1
|
Jiang X, Zhang Q, Zheng Z, Shen Z, Luo Q. Latent class analysis-derived classification for cancer-specific death stratification of hepatocellular carcinoma. Int J Cancer 2025; 157:325-335. [PMID: 40071657 DOI: 10.1002/ijc.35399] [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: 10/18/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 05/16/2025]
Abstract
The heterogeneity in prognostic survival and treatment response of hepatocellular carcinoma (HCC) limits the accurate assessment of HCC-specific mortality. This study aimed to identify potential HCC subtypes through latent class analysis (LCA) to improve HCC-specific mortality prediction and optimize treatment recommendations. We analyzed data from 7746 HCC patients in the Surveillance, Epidemiology, and End Results (SEER) databases, incorporating demographic and clinicopathological information and applying LCA to identify HCC subtypes. Prognostic survival and treatment response across different HCC subtypes were evaluated utilizing Cox proportional hazards regression and competing risks models. The classification was externally validated with data from 6791 patients. Four HCC subtypes (LCAC1-LCAC4) were determined. Compared with LCAC1, both LCAC2 (HR = 1.887, p < .001) and LCAC4 (HR = 1.317, p < .001) were associated with significantly shorter overall survival. LCAC2 had the highest HCC-specific mortality (HR: 2.395, p < .001), followed by LCAC4 (HR: 1.531, p < .001), and LCAC3 (HR: 1.424, p < .001). LCAC3 was associated with the lowest risk of non-HCC-specific mortality (HR: 0.613, p < .001). Surgical treatment, particularly preoperative systemic therapy, significantly improved survival across all HCC subtypes, whereas chemotherapy and radiotherapy had limited efficacy in LCAC1 and LCAC3 patients. External validation corroborated these findings. This study provides a classification system that differentiates HCC-specific mortality, facilitating accurate survival stratification and treatment recommendations, and provides valuable insight for clinical decision-making.
Collapse
Affiliation(s)
- Xiaoyan Jiang
- Department of Gestational and Toxic Hepatopathy, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| | - Qianyuan Zhang
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Ziying Zheng
- Department of Oncology Medicine, Fujian Medical University Union Hospital, Fuzhou, Fujian, People's Republic of China
| | - Zhiyong Shen
- Department of Radiotherapy, Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
- Department of Radiotherapy, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, Fujian, People's Republic of China
| | - Qiong Luo
- Department of Oncology Medicine, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, Fujian, People's Republic of China
| |
Collapse
|
2
|
Wang Z, Li Y, Wang X, Zhang W, Chen Y, Lu X, Jin C, Tu L, Jiang T, Yang Y, Ma X, Zeng J, Wen Y, Efferth T. Precision Strike Strategy for Liver Diseases Trilogy with Xiao-Chai-Hu Decoction: A Meta-Analysis with Machine Learning. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 142:156796. [PMID: 40347886 DOI: 10.1016/j.phymed.2025.156796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/30/2025] [Accepted: 04/20/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND AND PURPOSE The progression from hepatitis to liver fibrosis (LF) and ultimately to hepatic carcinoma (HCC) represents the advanced stages of various liver diseases. Currently, no universal treatment effectively addresses all three conditions. The Traditional Chinese Medicine formula Xiao-Chai-Hu decoction (XCHD) has shown promise in treating hepatitis, inhibiting LF, and serving as an adjunct therapy for HCC. This study evaluates the efficacy and optimal treatment durations of XCHD in managing these liver diseases using meta-analysis and machine learning techniques. METHODS Registered in the PROSPERO database (CRD42024534445), this meta-analysis systematically searched seven databases, including 54 studies with a total of 5,710 patients. Statistical analysis was performed using Stata 17.0. Five machine learning models-Random Forest (RF), XGBoost, Lasso, Multilayer Perceptron (MLP), and a stacking model combining these algorithms-were employed to analyze the data and predict the time-effect relationships. The optimal durations of XCHD treatment for the liver disease trilogy were subsequently projected. RESULTS XCHD significantly improved the primary outcome indicators for hepatitis, liver fibrosis, and HCC. Additionally, XCHD demonstrated a beneficial effect on liver dysfunction caused by these diseases. Machine learning predicted the optimal treatment durations of XCHD as 12 weeks for hepatitis, 20.31 weeks for liver fibrosis, and 12 weeks for HCC. CONCLUSION XCHD is effective in treating the liver disease trilogy, with optimal treatment durations of 12 weeks for hepatitis and HCC, and 20.31 weeks for liver fibrosis. These findings support the potential of XCHD in developing precise clinical strategies for managing liver diseases. This study innovatively integrates meta-analysis with machine learning to determine the optimal treatment durations, providing a novel approach for evidence-based precision medicine in Traditional Chinese Medicine.
Collapse
Affiliation(s)
- Zexin Wang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yubing Li
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaobao Wang
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenwen Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuan Chen
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Xiaohua Lu
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Chunmei Jin
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany
| | - Lang Tu
- West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Tao Jiang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yiqin Yang
- Hunan Provincial Key Laboratory of the Research and Development of Novel Pharmaceutical Preparations, Changsha Medical University, Changsha, China
| | - Xiao Ma
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Jinhao Zeng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Department of Gastroenterology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Yueqiang Wen
- School of Basic Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China.
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmaceutical and Biomedical Sciences, Johannes Gutenberg University, Mainz, Germany.
| |
Collapse
|
3
|
Li YF, Yao LQ, Li C, Ren H, Gong JB, Wu H, Gu LH, Liang YJ, Yang YZ, Lin KY, Li ZQ, Zheng QX, Chen TH, Zhou YH, Wang H, Guo HW, Xu JH, Chen Z, Shen F, Wang MD, Yang T. Statistical Cure After Hepatectomy for Hepatitis B Virus-Associated Hepatocellular Carcinoma: A Risk-Stratification Model. Ann Surg Oncol 2025; 32:4396-4407. [PMID: 40188279 DOI: 10.1245/s10434-025-17176-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/27/2025] [Indexed: 04/07/2025]
Abstract
BACKGROUND Statistical cure, defined as achieving life expectancy comparable with that of disease-free individuals, has not been specifically investigated in hepatitis B virus-associated hepatocellular carcinoma (HBV-HCC), which accounts for more than 50% of the global HCC burden. This study aimed to develop a cure model for HBV-HCC after hepatectomy using matched HBV carriers and the general population as reference groups. METHODS From a Chinese multicenter database, HBV-HCC patients who underwent curative-intent hepatectomy were retrospectively reviewed. Independent prognostic factors were identified through Cox regression. A spline-based cure model was applied using two reference populations: matched Chinese HBV carriers (from Shanghai Center for Disease Control and Prevention) and the general population (from the National Bureau of Statistics). RESULTS The study analyzed 740 HBV-HCC patients. The following eight independent risk factors were identified: preoperative high viral load (hazard ratio [HR] 1.27), Child-Pugh grade (HR 1.21 and 1.43), multiple tumors (HR 1.70), tumor size greater than 5.0 cm (HR 1.47), macrovascular invasion (HR 3.33), microvascular invasion (HR 1.25), intraoperative blood transfusion (HR 1.21), and postoperative HBV reactivation (HR 1.89). The overall cure probability was 21.2% versus that for HBV carriers and 11.1% versus that for the general population. Risk stratification identified distinct groups relative to HBV carriers. Low risk (64.2%) showed an initial cure rate of 30.3% and achieved a 95% cure probability by 8.6 years, whereas high risk (10.5%) showed negligible cure probability. CONCLUSIONS This first HBV-HCC-specific cure model demonstrated that statistical cure is achievable for a subset of patients after hepatectomy. Risk stratification identifies patients with varying cure probabilities, providing valuable guidance for personalized treatment strategies and surveillance protocols.
Collapse
Affiliation(s)
- Yi-Fan Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Lan-Qing Yao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Hong Ren
- Department of Viral Hepatitis Control and Prevention, Shanghai Municipal Center for Disease Control and Prevention, Shanghai, China
| | - Jin-Bo Gong
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Li-Hui Gu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ying-Jian Liang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Yu-Ze Yang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, China
| | - Kong-Ying Lin
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fujian, China
| | - Zi-Qiang Li
- Department of Liver Transplantation and Hepatic Surgery, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qi-Xuan Zheng
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Ting-Hao Chen
- Department of General Surgery, Ziyang First People's Hospital, Ziyang, China
| | - Ya-Hao Zhou
- Department of Hepatobiliary Surgery, Pu'er People's Hospital, Pu'er, China
| | - Hong Wang
- Department of General Surgery, Liuyang People's Hospital, Liuyang, China
| | - Hong-Wei Guo
- The 2nd Department of General Surgery, The Second People's Hospital of Changzhi, Changzhi, China
| | - Jia-Hao Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Saleh RO, Hamad HA, Najim MA, Menon SV, Kaur M, Sivaprasad GV, Abohassan M, Juan WT, Husseen B, Mustafa YF. Exosome-mediated Transfer of lncRNA in Liver Associated Diseases; Uncovered Truths. Cell Biochem Biophys 2025; 83:1465-1481. [PMID: 39567423 DOI: 10.1007/s12013-024-01617-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 11/22/2024]
Abstract
Exosomes are extracellular vesicles with a diameter ranging from 40 to 160 nm. They are produced by hepatocytes, cholangiocytes, hepatic stellate cells (HSCs), liver sinusoidal endothelial cells (LSECs) and Kupffer cells in liver tissue. The secretion of exosomes might vary in quantity and composition in reaction to multiple triggers and various stages of disease. They transport various payloads, such as proteins, DNAs, and RNAs, and enable cell interaction to regulate myriad physiological and pathological processes in liver tissue. Long non-coding RNAs (lncRNAs) are a crucial component of exosomes with an excellent capability to regulate multiple cellular activities such as differentiation, development, metabolism, proliferation, apoptosis, and activation. With the advancements in transcriptomic and genomic study methods and database management technology, the functions and mechanisms of exosomal lncRNAs in liver diseases have been well-studied. This article delves into the detailed role of exosomal lncRNAs in liver disease onset and progression, ranging from hepatocellular carcinoma (HCC) to liver fibrosis drug-induced liver damage (DILI) and steatotic liver diseases.
Collapse
Affiliation(s)
- Raed Obaid Saleh
- Medical Laboratory Techniques Department, College of Health and Medical Technology, University of Al Maarif, Anbar, Iraq.
| | - Hamad Ali Hamad
- Department of Pathological Analysis, Collage of Applied Sciences, University of Fallujah, Fallujah, Iraq
- Department of Biomedical Sciences, Faculty of Medicine and Health Sciences, University Putra Malaysia, Serdang, Malaysia
| | | | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mandeep Kaur
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, India
| | - G V Sivaprasad
- Department of Basic Science & Humanities, Raghu Engineering College, Visakhapatnam, India
| | - Mohammad Abohassan
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, King Khalid University, Abha, Saudi Arabia
| | - Wen-Tau Juan
- Department of Medical Research, China Medical University Hospital, Taichung, Taiwan
| | - Beneen Husseen
- Medical Laboratory Technique college, The Islamic University, Najaf, Iraq
- Medical Laboratory Technique college, The Islamic University of Al Diwaniyah, Al Diwaniyah, Iraq
- Medical Laboratory Technique college, The Islamic University of Babylon, Babylon, Iraq
| | - Yasser Fakri Mustafa
- Department of Pharmaceutical Chemistry, College of Pharmacy, University of Mosul, Mosul, Iraq
| |
Collapse
|
5
|
Lee JW, Lee SM, Kang B, Kim JS, An C, Chon HJ, Jang SJ. Prognostic Significance of Volumetric Parameters on Pretreatment FDG PET/CT in Patients With Hepatocellular Carcinoma Receiving Atezolizumab Plus Bevacizumab Therapy. Clin Nucl Med 2025; 50:486-494. [PMID: 40254801 DOI: 10.1097/rlu.0000000000005896] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Accepted: 03/06/2025] [Indexed: 04/22/2025]
Abstract
BACKGROUND This study aimed to assess prognostic significance of FDG PET/CT parameters in predicting progression-free survival (PFS) and overall survival (OS) in patients with hepatocellular carcinoma (HCC) treated with atezolizumab plus bevacizumab therapy. PATIENTS AND METHODS We retrospectively enrolled 78 patients with HCC who underwent FDG PET/CT before atezolizumab plus bevacizumab therapy and identified intrahepatic target tumor lesions on pretreatment imaging studies. From PET/CT images, we measured SUVmax, tumor-to-normal liver uptake ratio, metabolic tumor volume, and total lesion glycolysis (TLG) for intrahepatic tumor lesions, as well as SUVmax for extrahepatic metastatic lesions (extrahepatic SUVmax). RESULTS In comparisons of PET/CT parameters, patients with progressive disease demonstrated significantly higher TLG values than those achieving complete or partial response ( P < 0.05). In the multivariate survival analysis, TLG independently predicted both PFS ( P = 0.019) and OS ( P = 0.003). Metabolic tumor volume was significantly associated with OS alone ( P = 0.010), and extrahepatic SUVmax was significantly associated with only PFS ( P = 0.045). Patients with high TLG values experienced poorer PFS and OS than those with low TLG values ( P < 0.05). CONCLUSIONS TLG in intrahepatic HCC lesions was significantly associated with treatment response and served as an independent prognostic factor for PFS and OS. TLG could be a potential imaging biomarker for predicting clinical outcomes in patients with HCC receiving atezolizumab plus bevacizumab therapy.
Collapse
Affiliation(s)
- Jeong Won Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, Cheonan
| | - Sang Mi Lee
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, Cheonan
| | - Beodeul Kang
- Department of Internal Medicine, Division of Medical Oncology
| | - Jung Sun Kim
- Department of Nuclear Medicine, Soonchunhyang University Cheonan Hospital, Cheonan
| | | | - Hong Jae Chon
- Department of Internal Medicine, Division of Medical Oncology
| | - Su Jin Jang
- Department of Nuclear Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongam, Republic of Korea
| |
Collapse
|
6
|
Guo D, Liu L, Jin Y. Prediction early recurrence of hepatocellular carcinoma after hepatectomy using gadoxetic acid-enhanced MRI and IVIM. Eur J Radiol Open 2025; 14:100643. [PMID: 40166482 PMCID: PMC11957592 DOI: 10.1016/j.ejro.2025.100643] [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: 01/01/2025] [Revised: 02/26/2025] [Accepted: 03/09/2025] [Indexed: 04/02/2025] Open
Abstract
Objectives This study aims to develop and validate a predictive nomogram for early recurrence in hepatocellular carcinoma (HCC), utilizing gadoxetic acid-enhanced MRI and intravoxel incoherent motion (IVIM) imaging to improve preoperative assessment and decision-making. Materials and methods From March 2018 and June 2022, a total of 245 patients with pathologically confirmed HCC, who underwent preoperative gadoxetic acid-enhanced MRI and IVIM, were retrospectively enrolled from two hospitals. These patients were divided into a training cohort (n = 160) and a validation cohort (n = 85). All patients were followed until death or the last follow-up date, with a minimum follow-up period of two years. Clinical indicators and pathologic information were compared between train cohort and validation cohort. Radiological features and diffusion parameters were compared between recurrence and non-recurrence groups using the chi-square test, Mann-Whitney U test and independent sample t test in training cohort. Univariate and multivariate analyses were performed to identify significant clinical-radiological variables associated with early recurrence in the training cohort. Based on these findings, a predictive nomogram integrating risk factors and diffusion parameters was developed. The predictive performance of the nomogram was evaluated in both the training and validation cohorts. Results No statistically significant difference in clinical and pathologic characteristics were observed between the training and validation cohorts. In training cohort, significant differences were identified between the recurrence and non-recurrence groups in tumor size, nodule-in-nodule architecture, mosaic architecture, non-smooth tumor margin, intratumor necrosis, satellite nodule, and peritumoral hypo-intensity in the hepatobiliary phase (HBP). The results of multivariate analysis identified tumor size (HR, 1.435; 95 % CI, 0.702-2.026; p < 0.05), mosaic architecture (HR, 0.790; 95 % CI, 0.421-1.480; p < 0.05), non-smooth tumor margin (HR, 1.775; 95 % CI, 0.941-3.273; p < 0.05), intratumor necrosis (HR, 1.414; 95 % CI, 0.807-2.476; p < 0.05), satellite nodule (HR, 0.648; 95 % CI, 0.352-1.191; p < 0.01), peritumoral hypo-intensity on HBP (HR, 2.786; 95 % CI, 1.141-6.802; p < 0.001) and D (HR, 0.658; 95 % CI,0.487-0.889; p < 0.01) were the independent risk factor for recurrence. The nomogram exhibited excellent predictive performance with C-index of 0.913 and 0.875 in the training cohort and validation cohort, respectively. Also, based on the nomogram score, the patients were classified according to risk factor and the Kaplan-Meier curve analysis also showed that the nomogram had a good predictive efficacy. Conclusion The nomogram, integrating radiological risk factors and diffusion parameters, offers a reliable tool for preoperative prediction of early recurrence in HCC patients.
