1
|
Wang W, Sun C, Jing L, Xia Y, Zhang S, Shi Y. Light-driven secondary structural remodeling in biomimetic nanosystem to enhance tumor chemo-phototherapy. Mater Today Bio 2025; 33:101955. [PMID: 40538753 PMCID: PMC12178714 DOI: 10.1016/j.mtbio.2025.101955] [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: 04/10/2025] [Revised: 05/25/2025] [Accepted: 06/06/2025] [Indexed: 06/22/2025] Open
Abstract
The integration of chemotherapy and phototherapy for treating advanced liver cancer has gained considerable attention. However, challenges such as short drug retention times significantly impact patient prognosis. We introduce a light-triggered nanosystem that employs molecular aggregation control for PTT and sustained chemotherapy. This nanosystem, known as Reg/IR783@CM nanoparticles (RIMNPs), consists of a core-shell carrier-free nanodrug self-assembled from the chemotherapy drug regorafenib (Reg) and the photothermal agent IR783, coated with a homologous liver cancer cell membrane. The developed core-shell nanocarrier exhibits excellent water dispersibility, high drug load, extended blood circulation, and tumor site enrichment. Upon light exposure, the nanosystem provides outstanding near-infrared imaging and robust photothermal effects. Concurrently, light exposure accelerates the degradation of the outer IR783 layer, resulting in regorafenib exposure and triggering secondary assembly, which significantly enhances drug retention at the tumor site. Our findings indicate that the nanosystem effectively suppresses tumor growth by combining photothermal therapy with the inhibition of tumor cell proliferation and angiogenesis, and by modulating tumor-associated macrophages. Notably, this nanosystem also demonstrates low cytotoxicity and high biocompatibility. This study presents a novel light-driven in-situ assembly strategy, offering a simplified and effective approach for constructing tumor imaging and treatment systems.
Collapse
Affiliation(s)
- Weijie Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Chenguang Sun
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Linhao Jing
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yaning Xia
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Shuijun Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Yupeng Shi
- Department of MRI, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| |
Collapse
|
2
|
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
|
3
|
Luo J, Liang J, Xie H, Chen J, Shen X, Yang F, Li T. Evaluation and verification of MPDZ as a prognostic biomarker for hepatocellular carcinoma through multiple databases. Clin Exp Med 2025; 25:209. [PMID: 40531431 PMCID: PMC12176938 DOI: 10.1007/s10238-025-01729-5] [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: 12/15/2024] [Accepted: 05/09/2025] [Indexed: 06/22/2025]
Abstract
The aim of this study was to assess the prognostic value of the multi-PDZ domain protein (MPDZ) in hepatocellular carcinoma (HCC). MPDZ expression in HCC and normal liver tissue samples were analyzed using TCGA-LIHC data and validated in the GSE14520, GSE62232, GSE76427, GSE121248, and GSE136247 datasets. MPDZ's prognostic value in HCC was evaluated based on Kaplan-Meier survival analysis and Cox proportional risk models. The correlation of MPDZ with other prognostic factors was explored by combined survival analysis and stratified survival analysis. The differentially expressed genes between patient groups stratified on the basis of MPDZ levels in TCGA-LIHC were screened using R and functionally annotated by the GO and KEGG pathway analysis by DAVID. Furthermore, gene set enrichment analysis was conducted to determine the basic molecular mechanism of MPDZ in HCC, and the protein-protein interactions, gene-gene interactions, and immune infiltration status of MPDZ was analyzed by STRING, GeneMania, and TIMER. Our findings indicate that MPDZ is downregulated in HCC and portends worse prognosis. Bioinformatics analysis revealed a strong link between MPDZ and liver cancer progression, liver cancer survival, multiple metabolic pathways, and multiple signaling pathways. In addition, our findings indicate that MPDZ expression is associated with several key genes in the ferroptosis pathway and m6A methylation. Finally, immunohistochemical assessment of clinical specimens confirmed low MPDZ protein expression in HCC tissues relative to paraneoplastic tissues. Taken together, MPDZ is a promising biomarker for the diagnosis and prognosis of HCC.
