101
|
Xu X, Zhang Y, Wu S, Wu Y, Lin X, Chen K, Lin X. Hepatitis B Virus Promotes Angiogenesis in Hepatocellular Carcinoma by Increasing m6A Modification of VEGFA mRNA via IGF2BP3. J Med Virol 2025; 97:e70356. [PMID: 40260505 DOI: 10.1002/jmv.70356] [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: 02/11/2025] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/23/2025]
Abstract
Angiogenesis plays a crucial role in the development of HBV-related hepatocellular carcinoma (HCC). VEGFA is a key angiogenic factor, and while its transcriptional regulation by HBV has been extensively studied, its posttranscriptional regulation by HBV remains poorly understood. Building on our previous findings that delineated an RBM15/YTHDF2/IGF2BP3 regulatory axis in m6A-mediated RNA metabolism in HCC, this study further explores the posttranscriptional regulation of VEGFA by HBV. By MeRIP-qPCR and integrating MeRIP-seq data, we discovered that HBV enhances m6A methylation of VEGFA mRNA. Comprehensive cellular and molecular biology experiments demonstrated that HBV induces the upregulation of IGF2BP3, which serves as a key "reader" that recognizes and stabilizes VEGFA mRNA in an m6A methylation-dependent manner. This stabilization leads to elevated VEGFA expression, promoting enhanced cellular functions such as HUVEC migration and tube formation. Furthermore, in an HBV-associated HCC xenograft model, IGF2BP3 knockdown resulted in decreased VEGFA expression and inhibited tumor growth. This study expands our understanding of HBV-driven angiogenesis and identifies the IGF2BP3-VEGFA axis as a potential therapeutic target for antiangiogenic strategies in HBV-related HCC.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/virology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/metabolism
- Vascular Endothelial Growth Factor A/genetics
- Vascular Endothelial Growth Factor A/metabolism
- RNA-Binding Proteins/metabolism
- RNA-Binding Proteins/genetics
- Liver Neoplasms/virology
- Liver Neoplasms/pathology
- Liver Neoplasms/genetics
- Liver Neoplasms/metabolism
- Animals
- Neovascularization, Pathologic/virology
- Neovascularization, Pathologic/genetics
- RNA, Messenger/metabolism
- RNA, Messenger/genetics
- Hepatitis B virus/pathogenicity
- Hepatitis B virus/physiology
- Mice
- Methylation
- Cell Line, Tumor
- Human Umbilical Vein Endothelial Cells
- Mice, Nude
- Hepatitis B/virology
- Hepatitis B/complications
- Angiogenesis
Collapse
Affiliation(s)
- Xiaoxin Xu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
| | - Yi Zhang
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
| | - Shuxiang Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| | - Yuecheng Wu
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
| | - Xinjian Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
| | - Kunqi Chen
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
| | - Xu Lin
- Key Laboratory of Gastrointestinal Cancer (Fujian Medical University), Ministry of Education, School of Basic Medical Sciences, Fuzhou, China
- Fujian Key Laboratory of Tumor Microbiology, Department of Medical Microbiology, School of Basic Medical Sciences, Fujian Medical University, Fuzhou, China
| |
Collapse
|
102
|
Wang P, Qiu J, Fang Y, Li S, Liu K, Cao Y, Zhang G, Wang Z, Gu X, Wu J, Jiang C. SENP3 inhibition suppresses hepatocellular carcinoma progression and improves the efficacy of anti-PD-1 immunotherapy. Cell Death Differ 2025; 32:959-972. [PMID: 39755756 PMCID: PMC12089275 DOI: 10.1038/s41418-024-01437-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 12/08/2024] [Accepted: 12/16/2024] [Indexed: 01/06/2025] Open
Abstract
The importance of SUMOylation in tumorigenesis has received increasing attention, and research on therapeutic agents targeting this pathway has progressed. However, the potential function of SUMOylation during hepatocellular carcinoma (HCC) progression and the underlying molecular mechanisms remain unclear. Here, we identified that SUMO-Specific Peptidase 3 (SENP3) was upregulated in HCC tissues and correlated with a poor prognosis. Multiple functional experiments demonstrated that SENP3 promotes the malignant phenotype of HCC cells. Mechanistically, SENP3 deSUMOylates RACK1 and subsequently increases its stability and interaction with PKCβII, thereby promoting eIF4E phosphorylation and translation of oncogenes, including Bcl2, Snail and Cyclin D1. Additionally, tumor-intrinsic SENP3 promotes the infiltration of tumor-associated macrophages (TAMs) while reducing cytotoxic T cells to facilitate immune evasion. Mechanistically, SENP3 promotes translation of CCL20 via the RACK1 /eIF4E axis. Liver-specific knockdown of SENP3 significantly inhibits liver tumorigenesis in a chemically induced HCC model. SENP3 inhibition enhances the therapeutic efficacy of PD-1 blockade in an HCC mouse model. Collectively, SENP3 plays cell-intrinsic and cell-extrinsic roles in HCC progression and immune evasion by modulating oncogene and cytokine translation. Targeting SENP3 is a novel therapeutic target for boosting HCC responsiveness to immunotherapy.
Collapse
Affiliation(s)
- Peng Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Jiannan Qiu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
| | - Yuan Fang
- Department of Oncology, the First Affiliated Hospital of Nanjing Medical University, Nanjing, China
| | - Songmao Li
- Department of Immunology, Key Laboratory of Immune Microenvironment and Disease,, Nanjing Medical University, Nanjing, China
| | - Kua Liu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China
| | - Yin Cao
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Guang Zhang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhongxia Wang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
| | - Xiaosong Gu
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
| | - Junhua Wu
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- State Key Laboratory of Pharmaceutical Biotechnology, National Institute of Healthcare Data Science at Nanjing University, Jiangsu Key Laboratory of Molecular Medicine, Medical School, Nanjing University, Nanjing, China.
| | - Chunping Jiang
- Division of Hepatobiliary and Transplantation Surgery, Department of General Surgery, Nanjing Drum Tower Hospital, the Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, China.
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Fujian Medical University, Quanzhou, China.
| |
Collapse
|
103
|
Choudhury A, Roy A, Mukund A, Sharma D, Heo S, Choi WM. Managing Complex Hepatocellular Carcinoma Subtypes: Diffuse Infiltrative, Large Tumours, and Tumour Rupture-The Challenges and Strategies. J Clin Exp Hepatol 2025; 15:102505. [PMID: 40028241 PMCID: PMC11870255 DOI: 10.1016/j.jceh.2025.102505] [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: 06/02/2024] [Accepted: 01/11/2025] [Indexed: 03/05/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cause of cancer globally, third most common cause of cancer-related death, and most common primary liver malignancy. Whilst nodular well-defined HCC remains the classical phenotype, presentations with infiltrative phenotype, very large HCC, and complications as tumour rupture provide immense diagnostic and therapeutic challenges. Infiltrative HCC is difficult to distinguish against the background cirrhotic liver. They are ill defined on imaging, have poor vascularity, and aggressive biological behaviour. Vascular invasion, metastasis, and poor response to loco-regional, as well as systemic therapy, leads to dismal prognosis. Very large HCCs have a relatively better prognosis than infiltrative HCC and mandate multimodal therapies to downstage for a curative response including liver transplant. Improvement in interventional radiology techniques, emerging evidence with systemic therapies including immunotherapy, and better understanding of tumour biology have opened newer avenues in the management of such complex cases. HCC rupture is a catastrophic moment in the natural history of HCC which has an exponential increase in mortality. Clinical presentation of pain abdomen, hypotension/syncope, new onset, or sudden increase in ascites mandates a strong suspicion of rupture. Presence of hemoperitoneum on diagnostic tap and contrast extravasation in a computed tomography/magnetic resonance imaging are the diagnostic hallmarks. Emergency surgical intervention, locoregional therapies in the form of bland embolisation, or chemoembolisation forms the management cornerstone. The long-term survival and liver transplant as a curative therapy still needs more data as fear of tumour spread is a possibility. This review summarises the clinical challenges with this advance HCC and provides an algorithmic approach for management.
Collapse
Affiliation(s)
- Ashok Choudhury
- Department of Hepatology and Liver Transplant, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Akash Roy
- Apollo Multispeciality Hospitals, Kolkata and Apollo Hospitals Educational and Research Foundation), Institute of Gastrosciences and Liver Transplantation, Kolkata, India
| | - Amar Mukund
- Department of Intervention Radiology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Deepti Sharma
- Department of Radiation Oncology, Institute of Liver and Biliary Sciences, New Delhi, India
| | - Subin Heo
- Department of Radiology and Research Institute of Radiology, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Won-Mook Choi
- Department of Gastroenterology, Liver Centre, Asan Medical Centre, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
104
|
Huang P, Rodriguez-Matos FJ, Qi J, Trehan R, Myojin Y, Zhu XB, Greten TF, Ma C. Hepatic immune environment differences among common mouse strains in models of MASH and liver cancer. JHEP Rep 2025; 7:101380. [PMID: 40342632 PMCID: PMC12060451 DOI: 10.1016/j.jhepr.2025.101380] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 02/20/2025] [Accepted: 02/24/2025] [Indexed: 05/11/2025] Open
Abstract
Background & Aims Inbred mouse strains are critical tools for studying immune regulation of metabolic dysfunction-associated steatohepatitis (MASH) and hepatocellular carcinoma (HCC). Here, we profiled mouse strain-associated hepatic immune differences, and performed mice-human cross-species immune comparisons. Methods Immune landscapes of C57BL/6, BALB/c, and FVB/N mice were compared under healthy, MASH, or HCC state using high-dimensional spectral flow cytometry (n = 4 per condition). MASH was induced by feeding methionine- and choline-deficient or Western diet + carbon tetrachloride. HCC was caused by hydrodynamic plasmid injection of MYC/sg-p53. Public mouse and human scRNA-seq datasets were used for validation and cross-species comparisons. Results In healthy mice, liver CD4+ T (24% vs. 14% vs. 34%, p <0.05) and B cells (36.5% vs. 35% vs.18%, p <0.05) varied the most among three strains. C57BL/6 mice showed TH1 dominance, whereas BALB/c and FVB/N mice had TH2 dominance (log[TH1:TH2] = 0.17, -0.31, -0.17). In MASH mice, expansion of liver myeloid cells and innate lymphocytes were commonly found, but changes of B cells (log(fold-change) = -0.38, -0.28, -0.58, p <0.05) and T subsets (e.g. CD4+ T log(fold-change) = -0.21, -0.07, -0.15, p <0.05) varied greatly among strains. MYC/sg-p53 HCC induced a consistent expansion of liver Tregs and CD8+ T cells (p <0.05), but differential shifts of liver immune landscape were seen among strains. The flow cytometry data was supported by public scRNA-seq datasets matching C57BL/6 background. Further cross-species comparison in MASH condition confirmed shared changes of adaptive lymphocytes between mice and humans. In two MASH models, BALB/c or C57BL/6 mice were more consistent to recapture loss of CD4+ T or B cells, respectively (p <0.05). Conclusions Substantial liver immune differences exist among common mouse strains. Mice can recapitulate certain human liver immune changes with strain variations. Impact and implications Our immune cell profiling study revealed that the liver immune environment can be quite different among common mouse strains both under healthy and pathologic states, such as steatohepatitis or neoplastic processes. Our results serve as a data resource for studies investigating liver immunology and provide valuable insights for the design of studies on various immune cells in the livers of mice.
Collapse
Affiliation(s)
- Patrick Huang
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Francisco J. Rodriguez-Matos
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jonathan Qi
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Rajiv Trehan
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Yuta Myojin
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xiao Bin Zhu
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Tim F. Greten
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
- NCI CCR Liver Cancer Program, National Institutes of Health, Bethesda, MD, USA
| | - Chi Ma
- Gastrointestinal Malignancy Section, Thoracic and Gastrointestinal Malignancies Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
105
|
Wu J, Yang F, Huang G. Single-cell sequencing combined with bulk RNA seq reveals the roles of natural killer cell in prognosis and immunotherapy of hepatocellular carcinoma. Sci Rep 2025; 15:15314. [PMID: 40312525 PMCID: PMC12046010 DOI: 10.1038/s41598-025-99638-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: 07/19/2024] [Accepted: 04/22/2025] [Indexed: 05/03/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a type of highly heterogeneous tumor characterized by a high mortality rate and poor prognosis. Natural Killer cells (NK cells) are important immune cells that play an important role in anti-tumor activities, antiviral responses, and immune regulation. The relationship between NK cells and HCC remains unclear. It would be valuable to identify a NK-related prognostic signature for HCC. WGCNA and single-cell sequencing RNA were performed to identify NK cell related genes. Gene Enrichment Analysis were used to identify the potential signal pathway. After combing genes from WGCNA and scRNA, Unicox, LASSO + StepCox and Multicox analysis were used to filter prognostic-related gene and construct a prognostic model. Then we performed Proposed time analysis to identify the developmental trajectories of NK cells. Finally, ssGSEA and estimate methods were used to evaluate the immune microenvironment and sensitivity drugs. Using the scRNA-seq data, we identified 1396 genes with high NK cell scores. Based on the results of scRNA-seq, 250 NK-related genes were identified from WGCNA. We identified 223 intersecting genes between the scRNA-seq and WGCNA. After integrating clinical data with the bulk RNA-seq data of these intersecting genes, we constructed a prognostic model to accurately predict the prognosis of HCC patients. Eventually, we found that high-risk HCC patients exhibited worse survival outcomes and lower sensitivity to immunotherapy. We constructed a risk model based on NK cell-related genes that can predict the prognosis of HCC patients accurately. This model can also predict the immunotherapy response of HCC effectively.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/pathology
- Killer Cells, Natural/immunology
- Killer Cells, Natural/metabolism
- Liver Neoplasms/genetics
- Liver Neoplasms/therapy
- Liver Neoplasms/immunology
- Liver Neoplasms/mortality
- Liver Neoplasms/pathology
- Single-Cell Analysis/methods
- Prognosis
- Immunotherapy/methods
- RNA-Seq
- Gene Expression Regulation, Neoplastic
- Tumor Microenvironment/immunology
- Sequence Analysis, RNA
- Gene Expression Profiling
- Biomarkers, Tumor/genetics
- Male
Collapse
Affiliation(s)
- Jiahao Wu
- The Fifth Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
- Guangzhou Medical University, Guangzhou, China
| | - Fan Yang
- Hui Ya Hospital of The First Affiliated Hospital, Sun Yat-sen University, Huizhou, China
| | - Guanqun Huang
- Guangzhou Twelfth People's Hospital, Guangzhou, China.
| |
Collapse
|
106
|
Kohya R, Suda G, Ohara M, Hosoda S, Sho T, Chuma M, Komori A, Kugiyama Y, Yasui Y, Tsuchiya K, Kurosaki M, Tani J, Kaneko S, Nakagawa M, Asahina Y, Maekawa S, Enomoto N, Yamamoto Y, Baba M, Yamada R, Sasaki T, Yoda T, Yoshida S, Fu Q, Yang Z, Maehara O, Ohnishi S, Tokuchi Y, Kitagataya T, Kawagishi N, Nakai M, Natsuizaka M, Ogawa K, Sakamoto N. Serum FGF21 as a predictor of response to atezolizumab and bevacizumab in HCC. JHEP Rep 2025; 7:101364. [PMID: 40242310 PMCID: PMC11999275 DOI: 10.1016/j.jhepr.2025.101364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 02/01/2025] [Accepted: 02/13/2025] [Indexed: 04/18/2025] Open
Abstract
Background & Aims Fibroblast growth factor 21 (FGF21) is a crucial regulator of cell metabolism. Tumour-secreted FGF21 has shown immune-checkpoint factor functions, and high FGF21 levels are associated with a poor prognosis for patients. However, its prognostic value and impact on treatment response in patients with hepatocellular carcinoma (HCC) treated with immune-checkpoint inhibitors (ICIs) remain unclear. Thus, this study investigated the potential of high FGF21 levels as a prognostic marker and whether traditional ICI-based therapy can improve the prognosis of patients with high FGF21 levels. Methods In this retrospective multicentre study, patients with unresectable HCC who received atezolizumab/bevacizumab in the NORTE study group (n = 117) were classified into high (≥915 pg/ml; n = 29) and non-high (n = 88) FGF21 groups. For validation, we investigated patients treated with atezolizumab/bevacizumab in an independent cohort (n = 285). Overall survival, progression-free survival, and treatment response were compared between patients with and without high baseline FGF21 levels. Results The median overall survival (p <0.001) and progression-free survival (p = 0.045) were significantly shorter in the high FGF21 group than in the non-high FGF21 group. Independent cohort analysis validated these results. In the overall cohort, the median progression-free survival (5.75 vs. 8.84 months; p = 0.027) and median overall survival (14.13 vs. 22.08 months; p <0.001) were significantly shorter in the high FGF21 group than in the non-high FGF21 group. The durable response (≥6 months) + complete response rate was significantly decreased in the high FGF21 group (p = 0.045). No patient with a high FGF21 level achieved a complete response, whereas this was achieved in 4.1% (13/319) of patients with non-high FGF21 levels. Multivariate Cox regression analysis identified high baseline serum FGF21 as an independent poor prognostic factor for overall survival (hazard ratio 2.20, p <0.001). Conclusions Serum FGF21 may be a robust, non-invasive prognostic and treatment response marker for unresectable HCC treated with atezolizumab/bevacizumab. Impact and implications FGF21 has been reported to act as a secreted immune-checkpoint factor, and elevated levels of FGF21 are associated with a poor prognosis in patients with HCC. It is not fully understood whether ICIs can overcome the impact of high FGF21 levels on the shortened prognosis of patients with HCC. In this multicentre retrospective study, patients with HCC and high baseline levels of serum FGF21 who received atezolizumab/bevacizumab treatment exhibited a significantly shorter overall survival and shorter progression-free survival. These findings suggest serum FGF21 as a robust prognostic marker and an indicator of treatment response in unresectable HCC treated with ICI-based therapy. These findings could be crucial for the implementation of personalised treatment strategies for unresectable HCC. However, identifying optimal therapeutic options for patients with unresectable HCC and high serum FGF21 levels remains an urgent and critical clinical issue.