Collapse
Affiliation(s)
- Da Guo
- Department of Radiology, Physical and Mental Hospital of Nanchong City, Nanchong, Sichuan, PR China
| | - Liping Liu
- Department of Radiology, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, PR China
| | - Yu Jin
- Department of Radiology, Chengdu Second People’s Hospital, Chengdu, Sichuan, PR China
| |
Collapse
|
7
|
Liu Q, Liang Z, Wang J, Wang Y, Wang J, Wang S, Du Z, Zhao L, Wei Y, Huang D. Mannose-modified multifunctional iron-based nanozyme for hepatocellular carcinoma treatment by remodeling the tumor microenvironment. Colloids Surf B Biointerfaces 2025; 250:114548. [PMID: 39923382 DOI: 10.1016/j.colsurfb.2025.114548] [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: 10/19/2024] [Revised: 01/12/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths worldwide, with conventional treatments often accompanied by severe side effects. Recently, nanozymes have been extensively employed in cancer therapy due to their enhanced enzymatic activities, stability compared to native enzymes. However, a standalone nanozyme exhibits insufficient targeting capability and fails to specifically localize to the pathological site. In this study, we successfully synthesized a multifunctional iron-based-nanozyme delivery system - Fe3O4-OA-DHCA-PEI-MAN@DSF modified with PEI and MAN by the thermal decomposition method. This mannose-modified nanozyme can specifically target HCC cells via an external magnetic field and mannose-mannose receptor (MRC2) binding. In addition, it exhibits good biocompatibility and pH-dependent drug release characteristics. Within the acidic tumor microenvironment, the iron-based nanozyme initiates intracellular fenton reactions, boosting reactive oxygen species (ROS) production, which ultimately induces apoptosis in HCC cells. Concurrently, the disulfiram small molecule released from the Fe3O4-OA-DHCA-PEI-MAN@DSF nanozyme binds to the FROUNT factor within monocyte-macrophages, thereby inhibiting their response to chemotactic signals emitted by liver cancer cells. This process ultimately suppresses the recruitment of macrophages by HCC cells, reshaping the tumor microenvironment and supporting effective liver cancer treatment. Moreover, this nanozyme system holds potential for MRI-guided targeted chemotherapy combined with chemodynamic therapy, aiming to refine the early diagnosis and precision treatment of hepatic carcinoma, and paving the way for the creation of sophisticated integrated nanoplatforms melding diagnostic and therapeutic functionalities.
Collapse
Affiliation(s)
- Qi Liu
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Ziwei Liang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; NHC Key Laboratory of Glycoconjuates Research Department of Biochemistry and Molecular, Biology School of Basic Medical Sciences, Fudan University, Shanghai 200032, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| | - Jiapu Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Yuhui Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Jie Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Shaojie Wang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China
| | - Zhi Du
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Liqin Zhao
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China
| | - Yan Wei
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| | - Di Huang
- Department of Biomedical Engineering, Research Center for Nano-biomaterials & Regenerative Medicine, College of Artificial Intelligence, Shanxi Key Laboratory of Materials Strength & Structural Impact, Taiyuan University of Technology, Taiyuan 030024, China; Shanxi-Zheda Institute of Advanced Materials and Chemical Engineering, Taiyuan 030032, China; Shanxi Provincial Key Laboratory for Functional Proteins, Shanxi Jinbo Bio-Pharmaceutical Co., Ltd, Taiyuan 030032, China.
| |
Collapse
|
8
|
Yan F, Wang W, Yang Z, Huang Y, Rao Y, Qu G, Peng H, Shi M, Zeng W, Chen D, Xing W. Intra-arterial lidocaine improves long-term survival in patients with hepatocellular carcinoma undergoing transcatheter arterial chemoembolisation: a retrospective propensity score-matched study. Br J Anaesth 2025; 134:1628-1637. [PMID: 40118673 DOI: 10.1016/j.bja.2025.01.042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 12/14/2024] [Accepted: 01/07/2025] [Indexed: 03/23/2025] Open
Abstract
BACKGROUND Lidocaine, the most widely used local anaesthetic, has anticancer effects in both laboratory findings and retrospective clinical studies. We explored the potential benefits of intra-arterial lidocaine on long-term survival in patients with hepatocellular carcinoma (HCC) undergoing transcatheter arterial chemoembolisation (TACE). METHODS This retrospective cohort study included patients with HCC who received TACE as initial treatment from August 2011 to October 2016. Eligible patients were categorised into no lidocaine and lidocaine groups. Propensity score matching was undertaken. Progression-free survival (PFS) and overall survival were compared between the two groups. Subgroup analysis was performed to explore the survival benefit of combining intra-arterial lidocaine with platinum-based chemotherapy during TACE. RESULTS Of 374 eligible patients, 96 were in the lidocaine group and 278 were in the no lidocaine group. Survival analysis showed that intra-arterial lidocaine was associated with longer PFS (P=0.004) and overall survival (P<0.001). After propensity score matching, PFS (P<0.001) and overall survival (P=0.001) benefits of lidocaine remained. Multivariate analysis showed that intra-arterial lidocaine was an independent prognostic factor for PFS (P=0.011) and overall survival (P=0.044). The impact of intra-arterial lidocaine was similar in patients receiving the TACE regimen with platinum (PFS: P=0.014; overall survival: P=0.023). CONCLUSIONS Intra-arterial lidocaine might improve long-term survival in patients with HCC undergoing TACE and in the subgroup of patients receiving platinum. The study highlights the potential antitumour benefits of combining lidocaine and chemotherapeutics in patients with cancer.
Collapse
Affiliation(s)
- Fang Yan
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Wanyu Wang
- Department of Anesthesiology, West China Hospital of Stomatology, Sichuan University, Chengdu, Sichuan, PR China
| | - Zhiwen Yang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yang Huang
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Yan Rao
- Department of Anesthesiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian Province, PR China
| | - Ge Qu
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Hui Peng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Ming Shi
- Department of Hepatobiliary Oncology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Weian Zeng
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China
| | - Dongtai Chen
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| | - Wei Xing
- Department of Anesthesiology, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-sen University Cancer Center, Guangzhou, PR China.
| |
Collapse
|
9
|
Zhang H, Yuan F, Zhao N, Tang W, Zhao P, Liu C, Chen S, Hou X, Xia C, Chu J. Nanoparticle-mediated SIRT1 inhibition suppresses M2 macrophage polarization and hepatocarcinogenesis in chronic hepatitis B. J Nanobiotechnology 2025; 23:385. [PMID: 40426198 DOI: 10.1186/s12951-025-03447-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Accepted: 05/05/2025] [Indexed: 05/29/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a major complication of chronic hepatitis B (CHB), with macrophage M2 polarization playing a critical role in shaping the tumor-promoting hepatic immune microenvironment. Sirtuin 1 (SIRT1) has been implicated in immune modulation and liver carcinogenesis. This study investigates the potential of Mimetic Nanoparticles (MNPs) for delivering SIRT1 inhibitors to regulate macrophage polarization and remodel the hepatic immune microenvironment, aiming to prevent HCC development post-CHB. A transgenic mouse model of CHB was established, and RNA sequencing (RNA-seq) and proteomics analyses revealed significant dysregulation of genes associated with M2 macrophage polarization, particularly SIRT1. Functional enrichment analysis highlighted key pathways, including PI3K-Akt and NF-κB, that contribute to CHB-driven immune alterations. Synthesized and characterized MNPs successfully delivered SIRT1 inhibitors, effectively inhibiting M2 macrophage polarization in vitro. In vivo administration of MNPs-SIRT1-FN significantly reduced M2 macrophage infiltration and suppressed tumor growth. These findings suggest that nanoparticle-mediated SIRT1 inhibition is a promising strategy for immunomodulation and HCC prevention in CHB patients. This study provides novel insights into nanoparticle-based immunotherapy for CHB-related HCC and highlights a potential therapeutic avenue for liver cancer prevention.
Collapse
Affiliation(s)
- He Zhang
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Feng Yuan
- Laboratory of Inflammation and Vaccines, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Nan Zhao
- Institute of Clinical Medicine, National Infrastructures for Translational Medicine, Peking Union Medical College Hospital, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, 100730, China
| | - Wenqiang Tang
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China
| | - Pengwei Zhao
- The Fourth Affiliated Hospital, Zhejiang University School of Medicine, Yiwu, 322000, China
| | - Chunfa Liu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Shan Chen
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China
| | - Xiaolin Hou
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
| | - Changyou Xia
- State Key Laboratory for Animal Disease Control and Prevention, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China.
| | - Jun Chu
- College of Animal Science and Technology, Beijing University of Agriculture, No. 7 Beinong Road, Beijing, 102206, China.
- Institute of Animal Science, Tibet Academy of Agricultural and Animal Husbandry Science, Lhasa, 850009, China.
| |
Collapse
|
10
|
Ye M, Chen S, Lu C, Wu Y, Tian J, Han A, Zhu J, Li B, Li Q. Cinobufagin inhibits hepatocellular carcinoma EMT-like stemness via VEGF/VEGFR2 autocrine signaling. Discov Oncol 2025; 16:930. [PMID: 40418461 DOI: 10.1007/s12672-025-02707-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 05/13/2025] [Indexed: 05/27/2025] Open
Abstract
PURPOSE This study aimed to explore the role of the VEGFa/VEGFR2 autocrine pathway in cinobufagin-induced inhibition of hepatocellular carcinoma metastasis. METHODS A CCK-8 assay was performed to assess cell viability. Scratch healing, Transwell, and sphere formation assays were used to measure the effects of cinobufagin on cell migration, invasion, and tumor sphere formation. An immunofluorescence double staining method was used to detect the localization of VEGFa and VEGFR2. The effects of inhibiting the VEGFa/VEGFR2 autocrine pathway on Huh7 cell metastasis and the effects of the VEGFa/VEGFR2 autocrine pathway on the regulation of PI3K/AKT-dependent migration, invasion, and epithelial‒mesenchymal transition were examined through use of the VEGFR2 inhibitor apatinib and the PI3K inhibitor LY294002. The effects of cinobufagin on the VEGFa/VEGFR2 autocrine pathway and tumor metastasis were assessed in transplanted tumors. RESULTS Cinobufagin inhibited Huh7 cell viability, migration, invasion, and tumor sphere formation in a dose-dependent manner. In addition, colocalization between VEGFa and VEGFR2 was detected in Huh7 cells. The results revealed that apatinib treatment significantly inhibited PI3K/AKT-dependent migration, invasion, and epithelial‒mesenchymal transition. The VEGFa/VEGFR2 autocrine pathway, the PI3K/AKT pathway, and epithelial-mesenchymal transition-associated protein markers were attenuated by cinobufagin in Huh7 cells. Additionally, cinobufagin attenuated the growth of hepatocellular carcinoma tumors in the Huh7 xenograft model and significantly downregulated the VEGFa/VEGFR2 autocrine pathway, the PI3K/AKT pathway, and the epithelial‒mesenchymal transition. CONCLUSION Cinobufagin may attenuate the PI3K/AKT-dependent metastatic potential of hepatocellular carcinoma by inhibiting the VEGFa/VEGFR2 autocrine pathway.
Collapse
Affiliation(s)
- Mingjun Ye
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Shujun Chen
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Chen Lu
- The First Affiliated Hospital, Anhui University of Chinese Medicine, Hefei, 230038, China
| | - Yangpei Wu
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jiaming Tian
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Anqi Han
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Jimin Zhu
- School of Life Sciences, Anhui University of Chinese Medicine, Hefei, 230012, China
| | - Baikun Li
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China.
- School of Traditional Chinese Medicine, Anhui University of Chinese Medicine, Hefei, 230012, China.
| | - Qinglin Li
- Key Laboratory of Xin'an Medicine (Anhui University of Chinese Medicine), The Ministry of Education, Hefei, 230038, China.
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, China.
| |
Collapse
|
11
|
Li T, Liu S, Wang S, Sun S, Ji F, Li M, Zhang Y. Identification of metabolic reprogramming-related key genes in hepatocellular carcinoma after transcatheter arterial chemoembolization treatment. Discov Oncol 2025; 16:861. [PMID: 40404896 PMCID: PMC12098233 DOI: 10.1007/s12672-025-02606-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Accepted: 05/06/2025] [Indexed: 05/24/2025] Open
Abstract
BACKGROUND Metabolic reprogramming plays an important role in therapeutic efficacy of hepatocellular carcinoma (HCC). However, the metabolic reprogramming-related key genes associated with transcatheter arterial chemoembolization (TACE) treatment sensitivity in HCC remain further investigation. METHODS We analyzed data from public databases, The Cancer Genome Atlas and Gene Expression Omnibus, as well as metabolism-related genes (MRGs), to identify key genes associated with TACE treatment sensitivity. Further analysis was conducted on the relationship between key genes and immune cell infiltration, HCC-related genes, regulatory network construction, nomogram construction, and drug sensitivity analysis. Finally, the expression of key genes was validated based on databases and in vitro RT-qPCR. RESULTS Four key genes (CDC20, LPCAT1, PON1, and SPP1) associated with TACE treatment sensitivity were identified. Increased CDC20, LPCAT1, and SPP1 and reduced PON1 were found in tumor tissues than normal tissues, as well as in advanced patients than early-stage patients. Lower expression of CDC20, LPCAT1, and SPP1, and higher expression of PON1 were detected in responsive patients than non-responsive patients. Patients with high expression of CDC20, LPCAT1, and SPP1, and low expression of PON1 had poor prognosis. They were also correlated with tumor immune microenvironment and sensitivity to multiple chemotherapy drugs. The expressions of key genes at the gene and protein levels were validated. CONCLUSIONS Our study provided systematic insights into identification of biomarkers for TACE treatment sensitivity in HCC.