Collapse
Affiliation(s)
- Jianzhu Luo
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Jianhua Liang
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Haixiang Xie
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, 530021, Guangxi Zhuang Autonomous Region, People's Republic of China
- Guangxi Key Laboratory of Enhanced Recovery After Surgery for Gastrointestinal Cancer, 530021, Nanning, People's Republic of China
| | - Jiaguang Chen
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Xiaoqiang Shen
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Fuquan Yang
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China
| | - Tianman Li
- Department of Hepatobiliary Surgery, The First People's Hospital of Yulin, No. 495, Education Middle Road, Yulin, 537000, Guangxi Zhuang Autonomous Region, People's Republic of China.
| |
Collapse
|
4
|
Zhang Y, Liu H, Zhen W, Jiang T, Cui J. Advancement of drugs conjugated with GalNAc in the targeted delivery to hepatocytes based on asialoglycoprotein receptor. Carbohydr Res 2025; 552:109426. [PMID: 40068307 DOI: 10.1016/j.carres.2025.109426] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/11/2025] [Accepted: 02/12/2025] [Indexed: 04/22/2025]
Abstract
The asialoglycoprotein receptor (ASGPR) is specifically expressed in hepatocytes. Sugar molecules, such as asialoglycoprotein, galactose, galactosamine, and N-acetyl galactosamine (GalNAc), have a high affinity for ASGPR. This review summarizes the structure of ASGPR, the distribution of this molecule in different cells, and the factors influencing the binding of GalNAc to ASGPR. We introduce the application of GalNAc in targeted delivery into hepatocytes by forming conjugated compounds with RNAs and small molecules, and the standard methods for synthesizing GalNAc are also briefly presented. This is to provide an overview of the current research on GalNAc and to shed light on the design of the new GalNAc.
Collapse
Affiliation(s)
- Yafang Zhang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Hongliang Liu
- Pharmaron Beijing Co., Ltd. (China), Beijing, 100176, China
| | - Weina Zhen
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Tingting Jiang
- Baoding Key Laboratory for Precision Diagnosis and Treatment of Infectious Diseases in Children, Baoding Hospital of Beijing Children's Hospital, Capital Medical University, Baoding, 071000, Hebei Province, China
| | - Jingxuan Cui
- College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| |
Collapse
|
5
|
Jain A, Mishra AK, Hurkat P, Shilpi S, Mody N, Jain SK. Navigating liver cancer: Precision targeting for enhanced treatment outcomes. Drug Deliv Transl Res 2025; 15:1935-1961. [PMID: 39847205 DOI: 10.1007/s13346-024-01780-x] [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] [Accepted: 12/18/2024] [Indexed: 01/24/2025]
Abstract
Cancer treatments such as surgery and chemotherapy have several limitations, including ineffectiveness against large or persistent tumors, high relapse rates, drug toxicity, and non-specificity of therapy. Researchers are exploring advanced strategies for treating this life-threatening disease to address these challenges. One promising approach is targeted drug delivery using prodrugs or surface modification with receptor-specific moieties for active or passive targeting. While various drug delivery systems have shown potential for reaching hepatic cells, nano-carriers offer significant size, distribution, and targetability advantages. Engineered nanocarriers can be customized to achieve effective and safe targeting of tumors by manipulating physical characteristics such as particle size or attaching receptor-specific ligands. This method is particularly advantageous in treating liver cancer by targeting specific hepatocyte receptors and enzymatic pathways for both passive and active therapeutic strategies. It highlights the epidemiology of liver cancer and provides an in-depth analysis of the various targeting approaches, including prodrugs, liposomes, magneto-liposomes, micelles, glycol-dendrimers, magnetic nanoparticles, chylomicron-based emulsion, and quantum dots surface modification with receptor-specific moieties. The insights from this review can be immensely significant for preclinical and clinical researchers working towards developing effective treatments for liver cancer. By utilizing these novel strategies, we can overcome the limitations of conventional therapies and offer better outcomes for liver cancer patients.
Collapse
Affiliation(s)
- Ankit Jain
- Department of Pharmacy, Birla Institute of Technology and Science (BITS), Pilani Campus, Pilani, Rajasthan, 333031, India.