Collapse
Affiliation(s)
- Risako Kohya
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Goki Suda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masatsugu Ohara
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shunichi Hosoda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Takuya Sho
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Makoto Chuma
- Gastroenterology Centre, Yokohama City University Medical Centre, Minami-ku, Yokohama, Japan
| | - Atsumasa Komori
- Hepatology Division, NHO Nagasaki Medical Centre, Ōmura, Nagasaki, Japan
| | - Yuki Kugiyama
- Hepatology Division, NHO Nagasaki Medical Centre, Ōmura, Nagasaki, Japan
| | - Yutaka Yasui
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Musashino, Tokyo, Japan
| | - Kaoru Tsuchiya
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Musashino, Tokyo, Japan
| | - Masayuki Kurosaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Musashino, Tokyo, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, Takamatsu, Kagawa, Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
- Department of Liver Disease Control, Tokyo Medical and Dental University, Bunkyo-ku, Tokyo, Japan
| | - Shinya Maekawa
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Kofu City, Yamanashi, Japan
| | - Nobuyuki Enomoto
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Kofu City, Yamanashi, Japan
| | - Yoshiya Yamamoto
- Department of Gastroenterology and Hepatology, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Masaru Baba
- Centre for Gastroenterology and Hepatology, Japan Community Healthcare Organisation Hokkaido Hospital, Sapporo, Hokkaido, Japan
| | - Ren Yamada
- Kushiro Rosai Hospital, Kushiro, Hokkaido, Japan
| | - Takashi Sasaki
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Tomoka Yoda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Sonoe Yoshida
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Qingjie Fu
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Zijian Yang
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Osamu Maehara
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Shunsuke Ohnishi
- Laboratory of Molecular and Cellular Medicine, Faculty of Pharmaceutical Sciences, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Yoshimasa Tokuchi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
- Department of Gastroenterology and Hepatology, Hakodate Municipal Hospital, Hakodate, Hokkaido, Japan
| | - Takashi Kitagataya
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Naoki Kawagishi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Masato Nakai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Mitsuteru Natsuizaka
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Koji Ogawa
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| | - Naoya Sakamoto
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Hokkaido University, Sapporo, Hokkaido, Japan
| |
Collapse
|
107
|
Zhang L, Wang D, Zhang LZ, Yang WH, Yu C, Qin J, Feng LZ, Liu Z, Teng GJ. Pickering emulsion with tumor vascular destruction and microenvironment modulation for transarterial embolization therapy. Biomaterials 2025; 316:123018. [PMID: 39709852 DOI: 10.1016/j.biomaterials.2024.123018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 12/02/2024] [Accepted: 12/13/2024] [Indexed: 12/24/2024]
Abstract
In the clinic, Lipiodol chemotherapeutic emulsions remain a main choice for patients diagnosed with hepatocellular carcinoma (HCC) via the mini-invasive transarterial chemoembolization (TACE) therapy. However, the poor stability of conventional Lipiodol chemotherapeutic emulsions would result in the fast drug diffusion and incomplete embolization, inducing systemic toxicity and impairing the efficacy of TACE therapy. Therefore, it is of great importance to construct alternative formulations based on commercial Lipiodol to achieve the improved efficacy and safety of HCC treatment. Herein, calcium phosphate (CaP) nanoparticles-stabilized Lipiodol Pickering emulsion (CaP-LPE) with improved stability and pH-responsiveness is prepared and utilized for the encapsulation of combretastatin A4-phosphate (CA4P), a clinically approved vascular disrupting agent. The obtained CA4P-loaded CaP-LPE (CCaP-LPE) was shown to be enhanced stability compared to conventional Lipiodol emulsion and pH-responsive release of the encapsulated drugs. On one hand, the released CA4P could disrupt tumor vascular and cut off the blood supplying of tumor cells, thus starving cancer cells. Moreover, it was revealed that CCaP-LPE could reverse immunosuppressive tumor microenvironment (TME) by neutralizing tumor acidity, leading to the increased infiltration of CD8+ T cells and the decreased percentages of immunosuppressive cells. As the result, such CCaP-LPE could effectively shrink orthotopic N1S1 HCC tumors in rats by eliciting a potent antitumor immune response. Therefore, this study highlights a simple strategy to construct a novel LPE with the potencies of tumor vascular disruption and TME modulation, holding a great promise for TAE therapy of HCC.
Collapse
Affiliation(s)
- Lei Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Duo Wang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Lin-Zhu Zhang
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Wei-Hao Yang
- Department of Interventional Radiology, The First Affiliated Hospital of Soochow University, Suzhou, 215006, China
| | - Chao Yu
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Juan Qin
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China
| | - Liang-Zhu Feng
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China
| | - Zhuang Liu
- Institute of Functional Nano & Soft Materials (FUNSOM), Jiangsu Key Laboratory for Carbon-Based Functional Materials & Devices, Soochow University, Suzhou 215123, China.
| | - Gao-Jun Teng
- Center of Interventional Radiology and Vascular Surgery, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology (Southeast University), Department of Radiology, Zhongda Hospital, Medical School, Southeast University, 87 Dingjiaqiao Road, Nanjing, 210009, China; National Innovation Platform for Integration of Medical Engineering Education (NMEE) (Southeast University), Nanjing, 210009, China; Basic Medicine Research and Innovation Center of Ministry of Education, Zhongda Hospital, Southeast University, Nanjing, 210009, China; State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
108
|
Das S, Halder D, Jeyaprakash RS. Computational-guided approach for identification of PI3K alpha inhibitor in the treatment of hepatocellular carcinoma by virtual screening and water map analysis. J Biomol Struct Dyn 2025; 43:3886-3908. [PMID: 38197431 DOI: 10.1080/07391102.2023.2300131] [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: 10/16/2023] [Accepted: 12/22/2023] [Indexed: 01/11/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most deadly disorders, with a relative survival rate of 36% in the last 5 years. After an extensive literature survey and pathophysiology analysis, PI3Kα was found to be a promising biological target as PIK3CA gene upregulation was observed in HCC, resulting in the loss of apoptosis of cells, which leads to uncontrollable growth and proliferation. Due to superior selectivity and promising therapeutic activity, the PI3K-targeted molecule library was selected, and the ligand preparation was executed. The study mainly focused on e-pharmacophore development, virtual screening and receptor-ligand docking analysis. Then, MMGBSA and ADME prediction analysis was performed with the top 10 molecules; for further analysis of ligand-receptor binding affinity at the catalytic binding site, induced fit docking was performed with the top two molecules. The analysis of quantum chemical stability descriptors, i.e., frontier molecular orbital analysis, was performed followed by molecular dynamics simulation of 100 ns to better understand the ligand-receptor binding. In this study, water map analysis played a significant role in the hit optimization and analysis of the thermodynamic properties of the receptor-ligand complex. The two hit molecules K894-1435 and K894-1045 represented superior docking scores, enhanced stability, and inhibitory action targeting Valine 851 amino acid residue at the catalytic binding site. Hence, the study has significance for the quest for selective PI3Kα inhibitors through the process of hit-to-lead optimization.
Collapse
Affiliation(s)
- Subham Das
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - Debojyoti Halder
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| | - R S Jeyaprakash
- Department of Pharmaceutical Chemistry, Manipal College of Pharmaceutical Sciences, Manipal Academy of Higher Education, Manipal, Karnataka, India
| |
Collapse
|
109
|
Li Q, Chang X, Gu J, Yang Y, Ouyang J, Zhou Y, Zhao H, Zhou J. Adjuvant Transarterial Chemoembolization in Resected Macrotrabecular-massive Hepatocellular Carcinoma (ATAC-MACRO): A Multicenter Real-world Retrospective Study. Acad Radiol 2025; 32:2585-2595. [PMID: 39848885 DOI: 10.1016/j.acra.2024.12.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 12/08/2024] [Accepted: 12/22/2024] [Indexed: 01/25/2025]
Abstract
RATIONALE AND OBJECTIVES The purpose of this study was to demonstrate the impact of postoperative adjuvant transarterial chemoembolization (TACE) on the prognosis of patients with macrotrabecular-massive hepatocellular carcinoma (MTM-HCC). MATERIALS AND METHODS This retrospective study used the clinical records of patients with resected MTM-HCC with/without adjuvant TACE at three centers between January 2015 and December 2022. The primary end point was recurrence free survival (RFS). The secondary end points were overall survival (OS) and safety. RESULTS A total of 559 eligible patients were classified into the adjuvant TACE group and the observation group. After propensity score matching analysis, both RFS (HR 0.62 [95% CI, 0.48 to 0.80]; P < 0.001) and OS (HR 0.59 [95% CI, 0.42 to 0.84]; P = 0.013) in the adjuvant TACE group were significantly better than the observation group. By Cox regression models, mALBI grade, types of hepatectomy, number, satellite lesion, without adjuvant TACE were identified as independent risk factors for RFS, and mALBI grade, number, maximum tumor size, satellite lesion, microvascular invasion, high AFP level, without adjuvant TACE were identified as independent risk factors for OS. The incidence of surgery-related adverse events (AEs) had no significant difference between the two groups (P = 0.609). The majority of AEs associated with adjuvant TACE were grade I (84.4%), and no treatment-related deaths occurred in either group. CONCLUSIONS Adjuvant TACE significantly improved the RFS and OS of patients with resected MTM-HCC with acceptable toxicity.
Collapse
Affiliation(s)
- Qingjun Li
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Xu Chang
- Department of Interventional Therapy II, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China (X.C.)
| | - Jiaye Gu
- School of Ophthalmology & Optometry, Wenzhou Medical University, Wenzhou, Zhejiang, China (J.G.)
| | - Yi Yang
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.Y., H.Z.)
| | - Jingzhong Ouyang
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Yanzhao Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.)
| | - Hong Zhao
- Department of Liver Surgery, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China (Y.Y., H.Z.)
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Zhengzhou, Henan, China (Q.L., J.O., Y.Z., J.Z.).
| |
Collapse
|
110
|
Schulze K, Rose TD, Adlung L, Peschka M, Pagani F, Gorgulho J, Fründt TW, Labgaa I, Haber PK, Zimpel C, Castven D, Weinmann A, Garzia-Lezana T, Waldmann M, Renné T, Voß H, Moritz M, Orlikowski D, Schlüter H, Baumbach J, Schwartz M, Lohse AW, Huber S, Sangro B, Macias RI, Izquierdo-Sanchez L, Banales JM, Wege H, Marquardt JU, Villanueva A, Pauling JK, von Felden J. Metabolomic liquid biopsy dynamics predict early-stage HCC and actionable candidates of human hepatocarcinogenesis. JHEP Rep 2025; 7:101340. [PMID: 40290517 PMCID: PMC12023797 DOI: 10.1016/j.jhepr.2025.101340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Actionable candidates of hepatocarcinogenesis remain elusive, and tools for early detection are suboptimal. Our aim was to demonstrate that serum metabolome profiles reflect the initiation of hepatocellular carcinoma (HCC) and enable the identification of biomarkers for early HCC detection and actionable candidates for chemoprevention. Methods This global cohort study included 654 patients and 801 biospecimens. Following serum metabolome profiling across the spectrum of hepatocarcinogenesis, we conducted a phase II biomarker case-control study for early HCC detection. Findings were independently validated through in silico analysis, mRNA sequencing, and proteome profiling of primary HCC and non-tumoral tissue, and in vitro experiments. Results Aspartic acid, glutamic acid, taurine, and hypoxanthine were differentially abundant in the serum across chronic liver disease, cirrhosis, initial HCC, and progressed HCC, independent of sex, age, and etiology. In a phase II biomarker case-control study, a blood-based metabolite signature yielded an AUC of 94% to discriminate between patients with early-stage HCC and controls with cirrhosis, including independent validation. Unsupervised biclustering (MoSBi), lipid network analysis (LINEX2), and pathway enrichment analysis confirmed alterations in amino acid-, lipid-, and nucleotide-related pathways. In tumor tissue, these pathways were significantly deregulated regarding gene and protein expression in two independent datasets, including actionable targets RRM2, GMPS, BCAT1, PYCR2, and NEU1. In vitro knockdown confirmed a functional role in proliferation and migration, as exemplified for PYCR2. Conclusions These findings demonstrate that serum metabolome profiling indicates deregulated metabolites and pathways during hepatocarcinogenesis. Our liquid biopsy approach accurately detects early-stage HCC outperforming currently recommended surveillance tools and facilitates identification of actionable candidates for chemoprevention. Impact and implications Deregulated cellular metabolism is a hallmark of cancer. In smaller studies, circulating metabolite profiles have been associated with HCC, although mainly in the context of fatty liver disease. Translation strategies for primary prevention or early detection are lacking. In this global study, we present an unsupervised landscape of the altered serum metabolome profile during hepatocarcinogenesis, independent of age, sex, and etiology. We provide a blood-based metabolite signature that accurately identifies early-stage HCC in a phase II biomarker study including independent validation. Further RRM2, GMPS, BCAT1, PYCR2, and NEU1 are identified in tumor tissue as actionable candidates for prevention. Our data provide the rationale for clinical trials testing liquid biopsy metabolome-based signatures for early HCC detection and the development of chemoprevention strategies.
