51
|
Ananthakrishnan AI, Mahin A, Prasad TSK, Abhinand CS. Transcriptome Profiling and Viral-Human Interactome Insights Into HBV-Driven Oncogenic Alterations in Hepatocellular Carcinoma. Microbiol Immunol 2025. [PMID: 40243270 DOI: 10.1111/1348-0421.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is the primary form of liver cancer that poses a significant global health concern due to its increasing incidence rates and diverse etiology. Chronic infection induced by hepatitis B virus (HBV) is a prominent etiological factor influencing the development of HCC. Although recent advances in multi-omics approaches have facilitated extensive exploration of HCC molecular characteristics, translating the characteristics of subtypes into clinical applications has been challenging due to parameters like limited sample size and complex classifiers for early detection. In the present study, we performed transcriptomics profiling of HBV-infected HCC patient tissue data to gather comprehensive insights into the intricate molecular mechanisms underlying HBV-associated HCC, specifically, viral protein interactions that influence the expression of oncogenes. The 1059 differentially expressed genes (DEGs) identified across two GEO data sets revealed upregulation of cell cycle and mitosis-related genes, alongside downregulation of genes involved in fatty acid degradation and cytochrome P450 activity. CDK1 and CDC20 which are part of the top cluster and hub gene from interactome analysis were identified as potential markers for HBV-positive HCC through gene expression pattern and overall survival analysis. Additionally, 19 DEGs showing significance in HCC development were identified as interacting partners with HBV proteins. Among them, the interaction of HBsAg with ALB and SHBG and their downregulation correlates to the lower testosterone levels identified in HBV and HCC patients. Together, the study enhances the understanding of the heterogeneity and molecular pathogenesis of HBV-positive HCC.
Collapse
Affiliation(s)
- Anilkumar I Ananthakrishnan
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Althaf Mahin
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
- Department of Virus Genomics, Bioinformatics, and Statistics, Institute of Advanced Virology, Thiruvananthapuram, India
| |
Collapse
|
52
|
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
|
53
|
Yao RR, Zhang QB, Ge MX, Li L, Li J, Zhou XL, Wang ZX, Liang XH. The safety and efficacy of atezolizumab for recurrent primary liver cancer after liver transplantation. Discov Oncol 2025; 16:553. [PMID: 40244542 PMCID: PMC12006625 DOI: 10.1007/s12672-025-02299-4] [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: 11/20/2024] [Accepted: 04/02/2025] [Indexed: 04/18/2025] Open
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) have been proved to have certain therapeutic effects for primary liver cancer. However, the efficacy and safety of their applications in liver transplantation (LT) recipients with recurrent tumor remained unclear and even controversial. METHODS A retrospective study was conducted to evaluate the safety and efficacy of atezolizumab in LT recipients with recurrent PLC from August 1, 2019, to July 1, 2022. The primary endpoint was the incidence of allograft rejection, while secondary endpoints included overall response rate (ORR), progression-free survival (PFS), and overall survival (OS). Additionally, risk factors associated with ICI-related rejection were analyzed. RESULTS A total of eight LT recipients with recurrent PLC were included in the study, comprising six cases of hepatocellular carcinoma (HCC) and two cases of intrahepatic cholangiocarcinoma (ICC). The median number of atezolizumab treatment cycles was 3 (range, 1-10). Graft rejection occurred in 25% of patients (2/8). The median overall survival (mOS) from the initiation of atezolizumab was 6.2 months (range, 0.7-12.5 months), with a mortality rate of 75% (6/8). The ORR (complete response [CR] + partial response [PR]) was 28.5% (2/7), and the disease control rate (DCR; CR + PR + stable disease [SD]) was 42.9% (3/7). Time from LT to recurrence (P = 0.008) and Interval time from LT to atezolizumab (P = 0.005) were associated with liver rejection. CONCLUSIONS Atezolizumab demonstrated a certain degree of efficacy as a salvage treatment for patients with recurrent HCC and ICC after LT. However, given the potential risk of allograft rejection, careful evaluation of safety is essential before initiating ICI therapy. Moreover, close monitoring and timely intervention are necessary during treatment to mitigate the risk of rejection. Future studies should further explore the optimal timing and strategies for ICI administration in this patient population.
Collapse
Grants
- (2020QD010) Scientific Research Foundation of Huashan Hospital, Fudan University
- (2020QD010) Scientific Research Foundation of Huashan Hospital, Fudan University
- (2020QD010) Scientific Research Foundation of Huashan Hospital, Fudan University
- (2020QD010) Scientific Research Foundation of Huashan Hospital, Fudan University
- (2020QD010) Scientific Research Foundation of Huashan Hospital, Fudan University
- 82071797, 82173093, 82241225 Natural Science Foundation of China
- 82071797, 82173093, 82241225 Natural Science Foundation of China
- 82071797, 82173093, 82241225 Natural Science Foundation of China
- (SHDC2020CR2021B) the Shanghai Hospital Development Center
- (SHDC2020CR2021B) the Shanghai Hospital Development Center
- (SHDC2020CR2021B) the Shanghai Hospital Development Center
Collapse
Affiliation(s)
- Rong-Rong Yao
- Department of Oncology, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Quan-Bao Zhang
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
- Institute of Organ Transplantation, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Meng-Xi Ge
- Department of Oncology, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Li Li
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
- Institute of Organ Transplantation, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Jing Li
- Department of Oncology, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Xin-Li Zhou
- Department of Oncology, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China
| | - Zheng-Xin Wang
- Liver Transplantation Center, Department of General Surgery, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China.
- Institute of Organ Transplantation, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China.
| | - Xiao-Hua Liang
- Department of Oncology, Huashan Hospital, Fudan University, 12 Urumqi Road(M), Shanghai, 200040, China.
| |
Collapse
|
54
|
Zhang H, Wu B, Zhang L, Peng Z. Relationship between MTHFR 677C > T polymorphism and serum PIVKA-II levels in hepatocellular carcinoma. Arch Physiol Biochem 2025:1-8. [PMID: 40243201 DOI: 10.1080/13813455.2025.2493107] [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: 12/09/2024] [Revised: 03/04/2025] [Accepted: 04/09/2025] [Indexed: 04/18/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major public health problem with increasing incidence and mortality worldwide. The methylenetetrahydrofolate reductase (MTHFR) 677 C > T polymorphism is associated with the development and progression of various tumours, while protein induced by vitamin K absence II (PIVKA-II) is an important tumour marker for the diagnosis of HCC. This study aims to investigate the relationship between the MTHFR 677 C > T polymorphism and serum PIVKA-II levels in HCC patients, providing new insights for early diagnosis, risk assessment, and prognosis evaluation of HCC. METHODS This study included 120 HCC patients and 100 healthy controls. MTHFR 677 C > T genotyping was performed using fluorescent quantitative PCR, and serum PIVKA-II levels were measured. Bioinformatics analysis was used to explore the expression of the MTHFR gene in HCC and its relationship with prognosis. RESULTS MTHFR 677 C > T TT carriers had an increased risk of HCC (OR = 2.393; 95% CI 1.055-5.429; p = 0.037); the risk of HCC for T gene carriers was 58.3% higher than that for C gene carriers in the allele model (OR = 1.583; 95% CI 1.059-2.364; p = 0.025). The difference in serum PIVKA-II concentration was statistically significant between the controls, stage I-II patients, and stage III-IV patients (p < 0.05), and the difference in serum PIVKA-II concentration was statistically significant between patients with the TT genotype and patients with the CC and CT genotypes (all p values less than 0.05). UALCAN database analysis showed that MTHFR gene expression levels were increased in patients with HCC, and the high expression of the MTHFR gene was negatively correlated with patient survival rates. CONCLUSIONS There is an association between the MTHFR 677 C > T TT genotype and serum PIVKA-II levels in HCC. This could help identify high-risk individuals and assess disease severity, providing a potential genetic biomarker for the diagnosis of HCC.
Collapse
Affiliation(s)
- Hongyu Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Clinical Faculty of Guangxi University of Chinese Medicine, Liuzhou, Guangxi, China
| | - Baixiu Wu
- Department of Nuclear Medicine, the Fourth Affiliated Hospital of Guangxi Medical University, Liuzhou Worker's Hospital, Liuzhou, Guangxi, China
| | - Liang Zhang
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Clinical Faculty of Guangxi University of Chinese Medicine, Liuzhou, Guangxi, China
| | - Zheng Peng
- Department of Clinical Laboratory, The Third Affiliated Hospital of Guangxi University of Chinese Medicine, Liuzhou Traditional Chinese Medical Hospital, The Third Clinical Faculty of Guangxi University of Chinese Medicine, Liuzhou, Guangxi, China
| |
Collapse
|
55
|
Liu X, Liu Z, Kobayashi T, Lei PJ, Shi Y, Yuan D, Wang J, Li M, Matsui A, Mafra K, Huang P, Kuang M, Bod L, Duda DG. Inhibiting B-cell-mediated Immunosuppression to Enhance the Immunotherapy Efficacy in Hepatocellular Carcinoma. RESEARCH SQUARE 2025:rs.3.rs-6355345. [PMID: 40321752 PMCID: PMC12047993 DOI: 10.21203/rs.3.rs-6355345/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/08/2025]
Abstract
Background Immunotherapy is efficacious in hepatocellular carcinoma (HCC), but the benefits are limited to a minority of patients. Most HCC patients show resistance to immune checkpoint blockade (ICB). Agonists of the stimulator of interferon genes (STING), potent immune stimulators, showed limited effectiveness. Using preclinical models, we studied the mechanisms of resistance to ICB and STING agonism. Methods Murine HCA-1 and RIL-175 HCCs were orthotopically grown in mice with underlying liver fibrosis, to mimic the presentation of human HCC. Established tumors were treated with a STING agonist (BMS-986301) or anti-PD1 ICB, and mice were followed to evaluate safety and efficacy, as well as the mechanisms of treatment resistance by RNA sequencing, flow cytometry, and immunofluorescence, B-cell depletion and T-cell immunoglobulin and mucin domain 1 (TIM-1) ICB. Results Unbiased analyses of transcriptomic data from murine HCC tissues from ICB-treated mice showed an increased abundance of intratumoral CD8+ T cells and B cells. STING agonism alone showed efficacy in the ICB-responsive RIL-175 HCC model but more limited efficacy in the ICB-resistant HCA-1 model. STING agonism increased circulating IL-10 and intratumoral infiltration by B-cells, including TIM-1+ B cells, and promoted the formation of tertiary lymphoid structure (TLS)-like structures, especially in the peritumoral areas. Strikingly, adding B cell depletion to ICB or STING agonism treatment significantly increased survival. Interestingly, unlike ICB, STING agonism also had a pronounced anti-metastatic activity. In addition, the combination of STING agonism and TIM-1 blockade augmented B cell differentiation and antigen presentation in vitro and improved the anti-tumor effects in murine HCC in vivo. This approach decreased the number of TIM-1+ B cells in the tumor and shifted B cells to higher expression of CD86 and MHC class II, enhancing the antigen presentation capability and further boosting the antitumor efficacy of CD8+ cytotoxic T cells. Conclusion Our findings demonstrate that B cells are associated with ICB- and STING-mediated therapy resistance, and that depleting B-cells or targeting TIM-1 enhances both innate and acquired therapeutic efficacy in HCC.
Collapse
Affiliation(s)
- Xin Liu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zelong Liu
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Tatsuya Kobayashi
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Pin-Ji Lei
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Yue Shi
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dandan Yuan
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, China
| | - Jianguo Wang
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Affiliated Hangzhou First People’s Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Min Li
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- The First Affiliated Hospital of Anhui Medical University, Hefei, China
| | - Aya Matsui
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
- Kanazawa University Institute of Medical, Pharmaceutical and Health Sciences Faculty of Medicine, Kanazawa, Japan
| | - Kassiana Mafra
- Department of Medicine, Krantz Family Center for Cancer Research, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Peigen Huang
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| | - Ming Kuang
- The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Lloyd Bod
- Department of Medicine, Krantz Family Center for Cancer Research, Center for Immunology and Inflammatory Diseases, Massachusetts General Hospital, Boston, MA, USA; Harvard Medical School, Boston, MA, USA; Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Dan G. Duda
- Edwin. L. Steele Laboratories for Tumor Biology, Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, USA
| |
Collapse
|
56
|
Jiang J, Liu F, Cui D, Xu C, Chi J, Yan T, Guo F. Novel molecular mechanisms of immune evasion in hepatocellular carcinoma: NSUN2-mediated increase of SOAT2 RNA methylation. Cancer Commun (Lond) 2025. [PMID: 40227950 DOI: 10.1002/cac2.70023] [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: 03/01/2024] [Revised: 03/13/2025] [Accepted: 03/17/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a deadly malignancy known for its ability to evade immune surveillance. NOP2/Sun RNA methyltransferase family member 2 (NSUN2), an RNA methyltransferase involved in carcinogenesis, has been associated with immune evasion and energy metabolism reprogramming. This study aimed to examine the molecular mechanisms underlying the involvement of NSUN2 in immune evasion and metabolic reprogramming of HCC. METHODS Single-cell transcriptomic sequencing was applied to examine cellular composition changes, particularly immune cell dynamics, in HCC and adjacent normal tissues. Bulk RNA-seq and proteomics identified key genes and proteins. Methylation sequencing and methylated RNA immunoprecipitation (MeRIP) were carried out to characterize the role of NSUN2 in 5-methylcytosine (m5C) modification of sterol O-acyltransferase 2 (SOAT2). Clinical samples from 30 HCC patients were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blotting. Gene expression was manipulated using CRISPR/Cas9 and lentiviral vectors. In vitro co-culture models and metabolomics were used to study HCC cell-T cell interactions, energy metabolism, and immune evasion. Tumor growth in an orthotopic mouse model was monitored by bioluminescence imaging, with subsequent measurements of tumor weight, volume, and immunohistochemical staining. RESULTS Single-cell transcriptomic analysis identified a marked increase in malignant cells in HCC tissues. Cell communication analysis indicated that tumor cells might promote cancer progression by evading immune clearance. Multi-omics analyses identified NSUN2 as a key regulator in HCC development. MeRIP confirmed that NSUN2 facilitated the m5C modification of SOAT2. Analysis of human HCC tissue samples demonstrated pronounced upregulation of NSUN2 and SOAT2, along with elevated m5C levels in HCC tissues. In vitro experiments uncovered that NSUN2 augmented the reprogramming of energy metabolism and repressed the activity and cytotoxicity of CD8+ T cells, contributing to immune evasion. In vivo studies further substantiated the role of NSUN2 in fostering immune evasion and tumor formation of HCC by modulating the m5C modification of SOAT2. CONCLUSIONS The findings highlight the critical role of NSUN2 in driving HCC progression through the regulation of m5C modification on SOAT2. These findings present potential molecular markers for HCC diagnosis and therapeutic targets for its treatment.
