 [95% CI: 0.805, 0.904], 0.784 [95% CI: 0.721, 0.838], and 0.809 [95% CI: 0.748, 0.860]). The hybrid model showed optimal performance, with AUCs of 0.823 (95% CI: 0.728, 0.895) and 0.875 (95% CI: 0.725, 0.960) in the internal and external validation cohorts, respectively. The calibration curve demonstrated that the nomogram had good diagnostic ability, and decision curve analysis indicated good clinical utility across all cohorts. Importantly, patients with a predicted high risk of TLSs from the hybrid model gained a survival benefit from targeted immunotherapy. CONCLUSION The hybrid model showed satisfactory performance in predicting intra-tumoral TLS positivity and targeted immunotherapy benefit in patients with HCC, potentially assisting clinicians in selecting precise individualized therapies. KEY POINTS Question How can accurate preoperative risk stratification of tertiary lymphoid structures positivity HCC be achieved to support targeted immunotherapy decision-making? Findings A hybrid model combining radiomics model and clinical-radiological model may be a reliable marker for predicting tertiary lymphoid structures positivity HCC. Clinical relevance Using this hybrid model may be useful in predicting tertiary lymphoid structures and screening candidate patients for targeted immunotherapy based on multiparametric MRI, which has potential clinical value in guiding clinical decision-making and improving patient outcomes.
Collapse
Affiliation(s)
- Yiman Li
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Xiaofeng Li
- Department of Radiology, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Xixi Xiao
- Department of Oncology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Cheng
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Qingrui Li
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Chen Liu
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China
| | - Ping Cai
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Wei Chen
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| | - Huarong Zhang
- Institute of Pathology and Southwest Cancer Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China.
| | - Xiaoming Li
- 7T Magnetic Resonance Translational Medicine Research Center, Department of Radiology, Southwest Hospital, Army Medical University (Third Military Medical University), Chongqing, China.
| |
Collapse
|
2
|
Zhang X, Zhang X, Luo QK, Fu Q, Liu P, Pan CJ, Liu CJ, Zhang HW, Qin T. Pretreatment radiomic imaging features combined with immunological indicators to predict targeted combination immunotherapy response in advanced hepatocellular carcinoma. World J Clin Oncol 2025; 16:102735. [PMID: 40290677 PMCID: PMC12019258 DOI: 10.5306/wjco.v16.i4.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/16/2024] [Accepted: 01/23/2025] [Indexed: 03/26/2025] Open
Abstract
BACKGROUND Early symptoms of hepatocellular carcinoma (HCC) are not obvious, and more than 70% of which does not receive radical hepatectomy, when first diagnosed. In recent years, molecular-targeted drugs combined with immunotherapy and other therapeutic methods have provided new treatment options for middle and advanced HCC (aHCC). Predicting the effect of targeted combined immunotherapy has become a hot topic in current research. AIM To explore the relationship between nodule enhancement in hepatobiliary phase and the efficacy of combined targeted immunotherapy for aHCC. METHODS Data from 56 patients with aHCC for magnetic resonance imaging with gadolinium-ethoxybenzyl-diethylenetriamine pentaacetic acid were retrospectively collected. Signal intensity of intrahepatic nodules was measured, and the hepatobiliary relative enhancement ratio (RER) was calculated. Progression-free survival (PFS) of patients with high and low reinforcement of HCC nodules was compared. The model was validated using receiver operating characteristic curves. Univariate and multivariate logistic regression and Kaplan-Meier analysis were performed to explore factors influencing the efficacy of targeted immunization and PFS. RESULTS Univariate and multivariate analyses revealed that the RER, neutrophil-to-lymphocyte ratio, platelet-to-lymphocyte ratio, and prognostic nutritional index were significantly associated with the efficacy of tyrosine kinase inhibitors combined with immunotherapy (P < 0.05). The area under the curve of the RER for predicting the efficacy of tyrosine kinase inhibitors combined with anti-programmed death 1 antibody in patients with aHCC was 0.876 (95% confidence interval: 0.781-0.971, P < 0.05), the optimal cutoff value was 0.904, diagnostic sensitivity was 87.5%, and specificity was 79.2%. Kaplan-Meier analysis showed that neutrophil-to-lymphocyte ratio < 5, platelet-to-lymphocyte ratio < 300, prognostic nutritional index < 45, and RER < 0.9 significantly improved PFS. CONCLUSION AHCC nodules enhancement in the hepatobiliary stage was significantly correlated with PFS. Imaging information and immunological indicators had high predictive efficacy for targeted combined immunotherapy and were associated with PFS.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Xu Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Qian-Kun Luo
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Qiang Fu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Pan Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Chang-Jie Pan
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Chuan-Jiang Liu
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Hong-Wei Zhang
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| | - Tao Qin
- Department of Hepato-Biliary-Pancreatic Surgery, Zhengzhou University People’s Hospital & Henan Provincial People’s Hospital, Zhengzhou 450003, Henan Province, China
| |
Collapse
|
3
|
Guo X, Zhao Z, Zhu L, Liu S, Zhou L, Wu F, Fang S, Chen M, Zheng L, Ji J. The evolving landscape of biomarkers for systemic therapy in advanced hepatocellular carcinoma. Biomark Res 2025; 13:60. [PMID: 40221793 PMCID: PMC11993949 DOI: 10.1186/s40364-025-00774-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 03/29/2025] [Indexed: 04/14/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and deadliest cancers. With the approval of multiple first- and second-line agents, especially the combination therapies based on immune checkpoint inhibitor (ICI) regimens, the landscape of systemic therapy for advanced HCC (aHCC) is more diverse than ever before. The efficacy of current systemic therapies shows great heterogeneity in patients with aHCC, thereby identifying biomarkers for response prediction and patient stratification has become an urgent need. The main biomarkers for systemic therapy in hepatocellular carcinoma are derived from peripheral blood, tissues, and imaging. Currently, the understanding of the clinical response to systemic therapy indicates unequivocally that a single biomarker cannot be used to identify patients who are likely to benefit from these treatments. In this review, we provide an integrated landscape of the recent development in molecular targeted therapies and ICIs-based therapies, especially focusing on the role of clinically applicable predictive biomarkers. Additionally, we further highlight the latest advancements in biomarker-driven therapies, including targeted treatments, adoptive cell therapies, and bispecific antibodies.
Collapse
Affiliation(s)
- Xinyu Guo
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Zhongwei Zhao
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingyi Zhu
- The 2nd Clinical Medical College, Zhejiang Chinese Medical University, Hangzhou, 310000, China
| | - Shuang Liu
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Lingling Zhou
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Fazong Wu
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Shiji Fang
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Minjiang Chen
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China
| | - Liyun Zheng
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| | - Jiansong Ji
- Zhejiang Engineering Research Center of Interventional Medicine Engineering and Biotechnology, School of Medicine, Lishui Hospital, Zhejiaing University, Lishui, 323000, China.
- Department of Radiology, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui Central Hospital, Lishui, 323000, China.
- Zhejiang Key Laboratory of Imaging and Interventional Medicine, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui, Zhejiang, 323000, China.
| |
Collapse
|
4
|
Choi J, Gordon A, Eresen A, Zhang Z, Borhani A, Bagci U, Lewandowski R, Kim DH. Current applications of radiomics in the assessment of tumor microenvironment of hepatocellular carcinoma. Abdom Radiol (NY) 2025:10.1007/s00261-025-04916-w. [PMID: 40208284 DOI: 10.1007/s00261-025-04916-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: 11/30/2024] [Revised: 02/10/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025]
Abstract
The tumor microenvironment (TME) of hepatocellular carcinoma (HCC) has garnered significant attention, especially with the rise of immunotherapy as a treatment strategy. Radiomics, an innovative technique, offers valuable insights into the intricate structure of the TME. This review highlights recent advancements in radiomics for analyzing the HCC TME, identifies key areas that warrant further research, and explores comprehensive multi-omics approaches that extend the potential of radiomics to new frontiers.
Collapse
Affiliation(s)
- Junghwa Choi
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Andrew Gordon
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Aydin Eresen
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Zhuoli Zhang
- Department of Radiological Sciences, University of California, Irvine, Irvine, USA
| | - Amir Borhani
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Ulas Bagci
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Robert Lewandowski
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA
| | - Dong-Hyun Kim
- Department of Radiology, Feinberg School of Medicine, Northwestern University, Chicago, 60611, USA.
- Robert H. Lurie Comprehensive Cancer Center, Northwestern University, Chicago, 60611, USA.
| |
Collapse
|
5
|
Xu J, Li J, Wang T, Luo X, Zhu Z, Wang Y, Wang Y, Zhang Z, Song R, Yang LZ, Wang H, Wong STC, Li H. Predicting treatment response and prognosis of immune checkpoint inhibitors-based combination therapy in advanced hepatocellular carcinoma using a longitudinal CT-based radiomics model: a multicenter study. BMC Cancer 2025; 25:602. [PMID: 40181337 PMCID: PMC11967134 DOI: 10.1186/s12885-025-13978-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 03/19/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Identifying effective predictive strategies to assess the response of immune checkpoint inhibitors (ICIs)-based combination therapy in advanced hepatocellular carcinoma (HCC) is crucial. This study presents a new longitudinal CT-based radiomics model to predict treatment response and prognosis in advanced HCC patients undergoing ICIs-based combination therapy. METHODS Longitudinal CT images were collected before and during the treatment for HCC patients across three institutions from January 2019 to April 2022. A total of 1316 radiomic features were extracted from arterial and portal venous phase abdominal CT images for each patient. A model called Longitudinal Whole-liver CT-based Radiomics (LWCTR) was developed to categorize patients into responders or non-responders using radiomic features and clinical information through support vector machine (SVM) classifiers. The area under the curve (AUC) was used as the performance metric and subsequently applied for risk stratification and prognostic assessment. The Shapley Additive explanations (SHAP) method was used to calculate the Shapley value, which explains the contribution of each feature in the SVM model to the prediction. RESULTS This study included 395 eligible participants, with a median age of 57 years (IQR 51-66), comprising 344 males and 51 females. The LWCTR model performed well in predicting treatment response, achieving an AUC of 0.883 (95% confidence interval [CI] 0.881-0.888) in the training cohort, 0.876 (0.858-0.895) in the internal validation cohort, and 0.875 (0.860-0.887) in the external test cohort. The Rad-Nomo model, integrating the LWCTR model's prediction score (Rad-score) with the modified Response Evaluation Criteria in Solid Tumors (mRECIST), demonstrated strong prognostic performance. It achieved time-dependent AUC values of 0.902, 0.823, and 0.850 at 1, 2, and 3 years in the internal validation cohort and 0.893, 0.848, and 0.762 at the same intervals in the external test cohort. CONCLUSION The proposed LWCTR model performs well in predicting treatment response and prognosis in patients with HCC receiving ICIs-based combination therapy, potentially contributing to personalized and timely treatment decisions.
Collapse
Affiliation(s)
- Jun Xu
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Intervention, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Junjun Li
- Department of Radiology, The First Affiliated Hospital of University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Tengfei Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
- University of Science and Technology of China, Hefei, 230026, People's Republic of China.
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| | - Xin Luo
- Yangtze Delta Region Institute (Huzhou) & School of Resources and Environment, University of Electronic Science and Technology of China, Huzhou, Chengdu, 313099, 611731, China
| | - Zhangxiang Zhu
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, People's Republic of China
| | - Yimou Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Yong Wang
- Department of Radiology, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, 241001, People's Republic of China
| | - Zhenglin Zhang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
| | - Ruipeng Song
- Department of Hepatobiliary Surgerydivision of Life Sciences and Medicineanhui Province Key Laboratory of Hepatopancreatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, The First Affiliated Hospital of USTC, the University of Science and Technology of China, Hefei, 230001, People's Republic of China
| | - Li-Zhuang Yang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Hongzhi Wang
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
- University of Science and Technology of China, Hefei, 230026, People's Republic of China
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China
| | - Stephen T C Wong
- Department of Systems Medicine and Bioengineering, Houston Methodist Cancer Center, Houston Methodist Hospital, Houston, TX, 77030, USA
- Department of Radiology, Weill Cornell Medical College, New York, NY, 10065, United States
| | - Hai Li
- Anhui Province Key Laboratory of Medical Physics and Technology, Institute of Health and Medical Technology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
- University of Science and Technology of China, Hefei, 230026, People's Republic of China.
- Department of Oncology, Hefei Cancer Hospital, Chinese Academy of Sciences, Hefei, 230031, People's Republic of China.
| |
Collapse
|
6
|
Zhang F, Wang YS, Li SP, Zhao B, Huang N, Song RP, Meng FZ, Feng ZW, Zhang SY, Song HC, Chen XP, Liu LX, Wang JZ. Alpha-fetoprotein combined with initial tumor shape irregularity in predicting the survival of patients with advanced hepatocellular carcinoma treated with immune-checkpoint inhibitors: a retrospective multi-center cohort study. J Gastroenterol 2025; 60:442-455. [PMID: 39714631 PMCID: PMC11922967 DOI: 10.1007/s00535-024-02202-y] [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: 08/12/2024] [Accepted: 12/07/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are playing a significant role in the treatment of hepatocellular carcinoma (HCC). This study aims to explore the prognostic value of alpha-fetoprotein (AFP) and initial tumor shape irregularity in patients treated with ICIs. METHODS In this retrospective, multi-center study, 296 HCC patients were randomly divided into the training set and the validation set in a 3:2 ratio. The training set was used to evaluate prognostic factors and to develop an easily applicable ATSI (AFP and Tumor Shape Irregularity) score, which was verified in the validation set. RESULTS The ATSI score was developed from two independent prognostic risk factors: baseline AFP ≥ 400 ng/ml (HR 1.73, 95% CI 1.01-2.96, P = 0.046) and initial tumor shape irregularity (HR 1.94, 95% CI 1.03-3.65, P = 0.041). The median overall survival (OS) was not reached (95% CI 28.20-NA) in patients who met no criteria (0 points), 25.8 months (95% CI 14.17-NA) in patients who met one criterion (1 point), and 17.03 months (95% CI 11.73-23.83) in patients who met two criteria (2 points) (P = 0.001). The median progression-free survival (PFS) was 10.83 months (95% CI 9.27-14.33) for 0 points, 8.03 months (95% CI 6.77-10.57) for 1 point, and 5.03 months (95% CI 3.83-9.67) for 2 points (P < 0.001). The validation set effectively verified these results (median OS, 37.43/24.27/14.03 months for 0/1/2 points, P = 0.028; median PFS, 13.93/8.30/4.90 months for 0/1/2 points, P < 0.001). CONCLUSIONS The ATSI score can effectively predict prognosis in HCC patients receiving ICIs.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yong-Shuai Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shao-Peng Li
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Bin Zhao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Nan Huang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Rui-Peng Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Fan-Zheng Meng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Zhi-Wen Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China
| | - Shen-Yu Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Hua-Chuan Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xiao-Peng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China.
| | - Lian-Xin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Ji-Zhou Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
7
|
Tao R, Lu H, Dong X, Ren QQ, Fan H, Tang Z, Xia X. A nomogram based on quantitative MR signal intensity predicts early response to combined systemic treatment in patients with hepatocellular carcinoma. Front Oncol 2025; 15:1527108. [PMID: 40171262 PMCID: PMC11959652 DOI: 10.3389/fonc.2025.1527108] [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: 11/12/2024] [Accepted: 02/24/2025] [Indexed: 04/03/2025] Open
Abstract
Objective This study aimed to develop and evaluate the value of a nomogram based on quantitative MR signal intensity to predict response to combined systemic therapy of anti-angiogenesis and immune checkpoint inhibitor (ICI) in hepatocellular carcinoma (HCC) patients. Methods 117 HCC patients who underwent the combined systemic treatment at a tertiary hospital between September 2020 and May 2024 were enrolled and divided into a development cohort (n = 82) and a validation cohort (n = 35). The predictive value of the relative signal intensity attenuation index (rSIAI) based on enhanced MR parameters and laboratory parameters on disease control was evaluated using receiver operating characteristic (ROC) curves, with the determination of optimal cut-off values (COVs) accomplished via Youden's index. Univariate and multivariable analyses were conducted to evaluate the association between COVs and disease control. The validity of the COVs was further confirmed through chi-square testing and calculation of Cramer's V coefficient (V). A nomogram was constructed based on the multivariable logistic regression model and evaluated for clinical applicability. Results rSIAI from arterial to portal phase (rSI_ap) in combination with peripheral T-cell subset (CD4+) achieved the most accurate predictive performance for outcome compared to rSI_ap or CD4+ alone, with an area under the curve (AUC) of the ROC of 0.845 (95% CI, 0.748-0.915). A nomogram based on rSI_ap and CD4+ was constructed. Calibration and decision curve analyses confirmed the clinical relevance and value of the nomogram. Conclusion The nomogram based on rSI_ap has the potential to be a non-invasive tool for predicting disease control in advanced HCC patients who have received combined anti-angiogenesis and ICI therapies.