Collapse
Affiliation(s)
- Tongfei Li
- Department of Interventional Radiology, The Second Affiliated Hospital of Shandong First Medical University, Tai'an, 271000, China
| | - Shujuan Liu
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Shengjun Wang
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Shan Sun
- Department of Oncology, Jinan Seventh People's Hospital, Jinan, 250132, China
| | - Feng Ji
- Department of Oncology, Shanxian Dongda Hospital, Heze, 274399, China
| | - Mingliang Li
- Automation Department, Jigang Group International Engineering Technology Co., Ltd, Jinan, 250098, China
| | - Yong Zhang
- School of Preventive Medicine, Shandong First Medical University, No. 6699 Qingdao Road, Jinan, 250117, Shandong, People's Republic of China.
| |
Collapse
|
12
|
Zhang F, Wu Z, Xiang Y, He Q, Li W, Yang K, Yang Y. SOX4 reprograms fatty acid metabolism through the CHREBP to inhibit ferroptosis in hepatocellular carcinoma. Cell Death Discov 2025; 11:246. [PMID: 40399256 PMCID: PMC12095664 DOI: 10.1038/s41420-025-02527-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 04/20/2025] [Accepted: 05/08/2025] [Indexed: 05/23/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality, characterized by aggressive progression and poor prognosis. Pathological angiogenesis in HCC is closely linked to metabolic reprogramming, particularly concerning fatty acid metabolism. The interplay between fatty acid metabolism and ferroptosis, a type of cell death driven by lipid peroxidation, is emerging as a crucial area of study. The transcription factor SOX4 is known to be overexpressed in various cancers, including HCC, and may play a key role in these processes. We assessed SOX4 expression in HCC using clinical samples and data from online databases. Next-generation RNA sequencing was employed to explore the effects of SOX4 on fatty acid metabolism, focusing on the CHREBP pathway. Functional assays, including lipid peroxidation and angiogenesis studies, were conducted to investigate the role of SOX4 in regulating ferroptosis and angiogenesis in HCC. SOX4 was found to be significantly upregulated in HCC and associated with enhanced angiogenesis. Mechanistically, SOX4 activated the CHREBP/SCD1 pathway, leading to increased production of monounsaturated fatty acids, which in turn inhibited ferroptosis. This suppression of ferroptosis contributed to the promotion of angiogenesis and tumor progression in HCC. In conclusion, SOX4 reprograms fatty acid metabolism via the CHREBP/SCD1 pathway, thereby inhibiting ferroptosis and promoting angiogenesis in HCC. These findings suggest that targeting the SOX4-CHREBP axis could represent a novel therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Fan Zhang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou, China
| | - Zhiwei Wu
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | - Yang Xiang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China.
- Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou, China.
| | - Qing He
- Department of Organ Transplantation, Xiangya Hospital, Central South University, Changsha, China
| | - Wanqing Li
- Hunan Occupational Disease Prevention Hospital, Changsha, China
| | - Kaipeng Yang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou, China
| | - Yijun Yang
- Department of Hepatobiliary Surgery, Affiliated Haikou Hospital of Xiangya Medical College, Central South University, Haikou, China
- Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou, China
| |
Collapse
|
13
|
Zhang P, Shi Y, Zhou M, Mao Q, Tao Y, Yang L, Zhang X. A CECT-Based Radiomics Nomogram Predicts the Overall Survival of Patients with Hepatocellular Carcinoma After Surgical Resection. Biomedicines 2025; 13:1237. [PMID: 40427064 DOI: 10.3390/biomedicines13051237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/19/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Objective: The primary objective of this study was to develop and validate a predictive nomogram that integrates radiomic features derived from contrast-enhanced computed tomography (CECT) images with clinical variables to predict overall survival (OS) in patients with hepatocellular carcinoma (HCC) after surgical resection. Methods: This retrospective study analyzed the preoperative enhanced CT images and clinical data of 202 patients with HCC who underwent surgical resection at the Affiliated Hospital of North Sichuan Medical College (Institution 1) from June 2017 to June 2021 and at Nanchong Central Hospital (Institution 2) from June 2020 to June 2022. Among these patients, 162 patients from Institution 1 were randomly divided into a training cohort (112 patients) and an internal validation cohort (50 patients) at a 7:3 ratio, whereas 40 patients from Institution 2 were assigned as an independent external validation cohort. Univariate and multivariate Cox proportional hazards regression analyses were performed to identify clinical risk factors associated with OS after HCC resection. Using 3D-Slicer software, tumor lesions were manually delineated slice by slice on preoperative non-contrast-enhanced (NCE) CT, arterial phase (AP), and portal venous phase (PVP) images to generate volumetric regions of interest (VOIs). Radiomic features were subsequently extracted from these VOIs. LASSO Cox regression analysis was employed for dimensionality reduction and feature selection, culminating in the construction of a radiomic signature (Radscore). Cox proportional hazards regression models, including a clinical model, a radiomic model, and a radiomic-clinical model, were subsequently developed for OS prediction. The predictive performance of these models was assessed via the concordance index (C-index) and time-ROC curves. The optimal performance model was further visualized as a nomogram, and its predictive accuracy was evaluated via calibration curves and decision curve analysis (DCA). Finally, the risk factors in the optimal performance model were interpreted via Shapley additive explanations (SHAP). Results: Univariate and multivariate Cox regression analyses revealed that BCLC stage, the albumin-bilirubin index (ALBI), and the NLR-PLR score were independent predictors of OS after HCC resection. Among these three models, the radiomic-clinical model exhibited the highest predictive performance, with C-indices of 0.789, 0.726, and 0.764 in the training, internal and external validation cohorts, respectively. Furthermore, the time-ROC curves for the radiomic-clinical model showed 1-year and 3-year AUCs of 0.837 and 0.845 in the training cohort, 0.801 and 0.880 in the internal validation cohort, and 0.773 and 0.840 in the external validation cohort. Calibration curves and DCA demonstrated the model's excellent calibration and clinical applicability. Conclusions: The nomogram combining CECT radiomic features and clinical variables provides an accurate prediction of OS after HCC resection. This model is beneficial for clinicians in developing individualized treatment strategies for patients with HCC.
Collapse
Affiliation(s)
- Peng Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yue Shi
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Maoting Zhou
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Qi Mao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Yunyun Tao
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Lin Yang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| | - Xiaoming Zhang
- Medical Imaging Key Laboratory of Sichuan Province, Department of Radiology, Interventional Medical Center, Science and Technology Innovation Center, The Affiliated Hospital of North Sichuan Medical College, Nanchong 637000, China
| |
Collapse
|
14
|
Jiang Y, Dong X, Zhang Y, Su F, Zhao L, Shi X, Zhong J. Navigating the complexities: challenges and opportunities in conversion therapy for advanced hepatocellular carcinoma. Clin Exp Med 2025; 25:169. [PMID: 40382739 PMCID: PMC12086121 DOI: 10.1007/s10238-025-01698-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Primary liver cancer ranks as the sixth most prevalent malignant tumor and stands as the second leading cause of cancer-related mortality globally, posing a significant threat to public health. Hepatocellular carcinoma (HCC) is the most common type of liver cancer worldwide. Surgical resection remains the cornerstone treatment for achieving radical cure and prolonged survival in HCC patients. Contrary to Western countries, the majority of HCC patients in China present with hepatitis B virus infection and consequent liver cirrhosis, with most cases diagnosed at an intermediate or advanced stage. This complexity results in a poor prognosis. Recent advancements in local therapeutic techniques and the introduction of systemic therapies, including targeted and immunotherapy agents, have provided new avenues for both clinical and basic conversion therapy for advanced HCC. Integrating multi-dimensional local and systemic therapies, multi-modal sequential, and comprehensive multidisciplinary approaches into the management of HCC patients has demonstrated promising conversion success rates. This holistic management strategy involves combining multiple treatment modalities vertically and coordinating various disciplines horizontally. However, significant challenges remain, including the precise selection of patients eligible for conversion therapy, the optimal choice of conversion therapy regimens, and the accurate determination of surgical timing post-conversion therapy. Addressing these challenges is crucial for hepatobiliary surgeons. High-quality, randomized controlled trials are urgently needed to generate robust evidence for clinical practice. This review aims to synthesize the latest research developments both in China and internationally and examines key issues in the realm of HCC conversion therapy.
Collapse
Affiliation(s)
- Yubo Jiang
- Department of Gastroenterology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong Province, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yingying Zhang
- Department of Oncology, Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong Province, China
| | - Feiyan Su
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Lei Zhao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China.
| |
Collapse
|
15
|
Li R, Zhang G, Tao Q, Wu Z, Liu X, Wang R, Liu L, Niu Y, Du K, Wu R, Du F, Zheng X, Li Y, Shi X. Revealing the prognostic potential of natural killer cell-related genes in hepatocellular carcinoma: the key role of NRAS. Discov Oncol 2025; 16:807. [PMID: 40383831 PMCID: PMC12086133 DOI: 10.1007/s12672-025-02200-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/21/2025] [Indexed: 05/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy associated with high morbidity and mortality rates worldwide. To improve the prognosis of HCC, early diagnosis is crucial. However, to date, little is known about the role of natural killer cell-related genes (NKCRGs) in predicting the prognosis of hepatocellular carcinoma patients. In this study, we identified 24 differentially expressed NKCRGs in HCC specimens from the TCGA dataset, including 22 upregulated genes and 2 downregulated genes. Functional enrichment analysis revealed that these genes were mainly involved in immune response pathways and various cancer-related pathways. Univariate analysis identified 21 prognostic NKCRGs, with eight genes (PAK1, MAP2K2, MAPK3, PLCG1, SHC1, HRAS, NRAS, and MICB) confirmed to be involved in HCC prognosis through Venn diagram analysis. A prognostic model was developed using LASSO-Cox regression, incorporating four genes (MAP2K2, SHC1, HRAS, and NRAS). The model's risk score was significantly associated with overall survival (OS) in both the TCGA and ICGC cohorts. Patients with high-risk scores had poorer OS, as demonstrated by Kaplan-Meier curves and ROC analyses. The risk score was not significantly correlated with gender or age but was higher in patients with advanced tumor grades and stages. Immune status analysis using ssGSEA showed higher enrichment scores for various immune cells and pathways in the high-risk group. Additionally, the risk score was positively correlated with the immune score, indicating its potential role in tumor microenvironment modulation. Expression analysis revealed that HRAS, SHC1, MAP2K2, and NRAS were upregulated in HCC tissues, with higher expressions of HRAS, MAP2K2, and NRAS associated with shorter OS. Knockdown experiments confirmed that silencing NRAS suppressed the proliferation of HCC cells, highlighting its potential as a therapeutic target. Overall, our findings suggest that the identified NKCRGs, particularly NRAS, play crucial roles in HCC progression and could serve as valuable prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Qiang Tao
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Ziyun Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Xiaoping Liu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Rongrong Wang
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Lei Liu
- Department of Clinical Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yiran Niu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Kaile Du
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Runpeng Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Fei Du
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xiyan Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yingliang Li
- Department of Breast Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
16
|
Wu W, Mao H, Song J, Yang F. Bibliometric analysis of hepatocellular carcinoma and tyrosine kinase inhibitors. Medicine (Baltimore) 2025; 104:e42015. [PMID: 40388796 PMCID: PMC12091622 DOI: 10.1097/md.0000000000042015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/13/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor globally and in China, and its incidence and mortality rate are increasing year by year, and it faces many challenges and difficulties in treatment. Tyrosine kinase inhibitors (TKIs) have important roles in cell growth, proliferation, and differentiation, and have now become important drugs for cancer treatment. There are no bibliometric studies on liver cancer and TKIs to date. METHODS We retrieved 2848 records from the Web of Science™ Core Collection (WoSCC) database and analyzed them scientifically and metrically using CiteSpace and VOSviewer in terms of temporal and spatial distributions, author distributions, journal distributions, references, and keywords. RESULTS From January 1, 2004, to December 31, 2023, the WoSCC database documented 2848 publications related to tyrosinase inhibitors in HCC, comprising 2151 articles and 697 reviews. This literature involved 80 countries and regions, 3265 institutions, and 16,653 authors. Analysis shows a steady increase in publications annually since 2004, divided into 3 phases: 2004 to 2010 with fewer than 100 papers annually, suggesting minimal research attention; 2011 to 2019 with gradual growth, indicating increasing research interest; and a rapid surge post-2020, peaking in 2023, signaling heightened global interest in this field. CONCLUSION Our bibliometric analysis on TKIs and HCC spans years, countries, institutions, authors, disciplines, and journals. Since 2004, this field has gained attention, with current research focusing on inflammatory and immune mechanisms, associated diseases, cytokines, and TKIs' applications in liver cancer treatment, including combination therapies. These areas signify emerging research directions.
Collapse
Affiliation(s)
- Wurihan Wu
- Department of Neurology Department, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hejun Mao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jian Song
- Emergency Intensive Care Unit, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, China
| | - Fan Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
17
|
Liu MS, Zhong SS, Wang JK, Wang T, Zhang KH. Research Trends on Nanomaterials and Hepatocellular Carcinoma From 1999 to 2024: A Bibliometric Analysis. Drug Des Devel Ther 2025; 19:3949-3970. [PMID: 40395437 PMCID: PMC12091239 DOI: 10.2147/dddt.s516647] [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: 01/09/2025] [Accepted: 05/04/2025] [Indexed: 05/22/2025] Open
Abstract
Objective Extensive exploratory studies have been conducted and promising progress has been made in the use of nanomaterials for the diagnosis and treatment of hepatocellular carcinoma (HCC). Here, we aimed to reveal the evolution and trends in this field through bibliometric analysis. Methods English-language publications (1999-2024) in the field of nanomaterials and HCC were retrieved from the Web of Science database, and eligible articles were selected for bibliometric analysis (data extraction, statistical analysis, and visualization) using VOSviewer and Citespace software. Results A total of 1617 eligible publications were analyzed. The number of publications increased rapidly from 2012 and peaked in 2020. China contributed the most publications, and the United States had the most citations. The Chinese Academy of Sciences was the most influential institution. The "International Journal of Nanomedicine (DOVE Medical)" published the most articles, while "Biomaterials (Elsevier)" was the most influential journal. Jie Tian had the highest number of publications, and Dan Shao had the highest average citation per article. Keyword analysis revealed that nanoparticles for targeted drug delivery, therapy and imaging of HCC were research hotspots. Keywords with citation bursts in the last three years included photodynamic therapy, sorafenib, and tumor microenvironment. Nano-vaccines, nano-antibodies, and synergistic therapies were emerging therapeutic strategies. A total of seven clinical trials were published, but to date there have been no major breakthroughs in HCC therapy using nanomaterials. Conclusion Research on nanomaterials and HCC has shown an overall upward trend, with research hotspots and frontiers focusing on nanoparticle-targeted chemotherapies, photodynamic therapy, and related tumor microenvironment research.