| | - Ashwini Kumar Mishra
- Department of Pharmaceutics, School of Pharmacy & Technology Management, SVKM'S NMIMS Deemed-to-be University, Shirpur, Maharashtra, 425405, India
- Central Ayurveda Research Institute, Jhansi, Uttar Pradesh, 284003, India
| | - Pooja Hurkat
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | - Satish Shilpi
- School of Pharmaceuticals and Population Health Informatics, FOP, DIT University, Dehradun, Uttarakahnad, India
| | - Nishi Mody
- Dr. Hari Singh Gour Central University, Sagar, 470003, MP, India
| | | |
Collapse
|
6
|
Zhang W, Liu Z, Liu H, Huang Z, Huang X, Xu L, Che X, Zhan Z. The impact of immune checkpoint inhibitors on prognosis in unresectable hepatocellular carcinoma treated with TACE and lenvatinib: a meta-analysis. Front Immunol 2025; 16:1573505. [PMID: 40469305 PMCID: PMC12133757 DOI: 10.3389/fimmu.2025.1573505] [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: 02/09/2025] [Accepted: 04/25/2025] [Indexed: 06/19/2025] Open
Abstract
Background Combination of multiple therapies is a common approach to treating patients with unresectable hepatocellular carcinoma (uHCC). The impact of immune checkpoint inhibitors (ICIs) on prognosis in uHCC patients treated with transarterial chemoembolization (TACE) and lenvatinib remains unclear. Aim The purpose of this study was to compare the efficacy and safety of TACE plus lenvatinib plus ICIs (TACE+L+I) with TACE plus lenvatinib (TACE+L) in the treatment of patients with uHCC. Methods Publicly available studies comparing the efficacy and safety of TACE+L+I and TACE+L in the treatment of uHCC were collected from the databases PubMed, Embase and Cochrane Library, with a cut-off date of November 1, 2024. Stata SE 15 software was used for analysis. Results Fifteen studies with a total of 1365 patients were included, 688 in the TACE+L+I group and 677 in the TACE+L group. Meta-analysis showed that the TACE+L+I group was significantly higher than the TACE+L group in complete response (RR = 2.34, 95%CI:1.53, 3.59, p < 0.0001), partial response (RR = 1.45, 95%CI:1.28, 1.64, p < 0.0001), objective response rate (RR = 1.55, 95%CI:1.39, 1.73, p < 0.00001), and disease control rate (RR = 1.22, 95%CI:1.10, 1.36, p = 0.0003). The TACE+L+I group was significantly lower than the TACE+L group in progression of disease (RR = 0.39, 95%CI:0.30, 0.51, p < 0.00001). Moreover, TACE+L+I group was not significantly different from TACE+L group in stable disease (RR = 0.85, 95%CI:0.69, 1.03, p = 0.10). The TACE+L+I group was significantly higher than the TACE+L group in overall survival (HR = 2.32, 95%CI:1.95, 3.15, p<0.05) and progression-free survival (HR = 2.30, 95%CI:1.80, 2.93, p<0.05). The TACE+L+I group had a significantly higher incidence of hypothyroidism compared to the TACE+L group (RR = 1.81, 95%CI:1.20, 2.71, p<0.05), but there was no significant difference in other adverse events, such as hypertension, diarrhea, hand-foot syndrome, fatigue, elevated AST, elevated ALT, decreased appetite, hypothyroidism, abdominal pain, thrombocytopenia, rash, and nausea. Conclusion ICIs significantly improved the survival outcome of uHCC treated with TACE+L, and increased the incidence of hypothyroidism. However, this conclusion still needs further validation in the future with more high-quality randomized controlled trials and longer follow-up.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Zirong Liu
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Hongjin Liu
- 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, Guangdong, China
| | - Zhangkan Huang
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Xiaozhun Huang
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Lin Xu
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| | - Zhengyin Zhan
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital & Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, Guangdong, China
| |
Collapse
|
7
|
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
|
8
|
Xu W, Weng J, Zhao Y, Xie P, Xu M, Liu S, Yu Q, Yu M, Liang B, Chen J, Sun HC, Li H, Ye Q, Shen Y. FMO2 + cancer-associated fibroblasts sensitize anti-PD-1 therapy in patients with hepatocellular carcinoma. J Immunother Cancer 2025; 13:e011648. [PMID: 40316306 PMCID: PMC12049961 DOI: 10.1136/jitc-2025-011648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Accepted: 04/14/2025] [Indexed: 05/04/2025] Open
Abstract
BACKGROUND The efficacy of immune checkpoint inhibitors (ICIs) for hepatocellular carcinoma (HCC) is limited by heterogeneity in individual responses to therapy. The heterogeneous phenotypes and crucial roles of cancer-associated fibroblasts (CAFs) in immunotherapy resistance remain largely unclear. METHODS A specific CAF subset was identified by integrating comprehensive single-cell RNA sequencing, spatial transcriptomics and transcriptome profiling of patients with HCC with different responses to antiprogrammed cell death protein 1 (anti-PD-1) therapy. Mouse orthotopic HCC models and a coculture system were constructed, and cytometry by time-of-flight analysis was performed to investigate the functions and mechanisms of specific CAFs in the immune context of HCC. RESULTS We identified a distinct flavin-containing monooxygenase 2 (FMO2)+ CAF subset associated with a favorable response to anti-PD-1 therapy and better clinical outcomes. FMO2+ CAFs increase anti-PD-1 treatment efficacy by promoting tertiary lymphoid structure formation and increasing the infiltration of CD8+ T cells and M1-like macrophages through the C-C motif chemokine ligand 19 (CCL19)-C-C motif chemokine receptor 7 axis. Mechanistically, FMO2 promotes nuclear factor kappa B/p65-mediated CCL19 expression by competitively binding to glycogen synthase 1 (GYS1) with praja ring finger ubiquitin ligase 1 (PJA1), thereby suppressing the PJA1-mediated proteasomal degradation of GYS1. CCL19 treatment potentiated the therapeutic efficacy of anti-PD-1 therapy in mouse orthotopic HCC models. A favorable immunotherapy response was observed in patients with HCC with high serum levels of CCL19. CONCLUSIONS We identified a novel FMO2+ CAF subset that serves as a critical regulator of microenvironmental immune properties and a predictive biomarker of the immunotherapy response in patients with HCC. CCL19 in combination with anti-PD-1 therapy may constitute a novel therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Wenxin Xu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Jialei Weng
