1
|
Jaber F, El-Serag HB. HES V2.0 surpasses GALAD for HCC detection: a review of multi-dimensional biomarker scores and studies. Hepat Oncol 2025; 12:2494446. [PMID: 40308043 PMCID: PMC12051611 DOI: 10.1080/20450923.2025.2494446] [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: 09/17/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
This was a narrative review of select studies published through September of 2024. We review the shift toward multi-dimensional scores such as HCC early detection screening (HES), GALAD, ASAP, and mt-HBT represents a significant advancement in biomarker research for hepatocellular carcinoma (HCC) detection. Unlike single biomarker approaches, these scores integrate various clinical and biochemical factors to enhance predictive accuracy by reflecting different complementary aspects of disease progression and HCC oncogenesis. Proper testing and validation of biomarker scores in phase 3 biomarker studies is essential before wide use can be recommended. We also review the comparative performance of biomarker scores in phase 3 studies. The new version of HES (HES V2.0) which includes AFP, AFP L3, DCP, and changes in their levels the past one year, if available, in addition to age, platelets, albumin, ALT and underlying liver disease etiology outperforms GALAD in detecting early-stage HCC with overall 6.7% higher sensitivity, and ASAP with 13.4%-18.0% higher sensitivity, both at fixed 90% specificity. HES V2.0 is a leading candidate biomarker score for prospective testing in clinical studies of early HCC detection.
Collapse
Affiliation(s)
- Fouad Jaber
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Hashem B. El-Serag
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Liu G, Kuang S, Zhong W, Yang A, Huang X, Xie Y, Zhang X, Li Y, Qin Q, Liu G. Exploring the potential mechanisms of Jinglinzi powder in treating hepatocellular carcinoma based on LC-MS, network pharmacology, molecular docking, and experimental validation. J Pharm Biomed Anal 2025; 263:116899. [PMID: 40286672 DOI: 10.1016/j.jpba.2025.116899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/29/2025]
Abstract
This study systematically predicts the active components, targets, and mechanisms of JLZP against HCC by integrating LC-MS, network pharmacology, and molecular docking, with experimental validation of its pro-pyroptotic effects. Through HERB and NPASS databases, 81 bioactive components of JLZP and 78 overlapping HCC-related targets were identified. Protein-protein interaction network and KEGG enrichment analyses revealed that JLZP likely induces cell death via cancer-related pathways. Molecular docking (CB-Dock2) demonstrated high binding affinity between JLZP core components (e.g., protopine) and pyroptosis-associated targets (NLRP3, GSDMD). In vitro experiments confirmed that JLZP significantly suppressed MHCC-97L cell proliferation and migration, while upregulating pyroptosis markers (IL-1β, IL-18) at both mRNA and protein levels, with these effects reversed by a pyroptosis inhibitor. This study is the first to elucidate JLZP's anti-HCC mechanism through pyroptosis activation, identifying its pharmacodynamic material basis and multi-target action. The "component-target-pathway-experiment" multidimensional strategy provides methodological insights for deciphering traditional Chinese medicine formulas, offering a theoretical foundation for developing JLZP-based anticancer therapies.
Collapse
Affiliation(s)
- Gaofeng Liu
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Shanshan Kuang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Weixing Zhong
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Anming Yang
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China
| | - Xiaoli Huang
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Yin Xie
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Xin Zhang
- Laboratory Medicine Center, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China
| | - Yikai Li
- School of Traditional Chinese Medicine, Southern Medical University, Guangzhou 510515, China
| | - Qingguang Qin
- Department of acupuncture and moxibustion, Hainan Provincial People's Hospital, Haikou 570311, China.
| | - Guangjie Liu
- Department of Neurosurgery, Nanfang Hospital, Southern Medical University, Guangzhou 510510, China.
| |
Collapse
|
3
|
Wang X, Li C, Chen L, He B, Li Y. Therapeutic potential of dual HDAC6/SIRT2 inhibition in Alzheimer's disease. Eur J Med Chem 2025; 294:117733. [PMID: 40381221 DOI: 10.1016/j.ejmech.2025.117733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disorder characterized by hallmark pathological changes such as amyloid β (Aβ) plaques, neurofibrillary tangles (NFTs) due to tau hyperphosphorylation, and neuroinflammation. Current therapeutic approaches focusing on single-target strategies exhibit limited efficacy, necessitating the exploration of novel multi-target approaches. Histone deacetylase 6 (HDAC6) and SIRT2, as two types of cytosolic histone deacetylases, have emerged as promising targets for AD treatment. HDAC6 plays a role in tau protein phosphorylation, while SIRT2 is involved in Aβ production. Both enzymes regulate microtubule proteins, impacting the formation of NFTs and Aβ plaques. Inhibition of HDAC6 reduces tau hyperphosphorylation, improves microtubule stability, and mitigates neuroinflammation, whereas SIRT2 inhibition attenuates Aβ accumulation and neuroinflammation. Recent studies indicate that dual-targeted inhibition of HDAC6 and SIRT2 may exhibit synergistic effects, suggesting it as a promising strategy for AD treatment. This review summarizes the biological roles of HDAC6 and SIRT2 in AD pathology and examines the development of dual-target inhibitors. It also discusses the challenges, including selectivity and toxicity, emphasizing that the development of combined HDAC6 and SIRT2 inhibitors represents a new direction for future AD treatment.
Collapse
Affiliation(s)
- Xingyu Wang
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Cunjiang Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Lei Chen
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China
| | - Bin He
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China.
| | - Yan Li
- State Key Laboratory of Discovery and Utilization of Functional Components in Traditional Chinese Medicine, Engineering Research Center for the Development and Application of Ethnic Medicine and TCM (Ministry of Education), Guizhou Provincial Engineering Research Center for the Development and Application of Ethnic Medicine and TCM, School of Pharmacy, Guizhou Medical University, Guian New Area, 561113, China; School of Basic Medical Science, Guizhou Medical University, Guian New Area, 561113, China.
| |
Collapse
|
4
|
Vasudevan J, Vijayakumar R, Reales-Calderon JA, Lam MSY, Ow JR, Aw J, Tan D, Tan AT, Bertoletti A, Adriani G, Pavesi A. In vitro integration of a functional vasculature to model endothelial regulation of chemotherapy and T-cell immunotherapy in liver cancer. Biomaterials 2025; 320:123175. [PMID: 40043483 DOI: 10.1016/j.biomaterials.2025.123175] [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: 09/17/2024] [Revised: 01/31/2025] [Accepted: 02/05/2025] [Indexed: 04/06/2025]
Abstract
The complex tumor microenvironment (TME) presents significant challenges to the development of effective therapies against solid tumors, highlighting the need for advanced in vitro models that better recapitulate TME biology. To address this, we developed a vascularized human liver tumor model using a microfluidic platform, designed to test both drug and cell-based therapies. This model mimics critical tumorigenic features such as hypoxia, extracellular matrix (ECM), and perfusable vascular networks. Intravascular administration of Sorafenib demonstrated its ability to disrupt vascular structures significantly, while eliciting heterogeneous responses in two distinct liver tumor cell lines, HepG2 and Hep3b. Furthermore, treatment with engineered T-cells revealed that the tumor vasculature impeded T-cell infiltration into the tumor core but preserved their cytotoxic capacity, albeit with reduced exhaustion levels. Cytokine analysis and spatial profiling of vascularized tumor samples identified proinflammatory factors that may enhance T-cell-mediated antitumor responses. By capturing key TME characteristics, this microfluidic platform provides a powerful tool enabling detailed investigation of tumor-immune and tumor-vascular interactions. Its versatility could serve as a promising bridge between preclinical studies and clinical testing, offering opportunities for developing and optimizing personalized therapeutic strategies for solid tumors.
Collapse
Affiliation(s)
- Jyothsna Vasudevan
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore
| | - Ragavi Vijayakumar
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jose Antonio Reales-Calderon
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Maxine S Y Lam
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Jin Rong Ow
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Joey Aw
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Damien Tan
- Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore
| | - Anthony Tanoto Tan
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Antonio Bertoletti
- Duke-NUS Medical School, 8 College Road, Singapore, 169857, Republic of Singapore
| | - Giulia Adriani
- Singapore Immunology Network (SIgN), Agency for Science, Technology, and Research (A∗STAR), 8A Biomedical Grove, Immunos, Singapore, 138648, Republic of Singapore; Department of Biomedical Engineering, National University of Singapore (NUS), 4 Engineering Drive 3, Singapore, 117583, Republic of Singapore
| | - Andrea Pavesi
- Mechanobiology Institute, National University of Singapore (NUS), 5A Engineering Drive 1, Singapore, 117411, Republic of Singapore; Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A∗STAR), 61 Biopolis Drive, Singapore, 138673, Republic of Singapore; Lee Kong Chian School of Medicine (LKCMedicine), Cancer Discovery and Regenerative Medicine Program, Nanyang Technological University, 308232, Republic of Singapore.
| |
Collapse
|
5
|
Zhang W, Hong X, Xiao Y, Wang H, Zeng X. Sorafenib resistance and therapeutic strategies in hepatocellular carcinoma. Biochim Biophys Acta Rev Cancer 2025; 1880:189310. [PMID: 40187502 DOI: 10.1016/j.bbcan.2025.189310] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 03/30/2025] [Accepted: 03/30/2025] [Indexed: 04/07/2025]
Abstract
Hepatocellular carcinoma (HCC) remains one of the most prevalent and lethal cancers globally. While surgical resection and liver transplantation offer potential cures for early-stage HCC, the majority of patients are diagnosed at advanced stages where such interventions are not viable. Sorafenib, a multi-target kinase inhibitor, has been a cornerstone in the treatment of advanced HCC since its approval in 2007. Despite its significant clinical impact, less than half of the treated patients derive long-term benefits due to the emergence of resistance and associated side effects. This review focuses on the role of sorafenib, an FDA-approved multi-target kinase inhibitor, in treating advanced HCC, discusses the mechanisms underlying its therapeutic effects and associated resistance, and explores additional therapeutic strategies being investigated to improve patient outcomes.
Collapse
Affiliation(s)
- Weijing Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China
| | - Xuechuan Hong
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China
| | - Yuling Xiao
- Department of Cardiology, Zhongnan Hospital of Wuhan University, School of Pharmaceutical Sciences, Wuhan University, Wuhan 430071, China; Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China; State Key Laboratory of Drug Research & Center of Pharmaceutics, Shanghai Institute of Materia Medica, Chinese Academy of Sciences, Shanghai 201203, China
| | - Hongbo Wang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai 264005, China.
| | - Xiaodong Zeng
- Shandong Laboratory of Yantai Drug Discovery, Bohai Rim Advanced Research Institute for Drug Discovery, Yantai 264117, China.
| |
Collapse
|
6
|
Ma C, Yu X, Zhang X, Su L, Jiang O, Cui R. Combination of radiotherapy and ICIs in advanced hepatocellular carcinoma: A systematic review of current evidence and future prospects (Review). Oncol Lett 2025; 30:342. [PMID: 40438865 PMCID: PMC12117537 DOI: 10.3892/ol.2025.15088] [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: 11/14/2024] [Accepted: 03/24/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a global health concern because of its rising prevalence and high fatality rates. Conventional treatments for advanced HCC (aHCC) have limited success, emphasizing the need for novel treatment options. Radiotherapy (RT) treatments, such as stereotactic body radiation and proton therapy, improve local tumor management via precision targeting. Moreover, immune checkpoint inhibitors (ICIs) that target the programmed cell death protein 1(PD-1)/PD ligand 1 (PD-L1) and cytotoxic T lymphocyte associated protein 4 (CTLA-4) pathways have promise for systemic antitumor effectiveness. The combination of RT and ICIs takes advantage of their complementary mechanisms: RT kills immunogenic cells and controls the tumor microenvironment to increase antigen presentation, whereas ICIs enhance and maintain antitumor immune responses. This combination enhances tumor regression and immune response in aHCC, improving response rate and progression-free survival with manageable safety. The present review aimed to summarize the rationale for combining RT + ICIs in patients with aHCC and clinical outcomes, as well as ways to enhance this combination technique. The combination of these models is a promising technique for improving outcomes for patients with aHCC and warrants further investigation.
Collapse
Affiliation(s)
- Cheng Ma
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Xinlin Yu
- Department of Oncology, The Affiliated Hospital of Chengdu University, Chengdu, Sichuan 610000, P.R. China
| | - Xialin Zhang
- Department of Oncology, Affiliated Hospital of Southwest Medical University, Luzhou, Sichuan 646000, P.R. China
| | - Lihong Su
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ou Jiang
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| | - Ran Cui
- Department of Oncology, The First People's Hospital of Neijiang, Neijiang, Sichuan 641000, P.R. China
| |
Collapse
|
7
|
Li Y, Liu X, Dong Y, Zhou Y. Angiogenesis causes and vasculogenic mimicry formation in the context of cancer stem cells. Biochim Biophys Acta Rev Cancer 2025; 1880:189323. [PMID: 40239849 DOI: 10.1016/j.bbcan.2025.189323] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 04/10/2025] [Accepted: 04/10/2025] [Indexed: 04/18/2025]
Abstract
Tumor occurrence, development, invasion, and metastasis are regulated by multiple mechanisms. Among these, angiogenesis promotes tumor progression mainly by supplying tumor tissue and providing channels for tumor metastasis. Cancer stem cells (CSCs) are another important factor affecting tumor progression by involving in tumor initiation and development, while remaining insensitive to conventional antitumor treatments. Among treatment strategies for them, owing to the existence of alternative angiogenic pathways or the risk of damaging normal stem cells, the clinical effect is not ideal. Angiogenesis and CSCs may influence each other in this process. Tumor angiogenesis can support CSC self-renewal by providing a suitable microenvironment, whereas CSCs can regulate tumor neovascularization and mediate drug resistance to anti-angiogenic therapy. This review summarized the role of vascular niche formed by angiogenesis in CSC self-renewal and stemness maintenance, and the function of CSCs in endothelial progenitor cell differentiation and pro-angiogenic factor upregulation. We also elucidated the malignant loop between CSCs and angiogenesis promoting tumor progression. Additionally, we summarized and proposed therapeutic targets, including blocking tumor-derived endothelial differentiation, inhibiting pro-angiogenic factor upregulation, and directly targeting endothelial-like cells comprising CSCs. And we analyzed the feasibility of these strategies to identify more effective methods to improve tumor treatment.
Collapse
Affiliation(s)
- Ying Li
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Xiaofang Liu
- Department of Anus and Intestine Surgery, The First Affiliated Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China
| | - Yaodong Dong
- Department of Otolaryngology Head and Neck Surgery, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| | - Yingying Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang, Liaoning, People's Republic of China.
| |
Collapse
|
8
|
Liu W, Wu G, Wang J, Wu S, Chen Z. Co‑treatment with triptolide and RSL3 induces hepatocellular carcinoma cell apoptosis and ferroptosis. Mol Med Rep 2025; 32:202. [PMID: 40376993 DOI: 10.3892/mmr.2025.13567] [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: 02/19/2025] [Accepted: 04/15/2025] [Indexed: 05/18/2025] Open
Abstract
Glutathione peroxidase 4 (GPx4; also known as phospholipid hydroperoxide glutathione peroxidase) inhibits cell death, including apoptosis and ferroptosis, by reducing lipid peroxidation. In addition, western blot assays showed that GPx4 protein levels were elevated in hepatocellular carcinoma (HCC) cells following triptolide (TPL) treatment. Therefore, it was hypothesized that HCC cells might develop partial resistance to TPL‑induced cytotoxicity through upregulation of the GPx4 protein. To enhance anti‑proliferative efficacy, the present study co‑treated HCC cells with a combination of TPL and RAS‑selective lethal 3 (RSL3), a well‑characterized GPx4 activity inhibitor. Subsequent experimental data produced from Cell Counting Kit‑8 and flow cytometric analyses demonstrated that co‑administration of TPL and RSL3 promoted HCC cell apoptosis, elevated intracellular reactive oxygen species levels and induced ferroptosis. These collective findings suggested that co‑treatment with TPL and RSL3 may induce both apoptotic and ferroptotic pathways in HCC cells.