Collapse
Affiliation(s)
- Ran Tao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Haohao Lu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xiangjun Dong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qian Qian Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hongjie Fan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zhaoming Tang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
8
|
Hapaer G, Che F, Xu Q, Li Q, Liang A, Wang Z, Ziluo J, Zhang X, Wei Y, Yuan Y, Song B. Radiomics-based biomarker for PD-1 status and prognosis analysis in patients with HCC. Front Immunol 2025; 16:1435668. [PMID: 39944703 PMCID: PMC11813882 DOI: 10.3389/fimmu.2025.1435668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 01/13/2025] [Indexed: 03/17/2025] Open
Abstract
Purpose To investigate the impact of preoperative contrast-enhanced CT-based radiomics model on PD-1 prediction in hepatocellular carcinoma (HCC) patients. Methods The study included 105 HCC patients (training cohort: 72; validation cohort: 33) who underwent preoperative contrast-enhanced CT and received systemic sorafenib treatment after surgery. Radiomics score was built for each patient and was integrated with independent clinic radiologic predictors into the radiomics model using multivariable logistic regression analysis. Results Seventeen radiomics features were finally selected to construct the radiomics score. In multivariate analysis, serum creatine and peritumoral enhancement were significant independent factors for PD-1 prediction. The radiomics model integrated radiomics signature with serum creatine and peritumoral enhancement showed good discriminative performance (AUC of 0.897 and 0.794 in the training and validation cohort). Overall survival (OS) was significantly different between the radiomics-predicted PD-1-positive and PD-1-negative groups (OS: 29.66 months, CI:16.03-44.40 vs. 31.04 months, CI: 17.10-44.07, P<0.001). Radiomics-predicted PD-1 was an independent predictor of OS of patients treated with sorafenib after surgery. (Hazard ratio [HR]: 1.61 [1.23-2.1], P<0.001). Conclusion The proposed model based on radiomic signature helps to evaluate PD-1 status of HCC patients and may be used for evaluating patients most likely to benefit from sorafenib as a potentially combination therapy regimen with immune checkpoint therapies.
Collapse
Affiliation(s)
- Gulizaina Hapaer
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Che
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, 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, Chengdu, Sichuan, China
| | - Ailin Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhou Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jituome Ziluo
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zhang
- Pharmaceutical Diagnostics, General Electric (GE) Healthcare, Shanghai, China
| | - Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Yuan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, Sanya People’s Hospital, Sanya, China
| |
Collapse
|
9
|
Liao Z, Chen G, Cao X, Liu L, Li J, Zhu B, Cao Z. Cohort-based nomogram for forensic prediction of SCD: a single-center pilot study. Forensic Sci Med Pathol 2025:10.1007/s12024-024-00920-6. [PMID: 39797964 DOI: 10.1007/s12024-024-00920-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/14/2024] [Indexed: 01/13/2025]
Abstract
Forensic diagnosis of sudden cardiac death (SCD) is an extremely important part of routine forensic practice. The present study aimed to develop and validate nomograms for predicting the probability of SCD with special regards to ischemic heart disease-induced SCD (IHD-induced SCD) based on multiple autopsy variables. A total of 3322 cases, were enrolled and randomly assigned into a training cohort (n = 2325) and a validation cohort (n = 997), respectively. Prediction models of SCD and IHD-induced SCD were developed through multivariable logistic regression based on variables selected by LASSO regression or ridge regression, and prediction model with higher area under the curve (AUC) of the receiver operating characteristic (ROC) curve in the validation cohort was used to establish nomograms. For SCD prediction, discrimination of the nomogram was determined based on the ROC with AUC of 0.751 (95% CI, 0.726-0.775) and 0.735 (95% CI, 0.696-0.774) in the training cohort and validation cohort respectively. The AUC of IHD-induced SCD prediction nomogram in the training cohort and validation cohort were 0.742 (95% CI, 0.716-0.768) and 0.738 (95% CI, 0.698-0.777). To facilitate the use of nomograms in routine casework in forensic practice, web calculators ( https://forensic.shinyapps.io/Forensic_SCD/ , https://forensic.shinyapps.io/Forensic_IHDinducedSCD/ ) were constructed. In conclusion, the present study developed and validated simple and practical nomograms for predicting the probability of SCD and IHD-induced SCD based on multiple autopsy variables. The nomograms have certain efficiency for discrimination and calibration to provide a novel approach to diagnose cause of death, and may become a valuable tool in forensic practice.
Collapse
Affiliation(s)
- Zihan Liao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China
| | - Gaohan Chen
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China
| | - Xingrui Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China
| | - Longqiao Liu
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China
| | - Jiatong Li
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China
| | - Baoli Zhu
- Academy of Forensic Science, Liaoning University, No. 111, Nujiang Street, Huanggu Area, Shenyang, 110031, P. R. China.
| | - Zhipeng Cao
- Department of Forensic Pathology, School of Forensic Medicine, China Medical University, Shenyang, 110122, P. R. China.
- Liaoning Province Key Laboratory of Forensic Bio-evidence Sciences, Shenyang, 110122, P. R. China.
- Center of Forensic Investigation, China Medical University, No. 77, Puhe Road, Shenyang North New Area, Shenyang, 110122, P. R. China.
| |
Collapse
|
10
|
Peng K, Zhang X, Li Z, Wang Y, Sun HW, Zhao W, Pan J, Zhang XY, Wu X, Yu X, Wu C, Weng Y, Lin X, Liu D, Zhan M, Xu J, Zheng L, Zhang Y, Lu L. Myeloid response evaluated by noninvasive CT imaging predicts post-surgical survival and immune checkpoint therapy benefits in patients with hepatocellular carcinoma. Front Immunol 2024; 15:1493735. [PMID: 39687612 PMCID: PMC11646988 DOI: 10.3389/fimmu.2024.1493735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
Background The potential of preoperative CT in the assessment of myeloid immune response and its application in predicting prognosis and immune-checkpoint therapy outcomes in hepatocellular carcinoma (HCC) has not been explored. Methods A total of 165 patients with pathological slides and multi-phase CT images were included to develop a radiomics signature for predicting the imaging-based myeloid response score (iMRS). Overall survival (OS) and recurrence-free survival (RFS) were assessed according to the iMRS risk group and validated in a surgical resection cohort (n = 98). The complementary advantage of iMRS incorporating significant clinicopathologic factors was investigated by the Cox proportional hazards analysis. Additionally, the iMRS in inferring the benefits of immune checkpoint therapy was explored in an immunotherapy cohort (n = 36). Results We showed that AUCs of the optimal radiomics signature for iMRS were 0.941 [95% confidence interval (CI), 0.909-0.973] and 0.833 (0.798-0.868) in the training and test cohorts, respectively. High iMRS was associated with poor RFS and OS. The prognostic performance of the Clinical-iMRS nomogram was better than that of a single parameter (p < 0.05), with a 1-, 3-, and 5-year C-index for RFS of 0.729, 0.709, and 0.713 in the training, test, and surgical resection cohorts, respectively. A high iMRS score predicted a higher proportion of objective response (vs. progressive disease or stable disease; odds ratio, 2.311; 95% CI, 1.144-4.672; p = 0.020; AUC, 0.718) in patients treated with anti-PD-1 and PD-L1. Conclusions iMRS may provide a promising method for predicting local myeloid immune responses in HCC patients, inferring postsurgical prognosis, and evaluating benefits of immune checkpoint therapy.
Collapse
Affiliation(s)
- Kangqiang Peng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiao Zhang
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Medical AI Lab, Hebei Provincial Engineering Research Center for AI-Based Cancer Treatment Decision-Making, The First Hospital of Hebei Medical University, Shijiazhuang, China
| | - Zhongliang Li
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
| | - Yongchun Wang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Department of Pathology, Xiangya Hospital, Central South University, Changsha, China
| | - Hong-Wei Sun
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
| | - Wei Zhao
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Department of Management, School of Business, Macau University of Science and Technology, Macau, Macau SAR, China
| | - Jielin Pan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Department of Radiology, Zhuhai People’s Hospital, Jinan University, Zhuhai, China
| | - Xiao-Yang Zhang
- College of Medicine and Biological Information Engineering, Northeastern University, Shenyang, China
| | - Xiaoling Wu
- Department of Radiology, The First Affiliated Hospital of Jinan University, Guangzhou, China
| | - Xiangrong Yu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Department of Radiology, Zhuhai People’s Hospital, Jinan University, Zhuhai, China
| | - Chong Wu
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yulan Weng
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaowen Lin
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
| | - Dingjie Liu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- The Department of Cerebrovascular Disease, Zhuhai People’s Hospital, Jinan University, Zhuhai, China
| | - Meixiao Zhan
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Guangzhou First People’s Hospital, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| | - Jing Xu
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Limin Zheng
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
- Ministry of Education (MOE) Key Laboratory of Gene Function and Regulation, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yaojun Zhang
- State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Ligong Lu
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (Zhuhai Clinical Medical College), Jinan University, Zhuhai, China
- Guangzhou First People’s Hospital, The Second Affiliated Hospital, School of Medicine, South China University of Technology, Guangzhou, China
| |
Collapse
|
11
|
Hu Y, Zhang L, Qi Q, Ren S, Wang S, Yang L, Zhang J, Liu Y, Li X, Cai X, Duan S, Zhang L. Machine learning-based ultrasomics for predicting response to tyrosine kinase inhibitor in combination with anti-PD-1 antibody immunotherapy in hepatocellular carcinoma: a two-center study. Front Oncol 2024; 14:1464735. [PMID: 39610931 PMCID: PMC11602396 DOI: 10.3389/fonc.2024.1464735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Objective The objective of this study is to build and verify the performance of machine learning-based ultrasomics in predicting the objective response to combination therapy involving a tyrosine kinase inhibitor (TKI) and anti-PD-1 antibody for individuals with unresectable hepatocellular carcinoma (HCC). Radiomic features can reflect the internal heterogeneity of the tumor and changes in its microenvironment. These features are closely related to pathological changes observed in histology, such as cellular necrosis and fibrosis, providing crucial non-invasive biomarkers to predict patient treatment response and prognosis. Methods Clinical, pathological, and pre-treatment ultrasound image data of 134 patients with recurrent unresectable or advanced HCC who treated with a combination of TKI and anti-PD-1 antibody therapy at Henan Provincial People's Hospital and the First Affiliated Hospital of Zhengzhou University between December 2019 and November 2023 were collected and retrospectively analyzed. Using stratified random sampling, patients from the two hospitals were assigned to training cohort (n = 93) and validation cohort (n = 41) at a 7:3 ratio. After preprocessing the ultrasound images, regions of interest (ROIs) were delineated. Ultrasomic features were extracted from the images for dimensionality reduction and feature selection. By utilizing the extreme gradient boosting (XGBoost) algorithm, three models were developed: a clinical model, an ultrasomic model, and a combined model. By analyzing the area under the receiver operating characteristic (ROC) curve (AUC), specificity, sensitivity, and accuracy, the predicted performance of the models was evaluated. In addition, we identified the optimal cutoff for the radiomic score using the Youden index and applied it to stratify patients. The Kaplan-Meier (KM) survival curves were used to examine differences in progression-free survival (PFS) between the two groups. Results Twenty ultrasomic features were selected for the construction of the ultrasomic model. The AUC of the ultrasomic model for the training cohort and validation cohort were 0.999 (95%CI: 0.997-1.000) and 0.828 (95%CI: 0.690-0.966), which compared significant favorably to those of the clinical model [AUC = 0.876 (95%CI: 0.815-0.936) for the training cohort, 0.766 (95%CI: 0.597-0.935) for the validation cohort]. Compared to the ultrasomic model, the combined model demonstrated comparable performance within the training cohort (AUC = 0.977, 95%CI: 0.957-0.998) but higher performance in the validation cohort (AUC = 0.881, 95%CI: 0.758-1.000). However, there was no statistically significant difference (p > 0.05). Furthermore, ultrasomic features were associated with PFS, which was significantly different between patients with radiomic scores (Rad-score) greater than 0.057 and those with Rad-score less than 0.057 in both the training (HR = 0.488, 95% CI: 0.299-0.796, p = 0.003) and validation cohorts (HR = 0.451, 95% CI: 0.229-0.887, p = 0.02). Conclusion The ultrasomic features demonstrates excellent performance in accurately predicting the objective response to TKI in combination with anti-PD-1 antibody immunotherapy among patients with unresectable or advanced HCC.
Collapse
Affiliation(s)
- Yiwen Hu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Linlin Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qinghua Qi
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Ren
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Simeng Wang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lanling Yang
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Juan Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yuanyuan Liu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaoxiao Li
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiguo Cai
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Shaobo Duan
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Health Management, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Lianzhong Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
12
|
Tang Z, Wang W, Gao B, Liu X, Liu X, Zhuo Y, Du J, Ai F, Yang X, Gu H. Unveiling Tim-3 immune checkpoint expression in hepatocellular carcinoma through abdominal contrast-enhanced CT habitat radiomics. Front Oncol 2024; 14:1456748. [PMID: 39582537 PMCID: PMC11581969 DOI: 10.3389/fonc.2024.1456748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 10/11/2024] [Indexed: 11/26/2024] Open
Abstract
Introduction Immune checkpoint inhibitors (ICIs) are important systemic therapeutic agents for hepatocellular carcinoma (HCC), among which T-cell immunoglobulin and mucin-domain containing protein 3 (Tim-3) is considered an emerging target for ICI therapy. This study aims to evaluate the prognostic value of Tim-3 expression and develop a predictive model for Tim-3 infiltration in HCC. Methods We collected data from 424 HCC patients in The Cancer Genome Atlas (TCGA) and data from 102 pathologically confirmed HCC patients from our center for prognostic analysis. Multivariate Cox regression analyses were performed on both datasets to determine the prognostic significance of Tim-3 expression. In radiomics analysis, we used the K-means algorithm to cluster regions of interest in arterial phase enhancement and venous phase enhancement images from patients at our center. Radiomic features were extracted from three subregions as well as the entire tumor using pyradiomics. Five machine learning methods were employed to construct Habitat models based on habitat features and Rad models based on traditional radiomic features. The predictive performance of the models was compared using ROC curves, DCA curves, and calibration curves. Results Multivariate Cox analyses from both our center and the TCGA database indicated that high Tim-3 expression is an independent risk factor for poor prognosis in HCC patients. Higher levels of Tim-3 expression were significantly associated with worse prognosis. Among the ten models evaluated, the Habitat model constructed using the LightGBM algorithm showed the best performance in predicting Tim-3 expression status (training set vs. test set AUC 0.866 vs. 0.824). Discussion This study confirmed the importance of Tim-3 as a prognostic marker in HCC. The habitat radiomics model we developed effectively predicted intratumoral Tim-3 infiltration, providing valuable insights for the evaluation of ICI therapy in HCC patients.