Collapse
Affiliation(s)
- Kornelius Schulze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lorenz Adlung
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), and Center for Biomedical AI (bAIome), Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Peschka
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Newborn Screening and Metabolic Laboratory, Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Pagani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg–Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben W. Fründt
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Ismail Labgaa
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Visceral Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philipp K. Haber
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Arndt Weinmann
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Teresa Garzia-Lezana
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hannah Voß
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Moritz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorian Orlikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Myron Schwartz
- Recanati Miller Transplant Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Ansgar W. Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Bruno Sangro
- Liver Unit, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
| | - Rocio I.R. Macias
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Henning Wege
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Augusto Villanueva
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josch Konstantin Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Johann von Felden
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| |
Collapse
|
111
|
Altaf A, Khalil M, Akabane M, Rashid Z, Kawashima J, Zindani S, Ruzzenente A, Ratti F, Marques H, Cauchy F, Lam V, Poultsides G, Aucejo F, Kitago M, Popescu I, Martel G, Gleisner A, Bauer TW, Hugh T, Bhimani N, Shen F, Endo I, Pawlik TM. Up-front resection for hepatocellular carcinoma: Assessing futility in the preoperative setting. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109594. [PMID: 39826445 DOI: 10.1016/j.ejso.2025.109594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 12/28/2024] [Accepted: 01/09/2025] [Indexed: 01/22/2025]
Abstract
OBJECTIVE We sought to develop a predictive model to preoperatively identify patients with hepatocellular carcinoma (HCC) at risk of undergoing futile upfront liver resection (LR). METHODS Patients undergoing curative-intent LR for HCC were identified from a large multi-institutional database. Futile LR was defined by death or disease recurrence within six months postoperatively. Backward logistic regression was performed to identify factors associated with futility. Additionally, binary criteria were established for surgical candidacy, aiming to keep the likelihood of futility below 20 %. RESULTS Among 1633 patients with HCC, 264 (16.2 %) underwent futile upfront LR. Tumor burden score (TBS) (coefficient: 0.083, 95%CI: 0.067-0.099), alpha-fetoprotein (AFP) (coefficient: 0.254, 95%CI: 0.195-0.310), and albumin-bilirubin (ALBI) grade 2/3 (coefficient: 0.566, 95%CI: 0.420-0.718) were independently associated with an increased risk of futile LR. The model demonstrated strong discrimination and calibration in both derivation and validation cohorts. Low, intermediate, and high-risk groups were determined based on the risk model, each with an escalating likelihood of futility, worse histological features, and worse survival outcomes. Six distinct conditions based on AFP-adjusted-to-TBS criteria were established, all with a futility likelihood of less than 20 %. Patients fulfilling these criteria had significantly better long-term recurrence-free and overall survival. The futility risk model was made available online for wide clinical applicability: (https://altaf-pawlik-hcc-futilityofsurgery-calculator.streamlit.app/). CONCLUSION A preoperative risk model and AFP-adjusted-to-TBS criteria were developed and validated to predict the likelihood of futile LR among patients with HCC. This pragmatic clinical tool may assist clinicians in preoperative decision-making, helping them avoid futile surgery unlikely to offer long-term benefits.
Collapse
Affiliation(s)
- Abdullah Altaf
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Mujtaba Khalil
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Miho Akabane
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Zayed Rashid
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Jun Kawashima
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | - Shahzaib Zindani
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States
| | | | | | - Hugo Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - François Cauchy
- Department of Hepatobiliopancreatic Surgery, APHP, Beaujon Hospital, Clichy, France
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Sydney, NSW, Australia
| | - George Poultsides
- Department of Surgery, Stanford University, Stanford, CA, United States
| | - Federico Aucejo
- Department of Surgery, Cleveland Clinic., Cleveland, OH, United States
| | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ana Gleisner
- Department of Surgery, University of Colorado, Aurora, CO, United States
| | - Todd W Bauer
- Department of Surgery, University of Virginia, Charlottesville, VA, United States
| | - Tom Hugh
- Department of Surgery, School of Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Nazim Bhimani
- Department of Surgery, School of Medicine, The University of Sydney, Sydney, NSW, Australia
| | - Feng Shen
- Department of Surgery, Eastern Hepatobiliary Surgery Hospital, Shanghai, China
| | - Itaru Endo
- Department of Surgery, Yokohama City University School of Medicine, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, United States.
| |
Collapse
|
112
|
Oh JM, Park Y, Lee J, Shen K. Microfabricated Organ-Specific Models of Tumor Microenvironments. Annu Rev Biomed Eng 2025; 27:307-333. [PMID: 40310890 DOI: 10.1146/annurev-bioeng-110222-103522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Despite the advances in detection, diagnosis, and treatments, cancer remains a lethal disease, claiming the lives of more than 600,000 people in the United States alone in 2024. To accelerate the development of new therapeutic strategies with improved responses, significant efforts have been made to develop microfabricated in vitro models of tumor microenvironments (TMEs) that address the limitations of animal-based cancer models. These models incorporate several advanced tissue engineering techniques to better reflect the organ- and patient-specific TMEs. Additionally, microfabricated models integrated with next-generation single-cell omics technologies provide unprecedented insights into patient's cellular and molecular heterogeneity and complexity. This review provides an overview of the recent understanding of cancer development and outlines the key TME elements that can be captured in microfabricated models to enhance their physiological relevance. We highlight the recent advances in microfabricated cancer models that reflect the unique characteristics of their organs of origin or sites of dissemination.
Collapse
Affiliation(s)
- Jeong Min Oh
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
| | - Yongkuk Park
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
| | - Jungwoo Lee
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts, USA;
- Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts, USA
- Molecular and Cellular Biology Graduate Program, University of Massachusetts, Amherst, Massachusetts, USA
| | - Keyue Shen
- Alfred E. Mann Department of Biomedical Engineering, University of Southern California, Los Angeles, California, USA;
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, USA
| |
Collapse
|
113
|
Yang M, Song X, Zhang F, Li M, Chang W, Wang Z, Li M, Shan H, Li D. Spatial proteomic landscape of primary and relapsed hepatocellular carcinoma reveals immune escape characteristics in early relapse. Hepatology 2025; 81:1452-1467. [PMID: 38900411 DOI: 10.1097/hep.0000000000000979] [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: 03/22/2024] [Accepted: 06/03/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS Surgical resection serves as the principal curative strategy for HCC, yet the incidence of postoperative recurrence remains alarmingly high. However, the spatial molecular structural alterations contributing to postoperative recurrence in HCC are still poorly understood. APPROACH AND RESULTS We employed imaging mass cytometry to profile the in situ expression of 33 proteins within 358,729 single cells of 92 clinically annotated surgical specimens from 46 patients who were treated with surgical resections for primary and relapsed tumors. We revealed the recurrence progression of HCC was governed by the dynamic spatial distribution and functional interplay of diverse cell types across adjacent normal, tumor margin, and intratumor regions. Our exhaustive analyses revealed an aggressive, immunosuppression-related spatial ecosystem in relapsed HCC. Additionally, we illustrated the prominent implications of the tumor microenvironment of tumor margins in association with relapse HCC. Moreover, we identified a novel subpopulation of dendritic cells (PDL1 + CD103 + DCs) enriched in the peritumoral area that correlated with early postoperative recurrence, which was further validated in an external cohort. Through the analysis of single-cell RNA sequencing data, we found the interaction of PDL1 + CD103 + DCs with regulatory T cells and exhausted T cells enhanced immunosuppression and immune escape through multiple ligand-receptor pathways. CONCLUSIONS We comprehensively depicted the spatial landscape of single-cell dynamics and multicellular architecture within primary and relapsed HCC. Our findings highlight spatial organization as a prominent determinant of HCC recurrence and provide valuable insight into the immune evasion mechanisms driving recurrence.
Collapse
Affiliation(s)
- Meilin Yang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Xiaoyi Song
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Fan Zhang
- Department of Head and Neck Oncology, Cancer Center, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
| | - Mingan Li
- Department of Interventional Radiology, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wuguang Chang
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Zheyan Wang
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| | - Man Li
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Department of Information Technology and Data Center, The Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
- Biobank of the Fifth Affiliated Hospital, Sun Yat-Sen University, Zhuhai, China
| | - Hong Shan
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
- Center for Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong Province, China
| | - Dan Li
- Department of Nuclear Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong Provincial Engineering Research Center of Molecular Imaging, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, Guangdong, China
- Guangdong-Hong Kong-Macao University Joint Laboratory of Interventional Medicine, The Fifth Affiliated Hospital, Sun Yat-sen University, Zhuhai, China
| |
Collapse
|
114
|
Qiu Y, Xu J, Liao W, Yang S, Wen Y, Farag MA, Zheng L, Zhao C. Ulvan derived from Ulva lactuca suppresses hepatocellular carcinoma cell proliferation through miR-542-3p-mediated downregulation of SLC35F6. Int J Biol Macromol 2025; 308:142252. [PMID: 40118430 DOI: 10.1016/j.ijbiomac.2025.142252] [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: 12/12/2024] [Revised: 03/13/2025] [Accepted: 03/16/2025] [Indexed: 03/23/2025]
Abstract
Hepatocellular carcinoma (HCC) therapy still presents significant challenges, with a critical need for novel molecular targets and effective natural compound-based therapies. Despite its known oncogenic potential in other cancers, the role of SLC35F6 in HCC has not been previously reported, leaving a gap in our understanding of its function and therapeutic relevance. Here, we demonstrate that SLC35F6 is overexpressed in HCC and is associated with poor prognosis. Ulva lactuca polysaccharide (ULP), a natural extract with known antitumor properties, exerts its effects by upregulating miR-542-3p, which in turn inhibits SLC35F6 expression and significantly increases TP53 protein levels. Furthermore, TP53 is positively regulated by miR-542-3p, and our results indicate that SLC35F6 is a target gene of miR-542-3p. Knockdown of SLC35F6 in H22 and HepG2 cells markedly reduced cell growth while elevating TP53 expression, supporting SLC35F6 as a key regulatory factor in the miR-542-3p/TP53 axis. While this study did not confirm direct mutual regulation between SLC35F6 and TP53, our findings provide evidence that targeting SLC35F6 can suppress HCC progression. Collectively, these results identify SLC35F6 as a potential therapeutic target for HCC and provide mechanistic insights into its regulation through the miR-542-3p/SLC35F6/TP53 axis.
Collapse
Affiliation(s)
- Yinghui Qiu
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; School of Materials Science and Engineering, Huaqiao University, Xiamen 361021, China
| | - Jingxiang Xu
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Wei Liao
- State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Food Science and Resources, China-Canada Joint Lab of Food Science and Technology, Key Laboratory of Bioactive Polysaccharides of Jiangxi Province, Nanchang University, Nanchang 330047, China
| | - Shuxin Yang
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yuxi Wen
- University of Vigo, Nutrition and Bromatology Group, Department of Analytical Chemistry and Food Science, Faculty of Science, E32004 Ourense, Spain
| | - Mohamed A Farag
- Pharmacognosy Department, College of Pharmacy, Cairo University, Cairo, Egypt
| | - Lingjun Zheng
- School of Agriculture and Biology, Shanghai JiaoTong University, Shanghai 200240, China
| | - Chao Zhao
- College of Marine Sciences, Fujian Agriculture and Forestry University, Fuzhou 350002, China; State Key Laboratory of Mariculture Breeding, Key Laboratory of Marine Biotechnology of Fujian Province, Fujian Agriculture and Forestry University, Fuzhou 350002, China.
| |
Collapse
|
115
|
Roberto M, Arrivi G, Delle Cave D. Editorial: Targeting the tumor microenvironment for effective treatment of gastrointestinal cancers. Front Endocrinol (Lausanne) 2025; 16:1600639. [PMID: 40370781 PMCID: PMC12074961 DOI: 10.3389/fendo.2025.1600639] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/26/2025] [Accepted: 04/02/2025] [Indexed: 05/16/2025] Open
Affiliation(s)
- Michela Roberto
- Oncology Unit, Policlinico Umberto I, Haematological, Oncological and Dermatological Department, Sapienza, University of Rome, Roma, Italy
| | - Giulia Arrivi
- Oncology Unit, Department of Clinical and Molecular Medicine Sapienza University of Rome, Sant’Andrea University Hospital, Rome, Italy
| | - Donatella Delle Cave
- Institute of Genetics and Biophysics ‘Adriano Buzzati-Traverso’ (IGB), National Research Council of Italy (CNR), Naples, Italy
| |
Collapse
|
116
|
Kawashima J, Akabane M, Khalil M, Woldesenbet S, Endo Y, Sahara K, Ruzzenente A, Ratti F, Marques HP, Oliveira S, Balaia J, Cauchy F, Lam V, Poultsides GA, Kitago M, Popescu I, Martel G, Gleisner A, Hugh T, Weiss M, Aucejo F, Aldrighetti L, Endo I, Pawlik TM. Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden (MELD-AFP-TBS) score to stratify prognosis after liver resection for hepatocellular carcinoma. Surgery 2025; 183:109388. [PMID: 40311416 DOI: 10.1016/j.surg.2025.109388] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Revised: 03/06/2025] [Accepted: 03/31/2025] [Indexed: 05/03/2025]
Abstract
INTRODUCTION Morphologic criteria, such as the Barcelona Clinic Liver Cancer staging system often fail to accurately predict long-term survival among patients undergoing liver resection for hepatocellular carcinoma. We sought to develop a continuous risk score that incorporates established markers of tumor biology and liver function to improve the prediction of overall survival. METHODS Data from a multi-institutional database were used to identify patients who underwent curative-intent hepatectomy for hepatocellular carcinoma. A predictive score for overall survival was developed using weighted beta-coefficients from a multivariable Cox regression model. RESULTS Among 850 patients, 595 (70.0%) were assigned to the training cohort, and 255 (30.0%) to the test cohort. In the training cohort, multivariable analysis identified the Model of End-Stage Liver Disease (hazard ratio, 1.04; 95% confidence interval, 1.01-1.07), log-transformed alpha-fetoprotein (hazard ratio, 1.07; 95% confidence interval, 1.02-1.13), and tumor burden score (hazard ratio, 1.07; 95% confidence interval, 1.03-1.11) as independent predictors of worse overall survival. The Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score, based on the Cox model, stratified patients into low-risk (n = 466, 78.3%) with a 5-year OS of 70.5% and high-risk (n = 129, 21.7%) with a 5-year OS of 47.0% (P < .001). In the test cohort, the Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score demonstrated superior discriminative accuracy (C-index: 0.72, time-dependent area under the curve 1-year: 0.80, 3-year 0.76, 5-year 0.70) compared with the Barcelona Clinic Liver Cancer staging system (C-index: 0.53, time-dependent area under the curve 1-year: 0.61, 3-year 0.55, 5-year 0.56). An online tool was made accessible at https://jk-osu.shinyapps.io/MELD_AFP_TBS/. CONCLUSION The Model of End-Stage Liver Disease-alpha-fetoprotein-tumor burden score provides a novel, accurate tool for prognostic stratification of patients with hepatocellular carcinoma, identifying high-risk patients who may benefit from alternative treatments to improve outcomes.