Collapse
Affiliation(s)
- Weixia Liu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Guodi Wu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Jing Wang
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Shanshan Wu
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| | - Zhi Chen
- The State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310003, P.R. China
| |
Collapse
|
9
|
Pettas T, Lachanoudi S, Karageorgos FF, Ziogas IA, Fylaktou A, Papalois V, Katsanos G, Antoniadis N, Tsoulfas G. Immunotherapy and liver transplantation for hepatocellular carcinoma: Current and future challenges. World J Transplant 2025; 15:98509. [DOI: 10.5500/wjt.v15.i2.98509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 10/03/2024] [Accepted: 11/07/2024] [Indexed: 02/21/2025] Open
Abstract
Despite existing curative options like surgical removal, tissue destruction techniques, and liver transplantation for early-stage hepatocellular carcinoma (HCC), the rising incidence and mortality rates of this global health burden necessitate continuous exploration of novel therapeutic strategies. This review critically assesses the dynamic treatment panorama for HCC, focusing specifically on the burgeoning role of immunotherapy in two key contexts: early-stage HCC and downstaging advanced HCC to facilitate liver transplant candidacy. It delves into the unique immunobiology of the liver and HCC, highlighting tumor-mediated immune evasion mechanisms. Analyzing the diverse immunotherapeutic approaches including checkpoint inhibitors, cytokine modulators, vaccines, oncolytic viruses, antigen-targeting antibodies, and adoptive cell therapy, this review acknowledges the limitations of current diagnostic markers alpha-fetoprotein and glypican-3 and emphasizes the need for novel biomarkers for patient selection and treatment monitoring. Exploring the rationale for neoadjuvant and adjuvant immunotherapy in early-stage HCC, current research is actively exploring the safety and effectiveness of diverse immunotherapeutic approaches through ongoing clinical trials. The review further explores the potential benefits and challenges of combining immunotherapy and liver transplant, highlighting the need for careful patient selection, meticulous monitoring, and novel strategies to mitigate post-transplant complications. Finally, this review delves into the latest findings from the clinical research landscape and future directions in HCC management, paving the way for optimizing treatment strategies and improving long-term survival rates for patients with this challenging malignancy.
Collapse
Affiliation(s)
- Theodoros Pettas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Sofia Lachanoudi
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Filippos F Karageorgos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Ioannis A Ziogas
- Department of Surgery, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| | - Asimina Fylaktou
- Department of Immunology, National Peripheral Histocompatibility Center, Hippokration General Hospital, Thessaloniki 54642, Greece
| | - Vassilios Papalois
- Department of Transplant Surgery, Imperial College Renal and Transplant Centre, London W12 0HS, United Kingdom
| | - Georgios Katsanos
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| | - Nikolaos Antoniadis
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University School of Medicine, Thessaloniki 54642, Greece
| | - Georgios Tsoulfas
- Department of Transplantation Surgery, Center for Research and Innovation in Solid Organ Transplantation, Aristotle University of Thessaloniki, School of Medicine, Thessaloniki 54642, Greece
| |
Collapse
|
10
|
Shang LQ, Guo HX, Wang P, Sun XH, You JQ, Ma JT, Wang LK, Liu JX, Wang ZQ, Shao HB. Global scientific trends on hepatocellular carcinoma research from 2004 to 2023: A bibliometric and visualized analysis. World J Gastrointest Oncol 2025; 17:105781. [DOI: 10.4251/wjgo.v17.i6.105781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/22/2025] [Accepted: 04/17/2025] [Indexed: 06/13/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality worldwide, and the research landscape has rapidly evolved over the past two decades. Despite significant progress, an in-depth analysis of global research trends, collaborative networks, and emerging themes in HCC remains limited. This study aimed to fill this gap by conducting a bibliometric analysis to map the research output, identify key contributors, and highlight future directions in HCC research. We hypothesized that the analysis would reveal a growing focus on molecular mechanisms and immunotherapy, with increasing contributions from specific countries and institutions.
AIM To investigate global research trends, collaborative networks, and emerging themes in HCC from 2004 to 2023.
METHODS A bibliometric analysis was performed using 93987 publications from the Science Citation Index Expanded Database of the Web of Science Core Collection. Data were analyzed using the VOSviewer software to identify publication trends, leading contributors, and research themes. Key metrics included annual publication output, country and institutional contributions, journal impact, and thematic clusters. Statistical analysis was carried out to quantify trends and collaborations.
RESULTS The number of annual publications increased from 2341 in 2004 to 8756 in 2023, with 65583 papers (69.78%) published between 2014 and 2023. China, the United States, and Japan were the top contributors, constituting 58.3% of total publications. PLOS One published the most studies (n = 2145), while Gastroenterology had the highest average number of citations (78.4 citations per paper). Fudan University was the most prolific institution (n = 1872). Thematic analysis identified five main clusters, namely molecular mechanisms, therapeutic strategies, prognosis and immunology, risk factors, and diagnostic approaches.
CONCLUSION This study highlights the growing focus on HCC research, particularly in immunotherapy and molecular mechanisms, underscoring the significance of international collaboration to advance diagnosis and treatment strategies.
Collapse
Affiliation(s)
- Li-Qi Shang
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Hao-Xin Guo
- Department of Information Center, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Peng Wang
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Xiao-Han Sun
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Jia-Qi You
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Jun-Ting Ma
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Lu-Ke Wang
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Jia-Xi Liu
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Zhong-Qing Wang
- Department of Information Center, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| | - Hai-Bo Shao
- Department of Interventional Radiology, The First Hospital of China Medical University, Shenyang 110000, Liaoning Province, China
| |
Collapse
|
11
|
Cui X, Ruan M, Li Y, Yang C, Zhang J, Jin R, Wu D, Sun W, Wang R. Characteristics and outcomes of primary and secondary resistance to immune checkpoint inhibitors in hepatocellular carcinoma. Cancer Immunol Immunother 2025; 74:239. [PMID: 40481877 PMCID: PMC12145340 DOI: 10.1007/s00262-025-04089-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2025] [Accepted: 05/12/2025] [Indexed: 06/11/2025]
Abstract
Resistance limits the efficacy and durability of immune checkpoint inhibitors (ICIs) in hepatocellular carcinoma (HCC). Therefore, we conducted a retrospective cohort study to investigate the outcomes and characteristics of HCC patients with resistance to immunotherapy. Patients with HCC who have received ICIs at Eastern Hepatobiliary Surgery Hospital between 2016 and 2021 were retrospectively screened and divided into primary resistance, secondary resistance, and durable response group. Time to progression (TTP), overall survival (OS), subsequent management and post-progression survival (PPS) were analyzed. Of 496 patients included, 229 (46.2%) and 141 (28.4%) patients developed primary and secondary resistance, and 126 (25.4%) patients achieved a durable response, the median TTP was 2.83 [2.56-3.09] months, 11.93 [10.45-13.40] months, and not reached, respectively, whereas the median OS was 12.83 [10.36-15.30] months, 31.53 [28.09-34.97] and not reached, respectively. Multivariate logistic regression revealed that Child-Pugh score, BCLC stage, and combined systemic therapies (ICI plus bevacizumab or lenvatinib versus ICI monotherapy) were independently associated with primary resistance, and only combined systemic therapies (ICI plus bevacizumab versus ICI monotherapy) were independently associated with secondary resistance. AFP levels were independently associated with PPS in patients with primary resistance, while post-progression therapies (ICI-based therapies versus others) were independently associated with PPS in patients with resistance. The risk of resistance was notably lower in patients receiving the combination of ICI plus bevacizumab. High AFP levels were associated with the survival of patients with primary resistance. ICI-based maintenance therapy after resistance may provide a significant survival advantage for HCC patients.
Collapse
Affiliation(s)
- Xiaowen Cui
- Department of Oncology, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, Shanghai, China
| | - Minghao Ruan
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Yao Li
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Cheng Yang
- Department of Special Treatment I and Liver Transplantation, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, Shanghai, China
| | - Jin Zhang
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Riming Jin
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Dong Wu
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China
| | - Wen Sun
- National Center for Liver Cancer, The Naval Medical University, 800 Xiangyin Road, Shanghai, 200433, China.
| | - Ruoyu Wang
- The First Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, The Naval Medical University, 225 Changhai Road, Shanghai, 200438, China.
| |
Collapse
|
12
|
Ben Khaled N, Zarka V, Hobeika B, Schneider J, Rau M, Weich A, Leicht HB, Ye L, Piseddu I, Dill MT, Kandulski A, Pinter M, Ehmer U, Schirmacher P, Marquardt JU, Mayerle J, De Toni EN, Geier A, Reiter FP. Therapeutic Sequences of Systemic Therapy After Atezolizumab Plus Bevacizumab for Hepatocellular Carcinoma: Real-World Analysis of the IMMUreal Cohort. Aliment Pharmacol Ther 2025; 61:1755-1766. [PMID: 40181694 PMCID: PMC12074566 DOI: 10.1111/apt.70090] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 11/25/2024] [Accepted: 03/09/2025] [Indexed: 04/05/2025]
Abstract
BACKGROUND The introduction of several new systemic therapies in recent years has significantly altered the treatment landscape for advanced hepatocellular carcinoma. However, while the approval of the combination of atezolizumab and bevacizumab as the preferred first-line therapy over sorafenib represents progress, it has also raised uncertainties regarding optimal treatment sequencing for advanced disease. AIMS This study evaluates the sequential treatment of hepatocellular carcinoma following therapy with atezolizumab and bevacizumab, providing evidence from a prospective real-world cohort. METHODS Data were derived from the ongoing IMMUreal cohort, which investigates immunotherapy in hepatocellular carcinoma across two tertiary centres in Bavaria. A total of 124 patients treated with atezolizumab and bevacizumab as first-line therapy between June 2020 and December 2023 were analysed. Feasibility, treatment patterns, and outcomes of sequential therapy were assessed, with a focus on defined prognostic subgroups. RESULTS The median overall survival under real-world conditions was 19.8 months. Less than half of the patients (41.2%) proceeded to second-line therapy, and only 19.2% were eligible for third-line treatment. This decline in treatment eligibility corresponded to a marked reduction in therapy duration and progressive deterioration in liver function, as indicated by Albumin-Bilirubin and Child-Pugh scores. While patients with worse baseline liver function, such as patients with Child-Pugh B or ALBI > 1, had a significantly lower probability of transitioning to 2nd line therapy, no significant association was found between the number of treatment lines and factors such as liver cirrhosis, poor physical condition, extrahepatic disease, or macrovascular invasion. CONCLUSIONS Sequential therapy following atezolizumab and bevacizumab is feasible only for selected patients. However, preserving liver function seems crucial to optimising multi-line therapy and improving outcomes in advanced hepatocellular carcinoma.
Collapse
Affiliation(s)
- Najib Ben Khaled
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Valentina Zarka
- Division of Hepatology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| | - Bernard Hobeika
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Julia Schneider
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Monika Rau
- Division of Hepatology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| | - Alexander Weich
- Division of Gastroenterology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| | - Hans Benno Leicht
- Division of Hepatology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| | - Liangtao Ye
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
- Digestive Diseases CenterThe Seventh Affiliated Hospital, Sun Yat‐Sen UniversityShenzhenChina
| | - Ignazio Piseddu
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Michael T. Dill
- Department of Gastroenterology, Infectious Diseases and IntoxicationHeidelberg University HospitalHeidelbergGermany
- National Center for Tumor Diseases (NCT)NCT Heidelberg, a Partnership Between DKFZ and Heidelberg University HospitalHeidelbergGermany
- German Cancer Research Center (DKFZ) HeidelbergResearch Group Experimental Hepatology, Inflammation and CancerHeidelbergGermany
| | - Arne Kandulski
- Gastroenterology, Hepatology, Endocrinology, Rheumatology and Infectious Diseases, Department of Internal Medicine IUniversity Hospital RegensburgRegensburgGermany
| | - Matthias Pinter
- Division of Gastroenterology and Hepatology, Department of Medicine IIIMedical University of ViennaViennaAustria
| | - Ursula Ehmer
- Clinical Department for Internal Medicine II, Department of Clinical Medicine, TUM School of Medicine and Health, University Medical Center, Technical University of MunichMunichGermany
| | | | | | - Julia Mayerle
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Enrico N. De Toni
- Department of Medicine IIUniversity Hospital, LMU MunichMunichGermany
| | - Andreas Geier
- Division of Hepatology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| | - Florian P. Reiter
- Division of Hepatology, Department of Medicine IIUniversity Hospital WürzburgWürzburgGermany
| |
Collapse
|
13
|
Ng KYY, Teo AEK, Tan SH, Tan JJE, Tay DSH, Lee AWX, Ang AJS, Wong LWJ, Choo SP, Toh HC, Lee SY, Lee JJX, Tai DWM. Impact of Antibiotics and Chronic Medications on Efficacy of Immune Checkpoint Inhibitors in Patients With Hepatocellular Carcinoma. Asia Pac J Clin Oncol 2025; 21:256-265. [PMID: 39601254 DOI: 10.1111/ajco.14139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 08/29/2024] [Accepted: 10/28/2024] [Indexed: 11/29/2024]
Abstract
BACKGROUND AND AIMS The interaction of immune checkpoint inhibitors (ICI) and concomitant medications such as antibiotics, metformin, statins, beta-blockers, proton pump inhibitors (PPIs), nonsteroidal anti-inflammatory drugs (NSAIDs), and low-dose aspirin has been studied in other malignancies. Our study aims to investigate the relationship between these medications and ICI efficacy in patients with advanced hepatocellular carcinoma (aHCC). METHODS A retrospective review of patients who received at least one dose of ICIs between May 2015 and November 2019 was performed. The primary objectives were to compare the overall survival (OS) and progression-free survival (PFS) between patients with and without medication usage. Log rank test was used to assess for differences in survival. Hazard ratios were reported using Cox proportional hazard regression analysis. The data cutoff date was December 31, 2020. RESULTS A total of 168 patients were included. Median age was 69 years, 85.7% male, 60.7% ECOG 0, 78.0% Child-Pugh A liver cirrhosis, 57.7% hepatitis B etiology, 8.9% hepatitis C, and 33.3% nonviral. One hundred three patients (61.3%) received ICI monotherapy, while 38.7% received ICI in combination. Sixty-two patients (36.9%) had concomitant antibiotic usage, 26.8% metformin, 30.4% statin, 31.0% beta-blockers, 60.1% PPI, 6.5% NSAIDs, and 11.9% aspirin. Patients with aHCC receiving antibiotics did not have a shorter OS (adjusted HR [aHR] 1.40, 95% CI 0.94-2.09, p = 0.096) or shorter PFS (aHR 0.94, 95% CI 0.66-1.34, p = 0.73), as compared to those who did not receive antibiotics. However, patients with aHCC of viral hepatitis etiology receiving ICI treatment and concurrent antibiotics had shorter OS (5.5 vs. 14.2 months, aHR 1.93, 95% CI 1.17-3.17, p = 0.010) and PFS (1.1 vs. 2.6 months, aHR 2.69, 95% CI 1.28-5.65, p = 0.009), as compared to those who did not receive antibiotics. CONCLUSIONS The use of antibiotics may diminish ICI efficacy in patients with aHCC of viral hepatitis etiology, while the use of metformin, statins, beta-blockers, NSAIDs, and aspirin is not associated with significant clinical outcomes.
Collapse
Affiliation(s)
- Kennedy Yao Yi Ng
- Division of Population Health and Integrated Care, Singapore General Hospital, Singapore, Singapore
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| | | | - Sze Huey Tan
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
- Division of Clinical Trials and Epidemiological Sciences, National Cancer Centre Singapore, Singapore, Singapore
| | - Jack Jie En Tan
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Desiree Shu Hui Tay
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Ailica Wan Xin Lee
- Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Andrea Jing Shi Ang
- Division of Internal Medicine, Singapore General Hospital, Singapore, Singapore
| | | | | | - Han Chong Toh
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| | - Suat Ying Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| | - Joycelyn Jie Xin Lee
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| | - David Wai-Meng Tai
- Division of Medical Oncology, National Cancer Centre Singapore, Singapore, Singapore
- Oncology Academic Program, Duke-NUS Medical School, Singapore, Singapore
| |
Collapse
|
14
|
Collins AL, Kirkness K, Ramon-Gil E, Tzortzopoulou E, Geh D, Dishington J, Graham E, Muir R, Cameron R, Luli S, Khurram E, Storey D, Paish HL, Nelson G, McDonald D, Filby A, Borthwick LA, Oakley F, Mann DA, Leslie J. Precision-cut tumor slices for modeling hepatocellular carcinoma enable at-scale drug screening. Hepatol Commun 2025; 9:e0706. [PMID: 40377490 PMCID: PMC12088631 DOI: 10.1097/hc9.0000000000000706] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/11/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Disease modeling is vital for our understanding of disease mechanisms and for developing new therapeutic strategies. Accurately modeling the intact tumor microenvironment (TME) is increasingly recognized as essential for gaining insights into cancer biology and therapeutic response. Preclinical mouse models have provided utility for studying the evolving TME, but these models are costly and can lead to animal suffering and the discontinuation of drug investigations. To address these limitations, particularly in hepatocellular carcinoma (HCC), we have developed an ex vivo model using tumor precision-cut slices (TPCS) derived from orthotopic liver tumors. METHODS Murine HCC tumors were generated via intrahepatic injection of Hep-53.4 cells, providing a source of tumor tissue for TPCS generation. Subsequent scaling to a 96-well format and modification to include a secreted luciferase enabled longitudinal ex vivo screening of 26 drugs applied at 2 doses over an 8-day period, using just 5 tumors. One drug identified in the screen, salinomycin, was then validated in vivo via intraperitoneal injection of mice with orthotopic liver tumors. RESULTS Histological characterization determined that TPCS maintain the architecture, cellular complexity, and drug responsiveness of the original HCC-TME under simplified culture conditions that preserve viability and metabolic activity. In addition to typical HCC therapies, sorafenib and anti-PD1 immunotherapy, the screen identified 2 drugs as potent anticancer agents capable of impacting the viability of TPCS: salinomycin and rottlerin. Salinomycin was further validated in vivo, significantly reducing tumor burden without evidence of toxicity. CONCLUSIONS We present a 3Rs (Reduction, Refinement, Replacement) approach for studying HCC biology and performing 96-well-scale drug screening within an intact, metabolically active TME, offering a more ethical and effective platform for drug discovery.