- Department of Surgical Oncology, Zhejiang University, Hangzhou, Zhejiang, China
| | - Yufei Zhao
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Peiyi Xie
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Minghao Xu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Shaoqing Liu
- The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Qiang Yu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Mincheng Yu
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Bugang Liang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Junbo Chen
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hui-Chuan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Hui Li
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Qinghai Ye
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| | - Yinghao Shen
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital Fudan University, Shanghai, China
| |
Collapse
|
9
|
Guo X, Cui T, Sun L, Fu Y, Cheng C, Wu C, Zhu Y, Liang S, Liu Y, Zhou S, Li X, Ji C, Ma K, Zhang N, Chu Q, Xing C, Deng S, Wang J, Liu Y, Liu L. A STT3A-dependent PD-L1 glycosylation modification mediated by GMPS drives tumor immune evasion in hepatocellular carcinoma. Cell Death Differ 2025; 32:944-958. [PMID: 39690246 DOI: 10.1038/s41418-024-01432-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Revised: 12/01/2024] [Accepted: 12/05/2024] [Indexed: 12/19/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a malignant tumor characterized by rapid progression. To explore the regulatory mechanism of rapid tumor growth and metastasis, we conducted proteomic and scRNA-Seq analyses on advanced HCC tissues and identified a significant molecule, guanine monophosphate synthase (GMPS), closely associated with the immune evasion in HCC. We analyzed the immune microenvironment characteristics remodeled by GMPS using scRNA-Seq and found GMPS induced tumor immune evasion in HCC by impairing the tumor-killing function of CD8 + T cells. Further investigation revealed that GMPS increased PD-L1 expression by regulating its ubiquitination and glycosylation modification. Mechanistically, GMPS enhanced the bond between PD-L1 and the catalytic subunit STT3A of oligosaccharyltransferase (OST) by acting as an additional module connecting the Sec61 channel complex and STT3A, which aided in the translocation and modification of nascent peptides. Increased PD-L1 impaired the tumor-killing function of CD8 + T cells, leading to the immune evasion. Importantly, targeting GMPS with angustmycin A, an inhibitor of GMPS activity, significantly suppressed PD-L1 expression and tumor growth in HCC, which also increased the sensitivity to anti-CTLA-4 immunotherapy. These findings suggested the potential of targeting GMPS as a promising therapeutic approach for HCC.
Collapse
Affiliation(s)
- Xinyu Guo
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Tianming Cui
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Linmao Sun
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yumin Fu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Cheng Cheng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Chenghui Wu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yitong Zhu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shuhang Liang
- Department of Gastrointestinal Surgery, Anhui Province Key Laboratory of Hepatopancreatobiliary Surgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Yufeng Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shuo Zhou
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Xianying Li
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Changyong Ji
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Kun Ma
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Ning Zhang
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China
| | - Qi Chu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Changjian Xing
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Shumin Deng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China
| | - Jiabei Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Yao Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Lianxin Liu
- Department of Hepatic Surgery, Key Laboratory of Hepatosplenic Surgery, Ministry of Education, The First Affiliated Hospital of Harbin Medical University, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
10
|
Miri H, Rahimzadeh P, Hashemi M, Nabavi N, Aref AR, Daneshi S, Razzaghi A, Abedi M, Tahmasebi S, Farahani N, Taheriazam A. Harnessing immunotherapy for hepatocellular carcinoma: Principles and emerging promises. Pathol Res Pract 2025; 269:155928. [PMID: 40184729 DOI: 10.1016/j.prp.2025.155928] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/07/2025]
Abstract
HCC is considered as one of the leadin causes of death worldwide, with the ability of resistance towards therapeutics. Immunotherapy, particularly ICIs, have provided siginficant insights towards harnessing the immune system. The present review introduces the concepts and possibilities of immunotherapy for HCC treatment, emphasizing its underlying mechanisms and capacity to enhance patient results, focusing on both pre-clinical and clinical insights. The functions of TME and immune evasion mechanisms typical of HCC would be evaluated along with how contemporary immunotherapeutic approaches are designed to address these challenges. Furthermore, the clinical application of immunotherapy in HCC is discussed, emphasizing recent trial findings demonstrating the effectiveness and safety of drugs. In addition, the problems caused by immune evasion and resistance would be discussed to increase potential of immunotherapy along with combination therapy.