Collapse
Affiliation(s)
- Jinhua Jiang
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| | - Feng Liu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Dan Cui
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Caixia Xu
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Jiachang Chi
- Department of Interventional Oncology, Renji Hospital, School of Medicine, Shanghai Jiaotong University, Shanghai, P. R. China
| | - Tinghua Yan
- The First Clinical Medical College of Jinan University, Guangzhou, Guangdong, P. R. China
| | - Fang Guo
- Key Laboratory of Systems Biomedicine, Ministry of Education, Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, P. R. China
| |
Collapse
|
57
|
Li Z, Wang Y, Yang K, Li C, Zhang M. Reduction in Liver Cancer Risk by Quercetin via Modulation of Urate Levels: Insights from Drug-Target Mendelian Randomization. Genes (Basel) 2025; 16:449. [PMID: 40282409 PMCID: PMC12027248 DOI: 10.3390/genes16040449] [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: 02/18/2025] [Revised: 04/02/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025] Open
Abstract
BACKGROUND Quercetin, a dietary flavonoid and a widely used supplement, has hepatoprotective properties. Given its urate-lowering effects and epidemiological evidence linking elevated serum urate levels to liver cancer risk, we tested whether quercetin reduces liver cancer risk via modulation of urate levels by bioinformatics methods. METHODS We employed drug-target Mendelian randomization using genome-wide association study summary statistics from public databases (e.g., MRC-IEU) to assess genetic associations, and integrated these findings with GEO datasets (such as GSE138709 and GSE179443) and immune infiltration analyses using tools like xCell, TIMER. RESULTS Our analyses identified ABCG2-mediated urate elevation as a causal risk factor for hepatocellular carcinoma (OR = 1.001, p < 0.01), cholangiocarcinoma (OR = 3.424, p < 0.01), and liver fibrosis (OR = 2.528, p < 0.01). Single-cell transcriptomics revealed elevated ABCG2 expression in cholangiocarcinoma endothelial cells, while immune infiltration analysis showed significant associations between ABCG2 expression and both endothelial cell and macrophage infiltration. Survival analysis further indicated that ABCG2 was not associated with poor prognosis in cholangiocarcinoma or hepatocellular carcinoma. CONCLUSIONS Considering quercetin's multifaceted interactions with BCRP/ABCG2, our findings support its potential use as a preventive dietary supplement for hepatic diseases rather than as an adjunctive therapy for established liver cancer.
Collapse
Affiliation(s)
- Zhengwen Li
- School of Pharmacy, Chengdu University, 2025 Chengluo Avenue, Chengdu 610106, China;
| | - Yue Wang
- Cell Biology-Inspired Tissue Engineering, Institute for Technology-Inspired, Regenerative Medicine, Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Kaichuan Yang
- School of Pharmacy, Chengdu University, 2025 Chengluo Avenue, Chengdu 610106, China;
| | - Chujie Li
- Precision Medicine, Faculty of Health, Medicine and Life Sciences, Maastricht University, 6200 MD Maastricht, The Netherlands;
| | - Ming Zhang
- College of Food Science and Engineering, Hainan University, 58 Renmin Road, Haikou 570228, China
| |
Collapse
|
58
|
Ghosh D, Guin A, Kumar A, Das A, Paul S. Comprehensive insights of etiological drivers of hepatocellular carcinoma: Fostering targeted nano delivery to anti-cancer regimes. Biochim Biophys Acta Rev Cancer 2025; 1880:189318. [PMID: 40222420 DOI: 10.1016/j.bbcan.2025.189318] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 04/05/2025] [Accepted: 04/06/2025] [Indexed: 04/15/2025]
Abstract
Hepatocellular carcinoma (HCC) stands as one of the most prevalent and deadliest malignancies on a global scale. Its complex pathogenesis arises from multifactorial etiologies, including viral infections, metabolic syndromes, and environmental carcinogens, all of which drive genetic and molecular aberrations in hepatocytes. This intricate condition is associated with multiple causative factors, resulting in the abnormal activation of various cellular and molecular pathways. Given that HCC frequently manifests within the context of a compromised or cirrhotic liver, coupled with the tendency of late-stage diagnoses, the overall prognosis tends to be unfavorable. Systemic therapy, especially conventional cytotoxic drugs, generally proves ineffective. Despite advancements in therapeutic interventions, conventional treatments such as chemotherapy often exhibit limited efficacy and substantial systemic toxicity. In this context, nanomedicine, particularly lipid-based nanoparticles (LNPs), has emerged as a promising strategy for enhancing drug delivery specificity and reducing adverse effects. This review provides a comprehensive overview of the molecular and metabolic underpinnings of HCC. Furthermore, we explored the role of lipid-based nano-formulations including liposomes, solid lipid nanoparticles, and nanostructured lipid carriers in targeted drug delivery for HCC. We have highlighted recent advances in LNP-based delivery approaches, FDA-approved drugs, and surface modification strategies to improve liver-specific delivery and therapeutic efficacy. It will provide a comprehensive summary of various treatment strategies, recent clinical advances, receptor-targeting strategies and the role of lipid composition in cellular uptake. The review concludes with a critical assessment of existing challenges and future prospects in nanomedicines-driven HCC therapy.
Collapse
Affiliation(s)
- Dipanjan Ghosh
- Department of Biotechnology and Dr. B.C. Guha Centre for Genetic Engineering and Biotechnology, University of Calcutta, Kolkata 700019, West Bengal, India
| | - Aharna Guin
- Department of Chemistry, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Aryan Kumar
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India
| | - Amlan Das
- Department of Microbiology & Department of Biochemistry, Royal School of Biosciences, The Assam Royal Global University, Guwahati 781035, Assam, India.
| | - Santanu Paul
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, Tirupati 517619, Andhra Pradesh, India.
| |
Collapse
|
59
|
Lan X, Zhang H, Chen ZY, Wang J, Zhang SC, Li Q, Ke JY, Wei W, Huang R, Tang X, Chen SP, Huang TT, Zhou YW. Suppressor of cytokine signaling 2 modulates regulatory T cell activity to suppress liver hepatocellular carcinoma growth and metastasis. World J Gastroenterol 2025; 31:100566. [PMID: 40248063 PMCID: PMC12001165 DOI: 10.3748/wjg.v31.i13.100566] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/27/2024] [Accepted: 03/11/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND Liver hepatocellular carcinoma (LIHC) is a highly aggressive cancer with poor prognosis due to its complex tumor microenvironment (TME) and immune evasion. Regulatory T cells (Tregs) play a critical role in tumor progression. Suppressor of cytokine signaling 2 (SOCS2), a key immune regulator, may modulate Treg activity and impact LIHC growth and metastasis. AIM To explore how the SOCS2 affects Treg activity in LIHC and its impact on tumor growth and metastasis. METHODS LIHC transcriptome data from The Cancer Genome Atlas database were analyzed using Gene Set Enrichment Analysis, Estimation of Stromal and Immune Cells in Malignant Tumors Using Expression Data, and Cell-Type Identification by Estimating Relative Subsets of RNA Transcripts to evaluate immune pathways and Treg infiltration. Key prognostic genes were identified using Weighted Gene Co-expression Network Analysis and machine learning. In vitro, co-culture experiments, migration assays, apoptosis detection, and enzyme-linked immunosorbent assay were conducted. In vivo, tumor growth, metastasis, and apoptosis were assessed using subcutaneous and lung metastasis mouse models with hematoxylin and eosin staining, Terminal Deoxynucleotidyl Transferase dUTP Nick End Labeling, and immunohistochemistry analyses. RESULTS SOCS2 overexpression inhibited Treg cell activity, reducing LIHC cell migration and invasion while increasing apoptosis. In vivo, SOCS2 suppressed tumor growth and metastasis, confirming its therapeutic potential. CONCLUSION SOCS2 modulates CD4+ T function in the TME, contributing to LIHC progression. Targeting SOCS2 presents a potential therapeutic strategy for treating LIHC.
Collapse
Affiliation(s)
- Xi Lan
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Heng Zhang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Ze-Yan Chen
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Jing Wang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Shi-Chang Zhang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Qing Li
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Juan-Yu Ke
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Wei Wei
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Rong Huang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Xi Tang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Si-Ping Chen
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Ting-Ting Huang
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| | - Yi-Wen Zhou
- Clinical Laboratory Center, Shenzhen Hospital, Southern Medical University, Shenzhen 518101, Guangdong Province, China
| |
Collapse
|
60
|
Alibrahem W, Helu NK, Oláh C, Prokisch J. Potential of Carbon Nanodots (CNDs) in Cancer Treatment. NANOMATERIALS (BASEL, SWITZERLAND) 2025; 15:560. [PMID: 40214605 PMCID: PMC11990490 DOI: 10.3390/nano15070560] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Revised: 04/02/2025] [Accepted: 04/04/2025] [Indexed: 04/14/2025]
Abstract
Carbon Nanodots (CNDs) are characterized by their nanoscale size (<10 nm), biocompatibility, stability, fluorescence, and photoluminescence, making them a promising candidate for cancer therapy. The difference in the methods of synthesis of CNDs, whether top-down or bottom-up, affects the formation, visual, and surface characteristics of CNDs, which are crucial for their biomedical and pharmaceutical applications. The urgent need for innovative therapeutic strategies from CNDs is due to the limitations and barriers posed by conventional therapies including drug resistance and cytotoxicity. Nano-loaded chemotherapy treatments are highly effective and can enhance the solubility and targeted delivery of chemotherapeutic agents, generate reactive oxygen species (ROS) to induce cancer cell cytotoxicity, and regulate intracellular signaling pathways. Their ability to be designed for cellular uptake and exact intracellular localization further improves their therapeutic potential. In addition to working on drug delivery, CNDs are highlighted for their dual functionality in imaging and therapy, which allows real-time observing of treatment efficacy. Despite the development of these treatments and the promising results for the future, challenges still exist in cancer treatment.
Collapse
Affiliation(s)
- Walaa Alibrahem
- Doctoral School of Health Sciences, University of Debrecen, Egyetem tér 1, 4028 Debrecen, Hungary;
| | - Nihad Kharrat Helu
- Doctoral School of Health Sciences, University of Debrecen, Egyetem tér 1, 4028 Debrecen, Hungary;
| | - Csaba Oláh
- Mathias Institute, University of Tokaj, Eötvös Str. 7, 3950 Sárospatak, Hungary;
- Neurosurgery Department, Borsod County University Teaching Hospital, Szentpéteri kapu 72-76, 3526 Miskolc, Hungary
| | - József Prokisch
- Faculty of Agricultural and Food Sciences and Environmental Management, Institute of Animal Science, Biotechnology and Nature Conservation, University of Debrecen, Böszörményi Street 138, 4032 Debrecen, Hungary;
| |
Collapse
|
61
|
Custode BM, Annunziata F, Dos Santos Matos F, Schiano V, Maffia V, Lillo M, Colonna R, De Cegli R, Ballabio A, Pastore N. Folliculin depletion results in liver cell damage and cholangiocarcinoma through MiT/TFE activation. Cell Death Differ 2025:10.1038/s41418-025-01486-8. [PMID: 40189703 DOI: 10.1038/s41418-025-01486-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 02/11/2025] [Accepted: 03/17/2025] [Indexed: 05/11/2025] Open
Abstract
Mutations in the tumor suppressor gene Folliculin (FLCN) are responsible for Birt-Hogg-Dube' (BHD) syndrome, a rare inherited condition that predisposes affected individuals to skin tumors, pulmonary cysts, and kidney tumors. FLCN regulates key cellular pathways, including TFEB, TFE3, and mTORC1, which are critical for maintaining cell homeostasis. Loss of FLCN leads to both hyperactivation of mTORC1 and constitutive activation of TFEB and TFE3, contributing to tumorigenesis. While previous studies showed that Flcn liver-specific conditional knockout (FlcnLiKO) mice are protected from developing liver fibrosis and damage upon high-fat diet exposure, the potential role of FLCN loss in liver carcinogenesis remained unexplored. Here, we demonstrate that hepatic loss of FLCN in mice results in cancer associated with inflammation and fibrosis with features of cholangiocarcinoma (CCA). This phenotype emerges in mice over 90-week-old, with a male predominance. Moreover, FlcnLiKO mice are more prone to develop diethylnitrosamine (DEN)- or 3,5-diethoxycarbonyl-1,4-dihydrocollidine (DDC)- induced liver tumors with heterogenous histological features. Notably, depletion of TFE3, but not TFEB, in the liver of FlcnLiKO mice fully rescues the cancer phenotype and normalized mTORC1 signaling, highlighting TFE3 as the primary driver of liver cancer and mTORC1 hyperactivity in the absence of FLCN.
Collapse
Affiliation(s)
| | | | | | - Valentina Schiano
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Veronica Maffia
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Milena Lillo
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rita Colonna
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Rossella De Cegli
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
| | - Andrea Ballabio
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy
- Jan and Dan Duncan Neurological Research Institute, Texas Children Hospital, Houston, TX, USA
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy
| | - Nunzia Pastore
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli (NA), Italy.
- Department of Translational Medicine, Medical Genetics, Federico II University, Naples, Italy.
| |
Collapse
|
62
|
He H, Wu Y, Jia Z, Zhang Y, Pan Y, Zhang Y, Su K, Cui Y, Sun Y, Li D, Lv H, Yi J, Wang Y, Kou C, Sun X, Jiang J. A stratified precision screening strategy for enhancing hepatitis B- and C-associated liver cancer detection: a prospective study. Sci Rep 2025; 15:11396. [PMID: 40181083 PMCID: PMC11968812 DOI: 10.1038/s41598-025-95795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
This study explores new screening strategies to enhance liver cancer screening effectiveness. In a prospective study, 2605 participants underwent baseline, 6-months self-reported, and 1-year follow-up screenings using abdominal ultrasonography, AFP, AFP-L3%, and DCP. The results demonstrated the GALADUS protocol exhibited superior performance with higher AUC (0.935 vs. 0.836; DeLong P < 0.001), sensitivity (91.0% vs. 70.8%; P < 0.001), detection (3.1% vs. 2.4%; P < 0.001), and early diagnosis rates (64.2% vs. 58.7%) compared to the AFP/US protocol. Notably, among individuals with an aMAP score ≥ 60, GALADUS had significantly outperformed AFP/US in AUC (0.923 vs. 0.826; DeLong P < 0.001), sensitivity (94.2% vs. 69.6%; P < 0.001), detection (9.7% vs. 7.2%; P < 0.001), and early diagnosis rates (63.1% vs. 54.2%). However, for those with an aMAP score < 60, GALADUS offered no significant advantages. Introducing the "aMAP triage" protocol, combining GALADUS for aMAP ≥ 60 and AFP/US for aMAP < 60, further enhanced AUC to 0.925 (DeLong P < 0.001), improved sensitivity by 19.1% (89.9% vs. 70.8%; P < 0.001), and increased detection (3.1% vs. 2.4%; P < 0.001) and early diagnosis rates (65.0% vs. 58.7%), being cost-effective compared to GALADUS. In conclusion, this study highlights the potential of a stratified precision screening strategy in identifying high-risk individuals, applying tailored early detection protocols to improve liver cancer screening efficacy.
Collapse
Affiliation(s)
- Hua He
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yanhua Wu
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhifang Jia
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yangyu Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuchen Pan
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuzheng Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Kaisheng Su
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yingnan Cui
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuanlin Sun
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongming Li
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Haiyong Lv
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Jiaxin Yi
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuehui Wang
- Department of Geriatrics, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Changgui Kou
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaofeng Sun
- Department of Cadre's Wards Ultrasound Diagnostics, Ultrasound Diagnostic Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Jing Jiang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China.
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
63
|
Feng LH, Wei L, Hu B, Liang H, Li Q, Yin Q, Su T, Huang L, Liang H, Jiang J, Su M. Evaluation of serum ESPL1 as a biomarker for early diagnosis of HBV-related hepatocellular carcinoma. Front Oncol 2025; 15:1574317. [PMID: 40248196 PMCID: PMC12003130 DOI: 10.3389/fonc.2025.1574317] [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: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 04/19/2025] Open
Abstract
Objective To evaluate the reliability of serum human phosphorylated exospindle polar-like proteinase 1 (ESPL1) as a serum biomarker for early diagnosis of hepatitis B virus (HBV)-related hepatocellular carcinoma (HBV-HCC). Methods This retrospective study was conducted on 266 patients with chronic hepatitis B (CHB), liver cirrhosis (LC), and HBV-related HCC. Data on demographics and clinical information were collected, and ESPL1 levels were measured using enzyme linked immunosorbent assay. Levels of ESPL1, alpha-fetoprotein (AFP), and protein induced by vitamin K absence -II (PIVKA-II) were compared at different disease stages, and spearman correlation analysis was used to assess their relationship with clinical markers. The diagnostic accuracy of ESPL1, AFP, and PIVKA-II for early HBV- HCC was assessed using ROC curve analysis. Results The study comprised 121 patients diagnosed with CHB, 98 patients with LC, and 47 patients with HBV-HCC. Serum ESPL1 levels show an increasing trend across groups with chronic HBV infection, CHB, LC, and HBV-HCC, with levels at 224.6 ng/L, 285.8 ng/L, and 440.4 ng/L (in pairwise comparison, P<0.05). Serum AFP and PIVKA-II levels displayed no significant statistical differences between the CHB and LC groups. Spearman correlation analysis revealed that levels of ESPL1, PIVKA-II, and AFP are not influenced by clinical characteristics and show no correlation with each other. ROC curve analysis indicated that the optimal diagnostic threshold for ESPL1 in HBV-HCC is 345.7 ng/L, with AUC values for ESPL1, PIVKA-II, and AFP being 0.797 (95% CI: [0.708-0.886]), 0.788 (95% CI: [0.718-0.858]), and 0.572 (95% CI: [0.523-0.624]). In AFP and PIVKA-II negative patients, the AUC values for ESPL1 diagnosis of HBV-HCC were 0.79 and 0.83. Conclusion ESPL1 is a potential biomarker for tracking chronic HBV infection and predicting the development of HBV-HCC. Monitoring ESPL1 levels in serum could help with early detection and personalized screening HBV-HCC for individuals with chronic HBV infection.