Collapse
Affiliation(s)
- Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH; Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Mujtaba Khalil
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Selamawit Woldesenbet
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH
| | - Yutaka Endo
- Department of Transplant Surgery, University of Rochester Medical Center, Rochester, NY
| | - Kota Sahara
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Andrea Ruzzenente
- Division of General and Hepatobiliary Surgery, University of Verona, Verona, Italy
| | | | - Hugo P Marques
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Sara Oliveira
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - Jorge Balaia
- Department of Surgery, Curry Cabral Hospital, Lisbon, Portugal
| | - François Cauchy
- Department of HPB Surgery and Liver Transplantation, Beaujon Hospital, Clichy, France
| | - Vincent Lam
- Department of Surgery, Westmead Hospital, Westmead, New South Wales, Australia
| | | | - Minoru Kitago
- Department of Surgery, Keio University, Tokyo, Japan
| | - Irinel Popescu
- Department of Surgery, Fundeni Clinical Institute, Bucharest, Romania
| | | | - Ana Gleisner
- Department of Surgery, University of Colorado Denver, Denver, CO
| | - Tom Hugh
- Department of Surgery, The University of Sydney, Sydney, New South Wales, Australia
| | - Matthew Weiss
- Department of Surgery, Cancer Institute, Northwell Health, New Hyde Park, NY
| | - Federico Aucejo
- Department of Hepato-pancreato-biliary & Liver Transplant Surgery, Cleveland Clinic Foundation, Digestive Diseases and Surgery Institute, Cleveland, OH
| | | | - Itaru Endo
- Department of Gastroenterological Surgery, Yokohama City University, Yokohama, Japan
| | - Timothy M Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH.
| |
Collapse
|
117
|
Xie J, Wang X. Multiple-omics analysis of aggrephagy-related cellular patterns and development of an aggrephagy-related signature for hepatocellular carcinoma. World J Surg Oncol 2025; 23:175. [PMID: 40307857 PMCID: PMC12044776 DOI: 10.1186/s12957-025-03816-z] [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: 02/12/2025] [Accepted: 04/15/2025] [Indexed: 05/02/2025] Open
Abstract
BACKGROUND Protein aggrephagy, a selected autophagy process response for degrading protein aggregates, plays a critical role in various cancers. However, its regulatory mechanisms and clinical implications in hepatocellular carcinoma (HCC) remain largely unexplored. METHODS We integrated bulk RNA-seq data from TCGA and single-cell RNA sequencing (scRNA-seq) data from GEO databases to systematically analyze aggrephagy-related genes (AGGRGs) in HCC. Prognostic aggrephagy-related genes (AGGRGs) were identified through univariate Cox and LASSO regression analyses, followed by the construction of a risk prediction model. Patients were stratified into high- and low-risk groups based on the median risk score. Comparative analyses were performed to assess clinical outcomes, pathway enrichment, and drug sensitivity. Independent risk factors were incorporated a nomogram using univariate and multivariate Cox regression. At the single-cell level, the AGG scores were calculated using AUCell algorithm, and cell interactions and pseudotime trajectory analyses were conducted. Finally, protein levels of key AGGRG was assessed via tissue microarray. RESULTS Eight AGGRGs (PFKP, TPX2, UBE2S, GOT2, ST6GALNAC4, ADAM15, G6PD, and KPNA2) were identified as prognostic markers for HCC. The high-risk group exhibited significantly worse survival outcomes, heightened drug resistance, and enrichment in cell cycle, mTORC1 signaling, and reactive oxygen species pathways. Single-cell transcriptomic analysis revealed 11 distinct cell types within the HCC tumor microenvironment (TME), including hepatocytes, T cells, NK cells, macrophages, monocytes, dendritic cells, plasma B cells, mature B cells, mast cells, endothelial cells, and fibroblasts. Hepatocytes exhibited the highest AGGRG scores and were associated with metabolic reprograming, proliferation, and immune evasion. Further subclustering of malignant hepatocytes using inferCNV revealed eight functionally heterogeneous subpopulations with extensive intercellular crosstalk. Trajectory analysis showed G6PD- and CCNB1-expressing subpopulations in early-to-intermediate differentiation states, whereas C3 and ARGs marked terminal differentiation. Notably, G6PD was predominantly expressed in early and mid-stages, while KPNA2, PFKP, and TPX2 were upregulated in advanced tumor states. Immunohistochemical (IHC) validation confirmed significant overexpression of G6PD in HCC tissues compared to adjacent normal tissues. CONCLUSION These findings provide a molecular framework for targeting aggrephagy pathways in HCC treatment strategies.
Collapse
Affiliation(s)
- Jiafen Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Xi Road, Yuexiu District, Guangzhou, 510120, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangzhou Medical University, 151 Yanjiang Xi Road, Yuexiu District, Guangzhou, 510120, China.
| |
Collapse
|
118
|
Yu X, Zhang Q, Wang L, Zhang Y, Zhu L. Engineered nanoparticles for imaging and targeted drug delivery in hepatocellular carcinoma. Exp Hematol Oncol 2025; 14:62. [PMID: 40307921 PMCID: PMC12044934 DOI: 10.1186/s40164-025-00658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Accepted: 04/18/2025] [Indexed: 05/02/2025] Open
Abstract
Liver cancer, notably hepatocellular carcinoma (HCC), poses a significant global health burden due to its high fatality rates. Conventional antitumor medications face challenges, including poor targeting, high toxicity, and drug resistance, leading to suboptimal clinical outcomes. This review focused on nanoparticle use in diagnosing and delivering medication for HCC, aiming to advance the development of nanomedicines for improved treatment outcomes. As an emerging frontier science and technology, nanotechnology has shown great potential, especially in precision medicine and personalized treatment. The success of nanosystems is attributable to their smaller size, biocompatibility, selective tumor accumulation, and lower toxicity. Nanoparticles, as a central part of nanotechnology innovation, have emerged in the field of medical diagnostics and therapeutics to overcome the various limitations of conventional chemotherapy, thus offering promising applications for improved selectivity, earlier and more precise diagnosis of cancers, personalized treatment, and overcoming drug resistance. Nanoparticles play a crucial role in drug delivery and imaging of HCC, with the body acting as a delivery system to target and deliver drugs or diagnostic reagents to specific organs or tissues, helping to accurately diagnose and target therapies while minimizing damage to healthy tissues. They protect drugs from early degradation and increase their biological half-life.
Collapse
Affiliation(s)
- Xianzhe Yu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
- Department of Gastrointestinal Surgery, Chengdu Second People's Hospital, No. 10 Qinyun Nan Street, Chengdu, 610041, Sichuan, People's Republic of China
| | - Qin Zhang
- Department of Postgraduate Students, West China School of Medicine/West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China
| | - Leibo Wang
- Department of Surgery, Beijing Jishuitan Hospital Guizhou Hospital Guiyang, Guiyang, 550000, Guizhou, The People's Republic of China
| | - Yan Zhang
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| | - Lingling Zhu
- Department of Medical Oncology, Cancer Center & Lung Cancer Center/Lung Cancer Institute, West China Hospital, Sichuan University, Chengdu, 610041, Sichuan, People's Republic of China.
| |
Collapse
|
119
|
Kah J, Staffeldt L, Volz T, Schulze K, Heumann A, Rövenstrunk G, Goebel M, Peine S, Dandri M, Lüth S. Classification of the LC4 Primarily-like Cell Line-Recapitulating a CDK4 Overexpressing Immune Evasive HIV-HCV-Induced HCC. Viruses 2025; 17:653. [PMID: 40431665 PMCID: PMC12115383 DOI: 10.3390/v17050653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/23/2025] [Accepted: 04/25/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality. HCC is characterized by high heterogeneity and, subsequently, adaptation by developing resistance to current treatments. Applying individualized models is crucial to understanding the potential of approved therapies. Therefore, we classify a primary-like cell line derived from the core region of an HCC with underlying HIV-HCV co-infection employing deep analysis on the pathway regulation level. METHODS We employed DEG analysis, followed by pathway analysis, to characterize the preservation level of the LC4 cells and the level of adoption. Next, we classify the new model for HCC research by employing healthy donor samples, commonly used HCC cell lines, and global RNAseq datasets. RESULTS LC4 cells reflect the characteristics of the parental cancer region, including immunosuppression and metabolic reprogramming, characterized by the downregulation of drug-metabolizing enzymes compared to healthy individuals, indicating a transition to alternate metabolic pathways. Moreover, we identified specific biomarkers equally regulated in the parental tissue, in global datasets of the same entities as well as in LC4 cells. CONCLUSIONS We classified LC4 cells as an individual immunosuppressive and highly progressive primary-like HCC cell line. LC4 cells are applicable as a model for preclinical drug testing, minimizing the lack of preclinical models in HCV-HIV-induced HCC research.
Collapse
Affiliation(s)
- Janine Kah
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- Department of Gastroenterology, Center for Translational Medicine, University Hospital Brandenburg, 14770 Brandenburg, Germany
| | - Lisa Staffeldt
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| | - Tassilo Volz
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Kornelius Schulze
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
| | - Asmus Heumann
- Department of General, Visceral and Thoracic Surgery, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany;
| | - Götz Rövenstrunk
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
- III. Department of Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Meike Goebel
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Sven Peine
- Institute for Transfusion Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany
| | - Maura Dandri
- Department of Internal Medicine, University Medical Center Hamburg-Eppendorf, 20246 Hamburg, Germany; (L.S.); (T.V.)
- German Center for Infection Research, Hamburg-Lübeck-Borstel Partner Site, 38124 Braunschweig, Germany
| | - Stefan Lüth
- Faculty of Health Sciences Brandenburg, Brandenburg Medical School Theodor Fontane, 16816 Neuruppin, Germany; (G.R.)
| |
Collapse
|
120
|
Xie L, Liu J, Yang Z, Chen H, Wang Y, Du X, Fu Y, Song P, Yu J. Microrobotic Swarms for Cancer Therapy. RESEARCH (WASHINGTON, D.C.) 2025; 8:0686. [PMID: 40302783 PMCID: PMC12038165 DOI: 10.34133/research.0686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Revised: 03/27/2025] [Accepted: 04/04/2025] [Indexed: 05/02/2025]
Abstract
Microrobotic swarms hold great promise for the revolution of cancer treatment. The coordination of miniaturized microrobots offers a unique approach to treating cancers at the cellular level with enhanced delivery efficiency and environmental adaptability. Prior studies have summarized the design, functionalization, and biomedical applications of microrobotic swarms. The strategies for actuation and motion control of swarms have also been introduced. In this review, we first give a detailed introduction to microrobot swarming. We then explore the design of microrobots and microrobotic swarms specifically engineered for cancer therapy, with a focus on tumor targeting, infiltration, and therapeutic efficacy. Moreover, the latest developments in active delivery methods and imaging techniques that enhance the precision of these systems are discussed. Finally, we categorize and analyze the various cancer therapies facilitated by functional microrobotic swarms, highlighting their potential to revolutionize treatment strategies for different cancer types.
Collapse
Affiliation(s)
- Leiming Xie
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Jinbo Liu
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Zhen Yang
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Hui Chen
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yibin Wang
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Xingzhou Du
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| | - Yongping Fu
- Department of Cardiovascular Medicine,
Affiliated Hospital of Shaoxing University, Shaoxing 312000, China
| | - Peng Song
- Department of Interventional Therapy, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital,
Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen 518116, China
| | - Jiangfan Yu
- Shenzhen Institute of Artificial Intelligence and Robotics for Society (AIRS), Shenzhen 518129, China
- School of Science and Engineering,
The Chinese University of Hong Kong, Shenzhen, Shenzhen 518172, China
| |
Collapse
|
121
|
Shen W, Li Z, Wang L, Liu Q, Zhang R, Yao Y, Zhao Z, Ji L. Tumor-resident Malassezia can promote hepatocellular carcinoma development by downregulating bile acid synthesis and modulating tumor microenvironment. Sci Rep 2025; 15:15020. [PMID: 40301518 PMCID: PMC12041395 DOI: 10.1038/s41598-025-99973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
Bacterial dysbiosis coincides with the carcinogenesis in malignancies such as lung and colon cancer, and has recently been suggested to involve in the pathogenesis of hepatocellular carcinoma (HCC). However, the mycobiome has not yet been definitively linked to liver tumorigenesis. Here we showed that the microbiota composition of HCC tumors was distinct from that of the normal adjacent to tumor (NAT) on the basis of richness and beta-diversity indices. Specifically, the fungal community that infiltrated HCC tumors was markedly enriched for Malassezia spp. and genus Malassezia in tumors was substantially more abundant than that in NAT. We also discovered that the relative abundance of genus Malassezia was strongly correlated with the tumor microenvironment (TME) signatures, including stromal and immune components. In addition, tumor-resident Malassezia could inhibit bile acid synthesis by downregulating the expression level of CYP7 A1 and CYP27 A1. To improve clinical usability, we developed a set of Malassezia-related genes, called Malassezia.Sig, which could accurately predict patient survival. Collectively, our work shows that tumor-resident Malasseiza may promote HCC progression by downregulating bile acid synthesis and modulating the TME, although more studies are needed.
Collapse
Affiliation(s)
- Weixi Shen
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zhihong Li
- Ganzi Tibetan Autonomous Prefecture People's Hospital, Tibet, 850002, China
| | - Lei Wang
- Women's Health Section, Harbin Red Cross Central Hospital, Harbin, 150076, China
| | - Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Renjie Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou, 571158, China
| | - Zhicheng Zhao
- The Fourth Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150018, China.
| | - Lei Ji
- Geneis Beijing Co., Ltd, Beijing, 100102, China.
| |
Collapse
|
122
|
Sökeland G, Brönnimann MP, Vassella E, Stirnimann G, Montani M, Friemel J. Clinically Uncertain Liver Masses: A Guide to Distinguishing Poorly Differentiated Primary Liver Cancer. Biomedicines 2025; 13:1063. [PMID: 40426891 PMCID: PMC12109020 DOI: 10.3390/biomedicines13051063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 04/07/2025] [Accepted: 04/09/2025] [Indexed: 05/29/2025] Open
Abstract
Objectives: The distinction of clinically uncertain, poorly differentiated liver masses into primary liver cancer (PLC) of cholangiocytic origin (intrahepatic cholangiocarcinoma; CCA) or hepatocellular origin (hepatocellular carcinoma; HCC) vs. metastasis is highly relevant to guiding patient treatment. Protocols differ in terms of resection, local ablation, liver transplantation, or systemic therapies with immune checkpoint inhibitors (ICIs) and tyrosine kinase inhibitors (TKIs). Methods: This retrospective case series exemplifies a multidisciplinary, practical guide to clinically uncertain liver masses using imaging, histomorphology, immune phenotyping, and mutational testing of telomerase promoter (TERT) combined with a literature review. Results: In 2/3 patients with uncertain liver masses and inconclusive immunohistochemistry profiles, TERT testing supported the diagnosis of poorly differentiated hepatocellular carcinoma. The third case with a history of sclerosing cholangitis and vague adenoid morphology yielded mutations in ARID1a and TP53 and was identified as primary liver cancer, consistent with poorly differentiated intrahepatic cholangiocarcinoma or mixed hepatocellular cholangiocarcinoma (cHCC/CCA). Conclusions: Finding HCC-typical TERT promoter mutations is a useful diagnostic tool in poorly differentiated primary liver cancer.
Collapse
Affiliation(s)
- Greta Sökeland
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
- Department of Neurology, Bern University Hospital, 3010 Bern, Switzerland
| | - Michael P. Brönnimann
- Department of Diagnostic, Interventional and Pediatric Radiology, Bern University Hospital, 3010 Bern, Switzerland
| | - Erik Vassella
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Guido Stirnimann
- Department of Hepatology, Inselspital, Bern University Hospital, 3010 Bern, Switzerland
| | - Matteo Montani
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
| | - Juliane Friemel
- Institute of Tissue Medicine and Pathology, University of Bern, 3008 Bern, Switzerland
- Institute of Forensic Medicine, University of Zürich, 8057 Zürich, Switzerland
| |
Collapse
|
123
|
Wang Y, Pan S, Tian J, Wang J, Yu Y, Wang S, Li F, Yang L, Liu X, Shen Y, Qiu Q, Luan J, Jia M, Xiong C, Duan X, Wang FS, Meng F. Cadonilimab, a PD-1/CTLA-4 bispecific antibody in unresectable hepatocellular carcinoma: a real-world study. Cancer Immunol Immunother 2025; 74:186. [PMID: 40293533 PMCID: PMC12037968 DOI: 10.1007/s00262-025-04038-8] [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: 11/29/2024] [Accepted: 03/26/2025] [Indexed: 04/30/2025]
Abstract
OBJECTIVE This study retrospectively evaluated the safety and efficacy of cadonilimab combined with tyrosine kinase inhibitors (TKI) for the treatment of unresectable hepatocellular carcinoma (uHCC). PATIENTS AND METHODS Seventy-eight patients who received cadonilimab + TKI were included; 42 and 36 received it as first-line (1 L) and second-line and above (≥ 2 L) systemic treatment, respectively. Besides, ninety-five patients who received PD-1 inhibitor + TKI as first-line treatments were included. Safety was the primary endpoint; secondary endpoints were overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and disease control rate (DCR). RESULTS Treatment-related adverse events (TRAEs) of any grade occurred in 84.6% of the patients, with grade ≥ 3 in 20.5%. In patients with a Child-Pugh score of ≥ 8 (CP ≥ 8), any grade TRAEs occurred in 88.2%, and grade ≥ 3 in 20.6%. The overall cohort's median progression-free survival (mPFS) was 3.6 months, whereas the median overall survival (mOS) was 8.8 months. In the 1 L group, mPFS was 6.7 months versus 2.3 months in ≥ 2 L. In the 1 L group, mOS was 13.7 months versus 3.2 months in ≥ 2 L. For CP < 8, 1 L mPFS was 7.6 months, mOS not reached; CP ≥ 8 had mPFS of 5.2 months, mOS of 5.6 months. For CP < 8 in ≥ 2 L, mPFS was 3.1 months, mOS 8.8 months; CP ≥ 8 had mPFS of 1.4 months, mOS of 2.2 months. After propensity score matching (PSM), the incidence of TRAEs of any grade was 77.1%, with grade ≥ 3 accounting for 17.1% in the PD-1 group. In the PD-1/CTLA-4 group, the incidence of TRAEs of any grade was 80.0%, and that of grade ≥ 3 TRAEs was 17.1%. The mPFS was 6.7 months in the PD-1/CTLA-4 group versus 3.3 months in the PD-1 group. The mOS was 13.7 months in the PD-1/CTLA-4 group versus 6.7 months in the PD-1 group. CONCLUSION Cadonilimab + TKI showed a favorable trend in safety and efficacy, especially when applied as first-line systemic therapy for uHCC. This study offers a clinical reference for its use in systemic uHCC therapy, particularly in patients with advanced liver dysfunction.