Collapse
Affiliation(s)
- Zhishen Tang
- Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Wei Wang
- Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Bo Gao
- Department of Radiology, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| | - Xuyang Liu
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Xiangyu Liu
- Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Yingquan Zhuo
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Jun Du
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Fujun Ai
- Department of Pathology and Pathophysiology, Guizhou Medical University, Guiyang, China
| | - Xianwu Yang
- School of Clinical Medicine, Guizhou Medical University, Guiyang, China
| | - Huajian Gu
- Department of Pediatric Surgery, Affiliated Hospital of Guizhou Medical University, Guiyang, China
| |
Collapse
|
13
|
Huang Y, Qian H. Advancing Hepatocellular Carcinoma Management Through Peritumoral Radiomics: Enhancing Diagnosis, Treatment, and Prognosis. J Hepatocell Carcinoma 2024; 11:2159-2168. [PMID: 39525830 PMCID: PMC11546143 DOI: 10.2147/jhc.s493227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary liver cancer and is associated with high mortality rates due to late detection and aggressive progression. Peritumoral radiomics, an emerging technique that quantitatively analyzes the tissue surrounding the tumor, has shown significant potential in enhancing the management of HCC. This paper examines the role of peritumoral radiomics in improving diagnostic accuracy, guiding personalized treatment strategies, and refining prognostic assessments. By offering unique insights into the tumor microenvironment, peritumoral radiomics enables more precise patient stratification and informs clinical decision-making. However, the integration of peritumoral radiomics into routine clinical practice faces several challenges. Addressing these challenges through continued research and innovation is crucial for the successful implementation of peritumoral radiomics in HCC management, ultimately leading to improved patient outcomes.
Collapse
Affiliation(s)
- Yanhua Huang
- Department of Ultrasound, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
| | - Hongwei Qian
- Department of Hepatobiliary and Pancreatic Surgery, Shaoxing People’s Hospital, Shaoxing, People’s Republic of China
- Shaoxing Key Laboratory of Minimally Invasive Abdominal Surgery and Precise Treatment of Tumor, Shaoxing, People’s Republic of China
| |
Collapse
|
14
|
Yang C, Zhang ZM, Zhao ZP, Wang ZQ, Zheng J, Xiao HJ, Xu H, Liu H, Yang L. Radiomic analysis based on magnetic resonance imaging for the prediction of VEGF expression in hepatocellular carcinoma patients. Abdom Radiol (NY) 2024; 49:3824-3833. [PMID: 38896246 PMCID: PMC11519187 DOI: 10.1007/s00261-024-04427-0] [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: 04/15/2024] [Revised: 05/30/2024] [Accepted: 05/30/2024] [Indexed: 06/21/2024]
Abstract
OBJECTIVE The purpose of this study was to investigate the ability of radiomic characteristics of magnetic resonance images to predict vascular endothelial growth factor (VEGF) expression in hepatocellular carcinoma (HCC) patients. METHODS One hundred and twenty-four patients with HCC who underwent fat-suppressed T2-weighted imaging (FS-T2WI) and dynamic contrast-enhanced magnetic resonance imaging (DCE-MRI) one week before surgical resection were enrolled in this retrospective study. Immunohistochemical analysis was used to evaluate the expression level of VEGF. Radiomic features were extracted from the axial FS-T2WI, DCE-MRI (arterial phase and portal venous phase) images of axial MRI. Least absolute shrinkage and selection operator (LASSO) and stepwise regression analyses were performed to select the best radiomic features. Multivariate logistic regression models were constructed and validated using tenfold cross-validation. Receiver operating characteristic (ROC) curve analysis, calibration curve analysis and decision curve analysis (DCA) were employed to evaluate these models. RESULTS Our results show that there were 94 patients with high VEGF expression and 30 patients with low VEGF expression among the 124 HCC patients. The FS-T2WI, DCE-MRI and combined MRI radiomics models had AUCs of 0.8713, 0.7819, and 0.9191, respectively. There was no significant difference in the AUC between the FS-T2WI radiomics model and the DCE-MRI radiomics model (p > 0.05), but the AUC for the combined model was significantly greater than the AUCs for the other two models (p < 0.05) according to the DeLong test. The combined model had the greatest net benefit according to the DCA results. CONCLUSION The radiomic model based on multisequence MR images has the potential to predict VEGF expression in HCC patients. The combined model showed the best performance.
Collapse
Affiliation(s)
- Cui Yang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Ze-Ming Zhang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Zhang-Ping Zhao
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Zhi-Qing Wang
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Jing Zheng
- Medical Imaging Key Laboratory of Sichuan Province, Science and Technology Innovation Center, Interventional Medical Center, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, P. R. China
| | - Hua-Jing Xiao
- Department of Pathology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Hong Xu
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Hui Liu
- Department of Radiology, Panzhihua Central Hospital, Panzhihua, 617000, Sichuan, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Science and Technology Innovation Center, Interventional Medical Center, Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, 637000, Sichuan, P. R. China.
| |
Collapse
|
15
|
Qi L, Zhu Y, Li J, Zhou M, Liu B, Chen J, Shen J. CT radiomics-based biomarkers can predict response to immunotherapy in hepatocellular carcinoma. Sci Rep 2024; 14:20027. [PMID: 39198563 PMCID: PMC11358293 DOI: 10.1038/s41598-024-70208-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Accepted: 08/13/2024] [Indexed: 09/01/2024] Open
Abstract
Hepatocellular Carcinoma (HCC) remains a leading cause of cancer deaths. Despite the rise of immunotherapies, many HCC patients don't benefit. There's a clear need for biomarkers to guide treatment decisions. This research aims to identify such biomarkers by combining radiological data and machine learning. We analyzed clinical and CT imaging data of 54 HCC patients undergoing immunotherapy. Radiologic features were examined to develop a model predicting short-term immunotherapy effects. We utilized 9 machine learning and 2 ensemble learning techniques using RapidMiner for model construction. We conducted the validation of the above feature combination using 29 HCC patients who received immunotherapy from another hospital, and tested and validated it using XGBoost combined with a genetic algorithm. In 54 HCC patients, radiomics features varied significantly between those with partial response (PR) and stable disease (SD). Key features in Gray Level Run Length Matrix (GLRLM) and in adjacent tissues' Intensity Direct, Neighborhood Gray Tone Difference Matrix (NGTDM), and Shape correlated with short-term immunotherapy efficacy. Selected feature combinations of 15, 19, and 8/15 features yielded better outcomes. Deep learning, random forest, and naive bayes outperformed other methods, with Bagging being more reliable than Adaboost. In the validation set of 29 HCC patients, the mentioned feature combination also demonstrated favorable performance. Furthermore, we achieved improved results when employing XGBoost in conjunction with a genetic algorithm for testing and validation. The machine learning model built with CT image features derived from GLCM, GLRLM, IntensityDirect, NGTDM, and Shape can accurately forecast the short-term efficacy of immunotherapy for HCC.
Collapse
Affiliation(s)
- Liang Qi
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No.321 Zhongshan Road, Nanjing, 210008, China
| | - Yahui Zhu
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No.321 Zhongshan Road, Nanjing, 210008, China
| | - Jinxin Li
- Department of Li Ka Shing Faculty of Medicine, The University of Hong Kong, HKSAR, China
| | - Mingzhen Zhou
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China
| | - Baorui Liu
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No.321 Zhongshan Road, Nanjing, 210008, China.
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| | - Jiu Chen
- Department of Radiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No.321 Zhongshan Road, Nanjing, 210008, China.
- Institute of Medical Imaging and Artificial Intelligence, Nanjing University, Nanjing, China.
| | - Jie Shen
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, No.321 Zhongshan Road, Nanjing, 210008, China.
- The Comprehensive Cancer Centre, Department of Oncology, Nanjing Drum Tower Hospital Clinical College of Nanjing University of Chinese Medicine, Nanjing, China.
| |
Collapse
|
16
|
Hua Y, Sun Z, Xiao Y, Li H, Ma X, Luo X, Tan W, Xie Z, Zhang Z, Tang C, Zhuang H, Xu W, Zhu H, Chen Y, Shang C. Pretreatment CT-based machine learning radiomics model predicts response in unresectable hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors and interventional therapy. J Immunother Cancer 2024; 12:e008953. [PMID: 39029924 PMCID: PMC11261678 DOI: 10.1136/jitc-2024-008953] [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] [Accepted: 06/25/2024] [Indexed: 07/21/2024] Open
Abstract
BACKGROUND Lenvatinib plus PD-1 inhibitors and interventional (LPI) therapy have demonstrated promising treatment effects in unresectable hepatocellular carcinoma (HCC). However, biomarkers for predicting the response to LPI therapy remain to be further explored. We aimed to develop a radiomics model to noninvasively predict the efficacy of LPI therapy. METHODS Clinical data of patients with HCC receiving LPI therapy were collected in our institution. The clinical model was built with clinical information. Nine machine learning classifiers were tested and the multilayer perceptron classifier with optimal performance was used as the radiomics model. The clinical-radiomics model was constructed by integrating clinical and radiomics scores through logistic regression analysis. RESULTS 151 patients were enrolled in this study (2:1 randomization, 101 and 50 in the training and validation cohorts), of which three achieved complete response, 69 showed partial response, 46 showed stable disease, and 33 showed progressive disease. The objective response rate, disease control rate, and conversion resection rates were 47.7, 78.1 and 23.2%. 14 features were selected from the initially extracted 1223 for radiomics model construction. The area under the curves of the radiomics model (0.900 for training and 0.893 for validation) were comparable to that of the clinical-radiomics model (0.912 for training and 0.892 for validation), and both were superior to the clinical model (0.669 for training and 0.585 for validation). Meanwhile, the radiomics model can categorize participants into high-risk and low-risk groups for progression-free survival (PFS) and overall survival (OS) in the training (HR 1.913, 95% CI 1.121 to 3.265, p=0.016 for PFS; HR 4.252, 95% CI 2.051 to 8.816, p=0.001 for OS) and validation sets (HR 2.347, 95% CI 1.095 to 5.031, p=0.012 for PFS; HR 2.592, 95% CI 1.050 to 6.394, p=0.019 for OS). CONCLUSION The promising machine learning radiomics model was developed and validated to predict the efficacy of LPI therapy for patients with HCC and perform risk stratification, with comparable performance to clinical-radiomics model.
Collapse
Affiliation(s)
- Yonglin Hua
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Zhixian Sun
- Breast Disease Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Laboratory of General Surgery, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
- Breast Tumor Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuxin Xiao
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Huilong Li
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Xiaowu Ma
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Xuan Luo
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Wenliang Tan
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Zhiqin Xie
- Department of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Central South University Xiangya School of Medicine, Zhuzhou, Hunan, China
| | - Ziyu Zhang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Chenwei Tang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Hongkai Zhuang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Weikai Xu
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Haihong Zhu
- Department of General Surgery, Qinghai Provincial People’s Hospital, Xining, Qinghai, China
| | - Yajin Chen
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| | - Changzhen Shang
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, Guangdong, China
| |
Collapse
|
17
|
Huang Y, Chen L, Ding Q, Zhang H, Zhong Y, Zhang X, Weng S. CT-based radiomics for predicting pathological grade in hepatocellular carcinoma. Front Oncol 2024; 14:1295575. [PMID: 38690170 PMCID: PMC11059035 DOI: 10.3389/fonc.2024.1295575] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2023] [Accepted: 03/18/2024] [Indexed: 05/02/2024] Open
Abstract
Objective To construct and validate radiomics models for hepatocellular carcinoma (HCC) grade predictions based on contrast-enhanced CT (CECT). Methods Patients with pathologically confirmed HCC after surgery and underwent CECT at our institution between January 2016 and December 2020 were enrolled and randomly divided into training and validation datasets. With tumor segmentation and feature extraction, radiomic models were constructed using univariate analysis, followed by least absolute shrinkage and selection operator (LASSO) regression. In addition, combined models with clinical factors and radiomics scores (Radscore) were constructed using logistic regression. Finally, all models were evaluated using the receiver operating characteristic (ROC) curve with the area under the curve (AUC), calibration curve, and decision curve analysis (DCA). Results In total 242 patients were enrolled in this study, of whom 170 and 72 formed the training and validation datasets, respectively. The arterial phase and portal venous phase (AP+VP) radiomics model were evaluated as the best for predicting HCC pathological grade among all the models built in our study (AUC = 0.981 in the training dataset; AUC = 0.842 in the validation dataset) and was used to build a nomogram. Furthermore, the calibration curve and DCA indicated that the AP+VP radiomics model had a satisfactory prediction efficiency. Conclusions Low- and high-grade HCC can be distinguished with good diagnostic performance using a CECT-based radiomics model.
Collapse
Affiliation(s)
- Yue Huang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Lingfeng Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Qingzhu Ding
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Han Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Yun Zhong
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Xiang Zhang
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| | - Shangeng Weng
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Fujian Abdominal Surgery Research Institute, The First Affiliated Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
18
|
Sheng Y, Wang Q, Liu H, Wang Q, Chen W, Xing W. Prognostic nomogram model for selecting between transarterial chemoembolization plus lenvatinib, with and without PD-1 inhibitor in unresectable hepatocellular carcinoma. Br J Radiol 2024; 97:668-679. [PMID: 38303541 PMCID: PMC11027259 DOI: 10.1093/bjr/tqae018] [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: 09/10/2023] [Revised: 12/11/2023] [Accepted: 01/13/2024] [Indexed: 02/03/2024] Open
Abstract
OBJECTIVES To establish and verify a prognostic nomogram model for selecting in unresectable hepatocellular carcinoma (uHCC) treated by transarterial chemoembolization plus lenvatinib (TACE-L) with or without PD-1 inhibitor. METHODS Data of 241 uHCC patients who underwent TACE-L (n = 128) and TACE-L plus PD-1 inhibitor (TACE-L-P, n = 113) were retrospectively reviewed. The differences in tumour responses, progression-free survival (PFS), overall survival (OS), and adverse events (AEs) between two groups were compared, and a prognostic nomogram model was established based on independent clinical-radiologic factors and confirmed by Cox regression analysis for predicting PFS and OS. The treatment selection for uHCC patients was stratified by the nomogram score. RESULTS Compared to TACE-L, TACE-L-P presented prolonged PFS (14.0 vs. 9.0 months, P < .001), longer OS (24.0 vs. 15.0 months, P < .001), and a better overall objective response rate (54.0% vs. 32.8%, P = .001). There was no significant difference between the rate of AEs in the TACE-L-P and the TACE-L (56.64% vs. 46.09%, P = .102) and the rate of grade ≥ 3 AEs (11.50% vs. 9.38%, P = .588), respectively. The nomogram model presented good discrimination, with a C-index of 0.790 for predicting PFS and 0.749 for predicting OS. Patients who underwent TACE-L and obtained a nomogram score >9 demonstrated improved 2-year PFS when transferred to TACE-L-P, and those with a nomogram ≤25 had better 2-year OS when transferred to TACE-L-P. CONCLUSIONS TACE-L-P showed significant improvements in efficiency and safety for uHCC patients compared with TACE-L. The nomogram was useful for stratifying treatment decisions and selecting a suitable population for uHCC patients. ADVANCES IN KNOWLEDGE Prognostic nomogram model is of great value in predicting individualized survival benefits for uHCC patients after TACE-L or/and TACE-L-P. And the nomogram was helpful for selection between TACE-L-P and TACE-L among uHCC patients.