Collapse
Affiliation(s)
- Lu-Huai Feng
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Lu Wei
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Bobin Hu
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hengkai Liang
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qingmei Li
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Qianbing Yin
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Tumei Su
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Long Huang
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Hongqian Liang
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Jianning Jiang
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High Incidence Tumors (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| | - Minghua Su
- Department of Infectious Diseases, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Key Laboratory of Early Prevention and Treatment of Regional High Incidence Tumors (Guangxi Medical University), Ministry of Education, Nanning, Guangxi, China
| |
Collapse
|
64
|
Rafati I, Yazdani L, Barat M, Karam E, Fohlen A, Nguyen BN, Castel H, Tang A, Cloutier G. Ultrasound shear wave viscoelastography to characterize liver nodules. Phys Med Biol 2025; 70:075022. [PMID: 40127537 DOI: 10.1088/1361-6560/adc4b8] [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: 12/06/2024] [Accepted: 03/24/2025] [Indexed: 03/26/2025]
Abstract
Purpose. To investigate the diagnostic performance of ultrasound (US)-based shear wave speed (SWS), shear wave attenuation (SWA), and combination of them as shear wave viscoelastography (SWVE) methods in patients undergoing US to characterize focal liver nodules.Materials and methods. In this prospective cross-sectional study, 70 patients with 72 nodules were enrolled. Investigational US and clinical magnetic resonance imaging (MRI) examinations were performed in all participants. The composite reference standard included MRI or histopathology to differentiate benign and malignant nodules. A linear discriminant analysis (LDA) was used to assess the combination of SWVE methods. Analyzes included Mann-WhitneyUtest, receiver operating characteristic analysis, and computation of sensitivity and specificity at the point that maximized the Youden index.Results. Mean SWS was significantly higher in malignant than benign nodules (2.49 ± 0.76 m s-1vs. 1.72 ± 0.70,p< 0.001), whereas SWA was lower (0.56 ± 0.30 vs. 1.10 ± 0.43 Np/m/Hz,p< 0.001). To differentiate between malignant and benign nodules, SWS with a threshold of 2.43 m s-1achieved a sensitivity of 0.54 (95% confidence interval (CI): 0.38-0.69) and a specificity of 0.88 (CI: 0.74-0.95). SWA with a threshold of 0.81 Np/m/Hz yielded a sensitivity of 0.81 (CI: 0.66-0.90) and a specificity of 0.74 (CI: 0.58-0.86). Combining these SWVE methods using a LDA resulted in a sensitivity of 0.81 (CI: 0.66-0.91) and a specificity of 0.86 (CI: 0.71-0.94).Conclusion. Malignant nodules had higher SWS and lower SWA than benign ones. The combination of SWS and SWA in a LDA classification algorithm increased the diagnostic performance.
Collapse
Affiliation(s)
- Iman Rafati
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada
- Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - Ladan Yazdani
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada
- Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - Maxime Barat
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Elige Karam
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Audrey Fohlen
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Bich N Nguyen
- Department of Pathology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Hélène Castel
- Departments of Hepatology and Liver Transplantation, University of Montreal Hospital, Montréal, Québec, Canada
| | - An Tang
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
- Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada
- Laboratory of Clinical Image Processing, University of Montreal Hospital Research Center, Montréal, Québec, Canada
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada
- Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
- Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada
| |
Collapse
|
65
|
Li J, Xian L, Wang X, Liu Y, Li J. The role of TACE in the era of immune-targeted therapy for hepatocellular carcinoma: a meta-analysis based on PSM. Front Immunol 2025; 16:1573834. [PMID: 40242754 PMCID: PMC12000099 DOI: 10.3389/fimmu.2025.1573834] [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: 02/10/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major global health challenge, with over 50% of patients ineligible for curative treatments at diagnosis. The combination of molecular targeted therapies and immunotherapy has shown promise in improving outcomes for advanced HCC. Objective This meta-analysis aims to assess the efficacy of combining transarterial chemoembolisation (TACE) with immune-targeted therapies in patients with unresectable HCC. Methods A systematic review and meta-analysis conforming to PRISMA guidelines were conducted by searching PubMed, Embase, Web of Science, and the Cochrane Library for studies published up to January 5, 2025. Due to the limited clinical evidence, our study exclusively included retrospective studies based on propensity score matching (PSM) analysis that compared the efficacy of TACE in combination with immune-targeted therapy to immune-targeted therapy alone. Key outcomes assessed included objective response rate (ORR), disease control rate (DCR), one-year overall survival (1-OS), one-year progression-free survival (1-PFS), median overall survival (mOS), and median progression-free survival (mPFS). Results A total of 9 PSM studies involving 2119 patients were included. The meta-analysis revealed that TACE significantly improved ORR, DCR, 1-OS, and 1-PFS, in addition to extending mOS and mPFS. Conclusion The findings suggest that the inclusion of TACE in treatment regimens for unresectable HCC notably enhances tumour control and patient survival. This study provides moderate to high-quality evidence supporting the integration of TACE in advanced HCC management, particularly for those patients not meeting standard TACE criteria. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD 42025631817.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinsen Wang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingnan Liu
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiarui Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
66
|
Li YF, Wang HW, Peng HY, Zhang ZY, Yao ZJ, Meng Y, Yang DY. Association between allopurinol and hepatocellular carcinoma: analysis of genetic risk and patient survival. Discov Oncol 2025; 16:454. [PMID: 40175812 PMCID: PMC11965048 DOI: 10.1007/s12672-025-02176-0] [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: 01/04/2025] [Accepted: 03/18/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Despite the widespread clinical use of allopurinol for managing hepatocellular carcinoma (HCC) and gout, its potential hepatotoxicity and its effect on the risk of HCC remain unclear. This study aimed to comprehensively assess the potential correlations between allopurinol exposure and HCC risk. METHODS We utilized genome-wide association study data from the IEU OpenGWAS project as instrumental variables (IVs) for Mendelian randomization (MR) analysis to investigate the causal relationship between allopurinol and HCC. Subsequently, we investigated the potential mediating factors (gout, liver fat, and percentage of liver fat, etc.) between allopurinol use and HCC. Furthermore, we analyzed assessed survival outcomes using the Kaplan-Meier method to compare patient subgroups by differential Xanthine dehydrogenase (XDH) expression. RESULTS MR analysis established a causal link between allopurinol use and increased HCC risk (OR: 1.013, 95% CI 1.004-1.023, p = 0.006). Causal relationships were also observed between gout (OR: 1.011, p = 0.008) and HCC. Mediation analysis indicated that gout mediated 61.6% of the effect of allopurinol on HCC. Survival analysis showed that higher expression of XDH was associated with improved survival of HCC patients (HR = 0.62, 95% CI 0.441-0.884, p = 0.008), indicating a 38% decrease in mortality risk compared to the lower expression group. CONCLUSIONS This study demonstrated a causal relationship between allopurinol use and an increased risk of HCC based on genetic evidence. Allopurinol should be used with caution in patients with or at risk for HCC.
Collapse
Affiliation(s)
- Yu-Fu Li
- Shenzhen University Health Science Center, Shenzhen, Guangdong, 518071, China
- Division of Gastroenterology & Hepatology, The University of Hongkong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Hui-Wei Wang
- Department of Dermatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Huan-Yan Peng
- Division of Gastroenterology & Hepatology, The University of Hongkong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Zhen-Ying Zhang
- Department of Dermatology, The University of Hong Kong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Zhi-Jia Yao
- Division of Gastroenterology & Hepatology, The University of Hongkong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Yuan Meng
- Division of Gastroenterology & Hepatology, The University of Hongkong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China
| | - Dong-Ye Yang
- Division of Gastroenterology & Hepatology, The University of Hongkong-Shenzhen Hospital, Shenzhen, Guangdong, 518053, China.
| |
Collapse
|
67
|
Yu Z, Cao W, Du C, Liu J, Peng L, Wei F. Developing a novel predictive model for identifying risk factors associated with being lost to follow-up among high-risk patients for recurrence following radical resection of hepatocellular carcinoma: the first report. BMC Cancer 2025; 25:597. [PMID: 40175947 PMCID: PMC11967030 DOI: 10.1186/s12885-025-14030-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 03/27/2025] [Indexed: 04/04/2025] Open
Abstract
BACKGROUND Follow-up is essential especially for patients who are at a high risk of recurrence after radical resection of hepatocellular carcinoma (HCC). The aim of this study was to develop a predictive model aimed at identifying the risk factors associated with being lost to follow-up (LTFU) in high-risk patients for recurrence following radical resection of HCC. METHODS The retrospective study was conducted at our institution between October 2018 to May 2023. The patients who underwent radical liver resection for HCC and had high-risk factors for recurrence were categorized into an LTFU group and a control group. Multivariate logistic regression analysis was utilized to determine risk factors and construct a nomogram predictive model. RESULTS A total of 352 patients were included and subsequently classified into two distinct groups: the LTFU group (n = 123, 34.94%) and the control group (n = 229, 65.06%). Logistic regression analysis was then conducted to explore the potential associations between various factors and the occurrence of LTFU. The findings identified several independent risk factors for LTFU, including smoking (odds ratio, OR = 1.823, 95% confidence interval, CI 1.086-3.060, p = 0.023); residing more than 200 km away from the hospital (OR = 1.857, 95% CI 1.105-3.121, p = 0.019); having an unstable profession (OR = 1.918, 95% CI 1.112-3.311, p = 0.019); and lacking medical insurance (OR = 5.921, 95% CI 1.747-20.071, p = 0.004); the presence of liver cirrhosis (OR = 2.161, 95% CI 1.153-4.048, p = 0.016); an operation time less than 240 min (OR = 2.138, 95% CI 1.240-3.688, p = 0.006); and the absence of postoperative adjuvant therapy (OR = 2.641, 95% CI 1.504-4.637, p = 0.001). Based on these seven significant factors, a main effects model was established, designated as the Wei-LTFU model, which achieved an area under the curve value of 0.744 (95% CI 0.691-0.798) in predicting the likelihood of LTFU. CONCLUSION A main effects model, namely the Wei-LTFU model, incorporating the seven significant factors was formulated to predict the likelihood of LTFU occurrence, ultimately aiming to assist healthcare workers in developing effective strategies to improve follow-up outcomes for patients.
Collapse
Affiliation(s)
- Zichen Yu
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Wenli Cao
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
- Department of Public Health, Hangzhou Medical College, Hangzhou, 310059, Zhejiang Province, China
| | - Chengfei Du
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Jie Liu
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Liping Peng
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China
| | - Fangqiang Wei
- Department of General Surgery, Cancer Center, Division of Hepatobiliary and Pancreatic Surgery, Affiliated People's Hospital, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, 310014, Zhejiang Province, China.
| |
Collapse
|
68
|
Kim JS, Kim HS, Tak KY, Han JW, Nam H, Sung PS, Lee SW, Kwon JH, Bae SH, Choi JY, Yoon SK, Jang JW. Male preference for TERT alterations and HBV integration in young-age HBV-related HCC: implications for sex disparity. Clin Mol Hepatol 2025; 31:509-524. [PMID: 39743888 PMCID: PMC12016656 DOI: 10.3350/cmh.2024.0545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 12/24/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025] Open
Abstract
BACKGROUND/AIMS Hepatocellular carcinoma (HCC) exhibits significant sex disparities in incidence, yet its molecular mechanisms remain unclear. We explored the role of telomerase reverse transcriptase (TERT) genetic alterations and hepatitis B virus (HBV) integration, both known major contributors to HCC, in sex-specific risk for HBV-related HCC. METHODS We examined 310 HBV-related HCC tissues to investigate sex-specific TERT promoter (TERT-pro) mutations and HBV integration profiles, stratified by sex and age, and validated with single-cell RNA sequencing (scRNA-seq) data. RESULTS Tumors predominantly exhibited TERT-pro mutations (26.0% vs. 0%) and HBV-TERT integration (37.0% vs. 3.0%) compared to non-tumorous tissues. While TERT-pro mutations increased with age in both sexes, younger males (≤60 years) showed marked predominance compared to younger females. Males had significantly more HBV integrations at younger ages, while females initially had fewer integrations that gradually increased with age. Younger males' integrations showed significantly greater enrichment in the TERT locus compared to younger females, alongside a preference for promoters, PreS/S regions, and CpG islands. Overall, TERT genetic alterations were significantly sex-differential in younger individuals (75.3% in males vs. 23.1% in females) but not in older individuals (76.9% vs. 83.3%, respectively). These alterations were associated with increased TERT expression. The skewed TERT abnormalities in younger males were further corroborated by independent scRNA-seq data. CONCLUSION Our findings highlight the critical role of TERT alterations and HBV integration patterns in the male predominance of HCC incidence among younger HBV carriers, offering insights for future exploration to optimize sex-specific patient care and HCC surveillance strategies.
Collapse
Affiliation(s)
- Jin Seoub Kim
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Korea
- Department of Medical Informatics, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Hye Seon Kim
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Biomedicine & Health Sciences, Graduate School, The Catholic University of Korea, Seoul, Korea
- Department of Medical Informatics, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Kwon Yong Tak
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ji Won Han
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Heechul Nam
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Pil Soo Sung
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Sung Won Lee
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jung Hyun Kwon
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Si Hyun Bae
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jong Young Choi
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Seung Kew Yoon
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Jeong Won Jang
- The Catholic University Liver Research Center, The Catholic University of Korea, Seoul, Korea
- Department of Medical Informatics, Cancer Research Institute, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Korea
| |
Collapse
|
69
|
Li Y, Zheng X, Li J, Dai Q, Wang CD, Chen M. LKAN: LLM-Based Knowledge-Aware Attention Network for Clinical Staging of Liver Cancer. IEEE J Biomed Health Inform 2025; 29:3007-3020. [PMID: 39392729 DOI: 10.1109/jbhi.2024.3478809] [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: 10/13/2024]
Abstract
Clinical staging of liver cancer (CSoLC), an important indicator for evaluating primary liver cancer (PLC), is key in the diagnosis, treatment, and rehabilitation of liver cancer. In China, the current CSoLC adopts the China liver cancer (CNLC) staging, which is usually evaluated by clinicians based on radiology reports. Therefore, inferring clinical information from unstructured radiology reports can provide auxiliary decision support for clinicians. The key to solving the challenging task is to guide the model to pay attention to the staging-related words or sentences, and the following issues may occur: 1) Imbalanced categories: Early- and mid-stage liver cancer symptoms are subtle, resulting in more data in the end-stage. 2) Domain sensitivity of liver cancer data: The liver cancer dataset contains substantial domain knowledge, leading to out-of-vocabulary issues and reduced classification accuracy. 3) Free-text and lengthy report: Radiology reports sparsely describe various lesions using domain-specific terms, making it hard to mine staging-related information. To address these, this article proposes a large language model (LLM)-based Knowledge-aware Attention Network (LKAN) for CSoLC. First, for maintaining semantic consistency, LLM and a rule-based algorithm are integrated to generate more diverse and reasonable data. Second, an unlabeled radiology corpus is pre-trained to introduce domain knowledge for subsequent representation learning. Third, attention is improved by incorporating both global and local features to guide the model's focus on staging-relevant information. Compared with the baseline models, LKAN has achieved the best results with 90.3% Accuracy, 90.0% Macro_F1 score, and 90.0% Macro_Recall.