Collapse
Affiliation(s)
- Mao-Sheng Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Si-Si Zhong
- Department of Quality and Safety Management, the First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Jin-Ke Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
18
|
Hong Z, Wang J, Hu B, Tu X, Yang J, Sun W, Duan X. Esculetin inhibits liver cancer by targeting glucose-6-phosphate isomerase mediated glycolysis. Biomed Pharmacother 2025; 188:118118. [PMID: 40373632 DOI: 10.1016/j.biopha.2025.118118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2025] [Revised: 04/26/2025] [Accepted: 04/30/2025] [Indexed: 05/17/2025] Open
Abstract
BACKGROUND Liver cancer is challenging to detect in its early stages, and the global incidence rate and mortality associated with this disease have reached alarming levels. Currently, treatment options for liver cancer are limited, and there is a significant lack of safe and effective therapeutic agents. Esculetin is a natural product, exhibits almost non-toxic and inhibitory properties against various malignancies, making it a subject worthy of further investigation in liver cancer. METHODS In this study, potential targets of esculetin in liver cancer were identified through transcriptomics, network pharmacology, and molecular docking technologies, and gene interference. Direct binding targets of esculetin were identified using surface plasmon resonance (SPR). The molecular mechanisms by which esculetin affects glucose metabolism in liver cancer were also explored. Finally, the activity against liver cancer and mechanisms of action of esculetin were validated in vivo using a mouse tumor model. RESULTS Glucose-6-phosphate isomerase (GPI) was shown to have a direct binding affinity for this compound. Esculetin inhibits glycolysis in liver cancer through its interaction with GPI and it was shown to exert a significant inhibitory effect on the genes and proteins associated with glycolysis such as ALDOA, ENO1, GAPDH, LDHA, PFKL, PGAM1, PGK1, and PKM2. Furthermore, esculetin not only suppresses the growth of liver cancer cells in vitro but also exhibits notable anti-tumor effects in vivo. CONCLUSIONS This study demonstrated the inhibitory effects of esculetin against liver cancer both in vitro and in vivo, demonstrating inhibition of glycolysis in liver cancer cells. In addition, the key glycolysis enzyme GPI was identified as a direct target of esculetin.
Collapse
Affiliation(s)
- Zongchao Hong
- Wuling Mountain Traditional Chinese Medicine Inspection and Testing Center, Hubei Minzu University, Enshi, China; Health Science Center, Hubei Minzu University, Enshi, China; Hubei Provincial Key Laboratory of Occurrence and Intervention of Rheumatic diseases,Hubei Minzu University, Enshi, China.
| | - Jingbo Wang
- Department of Integrated Traditional Chinese and Western Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.
| | - Baodan Hu
- The Central Hospital of Enshi Tujia and Miao Autonomous Prefecture, Enshi, China.
| | - Xin Tu
- Wuling Mountain Traditional Chinese Medicine Inspection and Testing Center, Hubei Minzu University, Enshi, China
| | - Jin Yang
- Health Science Center, Hubei Minzu University, Enshi, China
| | - Wanjin Sun
- Hubei Key Laboratory of theory and application research of liver and kidney in traditional Chinese medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
| | - Xueyun Duan
- Hubei Key Laboratory of theory and application research of liver and kidney in traditional Chinese medicine, Hubei Provincial Hospital of Traditional Chinese Medicine, Wuhan, China.
| |
Collapse
|
19
|
Cressman E, Stolley D, Fowlkes N, Felix E, Priebe W, Parrish S, Fuentes D. Harnessing Electrophiles In Vivo: A Pilot Study in Swine Using a Hydrophobic Radiopaque Formulation of 2-Propylpentanoyl Chloride. Mol Pharm 2025. [PMID: 40359181 DOI: 10.1021/acs.molpharmaceut.4c01456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Manipulating biology through chemistry is older than the discipline of chemistry itself. Traditionally, selectivity of oral or intravenous drugs relied on preferential drug-receptor binding. A novel approach exploiting image-guided techniques to impart spatial selectivity opens up a wide range of new possibilities for study and manipulation of biology. Motivated initially by the poor prognosis for solid tumors such as liver cancer, we demonstrate the use of an extreme form of in vivo chemistry for targeted delivery of 2-propylpentanoyl chloride in a swine model. The ensuing reaction in tissue devitalizes it by multiple mechanisms with lasting effects and, critically, demonstrates very low systemic exposure compared to controls. This work points toward a new, powerful strategy for investigating the interface between chemistry and biology in vivo.
Collapse
Affiliation(s)
- Erik Cressman
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Danielle Stolley
- Flow Cytometry & Cellular Imaging Core Facility, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Natalie Fowlkes
- Department of Veterinary Medicine and Surgery, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Edd Felix
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Waldemar Priebe
- Department of Cancer Systems Imaging, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - Steve Parrish
- Department of Interventional Radiology, MD Anderson Cancer Center, Houston, Texas 77030, United States
| | - David Fuentes
- Department of Imaging Physics, MD Anderson Cancer Center, Houston, Texas 77030, United States
| |
Collapse
|
20
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [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: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
21
|
Zhou J, Gao M, Zhang S, Guo WW, He W, Zhang M, Chen X, Dongzhi C, Li X, Yuan Y, Ma W. PP1A Modulates the Efficacy of Lenvatinib Plus ICIs Therapy by Inhibiting Ferroptosis in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2501730. [PMID: 40344394 DOI: 10.1002/advs.202501730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 04/09/2025] [Indexed: 05/11/2025]
Abstract
Advanced hepatocellular carcinoma (HCC) is characterized by poor prognosis, primarily due to limited therapeutic options and resistance to treatment. Although the combination of tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) has shown promising potential, the underlying mechanisms remain inadequately understood. Here, serine/threonine-specific protein phosphatase (PP1A) is upregulated in Lenvatinib-resistant HCC cells and correlates with poor prognosis. Functional experiments revealed that PP1A promotes HCC progression both in vitro and in vivo. Transcriptomic analysis and ferroptosis metabolite profiling (e.g., ROS, Fe2⁺, lipid-ROS, and GSH) demonstrated that PP1A inhibits Lenvatinib-induced ferroptosis by dephosphorylating Keap1 at site 104. This disruption of the Keap1-Nrf2 interaction enhances the transcription of ferroptosis-related markers and immune checkpoint PD-L1. Notably, single-cell sequencing and co-culture experiments revealed that PP1A knockdown alleviates T cell exhaustion and immune evasion, thereby improving antitumor immunity. In vivo experiments further demonstrated that PP1A knockdown significantly enhances the efficacy of Lenvatinib-ICIs combination therapy. Overall, our findings highlight PP1A as a critical regulator of ferroptosis and antitumor immunity, suggesting its potential as a predictive biomarker and therapeutic target for improving outcomes in advanced HCC.
Collapse
Affiliation(s)
- Jitong Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Meng Gao
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Shikun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Wing-Wa Guo
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Wenzhi He
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Minghe Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Xi Chen
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Cairang Dongzhi
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Xiaomian Li
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
- TaiKang Center for Life and Medical Sciences, Wuhan University, Wuhan, 430071, P. R. China
| | - Weijie Ma
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Wuhan, 430071, P. R. China
- Clinical Medicine Research Center for Minimally Invasive Procedure of Hepatobiliary & Pancreatic Diseases of Hubei Province, Wuhan, 430071, P. R. China
| |
Collapse
|
22
|
Fu J, Chen X, Li J, Peng L. Research advances of Sappanone A in inflammation-related diseases. Front Med (Lausanne) 2025; 12:1569732. [PMID: 40406412 PMCID: PMC12095284 DOI: 10.3389/fmed.2025.1569732] [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: 02/01/2025] [Accepted: 04/21/2025] [Indexed: 05/26/2025] Open
Abstract
Sappanone A (SA), a kind of homoisoflavanone extracted from the dry heartwood of Caesalpinia sappan L., has been shown to possess diverse bioactivities involving anti-inflammatory, antioxidant, and anti-apoptotic properties. Sustained proinflammatory state is a major factor in the occurrence and development of various diseases. Given the characteristics of SA, many studies have explored the effect of SA on inflammation-related diseases, which uncovered the multifaceted therapeutic potential of SA in such diseases. In this mini-review, we summarized the current achievements of SA on inflammation-related diseases (such as myocardial ischemia-reperfusion injury, liver injury, respiratory diseases, and kidney injury, etc.), in order to provide useful insights into the role of SA in inflammation-related diseases and benefit future clinical applications.
Collapse
Affiliation(s)
- Jie Fu
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiu Chen
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jinglun Li
- Department of Neurology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lilei Peng
- Department of Neurosurgery, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
23
|
Li H, Jiao J, Gu Y, Zeng Y, Sheng Y. Risk factors and clinical outcomes in patients with HCV eradication by direct-acting antivirals: a systematic review and meta-analysis. Infect Dis (Lond) 2025:1-31. [PMID: 40333300 DOI: 10.1080/23744235.2025.2493370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/09/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND In hepatitis C patients with sustained virologic response (SVR) achieved after direct-acting antivirals (DAAs), the incidence of adverse clinical outcomes can be reduced but not completely eliminated. This meta-analysis aims at estimating the incidence of clinical outcomes in hepatitis C patients after achieving SVR with DAAs. METHODS Literature search was carried out in PubMed, Cochrane Library database, Web of Science, and Embase. The primary endpoint was the incidence of hepatocellular carcinoma (HCC) occurrence, HCC recurrence, decompensated cirrhosis, and liver-related mortality, following DAA-induced elimination of hepatitis C virus (HCV). Subgroup analyses were performed according to age, gender, comorbidities, region, fibrosis stage, presence of decompensation, duration of follow-up, start point of follow-up, and HCC treatment modality. Furthermore, meta-regression was performed to explore sources of high heterogeneity. RESULTS Finally, 132 articles were included in our study. The pooled HCC occurrence rate was 1.50/100 person-years (95% CI, 1.35-1.65), HCC recurrence rate was 17.00/100 person-years (95% CI, 13.83-20.42), decompensation rate was 0.30/100 person-years (95% CI, 0.16-0.48), and liver-related mortality was 0.32/100 person-years (95% CI, 0.14-0.56). Meta-regression showed that duration of follow-up and fibrosis grade were important contributors to HCC occurrence. Age, start point of follow-up, and duration of follow-up were important contributors to HCC recurrence rate. CONCLUSION Patients with DAA-induced HCV elimination remain at risk for adverse outcomes, particularly those with cirrhosis and HCC history. The exposure to adverse outcomes tended to decrease over time, and the frequency and intensity of follow-up might be reduced in the future, which will require new scoring models to identify these individuals.
Collapse
Affiliation(s)
- Hualing Li
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiahuan Jiao
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuyi Gu
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Zeng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yunjian Sheng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
24
|
Tsilimigras DI, Kurzrock R, Pawlik TM. Molecular Testing and Targeted Therapies in Hepatobiliary Cancers: A Review. JAMA Surg 2025; 160:576-585. [PMID: 40105823 DOI: 10.1001/jamasurg.2025.0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Importance Hepatobiliary cancers are heterogeneous and molecularly complex. Recent advances in next-generation sequencing (NGS) have enhanced the understanding of their molecular landscape and enabled deployment of biomarker-based gene- and immune-targeted therapies. This review examines the role of molecular testing and targeted therapies in these malignant neoplasms. Observations Patients with hepatobiliary cancers have poor outcomes. Precision oncology studies have shown that while many common molecular alterations are not currently targetable in hepatocellular carcinoma (HCC), a large number of actionable alterations characterize biliary tract cancers (BTCs), with several therapies now approved by the US Food and Drug Administration. Immunotherapy is increasingly adopted in clinical practice, either as monotherapy or combined with cytotoxic chemotherapy, for both HCC and BTCs. Moreover, multiple solid cancer tumor-agnostic therapies are approved (larotrectinib, entrectinib, and repotrectinib for NTRK fusions; selpercatinib for RET fusions; dabrafenib and trametinib combination for BRAF V600E mutations; dostarlimab or pembrolizumab for tumors with high microsatellite instability and pembrolizumab for tumor mutation burden ≥10 mutations/megabase), highlighting the need for NGS as well as ERBB2 (formerly HER2) immunohistochemistry (IHC) (with the recent approval of solid tissue-agnostic deruxtecan trastuzumab for ERBB2-positive [IHC 3+] cancer) across cancers. N-of-1 clinical trials using customized drug combinations matched to the tumor's molecular profile have yielded encouraging results and provide a promising framework for future clinical trial design. Conclusions and Relevance Molecular testing and gene- and immune-targeted therapies are transforming hepatobiliary cancer treatment. Tumor-agnostic and N-of-1 clinical trials have challenged traditional clinical trial paradigms and provide the foundation for truly personalized oncology for patients with these aggressive cancers. Further work is needed to determine how to leverage these novel approaches into the management of operable disease.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center and Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Milwaukee
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
- Deputy Editor, JAMA Surgery
| |
Collapse
|
25
|
Lin Z, Wang W, Yan Y, Ma Z, Xiao Z, Mao K. A deep learning-based clinical-radiomics model predicting the treatment response of immune checkpoint inhibitors (ICIs)-based conversion therapy in potentially convertible hepatocelluar carcinoma patients: a tumor marker prognostic study. Int J Surg 2025; 111:3342-3355. [PMID: 40085751 DOI: 10.1097/js9.0000000000002322] [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: 12/18/2024] [Accepted: 03/02/2025] [Indexed: 03/16/2025]
Abstract
BACKGROUND The majority of patients with hepatocellular carcinoma (HCC) miss the opportunity of radical resection, making immune check-point inhibitors (ICIs)-based conversion therapy a primary option. However, challenges persist in predicting response and identifying the optimal patient subset. The objective is to develop a CT-based clinical-radiomics model to predict durable clinical benefit (DCB) of ICIs-based treatment in potentially convertible HCC patients. METHODS The radiomics features were extracted by pyradiomics in training set, and machine learning models was generated based on the selected radiomics features. Deep learning models were created using two different protocols. Integrated models were constructed by incorporating radiomics scores, deep learning scores, and clinical variables selected through multivariate analysis. Furthermore, we analyzed the relationship between integrated model scores and clinical outcomes related to conversion therapy in the entire cohort. Finally, radiogenomic analysis was conducted on bulk RNA and DNA sequencing data. RESULTS The top-performing integrated model demonstrated excellent predictive accuracy with an area under the curve (AUC) of 0.96 (95% CI: 0.94-0.99) in the training set and 0.88 (95% CI: 0.77-0.99) in the test set, effectively stratifying survival risk across the entire cohort and revealing significant disparity in overall survival (OS), as evidenced by Kaplan-Meier survival curves ( P < 0.0001). Moreover, integrated model scores exhibited associations with sequential resection among patients who achieved DCB and pathological complete response (pCR) among those who underwent sequential resection procedures. Notably, higher radiomics model was correlated with MHC I expression, angiogenesis-related processes, CD8 T cell-related gene sets, as well as a higher frequency of TP53 mutations along with increased levels of mutation burden and neoantigen. CONCLUSION The deep learning-based clinical-radiomics model exhibited satisfactory predictive capability in forecasting the DCB derived from ICIs-based conversion therapy in potentially convertible HCC, and was associated with a diverse range of immune-related mechanisms.