Collapse
Affiliation(s)
- Yilin Wang
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Shida Pan
- Capital Medical University, Beijing, 100069, China
| | - Jiahe Tian
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Jianing Wang
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Yingying Yu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, China
| | - Siyu Wang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Fengyi Li
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Luo Yang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Xiaomeng Liu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Yingjuan Shen
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Qin Qiu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Junqing Luan
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China
| | - Mengdie Jia
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
| | - Chuyue Xiong
- Peking University 302 Clinical Medical School, Beijing, 100191, China
| | - Xuanxuan Duan
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China
| | - Fu-Sheng Wang
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China.
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China.
| | - Fanping Meng
- Medical School of Chinese PLA, No. 28, Fuxing Road, Beijing, 100853, China.
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing, 100853, China.
| |
Collapse
|
124
|
Fang X, Cai Y, Zhao Z, Yang S, Li Z, Peng X, Hang M, Liu P, Li Y. Mechanisms of HRAS regulation of liver hepatocellular carcinoma for prognosis prediction. BMC Cancer 2025; 25:797. [PMID: 40295971 PMCID: PMC12039069 DOI: 10.1186/s12885-025-14131-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
BACKGROUND Liver hepatocellular carcinoma (LIHC) often has a poor prognosis. Since the relationship between HRas proto-oncogene, GTPase (HRAS) and LIHC has not been elucidated, the aim of this study was to explore the mechanisms by which HRAS is involved in regulating the prognosis of LIHC. METHODS We usedThe Cancer Genome Atlas (TCGA) database to characterize differences in HRAS gene expression between LIHC patients and healthy individuals. In addition, we analysed the relationships between HRAS gene expression levels and the clinicopathological characteristics of LIHC patients. Next, we used univariate and multivariate Cox regression analyses to identify prognostic factors. Differentially expressed genes were identified between the low- and high-expression groups, and KEGG and GO analyses and GSEA were performed to study the underlying mechanisms. The effects of high and low HRAS expression on the prognosis of LIHC patients was determined according to CIBERSORT. We subsequently assayed HRAS gene expression at the cellular level, and these data were validated in a tumour xenograft model. RESULTS We established and validated the HRAS gene as a prognostic signature and analysed the relationships between HRAS expression levels and clinicopathological features. Patients were categorized into high and low HRAS gene expression groups. We determined that high HRAS expression is associated with carbon metabolism, the PPAR signalling pathway, and small molecule catabolism in cancer. Furthermore, we conclude that the poor prognosis that results from elevated HRAS expression is associated with immune cell infiltration. We used LASSO + KNN to build an AI classification model that shows good performance in distinguishing liver cancer tissues form normal tissues. Finally, we verified that HRAS is highly expressed in hepatocellular carcinoma cells and promotes tumour growth. CONCLUSION We identified and validated the role of HRAS in hepatocellular carcinoma to assess hepatocellular carcinoma prognosis. The results of this study can be applied to predict survival, for personalized liver cancer treatment strategies, and provide information for the development of potential targeted therapies and new ideas for liver cancer patient treatment.
Collapse
Affiliation(s)
- Xingbao Fang
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Yan Cai
- Pathology Teaching and Research Office, Qujing Medical College, Qujing, Yunnan Province, 655000, China
| | - Zhuoyu Zhao
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Shaohua Yang
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Zhaojun Li
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Xiongbing Peng
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Meifang Hang
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China
| | - Peiwan Liu
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China.
| | - Yuehong Li
- Hepatobiliary Pancreatic Surgery, Kunming Medical University Affiliated Qujing Hospital (The First People's Hospital of Qujing City, Yunnan Province), Qujing, Yunnan Province, 655000, China.
| |
Collapse
|
125
|
Wang X, Liu D, Wang S, He R. An immune cell activation signature reflected hepatocellular carcinoma heterogeneity and predicted clinical outcomes. Front Immunol 2025; 16:1534611. [PMID: 40356904 PMCID: PMC12066757 DOI: 10.3389/fimmu.2025.1534611] [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] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 04/01/2025] [Indexed: 05/15/2025] Open
Abstract
Background The prognosis of hepatocellular carcinoma (HCC) remains challenging, and immune activation plays a critical role in cancer treatment. Identifying reliable immune activation-related prognostic markers is critical for predicting HCC patient outcomes. Method A six-gene signature was developed. The prognostic value was assessed by correlating the signature and survival. The robustness of the signature was validated in three independent Gene Expression Omnibus (GEO) datasets. Associations with clinical, genomic, and transcriptomic features were also evaluated. Additionally, single-cell sequencing data were analyzed to explore cell-cell interaction heterogeneity reflected by the signature. The biological role of candidate gene RORC was investigated, including chemotherapy resistance and detailed regulatory mechanism in affecting progression. The clinical potential role of RORC and its downstream gene was also evaluated by immunohistochemical (IHC) microarray. Results The six-gene signature stratified patients into high-risk and low-risk groups, with high-risk samples exhibiting significantly shorter overall survival (median: 23.8 months, 95% CI: 20.6-41.8) than low-risk samples (median: 83.2 months, 95% CI: 69.6-NA, p < 0.001). Validation in independent GEO datasets confirmed the robustness of the signature. The signature was significantly associated with the pathological stage and negatively correlated with PD-L1 expression, outperforming clinical indicators in predicting 3-year survival. The signature was significantly associated with TP53 mutations, genomic stability, and canonical cancer-related pathways. Single-cell sequencing data indicated that the signature revealed cell-cell interaction heterogeneity in HCC. Candidate gene RORC promotes proliferation and migration by regulating CDC6 gene expression as a transcription factor. Furthermore, RORC is also associated with multiple drug resistance, especially docetaxel and paclitaxel. IHC revealed that RORC and candidate gene CDC6 were valuable predictive biomarkers for prognosis. Conclusion The six-gene signature provides valuable insights into the biological status of HCC patients and is a robust tool for clinical application.
Collapse
Affiliation(s)
- Xiaofeng Wang
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, ;China
| | - Dongli Liu
- Department of Radiation Oncology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, ;China
| | - Shuai Wang
- Department of Radiation, Chushi Orthopedic Hospital, Zhengzhou, Henan, ;China
| | - Rui He
- Department of Medical Oncology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, ;China
| |
Collapse
|
126
|
Tu L, Luo J, Yin Y, Yu H. Bioinformatics analysis across pan-cancer and experimental validation in hepatocellular carcinoma revealed the oncogenic role of SF3B6. Front Pharmacol 2025; 16:1516534. [PMID: 40356980 PMCID: PMC12067594 DOI: 10.3389/fphar.2025.1516534] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2025] [Accepted: 04/15/2025] [Indexed: 05/15/2025] Open
Abstract
Background Splicing factor 3b subunit 6(SF3B6), a subunit of the SF3B complex, regulates the process of RNA splicing by recognizing the branch point adenosine in pre-mRNA and facilitating the interaction between U2 snRNA and the branch point sequence. Currently, there is no systematic multi-omics study exploring the diagnostic, prognostic, and immunotherapy predictive value of SF3B6 in pan-cancer, nor is its role in hepatocellular carcinoma (HCC) clear. Methods We utilized various databases to systematically examine the expression and genetic variation of SF3B6 across multiple cancer types, assessing its relationship with diagnosis, prognosis, immune infiltration, immunotherapy response, and associated signaling pathways. Additionally, we investigated the correlation between SF3B6 and prognosis, clinicopathological features, and treatment responses in HCC, as well as the roles of its related alternative splicing isoforms. Finally, we conducted in vitro experiments to validate the effects of SF3B6 on the proliferation, migration, invasion, apoptosis, and cell cycle progression of liver cancer cells. Results Results indicate that SF3B6 was highly expressed in various cancers and regulated by copy number variations and DNA methylation. The elevated expression of SF3B6 demonstrated predictive value for cancer diagnosis, prognosis, and responses to immunotherapy. Functional enrichment analysis suggests that SF3B6 was closely associated with pathways related to tumor immunity, tumor metabolism, and cell cycle. Additionally, high SF3B6 expression was an independent risk factor for overall survival and correlated with poor alpha-fetoprotein levels, pathological grading, clinical staging, and reduced responses to sorafenib and transarterial chemoembolization treatment in HCC. Interestingly, SF3B6 was associated with variant splicing isotypes of genes involved in the G2M checkpoint and DNA repair pathways, including NEIL3, NEK2, KIF4A, TROAP, and FANCD2. Moreover, SF3B6 was highly expressed in liver cancer cells, promoting the proliferation, migration, and invasion of cancer cells, inhibiting apoptosis, and regulating the transition from the S phase to the G2M phase of the cell cycle. Conclusion We emphasize that SF3B6 has the potential to serve as a biomarker for predicting cancer diagnosis, prognosis, and immunotherapy responses, especially in HCC. SF3B6 and its related alternative splicing isoforms promote the occurrence and progression of HCC and may serve as potential therapeutic targets.
Collapse
Affiliation(s)
- Linshuang Tu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jiefu Luo
- Department of Hepatobiliary Surgery, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Ya Yin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Huihong Yu
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
127
|
Li L, Dong J, Xu C, Wang S. Lactate drives senescence-resistant lineages in hepatocellular carcinoma via histone H2B lactylation of NDRG1. Cancer Lett 2025; 616:217567. [PMID: 39978571 DOI: 10.1016/j.canlet.2025.217567] [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: 01/05/2025] [Revised: 02/11/2025] [Accepted: 02/15/2025] [Indexed: 02/22/2025]
Abstract
Hepatocellular carcinoma (HCC) treatment options remain limited despite advances in targeted therapies for molecularly-defined cancers. To address tumor heterogeneity, we reconstructed HCC clonal evolution through single-cell RNA sequencing trajectory analysis, identifying 902 signature genes across seven cellular states. Weighted gene co-expression network analysis of public HCC datasets revealed tumor-grade-associated modules and established a 14-gene prognostic model linked to clonal evolution. Central to this model is the LDHA-NDRG1 axis - two hypoxia-responsive regulators showing coordinated spatiotemporal expression patterns during cancer progression. Dual-expressing cell lineages correlated with poor prognosis and senescence resistance through LDHA-mediated lactylation of histone H2B at K58 on NDRG1, an epigenetic mechanism connecting metabolic reprogramming to senescence evasion. Therapeutically, dual inhibition of this axis extended survival in metastatic HCC murine models. Our findings reveal that lactate-driven epigenetic modification via the LDHA-NDRG1 axis creates a molecularly distinct subpopulation enabling senescence resistance, providing mechanistic insights into HCC heterogeneity. This work proposes a precision medicine strategy targeting lactylation-mediated epigenetic regulation, with implications for developing combination therapies and patient stratification based on clonal evolution patterns.
Collapse
Affiliation(s)
- Lu Li
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Department of Nephrology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310022, China
| | - Jinyun Dong
- Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| | - Chunwei Xu
- Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310000, China.
| | - Shiqun Wang
- Cancer Institute, Key Laboratory of Cancer Prevention and Intervention, Ministry of Education, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310009, China; Center for Innovative Drug Research, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, China.
| |
Collapse
|
128
|
Sequeira LM, Ozturk NB, Sierra L, Gurakar M, Toruner MD, Zheng M, Simsek C, Gurakar A, Kim AK. Hepatocellular Carcinoma and the Role of Liver Transplantation: An Update and Review. J Clin Transl Hepatol 2025; 13:327-338. [PMID: 40206277 PMCID: PMC11976436 DOI: 10.14218/jcth.2024.00432] [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: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. Multiple treatment modalities are available for the management of HCC, depending on its stage as determined by the Barcelona Clinic Liver Cancer staging system. Because liver transplantation (LT) theoretically removes the cancer and replaces the organ at risk for future malignancy, LT is often considered the most definitive and one of the most efficacious treatment options for HCC. Nevertheless, the success and efficacy of liver transplantation depend on various tumor characteristics. As a result, multiple criteria have been developed to assess the appropriateness of a case of HCC for LT, with the pioneering Milan Criteria established in 1996. Over the past 20 to 30 years, these criteria have been critically evaluated, expanded, and often liberalized to make LT for patients with HCC a more universally applicable option. Furthermore, the development of other treatment modalities has enabled downstaging and bridging strategies for HCC prior to LT. In this narrative and comprehensive review, we provided an update on recent trends in the epidemiology of HCC, selection criteria for LT, implementation of LT across different regions, treatment modalities available as bridges, downstaging strategies, alternatives to LT, and, finally, post-LT surveillance.
Collapse
Affiliation(s)
- Lynette M. Sequeira
- Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - N. Begum Ozturk
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Leandro Sierra
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Merve Gurakar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Melanie Zheng
- Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cem Simsek
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmet Gurakar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy K. Kim
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
129
|
Annadurai P, Isaac AE. Prediction of key pathways in hepatocellular carcinoma (HCC): A machine learning approach using a sample pathway information matrix. Comput Biol Chem 2025; 118:108481. [PMID: 40300216 DOI: 10.1016/j.compbiolchem.2025.108481] [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: 12/05/2024] [Revised: 03/20/2025] [Accepted: 04/16/2025] [Indexed: 05/01/2025]
Abstract
Hepatocellular carcinoma (HCC) represents the most prevalent form of primary liver cancer, accounting for 75 % of all cases. Individuals with metabolic dysfunctions are at risk of developing significant symptoms, including cirrhosis. To address this, we proposed a novel method to find the signalling pathway based on the patient's gene expression. The objective includes examining the predictive biomarkers associated with cirrhosis-related HCC. The study combines gene expression and pathway enrichment data to find the biologically important pathways in disease progression. The differential gene expression analysis showed 58 upregulated and 62 downregulating differentially expressed genes. These DEGs were utilized to construct a protein-protein interaction network, and then the clustered genes were determined. Subsequently, pathway enrichment analysis was performed for the clustered genes and the gene-pathway interaction matrix was developed. The sample-pathway information matrix (SPIM) was obtained by multiplying the gene-expression and gene-pathway matrix. The key pathways were predicted from the SPIM using random forest model and we achieved 94 % of accuracy. The arachidonic acid metabolism was the most important pathway and genes involved in this pathway includes CYP2C9, CYP2C8, CYP2B6. These genes are well known for promoting metabolic disorders in the liver. Hence, our novel method proves that it could distinguish the samples and extract important pathways that are involved in differentiating the diseased samples based on the gene expression. Therefore, integrating gene expression and their enriched biological pathways may effectively help in identifying the key signalling pathways.