Collapse
|
70
|
Su BB, Zhu CJ, Cao J, Peng R, Tu DY, Jiang GQ, Jin SJ, Wang Q, Zhang C, Bai DS. Enhanced prediction of 5-year postoperative recurrence in hepatocellular carcinoma by incorporating LASSO regression and random forest models. Surg Endosc 2025; 39:2540-2550. [PMID: 40032663 DOI: 10.1007/s00464-025-11631-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Accepted: 02/18/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Tumor recurrence post-operation of hepatocellular carcinoma (HCC) impacts patient prognosis. Identifying and predicting 5-year HCC recurrence following surgery remains a substantial challenge. METHODS We included 338 patients diagnosed with HCC who underwent surgery from January 2013 to December 2018. Traditional logistic regression, random forest (RF), and LASSO regression methods were used to develop a predictive model for 5-year recurrence. The findings were presented visually using nomogram. The accuracy and sensitivity of the predictive model were evaluated by receiver operating curves (ROC) and decision curve analysis (DCA). RESULTS Of the 338 patients, 172 (50.9%) experienced 5 years recurrence, with a gender distribution of 79.7% males. Univariate and multivariate logistic regression analysis identified that three independent predictors of 5-year HCC recurrence (all P < 0.001). The area under the curve (AUC) value of the model (Model-1) constructed was 0.678. Then we combined LASSO regression and RF construct a predictive model including six factors: age, transarterial chemoembolization (TACE), microvascular invasion (MVI), alcohol, size, and number. The AUC of the model (Model-2) constructed was 0.733. DeLong's test results showed that Model-2 had significantly better prediction ability compared with Model-1 (P = 0.004). DCA also demonstrated that Model-2 had better predictive accuracy (P < 0.05). Then we constructed a nomogram, and Kaplan-Meier analysis showed that patients in the low-risk group had significantly better prognosis than the high (P < 0.001). CONCLUSION The predictive accuracy of our model, incorporating factors, such as age, alcohol, size, number, MVI, and TACE, significantly enhances clinical practice management by accurately forecasting 5 years HCC recurrence.
Collapse
Affiliation(s)
- Bing-Bing Su
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Chao-Jie Zhu
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Hepatobiliary Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
| | - Jun Cao
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Hepatobiliary Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
| | - Rui Peng
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Dao-Yuan Tu
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Guo-Qing Jiang
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Sheng-Jie Jin
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Qian Wang
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
| | - Chi Zhang
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China
- Department of Hepatobiliary Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China
| | - Dou-Sheng Bai
- Department of Hepatobiliary Surgery, Northern Jiangsu People'S Hospital Affiliated to Yangzhou University, Yangzhou, China.
- Department of Hepatobiliary Surgery, The Yangzhou Clinical Medical College of Xuzhou Medical University, Yangzhou, China.
- General Surgery Institute of Northern Jiangsu People'S Hospital, Yangzhou, China.
| |
Collapse
|
71
|
Nam H, Sung PS, Lee SW, Song DS, Kwon JH, Jang JW, Kim CW, Bae SH. Incorporating ALBI Grade with Geriatric Nutritional Risk Index Enhances Hepatocellular Carcinoma Risk Stratification. Liver Cancer 2025; 14:193-210. [PMID: 40255876 PMCID: PMC12005708 DOI: 10.1159/000541647] [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: 07/11/2024] [Accepted: 09/22/2024] [Indexed: 01/03/2025] Open
Abstract
Introduction Despite its prognostic impact, nutritional status has not yet been integrated into the assessment of hepatocellular carcinoma (HCC). This study investigated the association between geriatric nutritional risk index (GNRI) and overall survival (OS) in patients with HCC using a nationwide registry. Methods Data from the Korea Central Cancer Registry between 2008 and 2019 were analyzed. We explored the integration of the GNRI with the albumin-bilirubin (ALBI) grade for prognostic stratification. Restricted cubic spline regression was used to assess the association between GNRI and survival, stratified by ALBI grade. Results Among the 16,416 treatment-naïve HCC patients, the ALBI grades were distributed as follows: grade 1, 7,409; grade 2, 7,445; and grade 3, 1,562. Patients were categorized according to Barcelona Clinic Liver Cancer (BCLC) stages: 5,132 stage 0/A, 2,608 stage B, 5,289 stage C, and 968 stage D. The median OS for all patients was 3.1 years (95% CI: 3.0-3.2) and significantly differed with the inclusion of ALBI grade and GNRI (p < 0.001). The effect of combining ALBI grade and GNRI was further evaluated for each BCLC stage. This risk stratification showed a significant correlation with OS for each BCLC stage (all p < 0.001), except for stage D (p = 0.082). Multivariate analysis revealed that a combination of favorable ALBI grade and high GNRI score was independently associated with decreased mortality risk. Conclusion The GNRI was significantly correlated with OS across ALBI grades and BCLC stages. Integrating the GNRI into the ALBI grade may enhance risk stratification for patients with HCC.
Collapse
Affiliation(s)
- Heechul Nam
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Pil Soo Sung
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Sung Won Lee
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Do Seon Song
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Jung Hyun Kwon
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Jeong Won Jang
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Chang Wook Kim
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| | - Si Hyun Bae
- Department of Internal Medicine, College of Medicine, The Catholic University of Korea, Seoul, Republic of Korea
- The Catholic University Liver Research Center, Seoul, Republic of Korea
| |
Collapse
|
72
|
Sanghvi G, Roopashree R, Kashyap A, Sabarivani A, Ray S, Bhakuni PN. KIFC1 in cancer: Understanding its expression, regulation, and therapeutic potential. Exp Cell Res 2025; 447:114510. [PMID: 40058447 DOI: 10.1016/j.yexcr.2025.114510] [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: 01/10/2025] [Revised: 03/06/2025] [Accepted: 03/07/2025] [Indexed: 03/25/2025]
Abstract
Kinesins are a family of motor proteins essential for intracellular transport and cellular dynamics, with kinesin family member C1 (KIFC1) emerging as a key regulator of cancer progression. Recent studies highlight KIFC1's crucial role in mitotic spindle assembly, chromosome segregation, and cell migration-processes frequently dysregulated in cancer. Its involvement in promoting malignant cell proliferation and metastasis underscores its significance in tumor biology. In various cancer types, aberrant KIFC1 expression correlates with poor prognosis and aggressive phenotypes, suggesting its potential as a biomarker for disease severity. Mechanistically, KIFC1 influences signaling pathways linked to cell cycle regulation and programmed cell death, reinforcing its role in oncogenesis. Given its pivotal function in cancer cell dynamics, KIFC1 represents a promising therapeutic target. Strategies aimed at modulating its activity, including small molecules or RNA interference, could disrupt cancer cell viability and proliferation. The current review article highlights KIFC1's importance in cancer biology, advocating for further investigation into its mechanisms and the development of KIFC1-targeted therapies to enhance treatment efficacy and improve patient outcomes across various malignancies.
Collapse
Affiliation(s)
- Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, 360003, Gujarat, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Aditya Kashyap
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, 140401, Punjab, India
| | - A Sabarivani
- Department of Biomedical, Sathyabama Institute of Science and Technology, Chennai, Tamil Nadu, India
| | - Subhashree Ray
- Department of Biochemistry, IMS and SUM Hospital, Siksha 'O' Anusandhan (Deemed to be University), Bhubaneswar, Odisha, 751003, India
| | - Pushpa Negi Bhakuni
- Department of Allied Science, Graphic Era Hill University, Bhimtal, Uttarakhand, 248002, India; Graphic Era Deemed to be University, Dehradun, Uttarakhand, India.
| |
Collapse
|
73
|
Zhong BY, Fan W, Guan JJ, Peng Z, Jia Z, Jin H, Jin ZC, Chen JJ, Zhu HD, Teng GJ. Combination locoregional and systemic therapies in hepatocellular carcinoma. Lancet Gastroenterol Hepatol 2025; 10:369-386. [PMID: 39993404 DOI: 10.1016/s2468-1253(24)00247-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2024] [Revised: 07/20/2024] [Accepted: 07/25/2024] [Indexed: 02/26/2025]
Abstract
Locoregional therapies play a fundamental role in the treatment of patients with early and intermediate and locally advanced hepatocellular carcinomas. With encouraging recent advances in immunotherapy-based systemic therapies, locoregional therapies are being both promoted and challenged by new systemic therapy options. Combined locoregional and systemic therapies might enhance treatment outcomes compared with either option alone. This Series paper summarises the existing data on locoregional and systemic therapies for hepatocellular carcinoma, and discusses evidence from studies investigating their combination with a focus on their synergistic efficacy and safety.
Collapse
Affiliation(s)
- Bin-Yan Zhong
- 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, Nanjing, China; Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, China
| | - Wenzhe Fan
- Department of Interventional Oncology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Justin J Guan
- Division of Interventional Radiology, Department of Radiology, Cleveland Clinic, Cleveland, OH, USA
| | - Zhenwei Peng
- Department of Radiation Oncology, Cancer Center, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China; Institute of Precision Medicine, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Zhongzhi Jia
- Department of Interventional and Vascular Surgery, The Affiliated Changzhou Second People's Hospital of Nanjing Medical University, Changzhou, China
| | - Haojie Jin
- Shanghai Cancer Institute, State Key Laboratory of Systems Medicine for Cancer, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Zhi-Cheng Jin
- 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, Nanjing, China
| | - Jian-Jian Chen
- 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, Nanjing, China
| | - Hai-Dong Zhu
- 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, Nanjing, 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, Nanjing, China.
| |
Collapse
|
74
|
Qu Z, Zeng J, Zeng L, Li X, Zhang F. Esculetin triggers ferroptosis via inhibition of the Nrf2-xCT/GPx4 axis in hepatocellular carcinoma. Chin J Nat Med 2025; 23:443-456. [PMID: 40274347 DOI: 10.1016/s1875-5364(25)60853-3] [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: 01/05/2024] [Revised: 04/27/2024] [Accepted: 05/04/2024] [Indexed: 04/26/2025]
Abstract
Esculetin, a natural dihydroxy coumarin derived from the Chinese herbal medicine Cortex Fraxini, has demonstrated significant pharmacological activities, including anticancer properties. Ferroptosis, an iron-dependent form of regulated cell death, has garnered considerable attention due to its lethal effect on tumor cells. However, the exact role of ferroptosis in esculetin-mediated anti-hepatocellular carcinoma (HCC) effects remains poorly understood. This study investigated the impact of esculetin on HCC cells both in vitro and in vivo. The findings indicate that esculetin effectively inhibited the growth of HCC cells. Importantly, esculetin promoted the accumulation of intracellular Fe2+, leading to an increase in ROS production through the Fenton reaction. This event subsequently induced lipid peroxidation (LPO) and triggered ferroptosis within the HCC cells. The occurrence of ferroptosis was confirmed by the elevation of malondialdehyde (MDA) levels, the depletion of glutathione peroxidase (GSH-Px) activity, and the disruption of mitochondrial morphology. Notably, the inhibitor of ferroptosis, ferrostatin-1 (Fer-1), attenuated the anti-tumor effect of esculetin in HCC cells. Furthermore, the findings revealed that esculetin inhibited the Nrf2-xCT/GPx4 axis signaling in HCC cells. Overexpression of Nrf2 upregulated the expression of downstream SLC7A11 and GPX4, consequently alleviating esculetin-induced ferroptosis. In conclusion, this study suggests that esculetin exerts an anti-HCC effect by inhibiting the activity of the Nrf2-xCT/GPx4 axis, thereby triggering ferroptosis in HCC cells. These findings may contribute to the potential clinical use of esculetin as a candidate for HCC treatment.
Collapse
Affiliation(s)
- Zhixin Qu
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Jing Zeng
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China
| | - Laifeng Zeng
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China
| | - Xianmei Li
- Fujian Key Laboratory of Integrative Medicine on Geriatrics, Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou 350122, China.
| | - Fenghua Zhang
- Key Laboratory of Gastrointestinal Cancer (Ministry of Education), School of Basic Medical Sciences, Fujian Medical University, Fuzhou 350122, China.
| |
Collapse
|
75
|
Lee CK, Yoo C, Hong JY, Park SJ, Kim JW, Tai DWM, Kim H, Korphaisarn K, Tanasanvimon S, Chen SC, Kim JW, Kim I, Kim M, Choo J, Oh SB, Chen CT, Bae WK, Kim H, Huh SJ, Yen CJ, Park S, Lee DK, Chan LL, Kang B, Kang M, Sundar R, Choi HJ, Chan SL, Chon HJ, Lee MA. Real-World Study of Systemic Treatment after First-Line Atezolizumab plus Bevacizumab for Hepatocellular Carcinoma in Asia-Pacific Countries. Liver Cancer 2025; 14:127-141. [PMID: 40255875 PMCID: PMC12005689 DOI: 10.1159/000540969] [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: 03/29/2024] [Accepted: 08/11/2024] [Indexed: 10/11/2024] Open
Abstract
INTRODUCTION Atezolizumab plus bevacizumab is a commonly used first-line regimen for advanced hepatocellular carcinoma (HCC) treatment owing to its superior outcomes compared to sorafenib. However, optimal subsequent treatment options for patients with HCC who progressed on first-line atezolizumab plus bevacizumab remain unclear. METHODS This multinational, multi-institutional, retrospective study included patients with HCC from 22 centers in five Asia-Pacific countries who were treated with first-line atezolizumab plus bevacizumab, which was discontinued for any reason. The endpoints included progression-free survival (PFS) and overall survival (OS) according to patient characteristics and second-line regimens. RESULTS Between June 2016 and May 2023, 1,141 patients were treated with first-line atezolizumab plus bevacizumab, of whom 629 (55.1%) received subsequent treatment. Sorafenib and lenvatinib were the most commonly administered second-line regimens (53.9% and 25.6%, respectively). Overall, the median PFS and OS were 2.9 and 8.0 months, respectively. Lenvatinib had longer PFS (4.0 vs. 2.3 months) and OS (8.0 vs. 6.3 months) than sorafenib. Patients treated with tyrosine kinase inhibitor (TKI) plus immune checkpoint inhibitor (ICI) (n = 50, 8.3%) showed PFS and OS of 5.4 and 12.6 months, respectively. Lower tumor burden and lenvatinib or TKI plus ICI use were associated with longer second-line PFS. Preserved liver function was associated with improved OS. CONCLUSIONS In patients with HCC who progressed on first-line atezolizumab plus bevacizumab, sorafenib and lenvatinib were the most commonly used second-line regimens in Asia-Pacific countries, with lenvatinib resulting in longer OS than sorafenib. The second-line TKI plus ICI combination exhibited promising efficacy, suggesting the potential role of continuing ICIs beyond disease progression.