Collapse
Affiliation(s)
- Ye Sheng
- Department of Interventional Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| | - Qing Wang
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| | - HaiFeng Liu
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| | - Qi Wang
- Department of Interventional Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| | - WenHua Chen
- Department of Interventional Radiology, Third Affiliated Hospital of Soochow University & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| | - Wei Xing
- Department of Radiology, Third Affiliated Hospital of Soochow University, Changzhou & Changzhou First People’s Hospital, Juqian street NO.185, Tianning district, Changzhou, Jiangsu, 213003, China
| |
Collapse
|
19
|
Ligero M, Gielen B, Navarro V, Cresta Morgado P, Prior O, Dienstmann R, Nuciforo P, Trebeschi S, Beets-Tan R, Sala E, Garralda E, Perez-Lopez R. A whirl of radiomics-based biomarkers in cancer immunotherapy, why is large scale validation still lacking? NPJ Precis Oncol 2024; 8:42. [PMID: 38383736 PMCID: PMC10881558 DOI: 10.1038/s41698-024-00534-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Accepted: 01/26/2024] [Indexed: 02/23/2024] Open
Abstract
The search for understanding immunotherapy response has sparked interest in diverse areas of oncology, with artificial intelligence (AI) and radiomics emerging as promising tools, capable of gathering large amounts of information to identify suitable patients for treatment. The application of AI in radiology has grown, driven by the hypothesis that radiology images capture tumor phenotypes and thus could provide valuable insights into immunotherapy response likelihood. However, despite the rapid growth of studies, no algorithms in the field have reached clinical implementation, mainly due to the lack of standardized methods, hampering study comparisons and reproducibility across different datasets. In this review, we performed a comprehensive assessment of published data to identify sources of variability in radiomics study design that hinder the comparison of the different model performance and, therefore, clinical implementation. Subsequently, we conducted a use-case meta-analysis using homogenous studies to assess the overall performance of radiomics in estimating programmed death-ligand 1 (PD-L1) expression. Our findings indicate that, despite numerous attempts to predict immunotherapy response, only a limited number of studies share comparable methodologies and report sufficient data about cohorts and methods to be suitable for meta-analysis. Nevertheless, although only a few studies meet these criteria, their promising results underscore the importance of ongoing standardization and benchmarking efforts. This review highlights the importance of uniformity in study design and reporting. Such standardization is crucial to enable meaningful comparisons and demonstrate the validity of biomarkers across diverse populations, facilitating their implementation into the immunotherapy patient selection process.
Collapse
Affiliation(s)
- Marta Ligero
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Bente Gielen
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Victor Navarro
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Pablo Cresta Morgado
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
- Prostate Cancer Translational Research Group, Institute of Oncology (VHIO), Vall d'Hebron University Hospital, Barcelona, Spain
| | - Olivia Prior
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Rodrigo Dienstmann
- Oncology Data Science (ODysSey) Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain
| | - Paolo Nuciforo
- Molecular Oncology Group, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Stefano Trebeschi
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
| | - Regina Beets-Tan
- Department of Radiology, Netherlands Cancer Institute, Amsterdam, The Netherlands
- GROW School for Oncology and Reproduction, Maastricht University, Maastricht, The Netherlands
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark
| | - Evis Sala
- Dipartimento Diagnostica per Immagini, Radioterapia Oncologica ed Ematologia, Policlinico Universitario A. Gemelli IRCCS, Rome, Italy
- Dipartimento di Scienze Radiologiche ed Ematologiche, Universita Cattolica del Sacro Cuore, Rome, Italy
| | - Elena Garralda
- Department of Medical Oncology, Vall d'Hebron University Hospital and Institute of Oncology (VHIO), Barcelona, Spain
| | - Raquel Perez-Lopez
- Radiomics Group, Vall d'Hebron Institute of Oncology (VHIO), Barcelona, Spain.
| |
Collapse
|
20
|
Maino C, Vernuccio F, Cannella R, Franco PN, Giannini V, Dezio M, Pisani AR, Blandino AA, Faletti R, De Bernardi E, Ippolito D, Gatti M, Inchingolo R. Radiomics and liver: Where we are and where we are headed? Eur J Radiol 2024; 171:111297. [PMID: 38237517 DOI: 10.1016/j.ejrad.2024.111297] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2023] [Revised: 01/03/2024] [Accepted: 01/07/2024] [Indexed: 02/10/2024]
Abstract
Hepatic diffuse conditions and focal liver lesions represent two of the most common scenarios to face in everyday radiological clinical practice. Thanks to the advances in technology, radiology has gained a central role in the management of patients with liver disease, especially due to its high sensitivity and specificity. Since the introduction of computed tomography (CT) and magnetic resonance imaging (MRI), radiology has been considered the non-invasive reference modality to assess and characterize liver pathologies. In recent years, clinical practice has moved forward to a quantitative approach to better evaluate and manage each patient with a more fitted approach. In this setting, radiomics has gained an important role in helping radiologists and clinicians characterize hepatic pathological entities, in managing patients, and in determining prognosis. Radiomics can extract a large amount of data from radiological images, which can be associated with different liver scenarios. Thanks to its wide applications in ultrasonography (US), CT, and MRI, different studies were focused on specific aspects related to liver diseases. Even if broadly applied, radiomics has some advantages and different pitfalls. This review aims to summarize the most important and robust studies published in the field of liver radiomics, underlying their main limitations and issues, and what they can add to the current and future clinical practice and literature.
Collapse
Affiliation(s)
- Cesare Maino
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy.
| | - Federica Vernuccio
- Institute of Radiology, University Hospital of Padova, Padova 35128, Italy
| | - Roberto Cannella
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Paolo Niccolò Franco
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy
| | - Valentina Giannini
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Michele Dezio
- Department of Radiology, Miulli Hospital, Acquaviva delle Fonti 70021, Bari, Italy
| | - Antonio Rosario Pisani
- Nuclear Medicine Unit, Interdisciplinary Department of Medicine, University of Bari, Bari 70121, Italy
| | - Antonino Andrea Blandino
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, Palermo 90127, Italy
| | - Riccardo Faletti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Elisabetta De Bernardi
- Bicocca Bioinformatics Biostatistics and Bioimaging Centre - B4, University of Milano Bicocca, Milano 20100, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Davide Ippolito
- Department of Radiology, Fondazione IRCCS San Gerardo dei Tintori, Monza 20900, Italy; School of Medicine, University of Milano Bicocca, Milano 20100, Italy
| | - Marco Gatti
- Department of Surgical Sciences, University of Turin, Turin 10126, Italy
| | - Riccardo Inchingolo
- Unit of Interventional Radiology, F. Miulli Hospital, Acquaviva delle Fonti 70021, Italy
| |
Collapse
|
21
|
Berbís MÁ, Godino FP, Rodríguez-Comas J, Nava E, García-Figueiras R, Baleato-González S, Luna A. Radiomics in CT and MR imaging of the liver and pancreas: tools with potential for clinical application. Abdom Radiol (NY) 2024; 49:322-340. [PMID: 37889265 DOI: 10.1007/s00261-023-04071-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 09/15/2023] [Accepted: 09/19/2023] [Indexed: 10/28/2023]
Abstract
Radiomics allows the extraction of quantitative imaging features from clinical magnetic resonance imaging (MRI) and computerized tomography (CT) studies. The advantages of radiomics have primarily been exploited in oncological applications, including better characterization and staging of oncological lesions and prediction of patient outcomes and treatment response. The potential introduction of radiomics in the clinical setting requires the establishment of a standardized radiomics pipeline and a quality assurance program. Radiomics and texture analysis of the liver have improved the differentiation of hypervascular lesions such as adenomas, focal nodular hyperplasia, and hepatocellular carcinoma (HCC) during the arterial phase, and in the pretreatment determination of HCC prognostic factors (e.g., tumor grade, microvascular invasion, Ki-67 proliferation index). Radiomics of pancreatic CT and MR images has enhanced pancreatic ductal adenocarcinoma detection and its differentiation from pancreatic neuroendocrine tumors, mass-forming chronic pancreatitis, or autoimmune pancreatitis. Radiomics can further help to better characterize incidental pancreatic cystic lesions, accurately discriminating benign from malignant intrapancreatic mucinous neoplasms. Nonetheless, despite their encouraging results and exciting potential, these tools have yet to be implemented in the clinical setting. This non-systematic review will describe the essential steps in the implementation of the radiomics and feature extraction workflow from liver and pancreas CT and MRI studies for their potential clinical application. A succinct overview of reported radiomics applications in the liver and pancreas and the challenges and limitations of their implementation in the clinical setting is also discussed, concluding with a brief exploration of the future perspectives of radiomics in the gastroenterology field.
Collapse
Affiliation(s)
- M Álvaro Berbís
- Department of Radiology, HT Médica, San Juan de Dios Hospital, 14960, Córdoba, Spain.
- Department of Radiology, HT Médica, San Juan de Dios Hospital, Av. del Brillante, 106, 14012, Córdoba, Spain.
| | | | | | - Enrique Nava
- Department of Communications Engineering, University of Málaga, 29016, Málaga, Spain
| | - Roberto García-Figueiras
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Sandra Baleato-González
- Abdominal Imaging Section, University Clinical Hospital of Santiago, 15706, Santiago de Compostela, A Coruña, Spain
| | - Antonio Luna
- Department of Radiology, HT Médica, Clínica las Nieves, 23007, Jaén, Spain
| |
Collapse
|
22
|
Wu J, Liu W, Qiu X, Li J, Song K, Shen S, Huo L, Chen L, Xu M, Wang H, Jia N, Chen L. A Noninvasive Approach to Evaluate Tumor Immune Microenvironment and Predict Outcomes in Hepatocellular Carcinoma. PHENOMICS (CHAM, SWITZERLAND) 2023; 3:549-564. [PMID: 38223688 PMCID: PMC10781918 DOI: 10.1007/s43657-023-00136-8] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Revised: 09/21/2023] [Accepted: 10/13/2023] [Indexed: 01/16/2024]
Abstract
It is widely recognized that tumor immune microenvironment (TIME) plays a crucial role in tumor progression, metastasis, and therapeutic response. Despite several noninvasive strategies have emerged for cancer diagnosis and prognosis, there are still lack of effective radiomic-based model to evaluate TIME status, let alone predict clinical outcome and immune checkpoint inhibitor (ICIs) response for hepatocellular carcinoma (HCC). In this study, we developed a radiomic model to evaluate TIME status within the tumor and predict prognosis and immunotherapy response. A total of 301 patients who underwent magnetic resonance imaging (MRI) examinations were enrolled in our study. The intra-tumoral expression of 17 immune-related molecules were evaluated using co-detection by indexing (CODEX) technology, and we construct Immunoscore (IS) with the least absolute shrinkage and selection operator (LASSO) algorithm and Cox regression method to evaluate TIME. Of 6115 features extracted from MRI, five core features were filtered out, and the Radiomic Immunoscore (RIS) showed high accuracy in predicting TIME status in testing cohort (area under the curve = 0.753). More importantly, RIS model showed the capability of predicting therapeutic response to anti-programmed cell death 1 (PD-1) immunotherapy in an independent cohort with advanced HCC patients (area under the curve = 0.731). In comparison with previously radiomic-based models, our integrated RIS model exhibits not only higher accuracy in predicting prognosis but also the potential guiding significance to HCC immunotherapy. Supplementary Information The online version contains supplementary material available at 10.1007/s43657-023-00136-8.
Collapse
Affiliation(s)
- Jianmin Wu
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438 China
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
| | - Wanmin Liu
- Department of Radiology, Tongji Hospital, School of Medicine, Tongji University, Shanghai, 200333 China
| | - Xinyao Qiu
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Jing Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022 China
| | - Kairong Song
- Department of Radiology, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438 China
| | - Siyun Shen
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
| | - Lei Huo
- Department of Radiology, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438 China
| | - Lu Chen
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
| | - Mingshuang Xu
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
| | - Hongyang Wang
- Shanghai Key Laboratory of Metabolic Remodeling and Health, Institute of Metabolism and Integrative Biology, Fudan University, Shanghai, 200438 China
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032 China
| | - Ningyang Jia
- Department of Radiology, Third Affiliated Hospital of Naval Medical University, Shanghai, 200438 China
| | - Lei Chen
- The International Cooperation Laboratory on Signal Transduction, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438 China
- National Center for Liver Cancer, Shanghai, 201805 China
| |
Collapse
|
23
|
Zhang R, Zhao H, Wang P, Guo Z, Liu C, Qu Z. Hepatocellular carcinoma immune prognosis score predicts the clinical outcomes of hepatocellular carcinoma patients receiving immune checkpoint inhibitors. BMC Cancer 2023; 23:1181. [PMID: 38041022 PMCID: PMC10693152 DOI: 10.1186/s12885-023-11678-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/24/2023] [Indexed: 12/03/2023] Open
Abstract
OBJECTIVE The predictive biomarkers of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC) still need to be further explored. This study aims to establish a new immune prognosis biomarker to predict the clinical outcomes of hepatocellular carcinoma patients receiving immune checkpoint inhibitors. METHODS The subjects of this study were 151 HCC patients receiving ICIs at Harbin Medical University Cancer Hospital from January 2018 to December 2021. This study collected a wide range of blood parameters from patients before treatment and used Cox's regression analysis to identify independent prognostic factors in blood parameters, as well as their β coefficient. The hepatocellular carcinoma immune prognosis score (HCIPS) was established through Lasso regression analysis and COX multivariate analysis. The cut-off value of HCIPS was calculated from the receiver operating characteristic (ROC) curve. Finally, the prognostic value of HCIPS was validated through survival analysis, stratified analyses, and nomograms. RESULTS HCIPS was composed of albumin (ALB) and thrombin time (TT), with a cut-off value of 0.64. There were 56 patients with HCIPS < 0.64 and 95 patients with HCIPS ≥ 0.64, patients with low HCIPS were significantly related to shorter progression-free survival (PFS) (13.10 months vs. 1.63 months, P < 0.001) and overall survival (OS) (14.83 months vs. 25.43 months, P < 0.001). HCIPS has also been found to be an independent prognostic factor in this study. In addition, the stratified analysis found a significant correlation between low HCIPS and shorter OS in patients with tumor size ≥ 5 cm (P of interaction = 0.032). The C-index and 95% CI of the nomograms for PFS and OS were 0.730 (0.680-0.779) and 0.758 (0.711-0.804), respectively. CONCLUSIONS As a new score established based on HCC patients receiving ICIs, HCIPS was significantly correlated with clinical outcomes in patients with ICIs and might serve as a new biomarker to predict HCC patients who cloud benefit from ICIs.