Collapse
Affiliation(s)
- Hossein Miri
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Amir Reza Aref
- Department of Vitro Vision, DeepkinetiX, Inc, Boston, MA, USA
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University Of Medical Sciences, Jiroft, Iran
| | - Alireza Razzaghi
- Social Determinants of Health Research Center, Research Institute for Prevention of Non-Communicable Diseases, Qazvin University of Medical Sciences, Qazvin, Iran
| | - Maryam Abedi
- Department of Pathology, Cancer Institute, Tehran University of Medical Sciences, Tehran, Iran.
| | - Safa Tahmasebi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
11
|
Cheng S, Li B, Tang L, Liu S, Xiao J. Tremelimumab plus durvalumab versus sorafenib in first-line treatment of unresectable hepatocellular carcinoma: a cost-effectiveness analysis from the US payer perspective. BMJ Open 2025; 15:e090992. [PMID: 40306910 PMCID: PMC12049948 DOI: 10.1136/bmjopen-2024-090992] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 04/11/2025] [Indexed: 05/02/2025] Open
Abstract
OBJECTIVE In a recently published 4-year overall survival (OS) update from the phase III clinical trial named HIMALAYA (NCT03298451), single tremelimumab plus regular interval durvalumab (a regimen termed STRIDE) demonstrated significantly improved OS compared with sorafenib in the first-line setting of unresectable hepatocellular carcinoma (uHCC). Although dual immunotherapy represents a novel treatment option for uHCC, the economic implications of these high-priced drugs require further exploration. This study aimed to evaluate the cost-effectiveness of STRIDE in uHCC to inform first-line treatment decisions and help allocate medical resources most effectively. DESIGN Using a partitioned survival model, we conducted a cost-effectiveness analysis comparing STRIDE to sorafenib in the first-line treatment of uHCC. Clinical information was gathered from the phase III HIMALAYA trial. Costs and health state utilities data were derived from previous literature. Uncertainty of the model was assessed through one-way sensitivity analysis and probabilistic sensitivity analysis. OUTCOME MEASURES Total costs, life years, quality-adjusted life years (QALYs), incremental QALYs and incremental cost-effectiveness ratio (ICER). SETTING US payer perspective. PARTICIPANTS 393 participants in the STRIDE group and 389 participants in the sorafenib group who were diagnosed with uHCC and had no previous systemic treatment. INTERVENTIONS Single-dose tremelimumab plus monthly durvalumab (STRIDE) versus sorafenib. RESULTS Treatment with STRIDE provided an additional 0.51 QALYs at an incremental total cost of United States dollar ($)9812. The ICER of STRIDE was $19 239 per QALY compared with sorafenib, which falls below the willingness-to-pay threshold of $150 000 per QALY. Sensitivity analyses indicated that our results were robust to the variation ranges of key inputs. CONCLUSION In this economic evaluation comparing two first-line systemic therapies for uHCC patients, STRIDE was cost-effective compared with sorafenib from a US payer perspective. Our study is the first to demonstrate that immunotherapy can provide both survival benefits and economic viability in uHCC.