Collapse
Affiliation(s)
- Choong-kun Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Changhoon Yoo
- Department of Oncology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jung Yong Hong
- Division of Hematology-Oncology, Department of Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Se Jun Park
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, South Korea
| | - Jin Won Kim
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - David Wai Meng Tai
- Division of Medical Oncology, National Cancer Center Singapore, Singapore, Singapore
| | - Hyeyeong Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan, South Korea
| | - Krittiya Korphaisarn
- Division of Medical Oncology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | | | - San-Chi Chen
- Division of Medical Oncology, Department of Oncology, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Ju Won Kim
- Division of Oncology/Hematology, Department of Internal Medicine, Korea University Anam Hospital, Seoul, South Korea
| | - Ilhwan Kim
- Division of Oncology, Department of Internal Medicine, Inje University College of Medicine, Haeundae Paik Hospital, Busan, South Korea
| | - Moonho Kim
- Department of Hematology and Oncology, University of Ulsan College of Medicine, Gangneung Asan Hospital, Gangneung, South Korea
| | - Joan Choo
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Sang-Bo Oh
- Division of Hematology-Oncology, Department of Internal Medicine, School of Medicine, Yangsan Pusan National University Hospital, Busan, South Korea
| | - Ching-Tso Chen
- National Taiwan University Hospital Hsin-Chu Branch, Hsinchu, Taiwan
| | - Woo Kyun Bae
- Division of Hematology-Oncology, Department of Internal Medicine, Chonnam National University Medical School and Hwasun Hospital, Gwangju, South Korea
| | - Hongsik Kim
- Division of Hematology-Oncology, Department of Internal Medicine, Chungbuk National University Hospital, Cheongju, South Korea
| | | | - Chia-Jui Yen
- Department of Oncology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sejung Park
- Sondang Institute for Cancer Research, Yonsei University College of Medicine, Seoul, South Korea
| | - Dong Ki Lee
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Landon Long Chan
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Beodeul Kang
- Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Minsu Kang
- Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, South Korea
| | - Raghav Sundar
- Department of Haematology-Oncology, National University Cancer Institute, Singapore, Singapore
| | - Hye Jin Choi
- Division of Medical Oncology, Department of Internal Medicine, Yonsei Cancer Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Stephen Lam Chan
- Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Hong Kong Cancer Institute, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, Hong Kong SAR
| | - Hong Jae Chon
- Division of Medical Oncology, Department of Internal Medicine, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | - Myung-Ah Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary’s Hospital, College of Medicine, Cancer Research Institute, The Catholic University of Korea, Seoul, South Korea
| |
Collapse
|
76
|
Emmamally M, Kotze U, Bernon M, Robertson B, Khan R, Sobnach S, Sonderup M, Spearman CW, Jonas E. The spectrum and outcome of metastatic hepatocellular carcinoma in a South African patient cohort. HPB (Oxford) 2025; 27:572-581. [PMID: 39875276 DOI: 10.1016/j.hpb.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 11/17/2024] [Accepted: 01/05/2025] [Indexed: 01/30/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a leading cause of cancer-related death globally, particularly in developing countries in Southeast Asia and sub-Saharan Africa (SSA), where chronic hepatitis B virus (HBV) dominates as a major aetiological factor. METHODS We conducted a retrospective cohort study to quantify the metastatic profile of HCC in a South African patient population managed at a tertiary centre. Demographic, clinical and treatment data were extracted from an institutional registry. Patients with and without metastases were compared to identify factors associated with an increased risk of developing metastases. RESULTS Of 676 patients, 194 (28.7 %) had metastases. Patients with metastases were younger (46.37 vs. 52.23 years; p < 0.00001) and more frequently had chronic HBV, HIV co-infection and schistosomiasis compared to non-metastatic patients. The most common metastatic sites were lungs, skeletal, and peritoneum. For non-treated patients, skeletal metastases had the poorest survival. CONCLUSION This study confirms a typical SSA disease profile of aggressive HCC in a young population. We reported on the metastatic profile of HCC and the impact of different metastases on survival. Future research should focus on defining the concept of oligometastatic disease in HCC to identify patients where intervention targeting metastatic disease may be of benefit.
Collapse
Affiliation(s)
- Muhammad Emmamally
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa
| | - Urda Kotze
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa
| | - Marc Bernon
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa
| | - Barbara Robertson
- Division of Radiation Oncology, Department of Radiation Medicine, Faculty of Health Sciences, University of Cape Town Faculty and Groote Schuur Hospital, Cape Town, South Africa
| | - Rufaida Khan
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa
| | - Sanju Sobnach
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa
| | - Mark Sonderup
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town Faculty and Groote Schuur Hospital, Cape Town, South Africa
| | - C Wendy Spearman
- Division of Hepatology, Department of Medicine, Faculty of Health Sciences, University of Cape Town Faculty and Groote Schuur Hospital, Cape Town, South Africa
| | - Eduard Jonas
- Surgical Gastroenterology Unit, Division of General Surgery, University of Cape Town Faculty of Health Sciences and Groote Schuur Hospital, Cape Town, South Africa.
| |
Collapse
|
77
|
Jayasekera CR, Rajasooriyar C, Richards C, Arora S, Borad MJ. Transforming Hepatocellular Carcinoma Treatment in Lower Income Countries. Mayo Clin Proc 2025; 100:605-608. [PMID: 40057864 DOI: 10.1016/j.mayocp.2024.12.011] [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: 05/25/2024] [Revised: 11/30/2024] [Accepted: 12/18/2024] [Indexed: 04/05/2025]
Affiliation(s)
- Channa R Jayasekera
- Liver Transplant Program, Mayo Clinic, Phoenix, AZ; Division of Gastroenterology and Hepatology, Mayo Clinic, Phoenix, AZ.
| | - Chrishanthi Rajasooriyar
- Teaching Hospital Jaffna, Jaffna, Sri Lanka; Tellipalai Trail Cancer Hospital, Jaffna, Sri Lanka
| | - Clayton Richards
- Cancer ECHO Initiative, Project ECHO; University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Sanjeev Arora
- Cancer ECHO Initiative, Project ECHO; University of New Mexico Health Sciences Center, Albuquerque, NM
| | - Mitesh J Borad
- Liver and Biliary Cancer Research Program, Division of Hematology and Medical Oncology, Mayo Clinic, Phoenix, AZ; Mayo Clinic Comprehensive Cancer Center, Phoenix, AZ
| |
Collapse
|
78
|
Chew V. Targeting TM4SF1 to overcome immunotherapy resistance in hepatocellular carcinoma: Editorial on "Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma". Clin Mol Hepatol 2025; 31:642-645. [PMID: 39815771 PMCID: PMC12016644 DOI: 10.3350/cmh.2025.0031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 01/12/2025] [Indexed: 01/18/2025] Open
Affiliation(s)
- Valerie Chew
- Translational Immunology Institute (TII), SingHealth-DukeNUS Academic Medical Centre, Singapore, Singapore
| |
Collapse
|
79
|
Galli E, Patelli G, Villa F, Gri N, Mazzarelli C, Mangoni I, Sgrazzutti C, Ghezzi S, Sartore-Bianchi A, Belli LS, De Carlis L, Vanzulli A, Siena S, Bencardino K. Circulating blood biomarkers for minimal residual disease in hepatocellular carcinoma: A systematic review. Cancer Treat Rev 2025; 135:102908. [PMID: 40058162 DOI: 10.1016/j.ctrv.2025.102908] [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: 11/09/2024] [Revised: 02/24/2025] [Accepted: 02/25/2025] [Indexed: 04/08/2025]
Abstract
BACKGROUND Relapse after radical treatment remains a major concern in hepatocellular carcinoma (HCC), affecting 50-75 % of early-stage cases within 5 years. Early recurrence prediction is a clinical unmet need. Circulating blood biomarkers could provide a minimally invasive approach to detect minimal residual disease (MRD) post-intervention. Although alpha-fetoprotein has been the primary biomarker in this setting, its MRD sensitivity is limited to 50-70 %. This systematic review aims to summarize available evidence regarding the clinical validity and potential utility of emerging circulating blood biomarkers for MRD detection in HCC patients. METHODS We searched PubMed and Embase for peer-reviewed articles and abstracts published up to 2025, and ClinicalTrials.gov for ongoing trials on circulating blood biomarkers for MRD in HCC. RESULTS A total of 91 studies (74 with results and 17 ongoing, out of 2,386) were retrieved. We evaluated various blood biomarkers, including circulating DNA (cDNA, N = 24), circulating tumor cells (CTCs, N = 20), circulating RNA (cRNA, N = 8), and other miscellaneous (N = 22) for MRD detection in HCC. These biomarkers demonstrated encouraging results, albeit with notable heterogeneity. In particular, circulating tumor DNA (ctDNA) and CTCs stand as the most robust novel approaches, with 50-80 % sensitivity and specificity up to 94 %. Nonetheless, none of the 17 ongoing studies involve biomarker-driven intervention to prove clinical utility. CONCLUSIONS Novel circulating blood biomarkers are mature for MRD detection in HCC. However, variability in methodologies and results highlights the need for further validation. We encourage the investigation of CTCs and/or ctDNA in interventional trials to assess clinical utility. This biomarker-driven approach may enhance adjuvant treatment effectiveness in MRD-positive cases while minimizing toxicity in MRD-negative patients.
Collapse
Affiliation(s)
- Edoardogregorio Galli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Giorgio Patelli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; IFOM ETS - The AIRC Institute of Molecular Oncology, Milan, Italy.
| | - Federica Villa
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Nicole Gri
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Chiara Mazzarelli
- Hepatology and Gastroenterology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Iacopo Mangoni
- Department of General Surgery and Transplantation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | | | - Silvia Ghezzi
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Andrea Sartore-Bianchi
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy; Division of Clinical Research and Innovation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Luca Saverio Belli
- Hepatology and Gastroenterology Unit, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Luciano De Carlis
- Department of General Surgery and Transplantation, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Angelo Vanzulli
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Department of Radiology, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Salvatore Siena
- Department of Oncology and Hemato-Oncology, Università degli Studi di Milano (La Statale), Milan, Italy; Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| | - Katia Bencardino
- Niguarda Cancer Center, Department of Hematology, Oncology and Molecular Medicine, Grande Ospedale Metropolitano Niguarda, Milan, Italy
| |
Collapse
|
80
|
Liang S, Yuan L, Wang A, Li S, Wei Y, Wen T, Li T, Yang X, Ren Q, Zhu C, Wu M. Effect of short-term exercise-based prehabilitation program for patients undergoing liver cancer surgery: A randomized controlled trial. Surgery 2025; 180:109115. [PMID: 39826234 DOI: 10.1016/j.surg.2024.109115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 12/14/2024] [Accepted: 12/18/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Patients with liver cancer usually experience postoperative complications and reduced perioperative functional capacity. This study aimed to assess the effect of a short-term, exercise-based prehabilitation program on postoperative clinical outcomes and perioperative functional capacity in patients with liver cancer undergoing hepatectomy. METHODS This single-center, prospective, open-labeled randomized controlled trial was conducted with 205 patients. Patients in the prehabilitation group (n = 104) received a 1-week exercise intervention program before surgery, including aerobic and resistance exercises, and respiratory training. Patients in the control group (n = 101) received the usual clinical care. The assessors were blinded to the patient allocation. The primary outcome was the incidence of postoperative pulmonary complication during hospitalization, which the multivariate logistic regression model analyzed. Other outcomes included functional capacity measured as the 6-minute walk distance, postoperative complications, length of stay, hospital readmissions, hospitalization cost, and patient-reported outcomes. Post hoc subgroup analyses were performed. RESULTS The median duration of prehabilitation was 8 days. There was no between-group difference in the incidence of postoperative pulmonary complication (adjusted odds ratio, 0.70; 95% confidence interval, 0.37-1.29; P = .249). There were no differences in postoperative clinical outcomes and patient-reported outcomes, except for 6-minute walk distance (33.36 m higher in the prehabilitation group, 95% CI, 22.02-44.70; P < .001). CONCLUSION This exercise prehabilitation program did not affect postoperative clinical outcomes or patient-reported outcomes of patients with liver cancer, but it showed improvement in preoperative functional capacity.
Collapse
Affiliation(s)
- Shiqi Liang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Linyan Yuan
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Ao Wang
- Rehabilitation Medicine Center, Institute of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China; Key Laboratory of Rehabilitation Medicine in Sichuan Province, Chengdu, China
| | - Siqin Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Yonggang Wei
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tianfu Wen
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Tingting Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Xiaoling Yang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Qiuping Ren
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China
| | - Cairong Zhu
- Department of Epidemiology and Health Statistics, West China School of Public Health and West China Fourth Hospital, Sichuan University, Chengdu, China
| | - Menghang Wu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, China.
| |
Collapse
|
81
|
Zhu Y, Huang F, Liu X, Hou Y, Huang Y. Phillyrin regulates the JAK2/STAT3 signaling pathway by inhibiting TOP2A expression to accelerate ferroptosis in hepatocellular carcinoma. Oncol Rep 2025; 53:43. [PMID: 39950325 PMCID: PMC11843411 DOI: 10.3892/or.2025.8876] [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: 10/18/2024] [Accepted: 01/14/2025] [Indexed: 02/23/2025] Open
Abstract
Despite advancements and refinements in the therapeutic approaches for hepatic malignancies, liver cancer remains a prevalent and deadly form of cancer, with its grim outlook posing as a significant clinical challenge. Phillyrin (PHN) has been reported to have anticancer effects, but the anticancer mechanism in liver cancer is ominous. By searching the potential target of PHN in the online database and liver cancer disease database, it was found that there is only one overlap gene, and DNA topoisomerase II alpha (TOP2A) is abnormally expressed in liver cancer tissues. TOP2A overexpression and downregulated hepatocellular carcinoma cell lines were then constructed in vitro, and it was examined whether PHN treatment induced ferroptosis in hepatocellular carcinoma by regulating TOP2A's inhibition of Janus kinase 2/Signal transducer and activator of transcription 3 (JAK2/STAT3) signaling pathway through phenotypic assay, western blot assay, reverse transcription‑quantitative PCR assay and electron microscopy. The results showed that PHN could inhibit the expression of TOP2A protein and JAK2/STAT3 signaling pathway in hepatoma cells. PHN could also downregulate glutathione peroxidase 4 by suppressing the expression of TOP2A protein. PHN impeded the activity of factor inhibiting hypoxia‑inducible factor 1 alpha, thereby augmenting the synthesis of iron‑dependent apoptosis‑related proteins including cytochrome c oxidase subunit II, long‑chain acyl‑CoA synthetase family member 4 and NADPH oxidase 1, thus facilitating an increase in Fe2+ concentration and accelerating oxidative harm within hepatocellular carcinoma cells, culminating in the induction of ferroptotic cell death in these liver malignancy cells.
Collapse
Affiliation(s)
- Ying Zhu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Fenghe Huang
- Hebei Yiling Medical Research Institute Co., LTDS, Shijiazhuang, Hebei 050000, P.R. China
| | - Xiyu Liu
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yunlong Hou
- Hebei Yiling Medical Research Institute Co., LTDS, Shijiazhuang, Hebei 050000, P.R. China
| | - Yong Huang
- State Key Laboratory of Targeting Oncology, National Center for International Research of Biotargeting Theranostics, Guangxi Key Laboratory of Biotargeting Theranostics, Collaborative Innovation Center for Targeting Tumor Diagnosis and Therapy, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
- Guangxi Talent Highland of Major New Drugs Innovation and Development, Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
82
|
Huang W, Zhong L, Shi Y, Ma Q, Yang X, Zhang H, Zhang J, Wang L, Wang K, Li J, Zou J, Yang X, Yang L, Zeng Q, Jing L, Chen Z, Zhao Y. An Anti-CD147 Antibody-Drug Conjugate Mehozumab-DM1 is Efficacious Against Hepatocellular Carcinoma in Cynomolgus Monkey. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410438. [PMID: 39985225 PMCID: PMC12005782 DOI: 10.1002/advs.202410438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/21/2025] [Indexed: 02/24/2025]
Abstract
Effective treatment strategies are urgently needed for hepatocellular carcinoma (HCC) patients due to frequent therapeutic resistance and recurrence. Antibody-drug conjugate (ADC) is a specific antibody-drug conjugated with small molecular compounds, which has potent killing activity against cancer cells. However, few ADC candidates for HCC are undergoing clinical evaluation. CD147 is a tumor-associated antigen that is highly expressed on the surface of tumor cells. Here CD147 is found significantly upregulated in tumor tissues of HCC. Mehozumab-DM1, a humanized anti-CD147 monoclonal antibody conjugated with Mertansine (DM1) is developed. Mehozumab-DM1 is effectively internalized by cancer cells and demonstrated potent antitumor efficacy in HCC cells. In vivo evaluation of Mehozumab-DM1 is conducted in a CRISPR-mediated PTEN and TP53 mutation cynomolgus monkey liver cancer model, which is poorly responsive to sorafenib treatment. Mehozumab-DM1 demonstrated potent tumor inhibitory efficacy at doses of 0.2 and 1.0 mg kg-1 treatment groups in cynomolgus monkey. No treatment-related adverse reactions or body weight loss are observed. Interestingly, Mehozumab-DM1 treatment induced RIPK-dependent tumor cell necroptosis through inhibiting IκB kinase/NF-κB pathway. In conclusion, Mehozumab-DM1 potently inhibits hepatoma through effective internalization to release payload and inducing cell necroptosis to enhance the bystander effect, which is a promising treatment for refractory HCC.