Collapse
Affiliation(s)
- Amy L Collins
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Keara Kirkness
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Erik Ramon-Gil
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Eleni Tzortzopoulou
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Daniel Geh
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Jack Dishington
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Eleanor Graham
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Rhys Muir
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Rainie Cameron
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Saimir Luli
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
| | - Eman Khurram
- Newcastle University Medicine Malaysia, Iskandar Puteri, Malaysia
| | - Daniel Storey
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Hannah L. Paish
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Glyn Nelson
- Bioimaging Unit, Newcastle University, Newcastle upon Tyne, UK
| | - David McDonald
- Flow Cytometry Core Facility, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Andrew Filby
- Flow Cytometry Core Facility, Biosciences Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Lee A. Borthwick
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Fiona Oakley
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
| | - Derek A. Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
- FibroFind Ltd, William Leech Building, Medical School, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey
| | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
15
|
Yao Z, Ren Y, Cao M, Li Y, Su X, Hu Z, Han P, Yuen HK, Cheung TT. Comparative analysis of hepatectomy for HCC with PVTT: Insights from a 30-year single-center experience: Hepatectomy for HCC with PVTT. Surg Oncol 2025; 60:102211. [PMID: 40120185 DOI: 10.1016/j.suronc.2025.102211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/20/2025] [Accepted: 03/06/2025] [Indexed: 03/25/2025]
Abstract
BACKGROUND AND AIM Portal vein tumor thrombosis (PVTT) is frequent in hepatocellular carcinoma (HCC). Although hepatectomy is the primary treatment for HCC, no consensus exists on its role in PVTT between Eastern and Western clinicians. This study aims to assess the efficacy of hepatectomy in HCC patients with PVTT by analyzing perioperative outcomes and prognosis. METHODS This retrospective, single-center study reviewed HCC patient data from Queen Mary Hospital, Hong Kong (1989-2020). Propensity score matching (PSM) was applied to match patients with and without PVTT undergoing hepatectomy, comparing perioperative and survival outcomes between groups. RESULTS Among 3981 HCC patients, 1842 had PVTT and were not operated (not-operated group), while 2139 underwent hepatectomy. Of the operated patients, 156 had PVTT (PVTT group) and 1983 did not (no-PVTT group). Median overall survival (mOS) in the not-operated group was 2.7 months, compared to 13.0 months in the PVTT group. After 1:3 PSM, the no-PVTT group (n = 468) had longer mOS (47.0 vs. 13.0 months, p < 0.001) and disease-free survival (10.6 vs. 4.2 months, p < 0.001). The PVTT group had longer operative times (449 vs. 390 min, p < 0.001), higher complication rates (37.8 % vs. 28.2 %, p = 0.024), and closer surgical margins (0.6 vs. 1.0 cm, p = 0.036), but similar hospital mortality (p = 0.898). mOS for low-AFP (<17400 ng/ml) and high-AFP (≥17400 ng/ml) patients was 16.2 vs. 8.2 months, respectively (p < 0.001). CONCLUSION Aggressive treatment of PVTT is necessary. For certain PVTT patients, hepatectomy may be potentially effective, with acceptable perioperative safety and seemingly no technical barriers.
Collapse
Affiliation(s)
- Zhicheng Yao
- Department of Hepatobiliary & Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China; Department of Surgery, School of Clinical Medicine, The University of Hong Kong, 102 Pok Fu Lam Road, 999077, Hong Kong, China.
| | - Yupeng Ren
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Mingbo Cao
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Yuxuan Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Xiaorui Su
- Department of Hepatobiliary & Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Ziyi Hu
- Department of Hepatobiliary & Pancreatic Surgery, The Third Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510630, China.
| | - Pei Han
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, 102 Pok Fu Lam Road, 999077, Hong Kong, China.
| | - Ho Kam Yuen
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, 102 Pok Fu Lam Road, 999077, Hong Kong, China.
| | - Tan To Cheung
- Department of Surgery, School of Clinical Medicine, The University of Hong Kong, 102 Pok Fu Lam Road, 999077, Hong Kong, China.
| |
Collapse
|
16
|
Zhu J, Wang L, Nie X, Ou S, Shen J, Zhang S, Wu G. RBMS3-loss impedes TRIM21-induced ubiquitination of ANGPT2 in an RNA-independent manner and drives sorafenib resistance in hepatocellular carcinoma. Oncogene 2025; 44:1620-1633. [PMID: 40069332 DOI: 10.1038/s41388-025-03335-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 02/12/2025] [Accepted: 02/27/2025] [Indexed: 05/23/2025]
Abstract
Sorafenib, a first-line targeted drug for advanced hepatocellular carcinoma (HCC), has limited clinical application due to intrinsic/acquired resistance. In this study, we have identified the RNA-binding protein RBMS3 as a pivotal regulator involved in sorafenib resistance among patients with HCC. Loss- and gain-of-function experiments further demonstrate that downregulation of RBMS3 promotes angiogenesis and confers resistance to sorafenib by augmenting the capacity of HCC cells to express and secrete ANGPT2, while upregulation of RBMS3 reverse these phenotypes.Through immunoprecipitation mass spectrometry experiments and co-immunoprecipitation (co-IP), we further verified that RBMS3 can facilitate the K48-linked ubiquitination and subsequent protein degradation of ANGPT2 by recruiting the ubiquitin E3 ligase TRIM21 in an RNA-independent manner.Additionally, RBMS3 is found to be deleted in HCC tissues and exhibits a significant positive correlation with angiogenesis and resistance to sorafenib treatment. Importantly, the combination of ANGPT2 antibody in RBMS3-deficient HCC cells restores sensitivity to sorafenib both in vitro and in vivo. These findings uncovered a novel molecular basis for post-translational upregulation of ANGPT2, suggesting that RBMS3-loss plays an oncogenic role in HCC by promoting angiogenesis and conferring resistance to sorafenib treatment.
Collapse
Affiliation(s)
- Jinrong Zhu
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Lei Wang
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China
| | - Xiaoya Nie
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Shengming Ou
- Guangdong Provincial Key Laboratory of Advanced Drug Delivery, Guangdong Provincial Engineering Center of Topical Precise Drug Delivery System, School of Life Sciences and Biopharmaceutics, Guangdong Pharmaceutical University, Guangzhou, 510006, PR China
| | - Jianfei Shen
- Department of Cardiothoracic Surgery, Taizhou Hospital of Zhejiang Province affiliated to Wenzhou Medical University, Taizhou, PR China
| | - Shuxia Zhang
- Department of Oncobiology, Department of Basic Medical Sciences, Shantou University Medical College, Shantou, Guangdong, PR China.
- The Key Laboratory of Molecular Biology for High Cancer Incidence Coastal Chaoshan Area, Cancer Research Center, Shantou University Medical College, Shantou, Guangdong, PR China.
| | - Geyan Wu
- Biomedicine Research Centre, Guangdong Provincial Key Laboratory of Major Obstetric Diseases; Guangdong Provicial Clinical Research Center for Obsterics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou, PR China.
| |
Collapse
|
17
|
Zhu X, Li Z, Chen L, Li L, Ouyang M, Zhou H, Xiao K, Lin L, Chu PK, Zhou C, Xun C, Yang L, Huang W, Ding X. Exosomes delivering miR-129-5p combined with sorafenib ameliorate hepatocellular carcinoma progression via the KCTD1/HIF-1α/VEGF pathway. Cell Oncol (Dordr) 2025; 48:743-760. [PMID: 40227531 PMCID: PMC12119701 DOI: 10.1007/s13402-025-01044-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/31/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND Potassium channel tetramerization domain-containing 1 (KCTD1) plays a critical role in transcriptional regulation and adipogenesis, but its significance in hepatocellular cancer (HCC) has not been reported. METHODS Immunohistochemistry, Western blotting and quantitative real-time PCR analysis were performed to assess the expression of KCTD1 and related genes in HCC cells. MTT assays, colony formation, cell migration, invasion and the in-vivo mouse models were utilized to evaluate the function of KCTD1 in HCC progression. Co-immunoprecipitation, chromatin immunoprecipitation and luciferase reporter assays were conducted to elucidate the molecular mechanisms of KCTD1 in HCC. RESULTS KCTD1 expression was increased in human HCC tissues and closely associated with advanced tumor stages. KCTD1 overexpression enhanced growth, migration, and invasion of Huh7 and HepG2 cells both in vitro and in vivo, while KCTD1 knockdown reversed these effects in MHCC97H cells. Mechanistically, KCTD1 interacted with hypoxia-inducible factor 1 alpha (HIF-1α) and enhanced HIF-1α protein stability with the inhibited prolyl-hydroxylases (PHD)/Von Hippel-Lindau (VHL) pathway, consequently activating the Vascular Endothelial Growth Factor (VEGF)/VEGFR2 pathway in HCC cells. Sorafenib and KCTD1 knockdown synergistically inhibited intrahepatic tumor growth following in situ injection of MHCC97H cells. miR-129-5p downregulated KCTD1 by binding to KCTD1 3'UTR. Finally, 45 µg exosomes from miR-129-5p-overexpressing MHCC97H cells combined with 25 mg/kg sorafenib to decrease HCC tumor size. CONCLUSIONS These results suggested that KCTD1 protects HIF-1α from degradation and activates the VEGF signaling cascade to enhance HCC progression. Therefore, KCTD1 may serve as a novel target of HCC and pave the way for an efficient combined therapy in advanced HCC.
Collapse
Affiliation(s)
- Xinyu Zhu
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Zhiwei Li
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Li Chen
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Limin Li
- College of Engineering and Design, Hunan Normal University, Taozihu Road No. 68, Changsha, 410081, China.
| | - Mi Ouyang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Hao Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China
| | - Kai Xiao
- Department of Neurosurgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Ling Lin
- Department of Hepatobiliary and Pancreatic Surgery, Xiangya Hospital of Central South University, Changsha, Hunan, 410008, China
| | - Paul K Chu
- Department of Physics, Department of Materials Science and Engineering, and Department of Biomedical Engineering, City University of Hong Kong, Tat Chee Avenue, Kowloon, Hong Kong, China
| | - Chang Zhou
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Chengfeng Xun
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Liu Yang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Wenhuan Huang
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China
| | - Xiaofeng Ding
- The National & Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Science, Hunan Normal University, Lushan Road No. 14, Changsha, 410081, China.
- Key Laboratory of Model Animals and Stem Cell Biology in Hunan Province, School of Medicine, Hunan Normal University, Changsha, 410013, China.
| |
Collapse
|
18
|
Mohamed NM, Mohamed RH, Kennedy JF, Elhefnawi MM, Hamdy NM. A comprehensive review and in silico analysis of the role of survivin (BIRC5) in hepatocellular carcinoma hallmarks: A step toward precision. Int J Biol Macromol 2025; 311:143616. [PMID: 40306500 DOI: 10.1016/j.ijbiomac.2025.143616] [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: 02/13/2025] [Revised: 04/25/2025] [Accepted: 04/27/2025] [Indexed: 05/02/2025]
Abstract
Hepatocellular carcinoma (HCC) is a complex malignancy driven by the dysregulation of multiple cellular pathways. Survivin, a key member of the inhibitor of apoptosis (IAP) family, plays a central role in HCC tumorigenesis and progression. Despite significant research, a comprehensive understanding of the contributions of survivin to the hallmarks of cancer, its molecular network, and its potential as a therapeutic target remains incomplete. In this review, we integrated bioinformatics analysis with an extensive literature review to provide deeper insights into the role of survivin in HCC. Using bioinformatics tools such as the Human Protein Atlas, GEPIA, STRING, TIMER, and Metascape, we analyzed survivin expression and its functional associations and identified the top 20 coexpressed genes in HCC. These include TK1, SPC25, SGO2, PTTG1, PRR11, PLK1, NCAPH, KPNA2, KIF2C, KIF11, HJURP, GTSE1, FOXM1, CEP55, CENPA, CDCA3, CDC45, CCNB2, CCNB1 and CTD-2510F5.4. Our findings also revealed significant protein-protein interactions among these genes, which were enriched in pathways associated with the FOXM1 oncogenic signaling cascade, and biological processes such as cell cycle regulation, mitotic checkpoints, and diseases such as liver neoplasms. We also discussed the involvement of survivin in key oncogenic pathways, including the PI3K/AKT, WNT/β-catenin, Hippo, and JAK/STAT3 pathways, and its role in modulating cell cycle checkpoints, apoptosis, and autophagy. Furthermore, we explored its interactions with the tumor microenvironment, particularly its impact on immune modulation through myeloid-derived suppressor cells (MDSCs), tumor-associated macrophages, and natural killer cell function in HCC. Additionally, we highlighted its involvement in alkylglycerone phosphate synthase (AGPS)-mediated lipid reprogramming and identified important gaps in the survivin network that warrant further investigation. This review also examined the role of survivin in cancer stemness, inflammation, and virally mediated hepatocarcinogenesis. We evaluated its potential as a diagnostic, prognostic, predictive, and pharmacodynamic biomarker in HCC, emphasizing its relevance in precision medicine. Finally, we summarized emerging survivin-targeted therapeutics and ongoing clinical trials, underscoring the need for novel strategies to effectively target survivin in HCC.
Collapse
Affiliation(s)
- Nermin M Mohamed
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - Rania Hassan Mohamed
- Department of Biochemistry, Faculty of Science, Ain Shams University, Abassia, 11566 Cairo, Egypt
| | - John F Kennedy
- Chembiotech Laboratories, Kyrewood House, Tenbury Wells, Worcestershire, United Kingdom
| | - Mahmoud M Elhefnawi
- Biomedical Informatics and Chemoinformatics Group, Informatics and Systems Department, National Research Centre, Cairo, Egypt.
| | - Nadia M Hamdy
- Department of Biochemistry, Faculty of Pharmacy, Ain Shams University, Abassia, 11566 Cairo, Egypt.
| |
Collapse
|
19
|
Molinelli AR, Cross SJ, Leggas M. Recent Advances in Therapeutic Drug Monitoring of Antineoplastic and Antimicrobial Agents in Children. Clin Lab Med 2025; 45:315-327. [PMID: 40348442 DOI: 10.1016/j.cll.2025.01.014] [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: 05/14/2025]
Abstract
Therapeutic drug monitoring (TDM) is used to optimize drug therapy by ensuring efficacy or preventing toxicity. For a limited number of cytotoxic antineoplastic drugs, for aminoglycoside antibiotics, and for vancomycin the use of TDM is common practice. In this article, we summarize recent advances and indications for the TDM of antineoplastic agents in children, focusing on protein kinase inhibitors and the cytotoxic drug fludarabine. We also summarize recent recommendations for antimicrobial TDM of beta-lactam antibiotics and vancomycin.