Collapse
Affiliation(s)
- Rujia Zhang
- Department of Operating Room, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, 150086, Heilongjiang, China
| | - Haoran Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Peng Wang
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Zuoming Guo
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Chunxun Liu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China
| | - Zhaowei Qu
- Department of Hepatobiliary and Pancreatic Surgery, Harbin Medical University Cancer Hospital, Harbin Medical University, Harbin, 150081, Heilongjiang, China.
| |
Collapse
|
24
|
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
|
25
|
Kuang T, Ma W, Zhang J, Yu J, Deng W, Dong K, Wang W. Construction of a Nomogram to Predict Overall Survival in Patients with Early-Onset Hepatocellular Carcinoma: A Retrospective Cohort Study. Cancers (Basel) 2023; 15:5310. [PMID: 38001570 PMCID: PMC10670167 DOI: 10.3390/cancers15225310] [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: 10/18/2023] [Revised: 10/31/2023] [Accepted: 11/03/2023] [Indexed: 11/26/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is a widespread and impactful cancer which has pertinent implications worldwide. Although most cases of HCC are typically diagnosed in individuals aged ≥60 years, there has been a notable rise in the occurrence of HCC among younger patients. However, there is a scarcity of precise prognostic models available for predicting outcomes in these younger patients. A retrospective analysis was conducted to investigate early-onset hepatocellular carcinoma (EO-LIHC) using data from the Surveillance, Epidemiology, and End Results (SEER) database from 2004 to 2018. The analysis included 1392 patients from the SEER database and our hospital. Among them, 1287 patients from the SEER database were assigned to the training cohort (n = 899) and validation cohort 1 (n = 388), while 105 patients from our hospital were assigned to validation cohort 2. A Cox regression analysis showed that age, sex, AFP, grade, stage, tumor size, surgery, and chemotherapy were independent risk factors. The nomogram developed in this study demonstrated its discriminatory ability to predict the 1-, 3-, and 5-year overall survival (OS) rates in EO-LIHC patients based on individual characteristics. Additionally, a web-based OS prediction model specifically tailored for EO-LIHC patients was created and validated. Overall, these advancements contribute to improved decision-making and personalized care for individuals with EO-LIHC.
Collapse
Affiliation(s)
- Tianrui Kuang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wangbin Ma
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jiacheng Zhang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Jia Yu
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Wenhong Deng
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Keshuai Dong
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| | - Weixing Wang
- Department of General Surgery, Renmin Hospital of Wuhan University, Wuhan 430060, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan 430060, China
| |
Collapse
|
26
|
Greten TF, Villanueva A, Korangy F, Ruf B, Yarchoan M, Ma L, Ruppin E, Wang XW. Biomarkers for immunotherapy of hepatocellular carcinoma. Nat Rev Clin Oncol 2023; 20:780-798. [PMID: 37726418 DOI: 10.1038/s41571-023-00816-4] [Citation(s) in RCA: 83] [Impact Index Per Article: 41.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/29/2023] [Indexed: 09/21/2023]
Abstract
Immune-checkpoint inhibitors (ICIs) are now widely used for the treatment of patients with advanced-stage hepatocellular carcinoma (HCC). Two different ICI-containing regimens, atezolizumab plus bevacizumab and tremelimumab plus durvalumab, are now approved standard-of-care first-line therapies in this setting. However, and despite substantial improvements in survival outcomes relative to sorafenib, most patients with advanced-stage HCC do not derive durable benefit from these regimens. Advances in genome sequencing including the use of single-cell RNA sequencing (both of tumour material and blood samples), as well as immune cell identification strategies and other techniques such as radiomics and analysis of the microbiota, have created considerable potential for the identification of novel predictive biomarkers enabling the accurate selection of patients who are most likely to derive benefit from ICIs. In this Review, we summarize data on the immunology of HCC and the outcomes in patients receiving ICIs for the treatment of this disease. We then provide an overview of current biomarker use and developments in the past 5 years, including gene signatures, circulating tumour cells, high-dimensional flow cytometry, single-cell RNA sequencing as well as approaches involving the microbiome, radiomics and clinical markers. Novel concepts for further biomarker development in HCC are then discussed including biomarker-driven trials, spatial transcriptomics and integrated 'big data' analysis approaches. These concepts all have the potential to better identify patients who are most likely to benefit from ICIs and to promote the development of new treatment approaches.
Collapse
Affiliation(s)
- Tim F Greten
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA.
| | - Augusto Villanueva
- Divisions of Liver Disease and Hematology/Medical Oncology, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Firouzeh Korangy
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Benjamin Ruf
- Gastrointestinal Malignancies Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Mark Yarchoan
- Bloomberg-Kimmel Institute for Cancer Immunotherapy, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Lichun Ma
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Eytan Ruppin
- Cancer Data Science Laboratory, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Xin W Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Liver Carcinogenesis Section, Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| |
Collapse
|
27
|
Zhao Y, Zhang J, Wang N, Xu Q, Liu Y, Liu J, Zhang Q, Zhang X, Chen A, Chen L, Sheng L, Song Q, Wang F, Guo Y, Liu A. Intratumoral and peritumoral radiomics based on contrast-enhanced MRI for preoperatively predicting treatment response of transarterial chemoembolization in hepatocellular carcinoma. BMC Cancer 2023; 23:1026. [PMID: 37875815 PMCID: PMC10594790 DOI: 10.1186/s12885-023-11491-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Accepted: 10/08/2023] [Indexed: 10/26/2023] Open
Abstract
BACKGROUND Noninvasive and precise methods to estimate treatment response and identify hepatocellular carcinoma (HCC) patients who could benefit from transarterial chemoembolization (TACE) are urgently required. The present study aimed to investigate the ability of intratumoral and peritumoral radiomics based on contrast-enhanced magnetic resonance imaging (CE-MRI) to preoperatively predict tumor response to TACE in HCC patients. METHODS A total of 138 patients with HCC who received TACE were retrospectively included and randomly divided into training and validation cohorts at a ratio of 7:3. Total 1206 radiomics features were extracted from arterial, venous, and delayed phases images. The inter- and intraclass correlation coefficients, the spearman's rank correlation test, and the gradient boosting decision tree algorithm were used for radiomics feature selection. Radiomics models on intratumoral region (TR) and peritumoral region (PTR) (3 mm, 5 mm, and 10 mm) were established using logistic regression. Three integrated radiomics models, including intratumoral and peritumoral region (T-PTR) (3 mm), T-PTR (5 mm), and T-PTR (10 mm) models, were constructed using TR and PTR radiomics scores. A clinical-radiological model and a combined model incorporating the optimal radiomics score and selected clinical-radiological predictors were constructed, and the combined model was presented as a nomogram. The discrimination, calibration, and clinical utilities were evaluated by receiver operating characteristic curve, calibration curve, and decision curve analysis, respectively. RESULTS The T-PTR radiomics models performed better than the TR and PTR models, and the T-PTR (3 mm) radiomics model demonstrated preferable performance with the AUCs of 0.884 (95%CI, 0.821-0.936) and 0.911 (95%CI, 0.825-0.975) in both training and validation cohorts. The T-PTR (3 mm) radiomics score, alkaline phosphatase, tumor size, and satellite nodule were fused to construct a combined nomogram. The combined nomogram [AUC: 0.910 (95%CI, 0.854-0.958) and 0.918 (95%CI, 0.831-0.986)] outperformed the clinical-radiological model [AUC: 0.789 (95%CI, 0.709-0.863) and 0.782 (95%CI, 0.660-0.902)] in the both cohorts and achieved good calibration capability and clinical utility. CONCLUSIONS CE-MRI-based intratumoral and peritumoral radiomics approach can provide an effective tool for the precise and individualized estimation of treatment response for HCC patients treated with TACE.
Collapse
Affiliation(s)
- Ying Zhao
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Jian Zhang
- Department of Interventional Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Nan Wang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Qihao Xu
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Yuhui Liu
- College of Medical Imaging, Dalian Medical University, Dalian, China
| | - Jinghong Liu
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Qinhe Zhang
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Xinyuan Zhang
- College of Medical Imaging, Dalian Medical University, Dalian, China
| | - Anliang Chen
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Lihua Chen
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Liuji Sheng
- College of Medical Imaging, Dalian Medical University, Dalian, China
| | - Qingwei Song
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China
| | - Feng Wang
- Department of Interventional Radiology, The First Affiliated Hospital, Dalian Medical University, Dalian, China
| | - Yan Guo
- GE Healthcare (China), Shanghai, China
| | - Ailian Liu
- Department of Radiology, The First Affiliated Hospital, Dalian Medical University, No. 222 Zhongshan Road, Xigang District, Dalian, Liaoning, China.
| |
Collapse
|
28
|
Wang X, Huang H, Zhang L, Wu Y, Wen Y, Weng X, Chen Q, Liu W. Elevated levels of neutrophil related chemokine citrullinated histone H3, interleukin-8 and C-reaction protein in patients with immune checkpoint inhibitor therapy: predictive biomarkers for response to treatment. Cancer Cell Int 2023; 23:167. [PMID: 37580733 PMCID: PMC10426204 DOI: 10.1186/s12935-023-02994-8] [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: 04/17/2023] [Accepted: 07/19/2023] [Indexed: 08/16/2023] Open
Abstract
BACKGROUND Immune checkpoint inhibitor (ICI) therapy has been used in various tumors. The biomarkers predictive of a response to ICI treatment remain unclear, and additional and combined biomarkers are urgently needed. Secreted factors related to the tumor microenvironment (TME) have been evaluated to identify novel noninvasive predictive biomarkers. METHODS We analyzed 85 patients undergoing ICI therapy as the primary cohort. The associations between ICI response and all biomarkers were evaluated. A prediction model and a nomogram were developed and validated based on the above factors. RESULTS Seventy-seven patients were enrolled in the validation cohort. In the primary cohort, the baseline serum levels of H3Cit, IL-8 and CRP were significantly higher in nonresponder patients. A model based on these three factors was developed, and the "risk score" of an ICI response was calculated with the formula: "risk score" = 3.4591×H3Cit + 2.5808×IL8 + 2.0045 ×CRP- 11.3844. The cutoff point of the "risk score" was 0.528, and patients with a "risk score" lower than 0.528 were more likely to benefit from ICI treatment (AUC: 0.937, 95% CI: 0.886-0.988, with sensitivity 80.60%, specificity 91.40%). The AUC was 0.719 (95% CI: 0.600-0.837, P = 0.001), with a sensitivity of 70.00% and specificity of 65.20% in the validation cohort. CONCLUSIONS A model incorporating H3Cit, IL-8 and CRP has an excellent prediction ability for ICI response; thus, patients with a lower "risk score" selectively benefit from ICI treatment, which may have significant clinical implications for the early detection of an ICI response.
Collapse
Affiliation(s)
- Xueping Wang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Hao Huang
- Department of Laboratory Medicine, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510060, China
| | - Lin Zhang
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yaxian Wu
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Yingsheng Wen
- Department of Thoracic Surgery, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Xuezi Weng
- Department of Blood Transfusion, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Qi Chen
- Department of Blood Transfusion, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China
| | - Wanli Liu
- Department of Laboratory Medicine, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Cancer Center, Sun Yat-sen University, Guangzhou, 510060, China.
| |
Collapse
|
29
|
Xu B, Dong SY, Bai XL, Song TQ, Zhang BH, Zhou LD, Chen YJ, Zeng ZM, Wang K, Zhao HT, Lu N, Zhang W, Li XB, Zheng SS, Long G, Yang YC, Huang HS, Huang LQ, Wang YC, Liang F, Zhu XD, Huang C, Shen YH, Zhou J, Zeng MS, Fan J, Rao SX, Sun HC. Tumor Radiomic Features on Pretreatment MRI to Predict Response to Lenvatinib plus an Anti-PD-1 Antibody in Advanced Hepatocellular Carcinoma: A Multicenter Study. Liver Cancer 2023; 12:262-276. [PMID: 37601982 PMCID: PMC10433098 DOI: 10.1159/000528034] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/02/2022] [Indexed: 08/22/2023] Open
Abstract
Introduction Lenvatinib plus an anti-PD-1 antibody has shown promising antitumor effects in patients with advanced hepatocellular carcinoma (HCC), but with clinical benefit limited to a subset of patients. We developed and validated a radiomic-based model to predict objective response to this combination therapy in advanced HCC patients. Methods Patients (N = 170) who received first-line combination therapy with lenvatinib plus an anti-PD-1 antibody were retrospectively enrolled from 9 Chinese centers; 124 and 46 into the training and validation cohorts, respectively. Radiomic features were extracted from pretreatment contrast-enhanced MRI. After feature selection, clinicopathologic, radiomic, and clinicopathologic-radiomic models were built using a neural network. The performance of models, incremental predictive value of radiomic features compared with clinicopathologic features and relationship between radiomic features and survivals were assessed. Results The clinicopathologic model modestly predicted objective response with an AUC of 0.748 (95% CI: 0.656-0.840) and 0.702 (95% CI: 0.547-0.884) in the training and validation cohorts, respectively. The radiomic model predicted response with an AUC of 0.886 (95% CI: 0.815-0.957) and 0.820 (95% CI: 0.648-0.984), respectively, with good calibration and clinical utility. The incremental predictive value of radiomic features to clinicopathologic features was confirmed with a net reclassification index of 47.9% (p < 0.001) and 41.5% (p = 0.025) in the training and validation cohorts, respectively. Furthermore, radiomic features were associated with overall survival and progression-free survival both in the training and validation cohorts, but modified albumin-bilirubin grade and neutrophil-to-lymphocyte ratio were not. Conclusion Radiomic features extracted from pretreatment MRI can predict individualized objective response to combination therapy with lenvatinib plus an anti-PD-1 antibody in patients with unresectable or advanced HCC, provide incremental predictive value over clinicopathologic features, and are associated with overall survival and progression-free survival after initiation of this combination regimen.