Collapse
Affiliation(s)
- Priyadharshini Annadurai
- Bioinformatics Programming Laboratory, Department of Bioscience, School of Bio Science and Technology, Vellore Institute of Technology, Katpadi, Vellore - 632014, Tamil Nadu, India
| | - Arnold Emerson Isaac
- Bioinformatics Programming Laboratory, Department of Bioscience, School of Bio Science and Technology, Vellore Institute of Technology, Katpadi, Vellore - 632014, Tamil Nadu, India.
| |
Collapse
|
130
|
Elsabaawy M, Badran H, Ragab A, Abdelhafiz R, Nageeb M, Ashour R. ALBI-sarcopenia score as a predictor of treatment outcomes in hepatocellular carcinoma. Sci Rep 2025; 15:14621. [PMID: 40287454 PMCID: PMC12033259 DOI: 10.1038/s41598-025-97295-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2024] [Accepted: 04/03/2025] [Indexed: 04/29/2025] Open
Abstract
The recently developed ALBI-Sarcopenia score has demonstrated effectiveness in predicting mortality in hepatocellular carcinoma (HCC), emerging as a crucial factor in guiding treatment decisions. To assess the utility of the ALBI-Sarcopenia score in predicting the success of HCC treatment. A prospective study involving 262 liver cirrhosis with HCC patients were assigned to various treatment strategies according to Barcelona clinics of liver disease (BCLC) classification. Patients were followed up for 12 months reporting laboratory data, sarcopenia, ALBI-Sarcopenia score, and outcomes. Sarcopenia was prevalent in 43.1% (48.35% males and 31.25% females, P = 0.042). Most patients were HCV-positive (88.9%) and classified as CTP A (55.7%) or BCLC B (54.2%). Over the study period, TACE was the most administered treatment (41.2% at baseline), followed by a progressive shift toward best supportive care as disease severity increased. Complete response rates declined from 31.7% at 1 month to 21.4% at 12 months, while progressive disease rates increased from 21.8 to 37.8% over the same period. At 12 months, the ALBI-Sarcopenia score demonstrated the highest predictive accuracy for treatment response (AUC:0.69, p = 0.001), outperforming both the ALBI (AUC: 0.631, p = 0.001) and MELD (AUC:0.623, p = 0.003) scores. Logistic regression identified ALBI-Sarcopenia as a significant independent predictor of response at 1 month (OR:1.25, 95% CI:0.881-1.971, p = 0.002) and 12 months (OR:2.189, 95% CI:0.992-4.937, p = 0.001). The ALBI-Sarcopenia score is a robust predictor of treatment outcomes in HCC, offering superior prognostic accuracy compared to traditional scoring systems, and enhancing patient stratification for optimized treatment planning.
Collapse
Affiliation(s)
- Maha Elsabaawy
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt.
| | - Hanaa Badran
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt
| | - Amr Ragab
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt
| | - Rasha Abdelhafiz
- Radiodiagnosis and Interventional Radiology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt
| | - Madiha Nageeb
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt
| | - Reham Ashour
- Hepatology and Gastroenterology Department, National Liver Institute, Menoufia University, Shebeen El-Koom, Egypt
| |
Collapse
|
131
|
Lin J, Zhou J, Ye K, Xie F. Prunella vulgaris: A potential molecule for the treatment of hepatocellular carcinoma. Medicine (Baltimore) 2025; 104:e42267. [PMID: 40295240 PMCID: PMC12040002 DOI: 10.1097/md.0000000000042267] [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: 11/14/2024] [Revised: 03/03/2025] [Accepted: 03/25/2025] [Indexed: 04/30/2025] Open
Abstract
Prunella vulgaris (PV) is widely used in treating various diseases, but its relationship with hepatocellular carcinoma (HCC) remains unclear. This study systematically evaluates PV's therapeutic potential in HCC and explores its molecular mechanisms. Active compounds and molecular targets of PV were obtained from the Traditional Chinese Medicine Systems Pharmacology Database and Analysis Platform database, and HCC-related targets were identified using the Gene Expression Omnibus database. A drug-disease target network was built to identify key hub genes, which were further investigated through immune analysis, single-cell RNA sequencing, molecular docking, and in vitro experiments. We identified 185 drug targets and 635 HCC-related targets, with 15 potential PV targets linked to HCC progression. In vitro validation confirmed significant expression of these targets in HCC cells. Mechanistic analysis indicated that these hub genes may influence HCC progression through pathways like tumor protein 53 signaling and are associated with immune cell subsets, including CD8+ T cells and natural killer cells. This study identifies key bioactive components of PV for HCC treatment and reveals their molecular mechanisms. Dysregulation of these targets correlates with HCC pathogenesis, suggesting their potential as novel biomarkers. Future research will focus on further validation in vitro and in vivo to explore the clinical applicability of these targets and the synergistic potential of PV in combination with other treatments.
Collapse
Affiliation(s)
- Jie Lin
- Department of Hepatobiliary and Pancreatic Surgery, The Neijiang First People’s Hospital, Neijiang, Sichuan, China
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, Jilin, China
| | - Jiale Zhou
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Medical College, Chengdu, Sichuan, China
| | - Kailin Ye
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Medical College, Chengdu, Sichuan, China
| | - Fei Xie
- Department of Hepatobiliary and Pancreatic Surgery, The Neijiang First People’s Hospital, Neijiang, Sichuan, China
| |
Collapse
|
132
|
Hochnadel I, Hoenicke L, Petriv N, Suo H, Groebe L, Olijnik C, Bondarenko N, Alfonso JC, Jarek M, Shi R, Jeron A, Timrott K, Hirsch T, Jedicke N, Bruder D, Klawonn F, Lichtinghagen R, Geffers R, Lenzen H, Manns MP, Yevsa T. In vivo RNAi screen and validation reveals Ngp, Hba-a1, and S100a8 as novel inhibitory targets on T lymphocytes in liver cancer. Front Immunol 2025; 16:1549229. [PMID: 40352930 PMCID: PMC12061932 DOI: 10.3389/fimmu.2025.1549229] [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] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Accepted: 03/21/2025] [Indexed: 05/14/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) represents the third deadliest cancer worldwide with limited treatment options. Immune checkpoint inhibitors (ICIs) have revolutionized HCC therapy, but immune suppression within the tumor microenvironment remains a major challenge. Therefore, in this study, we aimed to define novel ICI molecules arising on T cells during aggressive HCC development. Methods Using autochthonous HCC models, we performed microarray analyses followed by in vivo RNA interference screen and identified several new ICI molecules on CD4 and CD8 T lymphocytes in HCC-bearing mice. Short hairpin RNA (shRNA)-mediated knockdown of the ICI molecules was performed to validate their functional role in T cell activity and survival of HCC-bearing mice. Finally, we searched for the presence of the defined ICI molecules in HCC patients. Results We identified neutrophilic granule protein (Ngp), hemoglobin subunit alpha-1 (Hba-a1), and S100 calcium-binding protein a8 (S100a8) as novel inhibitory molecules of T cells in HCC. The specific shRNA-based knockdown of these inhibitory targets was safe, led to a downregulation of classical ICI molecules (PD-1, PD-L1, 4-1BBL, CD160), and kept liver parameters under control in murine HCC. Besides, we detected upregulation of S100A8 and S100A9 in blood and liver tissues in HCC patients, supporting their clinical relevance. Conclusion The obtained results pave the way for the use of the newly defined ICI molecules Ngp, Hba-a1, and S100a8 as novel immunotherapeutic targets in further preclinical and clinical studies in HCC patients.
Collapse
Affiliation(s)
- Inga Hochnadel
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lisa Hoenicke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Nataliia Petriv
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Huizhen Suo
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Lothar Groebe
- Experimental Immunology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Chantal Olijnik
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Nina Bondarenko
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Department of Pathological Anatomy, Forensic Medicine and Pathological Physiology, Dnipro State Medical University, Dnipro, Ukraine
| | - Juan C. Alfonso
- Department of Systems Immunology, Technical University Braunschweig and HZI, Braunschweig, Germany
| | | | - Ruibing Shi
- Biostatistics Research Group, HZI, Braunschweig, Germany
| | - Andreas Jeron
- Immune Regulation Group, HZI, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Kai Timrott
- Department of General, Visceral and Transplant Surgery, MHH, Hannover, Germany
| | | | - Nils Jedicke
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Dunja Bruder
- Immune Regulation Group, HZI, Braunschweig, Germany
- Infection Immunology Group, Institute of Medical Microbiology and Hospital Hygiene, Otto-von-Guericke University Magdeburg, Magdeburg, Germany
| | - Frank Klawonn
- Biostatistics Research Group, HZI, Braunschweig, Germany
- Munich Biomarker Research Center, Institute of Laboratory Medicine, German Heart Center, Technical University of Munich, Munich, Germany
- Department of Computer Science, Ostfalia University, Wolfenbüttel, Germany
| | | | | | - Henrike Lenzen
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
- Department of Gastroenterology, Hepatology, Interventional Endoscopy and Diabetology, Academic Teaching Hospital Braunschweig, Braunschweig, Germany
| | - Michael P. Manns
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| | - Tetyana Yevsa
- Department of Gastroenterology, Hepatology, Infectious Diseases and Endocrinology, Hannover Medical School (MHH), Hannover, Germany
| |
Collapse
|
133
|
Yuan G, Luo X, He K, Tan Y, Luo C, Liu B, Sun Y, Liu J. Intratumoral self-assembly of renal-clearable gold nanoparticles as precise photothermal nanomedicine for liver tumor therapy. SCIENCE ADVANCES 2025; 11:eadw7032. [PMID: 40267199 PMCID: PMC12017308 DOI: 10.1126/sciadv.adw7032] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Accepted: 03/19/2025] [Indexed: 04/25/2025]
Abstract
Noninvasive photothermal therapy (PTT) for cancer with photothermal agents (PTAs) has recently achieved success in both preclinical and clinical trials. However, traditional PTAs tend to nonspecifically accumulate in normal liver tissue, hampering their use in PTT of liver tumors. By taking advantage of extremely low liver accumulation from ultrasmall renal-clearable gold nanoparticles (AuNPs), we report a biosafe therapeutic PTT strategy to treat liver tumors precisely through the intratumoral self-assembly of renal-clearable AuNPs at the tumor site via host-guest interactions. After active tumor targeting from the host AuNPs functionalized with both cyclo (Arg-Gly-Asp-d-Phe-Cys) and cyclodextrin, the guest AuNPs functionalized with both pH-responsive doxorubicin and adamantane are designed to precisely trigger intratumoral self-assembly, enhancing both PTT and chemotherapy toward the liver tumor microenvironment. This smart design principle generates a precise therapeutic action toward liver tumors without causing any noticeable heating effect or damage to the surrounding normal liver tissue.
Collapse
Affiliation(s)
- Gangqiang Yuan
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Xiaoxi Luo
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Kui He
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yue Tan
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Caiming Luo
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Ben Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Yidan Sun
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
| | - Jinbin Liu
- Key Laboratory of Functional Molecular Engineering of Guangdong Province, School of Chemistry and Chemical Engineering, South China University of Technology, Guangzhou 510640, China
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou 510640, China
| |
Collapse
|
134
|
Wang W, Gao X, Niu W, Yin J, He K. Targeting Metabolism: Innovative Therapies for MASLD Unveiled. Int J Mol Sci 2025; 26:4077. [PMID: 40362316 PMCID: PMC12071536 DOI: 10.3390/ijms26094077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2025] [Revised: 04/01/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
The recent introduction of the term metabolic-dysfunction-associated steatotic liver disease (MASLD) has highlighted the critical role of metabolism in the disease's pathophysiology. This innovative nomenclature signifies a shift from the previous designation of non-alcoholic fatty liver disease (NAFLD), emphasizing the condition's progressive nature. Simultaneously, MASLD has become one of the most prevalent liver diseases worldwide, highlighting the urgent need for research to elucidate its etiology and develop effective treatment strategies. This review examines and delineates the revised definition of MASLD, exploring its epidemiology and the pathological changes occurring at various stages of the disease. Additionally, it identifies metabolically relevant targets within MASLD and provides a summary of the latest metabolically targeted drugs under development, including those in clinical and some preclinical stages. The review finishes with a look ahead to the future of targeted therapy for MASLD, with the goal of summarizing and providing fresh ideas and insights.
Collapse
Affiliation(s)
- Weixin Wang
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Xin Gao
- School of Public Health, Jilin University, Changchun 130021, China;
| | - Wentong Niu
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| | - Jinping Yin
- NHC Key Laboratory of Radiobiology, School of Public Health, Jilin University, Changchun 130041, China;
| | - Kan He
- Department of Pharmacology, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; (W.W.); (W.N.)
| |
Collapse
|
135
|
Dang H, Lucci A, Barry A, Johnson V, Ruiz B, Reynolds L, Wojnar A, Chewe K, Pomyen Y, Eckert C, Zhang K, Hill J, Shah A, Bodzin A, Grabocka E, Gaida M, Fawzi N. Phase separation of NELFE modulates chromatin accessibility to promote dichotomous signaling pathways in hepatocellular carcinoma. RESEARCH SQUARE 2025:rs.3.rs-5843408. [PMID: 40313774 PMCID: PMC12045374 DOI: 10.21203/rs.3.rs-5843408/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Abstract
Biomolecular condensates partition various cellular processes including transcription, DNA repair, and RNA metabolism. We report NELFE, a member of the Negative Elongation Factor complex required for Polymerase II (Pol II) pausing, forms distinct foci mediated by two low complexity sequences. We show NELFE is oncogenic in hepatocellular carcinoma (HCC) by undergoing liquid-liquid phase separation (LLPS) with SMARCB1 to modulate chromatin accessibility to downregulate pro-apoptotic genes through Pol II pausing while activating pro-growth signals to promote HCC progression. Our work highlights the importance of NELFE LLPS as a mechanism of chromatin accessibility to regulate both paused and non-paused genes to drive tumorigenesis in hepatocellular carcinoma.
Collapse
|
136
|
Li L, Xu L, Liao W, Wang P, Xu M, Li B, Zhang M. circCEP70 encoded protein inhibits the progression of hepatocellular carcinoma. Cell Mol Life Sci 2025; 82:174. [PMID: 40272569 PMCID: PMC12022199 DOI: 10.1007/s00018-025-05651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/14/2025] [Accepted: 03/06/2025] [Indexed: 04/25/2025]
Abstract
Cirrhosis is closely related to hepatocellular carcinoma (HCC), however, the regulation of circular RNA (circRNA) in HCC with cirrhotic background has not yet been well illustrated. In this study, high throughput circRNA sequencing was applied to identified candidate circRNAs in HCC samples with cirrhotic background. The biological function of candidate circRNA was validated in both in vitro and in vivo settings. Additionally, Alphafold 3, mass spectrometry analysis and immunofluorescence were employed to investigate the underlying mechanisms involved. We found circCEP70 exhibited significantly higher expression levels in cirrhotic HCC samples and showed a positive correlation with improved prognosis. The RNA binding protein U2AF2 was found to suppress the expression of circCEP70 in cirrhosis patients. In vitro and in vivo experiments, including CCK-8, EdU, plate cloning, transwell, scratch, subcutaneous tumor formation, liver metastasis in situ, and lung metastasis assays confirmed the anti-carcinogenic effects. Mechanistically, circCEP70 encoded a novel protein named CEP70-160aa, which interacted with PKM2 and hindered its translocation into the nucleus. This interaction led to reduce STAT3 phosphorylation in the nucleus, thus inhibiting HCC proliferation and metastasis. In cirrhotic microenvironment, circCEP70 prevented HCC proliferation and metastasis through PKM2/STAT3 axis, and RNA binding protein U2AF2 could inhibit circCEP70 expression.