Collapse
MESH Headings
- Female
- Humans
- Male
- Antibodies, Monoclonal/economics
- Antibodies, Monoclonal/therapeutic use
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/economics
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/economics
- Cost-Benefit Analysis
- Cost-Effectiveness Analysis
- Liver Neoplasms/drug therapy
- Liver Neoplasms/mortality
- Liver Neoplasms/economics
- Quality-Adjusted Life Years
- Sorafenib/economics
- Sorafenib/therapeutic use
- Sorafenib/administration & dosage
- United States
- Clinical Trials, Phase III as Topic
Collapse
Affiliation(s)
- Shuqiao Cheng
- Department of Pharmacy, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Bin Li
- Department of Oncology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Lanhua Tang
- Department of Oncology, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Shao Liu
- Department of Pharmacy, Xiangya Hospital Central South University, Changsha, Hunan, China
| | - Jian Xiao
- Department of Pharmacy, Xiangya Hospital Central South University, Changsha, Hunan, China
| |
Collapse
|
12
|
Yin X, Zhao X, Shen Y, Xie W, He C, Guo J, Li Z, Xuan F, Zeng S, Zeng X, Fang C. Nanoparticle-mediated dual targeting of stromal and immune components to overcome fibrotic and immunosuppressive barriers in hepatocellular carcinoma. J Control Release 2025; 383:113783. [PMID: 40306574 DOI: 10.1016/j.jconrel.2025.113783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2025] [Revised: 04/22/2025] [Accepted: 04/24/2025] [Indexed: 05/02/2025]
Abstract
Cancer-associated fibroblasts (CAFs) are key drivers of hepatocellular carcinoma (HCC) through their promotion of fibrosis and immune suppression activity. To overcome this stroma-immune barrier, we developed D/F@MRL, a stroma-immune co-targeting nanoplatform that enables the spatiotemporal coordination of CAF reprogramming and immune activation. In D/F@MRL, MMP-2-responsive hybrid liposomes (MRL) was employed to co-load digoxin (Dig) and PD-L1-degrading nanofibers (NFs). Upon encountering the MMP-2-enriched HCC stroma, D/F@MRL undergoes enzymatic cleavage, thereby enabling the targeted release of Dig and NFs within the HCC microenvironment. Mechanistically, Dig inhibits the phosphorylation of SMAD3 in CAFs, while PD-L1 degradation destabilizes the TGFβ receptor, synergistically silencing TGF-β/Smad signaling to reprogram CAFs. This combination not only disrupts the fibrotic barrier but also creates a feed-forward loop that further enhances drug penetration, while reinforcing the immune activation driven by Dig-induced immunogenic cell death (ICD) and PD-L1 degradation. In the humanized immune PDX model, D/F@MRL successfully reprogrammed CAFs and robustly remodeled the stromal and immune microenvironments without causing systemic toxicity, highlighting its promising potential for clinical translation. By integrating CAF reprogramming with ICD and immune checkpoint inhibition, this strategy overcame the limitations of single-target therapies, induced robust immune activation, further provided a clinic-transformative approach for fibrotic malignancies.
Collapse
Affiliation(s)
- Xiangyi Yin
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xingyang Zhao
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Yiming Shen
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Weizhong Xie
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Cheng He
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Jianan Guo
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Zirong Li
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Feichao Xuan
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Silue Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Xiaojun Zeng
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China
| | - Chihua Fang
- First Department of Hepatobiliary Surgery, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Institute of Digital Intelligent Minimally Invasive Surgery, Zhujiang Hospital, Southern Medical University, Guangzhou 510280, China; Guangdong Provincial Clinical and Engineering Center of Digital Medicine, Guangzhou 510280, China; South China Institute of National Engineering Research Center of Innovation and Application of Minimally Invasive Instruments, Guangzhou 510280, China.
| |
Collapse
|
13
|
Naffaa MM, Al-Ewaidat OA, Gogia S, Begiashvili V. Neoantigen-based immunotherapy: advancing precision medicine in cancer and glioblastoma treatment through discovery and innovation. EXPLORATION OF TARGETED ANTI-TUMOR THERAPY 2025; 6:1002313. [PMID: 40309350 PMCID: PMC12040680 DOI: 10.37349/etat.2025.1002313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 04/07/2025] [Indexed: 05/02/2025] Open
Abstract
Neoantigen-based immunotherapy has emerged as a transformative approach in cancer treatment, offering precision medicine strategies that target tumor-specific antigens derived from genetic, transcriptomic, and proteomic alterations unique to cancer cells. These neoantigens serve as highly specific targets for personalized therapies, promising more effective and tailored treatments. The aim of this article is to explore the advances in neoantigen-based therapies, highlighting successful treatments such as vaccines, tumor-infiltrating lymphocyte (TIL) therapy, T-cell receptor-engineered T cells therapy (TCR-T), and chimeric antigen receptor T cells therapy (CAR-T), particularly in cancer types like glioblastoma (GBM). Advances in technologies such as next-generation sequencing, RNA-based platforms, and CRISPR gene editing have accelerated the identification and validation of neoantigens, moving them closer to clinical application. Despite promising results, challenges such as tumor heterogeneity, immune evasion, and resistance mechanisms persist. The integration of AI-driven tools and multi-omic data has refined neoantigen discovery, while combination therapies are being developed to address issues like immune suppression and scalability. Additionally, the article discusses the ongoing development of personalized immunotherapies targeting tumor mutations, emphasizing the need for continued collaboration between computational and experimental approaches. Ultimately, the integration of cutting-edge technologies in neoantigen research holds the potential to revolutionize cancer care, offering hope for more effective and targeted treatments.