Collapse
Affiliation(s)
- Bin Xu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - San-Yuan Dong
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Xue-Li Bai
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Tian-Qiang Song
- Department of Hepatobiliary, National Clinical Research Center of Cancer, Oncology Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Bo-Heng Zhang
- Department of Hepatic Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Le-Du Zhou
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yong-Jun Chen
- Department of Hepatobiliary Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Ming Zeng
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kui Wang
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, China
| | - Hai-Tao Zhao
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Na Lu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wei Zhang
- Department of Hepatobiliary, National Clinical Research Center of Cancer, Oncology Key Laboratory of Cancer Prevention and Therapy, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Xu-Bin Li
- Department of Radiology, National Clinical Research Center of Cancer, Tianjin Medical University Cancer Institute and Hospital, Tianjin, China
| | - Su-Su Zheng
- Department of Hepatic Oncology, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Guo Long
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Yu-Chen Yang
- Department of Hepatobiliary Surgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hua-Sheng Huang
- Department of Medical Oncology, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Lan-Qing Huang
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Navy Medical University, Shanghai, China
| | - Yun-Chao Wang
- Department of Liver Surgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Fei Liang
- Department of Biostatistics, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Xiao-Dong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Cheng Huang
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ying-Hao Shen
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Meng-Su Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Sheng-Xiang Rao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai Institute of Medical Imaging, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
30
|
Sun C, Wang Q, Hou L, Zhang R, Chen Y, Niu L. A contrast-enhanced ultrasound-based nomogram for the prediction of therapeutic efficiency of anti-PD-1 plus anti-VEGF agents in advanced hepatocellular carcinoma patients. Front Immunol 2023; 14:1229560. [PMID: 37575236 PMCID: PMC10413126 DOI: 10.3389/fimmu.2023.1229560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Accepted: 07/11/2023] [Indexed: 08/15/2023] Open
Abstract
Background There is no study focusing on noninvasive predictors for the efficacy of sintilimab (anti-PD-1) plus IBI305 (a bevacizumab biosimilar) treatment in advanced hepatocellular carcinoma (HCC). Method A total of 33 patients with advanced HCC were prospectively enrolled and received sintilimab plus IBI305 treatment from November 2018 to October 2019. Baseline characteristics including clinical data, laboratory data, and tumor features based on pretreatment CT/MR were collected. Meanwhile, pretreatment contrast-enhanced ultrasound (CEUS) for target tumor was performed and quantitative parameters were derived from time-intensity curves (TICs). A nomogram was developed based on the variables identified by the univariable and multivariable logistic regression analysis. The discrimination, calibration, and clinical utility of the nomogram were evaluated. Results Tumor embolus and grad ratio were significant variables related to the efficacy of sintilimab plus IBI305 strategy. The nomogram based on these two variables achieved an excellent predictive performance with an area under curve (AUC) of 0.909 (95% CI, 0.813-1). A bootstrapping for 500 repetitions was performed to validate this model and the AUC of the bootstrap model was 0.91 (95% CI, 0.8-0.98). The calibration curve and decision curve analysis (DCA) showed that the nomogram had a good consistency and clinical utility. Conclusions This study has established and validated a nomogram by incorporating the quantitative parameters of pretreatment CEUS and baseline clinical characteristics to predict the anti-PD-1 plus anti-VEGF treatment efficacy in advanced HCC patients.
Collapse
Affiliation(s)
- Chao Sun
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Qian Wang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lu Hou
- Department of Radiotherapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Rui Zhang
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yu Chen
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Lijuan Niu
- Department of Ultrasound, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
31
|
Dong W, Ji Y, Pi S, Chen QF. Noninvasive imaging-based machine learning algorithm to identify progressive disease in advanced hepatocellular carcinoma receiving second-line systemic therapy. Sci Rep 2023; 13:10690. [PMID: 37393336 PMCID: PMC10314898 DOI: 10.1038/s41598-023-37862-y] [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: 03/23/2023] [Accepted: 06/28/2023] [Indexed: 07/03/2023] Open
Abstract
The aim of this study was to predict tyrosine kinase inhibitors (TKI) plus anti-PD-1 antibodies (TKI-PD-1) efficacy as second-line treatment in advanced hepatocellular carcinoma (HCC) using radiomics analysis. From November 2018 to November 2019, a total of 55 patients were included. Radiomic features were obtained from the CT images before treatment and filtered using intraclass correlation coefficients (ICCs) and least absolute shrinkage and selection operator (LASSO) methods. Subsequently, ten prediction algorithms were developed and validated based on radiomic characteristics. The accuracy of the constructed model was measured through area under the receiver operating characteristic curve (AUC) analysis; survival analysis was performed via Kaplan-Meier and Cox regression analyses. Overall, 18 (32.7%) out of 55 patients had progressive disease. Through ICCs and LASSO, ten radiomic features were entered into the algorithm construction and validation. Ten machine learning algorithms showed different accuracies, with the support vector machine (SVM) model having the highest AUC value of 0.933 in the training cohort and 0.792 in the testing cohort. The radiomic features were associated with overall survival. In conclsion, the SVM algorithm is a useful method to predict TKI-PD-1 efficacy in patients with advanced HCC using images taken prior to treatment.
Collapse
Affiliation(s)
- Wei Dong
- Department of Medical Oncology, Nanyang Second People's Hospital, Nanyang, China
| | - Ye Ji
- Department of Medical Oncology, Nanyang Central Hospital, Nanyang, China
| | - Shan Pi
- Department of Radiology, The Third Affiliated Hospital, Sun Yat-Sen University, No. 600 Tianhe Road, Guangzhou, 510630, Guangdong, China.
| | - Qi-Feng Chen
- Department of Medical Imaging and Interventional Radiology, Sun Yat-Sen University Cancer Center, 651 Dongfeng Road East, Guangzhou, 510060, Guangdong, China.
- State Key Laboratory of Oncology in South China, Guangzhou, Guangdong, China.
- Collaborative Innovation Center for Cancer Medicine, Guangzhou, Guangdong, China.
| |
Collapse
|
32
|
Feng Y, Zhang H, Ren Q, Li C, Liu S, Zheng C, Xia X. Contrast-enhanced CT parameters predict short-term tumor response in patients with hepatocellular carcinoma who received sequential combined anti-angiogenesis and immune checkpoint inhibitor treatment. Eur J Radiol 2023; 162:110784. [PMID: 36958125 DOI: 10.1016/j.ejrad.2023.110784] [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: 10/16/2022] [Revised: 03/06/2023] [Accepted: 03/13/2023] [Indexed: 03/18/2023]
Abstract
PURPOSE To evaluate whether relative Hounsfield unit attenuation index (rHUAI) on contrast-enhanced computed tomography (CECT) can predict tumor response in advanced hepatocellular carcinoma (HCC) patients who received sequential combined treatment of immune checkpoint inhibitor (ICI) and anti-angiogenesis therapy. METHOD One hundred seventeen advanced HCC patients who underwent the sequential combined treatment in a tertiary hospital between March 2020 and December 2021 were allocated to prediction and validation cohorts (with a ratio of 2:1) based on the time of initial ICI treatment. rHUAI from the arterial to the portal-venous phase (rHU_ap) and from the portal-venous to the delayed phase (rHU_pd) was calculated. The optimal cut-off values (COVs) of rHU_ap and rHU_pd for predicting tumor response were identified using Youden's index. Univariate and multivariable analyses were performed to assess the relationship between the COVs and tumor response. The validity of COVs was verified in the validation cohort using the chi-square test and Cramer's V coefficient (V). RESULTS The optimal COVs of the two observers were 0.5316 and 0.3265 for rHU_ap, and -0.0208 and -0.0048 for rHU_pd, respectively. Multivariable analysis suggested that the COVs were independently associated with tumor response in the prediction cohort (rHU_ap, Odds ratio: 7.727 and 7.808, 95 % CI: 2.516-23.728 and 2.399-25.410, p value < 0.001 and 0.001; rHU_pd, Odds ratio: 0.034 and 0.011, 95 % CI: 0.002-0.600 and 0.001-0.209, p value of 0.021 and 0.003). In the validation cohort, the optimal COVs of rHU_ap had a moderate to a strong association with tumor response (V = 0.362-0.545, p < 0.05). The association between COVs of rHU_pd and tumor response was slight to strong (V = 0.24-0.545, p = 0.001 to 0.134). CONCLUSION rHUAI obtained from CECT has the potential as a non-invasive tool for predicting tumor response in advanced HCC patients who have received combined ICI and anti-angiogenesis treatment.
Collapse
Affiliation(s)
- Yiming Feng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Hui Zhang
- Department of Internal Medicine, Wuhan Hankou Hospital, 172 Zhaojiatiao Road, Wuhan City, Hubei Province 430011, China
| | - Qianqian Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Changde Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Song Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China
| | - Xiangwen Xia
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; Hubei Province Key Laboratory of Molecular Imaging, Wuhan 430022, China.
| |
Collapse
|
33
|
Wei Y, Yang M, Xu L, Liu M, Zhang F, Xie T, Cheng X, Wang X, Che F, Li Q, Xu Q, Huang Z, Liu M. Novel Computed-Tomography-Based Transformer Models for the Noninvasive Prediction of PD-1 in Pre-Operative Settings. Cancers (Basel) 2023; 15:cancers15030658. [PMID: 36765615 PMCID: PMC9913645 DOI: 10.3390/cancers15030658] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 01/05/2023] [Accepted: 01/12/2023] [Indexed: 01/24/2023] Open
Abstract
The expression status of programmed cell death protein 1 (PD-1) in patients with hepatocellular carcinoma (HCC) is associated with the checkpoint blockade treatment responses of PD-1/PD-L1. Thus, accurately and preoperatively identifying the status of PD-1 has great clinical implications for constructing personalized treatment strategies. To investigate the preoperative predictive value of the transformer-based model for identifying the status of PD-1 expression, 93 HCC patients with 75 training cohorts (2859 images) and 18 testing cohorts (670 images) were included. We propose a transformer-based network architecture, ResTransNet, that efficiently employs convolutional neural networks (CNNs) and self-attention mechanisms to automatically acquire a persuasive feature to obtain a prediction score using a nonlinear classifier. The area under the curve, receiver operating characteristic curve, and decision curves were applied to evaluate the prediction model's performance. Then, Kaplan-Meier survival analyses were applied to evaluate the overall survival (OS) and recurrence-free survival (RFS) in PD-1-positive and PD-1-negative patients. The proposed transformer-based model obtained an accuracy of 88.2% with a sensitivity of 88.5%, a specificity of 88.9%, and an area under the curve of 91.1% in the testing cohort.
Collapse
Affiliation(s)
- Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Meiyi Yang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Lifeng Xu
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou 324000, China
| | - Minghui Liu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Feng Zhang
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou 324000, China
| | - Tianshu Xie
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610000, China
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324000, China
| | - Xuan Cheng
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Xiaomin Wang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 610000, China
| | - Feng Che
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Qing Xu
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu 610000, China
| | - Zixing Huang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610000, China
- Correspondence: (Z.H.); (M.L.)
| | - Ming Liu
- Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou 324000, China
- Correspondence: (Z.H.); (M.L.)
| |
Collapse
|
34
|
Miranda J, Horvat N, Fonseca GM, Araujo-Filho JDAB, Fernandes MC, Charbel C, Chakraborty J, Coelho FF, Nomura CH, Herman P. Current status and future perspectives of radiomics in hepatocellular carcinoma. World J Gastroenterol 2023; 29:43-60. [PMID: 36683711 PMCID: PMC9850949 DOI: 10.3748/wjg.v29.i1.43] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/21/2022] [Revised: 10/27/2022] [Accepted: 12/14/2022] [Indexed: 01/04/2023] Open
Abstract
Given the frequent co-existence of an aggressive tumor and underlying chronic liver disease, the management of hepatocellular carcinoma (HCC) patients requires experienced multidisciplinary team discussion. Moreover, imaging plays a key role in the diagnosis, staging, restaging, and surveillance of HCC. Currently, imaging assessment of HCC entails the assessment of qualitative characteristics which are prone to inter-reader variability. Radiomics is an emerging field that extracts high-dimensional mineable quantitative features that cannot be assessed visually with the naked eye from medical imaging. The main potential applications of radiomic models in HCC are to predict histology, response to treatment, genetic signature, recurrence, and survival. Despite the encouraging results to date, there are challenges and limitations that need to be overcome before radiomics implementation in clinical practice. The purpose of this article is to review the main concepts and challenges pertaining to radiomics, and to review recent studies and potential applications of radiomics in HCC.
Collapse
Affiliation(s)
- Joao Miranda
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-010, Brazil
| | - Natally Horvat
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | | | - Maria Clara Fernandes
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Charlotte Charbel
- Department of Radiology, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | - Jayasree Chakraborty
- Department of Surgery, Memorial Sloan-Kettering Cancer Center, New York, NY 10065, United States
| | | | - Cesar Higa Nomura
- Department of Radiology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| | - Paulo Herman
- Department of Gastroenterology, University of Sao Paulo, Sao Paulo 05403-000, Brazil
| |
Collapse
|
35
|
Tao YY, Shi Y, Gong XQ, Li L, Li ZM, Yang L, Zhang XM. Radiomic Analysis Based on Magnetic Resonance Imaging for Predicting PD-L2 Expression in Hepatocellular Carcinoma. Cancers (Basel) 2023; 15:365. [PMID: 36672315 PMCID: PMC9856314 DOI: 10.3390/cancers15020365] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/01/2023] [Accepted: 01/03/2023] [Indexed: 01/09/2023] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignant tumour and the third leading cause of cancer death in the world. The emerging field of radiomics involves extracting many clinical image features that cannot be recognized by the human eye to provide information for precise treatment decision making. Radiomics has shown its importance in HCC identification, histological grading, microvascular invasion (MVI) status, treatment response, and prognosis, but there is no report on the preoperative prediction of programmed death ligand-2 (PD-L2) expression in HCC. The purpose of this study was to investigate the value of MRI radiomic features for the non-invasive prediction of immunotherapy target PD-L2 expression in hepatocellular carcinoma (HCC). A total of 108 patients with HCC confirmed by pathology were retrospectively analysed. Immunohistochemical analysis was used to evaluate the expression level of PD-L2. 3D-Slicer software was used to manually delineate volumes of interest (VOIs) and extract radiomic features on preoperative T2-weighted, arterial-phase, and portal venous-phase MR images. Least absolute shrinkage and selection operator (LASSO) was performed to find the best radiomic features. Multivariable logistic regression models were constructed and validated using fivefold cross-validation. The area under the receiver characteristic curve (AUC) was used to evaluate the predictive performance of each model. The results show that among the 108 cases of HCC, 50 cases had high PD-L2 expression, and 58 cases had low PD-L2 expression. Radiomic features correlated with PD-L2 expression. The T2-weighted, arterial-phase, and portal venous-phase and combined MRI radiomics models showed AUCs of 0.789 (95% CI: 0.702-0.875), 0.727 (95% CI: 0.632-0.823), 0.770 (95% CI: 0.682-0.875), and 0.871 (95% CI: 0.803-0.939), respectively. The combined model showed the best performance. The results of this study suggest that prediction based on the radiomic characteristics of MRI could noninvasively predict the expression of PD-L2 in HCC before surgery and provide a reference for the selection of immune checkpoint blockade therapy.
Collapse
Affiliation(s)
- Yun-Yun Tao
- Medical Imaging Key Laboratory of Sichuan Province, Interventional Medical Center, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yue Shi
- Medical Imaging Key Laboratory of Sichuan Province, Interventional Medical Center, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Xue-Qin Gong
- Medical Imaging Key Laboratory of Sichuan Province, Interventional Medical Center, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Li Li
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Zu-Mao Li
- Department of Pathology, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Interventional Medical Center, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Interventional Medical Center, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| |
Collapse
|
36
|
Fahmy D, Alksas A, Elnakib A, Mahmoud A, Kandil H, Khalil A, Ghazal M, van Bogaert E, Contractor S, El-Baz A. The Role of Radiomics and AI Technologies in the Segmentation, Detection, and Management of Hepatocellular Carcinoma. Cancers (Basel) 2022; 14:cancers14246123. [PMID: 36551606 PMCID: PMC9777232 DOI: 10.3390/cancers14246123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/08/2022] [Accepted: 12/09/2022] [Indexed: 12/15/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common primary hepatic neoplasm. Thanks to recent advances in computed tomography (CT) and magnetic resonance imaging (MRI), there is potential to improve detection, segmentation, discrimination from HCC mimics, and monitoring of therapeutic response. Radiomics, artificial intelligence (AI), and derived tools have already been applied in other areas of diagnostic imaging with promising results. In this review, we briefly discuss the current clinical applications of radiomics and AI in the detection, segmentation, and management of HCC. Moreover, we investigate their potential to reach a more accurate diagnosis of HCC and to guide proper treatment planning.