Collapse
Affiliation(s)
- Wan Huang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Liping Zhong
- State Key Laboratory of Targeting OncologyNational Center for International Research of Biotargeting TheranosticsGuangxi Key Laboratory of Biotargeting TheranosticsCollaborative Innovation Center for Targeting Tumor Diagnosis and TherapyGuangxi Medical UniversityNanningGuangxi530021China
| | - Ying Shi
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Qingzhi Ma
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Xiangmin Yang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Hongmei Zhang
- Department of Clinical OncologyXijing HospitalFourth Military Medical UniversityXi'anShaanxi710032China
| | - Jing Zhang
- Department of PathologyXijing HospitalThe Fourth Military Medical UniversityXi'anShaanxi710032China
| | - Ling Wang
- Department of Health StatisticsSchool of Preventive MedicineFourth Military Medical UniversityXi'anShaanxi710032China
| | - Kun Wang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Jingzhuo Li
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Jie Zou
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Xu Yang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Liu Yang
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Qingmei Zeng
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Lin Jing
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Zhi‐Nan Chen
- Department of Cell BiologyNational Translational Science Center for Molecular MedicineFourth Military Medical UniversityXi'anShaanxi710032China
- State Key Laboratory of New Targets Discovery and Drug Development for Major DiseasesXi'anShaanxi710032China
| | - Yongxiang Zhao
- State Key Laboratory of Targeting OncologyNational Center for International Research of Biotargeting TheranosticsGuangxi Key Laboratory of Biotargeting TheranosticsCollaborative Innovation Center for Targeting Tumor Diagnosis and TherapyGuangxi Medical UniversityNanningGuangxi530021China
| |
Collapse
|
83
|
Hang T, Fan D, Sun T, Chen Z, Yang X, Yue X. Deep Learning and Hyperspectral Imaging for Liver Cancer Staging and Cirrhosis Differentiation. JOURNAL OF BIOPHOTONICS 2025; 18:e202400557. [PMID: 39873135 DOI: 10.1002/jbio.202400557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/14/2025] [Accepted: 01/16/2025] [Indexed: 01/30/2025]
Abstract
Liver malignancies, particularly hepatocellular carcinoma (HCC), pose a formidable global health challenge. Conventional diagnostic techniques frequently fall short in precision, especially at advanced HCC stages. In response, we have developed a novel diagnostic strategy that integrates hyperspectral imaging with deep learning. This innovative approach captures detailed spectral data from tissue samples, pinpointing subtle cellular differences that elude traditional methods. A sophisticated deep convolutional neural network processes this data, effectively distinguishing high-grade liver cancer from cirrhosis with an accuracy of 89.45%, a sensitivity of 90.29%, and a specificity of 88.64%. For HCC differentiation specifically, it achieves an impressive accuracy of 93.73%, sensitivity of 92.53%, and specificity of 90.07%. Our results underscore the potential of this technique as a precise, rapid, and non-invasive diagnostic tool that surpasses existing clinical methods in staging liver cancer and differentiating cirrhosis.
Collapse
Affiliation(s)
- Tianyi Hang
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Danfeng Fan
- Nanjing University of Chinese Medicine, Nanjing, China
| | - Tiefeng Sun
- Nanjing University of Chinese Medicine, Nanjing, China
- Shandong Academy of Chinese Medicine, Jinan, China
| | | | - Xiaoqing Yang
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Xiaoqing Yue
- Nanjing University of Chinese Medicine, Nanjing, China
- Yucheng People's Hospital, Dezhou, China
| |
Collapse
|
84
|
Zhuo Z, Wu H, Xu L, Ji Y, Li J, Liu L, Zhang H, Yang Q, Zheng Z, Lun W. Machine learning-based integration reveals immunological heterogeneity and the clinical potential of T cell receptor (TCR) gene pattern in hepatocellular carcinoma. Apoptosis 2025; 30:955-975. [PMID: 39904860 DOI: 10.1007/s10495-025-02080-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/13/2025] [Indexed: 02/06/2025]
Abstract
The T Cell Receptor (TCR) significantly contributes to tumor immunity, whereas the intricate interplay with the Hepatocellular Carcinoma (HCC) microenvironment and clinical significance remains largely unexplored. Here, we aimed to examine the function of TCR signaling in tumor immunity and its clinical significance in HCC. Our objective was to employ TCR signaling genes and a machine learning-based integrative methodology to construct a prognostic prediction system termed the TCR score. Herein, we revealed that the TCR score serves as an independent risk factor for overall survival in HCC patients, demonstrating stable and robust performance. The accuracy of the TCR score significantly exceeds that of traditional clinical variables and published signatures. Additionally, the immune infiltration was abundant in patients with low TCR scores. Single-cell cohort analysis further demonstrates that patients with low TCR scores possess an immune-active tumor microenvironment (TME), with T/NK cells enhancing interactions with myeloid cells through signaling networks such as MIF, MK, and SPP1. In response to these changes in the TME, patients with high TCR scores exhibit poorer outcomes and shorter survival in immunotherapy cohorts. In vitro experiments demonstrated that the key TCR signaling biomarker SOS1 knockdown significantly suppresses the HCC cells' capability to proliferate, invade, and migrate while enhancing tumor cell apoptosis. The TCR score could function as a robust and potential tool to predict immune activity and improve clinical outcomes for HCC patients.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- Machine Learning
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Tumor Microenvironment/immunology
- Tumor Microenvironment/genetics
- Male
- Prognosis
- Female
- Signal Transduction
- Gene Expression Regulation, Neoplastic
- Cell Line, Tumor
- Middle Aged
Collapse
Affiliation(s)
- Zewei Zhuo
- School of Medicine, South China University of Technology, Guangzhou, 510006, China
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Huihuan Wu
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China
| | - Lingli Xu
- Dadong Street Community Health Service Center, Guangzhou, 510080, China
| | - Yuran Ji
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Jiezhuang Li
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Liehui Liu
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China
| | - Hong Zhang
- Department of Lymphoma, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, 510080, China.
| | - Qi Yang
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, Guangdong, 510080, China.
| | - Zhongwen Zheng
- Heyuan People's Hospital, Heyuan, Guangdong, 517001, China.
| | - Weijian Lun
- Department of Gastroenterology, The Sixth Affiliated Hospital, South China University of Technology, Foshan, 510315, China.
| |
Collapse
|
85
|
Alshammari KI, Ginawi I, Sherfi H, Ahmed HG. Hepatobiliary Cancers in Saudi Arabia From 2000 to 2025. Cureus 2025; 17:e81994. [PMID: 40351895 PMCID: PMC12065086 DOI: 10.7759/cureus.81994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/14/2025] Open
Abstract
Hepatobiliary cancers present a significant challenge to global health. Saudi Arabia and adjacent Gulf nations experience considerable impacts from these cancers. Numerous risk factors have contributed to the increasing prevalence of these cancers. The primary cases are linked to several factors, including hepatitis viral infection, smoking, alcohol consumption, diabetes mellitus, obesity or being overweight, liver cirrhosis, nonalcoholic fatty liver disease, hemochromatosis, aflatoxins, anabolic steroids, and genetic predisposition. Data regarding hepatobiliary cancers is scarcely obtained from Saudi Arabia. This research aims to clarify the epidemiology and risk factors linked to hepatobiliary cancers in Saudi Arabia. Our investigation revealed a lack of studies that collectively examine hepatocellular cancers in Saudi Arabia, highlighting a distinctive element of the current review. To determine the incidence, prevalence, risk factors, and other epidemiological metrics of hepatobiliary cancer in Saudi Arabia, a search was conducted using Medline/PubMed, Scopus, Web of Knowledge, Google Scholar, and relevant public databases that fulfilled the inclusion criteria. An electronic search was conducted using various keywords related to hepatobiliary cancer in Saudi Arabia. In summary, hepatobiliary cancers exhibit significant prevalence in Saudi Arabia, especially liver cancer. Commonly recognized risk factors for type 2 diabetes mellitus encompass tobacco and alcohol consumption, obesity or overweight status, and viral hepatitis.
Collapse
Affiliation(s)
| | | | | | - Hussain G Ahmed
- Pathology, Prof Medical Research Consultancy Center, El-Obeid, SDN
- Histopathology and Cytology, Faculty of Medical Laboratory Sciences, University of Khartoum, Khartoum, SDN
| |
Collapse
|
86
|
Ramier C, Protopopescu C, Di Beo V, Parlati L, Marcellin F, Carrat F, Asselah T, Bourlière M, Carrieri P. Behaviour-Based Predictive Scores of Hepatocellular Carcinoma in People With Chronic Hepatitis B (ANRS CO22 HEPATHER). Liver Int 2025; 45:e70065. [PMID: 40087922 PMCID: PMC11909585 DOI: 10.1111/liv.70065] [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: 07/15/2024] [Revised: 01/21/2025] [Accepted: 03/05/2025] [Indexed: 03/17/2025]
Abstract
BACKGROUND AND AIMS Early assessment of hepatocellular carcinoma (HCC) risk could improve long-term outcomes in people with chronic hepatitis B virus (HBV) infection. Some existing HCC predictive scores are not easily implementable. We developed easy-to-use HCC predictive scores based on behavioural and routine bio-clinical data in people with chronic HBV infection. METHODS Eight-year follow-up data was analysed from people with chronic HBV infection enrolled in the French ANRS CO22 HEPATHER cohort. Patients were randomly split into two samples (training/testing). A multivariable Cox model for time to HCC was estimated on the training sample. The HCC predictive score was computed by summing the points assigned to model predictors, normalising their coefficients over a 10-year age increment, and rounding to the nearest integer. The Youden index identified the score's optimal risk threshold. Comparisons with existing predictive scores were performed on the testing sample. RESULTS In the study population (N = 4370; 63% of men; 65% of < 50 years old), 56 HCC cases occurred during 25,900 follow-up person-years. Two HCC predictive scores were defined: SADAPTT (daily soft drink consumption, age, hepatitis Delta infection, unhealthy alcohol use, platelet count, heavy tobacco smoking, and HBV treatment) and ADAPTT (the same predictors except for daily soft drink consumption), with ranges 0-13 and 0-14, respectively, and values ≥ 3 indicating a high HCC risk. Their performances were similar to existing scores. CONCLUSIONS We developed two effective behaviour-based HCC predictive scores, implementable in many settings, including primary care and decentralised areas. Further studies are needed to validate these scores in other datasets.
Collapse
Affiliation(s)
- Clémence Ramier
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
| | - Camelia Protopopescu
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
| | - Vincent Di Beo
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
| | - Lucia Parlati
- Département d'Hépatologie/AddictologieUniversité de Paris Cité; INSERM U1016, AP‐HP, Hôpital CochinParisFrance
| | - Fabienne Marcellin
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
| | - Fabrice Carrat
- Institut National de la Santé et de la Recherche Médicale (INSERM), Institut Pierre Louis d'Epidémiologie et de Santé Publique, Sorbonne UniversitéParisFrance
- Hôpital Saint‐Antoine, Unité de Santé Publique, Assistance Publique‐Hôpitaux de Paris (AP‐HP)ParisFrance
| | - Tarik Asselah
- Department of HepatologyCentre de Recherche Sur l'Inflammation, INSERM UMR 1149, Hôpital Beaujon, Université de Paris‐CitéClichyFrance
| | - Marc Bourlière
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
- Département d'hépatologie et GastroentérologieHôpital Saint JosephMarseilleFrance
| | - Patrizia Carrieri
- Aix Marseille Univ, INSERM, IRD, SESSTIM, Sciences Économiques and Sociales de la Santé and Traitement de l'Information MédicaleMarseilleFrance
| |
Collapse
|
87
|
Cheraghpour M, Hatami B, Singal AG. Lifestyle and Pharmacologic Approaches to Prevention of Metabolic Dysfunction-associated Steatotic Liver Disease-related Hepatocellular Carcinoma. Clin Gastroenterol Hepatol 2025; 23:685-694.e6. [PMID: 39800201 DOI: 10.1016/j.cgh.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 01/15/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major concern for public health. Fatty liver disease, related to alcohol misuse or metabolic syndrome, has become the leading cause of chronic liver disease and HCC. The strong association between type 2 diabetes mellitus and HCC can be partly attributed to the development of metabolic dysfunction-associated steatotic liver disease (MASLD). There is a strong interest in strategies that may mitigate HCC risk and reduce HCC incidence in this growing population of at-risk individuals. In this review, we describe the pathogenesis of HCC in patients with MASLD and discuss potential emerging pharmacological and lifestyle interventions for MASLD-related HCC. HCC risk has been observed to be lower with healthy lifestyle behaviors, such as healthy dietary patterns (eg, high consumption of vegetables, whole grains, fish and poultry, yogurt, and olive oil, and low consumption of red and processed meats and dietary sugar) and increased physical activity. Selecting an appropriate pharmacologic approach for individuals with MASLD may also decrease the occurrence of HCC. Metformin, PPAR activators, sodium-glucose cotransporter 2 inhibitors, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, aspirin, and statins have all shown promise to reduce the risk of HCC, although guidelines do not recommend their use for the sole purpose of chemoprevention at this time, given a dearth of data defining their risk-benefit ratio.
Collapse
Affiliation(s)
- Makan Cheraghpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
88
|
Huimin W, Xin W, Shan Y, Junwang Z, Jing W, Yuan W, Qingtong L, Xiaohui L, Jia Y, Lili Y. Lactate promotes the epithelial-mesenchymal transition of liver cancer cells via TWIST1 lactylation. Exp Cell Res 2025; 447:114474. [PMID: 39993459 DOI: 10.1016/j.yexcr.2025.114474] [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: 10/15/2024] [Revised: 02/15/2025] [Accepted: 02/16/2025] [Indexed: 02/26/2025]
Abstract
Elevated lactate levels increase the risk of liver cancer progression. However, the mechanisms by which lactate promotes liver cancer progression remain poorly understood. Epithelial-mesenchymal transition (EMT), characterized by the loss of epithelial cells polarity and cell-cell adhesion, leading to the acquisition of mesenchymal-like phenotypes, is widely recognized as a key contributor to liver cancer progression. TWIST1 (Twist Family BHLH Transcription Factor 1) plays a central role in inducing EMT. Here, we investigated the role of lactate in promoting EMT in liver cancer and the underlying regulatory mechanisms. High levels of lactate significantly promoted EMT progression in liver cancer cells. Mechanistically, lactate-induced lactylation of TWIST1 in vivo and in vitro. Mutation assay confirmed that Lysine 33 (K33) is the major site of TWIST1 lactylation. Moreover, cell fractionation & luciferase reporter assay results identified that TWIST1-K33R mutant impaired the EMT process via inhibiting nuclear import and the transcriptional activity. Thus, our findings provide novel insights into the regulatory role of lactate in EMT in liver cancer pathogenesis. Additionally, targeting of lactate-driven lactylation of TWIST1 may boost the therapeutic strategy for liver cancer.