Collapse
Affiliation(s)
- Zijian Lin
- Department of Hepatobiliary Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Weidong Wang
- Department of Interventional Radiography, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Yongcong Yan
- 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
| | - Zifeng Ma
- Shanghai Public Health Clinical Center, Shanghai, China
| | - Zhiyu 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
| | - Kai Mao
- 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
|
26
|
Zhao C, Qin G, Ling C, Zhao Y, Huang Y, Jiang Z, Zhou N, Liu J, Su D, Jiang J. MSNs-loaded HMME and Erastin-mediated ferroptosis combined with sonodynamic therapy for HCC treatment. J Cancer Res Ther 2025; 21:465-476. [PMID: 40317153 DOI: 10.4103/jcrt.jcrt_1531_24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/24/2025] [Indexed: 05/07/2025]
Abstract
BACKGROUND Ferroptosis can have a major impact on the development and advancement of hepatocellular carcinoma (HCC) due to its clear association with heightened vulnerability to the disease. This study aimed to develop a novel nanoplatform to evaluate its effectiveness in in vivo and in vitro models of HCC. METHODS Erastin, a compound that induces iron-dependent cell death, and HMME, a sonosensitizer, were enclosed within mesoporous silica nanoparticles (MSNs). The nanoparticles were engineered to exhibit a responsive assembly-disassembly mechanism. Hydrophilic hyaluronic acid (HA) was utilized for conjugation modification to synthesize Erastin/HMME@MSNs-HA. In vivo and in vitro experiments were conducted to elucidate the antitumor mechanisms of this nanomaterial. RESULTS In the in vitro cellular experiments, Erastin/HMME@MSNs-HA was rapidly degraded by hyaluronidase, leading to increased endocytosis of the cancer cells. Cellular breakdown led to the generation of harmful reactive oxygen species (ROS), decreased glutathione levels, and increased lipid peroxidation, resulting in a decrease in mitochondrial membrane potential, dysfunctional mitochondria, reduced cell growth, and increased cell death. Additionally, the Erastin/HMME@MSNs-HA nanotherapy platform, when combined with ultrasound (US) treatment, exhibited significant therapeutic effectiveness against tumors in vivo. It induced significant cell death in cancerous tissues, decreased tumor growth, worsened tissue oxygen deprivation, and exhibited good compatibility with the body. CONCLUSION These findings indicate that the nanoplatform can effectively alleviate tumor hypoxia while inducing apoptosis and ferroptosis, laying the foundation for enhancing the efficacy of ROS-mediated HCC therapy.
Collapse
Affiliation(s)
- Chang Zhao
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Guchun Qin
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Caixia Ling
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yang Zhao
- Department of Radiology, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Yunxi Huang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Zelong Jiang
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Niqiang Zhou
- Department of Interventional Therapy, Guangxi Medical University Cancer Hospital, Nanning, China
- Guangxi Key Laboratory of Early Prevention and Treatment for Regional High Frequency Tumor, Nanning, China
| | - Junjie Liu
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Danke Su
- Department of Imaging Center, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Jinghang Jiang
- Department of Medical Ultrasound, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
27
|
Meyer HJ, Leonhardi J, Potratz J, Jechorek D, Schramm KI, Borggrefe J, Surov A. Association between radiomics of diffusion-weighted imaging and histopathology in hepatocellular carcinoma. A preliminary investigation. Magn Reson Imaging 2025; 118:110356. [PMID: 39938670 DOI: 10.1016/j.mri.2025.110356] [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: 12/27/2024] [Revised: 02/06/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
OBJECTIVE Diffusion-weighted imaging and the quantified apparent diffusion coefficient (ADC) correlate with cell density and histopathological features in tumors. Radiomics analysis may provide more insight into the underlying microstructure and may better correlate with histopathology. The present study used cross-sectional guided biopsy specimens to exploit the precise spatial localization of the performed biopsy to correlate radiomics features of the ADC map with immunohistochemical features in hepatocellular carcinoma (HCC). MATERIALS AND METHODS A total of 51 patients (11 female patients, 21.6 %) were included in the present study. The mean age was 71.9 ± 9.9 years, ranging from 42 to 91 years. Prebioptic liver MRI with diffusion-weighted imaging was used to correlate the radiomics features of the ADC maps with the immunohistochemical features quantified in liver biopsy. Proliferation potential Ki 67, leukocyte count and tumor-stroma ratio were evaluated as histopathological parameters. RESULTS The following ADC texture features were correlated with the Ki 67 index _MinNorm (r = -0.307, p = 0.03), Vertl_RLNonUni (r = - 0.309, p = 0.03), 135dr_RLNonUni (r = -0.346, p = 0.01). The texture feature _MinNorm achieved the best diagnostic accuracy with an area under the curve of 0.76 (95 % CI 0.60-0.91, p < 0.01) to discriminate between low and high proliferative HCC. Multiple statistically significant correlations were found between ADC texture features and tumor-stroma-ratio, the highest for S(0,1)Contrast (r = 0.460, p = 0.001). No statistically significant correlations were found between the ADC texture features with the CD45+ leukocyte count and grading. CONCLUSION Radiomics features of the ADC maps can reflect the underlying histopathology in HCC patients including the proliferation potential and tumor-stroma ratio but not CD45 positive cells and tumor grading. The complex interactions between quantitative imaging and histopathology need to be further investigated in a validation cohort.
Collapse
Affiliation(s)
- Hans-Jonas Meyer
- Department of Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany.
| | - Jakob Leonhardi
- Department of Diagnostic and Interventional Radiology, University Hospital Leipzig, Leipzig, Germany
| | - Johann Potratz
- Department of Pathology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Dörthe Jechorek
- Department of Pathology, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Kai Ina Schramm
- Department of Radiology and Nuclear Medicine, Otto von Guericke University Magdeburg, Magdeburg, Germany
| | - Jan Borggrefe
- Institute for Radiology, Neuroradiology and Nuclear Medicine, Johannes Wesling University, Ruhr University Bochum, Minden, Germany
| | - Alexey Surov
- Institute for Radiology, Neuroradiology and Nuclear Medicine, Johannes Wesling University, Ruhr University Bochum, Minden, Germany
| |
Collapse
|
28
|
Sohail SK. Natural Products as Modulators of miRNA in Hepatocellular Carcinoma: A Therapeutic Perspective. J Gene Med 2025; 27:e70019. [PMID: 40296860 DOI: 10.1002/jgm.70019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2025] [Revised: 04/07/2025] [Accepted: 04/20/2025] [Indexed: 04/30/2025] Open
Abstract
Hepatocellular carcinoma (HCC) continues to pose a substantial worldwide health concern, marked by elevated mortality rates and restricted therapeutic alternatives. Recent studies have highlighted the potential of natural compounds as therapeutic agents in cancer management. This review focuses on the diagnostic and prognostic potential of microRNAs (miRNAs) as biomarkers in HCC, alongside the therapeutic promise of natural products. We explore the intricate role of miRNAs in the pathogenesis of HCC, detailing their regulatory functions in cellular processes such as proliferation, apoptosis, and metastasis. Additionally, we discuss the emerging evidence supporting the use of natural compounds, including phytochemicals, in modulating miRNA expression and their potential synergistic effects with conventional therapies. Key miRNAs discussed include miR-21, an oncogenic factor that promotes tumor growth by targeting the tumor suppressor phosphatase and tensin homolog (PTEN); miR-34a, which enhances apoptosis and may improve treatment efficacy when combined with c-MET inhibitors; miR-203, whose downregulation correlates with poor outcomes and may serve as a prognostic marker; miR-16, which acts as a tumor suppressor and has diagnostic potential when measured alongside traditional markers like alpha-fetoprotein (AFP); and miR-483-3p, associated with resistance to apoptosis and tumor progression. By integrating insights from recent studies, this review aims to highlight the dual role of miRNAs as both biomarkers and therapeutic targets, paving the way for enhanced diagnostic strategies and novel treatment modalities in HCC management.
Collapse
|
29
|
Zhang L, Wang D, Zhang LZ, Yang WH, Yu C, Qin J, Feng LZ, Liu Z, Teng GJ. Pickering emulsion with tumor vascular destruction and microenvironment modulation for transarterial embolization therapy. Biomaterials 2025; 316:123018. [PMID: 39709852 DOI: 10.1016/j.biomaterials.2024.123018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
In the clinic, Lipiodol chemotherapeutic emulsions remain a main choice for patients diagnosed with hepatocellular carcinoma (HCC) via the mini-invasive transarterial chemoembolization (TACE) therapy. However, the poor stability of conventional Lipiodol chemotherapeutic emulsions would result in the fast drug diffusion and incomplete embolization, inducing systemic toxicity and impairing the efficacy of TACE therapy. Therefore, it is of great importance to construct alternative formulations based on commercial Lipiodol to achieve the improved efficacy and safety of HCC treatment. Herein, calcium phosphate (CaP) nanoparticles-stabilized Lipiodol Pickering emulsion (CaP-LPE) with improved stability and pH-responsiveness is prepared and utilized for the encapsulation of combretastatin A4-phosphate (CA4P), a clinically approved vascular disrupting agent. The obtained CA4P-loaded CaP-LPE (CCaP-LPE) was shown to be enhanced stability compared to conventional Lipiodol emulsion and pH-responsive release of the encapsulated drugs. On one hand, the released CA4P could disrupt tumor vascular and cut off the blood supplying of tumor cells, thus starving cancer cells. Moreover, it was revealed that CCaP-LPE could reverse immunosuppressive tumor microenvironment (TME) by neutralizing tumor acidity, leading to the increased infiltration of CD8+ T cells and the decreased percentages of immunosuppressive cells. As the result, such CCaP-LPE could effectively shrink orthotopic N1S1 HCC tumors in rats by eliciting a potent antitumor immune response. Therefore, this study highlights a simple strategy to construct a novel LPE with the potencies of tumor vascular disruption and TME modulation, holding a great promise for TAE therapy of HCC.
Collapse
Affiliation(s)
- Lei Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Duo Wang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Lin-Zhu Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Wei-Hao Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chao Yu
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Juan Qin
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Liang-Zhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
30
|
Lu R, Long X, Li X, Li J, Liu Z, Chai K, Zhang Y, Lin Y, Liu Z, Tima S, Zhong Z, Sun X. Icaritin-Loaded Liposomes Mediated by Hyaluronic Acid Promoted the Anti-Proliferation and Senescence in Huh7 Cells. Drug Dev Res 2025; 86:e70104. [PMID: 40347050 DOI: 10.1002/ddr.70104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/17/2025] [Accepted: 04/27/2025] [Indexed: 05/12/2025]
Abstract
Icaritin (ICT) shows great potential in cancer therapy. To enhance the cancer-fighting properties of icaritin against hepatocellular carcinoma (HCC), we developed icaritin-loaded liposomes modified with hyaluronic acid (HA-Lip-ICT). We employed statistical design methods to analyze how various factors affected particle dimensions and drug encapsulation, creating an optimized HA-Lip-ICT formulation that could effectively suppress HCC cell growth and trigger cellular aging. The human HCC cell line Huh7 was then exposed to different icaritin preparations. We assessed tumor cell viability through multiple assays, including colony formation and DNA synthesis measurements. Our results demonstrated that the refined HA-Lip-ICT significantly impaired HCC cell proliferation. Moreover, at a concentration of 10 μmol/L, HA-Lip-ICT markedly accelerated cellular senescence in HCC cells. These observations support our initial hypothesis that HA-Lip-ICT can inhibit HCC cell growth and promote their aging. While further research is needed to elucidate the exact mechanisms, this approach shows the promise of HA-Lip-ICT as a targeted therapy for improving the HCC treatments.
Collapse
Affiliation(s)
- Ruilin Lu
- Suining First People's Hospital, Suining, China
| | - Xin Long
- School of Clinical Medical Sciences, Southwest Medical University, Luzhou, China
| | - Xiyao Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Jiaxin Li
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhen Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Kexin Chai
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yujie Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Yan Lin
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Zhongbing Liu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Singkome Tima
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
| | - Zhirong Zhong
- Key Laboratory of Medical Electrophysiology, Ministry of Education, School of Pharmacy, Southwest Medical University, Luzhou, China
- Key Laboratory of Luzhou City for Aging Medicine, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, China
| | - Xiaoduan Sun
- Department of Medical Technology, Faculty of Associated Medical Sciences, Chiang Mai University, Chiang Mai, Thailand
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, China
| |
Collapse
|
31
|
Altaf A, Khalil M, Akabane M, Rashid Z, Kawashima J, Zindani S, Ruzzenente A, Ratti F, Marques H, Cauchy F, Lam V, Poultsides G, Aucejo F, Kitago M, Popescu I, Martel G, Gleisner A, Bauer TW, Hugh T, Bhimani N, Shen F, Endo I, Pawlik TM. Up-front resection for hepatocellular carcinoma: Assessing futility in the preoperative setting. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109594. [PMID: 39826445 DOI: 10.1016/j.ejso.2025.109594] [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: 12/03/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE We sought to develop a predictive model to preoperatively identify patients with hepatocellular carcinoma (HCC) at risk of undergoing futile upfront liver resection (LR). METHODS Patients undergoing curative-intent LR for HCC were identified from a large multi-institutional database. Futile LR was defined by death or disease recurrence within six months postoperatively. Backward logistic regression was performed to identify factors associated with futility. Additionally, binary criteria were established for surgical candidacy, aiming to keep the likelihood of futility below 20 %. RESULTS Among 1633 patients with HCC, 264 (16.2 %) underwent futile upfront LR. Tumor burden score (TBS) (coefficient: 0.083, 95%CI: 0.067-0.099), alpha-fetoprotein (AFP) (coefficient: 0.254, 95%CI: 0.195-0.310), and albumin-bilirubin (ALBI) grade 2/3 (coefficient: 0.566, 95%CI: 0.420-0.718) were independently associated with an increased risk of futile LR. The model demonstrated strong discrimination and calibration in both derivation and validation cohorts. Low, intermediate, and high-risk groups were determined based on the risk model, each with an escalating likelihood of futility, worse histological features, and worse survival outcomes. Six distinct conditions based on AFP-adjusted-to-TBS criteria were established, all with a futility likelihood of less than 20 %. Patients fulfilling these criteria had significantly better long-term recurrence-free and overall survival. The futility risk model was made available online for wide clinical applicability: (https://altaf-pawlik-hcc-futilityofsurgery-calculator.streamlit.app/). CONCLUSION A preoperative risk model and AFP-adjusted-to-TBS criteria were developed and validated to predict the likelihood of futile LR among patients with HCC. This pragmatic clinical tool may assist clinicians in preoperative decision-making, helping them avoid futile surgery unlikely to offer long-term benefits.