Collapse
Affiliation(s)
- Dalia Fahmy
- Diagnostic Radiology Department, Mansoura University Hospital, Mansoura 35516, Egypt
| | - Ahmed Alksas
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Ahmed Elnakib
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Ali Mahmoud
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
| | - Heba Kandil
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
- Faculty of Computer Sciences and Information, Mansoura University, Mansoura 35516, Egypt
| | - Ashraf Khalil
- College of Technological Innovation, Zayed University, Abu Dhabi 4783, United Arab Emirates
| | - Mohammed Ghazal
- Electrical, Computer, and Biomedical Engineering Department, Abu Dhabi University, Abu Dhabi 59911, United Arab Emirates
| | - Eric van Bogaert
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA
| | - Sohail Contractor
- Department of Radiology, University of Louisville, Louisville, KY 40202, USA
| | - Ayman El-Baz
- Bioengineering Department, University of Louisville, Louisville, KY 40292, USA
- Correspondence:
| |
Collapse
|
37
|
Value of T2-weighted-based radiomics model in distinguishing Warthin tumor from pleomorphic adenoma of the parotid. Eur Radiol 2022; 33:4453-4463. [PMID: 36502461 DOI: 10.1007/s00330-022-09295-0] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2022] [Revised: 11/01/2022] [Accepted: 11/09/2022] [Indexed: 12/14/2022]
Abstract
OBJECTIVES The differentiation of Warthin tumor and pleomorphic adenoma before treatment is crucial for clinical strategies. The aim of this study was to develop and test a T2-weighted-based radiomics model for differentiating pleomorphic adenoma from Warthin tumor of the parotid gland. METHODS A total of 117 patients, including 61 cases of Warthin tumor and 56 cases of pleomorphic adenoma, were retrospectively enrolled from two centers between January 2010 and June 2022. The training set included 82 cases, and the validation set included 35 cases. From T2-weighted images, 971 radiomics features were extracted. Seven radiomics features remained after a two-step selection process. We used the seven radiomics features and clinical factors through multivariable logistic regression to build radiomics and clinical models, respectively. A radiomics-clinical model was also built that combined the independent clinical predictors with the radiomics features. Through ROC curves, the three models were evaluated and compared. RESULTS In the radiomics model, AUCs were 0.826 and 0.796 in training and validation sets, respectively. In the clinical model, the AUCs were 0.923 and 0.926 in the training and validation sets, respectively. Decision curve analysis revealed that the radiomics-clinical model had the best diagnostic performance for distinguishing Warthin tumor from pleomorphic adenoma of the parotid gland (AUC = 0.962 and 0.934 for the training and validation sets, respectively). CONCLUSION The radiomics-clinical model performed well in differentiating pleomorphic adenoma from Warthin tumor of the parotid gland. KEY POINTS • The clinical model outperformed the radiomics model in distinguishing pleomorphic adenoma from Warthin tumor of the parotid gland. • The radiomics features extracted from T2-weighted images could help differentiate pleomorphic adenoma from Warthin tumor of the parotid gland. • The radiomics-clinical model was superior to the radiomics and the clinical models for differentiating pleomorphic adenoma from Warthin tumor of the parotid gland.
Collapse
|
38
|
Li X, Sun W, Ding X, Li W, Chen J. Prognostic model of immune checkpoint inhibitors combined with anti-angiogenic agents in unresectable hepatocellular carcinoma. Front Immunol 2022; 13:1060051. [PMID: 36532029 PMCID: PMC9751696 DOI: 10.3389/fimmu.2022.1060051] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 11/21/2022] [Indexed: 12/03/2022] Open
Abstract
Background The combination of immune checkpoint inhibitors (ICIs) and anti-angiogenic agents has shown promising efficacy in unresectable hepatocellular carcinoma (HCC), but until now no clinical prognostic models or predictive biomarkers have been established. Methods From 2016 to 2021, a total of 258 HCCs treated with ICIs and tyrosine kinase inhibitors (TKIs) were retrospectively enrolled, as the study cohort. Patients' baseline data was extracted by least absolute and shrinkage selection operator (LASSO) and Cox regression. Finally, a prognostic model in the form of nomogram was developed. Model performance was assessed in terms of discrimination, calibration, and clinical utility. A 5-fold cross-validation was used to evaluate the internal repeatability of the model. In addition, the patient cohort was divided into three subgroups according to nomogram scores. Their survivals were estimated by Kaplan-Meier methods and the differences were analyzed using log-rank tests. Results Seven clinical parameters were selected: Eastern Cooperative Oncology Group performance status (ECOG PS), combination of transarterial chemoembolization (TACE), extrahepatic metastasis (EHM), platelet to lymphocyte ratio (PLR), alanine aminotransferase (ALT), alpha-fetoprotein (AFP), and Child-Pugh score. The model had an area under the curve (AUC) of 0.777 at 1 year and 0.772 at 2 years. Receiver operating characteristic (ROC) curve, calibration curve and decision curve analysis (DCA) showed that the discrimination, consistency and applicability of the model were good. In addition, cross-validation validated the discrimination of the model, and the C index value of the model is 0.7405. The median overall survival (OS) of the high-, medium- and low-risk subgroups was 7.58, 17.50 and 53.17 months, respectively, with a significant difference between the groups (P < 0.0001). Conclusion We developed a comprehensive and simple prognostic model for the combination of ICIs plus TKIs. And it may predict the efficacy of the combination regimen for unresectable HCC.
Collapse
Affiliation(s)
| | | | | | - Wei Li
- *Correspondence: Jinglong Chen, ; Wei Li,
| | | |
Collapse
|
39
|
Ter Maat LS, van Duin IAJ, Elias SG, van Diest PJ, Pluim JPW, Verhoeff JJC, de Jong PA, Leiner T, Veta M, Suijkerbuijk KPM. Imaging to predict checkpoint inhibitor outcomes in cancer. A systematic review. Eur J Cancer 2022; 175:60-76. [PMID: 36096039 DOI: 10.1016/j.ejca.2022.07.034] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2022] [Revised: 07/17/2022] [Accepted: 07/21/2022] [Indexed: 11/03/2022]
Abstract
BACKGROUND Checkpoint inhibition has radically improved the perspective for patients with metastatic cancer, but predicting who will not respond with high certainty remains difficult. Imaging-derived biomarkers may be able to provide additional insights into the heterogeneity in tumour response between patients. In this systematic review, we aimed to summarise and qualitatively assess the current evidence on imaging biomarkers that predict response and survival in patients treated with checkpoint inhibitors in all cancer types. METHODS PubMed and Embase were searched from database inception to 29th November 2021. Articles eligible for inclusion described baseline imaging predictive factors, radiomics and/or imaging machine learning models for predicting response and survival in patients with any kind of malignancy treated with checkpoint inhibitors. Risk of bias was assessed using the QUIPS and PROBAST tools and data was extracted. RESULTS In total, 119 studies including 15,580 patients were selected. Of these studies, 73 investigated simple imaging factors. 45 studies investigated radiomic features or deep learning models. Predictors of worse survival were (i) higher tumour burden, (ii) presence of liver metastases, (iii) less subcutaneous adipose tissue, (iv) less dense muscle and (v) presence of symptomatic brain metastases. Hazard rate ratios did not exceed 2.00 for any predictor in the larger and higher quality studies. The added value of baseline fluorodeoxyglucose positron emission tomography parameters in predicting response to treatment was limited. Pilot studies of radioactive drug tracer imaging showed promising results. Reports on radiomics were almost unanimously positive, but numerous methodological concerns exist. CONCLUSIONS There is well-supported evidence for several imaging biomarkers that can be used in clinical decision making. Further research, however, is needed into biomarkers that can more accurately identify which patients who will not benefit from checkpoint inhibition. Radiomics and radioactive drug labelling appear to be promising approaches for this purpose.
Collapse
Affiliation(s)
- Laurens S Ter Maat
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Isabella A J van Duin
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Sjoerd G Elias
- Department of Epidemiology, Julius Center for Health Sciences and Primary Care, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Paul J van Diest
- Department of Pathology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Josien P W Pluim
- Image Science Institute, University Medical Center Utrecht, Utrecht, the Netherlands; Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Joost J C Verhoeff
- Department of Radiotherapy, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Pim A de Jong
- Department of Radiology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Tim Leiner
- Utrecht University, Utrecht, the Netherlands; Department of Radiology, Mayo Clinical, Rochester, MN, USA
| | - Mitko Veta
- Medical Image Analysis, Department Biomedical Engineering, Eindhoven University of Technology, Eindhoven, the Netherlands; Utrecht University, Utrecht, the Netherlands
| | - Karijn P M Suijkerbuijk
- Department of Medical Oncology, University Medical Center Utrecht, Utrecht, the Netherlands; Utrecht University, Utrecht, the Netherlands.
| |
Collapse
|
40
|
Miao L, Ma ST, Jiang X, Zhang HH, Wang YM, Li M. Prediction of the therapeutic efficacy of epirubicin combined with ifosfamide in patients with lung metastases from soft tissue sarcoma based on contrast-enhanced CT radiomics features. BMC Med Imaging 2022; 22:131. [PMID: 35883116 PMCID: PMC9316811 DOI: 10.1186/s12880-022-00859-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2022] [Accepted: 07/18/2022] [Indexed: 11/24/2022] Open
Abstract
Objective To investigate the value of contrast-enhanced computed tomography (CECT) radiomics features in predicting the efficacy of epirubicin combined with ifosfamide in patients with pulmonary metastases from soft tissue sarcoma. Methods A retrospective analysis of 51 patients with pulmonary metastases from soft tissue sarcoma who received the chemotherapy regimen of epirubicin combined with ifosfamide was performed, and efficacy was evaluated by Recist1.1. ROIs (1 or 2) were selected for each patient. Lung metastases were used as target lesions (86 target lesions total), and the patients were divided into a progression group (n = 29) and a non-progressive group (n = 57); the latter included a stable group (n = 34) and a partial response group (n = 23). Information on lung metastases was extracted from CECT images before chemotherapy, and all lesions were delineated by ITK-SNAP software manually or semiautomatically. The decision tree classifier had a better performance in all radiomics models. A receiver operating characteristic curve was plotted to evaluate the predictive performance of the radiomics model. Results In total, 851 CECT radiomics features were extracted for each target lesion and finally reduced to 2 radiomics features, which were then used to construct a radiomics model. Areas under the curves of the model for predicting lesion progression were 0.917 and 0.856 in training and testing groups, respectively. Conclusion The model established based on the radiomics features of CECT before treatment has certain predictive value for assessing the efficacy of chemotherapy for patients with soft tissue sarcoma lung metastases. Supplementary Information The online version contains supplementary material available at 10.1186/s12880-022-00859-6.
Collapse
Affiliation(s)
- Lei Miao
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Shu-Tao Ma
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, No. 17, Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Xu Jiang
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Huan-Huan Zhang
- 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China
| | - Yan-Mei Wang
- GE Healthcare China, Pudong New Town, Shanghai, People's Republic of China
| | - Meng 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, No. 17 Panjiayuan Nanli, Chaoyang District, Beijing, 100021, China.
| |
Collapse
|
41
|
Zhang Y, Zou J, Chen R. An M0 macrophage-related prognostic model for hepatocellular carcinoma. BMC Cancer 2022; 22:791. [PMID: 35854246 PMCID: PMC9294844 DOI: 10.1186/s12885-022-09872-y] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Accepted: 07/04/2022] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND The role of M0 macrophages and their related genes in the prognosis of hepatocellular carcinoma (HCC) remains poorly characterized. METHODS Multidimensional bioinformatic methods were used to construct a risk score model using M0 macrophage-related genes (M0RGs). RESULTS Infiltration of M0 macrophages was significantly higher in HCC tissues than in normal liver tissues (P = 2.299e-07). Further analysis revealed 35 M0RGs that were associated with HCC prognosis; two M0RGs (OLA1 and ATIC) were constructed and validated as a prognostic signature for overall survival of patients with HCC. Survival analysis revealed the positive relationship between the M0RG signature and unfavorable prognosis. Correlation analysis showed that this risk model had positive associations with clinicopathological characteristics, somatic gene mutations, immune cell infiltration, immune checkpoint inhibitor targets, and efficacy of common drugs. CONCLUSIONS The constructed M0RG-based risk model may be promising for the clinical prediction of prognoses and therapeutic responses in patients with HCC.
Collapse
Affiliation(s)
- Yiya Zhang
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Dermatology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Hunan Key Laboratory of Aging Biology, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ju Zou
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.,Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China
| | - Ruochan Chen
- Hunan Key Laboratory of Viral Hepatitis, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China. .,Department of Infectious Disease, Xiangya Hospital, Central South University, Changsha, 410008, Hunan, China.
| |
Collapse
|
42
|
Yuan G, Xie F, Song Y, Li Q, Li R, Hu X, Zang M, Cheng X, Lu G, Huang J, Fan W, Rong X, Sun J, Chen J. Hepatic Tumor Stiffness Measured by Shear Wave Elastography Is Prognostic for HCC Progression Following Treatment With Anti-PD-1 Antibodies Plus Lenvatinib: A Retrospective Analysis of Two Independent Cohorts. Front Immunol 2022; 13:868809. [PMID: 35757765 PMCID: PMC9218245 DOI: 10.3389/fimmu.2022.868809] [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: 02/03/2022] [Accepted: 05/03/2022] [Indexed: 12/24/2022] Open
Abstract
Background The clinical significance of liver stiffness (LS) measured by shear wave elastography (SWE) in programmed cell death protein-1 (PD-1) inhibitors treated advanced hepatocellular carcinoma (HCC) patients remains unknown. This study aimed to explore the prognostic value of baseline LS by SWE prior to PD-1 inhibitor treatment in combination with lenvatinib. Methods We retrospectively evaluated patients (n=133) with HCC who received anti-PD-1 antibodies plus lenvatinib at two high-volume medical centres, between January 2020 and June 2021. Univariate and multivariate logistic regression analysis were used to develop a novel nomogram. RNA sequencing and immunohistochemical staining were used to assess the heterogeneity of biological and immune characteristics associated with tumor stiffness. Results The objective response rate (ORR) and disease control rate (DCR) of the whole population were 23.4% and 72.2%, respectively. A LS value of the baseline tumorous foci of 19.53 kPa had the maximum sum of sensitivity and specificity, making it the optimal cut-off value for predicting PD-1 inhibitor efficacy. The nomogram comprised baseline tumor LS and albumin-bilirubin grade (ALBI), which provided favorable calibration and discrimination in the training dataset with an AUC of 0.840 (95%CI: 0.750-0.931) and a C-index of 0.828. Further, it showed acceptable discrimination in the validation cohort, with an AUC of 0.827 (95%CI: 0.673-0.980) and C-index of 0.803. The differentially expressed genes enriched in high stiffness tumors were predominantly associated with metabolic pathways, while those enriched in low stiffness tumors were related to DNA damage repair. Furthermore, patients with high stiffness tumors had a relatively lower infiltration of immune cells and histone deacetylase pathway inhibitors were identified as candidate drugs to promote the efficacy of immunotherapy. Conclusions Baseline LS value of tumorous foci by SWE—that is, before administration of a PD-1 inhibitor in combination with lenvatinib—is a convenient predictor of PD-1 inhibitor efficacy in patients with advanced HCC, which has potential to be used for pretreatment stratification to optimize treatment of advanced HCC.