Collapse
Affiliation(s)
- Alejandro R Molinelli
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop #150, Memphis, TN 38105, USA.
| | - Shane J Cross
- Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop #150, Memphis, TN 38105, USA. https://twitter.com/shane6cross
| | - Markos Leggas
- Center for Translational Pharmacology, Department of Pharmacy and Pharmaceutical Sciences, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Mail Stop 313, I-5104, Memphis, TN 38105, USA
| |
Collapse
|
20
|
Aoki T, Kudo M, Nishida N, Ueshima K, Tsuchiya K, Tada T, Morita M, Chishina H, Takita M, Hagiwara S, Ida H, Minami Y, Kuroda H, Nakamura N, Hiraoka A, Tomonari T, Tani J, Naganuma A, Kakizaki S, Ogawa C, Hatanaka T, Ishikawa T, Kawata K, Takebe A, Matsumoto I, Hidaka M, Kurosaki M, Kumada T, Izumi N. Proposal of discontinuation criteria of atezolizumab plus bevacizumab after curative conversion therapy for unresectable early-to-intermediate-stage hepatocellular carcinoma: a multicenter proof-of-concept study. J Gastroenterol 2025; 60:738-753. [PMID: 40055288 PMCID: PMC12095402 DOI: 10.1007/s00535-025-02233-z] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/29/2024] [Accepted: 02/18/2025] [Indexed: 04/02/2025]
Abstract
BACKGROUND Achieving complete response (CR) is a desirable goal in early-to-intermediate-stage hepatocellular carcinoma (HCC). While systemic and locoregional therapies show promise, optimal drug discontinuation criteria remain unclear. This study aims to investigate drug-off criteria for atezolizumab plus bevacizumab as a proof-of-concept study. METHODS This retrospective multicenter study included child-pugh class A patients with unresectable HCC without extrahepatic spread or macrovascular invasion who received atezolizumab plus bevacizumab as first-line therapy. Modified clinical CR (mCCR) was defined as CR per mRECIST with sustained normal alpha-fetoprotein (AFP) levels (< 10.0 ng/dl). Recurrence-free survival (RFS) and overall survival (OS) were analyzed based on the "drug-off" criteria defined by following: (1) mRECIST CR with locoregional therapies, (2) sustained normalization of AFP/AFP-L3/ des-gamma-carboxy prothrombin (DCP) for 12-24 weeks, and (3) complete tumor vascularity disappearance by contrast-enhanced ultrasonography (CEUS) or pathological curative resection. RESULTS The median follow-up was 16.5 months (95% CI 15.2-17.8). Among 51 patients achieving mCCR, 11 underwent surgery, with pathological CR in three cases. In contrast, viable lesions were observed in 7 of 40 cases assessed using CEUS. All patients meeting the drug-off criteria (n = 9) showed no recurrence and none of them experienced mortality, while 45.2% (19/42) of those not meeting the criteria experienced recurrence (median RFS: 12.8 months, p = 0.007). The median OS was not reached in dug-off criteria met patients (n = 9), 37.7 months (95% CI: NA) in non-criteria met patients (n = 42), and 27.1 months (95% CI 16.7-37.6) in non-mCCR patients (n = 184) (p < 0.001). CONCLUSION In patients with unresectable and TACE-unsuitable early-to-intermediate-stage HCC who met the drug-off criteria, significantly improved RFS and OS were observed compared those who did not meet the criteria. However, further validation studies are required to confirm the utility of the criteria.
Collapse
Affiliation(s)
- Tomoko Aoki
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan.
| | - Naoshi Nishida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Kazuomi Ueshima
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Kaoru Tsuchiya
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Toshifumi Tada
- Department of Internal Medicine, Japanese Red Cross Himeji Hospital, Himeji, Japan
| | - Masahiro Morita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Hirokazu Chishina
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Masahiro Takita
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Satoru Hagiwara
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Hiroshi Ida
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Yasunori Minami
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, 377-2 Ohno-Higashi, Osaka-Sayama, 589-8511, Japan
| | - Hidekatsu Kuroda
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Iwate Medical University, Iwate, Japan
| | - Noriaki Nakamura
- Department of General Surgery, Shuuwa General Hospital, Saitama, Japan
| | - Atsushi Hiraoka
- Gastroenterology Center, Ehime Prefectural Central Hospital, Matsuyama, Japan
| | - Tetsu Tomonari
- Department of Gastroenterology and Oncology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Joji Tani
- Department of Gastroenterology and Neurology, Kagawa University, Kagawa, Japan
| | - Atsushi Naganuma
- Department of Gastroenterology, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Satoru Kakizaki
- Department of Clinical Research, NHO Takasaki General Medical Center, Takasaki, Japan
| | - Chikara Ogawa
- Department of Gastroenterology and Hepatology, Takamatsu Red Cross Hospital, Takamatsu, Japan
| | - Takeshi Hatanaka
- Department of Gastroenterology, Gunma Saiseikai Maebashi Hospital, Maebashi, Japan
| | - Toru Ishikawa
- Department of Gastroenterology, Saiseikai Niigata Hospital, Niigata, Japan
| | - Kazuhito Kawata
- Hepatology Division, Department of Internal Medicine II, Hamamatsu University School of Medicine, Hamamatsu, Japan
| | - Atsushi Takebe
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Ippei Matsumoto
- Department of Surgery, Kindai University Faculty of Medicine, Osaka, Japan
| | - Masaaki Hidaka
- Department of Surgery, Nagasaki University Graduate School of Biomedical Sciences, Nagasaki, Japan
| | - Masayuki Kurosaki
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| | - Takashi Kumada
- Department of Nursing, Gifu Kyoritsu University, Ogaki, Japan
| | - Namiki Izumi
- Department of Gastroenterology and Hepatology, Musashino Red Cross Hospital, Tokyo, Japan
| |
Collapse
|
21
|
Cappuyns S, Piqué-Gili M, Esteban-Fabró R, Philips G, Balaseviciute U, Pinyol R, Gris-Oliver A, Vandecaveye V, Abril-Fornaguera J, Montironi C, Bassaganyas L, Peix J, Zeitlhoefler M, Mesropian A, Huguet-Pradell J, Haber PK, Figueiredo I, Ioannou G, Gonzalez-Kozlova E, D'Alessio A, Mohr R, Meyer T, Lachenmayer A, Marquardt JU, Reeves HL, Edeline J, Finkelmeier F, Trojan J, Galle PR, Foerster F, Mínguez B, Montal R, Gnjatic S, Pinato DJ, Heikenwalder M, Verslype C, Van Cutsem E, Lambrechts D, Villanueva A, Dekervel J, Llovet JM. Single-cell RNA sequencing-derived signatures define response patterns to atezolizumab + bevacizumab in advanced hepatocellular carcinoma. J Hepatol 2025; 82:1036-1049. [PMID: 39709141 PMCID: PMC12086051 DOI: 10.1016/j.jhep.2024.12.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Revised: 11/29/2024] [Accepted: 12/07/2024] [Indexed: 12/23/2024]
Abstract
BACKGROUND & AIMS The combination of atezolizumab and bevacizumab (atezo+bev) is the current standard of care for advanced hepatocellular carcinoma (HCC), providing a median overall survival (OS) of 19.2 months. Here, we aim to uncover the underlying cellular processes driving clinical benefit vs. resistance to atezo+bev. METHODS We harnessed the power of single-cell RNA sequencing in advanced HCC to derive gene expression signatures recapitulating 21 cell phenotypes. These signatures were applied to 422 RNA-sequencing samples of patients with advanced HCC treated with atezo+bev (n = 317) vs. atezolizumab (n = 47) or sorafenib (n = 58) as comparators. RESULTS We unveiled two distinct patterns of response to atezo+bev. First, an immune-mediated response characterised by the combined presence of CD8+ T effector cells and pro-inflammatory CXCL10+ macrophages, representing an immune-rich microenvironment. Second, a non-immune, angiogenesis-related response distinguishable by a reduced expression of the VEGF co-receptor neuropilin-1 (NRP1), a biomarker that specifically predicts improved OS upon atezo+bev vs. sorafenib (p = 0.039). Primary resistance was associated with an enrichment of immunosuppressive myeloid populations, namely CD14+ monocytes and TREM2+ macrophages, and Notch pathway activation. Based on these mechanistic insights we define "Immune-competent" and "Angiogenesis-driven" molecular subgroups, each associated with a significantly longer OS with atezo+bev vs. sorafenib (p of interaction = 0.027), and a "Resistant" subset. CONCLUSION Our study unveils two distinct molecular subsets of clinical benefit to atezolizumab plus bevacizumab in advanced HCC ("Immune-competent" and "Angiogenesis-driven") as well as the main traits of primary resistance to this therapy, thus providing a molecular framework to stratify patients based on clinical outcome and guiding potential strategies to overcome resistance. IMPACT AND IMPLICATIONS Atezolizumab + bevacizumab (atezo+bev) is standard of care in advanced hepatocellular carcinoma (HCC), yet molecular determinants of clinical benefit to the combination remain unclear. This study harnesses the power of single-cell RNA sequencing, deriving gene expression signatures representing 21 cell subtypes in the advanced HCC microenvironment. By applying these signatures to RNA-sequencing samples, we reveal two distinct response patterns to atezo+bev and define molecular subgroups of patients ("Immune-competent" and "Angiogenesis-driven" vs. "Resistant") with differential clinical outcomes upon treatment with atezo+bev, pointing towards the role of immunosuppressive myeloid cell types and Notch pathway activation in primary resistance to atezo+bev. These results may help refine treatment strategies and improve outcomes for patients with advanced HCC, while also guiding future research aimed at overcoming resistance mechanisms.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Bevacizumab/administration & dosage
- Bevacizumab/therapeutic use
- Male
- Female
- Single-Cell Analysis/methods
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Sequence Analysis, RNA/methods
- Middle Aged
- Tumor Microenvironment
- Drug Resistance, Neoplasm/genetics
- Sorafenib
Collapse
Affiliation(s)
- Sarah Cappuyns
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium; Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Centre for Cancer Biology, Leuven, Belgium; Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Marta Piqué-Gili
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Roger Esteban-Fabró
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Gino Philips
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Centre for Cancer Biology, Leuven, Belgium
| | - Ugne Balaseviciute
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Roser Pinyol
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Albert Gris-Oliver
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Vincent Vandecaveye
- Radiology Department, University Hospitals Leuven, Leuven, Belgium; Laboratory of Translational MRI, Department of Imaging and Pathology, KU Leuven, Leuven, Belgium
| | - Jordi Abril-Fornaguera
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Carla Montironi
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Pathology Department and Molecular Biology Core, Hospital Clínic of Barcelona, Barcelona, Spain
| | - Laia Bassaganyas
- Institut de Génomique Fonctionnelle, Univ. Montpellier, CNRS, INSERM, Montpellier, France
| | - Judit Peix
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Marcus Zeitlhoefler
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Agavni Mesropian
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Júlia Huguet-Pradell
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain
| | - Philipp K Haber
- Department of Surgery, Campus Charité Mitte and Campus Virchow-Klinikum, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Igor Figueiredo
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Giorgio Ioannou
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Edgar Gonzalez-Kozlova
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Antonio D'Alessio
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London, UK
| | - Raphael Mohr
- Department of Hepatology and Gastroenterology, Charité - Universitätsmedizin Berlin, Campus Virchow Klinikum (CVK) and Campus Charité Mitte (CCM), Berlin, Germany
| | - Tim Meyer
- Research Department of Oncology, UCL Cancer Institute, University College London, Royal Free Hospital, London, UK
| | - Anja Lachenmayer
- Department of Visceral Surgery and Medicine, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jens U Marquardt
- Department of Medicine I, University Medical Center Schleswig Holstein Campus Lübeck, Lübeck, Germany
| | - Helen L Reeves
- Hepatopancreatobiliary Multidisciplinary Team, Newcastle upon Tyne NHS Foundation Trust, Freeman Hospital, Newcastle upon Tyne, UK; Newcastle University Translational and Clinical Research Institute and Newcastle University Centre for Cancer, Medical School, Framlington Place, Newcastle Upon Tyne, NE2 4HH, UK
| | - Julien Edeline
- Department of Medical Oncology, Centre Eugène Marquis, Rennes, France
| | - Fabian Finkelmeier
- Department of Gastroenterology, University Liver and Cancer Centre, Frankfurt, Germany
| | - Jörg Trojan
- Department of Gastroenterology, University Liver and Cancer Centre, Frankfurt, Germany
| | - Peter R Galle
- Department of Medicine I, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Friedrich Foerster
- Department of Medicine I, University Medical Center of the Johannes-Gutenberg University, Mainz, Germany
| | - Beatriz Mínguez
- Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Liver Diseases Research Group, Vall d'Hebron Institute of Research (VHIR), Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; CIBERehd, Universitat Autònoma de Barcelona, Barcelona, Spain
| | - Robert Montal
- Department of Medical Oncology, Cancer Biomarkers Research Group, Hospital Universitari Arnau de Vilanova, IRBLleida, University of Lleida (UdL), Catalonia, Spain
| | - Sacha Gnjatic
- Department of Immunology and Immunotherapy, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, London, UK; Department of Translational Medicine, Università Del Piemonte Orientale "A. Avogadro", Novara, Italy
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Chris Verslype
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Eric Van Cutsem
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium; VIB Centre for Cancer Biology, Leuven, Belgium
| | - Augusto Villanueva
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Jeroen Dekervel
- Digestive Oncology, Department of Gastroenterology, University Hospitals Leuven, Leuven, Belgium; Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium.
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program (Divisions of Liver Diseases, Department of Hematology/Oncology, Department of Medicine), Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Catalonia, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Catalonia, 08010, Spain.
| |
Collapse
|
22
|
Li J, Wang Z, Ma L, Gou J, Feng Y, Lou Y, Zuo L, Wang T, Liang Y, Zhang Y, Wang E, Bai Y. Identification of transarterial chemoembolization candidates in advanced hepatocellular carcinoma patients classified solely by performance status 1: a multicenter retrospective study. Sci Rep 2025; 15:18792. [PMID: 40442090 PMCID: PMC12122698 DOI: 10.1038/s41598-025-00344-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Accepted: 04/28/2025] [Indexed: 06/02/2025] Open
Abstract
The Barcelona Clinic Liver Cancer (BCLC) advanced stage of hepatocellular carcinoma (HCC) includes a heterogeneous population, and new patient-tailored therapeutic indications are needed. Emerging evidence suggest that patients in this stage with mild tumor-related symptoms may benefit from more aggressive treatments including transarterial chemoembolization (TACE) and obtain better outcomes. This study aimed to investigate the effects of TACE on HCC patients with mild tumor-related symptoms and risk-stratify them for selecting potential candidates for TACE. We retrospectively collected data from 745 patients with liver-confined HCC undergoing TACE at 15 different centers from January 2015 to November 2022. The prognostic abilities of performance status (PS score of 0 vs. 1) were separately evaluated in high- and low-risk groups using the Hepatoma Arterial-embolization Prognostic (HAP) scoring model and its variants. PS1 remained an independent prognostic factor for overall survival (OS) in the whole cohort (P = 0.035). Interestingly, it lost its prognostic value for patients in low-risk groups (grade A + B) in all the four HAP models. This population with PS1 alone achieved similar OS to their counterparts with PS0. Risk stratification based on HAP scoring models could discriminate potential candidates from HCC patients in BCLC-C stage with PS1 alone. These patients could be classified into BCLC-B stage and benefit from TACE treatment.
Collapse
Affiliation(s)
- Jing Li
- Department of Digestive Diseases, Shanxi Bethune Hospital, Shanxi Academy of Medical Science, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan, China
- Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Shanxi Academy of Advanced Research and Innovation, Taiyuan, China
| | - Zhexuan Wang
- Department of Hepatobiliary Surgery, General Hospital of Eastern Theater Command, Nanjing, China
| | - Litian Ma
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, China
| | - Jiakun Gou
- Department of Digestive Diseases, Air Force Hospital of Western Theater Command, No. 1 Gongnongyuan Road, Jinjiang District, ChengduSichuan, 610000, China
| | - Yunan Feng
- Department of Digestive Diseases, Air Force Hospital of Western Theater Command, No. 1 Gongnongyuan Road, Jinjiang District, ChengduSichuan, 610000, China
| | - Yanju Lou
- Department of Orthopedic Surgery, Air Force Hospital of Western Theater Command, Chengdu, China
| | - Luo Zuo
- Department of Digestive Diseases, The Second Affiliated Hospital of Chengdu Medical College, Chengdu, China
| | - Tao Wang
- Department of Interventional Radiology, The Affiliated Yantai Yuhuangding Hospital of Qingdao University, Yantai, Shandong, China
| | - Yong Liang
- Department of Neurosurgery, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, 110015, Liaoning, China
| | - Yongchao Zhang
- Department of Digestive Diseases, Air Force Hospital of Western Theater Command, No. 1 Gongnongyuan Road, Jinjiang District, ChengduSichuan, 610000, China
| | - Enxin Wang
- Department of Digestive Diseases, Air Force Hospital of Western Theater Command, No. 1 Gongnongyuan Road, Jinjiang District, ChengduSichuan, 610000, China.
| | - Yang Bai
- Department of Gastroenterology, Tangdu Hospital, Air Force Medical University, Xi'an, China.
- Department of Neurosurgery, General Hospital of Northern Theater Command, No. 83 Wenhua Road, Shenhe District, Shenyang, 110015, Liaoning, China.
| |
Collapse
|
23
|
Lu Y, Lin J, Lu Y, Lin L, Zheng S, Chen Y, Huang S. Hepatotoxicity of ICI monotherapy or combination therapy in HCC: A systematic review and meta-analysis. PLoS One 2025; 20:e0323023. [PMID: 40440305 PMCID: PMC12121757 DOI: 10.1371/journal.pone.0323023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Accepted: 04/01/2025] [Indexed: 06/02/2025] Open
Abstract
BACKGROUND The aim of this study was to reveal the hepatotoxicity profile of different immune checkpoint inhibitor (ICI) used strategies in patients with Hepatocellular carcinoma (HCC) by meta-analysis. METHODS Literature was searched through PubMed, Cochrane, Embase, and Web of Science up to October 14, 2023, and the subject terms were "Carcinoma, Hepatocellular" and "Immune Checkpoint Inhibitors". The main observations were alanine aminotransferase (ALT) and aspartate aminotransferase (AST). ALT and AST were graded according to CTCAE. RESULTS A total of 32 studies with 7662 patients were included in the analysis. The results of meta-analysis showed that among different ICI treatment regimens, ICI monotherapy had the lowest incidence of any grade of ALT and AST elevation, and the highest for ICI+multikinase inhibitor (MKI); ICI+anti-VEGFR/VEGFA and ICI monotherapy had a lower incidence of grade ≥3 ALT and AST elevations, while ICI + MKI, dual immunotherapy, and dual immunotherapy+MKI had a higher incidence of grade ≥3 ALT and AST elevations; ICI monotherapy was more prone to any grade ALT elevation than placebo, and ICI monotherapy was more prone to ≥ 3 grade AST elevation than MKI; combination immunotherapy was more prone than MKI to any grade ALT and AST elevations; in grade ≥3 ALT and AST elevations, combination immunotherapy was similar to ICI monotherapy and MKI; ICI + MKI was more likely to have grade ≥3 ALT. CONCLUSION ICI monotherapy was more likely to cause severe hepatotoxicity than MKI. Combination immunotherapy treatment increased the incidence of hepatotoxicity compared to monotherapy, and ICI + MKI was prone to develop severe hepatotoxicity.