Collapse
Affiliation(s)
- Moawiah M Naffaa
- Department of Psychology and Neuroscience, Duke University, Durham, NC 27708, USA
- Department of Cell Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ola A Al-Ewaidat
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Sopiko Gogia
- Department of Internal Medicine, Ascension Saint Francis Hospital, Evanston, IL 60202, USA
| | - Valiko Begiashvili
- Department of Internal Medicine, University of Kansas Medical Center, Kansas City, KS 66103, USA
| |
Collapse
|
14
|
Fu X, Guo Y, Zhang K, Cheng Z, Liu C, Ren Y, Miao L, Liu W, Jiang S, Zhou C, Su Y, Yang L. Prognostic impact of extracellular volume fraction derived from equilibrium contrast-enhanced CT in HCC patients receiving immune checkpoint inhibitors. Sci Rep 2025; 15:13643. [PMID: 40254627 PMCID: PMC12009984 DOI: 10.1038/s41598-025-97677-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 04/07/2025] [Indexed: 04/22/2025] Open
Abstract
This study aimed to investigate whether extracellular volume (ECV) fraction derived from equilibration contrast-enhanced computed tomography (CECT) affects prognosis in HCC patients receiving ICIs. This retrospective study ultimately included 211 HCC patients undergoing ICIs, of whom 60 were included in an internal validation to assess the reproducibility of the results. Baseline unenhanced and equilibrated CECT were used to measure CT values of the tumor, liver and aorta, which were combined with hematocrit to calculate the ECV fraction. Correlation analysis was used to investigate the association between tumor ECV and liver ECV fractions. The effects of clinical variables and ECV fraction on progression-free survival (PFS) and overall survival (OS) were evaluated using Cox proportional hazards models and Kaplan-Meier curves. Of these 151 patients, tumor ECV fraction positively correlated with liver ECV fraction. In the Lower tumor ECV group, PFS (5.6 vs. 7.6 months) and OS (10.5 vs. 15.5 months) were notably shorter than in the Higher tumor ECV group, while no significant differences were found between the Higher and Lower liver ECV groups. Furthermore, the multivariable Cox regression model demonstrated that higher tumor ECV fraction level was an independent protective factor for PFS and OS (all P < 0.001). Internal validation cohort preliminary demonstrated reproducibility of results. The tumor ECV fraction is expected to become a routine indicator before ICIs therapy for HCC patients in contrast to liver ECV fraction, contributing to their subsequent management.
Collapse
Affiliation(s)
- Xiaona Fu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yusheng Guo
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Kailu Zhang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Zhixuan Cheng
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chanyuan Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Yi Ren
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Lianwei Miao
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Weiwei Liu
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Shanshan Jiang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China
| | - Chen Zhou
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Yangbo Su
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| | - Lian Yang
- Department of Radiology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China.
- Hubei Key Laboratory of Molecular Imaging, Wuhan, 430022, China.
| |
Collapse
|
15
|
Su P, Han Y, Yi J, Hou Y, Xiao Y. Research status and frontiers in liver cancer immunotherapy: a bibliometric perspective on highly cited literature. Front Oncol 2025; 15:1587252. [PMID: 40276056 PMCID: PMC12018336 DOI: 10.3389/fonc.2025.1587252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Accepted: 03/14/2025] [Indexed: 04/26/2025] Open
Abstract
Background Liver cancer is one of the major causes of cancer-related death in the world. As a breakthrough therapy, immunotherapy had significantly improved the prognosis of patients. However, the current research status and research hotspots in the field of liver cancer immunotherapy still lack systematic review. Based on the bibliometric analysis of highly cited papers, this study intended to reveal the current research status, research hotspots and future research trends in this field. Objective The purpose of this study was to analyze the national/regional contributions, authors and institutions cooperation network, keywords clustering and keywords burst analysis of highly cited papers on liver cancer immunotherapy through bibliometrics, so as to clarify the research frontier and development direction, and provide objective data support for future research direction and clinical practice. Methods The highly cited papers on liver cancer immunotherapy from the Web of Science core collection up to February 23, 2025 were retrieved, and 232 studies were included. CiteSpace was used to build a knowledge map, analyze the distribution of years, countries, authors, institutions and cooperation networks, and identify research hotspots and emerging trends through keyword clustering and burst detection. Results The number of highly cited papers continued to increase from 2014 and reached a peak in 2022. China and the United States had the highest number of publications and the centrality of cooperation networks. The author with the highest number of papers was Llovet, Josep M, whose research direction mainly focused on immune checkpoint inhibitor combination therapy and molecular typing. The author with the highest cooperation network centrality was Duda, Dan G, whose research team focused on tumor microenvironment regulation. Harvard University and the University of Barcelona played an important central role in the institutional collaboration. Keywords analysis showed that immune checkpoint inhibitors, tumor microenvironment and combination therapy were the core of liver cancer immunotherapy. Burst keywords such as cell lung cancer, pembrolizumab, advanced melanoma, blockade, lymphocytes, etc. had revealed the research frontier of liver cancer immunotherapy research. Conclusion The research on liver cancer immunotherapy had made multi-dimensional progress, with China and the United States leading the global cooperation. The main research directions were the combination strategy of immunization, the regulation of tumor microenvironment and the exploration of novel targets. In the future, it is necessary to optimize treatment resistance solutions, integrate interdisciplinary resources, and promote the development of precision and personalized treatment.