Collapse
Affiliation(s)
- Guosheng Yuan
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Fuli Xie
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yangda Song
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Qi Li
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Rong Li
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiaoyun Hu
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Mengya Zang
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Xiao Cheng
- Department of Hepatology, Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Guanting Lu
- Department of Hepatology, Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jing Huang
- Department of Hepatology, Zengcheng Branch, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoxiang Rong
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jian Sun
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Jinzhang Chen
- Department of Infectious Diseases and Hepatology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| |
Collapse
|
43
|
When artificial intelligence meets PD-1/PD-L1 inhibitors: Population screening, response prediction and efficacy evaluation. Comput Biol Med 2022; 145:105499. [DOI: 10.1016/j.compbiomed.2022.105499] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2022] [Revised: 03/26/2022] [Accepted: 04/03/2022] [Indexed: 02/07/2023]
|
44
|
Liu ZP, Chen WY, Wang ZR, Liu XC, Fan HN, Xu L, Pan Y, Zhong SY, Xie D, Bai J, Jiang Y, Zhang YQ, Dai HS, Chen ZY. Development and Validation of a Prognostic Model to Predict Recurrence-Free Survival After Curative Resection for Perihilar Cholangiocarcinoma: A Multicenter Study. Front Oncol 2022; 12:849053. [PMID: 35530316 PMCID: PMC9071302 DOI: 10.3389/fonc.2022.849053] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2022] [Accepted: 03/24/2022] [Indexed: 12/29/2022] Open
Abstract
Background Recurrence is the main cause of death in perihilar cholangiocarcinoma (pCCA) patients after surgery. Identifying patients with a high risk of recurrence is important for decision-making regarding neoadjuvant therapy to improve long-term outcomes. Aim The objective of this study was to develop and validate a prognostic model to predict recurrence-free survival (RFS) after curative resection of pCCA. Methods Patients following curative resection for pCCA from January 2008 to January 2016 were identified from a multicenter database. Using random assignment, 70% of patients were assigned to the training cohort, and the remaining 30% were assigned to the validation cohort. Independent predictors of RFS after curative resection for pCCA were identified and used to construct a prognostic model. The predictive performance of the model was assessed using calibration curves and the C-index. Results A total of 341 patients were included. The median overall survival (OS) was 22 months, and the median RFS was 14 months. Independent predictors associated with RFS included lymph node involvement, macrovascular invasion, microvascular invasion, maximum tumor size, tumor differentiation, and carbohydrate antigen 19-9. The model incorporating these factors to predict 1-year RFS demonstrated better calibration and better performance than the 8th American Joint Committee on Cancer (AJCC) staging system in both the training and validation cohorts (C-indexes: 0.723 vs. 0.641; 0.743 vs. 0.607). Conclusions The prognostic model could identify patients at high risk of recurrence for pCCA to inform patients and surgeons, help guide decision-making for postoperative adjuvant therapy, and improve survival.
Collapse
Affiliation(s)
- Zhi-Peng Liu
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Wei-Yue Chen
- Department of Clinical Research Institute, Lishui Hospital of Zhejiang University, Lishui, China
| | - Zi-Ran Wang
- Department of General Surgery, 903rd Hospital of People’s Liberation Army, Hangzhou, China
| | - Xing-Chao Liu
- Department of Hepatobiliary Surgery, Sichuan Provincial People’s Hospital, Chengdu, China
| | - Hai-Ning Fan
- Department of Hepatobiliary Surgery, Affiliated Hospital of Qinghai University, Xining, China
| | - Lei Xu
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yu Pan
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shi-Yun Zhong
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Dan Xie
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jie Bai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Jiang
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan-Qi Zhang
- Department of Health Statistics, College of Military Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Hai-Su Dai
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Zhi-Yu Chen, ; Hai-Su Dai,
| | - Zhi-Yu Chen
- Department of Hepatobiliary Surgery, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- *Correspondence: Zhi-Yu Chen, ; Hai-Su Dai,
| |
Collapse
|
45
|
Xiang YJ, Wang K, Zheng YT, Feng S, Yu HM, Li XW, Cheng X, Cheng YQ, Feng JK, Zhou LP, Meng Y, Zhai J, Shan YF, Cheng SQ. Effects of Stereotactic Body Radiation Therapy Plus PD-1 Inhibitors for Patients With Transarterial Chemoembolization Refractory. Front Oncol 2022; 12:839605. [PMID: 35387113 PMCID: PMC8978966 DOI: 10.3389/fonc.2022.839605] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2021] [Accepted: 03/01/2022] [Indexed: 12/31/2022] Open
Abstract
Background and Aims Patients with intermediate-stage hepatocellular carcinoma (HCC) who are refractory to transarterial chemoembolization (TACE) have a poor prognosis. This study aimed to explore whether stereotactic body radiation therapy (SBRT) combined with PD-1 inhibitors could improve the clinical outcomes of such patients. Methods This retrospective cohort study included patients with intermediate-stage HCC who were diagnosed with TACE refractoriness between January 2019 and December 2020 in the Eastern Hepatobiliary Surgery Hospital and the First Affiliated Hospital of Wenzhou Medical University. The patients were divided into two groups: (1) those who switched from TACE to receive stereotactic body radiotherapy (SBRT) combined with PD-1 inhibitors; (2) those who continued TACE treatment and added PD-1 inhibitors. Progression-free survival (PFS), overall survival (OS), and tumour response were assessed in both groups after becoming refractory to TACE treatment. Results Of the seventy-six patients included in this study, the median PFS was 19.6 months in the SBRT-IO group (n=31) and 10.1 months in the TACE-IO group (n=45, p<0.05). The SBRT-IO group also had a significantly higher OS than the TACE-IO group (p<0.05). The objective response rate (ORR) and disease control rate (DCR) were also better in the SBRT-IO group (ORR, 71.0% vs. 15.6%, OR=8.483, 95% CI 3.319-21.680, P < 0.001; DCR, 80.6% vs. 31.1%, OR=9.226, 95% CI 3.096-27.493, P < 0.001). Conclusions SBRT combined with a PD-1 inhibitor improves PFS and OS in TACE-refractory patients with intermediate-stage HCC. Therefore, this therapy is a suitable option in cases of TACE treatment failure.
Collapse
Affiliation(s)
- Yan-Jun Xiang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Kang Wang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yi-Tao Zheng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shuang Feng
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hong-Ming Yu
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Xiao-Wei Li
- Department II of Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Xi Cheng
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yu-Qiang Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jin-Kai Feng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Li-Ping Zhou
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yan Meng
- Department of Radiotherapy, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Jian Zhai
- Department II of Interventional Radiology, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Yun-Feng Shan
- Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.,Department of Hepatobiliary Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
46
|
Granata V, Fusco R, Setola SV, Simonetti I, Cozzi D, Grazzini G, Grassi F, Belli A, Miele V, Izzo F, Petrillo A. An update on radiomics techniques in primary liver cancers. Infect Agent Cancer 2022; 17:6. [PMID: 35246207 PMCID: PMC8897888 DOI: 10.1186/s13027-022-00422-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2022] [Accepted: 02/28/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Radiomics is a progressing field of research that deals with the extraction of quantitative metrics from medical images. Radiomic features detention indirectly tissue features such as heterogeneity and shape and can, alone or in combination with demographic, histological, genomic, or proteomic data, be used for decision support system in clinical setting. METHODS This article is a narrative review on Radiomics in Primary Liver Cancers. Particularly, limitations and future perspectives are discussed. RESULTS In oncology, assessment of tissue heterogeneity is of particular interest: genomic analysis have demonstrated that the degree of tumour heterogeneity is a prognostic determinant of survival and an obstacle to cancer control. Therefore, that Radiomics could support cancer detection, diagnosis, evaluation of prognosis and response to treatment, so as could supervise disease status in hepatocellular carcinoma (HCC) and Intrahepatic Cholangiocarcinoma (ICC) patients. Radiomic analysis is a convenient radiological image analysis technique used to support clinical decisions as it is able to provide prognostic and / or predictive biomarkers that allow a fast, objective and repeatable tool for disease monitoring. CONCLUSIONS Although several studies have shown that this analysis is very promising, there is little standardization and generalization of the results, which limits the translation of this method into the clinical context. The limitations are mainly related to the evaluation of data quality, repeatability, reproducibility, overfitting of the model. TRIAL REGISTRATION Not applicable.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Via Mariano Semmola 80131, Naples, Italy.
| | | | - Sergio Venazio Setola
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Via Mariano Semmola 80131, Naples, Italy
| | - Igino Simonetti
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Via Mariano Semmola 80131, Naples, Italy
| | - Diletta Cozzi
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via Della Signora 2, 20122, Milan, Italy
| | - Giulia Grazzini
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via Della Signora 2, 20122, Milan, Italy
| | - Francesca Grassi
- Division of Radiology, "Università Degli Studi Della Campania Luigi Vanvitelli", Naples, Italy
| | - Andrea Belli
- Division of Hepatobiliary Surgical Oncology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", 80131, Naples, Italy
| | - Vittorio Miele
- Department of Radiology, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy.,Italian Society of Medical and Interventional Radiology (SIRM), SIRM Foundation, Via Della Signora 2, 20122, Milan, Italy
| | - Francesco Izzo
- Division of Hepatobiliary Surgical Oncology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", 80131, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, "Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli", Via Mariano Semmola 80131, Naples, Italy
| |
Collapse
|
47
|
Wei J, Niu M, Yabo O, Zhou Y, Ma X, Yang X, Jiang H, Hui H, Cao H, Duan B, Li H, Ding D, Tian J. Advances in artificial intelligence techniques drive the application of radiomics in the clinical research of hepatocellular carcinoma. ILIVER 2022; 1:49-54. [DOI: 10.1016/j.iliver.2022.02.005] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
48
|
Gong XQ, Tao YY, Wu Y, Liu N, Yu X, Wang R, Zheng J, Liu N, Huang XH, Li JD, Yang G, Wei XQ, Yang L, Zhang XM. Progress of MRI Radiomics in Hepatocellular Carcinoma. Front Oncol 2021; 11:698373. [PMID: 34616673 PMCID: PMC8488263 DOI: 10.3389/fonc.2021.698373] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 08/31/2021] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the sixth most common cancer in the world and the third leading cause of cancer-related death. Although the diagnostic scheme of HCC is currently undergoing refinement, the prognosis of HCC is still not satisfactory. In addition to certain factors, such as tumor size and number and vascular invasion displayed on traditional imaging, some histopathological features and gene expression parameters are also important for the prognosis of HCC patients. However, most parameters are based on postoperative pathological examinations, which cannot help with preoperative decision-making. As a new field, radiomics extracts high-throughput imaging data from different types of images to build models and predict clinical outcomes noninvasively before surgery, rendering it a powerful aid for making personalized treatment decisions preoperatively. OBJECTIVE This study reviewed the workflow of radiomics and the research progress on magnetic resonance imaging (MRI) radiomics in the diagnosis and treatment of HCC. METHODS A literature review was conducted by searching PubMed for search of relevant peer-reviewed articles published from May 2017 to June 2021.The search keywords included HCC, MRI, radiomics, deep learning, artificial intelligence, machine learning, neural network, texture analysis, diagnosis, histopathology, microvascular invasion, surgical resection, radiofrequency, recurrence, relapse, transarterial chemoembolization, targeted therapy, immunotherapy, therapeutic response, and prognosis. RESULTS Radiomics features on MRI can be used as biomarkers to determine the differential diagnosis, histological grade, microvascular invasion status, gene expression status, local and systemic therapeutic responses, and prognosis of HCC patients. CONCLUSION Radiomics is a promising new imaging method. MRI radiomics has high application value in the diagnosis and treatment of HCC.
Collapse
Affiliation(s)
- Xue-Qin Gong
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yun-Yun Tao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Yao–Kun Wu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ning Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xi Yu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Ran Wang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing Zheng
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Nian Liu
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Hua Huang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Jing-Dong Li
- Department of Hepatocellular Surgery, Institute of Hepato-Biliary-Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Gang Yang
- Department of Hepatocellular Surgery, Institute of Hepato-Biliary-Intestinal Disease, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Qin Wei
- School of Medical Imaging, North Sichuan Medical College, Nanchong, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Xiao-Ming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Medical Research Center, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| |
Collapse
|
49
|
Granata V, Grassi R, Fusco R, Belli A, Cutolo C, Pradella S, Grazzini G, La Porta M, Brunese MC, De Muzio F, Ottaiano A, Avallone A, Izzo F, Petrillo A. Diagnostic evaluation and ablation treatments assessment in hepatocellular carcinoma. Infect Agent Cancer 2021; 16:53. [PMID: 34281580 PMCID: PMC8287696 DOI: 10.1186/s13027-021-00393-0] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Accepted: 07/06/2021] [Indexed: 02/07/2023] Open
Abstract
This article provides an overview of diagnostic evaluation and ablation treatment assessment in Hepatocellular Carcinoma (HCC). Only studies, in the English language from January 2010 to January 202, evaluating the diagnostic tools and assessment of ablative therapies in HCC patients were included. We found 173 clinical studies that satisfied the inclusion criteria.HCC may be noninvasively diagnosed by imaging findings. Multiphase contrast-enhanced imaging is necessary to assess HCC. Intravenous extracellular contrast agents are used for CT, while the agents used for MRI may be extracellular or hepatobiliary. Both gadoxetate disodium and gadobenate dimeglumine may be used in hepatobiliary phase imaging. For treatment-naive patients undergoing CT, unenhanced imaging is optional; however, it is required in the post treatment setting for CT and all MRI studies. Late arterial phase is strongly preferred over early arterial phase. The choice of modality (CT, US/CEUS or MRI) and MRI contrast agent (extracelllar or hepatobiliary) depends on patient, institutional, and regional factors. MRI allows to link morfological and functional data in the HCC evaluation. Also, Radiomics is an emerging field in the assessment of HCC patients.Postablation imaging is necessary to assess the treatment results, to monitor evolution of the ablated tissue over time, and to evaluate for complications. Post- thermal treatments, imaging should be performed at regularly scheduled intervals to assess treatment response and to evaluate for new lesions and potential complications.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Roberta Grassi
- Division of Radiology, Università degli Studi della Campania Luigi Vanvitelli, Naples, Italy
- Italian Society of Medical and Interventional Radiology SIRM, SIRM Foundation, Milan, Italy
| | | | - Andrea Belli
- Division of Hepatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Carmen Cutolo
- Department of Medicine, Surgery and Dentistry, University of Salerno, Salerno, Italy
| | - Silvia Pradella
- Radiology Division, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | - Giulia Grazzini
- Radiology Division, Azienda Ospedaliero-Universitaria Careggi, Florence, Italy
| | | | - Maria Chiara Brunese
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Federica De Muzio
- Department of Medicine and Health Sciences "Vincenzo Tiberio", University of Molise, Campobasso, Italy
| | - Alessandro Ottaiano
- Abdominal Oncology Division, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Antonio Avallone
- Abdominal Oncology Division, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Francesco Izzo
- Division of Hepatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale - IRCCS di Napoli, Naples, Italy
| |
Collapse
|