Collapse
Affiliation(s)
- Yuping Lu
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, PR China
| | - Jing Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, PR China
| | - Yufeng Lu
- School of Mathematics and Computer Science, Fuzhou University, Fujian, PR China
| | - Luping Lin
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, PR China
| | - Shicheng Zheng
- School of Basic Medical Sciences, Fujian Medical University, Fujian, PR China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, PR China
| | - Sha Huang
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fujian, PR China
| |
Collapse
|
24
|
Xie X, Wang Y, Wang Z, Zhang L, Li J, Li Y. Hepatocellular carcinoma drug resistance models. Cancer Cell Int 2025; 25:195. [PMID: 40437577 PMCID: PMC12121111 DOI: 10.1186/s12935-025-03821-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Accepted: 05/08/2025] [Indexed: 06/01/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the second leading cause of cancer-related death worldwide. Although drug therapy has been well developed and applied, its clinical efficacy is limited due to primary or acquired drug resistance in most HCC patients. Therefore, it is of great clinical significance to elucidate the key molecular mechanisms of resistance and improve the sensitivity of HCC cells to drugs. At present, a variety of HCC drug resistance models have been developed to find out resistance mechanisms, screen biomarkers, and explore strategies to reverse drug resistance, including traditional HCC drug resistance models, HCC patient-derived drug resistance models, three-dimensional drug resistance models, transgenic drug resistance models, and multi-drug resistance models. Here, we searched PubMed, Embase and Web of science for studies related to HCC drug resistance models in recent years, systematically summarized the established methods and characteristics of these models, reviewed their applications and compared their advantages and disadvantages, aiming to provide reference for the selection of appropriate models for HCC drug resistance research.
Collapse
Affiliation(s)
- Xiaolu Xie
- Department of Pharmacy, Yibin Hospital Affiliated to Children's Hospital of Chongqing Medical University, No. 108, Shangmao road, Xuzhou district, Yibin, Sichuan, China
| | - Yaomin Wang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Ziyi Wang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Lei Zhang
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Jun Li
- Department of Traditional Chinese Medicine, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
25
|
Qian S, He Y, Li R, Sun P, Zhang X, Pan L, Xu Z, Feng Z, Lian R, Yu L. Polymeric immunoglobulin receptor (pIgR) in cancer progression: a critical role and potential therapeutic target. Apoptosis 2025:10.1007/s10495-025-02116-x. [PMID: 40415061 DOI: 10.1007/s10495-025-02116-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/07/2025] [Indexed: 05/27/2025]
Abstract
Polymeric immunoglobulin receptor (pIgR) is a crucial receptor that primarily mediates the transcytosis of immunoglobulins A and M across epithelial cells, emerging as an essential participant in modulating both mucosal immunity and innate immunity. Recently, pIgR dysregulation in cancer has garnered widespread attention. It exhibits distinct mechanisms and effects across various cancer types with significant clinical value as a biomarker for malignant tumor diagnosis and prognosis evaluation. Recent therapeutic advances have revealed promising strategies, including dimeric IgA-based approaches targeting intracellular oncogenic drivers through pIgR-mediated transcytosis, small molecule modulators such as bufalin, and targeting EV-pIgR with neutralizing antibodies. Integrating these approaches with conventional therapies presents opportunities for enhanced treatment efficacy. Specifically, blocking EV-pIgR with neutralizing antibodies, when integrated with conventional hepatocellular carcinoma therapies such as sorafenib or other therapeutic agents, or a dIgA-targeting approach combined with immune checkpoint inhibitors, may enhance treatment efficacy. This review also addresses current challenges and future directions in pIgR-targeted cancer therapy, emphasizing the need for a deeper understanding of pIgR's regulatory mechanisms. These insights reveal that pIgR is an emerging therapeutic target with significant potential for the development of novel cancer treatment strategies.
Collapse
Affiliation(s)
- Shaoju Qian
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Yeqing He
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
| | - Ruixue Li
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Panpan Sun
- Department of Pathology, The Third Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Xingyi Zhang
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
| | - Lin Pan
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
| | - Zhishan Xu
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Zhiwei Feng
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan, 453003, China
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China
| | - Rong Lian
- Department of Otolaryngology, The First Affiliated Hospital of Xinxiang Medical University, Xinxiang, 453003, People's Republic of China.
| | - Lili Yu
- School of Basic Medical Sciences, Xinxiang Medical University, #601 Jinsui Road, Xinxiang, 453003, Henan, People's Republic of China.
- Xinxiang Engineering Technology Research Center of Immune Checkpoint Drug for Liver-Intestinal Tumors, Henan, 453003, China.
- Xinxiang Key Laboratory of Tumor Vaccine and Immunotherapy, School of Basic Medical Sciences, Xinxiang Medical University, Xinxiang, 453003, People's Republic of China.
| |
Collapse
|
26
|
Mo PL, Lin M, Gao BW, Zhang SB, Chen JP. Knowledge structure analysis and network visualization of tumor-associated macrophages in hepatocellular carcinoma research: A bibliometric mapping. World J Clin Oncol 2025; 16:102747. [DOI: 10.5306/wjco.v16.i5.102747] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 02/13/2025] [Accepted: 04/11/2025] [Indexed: 05/19/2025] Open
Abstract
BACKGROUND Tumor-associated macrophages (TAMs) have demonstrated significant potential as a research and treatment approach for hepatocellular carcinoma (HCC). Nevertheless, a comprehensive quantitative analysis of TAMs in HCC remained insufficient. Therefore, the objective of this study was to employ bibliometric methods to investigate the development trends and research frontiers pertaining to this field.
AIM To determine the knowledge structure and current research hotspots by bibliometric analysis of scholarly papers pertaining to TAMs in HCC.
METHODS The present study employed the Web of Science Core Collection to identify all papers related to TAMs in HCC research. Utilizing the Analysis Platform of Bibliometrics, CiteSpace 6.2.R4, and Vosviewer 1.6.19, the study conducted a comprehensive analysis encompassing multiple dimensions such as publication quantity, countries of origin, affiliated institutions, publishing journals, contributing authors, co-references, author keywords, and emerging frontiers within this research domain.
RESULTS A thorough examination was undertaken on 818 papers within this particular field, published between January 1, 1985 to September 1, 2023, which has witnessed a substantial surge in scholarly contributions since 2012, with a notable outbreak in 2019. China was serving as the central hub in this field, with Fudan University leading in terms of publications and citations. Chinese scholars have taken the forefront in driving the research expansion within this field. Hepatology emerged as the most influential journal in this field. The study by Qian and Pollard in 2010 received the highest number of co-citations. It was observed that the citation bursts of references coincided with the outbreak of publications. Notably, “tumor microenvironment”, “immunotherapy”, “prognostic”, “inflammation”, and “polarization”, etc. emerged as frequently occurring keywords in this field. Of particular interest, “immune evasion”, “immune infiltration”, and “cancer genome atlas” were identified as emerging frontiers in recent research.
CONCLUSION The field of TAMs in HCC exhibited considerable potential, as evidenced by the promising prospects of immunotherapeutic interventions targeting TAMs for the amelioration of HCC. The emerging frontiers in this field primarily revolved around modulating the immunosuppressive characteristics of TAMs within a liver-specific immune environment, with a focus on how to counter immune evasion and reduce immune infiltration.
Collapse
Affiliation(s)
- Ping-Li Mo
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Ming Lin
- Department of Hepatology, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510120, Guangdong Province, China
| | - Bo-Wen Gao
- Department of Traditional Chinese Medicine, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, Guangdong Province, China
| | - Shang-Bin Zhang
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| | - Jian-Ping Chen
- Shenzhen Key Laboratory of Hospital Chinese Medicine Preparation, Shenzhen Traditional Chinese Medicine Hospital, The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen 518033, Guangdong Province, China
| |
Collapse
|
27
|
Yau T, Galle PR, Decaens T, Sangro B, Qin S, da Fonseca LG, Karachiwala H, Blanc JF, Park JW, Gane E, Pinter M, Peña AM, Ikeda M, Tai D, Santoro A, Pizarro G, Chiu CF, Schenker M, He A, Chon HJ, Wojcik-Tomaszewska J, Verset G, Wang QQ, Stromko C, Neely J, Singh P, Jimenez Exposito MJ, Kudo M, CheckMate 9DW investigators. Nivolumab plus ipilimumab versus lenvatinib or sorafenib as first-line treatment for unresectable hepatocellular carcinoma (CheckMate 9DW): an open-label, randomised, phase 3 trial. Lancet 2025; 405:1851-1864. [PMID: 40349714 DOI: 10.1016/s0140-6736(25)00403-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/21/2025] [Accepted: 02/26/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Patients with unresectable hepatocellular carcinoma have a poor prognosis, and treatments with long-term benefits are needed. We report results from the preplanned interim analysis of the CheckMate 9DW trial assessing nivolumab plus ipilimumab versus lenvatinib or sorafenib for unresectable hepatocellular carcinoma in the first-line setting. METHODS This open-label, randomised, phase 3 trial enrolled patients aged 18 years or older with unresectable hepatocellular carcinoma without previous systemic therapy at 163 hospitals and cancer centres across 25 countries in Asia, Australia, Europe, North America, and South America. Patients had at least one measurable untreated lesion per Response Evaluation Criteria in Solid Tumours (RECIST) version 1.1, a Child-Pugh score of 5 or 6, and an Eastern Cooperative Oncology Group performance status of 0 or 1. Patients were randomly assigned (1:1) via an interactive response technology system to receive nivolumab (1 mg/kg) plus ipilimumab (3 mg/kg) intravenously every 3 weeks for up to four doses, followed by nivolumab 480 mg every 4 weeks or investigator's choice of either oral lenvatinib (8 mg or 12 mg mg daily depending on bodyweight) or oral sorafenib (400 mg twice daily). Randomisation was stratified by aetiology; the presence of macrovascular invasion, extrahepatic spread, or both; and baseline alpha-fetoprotein concentration. The primary endpoint was overall survival, which was assessed in all randomly assigned patients; safety was an exploratory endpoint and was assessed in all randomly assigned patients who received at least one dose of study medication. This trial is registered with ClinicalTrials.gov, NCT04039607 (ongoing). FINDINGS Between Jan 6, 2020, and Nov 8, 2021, 668 patients were randomly assigned to nivolumab plus ipilimumab (n=335) or lenvatinib or sorafenib (n=333). Early crossing of the overall survival Kaplan-Meier curves reflected a higher number of deaths during the first 6 months after randomisation with nivolumab plus ipilimumab (hazard ratio 1·65 [95% CI 1·12-2·43]) but was followed by a sustained separation of the curves thereafter in favour of nivolumab plus ipilimumab (0·61 [0·48-0·77]). After a median follow-up of 35·2 months (IQR 31·1-39·9), overall survival was significantly improved with nivolumab plus ipilimumab versus lenvatinib or sorafenib (median 23·7 months [95% CI 18·8-29·4] vs 20·6 months [17·5-22·5]; hazard ratio 0·79 [0·65-0·96]; two-sided stratified log-rank p=0·018); respective overall survival rates were 49% (95% CI 44-55) versus 39% (34-45) at 24 months and 38% (32-43) versus 24% (19-30) at 36 months. Overall, 137 (41%) of 332 patients receiving nivolumab plus ipilimumab and 138 (42%) of 325 patients receiving lenvatinib or sorafenib had grade 3-4 treatment-related adverse events. 12 deaths were attributed to treatment with nivolumab plus ipilimumab and three were attributed to treatment with lenvatinib or sorafenib. INTERPRETATION Nivolumab plus ipilimumab showed a significant overall survival benefit versus lenvatinib or sorafenib and manageable safety in patients with previously untreated unresectable hepatocellular carcinoma. These results support nivolumab plus ipilimumab as a first-line treatment in this setting. FUNDING Bristol Myers Squibb.
Collapse
Affiliation(s)
- Thomas Yau
- Centre of Cancer Medicine and University Department of Medicine, The University of Hong Kong, Hong Kong
| | - Peter R Galle
- University Medical Center, I Medical Department, Mainz, Germany.
| | - Thomas Decaens
- University of Grenoble Alpes, CHU Grenoble Alpes, Institute for Advanced Biosciences, CNRS UMR 5309-INSERM U1209, Grenoble, France
| | - Bruno Sangro
- Clinica Universidad de Navarra and CIBEREHD, Pamplona-Madrid, Spain
| | - Shukui Qin
- Nanjing Tianyinshan Hospital of China Pharmaceutical University, Nanjing, China
| | - Leonardo G da Fonseca
- Instituto do Cancer do Estado de São Paulo, ICESP, University of São Paulo School of Medicine, São Paulo, Brazil
| | | | | | - Joong-Won Park
- National Cancer Center and Myongji Hospital, Goyang, South Korea
| | - Edward Gane
- University of Auckland, Auckland, New Zealand
| | | | - Ana Matilla Peña
- Hospital General Universitario Gregorio Marañón, CIBEREHD, Madrid, Spain
| | - Masafumi Ikeda
- National Cancer Center Hospital East, Kashiwa Chiba, Japan
| | | | - Armando Santoro
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, and IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | | | | | | | - Aiwu He
- MedStar Georgetown University Hospital, Washington, DC, USA
| | - Hong Jae Chon
- Medical Oncology, CHA Bundang Medical Center, CHA University School of Medicine, Seongnam, South Korea
| | | | - Gontran Verset
- HUB-Hôpital Erasme, Université Libre de Bruxelles, Brussels, Belgium
| | - Qi Qi Wang
- Bristol Myers Squibb, Princeton, NJ, USA
| | | | | | | | | | | | | |
Collapse
|
28
|
Guo M, Chen S, Sun J, Xu R, Qi Z, Li J, Zhou L, Fang Y, Liu T, Xia J. PIP5K1A Suppresses Ferroptosis and Induces Sorafenib Resistance by Stabilizing NRF2 in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e04372. [PMID: 40405713 DOI: 10.1002/advs.202504372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/23/2025] [Indexed: 05/24/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Ferroptosis, an iron-dependent form of programmed cell death driven by lipid peroxidation, has emerged as a promising strategy for cancer treatment. However, the development of ferroptosis resistance limits the efficacy of such treatments. This study reports that phosphatidylinositol 4-phosphate 5-kinase 1 alpha (PIP5K1A) promotes HCC tumorigenesis and predicts poor prognosis in HCC patients. Knockdown of PIP5K1A enhances lipid peroxidation and increases sensitivity to sorafenib-induced ferroptosis by inhibiting the activation of downstream ferroptosis-related genes regulated by nuclear factor erythroid-2-related factor 2 (NRF2). Mechanistically, PIP5K1A competitively binds to the Kelch domain of Kelch-like ECH-associated protein 1 in a kinase-independent manner, leading to NRF2 escaping from ubiquitination degradation, thereby promoting NRF2-dependent transcription and suppressing ferroptosis. Furthermore, ISA-2011B, a PIP5K1A-specific inhibitor, effectively inhibits HCC growth and sensitized HCC cells to sorafenib. In conclusion, PIP5K1A is a promising therapeutic target for improving the efficacy of sorafenib and other ferroptosis inducers in HCC.