Collapse
Affiliation(s)
- Lian Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Cardiovascular Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Liangliang Xu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Wenwei Liao
- Department of Thoracic Surgery, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, 518020, China
- The First Affiliated Hospital, Jinan University, Guangzhou, 510630, China
| | - Peng Wang
- Department of Burns, Sichuan Academy of Medical Science, Sichuan Provincial People'S Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, China
| | - Mingqing Xu
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Hepato-Pancreato-Biliary Surgery, Meishan City People'S Hospital, Meishan Hospital of West China Hospital, Sichuan University, Meishan, 620010, China
| | - Bo Li
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Ming Zhang
- Division of Liver Surgery, Department of General Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China.
| |
Collapse
|
137
|
Kim MY, Hwangbo H, Ji SY, Kim DH, Park SH, Hong SH, Kim GY, Bang E, Choi YH. Cynaropicrin Suppresses Cell Proliferation by Inducing Mitophagy through p38 MAPK-Mediated Mitochondrial ROS Generation in Human Hepatocellular Carcinoma Cells. J Microbiol Biotechnol 2025; 35:e2501025. [PMID: 40295200 PMCID: PMC12089954 DOI: 10.4014/jmb.2501.01025] [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] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/25/2025] [Accepted: 02/26/2025] [Indexed: 04/30/2025]
Abstract
Cynaropicrin, a sesquiterpene lactone, has diverse pharmacological activities. However, its anticancer activity against hepatocellular carcinoma (HCC) has not been fully elucidated. Here, we investigated the cytotoxic effects of cynaropicrin and examined its mechanism of action in human HCC cells. The results demonstrated that cynaropicrin significantly induced cytotoxicity and autophagy in HCC cells, but not in immortalized non-cancerous hepatocytes, which was related to the generation of mitochondrial reactive oxygen species (mtROS) and induction of mitochondrial membrane potential loss. Under cynaropicrin treatment, the expression of microtubule-associated protein light chain 3, which is involved in the elongation of the phagophore membrane, was upregulated, whereas the expression of Beclin-1 and p62, which are essential for the formation of autophagosomes, was downregulated. In addition, the expression of mitophagy regulators PTEN-induced kinase 1 (PINK1) and Parkin in the mitochondria increased, suggesting the induction of autophagic flux in the mitochondria. However, N-acetyl-l-cysteine, a ROS scavenger, counteracted cynaropicrin-induced effects. Moreover, cynaropicrin increased the phosphorylation of p38 mitogen-activated protein kinase (MAPK), and the p38 MAPK inhibitor, SB203580, specifically attenuated cynaropicrin-induced cytotoxicity and mtROS production. Importantly, SB203580 reversed cynaropicrin-induced expression of PINK1 and Parkin in the mitochondria. Collectively, our findings demonstrate that cynaropicrin exerts cytotoxic effects against HCC cells by inducing mitochondrial autophagy through the activation of the p38 MAPK-ROS pathway, indicating that cynaropicrin could be a potential therapeutic agent for liver cancer treatment.
Collapse
Affiliation(s)
- Min Yeong Kim
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Hyun Hwangbo
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Seon Yeong Ji
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Da Hye Kim
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Shin-Hyung Park
- Department of Pathology, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Su Hyun Hong
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Gi Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 63243, Republic of Korea
| | - EunJin Bang
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| | - Yung Hyun Choi
- Basic Research Laboratory for the Regulation of Microplastic-Mediated Diseases and Anti-Aging Research Center, Dong-eui University, Busan 47340, Republic of Korea
- Department of Biochemistry, Dong-eui University College of Korean Medicine, Busan 47227, Republic of Korea
| |
Collapse
|
138
|
Xiong K, Huang K, Liu Y, Pang H, Chen P, Zheng Y, Li T, Li Z, Zhang M, Zheng D, Huang X, Cao M, Li Q, Liang J, Fan H, Li D, Sun J, Wen Z, Jiang Y. Efficacy and safety of TACE combined with TKIs and PD-1 inhibitors in HCC patients with prior TIPS. Front Oncol 2025; 15:1570029. [PMID: 40342829 PMCID: PMC12058502 DOI: 10.3389/fonc.2025.1570029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Accepted: 03/31/2025] [Indexed: 05/11/2025] Open
Abstract
Purpose To compare the efficacy and safety of TACE combined with TKIs and PD-1 inhibitors between HCC patients with and without prior TIPS. Methods This retrospective propensity score matching (PSM) study included advanced HCC patients treated with prior TIPS followed by TKIs, PD-1 inhibitors, and TACE between January 2021 and January 2023. Patients were matched with a control group of HCC patients who had not undergone TIPS (non-TIPS). Outcome measures included objective response rate (ORR) using modified RECIST (mRECIST v1.1), disease control rate (DCR), progression-free survival (PFS), overall survival (OS), and safety assessed by CTCAE v5.0. Results A total of 172 patients were included before PSM. After PSM, 42 patients with prior TIPS were matched with 71 non-TIPS patients. ORR was 31.0% in the TIPS group and 57.7% in the non-TIPS group (p = 0.007), Both PFS and OS were longer in the non-TIPS group, with a median PFS of 7.9 months for TIPS patients versus 12.3 months for non-TIPS patients (hazard ratio [HR] = 2.253, p < 0.001), and a median OS of 13.5 months versus 21.1 months, respectively (HR = 2.282, p = 0.002). Treatment-related adverse events showed no significant differences between the two groups. Conclusion TACE combined with TKIs and PD-1 inhibitors showed lower efficacy in HCC patients with prior TIPS, but it remains a viable option, providing a favorable safety profile and effective disease control.
Collapse
Affiliation(s)
- Kai Xiong
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Kuiyuan Huang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Yulong Liu
- Department of Vascular Intervention, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Huajin Pang
- Division of Vascular and Interventional Radiology, Department of General Surgery, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Peng Chen
- State Key Laboratory of Organ Failure Research, Guangdong Provincial Key Laboratory of Viral Hepatitis Research, Department of Infectious Diseases, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Yalu Zheng
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Tengzheng Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Zhangyun Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Moran Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Dandan Zheng
- Department of Radiation Oncology, The First Affiliated Hospital of Zhejiang University, Hangzhou, Zhejiang, China
| | - Xiaohong Huang
- Department of Pathology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Mingrong Cao
- Department of Hepatobiliary and pancreatic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Qiang Li
- Department of Hepatobiliary and pancreatic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Junjie Liang
- Department of Hepatobiliary and pancreatic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Huizhen Fan
- Department of Gastroenterology, The People′s Hospital of Yichun City, Yichun, Jiangxi, China
| | - Deju Li
- Department of Vascular Surgery, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| | - Jian Sun
- Department of Hepatobiliary and pancreatic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| | - Zhili Wen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
| | - Yuchuan Jiang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, Nanchang University, Nanchang, Jiangxi, China
- Department of Hepatobiliary and pancreatic Surgery, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, China
| |
Collapse
|
139
|
Wang J, Shi J, Mi L, Li N, Han X, Zhao M, Duan X, Han G, Hou J, Yin F. Identification and validation of a lenvatinib resistance-related prognostic signature in HCC, in which PFKFB4 contributes to tumor progression and lenvatinib resistance. BMC Gastroenterol 2025; 25:287. [PMID: 40269756 PMCID: PMC12020327 DOI: 10.1186/s12876-025-03861-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 04/07/2025] [Indexed: 04/25/2025] Open
Abstract
BACKGROUND Drug resistance reflects the evolution of tumors and represents the leading factor behind recurrence and death. Lenvatinib is the first-line therapy for hepatocellular carcinoma (HCC), but its effectiveness is limited by rapid development of resistance. Therefore, we aimed to identify lenvatinib resistance-related genes and assess their influence on prognosis and treatment response in HCC. METHODS The GSE186191 dataset served as the discovery cohort to identify lenvatinib resistance-related genes. A Venn diagram analysis delineated the intersection between lenvatinib resistance-related genes and prognostic-associated genes derived from The Cancer Genome Atlas (TCGA) database. The LASSO Cox regression model was implemented to construct a multigene signature in the TCGA cohort. A nomogram was built by integrating the TNM stage and our prognostic model. The gene signature and nomogram were further validated using HCC patients from the International Cancer Genome Consortium (ICGC) cohort. Mutation signatures, therapeutic response, functional enrichment, and immune profile analyses were performed in the two groups. Two lenvatinib-resistant (LR) HCC cells were established using a concentration gradient increment method. PFKFB4 expression was detected via qRT-PCR and western blot assay. The CCK-8 assay and flow cytometry were utilized to evaluate the proliferation and apoptosis of LR cells under different interventions. RESULTS We developed a lenvatinib resistance-related gene signature (ALPK3, SLC2A2, CTSV, and PFKFB4), and demonstrated that's a precise, independent, and specific prognostic model for HCC patients. High-risk patients were characterized by a predisposition to TP53 mutations, aggressive tumor features, and treatment resistance. The risk score was significantly associated with immune cell infiltration, immune checkpoint expression, angiogenesis, and tumor stemness. PFKFB4 was overexpressed in LR cells, and its knockdown significantly enhances the antiproliferative and pro-apoptotic effects of lenvatinib on resistant cells. CONCLUSIONS The lenvatinib resistance-related prognostic signature exhibits strong predictive power for prognosis in HCC patients and may serve as an effective tool for guiding treatment decisions. PFKFB4 promotes tumor progression and lenvatinib resistance, highlighting its potential as a novel therapeutic target for HCC. CLINICAL TRIAL NUMBER Not applicable.
Collapse
Affiliation(s)
- Jinfeng Wang
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Jianfei Shi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Lili Mi
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Ning Li
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Xin Han
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Man Zhao
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Xiaoling Duan
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Guangjie Han
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Jiaojiao Hou
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China
| | - Fei Yin
- Department of Gastroenterology, The Fourth Hospital of Hebei Medical University, Tianshan Street 169, Shijiazhuang, Hebei, China.
| |
Collapse
|
140
|
Lin KT, Muneer G, Huang PR, Chen CS, Chen YJ. Mass Spectrometry-Based Proteomics for Next-Generation Precision Oncology. MASS SPECTROMETRY REVIEWS 2025. [PMID: 40269546 DOI: 10.1002/mas.21932] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/25/2025]
Abstract
Cancer is the leading cause of death worldwide characterized by patient heterogeneity and complex tumor microenvironment. While the genomics-based testing has transformed modern medicine, the challenge of diverse clinical outcomes highlights unmet needs for precision oncology. As functional molecules regulating cellular processes, proteins hold great promise as biomarkers and drug targets. Mass spectrometry (MS)-based clinical proteomics has illuminated the molecular features of cancers and facilitated discovery of biomarkers or therapeutic targets, paving the way for innovative strategies that enhance the precision of personalized treatment. In this article, we introduced the tools and current achievements of MS-based proteomics, choice of discovery and targeted MS from discovery to validation phases, profiling sensitivity from bulk samples to single-cell level and tissue to liquid biopsy specimens, current regulatory landscape of MS-based protein laboratory-developed tests (LDTs). The challenges, success and future perspectives in translating research MS assay into clinical applications are also discussed. With well-designed validation studies to demonstrate clinical benefits and meet the regulatory requirements for both analytical and clinical performance, the future of MS-based assays is promising with numerous opportunities to improve cancer diagnosis, treatment, and monitoring.
Collapse
Affiliation(s)
- Kuen-Tyng Lin
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | - Gul Muneer
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
| | | | - Ciao-Syuan Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| | - Yu-Ju Chen
- Institute of Chemistry, Academia Sinica, Taipei, Taiwan
- Department of Chemistry, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
141
|
Oji K, Urade T, Omiya S, Kido M, Komatsu S, Gon H, Fukushima K, Yanagimoto H, Toyama H, Fukumoto T. Achieving textbook outcome in liver resection for hepatocellular carcinoma: malnutrition's pivotal role. Langenbecks Arch Surg 2025; 410:139. [PMID: 40266358 PMCID: PMC12018603 DOI: 10.1007/s00423-025-03703-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 04/06/2025] [Indexed: 04/24/2025]
Abstract
PURPOSE To investigate the impact of textbook outcome (TO) achievement on survival post-liver resection for hepatocellular carcinoma (HCC) and explore the associated factors. METHODS We retrospectively reviewed 330 patients diagnosed with HCC who underwent initial liver resection at our hospital between January 2011 and December 2019. We also investigated the achievement rates of five TOs and sub-analyzed the relationship between them and malnutrition. The patient's nutritional status was classified following the Global Leadership Initiative on Malnutrition (GLIM) criteria. RESULTS The TO achievement rate was 72.7%. In the prognostic analysis, the TO-achieving group showed significantly longer overall survival (OS) and recurrence-free survival (RFS). Significant differences in age, body mass index, weight loss, muscle mass, serum aspartate aminotransferase level, serum protein induced by vitamin K absence or antagonist-II, tumor characteristics, intraoperative blood loss, perioperative transfusion, and nutritional status were found between the groups. CONCLUSIONS TO achievement is associated with OS and RFS post-liver resection for HCC. The TO is valuable for evaluating treatment quality in liver resection. Additionally, malnutrition graded following the GLIM criteria, age, tumor stage, and intraoperative blood loss are independent factors for achieving a TO post-liver resection for HCC.
Collapse
Affiliation(s)
- Kentaro Oji
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takeshi Urade
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan.
| | - Satoshi Omiya
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Masahiro Kido
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Shohei Komatsu
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hidetoshi Gon
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Kenji Fukushima
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hiroaki Yanagimoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Hirochika Toyama
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| | - Takumi Fukumoto
- Department of Surgery, Division of Hepato-Biliary-Pancreatic Surgery, Kobe University Graduate School of Medicine, Kobe University, 7-5-2 Kusunoki-cho, Chuo-ku, Kobe, 650-0017, Japan
| |
Collapse
|
142
|
Zhong W, Zhao Z, Fang X, Sun J, Wei Y, Li F, Han B, Jin C. Constructing a neural network model based on tumor-infiltrating lymphocytes (TILs) to predict the survival of hepatocellular carcinoma patients. PeerJ 2025; 13:e19351. [PMID: 40292102 PMCID: PMC12032962 DOI: 10.7717/peerj.19351] [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] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary liver cancer worldwide, and early pathological diagnosis is crucial for formulating treatment plans. Despite the widespread attention to pathology in the treatment of HCC patients, a large amount of information contained in pathological images is often overlooked. Methods We retrospectively collected clinical data and pathological slide images from (a) 331 HCC patients at Qingdao University Affiliated Hospital between January 2013 and December 2016 and (b) 180 HCC patients from The Cancer Genome Atlas (TCGA). After data screening, precise quantification of various cell types was achieved using QuPath software. Key factors related to the survival prognosis of pathologically confirmed HCC patients were identified through Cox regression and neural network models, and potential therapeutic targets were screened. Results Our study showed that tumour-infiltrating lymphocytes (TILs) had a protective effect. We quantified the TILs index by machine learning and built a neural network model to predict the prognostic risk of patients (ROC = 0.836 for training set ROC validation set). 95% CI [0.7688-0.896], and there was a significant difference in prognosis in the high-low risk group predicted by the model (p = 2.6e-18, HR = 0.18, 95% CI [0.12-0.27], and TNFSF4 was identified as a possible immunotherapy target. Conclusion This study included a total of 511 patients, divided into a training cohort of 331 cases (from Qingdao University Hospital between January 2013 and December 2016) and a validation cohort of 180 cases (TCGA). The results revealed that tumor-infiltrating lymphocytes (TILs) have a protective effect and successfully predicted the survival risk of liver cancer patients using machine learning and neural network technology. The discovery of TNFSF4 provides a new potential target for immunotherapy.