Collapse
Affiliation(s)
- Wang Huimin
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Wu Xin
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Yu Shan
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Zhang Junwang
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Wen Jing
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Wang Yuan
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Liu Qingtong
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Li Xiaohui
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China
| | - Yao Jia
- Department of Gastroenterology, Shanxi Bethune Hospital, No.99 Longcheng Road, Taiyuan, 030032, China
| | - Yuan Lili
- Department of Gastroenterology, Second Hospital of Shanxi Medical University, No.382 Wuyi Road, Taiyuan, 030000, China.
| |
Collapse
|
89
|
Schürch CM. Liver cancer recurrence predicted by immune-cell location and gene expression. Nature 2025; 640:885-887. [PMID: 40075195 DOI: 10.1038/d41586-025-00734-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
|
90
|
Chen K, Tong AK, Moe FN, Ng DC, Lo RH, Gogna A, Yan SX, Thang SP, Loke KS, Venkatanarasimha NK, Huang HL, Too CW, Ong TS, Yeo EX, Peh DYY, Ng AW, Yang L, Chan WY, Chang JP, Goh BK, Toh HC, Chow PK. The Impact of Radiation Dose and Tumour Burden on Outcomes in Hepatocellular Carcinoma: 11-Year Experience in a 413-Patient Cohort Treated with Yttrium-90 Resin Microsphere Radioembolisation. Liver Cancer 2025; 14:158-179. [PMID: 40255874 PMCID: PMC12005707 DOI: 10.1159/000541539] [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: 11/09/2023] [Accepted: 09/16/2024] [Indexed: 04/22/2025] Open
Abstract
Introduction Transarterial radioembolisation (RE) using yttrium-90 (Y-90) microspheres is a widely used locoregional therapy for a broad spectrum of hepatocellular carcinoma (HCC) given its favourable safety profile. We evaluated the real-world outcomes of unresectable HCC treated with resin Y-90 RE and the relationship between tumour absorbed dose and subsequent curative therapy with survival. Methods Included were consecutive patients treated with Y-90 resin microspheres RE for unresectable HCC between January 2008 and May 2019 at the National Cancer Centre Singapore/Singapore General Hospital. The outcomes were stratified by tumour burden, distribution, presence of portal vein invasion (PVI) and liver function to improve prognostication. Results The median overall survival (OS) evaluated on 413 included patients was 20.9 months (95% CI: 18.2-24.0). More than half of the patients (214/413, 51.8%) had HCC beyond up-to-seven criteria, and 37.3% had portal vein invasion (154/413, 37.3%). Majority (71.7%) had dosimetry calculated based on the partition model. Patients who received ≥150 Gy to tumour had significantly better outcomes (OS 32.2 months, 95% CI: 18.3-46.4) than those who did not (OS 17.5 months, 95% CI: 13.7-22.7, p < 0.001). Seventy patients (17%) received curative therapies after tumour was downstaged by Y-90 RE and had better OS of 79.7 months (95% CI: 40.4 - NE) compared to those who did not receive curative therapies (OS 17.1 months; 95% CI: 13.5-20.4, p < 0.001). RE-induced liver injury was observed in 5.08% of the patients while 3.2% of the patients had possible radiation pneumonitis but none developed Grade 3-4 toxicity. For HCC without PVI, OS differed significantly with performance status, albumin-bilirubin grade, tumour distribution, and radiation dose; for HCC with PVI, Child-Pugh class and AFP were significant predictors of survival. Conclusions Treatment outcomes for unresectable HCC using Y-90 RE were favourable. Incorporating tumour burden and distribution improved prognostication. Patients who received tumour absorbed dose above 150 Gy had better OS. Patients who subsequently received curative therapies after being downstaged by Y-90 RE had remarkable clinical outcomes.
Collapse
Affiliation(s)
- Kaina Chen
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Aaron K.T. Tong
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Fiona N.N. Moe
- National Cancer Centre Singapore, Program in Translational and Clinical Liver Cancer Research, Singapore, Singapore
| | - David C.E. Ng
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Richard H.G. Lo
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Apoorva Gogna
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Sean X. Yan
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Sue Ping Thang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Kelvin S.H. Loke
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | | | - Hian Liang Huang
- Department of Nuclear Medicine and Molecular Imaging, Singapore General Hospital, Singapore, Singapore
| | - Chow Wei Too
- Duke-NUS Medical School Singapore, Singapore, Singapore
- Department of Vascular and Interventional Radiology, Singapore General Hospital, Singapore, Singapore
| | - Timothy S.K. Ong
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Eng Xuan Yeo
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel Yang Yao Peh
- Lee Kong Chian School of Medicine, Nanyang Technological University Singapore, Singapore, Singapore
| | - Ashley W.Y. Ng
- National Cancer Centre Singapore, Program in Translational and Clinical Liver Cancer Research, Singapore, Singapore
| | - Lu Yang
- Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Wan Ying Chan
- National Cancer Centre Singapore, Division of Oncologic Imaging, Singapore, Singapore
| | - Jason P.E. Chang
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
- Duke-NUS Medical School Singapore, Singapore, Singapore
| | - Brian K.P. Goh
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School Singapore, Surgery Academic Clinical Program, Singapore, Singapore
| | - Han Chong Toh
- Duke-NUS Medical School Singapore, Singapore, Singapore
- National Cancer Centre Singapore, Division of Medical Oncology, Singapore, Singapore
| | - Pierce K.H. Chow
- National Cancer Centre Singapore, Program in Translational and Clinical Liver Cancer Research, Singapore, Singapore
- Department of Hepatopancreatobiliary and Transplant Surgery, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore
- Duke-NUS Medical School Singapore, Surgery Academic Clinical Program, Singapore, Singapore
| |
Collapse
|
91
|
Lv J, Gan FY, Li MH, Yin QJ. Silencing NCAPD3 Inhibits Tumor Growth and Metastasis in Hepatocellular Carcinoma by Suppressing PI3K-AKT Signalling Pathway. Curr Med Sci 2025; 45:253-263. [PMID: 40029498 DOI: 10.1007/s11596-025-00026-2] [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: 11/29/2024] [Revised: 01/23/2025] [Accepted: 01/26/2025] [Indexed: 03/05/2025]
Abstract
OBJECTIVE To evaluate the expression pattern of non-SMC condensin II complex subunit D3 (NCAPD3) in hepatocellular carcinoma (HCC) tissues, assess its association with clinical characteristics, and explore the effects of NCAPD3 on HCC cells and the potential underlying mechanisms. METHODS NCAPD3 expression in HCC tumors and adjacent noncancerous tissues was quantified via quantitative PCR. Patients were divided into high- and low-expression groups on the basis of NCAPD3 levels, and associations with clinical parameters were assessed. The effects of NCAPD3 knockdown and the phosphatidylinositol-3-kinase (PI3K) agonist Y-P 740 on cell functions were examined via cell proliferation, Transwell migration, and invasion assays. Differentially expressed genes following NCAPD3 knockdown in SMMC-7721 cells were identified via mRNA sequencing. Western blotting was performed to measure NCAPD3, AKT serine/threonine kinase 1 (AKT1), and phosphorylated AKT1 levels. RESULTS NCAPD3 mRNA expression was notably upregulated in HCC tissues as compared with that in adjacent noncancer tissues. A positive correlation was observed between NCAPD3 expression and both lymphatic and distant metastases in patients with HCC. NCAPD3 knockdown reduced the proliferation and metastasis of SMMC-7721 and Huh-7 cells. mRNA sequencing revealed 140 downregulated genes and 125 upregulated genes. Further validation experiments confirmed that NCAPD3 modulated the PI3K-AKT signalling pathway and that the PI3K agonist Y-P 740 counteracted the effects of NCAPD3 knockdown. CONCLUSIONS Elevated NCAPD3 expression was strongly correlated with HCC metastasis. NCAPD3 inhibition impedes HCC cell growth and metastatic potential by suppressing the PI3K-AKT signalling pathway.
Collapse
Affiliation(s)
- Jun Lv
- Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China.
| | - Fu-Yuan Gan
- Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Ming-Hao Li
- Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| | - Qing-Jun Yin
- Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, China
| |
Collapse
|
92
|
Jia G, He P, Dai T, Goh D, Wang J, Sun M, Wee F, Li F, Lim JCT, Hao S, Liu Y, Lim TKH, Ngo NT, Tao Q, Wang W, Umar A, Nashan B, Zhang Y, Ding C, Yeong J, Liu L, Sun C. Spatial immune scoring system predicts hepatocellular carcinoma recurrence. Nature 2025; 640:1031-1041. [PMID: 40074893 DOI: 10.1038/s41586-025-08668-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2023] [Accepted: 01/17/2025] [Indexed: 03/14/2025]
Abstract
Given the high recurrence rates of hepatocellular carcinoma (HCC) post-resection1-3, improved early identification of patients at high risk for post-resection recurrence would help to improve patient outcomes and prioritize healthcare resources4-6. Here we observed a spatial and HCC recurrence-associated distribution of natural killer (NK) cells in the invasive front and tumour centre from 61 patients. Using extreme gradient boosting and inverse-variance weighting, we developed the tumour immune microenvironment spatial (TIMES) score based on the spatial expression patterns of five biomarkers (SPON2, ZFP36L2, ZFP36, VIM and HLA-DRB1) to predict HCC recurrence risk. The TIMES score (hazard ratio = 88.2, P < 0.001) outperformed current standard tools for patient risk stratification including the TNM and BCLC systems. We validated the model in 231 patients from five multicentred cohorts, achieving a real-world accuracy of 82.2% and specificity of 85.7%. The predictive power of these biomarkers emerged through the integration of their spatial distributions, rather than individual marker expression levels alone. In vivo models, including NK cell-specific Spon2-knockout mice, revealed that SPON2 enhances IFNγ secretion and NK cell infiltration at the invasive front. Our study introduces TIMES, a publicly accessible tool for predicting HCC recurrence risk, offering insights into its potential to inform treatment decisions for early-stage HCC.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Middle Aged
- Biomarkers, Tumor/analysis
- Biomarkers, Tumor/genetics
- Carcinoma, Hepatocellular/diagnosis
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/surgery
- Cohort Studies
- Extracellular Matrix Proteins/genetics
- Extracellular Matrix Proteins/deficiency
- Extracellular Matrix Proteins/metabolism
- Interferon-gamma/metabolism
- Killer Cells, Natural/immunology
- Killer Cells, Natural/cytology
- Liver Neoplasms/diagnosis
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/surgery
- Mice, Knockout
- Neoplasm Recurrence, Local/immunology
- Neoplasm Recurrence, Local/diagnosis
- Neoplasm Recurrence, Local/pathology
- Reproducibility of Results
- Tumor Microenvironment
Collapse
Affiliation(s)
- Gengjie Jia
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Peiqi He
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tianli Dai
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Denise Goh
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Mengyuan Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Felicia Wee
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Fuling Li
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Jeffrey Chun Tatt Lim
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore
| | - Shuxia Hao
- Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen, China
| | - Yao Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Tony Kiat Hon Lim
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Duke-NUS Medical School, Singapore, Singapore
| | | | - Qingping Tao
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Wei Wang
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
- Clinical Research Hospital of Chinese Academy of Sciences, University of Science and Technology of China, Hefei, China
| | - Ahitsham Umar
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Björn Nashan
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China
| | - Yongchang Zhang
- Hunan Cancer Hospital, The Affiliated Cancer Hospital of Central South University, Changsha, China
| | - Chen Ding
- State Key Laboratory of Genetic Engineering and Collaborative Innovation Center for Genetics and Development, School of Life Sciences, Institutes of Biomedical Sciences, Human Phenome Institute, Fudan University, Shanghai, China
| | - Joe Yeong
- Institute of Molecular and Cell Biology (IMCB), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Department of Anatomical Pathology, Singapore General Hospital, Singapore, Singapore.
- Singapore Immunology Network (SIgN), Agency for Science Technology and Research (A*STAR), Singapore, Singapore.
- Cancer Science Institute, National University of Singapore, Singapore, Singapore.
| | - Lianxin Liu
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
| | - Cheng Sun
- Department of Hepatobiliary Surgery, Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of University of Science and Technology of China (USTC), University of Science and Technology of China, Hefei, China.
- Chinese Academy of Sciences Key Laboratory of Innate Immunity and Chronic Disease, Key Laboratory of Immune Response and Immunotherapy, Institute of Immunology, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.
| |
Collapse
|
93
|
Ito T, Shimose S, Tani J, Tomonari T, Saeki I, Takeuchi Y, Hatanaka T, Sasaki K, Kakizaki S, Kanayama Y, Yoshioka N, Naito T, Takeuchi M, Yasunaka T, Sakata M, Iwamoto H, Itano S, Shirono T, Tanabe N, Yamamoto T, Naganuma A, Nishina S, Otsuka M, Takami T, Takayama T, Kawaguchi T, Kawashima H. Antitumor effects and immune-mediated adverse events of durvalumab plus tremelimumab treatment for unresectable hepatocellular carcinoma. Hepatol Res 2025; 55:577-587. [PMID: 40318087 DOI: 10.1111/hepr.14151] [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: 10/04/2024] [Revised: 11/25/2024] [Accepted: 11/26/2024] [Indexed: 01/11/2025]
Abstract
AIM Durvalumab plus tremelimumab (Dur/Tre) is a first-line systemic treatment option for unresectable hepatocellular carcinoma (uHCC). However, the management of severe immune-mediated adverse events (imAEs) is challenging. Therefore, we investigated the relationship between severe imAEs and antitumor responses in patients with uHCC treated with Dur/Tre. METHODS We included 157 patients with uHCC treated with Dur/Tre in this multicenter, retrospective study and analyzed the relationship between progression-free survival (PFS)/antitumor response and severe imAEs requiring high-dose corticosteroid treatment. RESULTS Thirty-two patients (20.4%) developed severe imAEs, including enterocolitis/diarrhea (n = 10), liver injury (n = 9), interstitial lung disease (n = 5), rashes (n = 4), cytokine-release syndrome/fever (n = 2), pancreatitis (n = 2), and others (n = 4) (median follow-up period, 6.8 months). Infliximab was administered in six patients with steroid-refractory enterocolitis. Although the objective response rate (ORR) and disease control rate (DCR) were significantly higher with first-line therapy than with later-line therapy (p = 0.026), the frequency of severe imAEs was not significantly different (p = 0.221). The ORR and DCR in patients with and without severe imAEs were 15.6% and 17.6% and 65.6% and 47.2%, respectively, with no significant differences. Five patients with severe imAEs, including rashes and liver injury, showed objective responses (partial response + complete response). Among patients who achieved an objective response, the PFS at 10 months was good (100% and 70.3% with and without high-dose corticosteroids, respectively). CONCLUSIONS Severe imAEs of Dur/Tre treatment requiring high-dose corticosteroid treatment did not affect antitumor efficacy, which differed depending on the type of imAEs. Therefore, appropriately managing imAEs is essential to guide sequential treatment.