Collapse
Affiliation(s)
- Abdullah Altaf
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Mujtaba Khalil
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Miho Akabane
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Zayed Rashid
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Jun Kawashima
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Shahzaib Zindani
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | | | | | - Hugo Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - François Cauchy
- Department of Hepatobiliopancreatic Surgery, APHP, Beaujon Hospital, Clichy, France
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Sydney, NSW, Australia
| | - George Poultsides
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Federico Aucejo
- Department of Surgery, Cleveland Clinic., Cleveland, OH, United States
| | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ana Gleisner
- Department of Surgery, University of Colorado, Aurora, CO, United States
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Tom Hugh
- Department of Surgery, School of Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Nazim Bhimani
- Department of Surgery, School of Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Feng Shen
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Itaru Endo
- Department of Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States.
| |
Collapse
|
32
|
Kawashima J, Akabane M, Khalil M, Woldesenbet S, Endo Y, Sahara K, Ruzzenente A, Ratti F, Marques HP, Oliveira S, Balaia J, Cauchy F, Lam V, Poultsides GA, Kitago M, Popescu I, Martel G, Gleisner A, Hugh T, Weiss M, Aucejo F, Aldrighetti L, Endo I, Pawlik TM. Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden (MELD-AFP-TBS) score to stratify prognosis after liver resection for hepatocellular carcinoma. Surgery 2025; 183:109388. [PMID: 40311416 DOI: 10.1016/j.surg.2025.109388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/06/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Morphologic criteria, such as the Barcelona Clinic Liver Cancer staging system often fail to accurately predict long-term survival among patients undergoing liver resection for hepatocellular carcinoma. We sought to develop a continuous risk score that incorporates established markers of tumor biology and liver function to improve the prediction of overall survival. METHODS Data from a multi-institutional database were used to identify patients who underwent curative-intent hepatectomy for hepatocellular carcinoma. A predictive score for overall survival was developed using weighted beta-coefficients from a multivariable Cox regression model. RESULTS Among 850 patients, 595 (70.0%) were assigned to the training cohort, and 255 (30.0%) to the test cohort. In the training cohort, multivariable analysis identified the Model of End-Stage Liver Disease (hazard ratio, 1.04; 95% confidence interval, 1.01-1.07), log-transformed alpha-fetoprotein (hazard ratio, 1.07; 95% confidence interval, 1.02-1.13), and tumor burden score (hazard ratio, 1.07; 95% confidence interval, 1.03-1.11) as independent predictors of worse overall survival. The Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score, based on the Cox model, stratified patients into low-risk (n = 466, 78.3%) with a 5-year OS of 70.5% and high-risk (n = 129, 21.7%) with a 5-year OS of 47.0% (P < .001). In the test cohort, the Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score demonstrated superior discriminative accuracy (C-index: 0.72, time-dependent area under the curve 1-year: 0.80, 3-year 0.76, 5-year 0.70) compared with the Barcelona Clinic Liver Cancer staging system (C-index: 0.53, time-dependent area under the curve 1-year: 0.61, 3-year 0.55, 5-year 0.56). An online tool was made accessible at https://jk-osu.shinyapps.io/MELD_AFP_TBS/. CONCLUSION The Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score provides a novel, accurate tool for prognostic stratification of patients with hepatocellular carcinoma, identifying high-risk patients who may benefit from alternative treatments to improve outcomes.
Collapse
Affiliation(s)
- Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH; Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Mujtaba Khalil
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Yutaka Endo
- Department of Transplant Surgery, University of Rochester Medical Center, Rochester, NY
| | - Kota Sahara
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Andrea Ruzzenente
- Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | | | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Sara Oliveira
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Jorge Balaia
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - François Cauchy
- Department of HPB Surgery and Liver Transplantation, Beaujon Hospital, Clichy, France
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | | | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ana Gleisner
- Department of Surgery, University of Colorado Denver, Denver, CO
| | - Tom Hugh
- Department of Surgery, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew Weiss
- Department of Surgery, Cancer Institute, Northwell Health, New Hyde Park, NY
| | - Federico Aucejo
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, OH
| | | | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| |
Collapse
|
33
|
Yu X, Zhang Q, Wang L, Zhang Y, Zhu L. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 2025; 14:62. [PMID: 40307921 PMCID: PMC12044934 DOI: 10.1186/s40164-025-00658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, No. 10 Qinyun Nan Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qin Zhang
- Department of Postgraduate Students, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Leibo Wang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital Guiyang, Guiyang, 550000, Guizhou, The People's Republic of China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
34
|
Zhang CY, Gu K, Chi MY, Gao XY, Gao L, Zhang NN, Liu YX, Li TZ. The application progress of PAMAM dendrimer in cancer imaging and treatment. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-38. [PMID: 40293953 DOI: 10.1080/09205063.2025.2497623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 12/03/2024] [Indexed: 04/30/2025]
Abstract
Polyamidoamine dendrimer (PAMAM) are effective carriers that transport diagnostic imaging reagents and drugs to the tumor site. Their excellent bio-compatibility and bio-degradability reduce damage to healthy tissues, resulting in improved treatment efficacy. Dendrimer molecules are particularly useful in targeted drug delivery within malignant cells. This article reviews recent progress of PAMAM in imaging and treating breast cancer, lung cancer, hepatocellular cancer, colorectal cancer, gastric cancer, prostate cancer, and glioblastoma. This review aims to provide new and feasible ideas for cancer diagnosis imaging and treatment while also serving as a significant reference point for personalized tumor therapy based on PAMAM materials.
Collapse
Affiliation(s)
- Cong-Ying Zhang
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Kai Gu
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Meng-Yi Chi
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Xiao-Yan Gao
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Ling Gao
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Nan-Nan Zhang
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| | - Yu-Xi Liu
- Shaanxi Academy of Traditional Chinese Medicine, Shaanxi, China
| | - Tian-Zhu Li
- Key Laboratory of Research on Human Genetic Diseases at Universities of Inner Mongolia Autonomous Region, Chifeng University, Inner Mongolia, China
- Basic Medical College, Chifeng University, Inner Mongolia, China
- Key Laboratory of Mechanism and Evaluation of Traditional Chinese & Mongolian Medicine, Chifeng University, Inner Mongolia, China
| |
Collapse
|
35
|
Tu L, Luo J, Yin Y, Yu H. Bioinformatics analysis across pan-cancer and experimental validation in hepatocellular carcinoma revealed the oncogenic role of SF3B6. Front Pharmacol 2025; 16:1516534. [PMID: 40356980 PMCID: PMC12067594 DOI: 10.3389/fphar.2025.1516534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Background Splicing factor 3b subunit 6(SF3B6), a subunit of the SF3B complex, regulates the process of RNA splicing by recognizing the branch point adenosine in pre-mRNA and facilitating the interaction between U2 snRNA and the branch point sequence. Currently, there is no systematic multi-omics study exploring the diagnostic, prognostic, and immunotherapy predictive value of SF3B6 in pan-cancer, nor is its role in hepatocellular carcinoma (HCC) clear. Methods We utilized various databases to systematically examine the expression and genetic variation of SF3B6 across multiple cancer types, assessing its relationship with diagnosis, prognosis, immune infiltration, immunotherapy response, and associated signaling pathways. Additionally, we investigated the correlation between SF3B6 and prognosis, clinicopathological features, and treatment responses in HCC, as well as the roles of its related alternative splicing isoforms. Finally, we conducted in vitro experiments to validate the effects of SF3B6 on the proliferation, migration, invasion, apoptosis, and cell cycle progression of liver cancer cells. Results Results indicate that SF3B6 was highly expressed in various cancers and regulated by copy number variations and DNA methylation. The elevated expression of SF3B6 demonstrated predictive value for cancer diagnosis, prognosis, and responses to immunotherapy. Functional enrichment analysis suggests that SF3B6 was closely associated with pathways related to tumor immunity, tumor metabolism, and cell cycle. Additionally, high SF3B6 expression was an independent risk factor for overall survival and correlated with poor alpha-fetoprotein levels, pathological grading, clinical staging, and reduced responses to sorafenib and transarterial chemoembolization treatment in HCC. Interestingly, SF3B6 was associated with variant splicing isotypes of genes involved in the G2M checkpoint and DNA repair pathways, including NEIL3, NEK2, KIF4A, TROAP, and FANCD2. Moreover, SF3B6 was highly expressed in liver cancer cells, promoting the proliferation, migration, and invasion of cancer cells, inhibiting apoptosis, and regulating the transition from the S phase to the G2M phase of the cell cycle. Conclusion We emphasize that SF3B6 has the potential to serve as a biomarker for predicting cancer diagnosis, prognosis, and immunotherapy responses, especially in HCC. SF3B6 and its related alternative splicing isoforms promote the occurrence and progression of HCC and may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Linshuang Tu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiefu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya Yin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huihong Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
36
|
Li L, Xu L, Liao W, Wang P, Xu M, Li B, Zhang M. circCEP70 encoded protein inhibits the progression of hepatocellular carcinoma. Cell Mol Life Sci 2025; 82:174. [PMID: 40272569 PMCID: PMC12022199 DOI: 10.1007/s00018-025-05651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 04/25/2025]
Abstract
Cirrhosis is closely related to hepatocellular carcinoma (HCC), however, the regulation of circular RNA (circRNA) in HCC with cirrhotic background has not yet been well illustrated. In this study, high throughput circRNA sequencing was applied to identified candidate circRNAs in HCC samples with cirrhotic background. The biological function of candidate circRNA was validated in both in vitro and in vivo settings. Additionally, Alphafold 3, mass spectrometry analysis and immunofluorescence were employed to investigate the underlying mechanisms involved. We found circCEP70 exhibited significantly higher expression levels in cirrhotic HCC samples and showed a positive correlation with improved prognosis. The RNA binding protein U2AF2 was found to suppress the expression of circCEP70 in cirrhosis patients. In vitro and in vivo experiments, including CCK-8, EdU, plate cloning, transwell, scratch, subcutaneous tumor formation, liver metastasis in situ, and lung metastasis assays confirmed the anti-carcinogenic effects. Mechanistically, circCEP70 encoded a novel protein named CEP70-160aa, which interacted with PKM2 and hindered its translocation into the nucleus. This interaction led to reduce STAT3 phosphorylation in the nucleus, thus inhibiting HCC proliferation and metastasis. In cirrhotic microenvironment, circCEP70 prevented HCC proliferation and metastasis through PKM2/STAT3 axis, and RNA binding protein U2AF2 could inhibit circCEP70 expression.
Collapse
Affiliation(s)
- Lian Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangliang Xu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenwei Liao
- Department of Thoracic Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Peng Wang
- Department of Burns, Sichuan Academy of Medical Science, Sichuan Provincial People'S Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mingqing Xu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Hepato-Pancreato-Biliary Surgery, Meishan City People'S Hospital, Meishan Hospital of West China Hospital, Sichuan University, Meishan, 620010, China
| | - Bo Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ming Zhang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
37
|
Wilcox S, Sengupta S, Huang C, Tokuda J, Lu A, Woodrum D, Chen Y. Development of a Low-Profile, Piezoelectric Robot for MR-Guided Abdominal Needle Interventions. Ann Biomed Eng 2025:10.1007/s10439-025-03719-w. [PMID: 40266438 DOI: 10.1007/s10439-025-03719-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: 12/08/2024] [Accepted: 03/25/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE Minimally invasive needle-based interventions are commonly used in cancer diagnosis and treatment, including procedures, such as biopsy, brachytherapy, and microwave ablation. Although MR-guided needle placement offers several distinct advantages, such as high-resolution target visualization and accurate device tracking, one of the primary limitations that affect its widespread adoption is the ergonomic constraints of the closed-bore MRI environment, requiring the patients to be frequently moved in and out to perform the needle-based procedures. This paper introduces a low-profile, body-mounted, MR-guided robot designed to address this limitation by streamlining the operation workflow and enabling accurate needle placement within the MRI scanner. METHODS The robot employs piezoelectric linear actuators and stacked Cartesian XY stages to precisely control the position and orientation of a needle guide. A kinematic model and control framework was developed to facilitate accurate targeting. Additionally, clinical workflow for the liver interventions was developed to demonstrate the robot's capability to replicate existing procedures. The proposed system was validated in benchtop environment and 3T MRI scanner to quantify the system performance. RESULTS Experimental validations conducted in free space demonstrated a position accuracy of 2.38 ± 0.94 mm and orientation error of 1.40 ± 2.89°. Additional tests to confirm MR-conditionality and MR-guided phantom placements were carried out to assess the system's performance and safety in MRI suite, yielding a position error of 2.01 ± 0.77 mm and an orientation error of 1.57 ± 1.31°. CONCLUSION The presented robot shows exceptional compatibility with a wide range of patients and bore sizes while maintaining clinically significant accuracy. Future work will focus on the validations in dynamic liver environments.
Collapse
Affiliation(s)
- Samuel Wilcox
- Institute of Robotics and Intelligent Machines, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, 30332, USA
| | - Saikat Sengupta
- Vanderbilt University Institute of Imaging Science, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Chuan Huang
- Department of Radiology and Imaging Sciences, Emory University, Atlanta, GA, 30322, USA
| | - Junichi Tokuda
- Department of Radiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, 02115, USA
| | - Aiming Lu
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - David Woodrum
- Department of Radiology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Yue Chen
- Institute of Robotics and Intelligent Machines, Georgia Institute of Technology, Atlanta, GA, 30332, USA.
- Department of Biomedical Engineering, Georgia Institute of Technology/Emory University, Atlanta, GA, 30332, USA.
| |
Collapse
|
38
|
Li X, Lu S, Huang CK. The Complexity of SIRT2 in Chronic Liver Disease: Liver SIRT2 Promotes Hepatocellular Carcinoma Development. Cell Mol Gastroenterol Hepatol 2025:101512. [PMID: 40280175 DOI: 10.1016/j.jcmgh.2025.101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Xinjian Li
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Chiung-Kuei Huang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
39
|
Xie L, Song D, Lan J, Liu P, Qin S, Ning Y, Liu Q. Plasma protein levels and hepatocellular carcinoma: a Mendelian randomization study with drug screening implications. Discov Oncol 2025; 16:567. [PMID: 40252200 PMCID: PMC12009266 DOI: 10.1007/s12672-025-02307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a significant cause of cancer-related mortality, highlighting the need for novel therapeutic strategies. Identifying key proteins and potential therapeutic agents is critical for improving treatment outcomes. METHODS We employed Mendelian randomization to identify proteins associated with HCC risk and utilized drug enrichment and molecular docking analyses to discover potential therapeutic agents. The efficacy of identified drugs was evaluated in vitro using immune-tumor co-culture systems and in vivo in a murine HCC model. Single-cell expression profiling and clinical sample analyses were conducted to explore expression patterns. RESULTS Our analyses identified 16 proteins linked to HCC pathogenesis. Among the therapeutic agents tested, Belinostat significantly enhanced T cell-mediated cytotoxicity against HCC cells and effectively reduced tumor growth in vivo. Single-cell analysis revealed significant modulation of immune cells within the tumor microenvironment, suggesting potential mechanisms for the observed therapeutic effects. CONCLUSION This study highlights the potential of Belinostat as a promising therapeutic agent for HCC. By modulating immune responses and tumor growth, Belinostat offers a novel approach to HCC treatment, warranting further clinical investigation to validate its efficacy and therapeutic potential.