Collapse
Affiliation(s)
- Mengzhou Guo
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Sinuo Chen
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jialei Sun
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Gastroenterology and Hepatology and Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Ruchen Xu
- Department of Gastroenterology and Hepatology and Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Zhuoran Qi
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Gastroenterology and Hepatology and Shanghai Institute of Liver Diseases, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jie Li
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Lianer Zhou
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Yuan Fang
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
- Department of Liver Surgery, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Tianshu Liu
- Department of Medical Oncology, Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| | - Jinglin Xia
- Liver Cancer Institute and Key Laboratory of Carcinogenesis and Cancer Invasion (Ministry of Education), Zhongshan Hospital, Fudan University, 180 Fenglin Road, Shanghai, 200032, China
| |
Collapse
|
29
|
Yang J, Chen R. Radiosensitization Strategies for Hepatocellular Carcinoma: Mechanisms, Therapeutic Advances, and Clinical Perspectives. Crit Rev Oncol Hematol 2025:104773. [PMID: 40412577 DOI: 10.1016/j.critrevonc.2025.104773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2025] [Revised: 05/17/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with treatment efficacy limited by late-stage diagnosis, frequent recurrence, and therapeutic resistance. Radiotherapy is a key local treatment for HCC; however, its efficacy is frequently limited by intrinsic tumor radioresistance. This review discusses strategies to improve the therapeutic response of HCC to radiotherapy. Targeting DNA repair mechanisms can block tumor cells from recovering after radiation-induced damage, whereas modulating cell cycle arrest and programmed cell death pathways (e.g., apoptosis, autophagy) diminishes their survival capacity. Furthermore, remodeling the tumor microenvironment-through hypoxia alleviation, metabolic reprogramming, oxidative stress regulation, and immune activation-may potentiate radiotherapy efficacy. Technological advances, such as stereotactic body radiotherapy and nanomaterial-based approaches, have also improved the precision and effectiveness of radiotherapy. Clinically, combining radiotherapy with systemic therapies (e.g., immune checkpoint inhibitors and antiangiogenic agents) has demonstrated preliminary promise in enhancing treatment outcomes. However, translating preclinical findings into clinical practice remains challenging due to tumor heterogeneity, normal tissue toxicity, and the lack of predictive biomarkers for treatment selection. Future research should focus on integrating molecular profiling with multimodal therapies to enable personalized radiosensitization and bridge the gap between mechanistic insights and clinical outcomes.
Collapse
Affiliation(s)
- Jiahui Yang
- Medical School of Southeast University, Nanjing, Jiangsu Province, China
| | - Rong Chen
- Department of Radiation Oncology, Affiliated ZhongDa Hospital, Southeast University, Nanjing, Jiangsu Province, China.
| |
Collapse
|
30
|
Prosperi E, Cescon M, Lai Q, Bonatti C, Prosperi E, Rizzo F, Maroni L, Laurenzi A, Serenari M, Morelli MC, Ravaioli M. The Italian Score for Organ Allocation: A Ten-Year Monocentric Retrospective Analysis in Liver Transplantation for Hepatocellular Carcinoma. Cancers (Basel) 2025; 17:1720. [PMID: 40427217 PMCID: PMC12110210 DOI: 10.3390/cancers17101720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2025] [Revised: 05/10/2025] [Accepted: 05/14/2025] [Indexed: 05/29/2025] Open
Abstract
BACKGROUND The Italian Score for Organ Allocation (ISO), a transplant benefit oriented allocation system, was introduced in Italy in 2016. The main objective of this study is to identify risk factors for Drop-Out in hepatocellular (HCC) patients enlisted for LT before (Pre-ISO Era) and after ISO (ISO Era) introduction, while the secondary objective is to evaluate the survival results. METHODS CIFs for liver transplantation and Drop-Out were estimated and compared between eras. Factors associated with Drop-Out were identified through multivariable competing risks regression. Survival results were compared using the log-rank test. RESULTS Between 2011 and 2020, 410 patients with HCC were listed for LT. We observed 103 vs. 217 LT and 49 vs. 41 Drop-Outs (p < 0.001) during the Pre-ISO and ISO Era, respectively. In the multivariable analysis, ISO ([sHR] 0.43; 95%CI 0.28-0.66, p < 0.001) and Alcoholic Cirrhosis ([sHR] 0.27, 95%CI 0.11-0.70; p = 0.007) were revealed to be protective factors for Drop-Out. One year after listing, the CI for Drop-Out decreased from 13.2% to 6.2% (p = 0.02). Despite no differences observed in post-LT survival, a significant difference in the intention-to-treat survival from enlisting was found (p = 0.0019). CONCLUSIONS Among other factors, ISO results were protective for the Drop-Out risk in HCC patients awaiting LT, with a benefit in ITT-OS survival.
Collapse
Affiliation(s)
- Enrico Prosperi
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Matteo Cescon
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Quirino Lai
- General Surgery and Organ Transplantation Unit, AOU Policlinico Umberto I, Sapienza University of Rome, 00185 Rome, Italy;
| | - Chiara Bonatti
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Edoardo Prosperi
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Francesca Rizzo
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Lorenzo Maroni
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Andrea Laurenzi
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
| | - Matteo Serenari
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| | - Maria Cristina Morelli
- Internal Medicine Unit for the Treatment of Severe Organ Failure, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy;
| | - Matteo Ravaioli
- Hepatobiliary and Transplant Surgery Unit, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40138 Bologna, Italy; (M.C.); (C.B.); (E.P.); (F.R.); (L.M.); (A.L.); (M.S.)
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, 40126 Bologna, Italy
| |
Collapse
|
31
|
Moris D, Martinino A, Schiltz S, Allen PJ, Barbas A, Sudan D, King L, Berg C, Kim C, Bashir M, Palta M, Morse MA, Lidsky ME. Advances in the treatment of hepatocellular carcinoma: An overview of the current and evolving therapeutic landscape for clinicians. CA Cancer J Clin 2025. [PMID: 40392748 DOI: 10.3322/caac.70018] [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: 11/25/2024] [Revised: 04/10/2025] [Accepted: 04/11/2025] [Indexed: 05/22/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common malignancy and the third leading cause of cancer-related death worldwide. Contemporary advances in systemic and locoregional therapies have led to changes in peer-reviewed guidelines regarding systemic therapy as well as the possibility of downstaging disease that may enable some patients with advanced disease to ultimately undergo partial hepatectomy or transplantation with curative intent. This review focuses on all modalities of therapy for HCC, guided by modern-day practice-changing randomized data where available. The surgical management of HCC, including resection and transplantation, both of which have evolving criteria for what is considered biologically resectable and transplantable, as well as locoregional therapy (i.e., therapeutic embolization, ablation, radiation, and hepatic arterial infusion), are discussed. Historical and modern-day practice-changing trials evaluating immunotherapy with targeted therapies for advanced disease, as well as adjuvant systemic therapy, are also summarized. In addition, this article examines the critical dimension of toxicities and patient-oriented considerations to ensure a comprehensive and balanced discourse on treatment implications.
Collapse
Affiliation(s)
- Dimitrios Moris
- Division of Surgical Oncology, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Alessandro Martinino
- Division of Abdominal Transplantation, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Sarah Schiltz
- Patient Advocate Steering Committee, National Cancer Institute Hepatobiliary Task Force, Los Gatos, California, USA
- Blue Faery, Simi Valley, California, USA
- Cancer CAREpoint, Los Gatos, California, USA
| | - Peter J Allen
- Division of Surgical Oncology, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Andrew Barbas
- Division of Abdominal Transplantation, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Debra Sudan
- Division of Abdominal Transplantation, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| | - Lindsay King
- Division of Gastroenterology and Hepatology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Carl Berg
- Division of Gastroenterology and Hepatology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Charles Kim
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Mustafa Bashir
- Department of Radiology, Duke University Medical Center, Durham, North Carolina, USA
| | - Manisha Palta
- Department of Radiation Oncology, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael A Morse
- Division of Medical Oncology, Department of Medicine, Duke University Medical Center, Durham, North Carolina, USA
| | - Michael E Lidsky
- Division of Surgical Oncology, Department of Surgery, Duke University Medical Center, Durham, North Carolina, USA
| |
Collapse
|
32
|
Xu X, Jiang X, Jiang H, Yuan X, Zhao M, Wang Y, Chen G, Li G, Duan Y. Prediction of prognosis of immune checkpoint inhibitors combined with anti-angiogenic agents for unresectable hepatocellular carcinoma by machine learning-based radiomics. BMC Cancer 2025; 25:888. [PMID: 40389888 PMCID: PMC12087138 DOI: 10.1186/s12885-025-14247-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Accepted: 04/29/2025] [Indexed: 05/21/2025] Open
Abstract
OBJECTIVES This study aims to develop and validate a novel radiomics model utilizing magnetic resonance imaging (MRI) to predict progression-free survival (PFS) in patients with unresectable hepatocellular carcinoma (uHCC) who are receiving a combination of immune checkpoint inhibitors (ICIs) and antiangiogenic agents. This is an area that has not been previously explored using MRI-based radiomics. METHODS 111 patients with uHCC were enrolled in this study. After performing univariate cox regression and the least absolute shrinkage and selection operator (LASSO) algorithms to extract radiological features, the Rad-score was calculated through a Cox proportional hazards regression model and a random survival forest (RSF) model. The optimal calculation method was selected by comparing the Harrell's concordance index (C-index) values. The Rad-score was then combined with independent clinical risk factors to create a nomogram. C-index, time-dependent receiver operating characteristics (ROC) curves, calibration curves, and decision curve analysis were employed to assess the forecast ability of the risk models. RESULTS The combined nomogram incorporated independent clinical factors and Rad-score calculated by RSF demonstrated better prognosis prediction for PFS, with C-index of 0.846, 0.845, separately in the training and the validation cohorts. This indicates that our model performs well and has the potential to enable more precise patient stratification and personalized treatment strategies. Based on the risk level, the participants were classified into two distinct groups: the high-risk signature (HRS) group and the low-risk signature (LRS) group, with a significant difference between the groups (P < 0.01). CONCLUSION The effective clinical-radiomics nomogram based on MRI imaging is a promising tool in predicting the prognosis in uHCC patients receiving ICIs combined with anti-angiogenic agents, potentially leading to more effective clinical outcomes.
Collapse
Affiliation(s)
- Xuni Xu
- Department of Radiology, Shaoxing Central Hospital, The Central Affiliated Hospital, Shaoxing University, Shaoxing, 312000, China
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xue Jiang
- Department of Pathology, Jinhua Municipal Central Hospital, Affiliated Jinhua Hospital, Zhejiang University School of Medicine, Jinhua, 321000, China
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Haoran Jiang
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Xiaoye Yuan
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Mengjing Zhao
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Yuqi Wang
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China
| | - Gang Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| | - Gang Li
- Department of Radiation and Chemotherapy Oncology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325000, China.
| | - Yuxia Duan
- Department of Radiology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China.
| |
Collapse
|
33
|
Yousef EH, El Gayar AM, El-Magd NFA. Insights into Sorafenib resistance in hepatocellular carcinoma: Mechanisms and therapeutic aspects. Crit Rev Oncol Hematol 2025; 212:104765. [PMID: 40389183 DOI: 10.1016/j.critrevonc.2025.104765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Revised: 05/07/2025] [Accepted: 05/11/2025] [Indexed: 05/21/2025] Open
Abstract
The most prevalent primary hepatic cancer, hepatocellular carcinoma (HCC), has a bad prognosis. HCC prevalence and related deaths have increased in recent decades. Food and Drug Administration (FDA) has licensed Sorafenib as a first-line treatment for individuals with advanced HCC. Despite this, some clinical studies indicate that a significant percentage of liver cancer patients exhibit insensitivity to sorafenib. Furthermore, the overall effectiveness of sorafenib is far from adequate, and the number of patients who benefit from therapy is low. In recent years, many researchers have focused on the mechanisms underlying sorafenib resistance. Acquired resistance to sorafenib in HCC cells has been reported to be facilitated by dysregulation of signal transducer and activator of transcription 3 (STAT3) activation, angiogenesis, autophagy, hypoxia-induced pathways, epithelial-mesenchymal transition (EMT), cancer stem cells (CSCs), ferroptosis, and non-coding RNAs (ncRNAs). Recent clinical trials, including comparisons of sorafenib with immune checkpoint inhibitors like tislelizumab, have shown promise in improving patient outcomes. Additionally, combination therapies targeting complementary pathways are under investigation to overcome resistance and enhance treatment efficacy. The limitation of Sorafenib's effectiveness has been partially but not completely clarified. Furthermore, while certain regimens have demonstrated positive results, more clinical trials are required to confirm them. Future research should focus on identifying predictive biomarkers for therapy response, targeting the tumor microenvironment, and exploring novel therapeutic agents and personalized medicine strategies. A deeper understanding of these mechanisms will be essential for developing more effective therapeutic approaches and improving the prognosis of patients with advanced HCC. This article discusses strategies that may be employed to enhance the success of treatment and summarizes new research on the possible pathways that lead to sorafenib resistance.
Collapse
Affiliation(s)
- Eman H Yousef
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt; Pharmacology and Biochemistry department, Faculty of Pharmacy, Horus University-Egypt, New Damietta 34511, Egypt.
| | - Amal M El Gayar
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| | - Nada F Abo El-Magd
- Biochemistry department, Faculty of Pharmacy, Mansoura University, Mansoura 35516, Egypt
| |
Collapse
|
34
|
Wu W, Mao H, Song J, Yang F. Bibliometric analysis of hepatocellular carcinoma and tyrosine kinase inhibitors. Medicine (Baltimore) 2025; 104:e42015. [PMID: 40388796 PMCID: PMC12091622 DOI: 10.1097/md.0000000000042015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/13/2025] [Indexed: 05/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is a common malignant tumor globally and in China, and its incidence and mortality rate are increasing year by year, and it faces many challenges and difficulties in treatment. Tyrosine kinase inhibitors (TKIs) have important roles in cell growth, proliferation, and differentiation, and have now become important drugs for cancer treatment. There are no bibliometric studies on liver cancer and TKIs to date. METHODS We retrieved 2848 records from the Web of Science™ Core Collection (WoSCC) database and analyzed them scientifically and metrically using CiteSpace and VOSviewer in terms of temporal and spatial distributions, author distributions, journal distributions, references, and keywords. RESULTS From January 1, 2004, to December 31, 2023, the WoSCC database documented 2848 publications related to tyrosinase inhibitors in HCC, comprising 2151 articles and 697 reviews. This literature involved 80 countries and regions, 3265 institutions, and 16,653 authors. Analysis shows a steady increase in publications annually since 2004, divided into 3 phases: 2004 to 2010 with fewer than 100 papers annually, suggesting minimal research attention; 2011 to 2019 with gradual growth, indicating increasing research interest; and a rapid surge post-2020, peaking in 2023, signaling heightened global interest in this field. CONCLUSION Our bibliometric analysis on TKIs and HCC spans years, countries, institutions, authors, disciplines, and journals. Since 2004, this field has gained attention, with current research focusing on inflammatory and immune mechanisms, associated diseases, cytokines, and TKIs' applications in liver cancer treatment, including combination therapies. These areas signify emerging research directions.
Collapse
Affiliation(s)
- Wurihan Wu
- Department of Neurology Department, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Hejun Mao
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| | - Jian Song
- Emergency Intensive Care Unit, Inner Mongolia Autonomous Region People’s Hospital, Hohhot, China
| | - Fan Yang
- Department of Hepatobiliary Surgery, Affiliated Hospital of Inner Mongolia Medical University, Hohhot, China
| |
Collapse
|
35
|
Liu MS, Zhong SS, Wang JK, Wang T, Zhang KH. Research Trends on Nanomaterials and Hepatocellular Carcinoma From 1999 to 2024: A Bibliometric Analysis. Drug Des Devel Ther 2025; 19:3949-3970. [PMID: 40395437 PMCID: PMC12091239 DOI: 10.2147/dddt.s516647] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 05/04/2025] [Indexed: 05/22/2025] Open
Abstract
Objective Extensive exploratory studies have been conducted and promising progress has been made in the use of nanomaterials for the diagnosis and treatment of hepatocellular carcinoma (HCC). Here, we aimed to reveal the evolution and trends in this field through bibliometric analysis. Methods English-language publications (1999-2024) in the field of nanomaterials and HCC were retrieved from the Web of Science database, and eligible articles were selected for bibliometric analysis (data extraction, statistical analysis, and visualization) using VOSviewer and Citespace software. Results A total of 1617 eligible publications were analyzed. The number of publications increased rapidly from 2012 and peaked in 2020. China contributed the most publications, and the United States had the most citations. The Chinese Academy of Sciences was the most influential institution. The "International Journal of Nanomedicine (DOVE Medical)" published the most articles, while "Biomaterials (Elsevier)" was the most influential journal. Jie Tian had the highest number of publications, and Dan Shao had the highest average citation per article. Keyword analysis revealed that nanoparticles for targeted drug delivery, therapy and imaging of HCC were research hotspots. Keywords with citation bursts in the last three years included photodynamic therapy, sorafenib, and tumor microenvironment. Nano-vaccines, nano-antibodies, and synergistic therapies were emerging therapeutic strategies. A total of seven clinical trials were published, but to date there have been no major breakthroughs in HCC therapy using nanomaterials. Conclusion Research on nanomaterials and HCC has shown an overall upward trend, with research hotspots and frontiers focusing on nanoparticle-targeted chemotherapies, photodynamic therapy, and related tumor microenvironment research.