Collapse
Affiliation(s)
- Wenqing Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ziyin Zhao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xin Fang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jingyi Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanbing Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengda Li
- Department of Hepatobiliary Surgery, Gao mi People’s Hospital, Weifang, Shandong, China
| | - Bing Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Jin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
143
|
Li X, Lu S, Huang CK. The Complexity of SIRT2 in Chronic Liver Disease: Liver SIRT2 Promotes Hepatocellular Carcinoma Development. Cell Mol Gastroenterol Hepatol 2025:101512. [PMID: 40280175 DOI: 10.1016/j.jcmgh.2025.101512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/20/2025] [Accepted: 04/03/2025] [Indexed: 04/29/2025]
Affiliation(s)
- Xinjian Li
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana
| | - Shaolei Lu
- Department of Pathology and Laboratory Medicine, Warren Alpert Medical School of Brown University, Rhode Island Hospital, Providence, Rhode Island
| | - Chiung-Kuei Huang
- Department of Pathology and Laboratory Medicine, Tulane University School of Medicine, New Orleans, Louisiana.
| |
Collapse
|
144
|
Sanchez CA, Treviso EM, Rocha CDS, Antunes LMG. Diallyl Disulfide Reduces Ethyl Carbamate-Induced Cytotoxicity and Apoptosis in Intestinal and Hepatic Cells. Chem Res Toxicol 2025; 38:623-634. [PMID: 40145834 PMCID: PMC12015961 DOI: 10.1021/acs.chemrestox.4c00439] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 03/11/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025]
Abstract
Epidemiological studies indicate that lifestyle and dietary habits are associated with an increasing cancer incidence. Consuming fermented foods and alcoholic beverages and smoking can expose humans to ethyl carbamate (EC), a probable human carcinogen classified as group 2A by the International Agency for Research on Cancer (IARC). Increasing the intake of bioactive compounds can reduce EC-induced toxicity. Diallyl disulfide (DADS), found in garlic, may protect against damage induced by chemical agents and natural compounds. Here, the potential protective effect of DADS against EC was investigated by evaluating EC-induced cytotoxicity, DNA damage, apoptosis, and reactive oxygen species production in colorectal adenocarcinoma (Caco-2) and hepatocarcinoma (HepG2) cells. To this end, resazurin, comet, and annexin V-FITC staining assays and CM-H2DCFDA markers were used to evaluate the effect on Caco-2 and HepG2 cells of protocols combining DADS (10-120 μM) and EC (80 mM). The protocols were as follows: (i) cells pretreated with DADS for 2 h and exposed to EC for 24 h; (ii) cells pretreated with DADS for 24 h and exposed to EC for 24 h; (iii) cells simultaneously exposed to DADS and EC for 24 h; (iv) cells exposed to EC for 24 h and treated with DADS for 2 h. EC induced cytotoxicity and apoptosis in Caco-2 and HepG2 cells and oxidative damage in Caco-2 cells. Combined exposure to DADS and EC for 24 h decreased EC-mediated cytotoxicity and apoptosis in both Caco-2 and HepG2 cells. These findings encourage further studies on the mechanisms of action of the combined DADS and EC.
Collapse
Affiliation(s)
- Caroline Andolfato Sanchez
- Department of Clinical Analysis,
Toxicology Food Science, School of Pharmaceutical Sciences of Ribeirão
Preto, University of São Paulo, Av. do Café, Vila Monte Alegre, Ribeirão Preto, SP 14040-903, Brazil
| | - Estefani Maria Treviso
- Department of Clinical Analysis,
Toxicology Food Science, School of Pharmaceutical Sciences of Ribeirão
Preto, University of São Paulo, Av. do Café, Vila Monte Alegre, Ribeirão Preto, SP 14040-903, Brazil
| | - Cecília
Cristina de Souza Rocha
- Department of Clinical Analysis,
Toxicology Food Science, School of Pharmaceutical Sciences of Ribeirão
Preto, University of São Paulo, Av. do Café, Vila Monte Alegre, Ribeirão Preto, SP 14040-903, Brazil
| | - Lusânia Maria Greggi Antunes
- Department of Clinical Analysis,
Toxicology Food Science, School of Pharmaceutical Sciences of Ribeirão
Preto, University of São Paulo, Av. do Café, Vila Monte Alegre, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
145
|
Liao L, Yi Q, Zhao Z, Xu M, Wu T, Chen S, Liu Y. miR-6844/HSD17B13 Axis Contributes the Malignant Phenotype of Hepatocellular Carcinoma Cells. Cell Biol Int 2025. [PMID: 40255210 DOI: 10.1002/cbin.70025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 04/03/2025] [Accepted: 04/09/2025] [Indexed: 04/22/2025]
Abstract
Hepatocellular carcinoma (HCC) is a prevalent form of primary liver cancer, and aberrant miRNAs expression significantly contributes to its progression. Although the abnormal expression of miR-6844 in HCC has been reported, its impact on the malignant phenotype of HCC cells and its potential mechanism remains unclear. In this study, we initially conducted a bioinformatics analysis to investigate the differential expression of miR-6844 in HCC tissues and its impact on patient prognosis. The association between miR-6844 expression levels and clinical characteristics of HCC patients was subsequently investigated by integrating data from clinical samples. Ultimately, the impact of miR-6844 on the malignant phenotype of HCC cells and the underlying mechanisms were examined through in vitro cellular experiments. The results showed that a high expression of miR-6844 in HCC, which was associated with poor prognosis and exhibited significant correlations with intrahepatic metastasis and clinical stage among patients. The upregulation of miR-6844 promoted the proliferation, migration, and invasion of HCC cells while suppressing apoptosis. Conversely, the downregulation of miR-6844 significantly attenuated the malignant phenotype of HCC cells. In addition, HSD17B13 was identified as a target gene of miR-6844, and the overexpression of HSD17B13 partially counteracted the oncogenic effects induced by miR-6844 in HCC cells, otherwise the opposite. Taken together, the above results suggest that miR-6844 plays a regulatory role in the malignant phenotype of HCC cells through its targeting of HSD17B13.
Collapse
Affiliation(s)
- Li Liao
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Qilin Yi
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Zhen Zhao
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Ming Xu
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Tao Wu
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Shuai Chen
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| | - Yu Liu
- Department of Hepatobiliary Surgery, Yueyang Central Hospital, Yueyang, Hunan, People's Republic of China
| |
Collapse
|
146
|
Wang TC, Huang W, Li SX, Li JX, Zhou P, Wang LZ, Wei N, Cai WW, Hu JJ, Xiao YD. High Fibroblast Activation Protein Expression in Hepatocellular Carcinoma: CT Imaging Features and Histological Characteristics. Acad Radiol 2025:S1076-6332(25)00315-0. [PMID: 40254479 DOI: 10.1016/j.acra.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 04/22/2025]
Abstract
RATIONALE AND OBJECTIVES To clarify computed tomography (CT) imaging features of high fibroblast activation protein (FAP) expression in hepatocellular carcinoma (HCC) and to investigate their correlation to histological characteristics and prognosis. METHODS This retrospective multicenter study evaluated patients with HCC who underwent liver resection between August 2013 and June 2023. Histological staining for FAP was performed, and patients were classified into low and high-FAP expression groups. A predictive model was developed and validated to identify FAP expression levels among CT imaging features. Moreover, CT imaging-related histological characteristics were evaluated. With the predictive model, patients in training cohort were stratified into predicted low and high-FAP expression groups. Overall survival (OS) and recurrence-free survival (RFS) were compared accordingly. Besides, propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) analysis were performed, and OS and RFS between the two groups were also compared. RESULTS In total, 1197 patients were included (high-FAP HCCs, n=267). Delayed central enhancement (OR: 15.196, 95% CI: 8.996-25.670, P<0.001) and dilated vasculature (OR: 7.455, 95% CI: 4.928-11.277, P<0.001) independently predicted high-FAP expression (AUCs: 0.779, training; 0.766, external validation). Based on these two CT imaging features, intratumoral fibrosis (ITF) and vessel-encapsulating tumor cluster (VETC) pattern were determined. Compared to low-FAP HCCs, high-FAP HCCs were more frequently with high ITF grade (54.7% vs. 22.4%; P<0.001) and VETC pattern (52.1% vs. 22.6%; P<0.001). Patients were divided into predicted low (n=564) and high (n=262) FAP expression groups. The predicted low-FAP group had significantly better or favorable trend toward improved OS and RFS than predicted high-FAP group before (P<0.001, OS and RFS), and after PSM (P=0.009, OS; P=0.005, RFS) and IPTW (P=0.019, OS; P=0.077, RFS). CONCLUSION Delayed central enhancement and dilated vasculature are independent factors of high-FAP expression in HCC. Noninvasive identifying FAP expression may offer potential prognostic and therapeutic insights.
Collapse
Affiliation(s)
- Tian-Cheng Wang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Wei Huang
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Shu-Xian Li
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Jun-Xiang Li
- Department of Interventional Radiology, Guizhou Medical University Affiliated Cancer Hospital, Guiyang 550004, China (J.X.L.)
| | - Peng Zhou
- Department of Pathology, the Second Xiangya Hospital of Central South University, Changsha 410011, China (P.Z.)
| | - Li-Zhou Wang
- Department of Interventional Radiology, the Affiliated Hospital of Guizhou Medical University, Guiyang 550004, China (L.Z.W.)
| | - Nan Wei
- Department of Diagnostic and Interventional Radiology, University Hospital Heidelberg, Heidelberg 69120, Germany (N.W.)
| | - Wen-Wu Cai
- Department of Liver Surgery, the Second Xiangya Hospital of Central South University, Changsha 410011, China (W.W.C.)
| | - Jun-Jiao Hu
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.)
| | - Yu-Dong Xiao
- Department of Radiology, the Second Xiangya Hospital of Central South University, No.139 Middle Renmin Road, Changsha 410011, China (T.C.W., W.H., S.X.L., J.J.H., Y.D.X.).
| |
Collapse
|
147
|
Liu H, Ge W, Yu X, Luo J, Zhang J, Yang M, Cao L, Zhang Y, Wang R, Yang C, Li P, Tian M, Peng X, Peng L, Wu D, Liu M, Liang Q, Zhang S, Li W, Rong P, Li H, Ma X, Wang W. CRISPR/Cas9-mediated SHP-1-knockout T cells combined with simvastatin enhances anti-tumor activity in humanized-PDX HCC model. iScience 2025; 28:112266. [PMID: 40241752 PMCID: PMC12003012 DOI: 10.1016/j.isci.2025.112266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 12/04/2024] [Accepted: 03/18/2025] [Indexed: 04/18/2025] Open
Abstract
Hepatocellular carcinoma (HCC) resists immunotherapy due to its immunosuppressive microenvironment. Sarcoma homology 2 domain-containing protein tyrosine phosphatase-1 (SHP-1) inhibits T cell receptor signaling, and its pharmacological inhibition is limited by poor selectivity and membrane permeability. Here, we generated CRISPR-edited SHP-1-knockout (KO) CD8+ T cells to enhance adoptive therapy against HCC. Single-cell RNA sequencing of HCC patient T cells revealed elevated SHP-1 in exhausted subsets. SHP-1-KO T cells exhibited increased effector memory T cells (TEM) proportions and enhanced IFN-γ/Granzyme B/perforin secretion, improving cytotoxicity against HCC lines. In humanized PDX models, SHP-1-KO T cells demonstrated superior tumor-killing activity. Transcriptomics identified upregulated lipid metabolism pathways, with HMGCR as a hub gene. Combining SHP-1-KO T cells with simvastatin (HMGCR inhibitor) synergistically amplified anti-HCC efficacy. This study proposes a dual strategy combining SHP-1-targeted cell therapy and metabolic modulation to overcome immunotherapy resistance, offering a translatable approach for HCC treatment.
Collapse
Affiliation(s)
- Huaping Liu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- Department of Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Wu Ge
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Xiaoping Yu
- Department of Radiology, Hunan Cancer Hospital and the Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Jianwei Luo
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Juan Zhang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Min Yang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Lu Cao
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Yangnan Zhang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Ruike Wang
- Xiangya School of Medicine, Central South University, Changsha, Hunan, China
| | - Cejun Yang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Pei Li
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Mengyu Tian
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - XiaoPei Peng
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Lei Peng
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Di Wu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Muqi Liu
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Qi Liang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Shengwang Zhang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Wei Li
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
| | - Pengfei Rong
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Hailan Li
- Department of Radiology, Hunan Provincial People’s Hospital, The First Affiliated Hospital of Hunan Normal University
| | - Xiaoqian Ma
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| | - Wei Wang
- Department of Radiology, the 3 Xiangya Hospital of Central South University, Changsha, Hunan, China
- The Institute for Cell Transplantation and Gene Therapy, Central South University, Changsha, Hunan, China
- Molecular Imaging Research Center of Central South University, Changsha, Hunan, China
| |
Collapse
|
148
|
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
|
149
|
Chen M, Bie L. Intratumoral microbiota for hepatocellular carcinoma: from preclinical mechanisms to clinical cancer treatment. Cancer Cell Int 2025; 25:152. [PMID: 40247312 PMCID: PMC12007317 DOI: 10.1186/s12935-025-03745-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Accepted: 03/10/2025] [Indexed: 04/19/2025] Open
Abstract
Intratumoral microbiota has been found to be a crucial component of hepatocellular carcinoma (HCC). Due to insufficient recognition, technical limitations, and low biomass of intratumoral microbiota, it is poorly understood. Intratumoral microbiota exhibit significant diversity in HCC tissues. It is involved in the development of HCC through several mechanisms, such as remodeling the immunosuppressive microenvironment, metabolic reprogramming, and genetic alterations. Moreover, intratumoral microbiota is associated with the metastasis of HCC cells. Herein, we reviewed the history of intratumoral microbiota, applied biotechnology to depict the signatures of intratumoral microbiota, investigated the potential sources of intratumoral microbiota, and assessed their functions, mechanisms, and heterogeneity. Furthermore, in this review, we summarized the development of therapeutics that can be used in the treatment of HCC and proposed future perspectives for research in this field.
Collapse
Affiliation(s)
- Muhua Chen
- Department of Hepato-Pancreato-Biliary & Gastric Medical Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, 310022, Zhejiang, China.
| | - Lei Bie
- Department of Thoracic Surgery, Wuhan No.1 Hospital, Wuhan, 430030, Hubei, China
| |
Collapse
|
150
|
Lin Y, Ma Y, Chen Y, Huang Y, Lin J, Xiao Z, Cui Z. Diagnostic and prognostic performance of serum GPC3 and PIVKA-II in AFP-negative hepatocellular carcinoma and establishment of nomogram prediction models. BMC Cancer 2025; 25:721. [PMID: 40247208 PMCID: PMC12007284 DOI: 10.1186/s12885-025-14025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE A significant proportion, ranging from 20 to 40%, of individuals with hepatocellular carcinoma (HCC) do not exhibit elevated Alpha-fetoprotein (AFP) levels. This study aimed to evaluate the utility of serum glypican-3 (GPC3) and protein induced by vitamin K absence or antagonist II (PIVKA-II) in an AFP-negative HCC (N-HCC) population, and to develop nomogram diagnostic and prognostic prediction models utilizing GPC3 and PIVKA-II. METHODS Serum GPC3 and PIVKA-II levels were measured in this case-control study, followed by the establishment of a receiver operating characteristic (ROC) curve, restricted cubic spline (RCS), and Kaplan-Meier survival curve. Additionally, a diagnostic prediction nomogram was constructed using univariate and multivariate logistic regression. Furthermore, we utilized least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression to develop a prognostic prediction nomogram. The performance of these models was evaluated using ROC curve analysis and decision curve analysis (DCA). RESULTS Serum GPC3 and PIVKA-II expression levels were significantly elevated in untreated patients with N-HCC (especially stageI and tumor size < 3 cm) compared to those with AFP-negative benign liver disease (N-BLD). Derived from ROC analysis, the diagnostic cutoff points for GPC3 and PIVKA-II were set at 0.100 ng/mL and 40.00 mAU/mL, respectively. PIVKA-II demonstrated sensitivity and specificity of 84.62% and 90.38%, surpassing GPC3's 76.92% and 73.08%. The area under the ROC curve (AUC) for a diagnostic prediction nomogram incorporating GPC3, PIVKA-II, and gamma-glutamyltransferase (GGT) was 0.943 (95% CI: 0.912-0.974), superior to models using GPC3 or PIVKA-II alone. This model showed 95.20% sensitivity and 81.70% specificity in differentiating N-HCC from N-BLD. Stratifying patients into high-risk and low-risk groups using cutoff values established by RCS for GPC3 (0.124 ng/mL) and PIVKA-II (274 mAU/mL) revealed significant associations between these risk stratifications and patient survival. Finally, the use of GPC3-highrisk, cirrhosis, albumin (ALB), portal venous thrombosis (PVT), and surgical treatment as five parameters in the nomogram prognostic prediction model effectively differentiated between high- and low-risk prognostic patients with N-HCC with relatively high accuracy. CONCLUSIONS Serum GPC3 and PIVKA-II demonstrate clinical significance in the timely detection and prognosis assessment of N-HCC. The application of nomogram prediction models based on GPC3 and PIVKA-II stands as an important adjunctive tool for diagnosing and prognosticating N-HCC.
Collapse
Affiliation(s)
- Yingying Lin
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Yuefei Ma
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yan Chen
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Yepei Huang
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Jinchuan Lin
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhenzhou Xiao
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China.
| | - Zhaolei Cui
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China.
| |
Collapse
|