Collapse
Affiliation(s)
- Longhui Xie
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Yongzhou Central Hospital, Yongzhou, Hunan, China
| | - Dekun Song
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, China
| | - Jianwei Lan
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Pengpeng Liu
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuang Qin
- Yongzhou Central Hospital, Yongzhou, Hunan, China
| | - Yinkuan Ning
- Department of Interventional Vascular Surgery, Shaoyang Central Hospital Shaoyang, Shaoyang, Hunan, China.
| | - Quanyan Liu
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
40
|
Li J, Yong T, Chen Y, Zeng T, Zhang K, Wang S, Zhang Y. Targeting PCNA/PARP1 axis inhibits the malignant progression of hepatocellular carcinoma. Front Pharmacol 2025; 16:1571786. [PMID: 40313621 PMCID: PMC12043649 DOI: 10.3389/fphar.2025.1571786] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025] Open
Abstract
Introduction Proliferating cell nuclear antigen (PCNA) is associated with the proliferation and recurrence of various cancers, and its high expression is associated with poor prognosis in hepatocellular carcinoma (HCC) patients. However, the mechanistic role of PCNA in HCC progression remains poorly understood. This study aimed to investigate how PCNA regulates DNA damage repair and cell cycle progression in HCC, with a focus on its interaction with poly (ADP-ribose) polymerase 1 (PARP1) and therapeutic implications. Methods PCNA was targeted genetically and pharmacologically in HCC cells to assess its effects on DNA damage repair and cell cycle arrest. Protein-protein interactions between PCNA and PARP1 were validated through co-immunoprecipitation and functional assays. The sensitivity of HCC cells to the PARP1 inhibitor Olaparib was evaluated under PCNA inhibition. Synergistic effects of AOH1160 (a PCNA inhibitor) and Olaparib were tested in vitro and in vivo using proliferation assays, DNA damage quantification, and cell cycle analysis. Prognostic relevance of PCNA expression was analyzed using TCGA datasets. Results Targeting PCNA suppressed DNA damage repair and induced cell cycle arrest in HCC cells. Mechanistically, PARP1 was identified as a downstream target of PCNA and directly interacted with PCNA. Inhibiting the expression or activity of PCNA increased the sensitivity of HCC cells to the PARP1 inhibitor, Olaparib. In addition, AOH1160 and Olaparib synergistically inhibited the proliferation, DNA damage repair and cell cycle progression of HCC cells. Elevated PCNA levels correlated with unfavorable HCC prognosis, supporting its role as a therapeutic biomarker. In vivo experiments also confirmed that repression of the PCNA/PARP1 axis significantly reduced HCC tumor growth. Discussion This study elucidates the relationship between PCNA and PARP1 in regulating the malignant progression of HCC, and highlight the pivotal role of PCNA/PARP1 axis in DNA damage repair and cell cycle progression. The correlation between elevated PCNA levels and unfavorable prognosis underscores its potential as a therapeutic biomarker. Repression of PCNA/PARP1 axis significantly inhibits the malignant proliferation of HCC cells both in vitro and in vivo. Collectively, the study provides a mechanistic foundation for therapies targeting PCNA/PARP1 axis.
Collapse
Affiliation(s)
- Jipin Li
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Tao Yong
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Yali Chen
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Tingyu Zeng
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Kaifeng Zhang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Shuping Wang
- Key Laboratory of Preclinical Study for New Drugs of Gansu Province, Institute of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Youcheng Zhang
- Department of General Surgery, The Second Hospital of Lanzhou University, Lanzhou, China
| |
Collapse
|
41
|
Granata V, Fusco R, Setola SV, Borzacchiello A, Della Sala F, Rossi I, Ravo L, Albano D, Vanzulli A, Petrillo A, Izzo F. Treatments and cancer: implications for radiologists. Front Immunol 2025; 16:1564909. [PMID: 40308594 PMCID: PMC12040653 DOI: 10.3389/fimmu.2025.1564909] [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: 01/22/2025] [Accepted: 03/24/2025] [Indexed: 05/02/2025] Open
Abstract
This review highlights the critical role of radiologists in personalized cancer treatment, focusing on the evaluation of treatment outcomes using imaging tools like Computed Tomography (CT), Magnetic Resonance Imaging (MRI), and Ultrasound. Radiologists assess the effectiveness and complications of therapies such as chemotherapy, immunotherapy, and ablative treatments. Understanding treatment mechanisms and consistent imaging protocols are essential for accurate evaluation, especially in managing complex cases like liver cancer. Collaboration between radiologists and oncologists is key to optimizing patient outcomes through precise imaging assessments.
Collapse
Affiliation(s)
- Vincenza Granata
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale – IRCCS di Napoli, Naples, Italy
| | - Roberta Fusco
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale – IRCCS di Napoli, Naples, Italy
| | - Sergio Venanzio Setola
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale – IRCCS di Napoli, Naples, Italy
| | - Assunta Borzacchiello
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Naples, Italy
| | - Francesca Della Sala
- Institute of Polymers, Composites and Biomaterials, National Research Council (IPCB-CNR), Naples, Italy
| | - Ivano Rossi
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale – IRCCS di Napoli, Naples, Italy
| | - Ludovica Ravo
- Division of Radiology, Università degli Studi di Napoli Federico II, Naples, Italy
| | - Domenico Albano
- Diagnostic and Interventional Radiology Unit, IRCCS Istituto Ortopedico Galeazzi, Milan, Italy
- Dipartimento di Scienze Biomediche, Chirurgiche ed Odontoiatriche, Università degli Studi di Milano, Milano, Italy
| | - Angelo Vanzulli
- Department of Radiology, ASST Grande Ospedale Metropolitano Niguarda, Milan, Italy
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano, Milan, Italy
| | - Antonella Petrillo
- Division of Radiology, Istituto Nazionale Tumori IRCCS Fondazione Pascale – IRCCS di Napoli, Naples, Italy
| | - Francesco Izzo
- Division of Epatobiliary Surgical Oncology, Istituto Nazionale Tumori IRCCS Fondazione Pascale—IRCCS di Napoli, Naples, Italy
| |
Collapse
|
42
|
Jin S, Zhao Q, Sun X, Su J, Wang P, Li P, Guo J, Zhang Y, Zong H, Gan X. L-741626 inhibits hepatocellular carcinoma progression by targeting Ref-1 to suppress MAPK/ERK signalling pathway activity. Biol Direct 2025; 20:54. [PMID: 40241114 PMCID: PMC12001403 DOI: 10.1186/s13062-025-00624-z] [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: 10/21/2024] [Accepted: 02/24/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common and challenging malignancy of the digestive tract. Unfortunately, patients with advanced HCC frequently experience limited long-term benefits from current treatments, highlighting the critical need for innovative therapeutic agents. The discovery and development of new small-molecule compounds that target tumours have become crucial aspects of cancer research. In this study, we report on L-741626, a compound that has significant inhibitory effects on HCC. Both in vivo and in vitro experiments confirmed that L-741626 inhibited the growth of HCC by suppressing the MAPK/ERK signalling pathway. Molecular docking simulations and drug affinity responsive target stability assays further identified redox Factor 1 (Ref-1) as a target of L-741626. Ref-1 is overexpressed in HCC and is correlated with poor prognosis and high stage. Further studies demonstrated that Ref-1 interacts with CRAF, a crucial component of the MAPK/ERK signalling pathway. Knockdown of Ref-1 in HCC cells led to inhibition of the MAPK/ERK pathway. Sorafenib is a well-established targeted therapy for the treatment of HCC, with its primary antitumor mechanism being the inhibition of the MAPK/ERK signalling pathway. However, the presence of tumor stem cells is a key factor contributing to resistance to sorafenib. Our study demonstrates that L-741626 can suppress tumor stemness in HCC. The combination of L-741626 and sorafenib significantly enhances the sensitivity of HCC, resulting in increased tumoricidal effects. Our findings reveal a novel pharmacological effect of L-741626, which inhibits MAPK/ERK signalling activity in HCC by targeting Ref-1. Furthermore, L-741626 exhibits a synergistic effect when combined with sorafenib, suggesting a new potential approach for HCC treatment.
Collapse
Affiliation(s)
- Shuiling Jin
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China.
| | - Qi Zhao
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Xiao Sun
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jinsong Su
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, Henan, China
| | - Peiwen Wang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Peixian Li
- Tianjian Laboratory of Advanced Biomedical Sciences, Academy of Medical Sciences, Zhengzhou University, Zhengzhou, China
| | - Jing Guo
- China-US (Henan) Hormel Cancer Institute, No.127, Dongming Road, Jinshui District, Zhengzhou, 450008, Henan, China
| | - Yibing Zhang
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Hong Zong
- Department of Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China.
| | - Xiaoli Gan
- School of Life Sciences, Zhengzhou University, Zhengzhou, 450001, China.
| |
Collapse
|
43
|
Pervez A, Khan B, Khan GN, Khattak S, Ali M, Mujeeb K, Nasib B, Kim HG, Qureshi IZ, Arshad M. Evaluation of hepatic cancer stem cells (CD 73+, CD 44+, and CD 90+) induced by diethylnitrosamine in male rats and treatment with biologically synthesized silver nanoparticles. Mol Biol Rep 2025; 52:393. [PMID: 40232523 DOI: 10.1007/s11033-025-10495-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 04/16/2025]
Abstract
BACKGROUND Cancer stem cells (CSCs) play a critical role in the initiation and heterogeneity of a variety of cancers due to their pluripotent nature and capacity for asymmetric cell division. Therefore, uncovering the carcinogens that increase the CSC population in target tissues is crucial for understanding the genesis of cancer. The therapeutic potential of Operculina turpethum (OT) derived silver nanoparticles (AgNPs) was assessed in diethylnitrosamine (DEN)-induced CSC populations; CD73+, CD44+, and CD90 + of hepatic tissues in male rats. METHODS Histopathology, fluorescence-activated cell sorting (FACS), and RT-qPCR were performed on the control, DEN, DEN + AgNPs, and AgNPs-treated groups. AgNPs were characterized by FTIR, EDX, XRD, and SEM. RESULTS AgNPs were confirmed by intense surface plasmon resonance at 425 nm. Antioxidants, the reducing sugars responsible for Ag+ 1 reduction and subsequent conjugate formation with nanoparticles, were confirmed by vibrational spectra. The spherical morphology, composition, and conjugation of silver nanoparticles to phytoconstituents with partially crystalline, face-centered cubic structure were established through SEM, EDX spectrum, and XRD, respectively. Disrupted tissue architecture, cell enlargement, mild pleomorphism, and expanded central veins were observed in hepatic tissues of DEN-treated animals. However, a moderate inflammatory response was observed in the DEN + AgNPs-treated group. CSC populations were significantly increased in the DEN-treated group, but decreased with AgNPs-treatment. The mRNA expression levels of CD90, CD44, and CD73 genes were significantly up-regulated in the DEN-treated group compared to control group however, in DEN + AgNPs and AgNPs groups it were similar to control group. CONCLUSION All together, DEN-induced the hepatic CSC cell populations and the OT mediated AgNPs have therapeutic potential to attenuate the harmful effects of DEN. This study provides evidenced that OT-synthesised AgNPs may be considered as a therapeutic agent for liver related malignancies.
Collapse
Affiliation(s)
- Amber Pervez
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Behramand Khan
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Gul Nabi Khan
- Department of Zoology, Islamia College University, Peshawar, Pakistan.
| | - Sumayya Khattak
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Mazhar Ali
- Department of Zoology, Islamia College University, Peshawar, Pakistan
| | - Komal Mujeeb
- Department of Chemistry, Islamia College University, Peshawar, Pakistan
| | - Bushra Nasib
- Institute of Pathology and Diagnostic Medicine, Khyber Medical University, Peshawar, Pakistan
| | - Hyung Goo Kim
- Department of Neurosurgery, Robert Wood Johnson Medical School, The State University of New Jersey, Rutgers, USA
| | | | - Muhammad Arshad
- Biochemistry Section, Jhang Campus, University of Veterinary and Animal Sciences, Lahore, Pakistan.
| |
Collapse
|
44
|
Liang LW, Luo RH, Huang ZL, Tang LN. Clinical observation of nivolumab combined with cabozantinib in the treatment of advanced hepatocellular carcinoma. World J Gastrointest Oncol 2025; 17:103631. [PMID: 40235875 PMCID: PMC11995320 DOI: 10.4251/wjgo.v17.i4.103631] [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: 11/26/2024] [Revised: 12/25/2024] [Accepted: 02/07/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a particularly serious kind of liver cancer. Liver cancer ranks third in terms of mortality rate worldwide, putting it among the leading causes of deaths from cancer. HCC is the primary kind of liver cancer and makes up the vast majority of cases, accounting for approximately 90% of occurrences. Numerous research have verified this information. the progress of fatty liver, alcohol induced cirrhosis, smoking habits, obesity caused by overweight, and metabolic diseases such as diabetes. The treatment strategies for HCC can be divided into two categories: One is curative treatment, including liver transplantation, surgical resection, and ablation therapy or selective arterial radiation embolization, aimed at completely eliminating the lesion; Another type is non curative treatment options, including transarterial chemoembolization and systemic therapy, which focus on controlling disease progression and prolonging patient survival. The majority of HCC patients are found to be in an advanced stage and need systemic therapy. Sorafenib and lenvatinib are frequently used as first-line medications in traditional HCC treatment to slow the disease's progression. For second-line treatment, regorafenib, cabozantinib, or remdesizumab are used to inhibit tumors through different mechanisms and prolong survival. In recent years, with the in-depth exploration of the pathogenesis and progression mechanism of HCC, as well as the rapid progress within the domain of tumor immunotherapy, the treatment prospects for advanced HCC patients have shown a positive transformation. This transformation is reflected in the fact that more and more patients are gradually gaining significant and considerable therapeutic advantages from advanced immunotherapy regimens, bringing unprecedented improvements to their treatment outcomes. In order to enable activated T cells to attack tumor cells, immune checkpoint inhibitors interfere with the inhibitory. AIM To evaluate the effects of nivolumab in combination with cabozantinib on patient tumor markers and immune function, as well as the therapeutic efficacy of this combination in treating advanced HCC, a study was conducted. METHODS In all, 100 patients with advanced HCC who were brought to our hospital between July 2022 and July 2023 and who did not match the requirements for surgical resection had their clinical data thoroughly analyzed retrospectively in this study. Among them, half of the patients (50 cases) only received oral cabozantinib as a single treatment regimen (set as the control group), while the other half of the patients (50 cases) received intravenous infusion of nivolumab in addition to oral cabozantinib (set as the observation group). The objective of the probe is to examine the variations in disease control rate (DCR) and objective response rate (ORR) between two groups; At the same time, changes in the levels of T lymphocyte subsets (CD3+, CD4+, CD8+) and tumor markers, including AFP, GP-73, and AFP-L3, were evaluated; In addition, changes in liver and kidney function indicators and adverse reactions during treatment were also monitored. For patients with advanced HCC, this research also calculated and analyzed the progression free survival of two patient groups throughout the course of a 12-month follow-up to assess the effectiveness and safety of this therapeutic approach. RESULTS Upon comparing baseline information for both groups of subjects before treatment, it was found that no statistically significant alterations had occurred (P > 0.05). After the therapeutic intervention, the observation group and control group's ORR and DCR differed statistically significantly (P < 0.05). The observation group's scores significantly improved. Subsequent examination revealed that the observation group's T lymphocyte subset levels had significantly changed, mostly exhibiting an increase in CD3+, CD4+, and CD4+/CD8+ levels while CD8+ levels had comparatively dropped. There was a significant difference (P < 0.05) between these changes and those in the control group. The observation group also showed positive improvements in tumor markers; AFP, GP-73, and AFP-L3 levels were considerably lower in the group under observation than in the control group, with statistically significant differences (P < 0.05). When liver function was assessed, total bilirubin and alanine aminotransferase were found to be considerably lower in the observation group than in the control group (P < 0.05). The incidence of adverse responses was not statistically significant (P > 0.05), indicating that the incidence of adverse responses did not differ significantly between the two groups. CONCLUSION When treating advanced HCC, nivolumab and cabozantinib together have the ability to increase T lymphocyte numbers, reduce tumor marker levels, effectively prolong survival time, and have better efficacy than simple control treatment, with good safety.