Collapse
Affiliation(s)
- Takanori Ito
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Shigeo Shimose
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Joji Tani
- Faculty of Medicine, Department of Gastroenterology and Neurology, Kagawa University, Takamatsu, Kagawa, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Issei Saeki
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Yasuto Takeuchi
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Kyo Sasaki
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Yuki Kanayama
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Naoki Yoshioka
- Department of Gastroenterology and Hepatology, Japanese Red Cross Aichi Medical Center Nagoya Daiichi Hospital, Nagoya, Japan
| | - Takehito Naito
- Department of Gastroenterology, Toyohashi Municipal Hospital, Toyohashi, Japan
| | - Mamiko Takeuchi
- Department of Gastroenterology, Anjo Kosei Hospital, Anjo, Japan
| | - Tetsuya Yasunaka
- Department of Gastroenterology, Fukuyama City Hospital, Fukuyama, Japan
| | - Masahiro Sakata
- Department of Gastroenterology, National Hospital Organization Fukuyama Medical Center, Fukuyama, Japan
| | - Hideki Iwamoto
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
- Department of Gastroenterology and Hepatology, Iwamoto Internal Medical Clinic, Kitakyusyu, Japan
| | - Satoshi Itano
- Department of Gastroenterology and Hepatology, Kurume Central Hospital, Kurume, Japan
| | - Tomotake Shirono
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Norikazu Tanabe
- Division of Laboratory, Yamaguchi University Hospital, Ube, Japan
| | - Takafumi Yamamoto
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Sohji Nishina
- Department of Gastroenterology and Hepatology, Kawasaki Medical School, Kurashiki, Japan
| | - Motoyuki Otsuka
- Department of Gastroenterology, Okayama University Hospital, Okayama, Japan
| | - Taro Takami
- Department of Gastroenterology and Hepatology, Yamaguchi University Graduate School of Medicine, Yamaguchi, Japan
| | - Tetsuji Takayama
- Department of Gastroenterology and Oncology, Institute of Biomedical Sciences, Tokushima University Graduate School of Medicine, Tokushima, Japan
| | - Takumi Kawaguchi
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Japan
| | - Hiroki Kawashima
- Department of Gastroenterology and Hepatology, Nagoya University Hospital, Nagoya, Japan
| |
Collapse
|
94
|
Uluk D, Pein J, Herda S, Schliephake F, Schneider CV, Bitar J, Dreher K, Eurich D, Zhang IW, Schaffrath L, Auer TA, Collettini F, Engelmann C, Tacke F, Pratschke J, Lurje I, Lurje G. Metabolic Dysfunction-Associated Steatotic Liver Disease (MASLD) Impacts Long-Term Outcomes After Curative-Intent Surgery for Hepatocellular Carcinoma. Aliment Pharmacol Ther 2025; 61:1318-1332. [PMID: 39964081 PMCID: PMC11950813 DOI: 10.1111/apt.70002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 09/18/2024] [Accepted: 01/17/2025] [Indexed: 03/29/2025]
Abstract
BACKGROUND Curative surgery for hepatocellular carcinoma (HCC) includes liver resection (LR) and orthotopic liver transplantation (OLT). Due to the obesity epidemic, metabolic dysfunction-associated steatotic liver disease (MASLD) is a frequent HCC aetiology that often coincides with increased alcohol consumption, termed MetALD, or even alcohol-associated liver disease (ALD). METHODS Patients undergoing LR or OLT for HCC at Charité-Universitätsmedizin Berlin (2010-2020) were included in this retrospective cohort study investigating disease aetiology, time to recurrence (TTR), overall survival (OS) and CT-based body composition. RESULTS Out of 579 patients with HCC, 417 underwent LR and 162 OLT. Tumour aetiologies were viral n = 191 (33.0%), MASLD n = 158 (27.3%), MetALD n = 51 (8.8%), ALD n = 68 (11.7%) and other/cryptogenic n = 111 (19.2%). Patients with MASLD and MetALD had more intramuscular (p < 0.001, p = 0.015) and visceral fat (both p < 0.001) than patients with non-metabolic dysfunction aetiologies. Patients with MASLD-HCC had comparable TTR (median 26 months, [95% CI: 23-31] vs. 30 months [95% CI: 4-57], p = 0.425) but shorter OS than patients with other HCC aetiologies (63 months [95% CI: 42-84] vs. 80 months [95% CI: 60-100], hazard ratio: 1.53 [95% CI: 1.050-2.229], p = 0.026) after LR. Multivariate analysis confirmed MASLD aetiology, portal vein thrombosis and MELD score ≥ 10 as independent prognostic factors for OS in LR (adjusted p = 0.021,p < 0.001,p = 0.003), even after excluding in-hospital mortality (adjusted p = 0.016,p = 0.002,p = 0.002). Causes of death were similar in MASLD and non-MASLD aetiology. CONCLUSIONS Patients with HCC undergoing LR and meeting the new MASLD criteria have significantly shorter OS. This study provides empirical prognostic evidence for the novel MASLD/MetALD classification in a large European cohort of patients undergoing curative-intent HCC therapy.
Collapse
Affiliation(s)
- Deniz Uluk
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
- Department of General, Visceral and Transplantation SurgeryHeidelberg University HospitalHeidelbergGermany
| | - Justus Pein
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Sophia Herda
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Frederik Schliephake
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
- Department of General, Visceral and Transplantation SurgeryHeidelberg University HospitalHeidelbergGermany
| | | | - Jude Bitar
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Katharina Dreher
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Dennis Eurich
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Ingrid W. Zhang
- Department of Gastroenterology and Hepatology, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Lukas Schaffrath
- Department of Gastroenterology and Hepatology, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Timo A. Auer
- Department of RadiologyCharité – Universitätsmedizin BerlinBerlinGermany
| | | | - Cornelius Engelmann
- Department of Gastroenterology and Hepatology, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Frank Tacke
- Department of Gastroenterology and Hepatology, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Johann Pratschke
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Isabella Lurje
- Department of General, Visceral and Transplantation SurgeryHeidelberg University HospitalHeidelbergGermany
- Department of Gastroenterology and Hepatology, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
| | - Georg Lurje
- Department of Surgery, Campus Charité Mitte, Campus Virchow KlinikumCharité‐Universitätsmedizin BerlinBerlinGermany
- Department of General, Visceral and Transplantation SurgeryHeidelberg University HospitalHeidelbergGermany
| |
Collapse
|
95
|
Yang H, Kong P, Hou S, Dong X, Abula I, Yan D. Potential prognostic biomarker SERPINA12: implications for hepatocellular carcinoma. Clin Transl Oncol 2025; 27:1597-1611. [PMID: 39235554 PMCID: PMC12000224 DOI: 10.1007/s12094-024-03689-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Accepted: 08/21/2024] [Indexed: 09/06/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains one of the most prevalent malignant tumors, exhibiting a high morbidity and mortality rate. The mechanism of its occurrence and development requires further study. The objective of this study was to investigate the role of SERPINA12 in the diagnosis, prognosis prediction and biological function within HCC. METHODS The Cancer Genome Atlas (TCGA) data were employed to analyze the relationship between clinical features and SERPINA12 expression in HCC. Kaplan-Meier curves were utilized to analyze the correlation between SERPINA12 expression and prognosis in HCC. The function of SERPINA12 was determined by enrichment analysis, and the relationship between SERPINA12 expression and immune cell infiltration was investigated. The expression of SERPINA12 was examined in 75 patients with HCC using RT-qPCR and immunohistochemistry, and survival analysis was performed. RESULTS The expression of SERPINA12 from TCGA database was found to be significantly higher in HCC tissues than in normal tissues and carried a poor prognosis. ROC curve demonstrated the diagnostic potential of SERPINA12 for HCC. The multivariate Cox regression analysis showed that pathologic T stage, tumor status, and SERPINA12 expression were independently associated with patient survival. The SERPINA12 expression was found to correlate with immune cell infiltration. Our RT-qPCR and immunohistochemical analysis revealed high expression of SERPINA12 in tumor tissues. Survival analysis indicated its association with poor prognosis. CONCLUSION SERPINA12 is a promising biomarker for diagnosis and prognosis, and it is associated with immune cell infiltration.
Collapse
Affiliation(s)
- Huan Yang
- The Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Panpan Kong
- The Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Songyu Hou
- The Department of Daily Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, UrumqiXinjiang, 830011, China
| | - Xiaogang Dong
- The Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Imamumaimaitijiang Abula
- The Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China
| | - Dong Yan
- The Department of Hepatopancreatobiliary Surgery, The Affiliated Tumor Hospital of Xinjiang Medical University, Urumqi, 830011, Xinjiang, China.
| |
Collapse
|
96
|
Jin SH, Kim DJ, Lee JW. Tetraspan(in)-mediated immune regulation in hepatocellular carcinoma: Editorial on "Targeting TM4SF1 promotes tumor senescence enhancing CD8+ T cell cytotoxic function in hepatocellular carcinoma". Clin Mol Hepatol 2025; 31:650-653. [PMID: 39930836 PMCID: PMC12016603 DOI: 10.3350/cmh.2025.0109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 02/07/2025] [Indexed: 04/18/2025] Open
Affiliation(s)
- Seo Hee Jin
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Dong Joo Kim
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
| | - Jung Weon Lee
- Department of Pharmacy, College of Pharmacy, Seoul National University, Seoul, Korea
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul, Korea
| |
Collapse
|
97
|
Bazzani V, Kundnani DL, Redin ME, Agostini F, Chhatlani K, Faini AC, Poziemski J, Baccarani U, Siedlecki P, Deaglio S, Storici F, Vascotto C. Characterization of a new mutation of mitochondrial ND6 gene in hepatocellular carcinoma and its effects on respiratory complex I. Sci Rep 2025; 15:10925. [PMID: 40157968 PMCID: PMC11954883 DOI: 10.1038/s41598-025-91746-x] [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: 12/03/2024] [Accepted: 02/24/2025] [Indexed: 04/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the most common form of liver cancer, which often arises from previous liver pathologies such as HBV, HCV, and alcohol abuse. It is typically associated with an enlarged cirrhotic organ. In this study, we analyzed tumor and distal tissues from a patient who underwent liver resection for HCC with no previous pathologies and whose liver showed normal function without signs of cirrhosis. Genetic analysis of mitochondrial DNA (mtDNA) revealed a novel variant of the gene encoding the NADH dehydrogenase subunit 6 (ND6) protein in the tumor tissue. The deletion of a thymidine generated an early stop codon, resulting in a truncated form of the protein (ΔND6) with 50% of the C-terminal primary sequence missing. ND6 is a subunit of the NADH dehydrogenase complex, also known as Complex I, the largest complex in the electron transport chain. Previous studies have linked mtDNA Complex I mutations to mitochondrial disorders and cancer. Through biochemical analyses, we characterized this new mutation and showed that the expression of ΔND6 negatively affects the stability and functionality of Complex I. Data were confirmed by molecular dynamics simulations suggesting conformational rearrangements, overall revealing a leading role of ND6 in the assembly of Complex I.
Collapse
Affiliation(s)
| | - Deepali L Kundnani
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | | | | | - Kirti Chhatlani
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Angelo Corso Faini
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin, 10126, Italy
| | - Jakub Poziemski
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | | | - Pawel Siedlecki
- Institute of Biochemistry and Biophysics, Polish Academy of Sciences, Warsaw, 02-106, Poland
| | - Silvia Deaglio
- Department of Medical Sciences, University of Turin, Turin, 10126, Italy
- Immunogenetics and Transplant Biology Unit, Città della Salute e della Scienza Hospital, Turin, 10126, Italy
| | - Francesca Storici
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Carlo Vascotto
- IMol Polish Academy of Sciences, Warsaw, 02-247, Poland.
- Department of Medicine, University of Udine, Udine, 33100, Italy.
| |
Collapse
|
98
|
Kaseb AO. Access to Medical Research is a Basic Human Right: A Roadmap to Research Integration Into a Practical Hepatocellular Carcinoma Treatment Allocation Algorithm (HCC-TAA). J Hepatocell Carcinoma 2025; 12:649-657. [PMID: 40171232 PMCID: PMC11960483 DOI: 10.2147/jhc.s523470] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2025] [Accepted: 03/14/2025] [Indexed: 04/03/2025] Open
Affiliation(s)
- Ahmed Omar Kaseb
- Department of Gastrointestinal Medical Oncology, Unit 426, University of Texas MD Anderson Cancer Center, Houston, TX, 77030, USA
| |
Collapse
|
99
|
Shi H, Hu C. A prediction model based on machine learning: prognosis of HBV-induced HCC male patients with smoking and drinking habits after local ablation treatment. Front Immunol 2025; 16:1464863. [PMID: 40226611 PMCID: PMC11985516 DOI: 10.3389/fimmu.2025.1464863] [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: 07/15/2024] [Accepted: 03/11/2025] [Indexed: 04/15/2025] Open
Abstract
Background Liver cancer, particularly hepatocellular carcinoma (HCC), is a major health concern globally and in China, possibly shows recurrence after ablation treatment in high-risk patients. This study investigates the prognosis of early-stage male HCC patients with chronic hepatitis virus B (HBV) infection who also have long-term smoking and drinking habits, following local ablation treatment. Methods Data from 257 patients treated at Capital Medical University, Beijing Youan Hospital from 2014 to 2022 were retrospectively analyzed. We first screened the variables by Lasso regression and random survival forest (RSF), followed by multivariate Cox regression analysis. Based on the screened variables after these steps, we performed and validated a nomogram to predict the survival status of these patients. Results Our results indicated that monocytes and globulin are risk factors while pre-albumin (PALB) is protective after selected by Lasso, RSF and multivariate Cox regression, providing a robust tool for predicting overall survival and guiding treatment for high-risk HCC patients. With promising discrimination, accuracy and clinical applicability, our model was translated into a nomogram for practical use. Conclusion Our prognostic model effectively identifies key risk factors such as monocytes, globulin and PALB, providing accurate predictions for HBV-induced male patients with smoking and drinking habits.
Collapse
Affiliation(s)
- Han Shi
- Beijing You’an Hospital, Capital Medical University, Beijing, China
| | - Caixia Hu
- Beijing You’an Hospital, Capital Medical University, Beijing, China
- Interventional therapy center for oncology, Beijing You ‘an Hospital, Capital Medical University, Beijin, China
| |
Collapse
|
100
|
Yang Z, Feng X, Yu H, Lv L, Gao C, Liu W, Yi S, Jia C, Fu B. Identification of tumor immune infiltration-associated VPS72 and prognostic significance of VPS72 and CD8A in hepatocellular carcinoma. Discov Oncol 2025; 16:410. [PMID: 40146476 PMCID: PMC11950588 DOI: 10.1007/s12672-025-02017-0] [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: 09/29/2024] [Accepted: 02/25/2025] [Indexed: 03/28/2025] Open
Abstract
BACKGROUND Copy Number Alterations (CNAs)-driven genes have gained attention as potential markers for predicting the response to immune checkpoint blockade in cancer treatment. Among them, VPS72 has emerged as a promising candidate in hepatocellular carcinoma (HCC). However, the relationship between VPS72 and immune infiltration remains unclear. METHODS TIMER analysis was performed to identify immune populations in bulk-RNAseq data. Then, we investigated the relationship between VPS72 and immune infiltration in HCC using diverse data sources, including the TCGA and GEO databases, clinical specimens, and animal models. RESULTS Our findings in the immunogenomic and TCGA-LIHC studies revealed significant enrichment of VPS72 among IRG in the altered group. Differential analysis and KEGG pathway analysis further highlighted the involvement of differentially expressed genes (DETs) in pathways related to the T cell receptor signaling pathway. Importantly, TIMER analysis suggested that low expression of VPS72 was associated with high infiltration of CD8 + T cells in multiple publicly available HCC datasets. To validate these findings, we conducted in vivo experiments and observed higher CD8A expression in VPS72-knockdown tumors. Additionally, in our patient cohort, individuals with low VPS72 expression exhibited higher CD8A expression. Furthermore, we identified a co-expression subtype characterized by low VPS72 and high CD8A levels, which showed a more favorable disease-free survival outcome in HCC. CONCLUSIONS The expression of VPS72 in tumors is associated with the tumor infiltration. VPS72 and CD8A coexpression are prognostic biomarkers in HCC.
Collapse
Affiliation(s)
- Zhou Yang
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Xiao Feng
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Haoyuan Yu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Lei Lv
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Chengli Gao
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Wei Liu
- Guangdong Provincial Key Laboratory of Liver Disease Research, Guangdong Province Engineering Laboratory for Transplantation Medicine, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Shuhong Yi
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China
| | - Changchang Jia
- Cell-Gene Therapy Translational Medicine Research Center, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China.
| | - Binsheng Fu
- Department of Hepatic Surgery and Liver Transplantation Center of the Third Affiliated Hospital, Organ Transplantation Institute, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|