Collapse
Affiliation(s)
- Mao-Sheng Liu
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Si-Si Zhong
- Department of Quality and Safety Management, the First Affiliated Hospital of Gannan Medical University, Ganzhou, People’s Republic of China
| | - Jin-Ke Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Ting Wang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| | - Kun-He Zhang
- Department of Gastroenterology, Jiangxi Provincial Key Laboratory of Digestive Diseases, Jiangxi Clinical Research Center for Gastroenterology, Digestive Disease Hospital, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, Jiangxi, People’s Republic of China
| |
Collapse
|
36
|
Li J, Bai Y, Xiong F, Liu X, Hu J, Zhang G, Liu J, Wu S, Zheng C, Kan X. Atezolizumab Plus Bevacizumab Combined with or without Transarterial Chemoembolization in the Treatment of Advanced Hepatocellular Carcinoma: A Single-Center Retrospective Study. J Hepatocell Carcinoma 2025; 12:973-984. [PMID: 40395491 PMCID: PMC12090845 DOI: 10.2147/jhc.s515453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 05/09/2025] [Indexed: 05/22/2025] Open
Abstract
Purpose This study aimed to compare the efficacy and safety of atezolizumab plus bevacizumab (T+A) in combination with transarterial chemoembolization (TACE) (T+A+TACE) and T+A for patients with advanced hepatocellular carcinoma (HCC). Patients and Methods From December 2020 to August 2024, 83 patients with advanced HCC who received T+A+TACE treatment or T+A treatment in our hospital were included, and these patients were categorized into TACE+T+A group (n=52) and T+A group (n=31). The clinical outcomes between the two groups were analyzed and compared, and the prognostic factors that affected the efficacy were analyzed. Results The median overall survival (OS) and median progression-free survival (PFS) in the T+A+TACE group were significantly longer than those of in the T+A group (OS: 22.8 vs 16.9 months, P = 0.015; PFS: 7.1 vs 4.9 months, P = 0.006). A significantly higher objective response rate (ORR) and disease control rate (DCR) that are based on the modified RECIST were achieved in the T+A+TACE group than those of in the T+A group (ORR: 51.9% vs 6.5%, P < 0.001; DCR: 88.5% vs 54.8%, P < 0.001). No significant differences in adverse events (AEs) were observed between the two groups (P > 0.05). The T+A+TACE treatment was identified as a protective factor for OS and PFS. Conclusion TACE further improved the efficacy of T+A treatment for patients with advanced HCC, and it did not increase the incidence of AEs. T+A+TACE treatment is a promising treatment option for patients with advanced HCC.
Collapse
Affiliation(s)
- Jing Li
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Yaowei Bai
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Fu Xiong
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Xiaocui Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Junwen Hu
- Department of Oncology, The Third People’s Hospital of Yibin, Sichuan, 644000, People’s Republic of China
| | - Guilin Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Jiayun Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Suyue Wu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Chuansheng Zheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| | - Xuefeng Kan
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People’s Republic of China
- Hubei Provincial Clinical Research Center for Precision Radiology & Interventional Medicine, Wuhan, 430022, People’s Republic of China
- Hubei Province Key Laboratory of Molecular Imaging, Wuhan, Hubei, 430022, People’s Republic of China
| |
Collapse
|
37
|
Ahn J, Song YS, Kim B, Yang S, Jung K, Lee JC, Kim J, Hwang JH. Prognostic Value of the Metabolic Response on Serial 18F-FDG PET/CT in Pancreatic Cancer. Gut Liver 2025; 19:462-472. [PMID: 40051310 PMCID: PMC12070205 DOI: 10.5009/gnl240458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 05/14/2025] Open
Abstract
Background/Aims The prognostic value of serial 18F-fluorodeoxyglucose positron emission tomography/ computed tomography (18F-FDG PET/CT) for patients with borderline resectable or locally advanced pancreatic cancer who undergo conversion surgery or continue chemotherapy without surgery has not been well-established. Methods A retrospective analysis of patients with pancreatic ductal adenocarcinoma was conducted at Seoul National University Bundang Hospital between March 2013 and February 2022. Patients underwent PET/CT at baseline and subsequent radiologic evaluations following chemotherapy. Changes in the maximum standardized uptake value (SUVmax), mean standardized uptake value (SUVmean), metabolic tumor volume, and total lesion glycolysis were analyzed. Based on their treatment regimens, patients were stratified into the conversion surgery group or nonconversion surgery group. Survival outcomes and various clinical factors were assessed. Results Among 121 patients, 52 underwent conversion surgery, and 69 continued to receive chemotherapy without surgery. A significant reduction in the SUVmax was correlated with prolonged recurrence-free survival and overall survival in the conversion surgery group. Confirmation of a pathologic response indicated a significant association between reductions in the SUVmax and positive outcomes. Reductions in the metabolic tumor volume and total lesion glycolysis were associated with improved progression-free survival and overall survival in the nonconversion surgery group. Conclusions Serial PET/CT scans demonstrated prognostic value in pancreatic ductal adenocarcinoma patients, revealing distinct correlations in the conversion surgery group and nonconversion surgery group.
Collapse
Affiliation(s)
- Jinwoo Ahn
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Yoo Sung Song
- Department of Nuclear Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Bomi Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Soomin Yang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Kwangrok Jung
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jong-Chan Lee
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jaihwan Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| | - Jin-Hyeok Hwang
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seoul National University College of Medicine, Seongnam, Korea
| |
Collapse
|
38
|
Liu M, Wei Y, Xie T, Yang M, Cheng X, Xu L, Li Q, Che F, Xu Q, Song B, Liu M. Deep Reinforcement Learning for CT-Based Non-Invasive Prediction of SOX9 Expression in Hepatocellular Carcinoma. Diagnostics (Basel) 2025; 15:1255. [PMID: 40428248 PMCID: PMC12110404 DOI: 10.3390/diagnostics15101255] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2025] [Revised: 04/24/2025] [Accepted: 04/30/2025] [Indexed: 05/29/2025] Open
Abstract
Background: The transcription factor SOX9 plays a critical role in various diseases, including hepatocellular carcinoma (HCC), and has been implicated in resistance to sorafenib treatment. Accurate assessment of SOX9 expression is important for guiding personalized therapy in HCC patients; however, a reliable non-invasive method for evaluating SOX9 status remains lacking. This study aims to develop a deep learning (DL) model capable of preoperatively and non-invasively predicting SOX9 expression from CT images in HCC patients. Methods: We retrospectively analyzed a dataset comprising 4011 CT images from 101 HCC patients who underwent surgical resection followed by sorafenib therapy at West China Hospital, Sichuan University. A deep reinforcement learning (DRL) approach was proposed to enhance prediction accuracy by identifying and focusing on image regions highly correlated with SOX9 expression, thereby reducing the impact of background noise. Results: Our DRL-based model achieved an area under the curve (AUC) of 91.00% (95% confidence interval: 88.64-93.15%), outperforming conventional DL methods by over 10%. Furthermore, survival analysis revealed that patients with SOX9-positive tumors had significantly shorter recurrence-free survival (RFS) and overall survival (OS) compared to SOX9-negative patients, highlighting the prognostic value of SOX9 status. Conclusions: This study demonstrates that a DRL-enhanced DL model can accurately and non-invasively predict SOX9 expression in HCC patients using preoperative CT images. These findings support the clinical utility of imaging-based SOX9 assessment in informing treatment strategies and prognostic evaluation for patients with advanced HCC.
Collapse
Affiliation(s)
- Minghui Liu
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China; (M.L.); (T.X.); (M.Y.); (X.C.)
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324003, China
| | - Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (Q.L.); (F.C.)
| | - Tianshu Xie
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China; (M.L.); (T.X.); (M.Y.); (X.C.)
| | - Meiyi Yang
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China; (M.L.); (T.X.); (M.Y.); (X.C.)
| | - Xuan Cheng
- School of Computer Science and Engineering, University of Electronic Science and Technology of China, Chengdu 611731, China; (M.L.); (T.X.); (M.Y.); (X.C.)
| | - Lifeng Xu
- Department of Medical Laboratory Science, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou 324000, China;
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (Q.L.); (F.C.)
| | - Feng Che
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (Q.L.); (F.C.)
| | - Qing Xu
- Institute of Clinical Pathology, West China Hospital, Sichuan University, Chengdu 610041, China;
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu 610041, China; (Y.W.); (Q.L.); (F.C.)
- Department of Radiology, Sanya People’s Hospital, Sanya 572000, China
| | - Ming Liu
- Yangtze Delta Region Institute (Quzhou), University of Electronic Science and Technology of China, Quzhou 324003, China
- Department of Medical Laboratory Science, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People’s Hospital, Quzhou 324000, China;
| |
Collapse
|
39
|
Di Benedetto G, Varvarà P, Drago SE, Cantone AF, Mauro N, Gaudio G, Burgaletto C, Bellanca CM, Broggi G, Caltabiano R, Pitarresi G, Cantarella G, Giammona G, Bernardini R. Targeted delivery of sorafenib via biotin decorated polyaminoaspartamide-based nanoparticles for the hepatocarcinoma treatment. Int J Pharm 2025; 678:125729. [PMID: 40379225 DOI: 10.1016/j.ijpharm.2025.125729] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2025] [Revised: 05/12/2025] [Accepted: 05/13/2025] [Indexed: 05/19/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common primary liver cancer, faces treatment challenges due to drug resistance and poor bioavailability, with sorafenib, a key therapy, characterized by rapid clearance and significant side effects. This paper describes the development of amphiphilic graft copolymers for efficient loading and delivery of sorafenib through controlled Atom Transfer Radical Polymerization (ATRP). The amphiphilic graft copolymer PHEA-g-IB-(pButMA)-g-PEG-Bt was synthesized to enhance tumor specificity via biotin-mediated targeting. The synthesis involved a three-step process, with successful functionalization confirmed through NMR and Size Exclusion Chromatography (SEC) analyses. Sorafenib-loaded nanoparticles, prepared via dialysis-based nanoprecipitation, exhibited a mean size of ∼ 300 nm, suitable for oral and parenteral administration, while drug release studies confirmed a sustained release profile, minimizing premature systemic loss and reducing the need for frequent administration. Evaluation of cytocompatibility and anticancer efficacy tested in vitro on HepG2 and HuH-7 cell lines revealed that biotinylated sorafenib-loaded nanoparticles had the highest ability to reduce cell viability. The enhanced anticancer effect of biotinylated NPs was validated in vivo using a murine tumor xenograft model, as evidenced by reduced tumor growth, lower Ki-67 proliferation index, and diminished CD31-positive vasculature. Protein expression analysis demonstrated that PBB-Bt@SOR elicited the strongest activation of p-p38 MAPK and caspase-8-mediated apoptosis, while enhancing the expression of the pro-survival AKT pathway. Overall, the study confirms that biotinylated sorafenib-loaded nanoparticles improve tumor suppression in HCC models, demonstrating their effectiveness in targeted drug delivery. These findings suggest biotin decorated polyamino aspartamide-based nanoparticles as a promising strategy to optimize chemotherapy regimens, minimizing systemic toxicity in HCC treatment.
Collapse
Affiliation(s)
- Giulia Di Benedetto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Paola Varvarà
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Salvatore Emanuele Drago
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Anna Flavia Cantone
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Nicolò Mauro
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Gabriella Gaudio
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Chiara Burgaletto
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Carlo Maria Bellanca
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| | - Giuseppe Broggi
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Via Santa Sofia 87, 95123 Catania, Italy
| | - Rosario Caltabiano
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", Anatomic Pathology, University of Catania, Via Santa Sofia 87, 95123 Catania, Italy
| | - Giovanna Pitarresi
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy.
| | - Giuseppina Cantarella
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy.
| | - Gaetano Giammona
- Laboratory of Biocompatible Polymers, Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, 90123 Palermo, Italy
| | - Renato Bernardini
- Department of Biomedical and Biotechnological Sciences, Section of Pharmacology, University of Catania, Via Santa Sofia 97, 95123 Catania, Italy
| |
Collapse
|
40
|
Fu T, Duan B, Sun P, Ma W, Wang T, Wang T, Tong Z, Wang Y. Innovative applications of silicon dioxide nanoparticles for targeted liver cancer treatment. Front Bioeng Biotechnol 2025; 13:1595772. [PMID: 40421114 PMCID: PMC12104587 DOI: 10.3389/fbioe.2025.1595772] [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: 03/25/2025] [Accepted: 04/24/2025] [Indexed: 05/28/2025] Open
Abstract
Liver cancer remains a major global health challenge, characterized by high mortality and limited treatment efficacy. Conventional therapies, including chemotherapy, immunotherapy, and viral vectors, are hindered by systemic toxicity, drug resistance, and high costs. Silica nanoparticles (SiO2NPs) have emerged as promising platforms for liver cancer therapy, offering precise drug delivery, stimuli-responsive release, and integrated diagnostic-therapeutic capabilities. This review critically examines the potential of SiO2NPs to overcome these therapeutic limitations. Notable advances include their high drug-loading capacity, customizable surface modifications, and dual-responsive systems (pH/redox/NIR-II) that enable >90% tumor-specific drug release. Preclinical studies have demonstrated synergistic efficacy in combination therapies. Additionally, theranostic SiO2NPs enable MRI-guided tumor delineation and real-time treatment monitoring. Despite promising results, challenges remain in long-term biosafety, scalable synthesis, and regulatory compliance. Early-phase clinical trials, including those using NIR-II-responsive platforms, highlight their translational potential but underscore the need for further validation of toxicity profiles and manufacturing standards. Future research should focus on optimizing combinatory treatment strategies, scaling up production, and aligning with evolving regulatory frameworks. By bridging nanomaterial innovation with clinical applications, SiO2NPs offer unparalleled potential for advancing precision oncology in hepatocellular carcinoma.
Collapse
Affiliation(s)
- Tiantian Fu
- Department of Thoracic Radiation Oncology Ward 1, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Boshi Duan
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Peng Sun
- Department of Hand Surgery 4 Ward, Central Hospital Affiliated to Shenyang Medical College, Shenyang, China
| | - Wei Ma
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | | | | | - Zhuang Tong
- Department of Thoracic Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| | - Yue Wang
- Department of General Surgery, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, Shenyang, China
| |
Collapse
|
41
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
42
|
Wang Z, Sheng J, Zhang X. Characterization of adverse reactions to four common targeted drugs for hepatocellular carcinoma in WHO-VigiAccess. Sci Rep 2025; 15:16188. [PMID: 40346128 PMCID: PMC12064674 DOI: 10.1038/s41598-025-00004-7] [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: 11/22/2024] [Accepted: 04/24/2025] [Indexed: 05/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related mortality, with limited therapeutic options available for advanced stages of the disease. Treatment strategies for HCC are multimodal and largely depend on the disease stage, liver function, and individual patient factors. Based on the WHO's VigiAccess database, this study employed a retrospective descriptive analysis of adverse drug reaction (ADR) reports associated with four widely used tyrosine kinase inhibitors (TKIs) for HCC, including Sorafenib, Cabozantinib, Lenvatinib, and Regorafenib. The analysis included demographic data such as patient age, gender, and geographical distribution, alongside clinical information on the systems and symptoms associated with ADR reports. A total of 112,975 ADR reports related to the four TKI-targeted drugs were identified. Sorafenib exhibited the highest ADR reporting rate (30.7%), followed by Cabozantinib (29.4%), Lenvatinib (24.5%), and Regorafenib (15.4%). The odds ratio method was employed to assess the statistical correlation between the use of these targeted drugs and the occurrence of ADRs. Notably, Sorafenib (3,746) and Regorafenib (2,496) served to have the highest number of reported palmar-plantar erythrodysaesthesia syndrome. Chi-square analyses suggested that ADRs related to Lenvatinib were reported significantly more frequently in female patients compared to their male counterparts. The findings of this study can enhance public awareness of drug-related adverse events and provide an evidence-based foundation for prioritizing the management of ADRs associated with TKIs in second-line HCC therapy.
Collapse
Affiliation(s)
- Zeyu Wang
- Department of the Hepatobiliary and Pancreatic Surgery, Jilin University Second Hospital, Changchun, 130000, China
| | - Jiyao Sheng
- Department of the Hepatobiliary and Pancreatic Surgery, Jilin University Second Hospital, Changchun, 130000, China.
| | - Xuewen Zhang
- Department of the Hepatobiliary and Pancreatic Surgery, Jilin University Second Hospital, Changchun, 130000, China.
| |
Collapse
|
43
|
Zhu S, Fu K, Li S, Yang C, Pan C, Wang X, Wang F, Yu X, To KKW, Fu L. Cardiotoxicity of small-molecule kinase inhibitors in cancer therapy. Exp Hematol Oncol 2025; 14:68. [PMID: 40346640 PMCID: PMC12063284 DOI: 10.1186/s40164-025-00660-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2025] [Accepted: 04/22/2025] [Indexed: 05/11/2025] Open
Abstract
Cancer is one of the leading causes of death worldwide. Recent advances in precision oncology have enabled many specific cancer patient populations to respond well and achieve longer survival with small-molecule kinase inhibitors, which have become a new therapeutic strategy for tumors. Since 2001, the Food and Drug Administration has approved 108 and 63 new anticancer drugs for treating solid tumors and hematological malignancies, respectively, 89 of which belong to the large group of small-molecule kinase inhibitors (SMKIs). Compared to conventional chemotherapeutic agents such as cyclophosphamide, doxorubicin, and 5-FU, SMKIs offer better efficacy with fewer toxic side effects. Nevertheless, with the development of more novel SMKIs and their wider clinical application to a larger population of cancer patients, variable degrees of cardiotoxic adverse events have emerged for some SMKIs during cancer therapy. This review comprehensively summarizes the most updated progress in the cardiotoxicity of SMKIs in cancer therapy and discusses the new findings and mechanisms, which will provide emerging strategies for the prevention of cardiotoxicity caused by small molecule targeted drugs and the design of the next generation of low cardiotoxicity targeted drugs.