Collapse
Affiliation(s)
- Lu-Wen Liang
- Infection and Liver Disease Center, The Second Affiliated Hospital of Chongqing Medical University, Chongqing 400000, China
| | - Rong-Hong Luo
- Department of Infectious Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Zhi-Li Huang
- Department of Infectious Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| | - Li-Na Tang
- Department of Infectious Diseases, The Third Affiliated Hospital of Chongqing Medical University, Chongqing 401120, China
| |
Collapse
|
45
|
Chen F, Cai Y, Zhou J. Relationship between retinol metabolism and hepatocellular carcinoma: a comprehensive analysis of Mendelian randomization, prognostic characteristic and experiment. Discov Oncol 2025; 16:513. [PMID: 40210831 PMCID: PMC11985832 DOI: 10.1007/s12672-025-02295-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Accepted: 04/02/2025] [Indexed: 04/12/2025] Open
Abstract
PURPOSE Retinol metabolism is intricately linked to the occurrence and progression of hepatocellular carcinoma (HCC); however, the precise pathogenic relationship between them remains elusive. The aim of this study was to elucidate the characteristics of retinol metabolism in HCC through Mendelian randomization, prognostic model and experimental validation. METHODS We used transcriptomic data related to HCC in TCGA and GEO databases for a variety of machine learning, including differential gene expression analysis, functional enrichment analysis, protein-protein network interaction, ceRNA regulatory network, and single-cell sequencing analysis. Mendelian randomization analysis was used to elucidate the causal analysis of retinol metabolism and the occurrence of HCC. Consensus cluster analysis was performed based on 11 retinol metabolism-related genes, and the prognostic model was constructed by Lasso regression and Cox regression analysis. The expression level of RDH16 gene was detected in cell lines and clinical samples, and finally the function of RDH16 gene and its regulatory relationship with miR- 665 were verified by in vitro cell experiments. RESULTS Differentially expressed genes were mainly concentrated in the retinol metabolic pathway. Mendelian randomization analysis showed that decreased retinol metabolic activity was causally associated with the occurrence of HCC. RDH16 gene was significantly lower expressed in HCC, and inhibition of RDH16 gene expression could promote the proliferation, migration and invasion of HCC cells and inhibit cell apoptosis. miR- 665 is an upstream regulator of RDH16 gene, which can inhibit the expression and function of RDH16. CONCLUSION The decrease of retinol metabolic activity can promote the occurrence and development of HCC. Targeting retinol metabolic pathway may be a new direction for the treatment of HCC.
Collapse
Affiliation(s)
- Fuqing Chen
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, 201 - 209 Hubin South Road, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Yifan Cai
- Department of Gastrointestinal Surgery, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, 361004, Fujian Province, People's Republic of China
| | - Jianyin Zhou
- Department of Hepatobiliary Surgery, Xiamen Key Laboratory of Translational Medical of Digestive System Tumor, Fujian Provincial Key Laboratory of Chronic Liver Disease and Hepatocellular Carcinoma, Zhongshan Hospital of Xiamen University, School of Medicine, Xiamen University, 201 - 209 Hubin South Road, Xiamen, 361004, Fujian Province, People's Republic of China.
| |
Collapse
|
46
|
Zhu Z, Lian X, Hu J, Wang Z, Zhong Y, Zhao Y, Lu L, Pan Y, Zhou M, Xu J. DPHC from Alpinia officinarum Hance specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate hepatocellular carcinoma. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119598. [PMID: 40058474 DOI: 10.1016/j.jep.2025.119598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/18/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Alpinia officinarum Hance (A. officinarum), a perennial herb used in the treatment of digestive system cancers, holds significant value for the Li people of Hainan as a traditional Chinese medicine. (R)-5-hydroxy-1,7-diphenyl-3-heptanone (DPHC), a diarylheptanoid component is derived from A. officinarum. Diarylheptanoids have demonstrated anti-proliferative effects on breast cancer cells, neuroblastoma cells, and other tumor cells. However, the pharmacological activity of DPHC in improving hepatocellular carcinoma (HCC) remains undefined. AIM OF THE STUDY To elucidate the anti-HCC effects of DPHC derived from A. officinarum and explore its underlying mechanistic pathways both in vivo and in vitro. MATERIAL AND METHODS The effects of DPHC on HCC cell lines were evaluated in vitro using cell counting kit-8, EdU cell proliferation assays, a wound healing assay, a three-dimensional tumor spheroid model, and flow cytometry. The ability of DPHC to ameliorate HCC was assessed in vivo via a nude mouse subcutaneous xenograft tumor model, serum biochemical marker detection, and hematoxylin-eosin staining. The molecular mechanism of DPHC in HCC was elucidated through a combination of transcriptome sequencing, cell transfection, immunohistochemistry assay, immunofluorescence staining, quantitative reverse transcription-PCR, and western blot analysis. RESULTS DPHC induced significant G0/G1 phase arrest and apoptosis in HepG2 and HCCLM3 cells while also markedly inhibiting tumor growth in nude mice. Mechanically, DPHC directly interacted with centromere-associated protein U (CENPU) to suppress its expression. The reduced expression of CENPU results in decreased interaction with the transcription factor E2F6, thereby affecting the transcriptional activity of the transcription factor E2F1. This subsequently inhibits the expression of downstream cell cycle factors (CCND1, CDK4, and CDK1) and increases apoptosis factors (Caspase 3 and Caspase 9). CONCLUSIONS DPHC from A. officinarum specifically modulates the function of CENPU in the cell cycle and apoptosis to ameliorate HCC. Our study revealed the anti-HCC effect and underlying mechanism of DPHC, offering new insights and potential targets for HCC treatment.
Collapse
Affiliation(s)
- Zhe Zhu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Xiuxia Lian
- Engineering Research Center of Tropical Medicine Innovation and Transformation of Ministry of Education & International Joint Research Center of Human-machine Intelligent Collaborative for Tumor Precision Diagnosis and Treatment of Hainan Province & Hainan Provincial Key Laboratory of Research and Development on Tropical Herbs, School of Pharmacy, Hainan Medical University, Haikou, Hainan, 571199, China
| | - Jicheng Hu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Zhe Wang
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yinghong Zhong
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yuan Zhao
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Lu Lu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China
| | - Yipeng Pan
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Mingyan Zhou
- Institute of Clinical Medicine, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China.
| | - Jian Xu
- Hepatobiliary and Liver Transplantation Department of Hainan Digestive Disease Center, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, 570311, China; Key Laboratory of Emergency and Trauma of Ministry of Education, The First Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China.
| |
Collapse
|
47
|
Xu Y, Xu L, Chen Q, Zou C, Huang J, Zhang L. Crosstalk between exosomes and tumor-associated macrophages in hepatocellular carcinoma: implication for cancer progression and therapy. Front Immunol 2025; 16:1512480. [PMID: 40264760 PMCID: PMC12011854 DOI: 10.3389/fimmu.2025.1512480] [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: 10/16/2024] [Accepted: 02/26/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the most prevalent type of primary liver cancer, represents a significant cause of cancer-related mortality. While our understanding of its pathogenesis is comparatively comprehensive, the influence of the tumor microenvironment (TME) on its progression warrants additional investigation. Tumor-associated macrophages (TAMs) have significant impacts on cancer cell proliferation, migration, invasion, and immune response, facilitating a complex interaction within the TME. Exosomes, which measure between 30 and 150 nanometers in size, are categorized into small extracellular vesicles, secreted by a wide range of eukaryotic cells. They can transfer biological molecules including proteins, non-coding RNAs, and lipids, which mediates the intercellular communication within the TME. Emerging evidence has revealed that exosomes regulate macrophage polarization, thus impacting cancer progression and immune responses within the TME of HCC. Moreover, TAM-derived exosomes also play crucial roles in malignant transformation, which hold immense potential for cancer therapy. In this review, we elaborate on the crosstalk between exosomes and TAMs within TME during HCC development. Moreover, we delve into the feasible treatment approaches for exosomes in cancer therapy and emphasize the limitations and challenges for the translation of exosomes derived from TAMs into clinical courses for cancer therapy, which may provide new perspectives on further ameliorations of therapeutic regimes based on exosomes to advance their clinical applications.
Collapse
Affiliation(s)
- Ying Xu
- Department of Anesthesiology Operating Room, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Linyue Xu
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Qiuyan Chen
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Can Zou
- School of Sports Medicine and Health, Chengdu Sport University, Chengdu, China
| | - Ju Huang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| | - Limei Zhang
- Department of Respiratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan, China
| |
Collapse
|
48
|
Li H, Zhang L, Shu B, Wang X, Yang S. Endoplasmic reticulum stress-related signatures: a game-changer in prognostic stratification for hepatocellular carcinoma. Eur J Gastroenterol Hepatol 2025; 37:454-465. [PMID: 39589828 DOI: 10.1097/meg.0000000000002894] [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] [Indexed: 11/28/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) has limited therapeutic options and a poor prognosis. The endoplasmic reticulum (ER) plays a crucial role in tumor progression and response to stress, making it a promising target for HCC stratification. This study aimed to develop a risk stratification model using ER stress-related signatures. METHODS We utilized transcriptome data from The Cancer Genome Atlas and Gene Expression Omnibus, which encompass whole-genome expression profiles and clinical annotations. Machine learning algorithms, including the least absolute shrinkage and selection operator, random forest, and support vector machine recursive feature elimination, were applied to the key genes associated with HCC prognosis. A prognostic system was developed using univariate Cox hazard analysis and least absolute shrinkage and selection operator Cox regression, followed by validation using Kaplan-Meier analysis and receiver operating characteristic curves. Tumor immune dysfunction and exclusion tools were used to predict immunotherapy responsiveness. RESULTS Two distinct clusters associated with ER stress were identified in HCC, each exhibiting unique clinical and biological features. Using a computational approach, a prognostic risk model, namely the ER stress-related signature, was formulated, demonstrating enhanced predictive accuracy compared with that of existing prognostic models. An effective clinical nomogram was established by integrating the risk model with clinicopathological factors. Patients with lower risk scores exhibited improved responsiveness to various chemotherapeutic, targeted, and immunotherapeutic agents. CONCLUSION The critical role of ER stress in HCC is highlighted. The ER stress-related signature developed in this study is a powerful tool to assess the risk and clinical treatment of HCC.
Collapse
Affiliation(s)
| | - Lei Zhang
- Department of Ultrasound, Beijing Tsinghua Changgung Hospital, School of Clinical Medicine, Tsinghua University
| | - Bin Shu
- Hepatopancereatobiliary Center
| | | | - Shizhong Yang
- Hepatopancreatobiliary Center, Beijing Tsinghua Changgung Hospital, Institute for Precision Medicine, Key Laboratory of Digital Intelligence Hepatology (Ministry of Education), Tsinghua University
- Research Unit of Precision Hepatobiliary Surgery Paradigm, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
49
|
Li G, Li W, Zhou Y, Tan X, Huang Q, Liang J, Zhou Z. Label-free determination of glypican-3 using PtPd@H-rGO nanocomposites decorated light-addressable potentiometric sensor. Bioelectrochemistry 2025; 162:108855. [PMID: 39608318 DOI: 10.1016/j.bioelechem.2024.108855] [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: 10/14/2024] [Revised: 11/11/2024] [Accepted: 11/18/2024] [Indexed: 11/30/2024]
Abstract
Glypican-3 (GPC3) is exclusively overexpressed in most Hepatocellular carcinoma (HCC) tissue but not in normal liver tissue, making it a promising biomarker for the precise detection of HCC. In this paper, a label-free light-addressable potentiometric sensor (LAPS) decorated by platinumpalladium-hemin-reduced graphene oxide nanocomposites (PtPd@H-rGO NCs) was constructed for determination of GPC3. The GPC3 aptamer (GPC3Apt) and PtPd@H-rGO NCs were modified on the surface of silicon-based LAPS chip to build sensitive unit of LAPS system. A readout photocurrent elicited from a modulated light source, registers the localized surface potential change. When a bias voltage is provided to the LAPS system, the GPC3-GPC3Apt complexes formed by the specific reaction between GPC3 and GPC3Apt at the sensing interface can cause the sensitive membrane surface potential to change, resulting in the photocurrent-voltage (I-V) curves generate a corresponding offset response. Therefore GPC3 concentration can be determined by monitoring the potential shifts (△V). Under optimal conditions, the potential shift is linearly related to the concentration of GPC3 in the range of 0.001-3.00 μg/mL with the limit of detection (LOD) of 0.0001 μg/mL. The LAPS has a good analytical performance with good specificity, reproducibility and stability, and can be used for the detection of GPC3 in actual serum samples, which provides a broad application prospect for the combined application of LAPS and aptamers in biooassay.
Collapse
Affiliation(s)
- Guiyin Li
- College of Chemistry, Guangdong University of Petrochemical Technology, Guandu Road, Maoming, Guangdong 525000, PR China
| | - Wenzhan Li
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, Guangxi 541004, PR China
| | - Yu Zhou
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, Guangxi 541004, PR China
| | - Xiaohong Tan
- College of Chemistry, Guangdong University of Petrochemical Technology, Guandu Road, Maoming, Guangdong 525000, PR China
| | - Qing Huang
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, Guangxi 541004, PR China
| | - Jintao Liang
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, Guangxi 541004, PR China.
| | - Zhide Zhou
- School of Life and Environmental Sciences, Guilin University of Electronic Technology, Guilin, Guangxi 541004, PR China.
| |
Collapse
|
50
|
Peng B, Zhang J, Xiang Y. CDCA8 and its multifaceted role in tumorigenesis. Biomed Pharmacother 2025; 185:117951. [PMID: 40056827 DOI: 10.1016/j.biopha.2025.117951] [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: 12/20/2024] [Revised: 02/11/2025] [Accepted: 02/27/2025] [Indexed: 03/10/2025] Open
Abstract
Human cell division cycle-associated 8 (CDCA8), also known as Borealin or Dasra-B, is a critical component of the vertebrate Chromosomal Passenger Complex (CPC). It plays a pivotal role in the segregation of sister chromatids during the cell cycle and is essential for preventing the formation of aneuploid chromosomes and ensuring successful cytokinesis. Numerous studies have demonstrated that CDCA8 is upregulated in various cancers, including hepatocellular carcinoma, lung cancer, glioma, and bladder cancer. By influencing key biological processes such as cell proliferation, apoptosis, invasion, and metastasis, CDCA8 drives tumor progression. Clinically, the expression of CDCA8 correlates closely with tumor staging and histological grading, providing significant prognostic value for patients with diverse cancers. Moreover, CDCA8 modulates tumor biology through multiple signaling pathways, including P53, PI3K/Akt, E2F/Rb, and mTOR. In summary, CDCA8 represents a promising diagnostic and therapeutic target across multiple cancer types and serves as a potential prognostic biomarker. This review highlights the critical roles of CDCA8 in cancer diagnosis, treatment, and prognosis, as well as the underlying mechanisms through which it exerts its effects. These insights offer a theoretical basis and research direction for early cancer diagnosis, targeted therapy, and prognostic evaluation.
Collapse
Affiliation(s)
- Boming Peng
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China
| | - Jianquan Zhang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China.
| | - Yang Xiang
- Department of Hepatobiliary Surgery, Haikou Affiliated Hospital of Central South University Xiangya School of Medicine, Haikou 570208, China; Haikou Key Laboratory of Clinical Research and Transformation of Digestive Diseases, Haikou 570208, China
| |
Collapse
|