Collapse
Affiliation(s)
- Shuangli Zhu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Kai Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Sijia Li
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Chuan Yang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Can Pan
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xueping Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Fang Wang
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China
| | - Xiyong Yu
- Key Laboratory of Molecular Target & Clinical Pharmacology and the State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences & The Fifth Affiliated Hospital, Guangdong, Guangzhou Medical University, Guangzhou, 511436, China
| | - Kenneth Kin Wah To
- School of Pharmacy, The Chinese University of Hong Kong, Hong Kong, 999077, China
| | - Liwu Fu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangdong Esophageal Cancer Institute, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, Guangdong, People's Republic of China.
| |
Collapse
|
44
|
Shen L, Cao F, Liu Y, Nuerhashi G, Lin L, Tan H, Wen C, Wang Y, Chen S, Zou H, Xie L, Fan W. Hepatic artery infusion of FOLFOX chemotherapy plus camrelizumab combined with sorafenib for advanced hepatocellular carcinoma in Barcelona Clinic Liver Cancer stage C (Double-IA-001): a phase II trial. BMC Med 2025; 23:275. [PMID: 40346494 PMCID: PMC12065160 DOI: 10.1186/s12916-025-04110-1] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 04/28/2025] [Indexed: 05/11/2025] Open
Abstract
BACKGROUND Hepatic arterial infusion chemotherapy (HAIC) with a combination of oxaliplatin, fluorouracil, and leucovorin (FOLFOX) has shown excellent local control for patients with Barcelona Clinic Liver Cancer (BCLC) stage C hepatocellular carcinoma (HCC). In China, both camrelizumab (a programmed cell death-1 [PD-1] inhibitor) and sorafenib have been approved for the first-line treatment of advanced HCC. This study aimed to investigate the efficacy and safety of hepatic artery infusion of FOLFOX chemotherapy plus camrelizumab combined with sorafenib in BCLC stage C advanced HCC. METHODS This was a single-arm phase II trial (ChiCTR2100041874) with a Simon's two-stage design. Eligible patients were given a maximum of 6 cycles of hepatic artery infusion with FOLFOX chemotherapy plus camrelizumab (200 mg once every 3 weeks). Sorafenib (400 mg orally twice daily) was given since day 3 after the completion of the first cycle of hepatic artery infusion until disease progression, intolerable toxicity, or conversion to surgical resection. The primary endpoint was objective response rate (ORR) based on the modified Response Evaluation Criteria In Solid Tumors (mRECIST). RESULTS Between January 4, 2021, and December 11, 2023, 25 patients were enrolled. Eleven patients had partial response, with an ORR of 44.0% (95% CI, 24.6-63.5%). The primary endpoint was not met, and the study failed to enter the second stage. Median progression-free survival was 4.87 months (95% CI, 2.07-7.66), with a 12-month rate of 23.2%. Median overall survival was 8.87 months (95% CI, 8.17-9.57), with 12- and 24-month rates of 40.3% and 26.9%, respectively. Two (8.0%) patients received curative resection after the study treatment. Grade ≥ 3 treatment-related adverse events occurred in 19 (76.0%) patients, with the most common being decreased lymphocyte count (13 [52.0%]), increased aspartate aminotransferase (11 [44.0%]), and increased alanine aminotransferase (seven [28.0%]). CONCLUSIONS Hepatic artery infusion of FOLFOX chemotherapy plus camrelizumab combined with oral sorafenib shows manageable safety profile but modest antitumor activity in patients with BCLC stage C advanced HCC.
Collapse
MESH Headings
- Humans
- Sorafenib/administration & dosage
- Sorafenib/therapeutic use
- Sorafenib/adverse effects
- Male
- Liver Neoplasms/drug therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- Female
- Middle Aged
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/adverse effects
- Fluorouracil/administration & dosage
- Fluorouracil/therapeutic use
- Fluorouracil/adverse effects
- Infusions, Intra-Arterial
- Hepatic Artery
- Aged
- Adult
- Leucovorin/administration & dosage
- Leucovorin/therapeutic use
- Leucovorin/adverse effects
- Organoplatinum Compounds/administration & dosage
- Organoplatinum Compounds/therapeutic use
- Neoplasm Staging
Collapse
Affiliation(s)
- Lujun Shen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Fei Cao
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Ying Liu
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
- Department of Medical Oncology, Guangzhou University of Traditional Chinese Medicine, Guangzhou, 510006, China
| | - Gulijiayina Nuerhashi
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Letao Lin
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Hontong Tan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Chunyong Wen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Yujia Wang
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Shuanggang Chen
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Hongliang Zou
- Department of Interventional Oncology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510080, China
| | - Lin Xie
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China
| | - Weijun Fan
- Department of Minimally Invasive Interventional Therapy, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, China.
- State Key Laboratory of Oncology in South China, Collaborative Innovation Center of Cancer Medicine, Sun Yat-Sen University, Guangzhou, 510060, China.
| |
Collapse
|
45
|
Taha S, Aljishi M, Sultan A, Bakhiet M. Calcium Homeostasis Disrupted-How Store-Operated Calcium Entry Factor SARAF Silencing Impacts HepG2 Liver Cancer Cells. Int J Mol Sci 2025; 26:4426. [PMID: 40362663 PMCID: PMC12072481 DOI: 10.3390/ijms26094426] [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: 03/15/2025] [Revised: 04/14/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
Hepatocellular carcinoma (HCC), a highly aggressive liver malignancy, is often associated with disrupted calcium homeostasis. Store-operated calcium entry (SOCE), involving components such as STIM1, Orai1, and SARAF, plays a critical role in calcium signaling and cancer progression. While STIM1 and Orai1 have been extensively studied, SARAF's role as a negative regulator of SOCE in HCC remains poorly understood. This preliminary study investigated SARAF's effects on calcium homeostasis, proliferation, and migration in HepG2 liver cancer cells, providing initial evidence of its tumor-suppressive role. SARAF expression was modulated using siRNA knockdown and overexpression plasmids, with validation by qRT-PCR. Functional assays demonstrated that SARAF silencing increased proliferation by 50% and migration by 40% (p < 0.05), while SARAF overexpression reduced proliferation by 50% and migration by 45% (p < 0.01), highlighting its tumor-suppressive role. Intracellular calcium levels, elevated in HepG2 cells, were partially restored by SARAF overexpression, though SARAF silencing did not further disrupt calcium regulation. These findings suggest that SARAF negatively regulates proliferation and migration in HCC, potentially through its role in maintaining calcium homeostasis. SARAF represents a promising therapeutic target in HCC. Future studies should explore the downstream molecular mechanisms governing SARAF's effects, investigate its role in other cancers, and assess its clinical potential for liver cancer therapy.
Collapse
Affiliation(s)
- Safa Taha
- Princess Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, Department of Molecular Medicine, College of Medicine and Health Sciences, Arabian Gulf University, Manama P.O. Box 26671, Bahrain; (M.A.); (A.S.); (M.B.)
| | | | | | | |
Collapse
|
46
|
Chen J, Trindl CA, Ye H, Huang D, Ooi A, Garcia JGN, Chapman E, Zhang DD. CYP4F11, an NRF2 Target Gene, Promotes Hepatocellular Carcinoma Cell Growth. Mol Carcinog 2025. [PMID: 40329467 DOI: 10.1002/mc.23925] [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: 01/27/2025] [Revised: 03/27/2025] [Accepted: 04/13/2025] [Indexed: 05/08/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, is the third leading cause of cancer-related mortality globally. Current systemic therapies for HCC are limited and often exhibit unsatisfactory efficacy, underscoring the need for novel therapeutic approaches. Nuclear factor erythroid 2-related factor-2 (NRF2), a master transcription factor regulating cellular redox and metabolic homeostasis, is frequently overexpressed in HCC due to mutations in NFE2L2/NRF2 or its negative regulator Kelch-like ECH-associated protein 1 (KEAP1), contributing to tumor progression. In this study, we identify CYP4F11, a member of the Cytochrome P450 family, as a direct target gene of NRF2. CYP4F11, primarily expressed in the liver, is crucial in fatty acid oxidation and eicosanoid metabolism. We demonstrate that CYP4F11 expression is driven by NRF2 and is significantly elevated in HCC patients harboring NFE2L2 gain of function or KEAP1 loss of function mutations. Functionally, CYP4F11 promotes HCC cell growth, and reduced expression of CYP4F11 not only suppresses HCC cell proliferation but also enhances sorafenib-induced HCC cell death. Further, NRF2 inhibition sensitizes HCC to sorafenib through downregulation of CYP4F11. These findings position CYP4F11 as a novel contributor to HCC progression and highlight the potential of targeting the NRF2-CYP4F11 axis for HCC treatment.
Collapse
Affiliation(s)
- Jinjing Chen
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Carlee A Trindl
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Haofeng Ye
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Dichun Huang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Aikseng Ooi
- Department of Pharmacology and Toxicology, College of Pharmacy, University of Arizona, Tucson, Arizona, USA
| | - Joe G N Garcia
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
| | - Eli Chapman
- Department of Pharmacology and Therapeutics, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, University of Florida, College of Medicine, Jupiter, Florida, USA
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| | - Donna D Zhang
- Department of Molecular Medicine, Center for Inflammation Science and Systems Medicine, The Herbert Wertheim UF Scripps Institute for Biomedical Innovation and Technology, Jupiter, Florida, USA
- University of Florida Health Cancer Center, University of Florida, Gainesville, Florida, USA
| |
Collapse
|
47
|
An Y, Liu W, Deng Y, Huang W, Huang J. SLC7A11-HSPB1 Axis: A Novel Mechanism for Hepatocellular Carcinoma Progression and Ferroptosis Regulation. Biomed J 2025:100869. [PMID: 40339903 DOI: 10.1016/j.bj.2025.100869] [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: 11/19/2024] [Revised: 03/27/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND SLC7A11, a plasma membrane protein, has been implicated as an oncogene in various cancers, including hepatocellular carcinoma (HCC). Its role in HCC pathogenesis, particularly in relation to ferroptosis, is not well understood. This study aims to investigate the function of SLC7A11 with ferroptosis and its interaction in development of HCC. METHODS AND MATERIAL Clinical HCC tissue samples were used to analyze the expression of SLC7A11 by RT-PCR. The impact of SLC7A11 on HCC cell viability, proliferation, and migration was assessed by CCK-8, AlamarBlue and Transwell. Protein-protein interactions were explored using co-immunoprecipitation and immunofluorescence. The effect of SLC7A11 on ferroptosis was evaluated by iron levels, ROS, and GSH. The impact of sorafenib and doxorubicin (DOX) on HCC cells was analyzed using cell viability assay. RESULTS SLC7A11 was found to be highly expressed in HCC tissues and was correlated with tumor size and poor prognosis. Overexpression of SLC7A11 in HCC cells promoted cell viability, proliferation, and migration. Additionally, SLC7A11 overexpression mitigated erastin-induced ferroptosis, as evidenced by decreased ROS levels and increased GSH levels. We also discovered that SLC7A11 interacted with HSPB1. HSPB1 inhibited erastin-induced ferroptosis. Furthermore, a portion of the cell death induced by sorafenib and DOX is attributed to ferroptosis, with HSPB1 and SLC7A11 inhibiting the death induced by the two drugs, respectively. CONCLUSIONS SLC7A11 plays a significant role in HCC progression by inhibiting ferroptosis, and its interaction with HSPB1 is a critical pathway in this process. Targeting the SLC7A11-HSPB1 axis may provide a novel therapeutic strategy for HCC treatment, highlighting the importance of understanding the mechanisms of ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yan An
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Weilong Liu
- Institute of Hepatology, National clinical research center for infectious diseases, Guangdong Key Lab for Diagnosis &Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
48
|
Singal AG, Salem R, Pinato DJ, Pillai A. Advances in Locoregional and Systemic Treatments for Hepatocellular Carcinoma. Gastroenterology 2025:S0016-5085(25)00660-2. [PMID: 40320088 DOI: 10.1053/j.gastro.2025.03.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 05/29/2025]
Abstract
Significant advances have occurred in the locoregional and systemic therapy landscape for hepatocellular carcinoma (HCC), with the most notable being the introduction of immune checkpoint inhibitor (ICI) combinations. ICI combinations have significantly improved the overall survival of patients with unresectable HCC, affording median survival over 2 years and long-term survival exceeding 5 years in a subset of patients. Accordingly, there has been increased interest in the earlier application of systemic therapies, including (neo)adjuvant therapy in the perioperative setting or in combination with intra-arterial therapies. However, recent data failed to demonstrate improved recurrence-free survival with use of adjuvant ICI therapy. Conversely, 2 trials showed improved progression-free survival when ICI therapies were combined with transarterial chemoembolization, although data regarding the impact on overall survival are still immature. These improved outcomes raise several new questions, including which patients with liver-localized HCC should receive systemic therapy, how should this be sequenced or combined with other available therapies, and how to manage those patients with marked responses, including consideration of liver transplantation. These questions are often determined on a case-by-case basis and best made in a multidisciplinary manner considering several factors, including tumor burden, degree of liver dysfunction, performance status, and patient's long-term goals of care.
Collapse
Affiliation(s)
- Amit G Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas (UT) Southwestern Medical Center, Dallas Texas.
| | - Riad Salem
- Department of Radiology, Northwestern University, Chicago, Illinois
| | - David J Pinato
- Department of Surgery & Cancer, Imperial College London, London, United Kingdom; Department of Translational Medicine (DIMET), University of Piemonte Orientale, Novara, Italy
| | - Anjana Pillai
- Department of Internal Medicine, University of Chicago, Chicago, Illinois
| |
Collapse
|
49
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
50
|
Tsilimigras DI, Kurzrock R, Pawlik TM. Molecular Testing and Targeted Therapies in Hepatobiliary Cancers: A Review. JAMA Surg 2025; 160:576-585. [PMID: 40105823 DOI: 10.1001/jamasurg.2025.0242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Importance Hepatobiliary cancers are heterogeneous and molecularly complex. Recent advances in next-generation sequencing (NGS) have enhanced the understanding of their molecular landscape and enabled deployment of biomarker-based gene- and immune-targeted therapies. This review examines the role of molecular testing and targeted therapies in these malignant neoplasms. Observations Patients with hepatobiliary cancers have poor outcomes. Precision oncology studies have shown that while many common molecular alterations are not currently targetable in hepatocellular carcinoma (HCC), a large number of actionable alterations characterize biliary tract cancers (BTCs), with several therapies now approved by the US Food and Drug Administration. Immunotherapy is increasingly adopted in clinical practice, either as monotherapy or combined with cytotoxic chemotherapy, for both HCC and BTCs. Moreover, multiple solid cancer tumor-agnostic therapies are approved (larotrectinib, entrectinib, and repotrectinib for NTRK fusions; selpercatinib for RET fusions; dabrafenib and trametinib combination for BRAF V600E mutations; dostarlimab or pembrolizumab for tumors with high microsatellite instability and pembrolizumab for tumor mutation burden ≥10 mutations/megabase), highlighting the need for NGS as well as ERBB2 (formerly HER2) immunohistochemistry (IHC) (with the recent approval of solid tissue-agnostic deruxtecan trastuzumab for ERBB2-positive [IHC 3+] cancer) across cancers. N-of-1 clinical trials using customized drug combinations matched to the tumor's molecular profile have yielded encouraging results and provide a promising framework for future clinical trial design. Conclusions and Relevance Molecular testing and gene- and immune-targeted therapies are transforming hepatobiliary cancer treatment. Tumor-agnostic and N-of-1 clinical trials have challenged traditional clinical trial paradigms and provide the foundation for truly personalized oncology for patients with these aggressive cancers. Further work is needed to determine how to leverage these novel approaches into the management of operable disease.
Collapse
Affiliation(s)
- Diamantis I Tsilimigras
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
| | - Razelle Kurzrock
- Medical College of Wisconsin Cancer Center and Linda T. and John A. Mellowes Center for Genomic Sciences and Precision Medicine, Milwaukee
| | - Timothy M Pawlik
- Department of Surgery, Division of Surgical Oncology, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus
- Deputy Editor, JAMA Surgery
| |
Collapse
|