Collapse
Affiliation(s)
- Pan Su
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yeqiong Han
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Jindong Yi
- Teaching and Research Section of Clinical Nursing, Xiangya Hospital, Central South University, Changsha, China
| | - Yu Hou
- Department of Pulmonology, Children’s Hospital, National Clinical Research Center For Child Health, Zhejiang University School of Medicine, Hangzhou, China
| | - Yao Xiao
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
- International Joint Research Center of Minimally Invasive Endoscopic Technology Equipment & Standards, Xiangya Hospital, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
16
|
Li J, Xian L, Wang X, Liu Y, Li J. The role of TACE in the era of immune-targeted therapy for hepatocellular carcinoma: a meta-analysis based on PSM. Front Immunol 2025; 16:1573834. [PMID: 40242754 PMCID: PMC12000099 DOI: 10.3389/fimmu.2025.1573834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Accepted: 03/12/2025] [Indexed: 04/18/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a major global health challenge, with over 50% of patients ineligible for curative treatments at diagnosis. The combination of molecular targeted therapies and immunotherapy has shown promise in improving outcomes for advanced HCC. Objective This meta-analysis aims to assess the efficacy of combining transarterial chemoembolisation (TACE) with immune-targeted therapies in patients with unresectable HCC. Methods A systematic review and meta-analysis conforming to PRISMA guidelines were conducted by searching PubMed, Embase, Web of Science, and the Cochrane Library for studies published up to January 5, 2025. Due to the limited clinical evidence, our study exclusively included retrospective studies based on propensity score matching (PSM) analysis that compared the efficacy of TACE in combination with immune-targeted therapy to immune-targeted therapy alone. Key outcomes assessed included objective response rate (ORR), disease control rate (DCR), one-year overall survival (1-OS), one-year progression-free survival (1-PFS), median overall survival (mOS), and median progression-free survival (mPFS). Results A total of 9 PSM studies involving 2119 patients were included. The meta-analysis revealed that TACE significantly improved ORR, DCR, 1-OS, and 1-PFS, in addition to extending mOS and mPFS. Conclusion The findings suggest that the inclusion of TACE in treatment regimens for unresectable HCC notably enhances tumour control and patient survival. This study provides moderate to high-quality evidence supporting the integration of TACE in advanced HCC management, particularly for those patients not meeting standard TACE criteria. Systematic review registration https://www.crd.york.ac.uk/PROSPERO/, identifier CRD 42025631817.
Collapse
Affiliation(s)
- Jiahao Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Lei Xian
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Xinsen Wang
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Yingnan Liu
- Department of Radiology, The First Hospital of Jilin University, Changchun, Jilin, China
| | - Jiarui Li
- Department of Interventional Therapy, The First Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
17
|
Zeng C, Hua S, Zhou J, Zeng T, Chen J, Su L, Jiang A, Zhou M, Tang Z. Oral Microalgae-Based Biosystem to Enhance Irreversible Electroporation Immunotherapy in Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2409381. [PMID: 39874200 PMCID: PMC12005737 DOI: 10.1002/advs.202409381] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Revised: 12/22/2024] [Indexed: 01/30/2025]
Abstract
Irreversible electroporation (IRE) is a novel local tumor ablation technique that can potentially stimulate immune responses. However, IRE alone cannot effectively activate the immune system or prevent distant metastases. Therefore, this study utilized the biocompatibility of Chlorella vulgaris (C. vulgaris) and polydopamine (PDA) adhesive properties to encapsulate a PD-1 inhibitor (PI). The PDA coating protects the drug from degradation by stomach acid and enhances its intestinal absorption. This carrier demonstrates excellent in vivo drug release control and biodistribution, significantly increasing the oral bioavailability of PI. Combining IRE with this natural carrier significantly improves the therapeutic efficacy, which increases the local drug concentration and activates the immune system. This system demonstrates significantly improved therapeutic efficacy against local tumors compared with PI or IRE alone and significantly reduces PI-associated side effects. A convenient oral delivery system is developed using this readily available natural micro-carrier that not only improves the therapeutic effect of IRE but also mitigates its adverse effects, indicating significant potential for clinical applications. This discovery offers a new strategy for hepatocellular carcinoma treatment with the potential to improve patient outcomes.
Collapse
Affiliation(s)
- Cheng Zeng
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Shiyuan Hua
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- Institute of Translational MedicineZhejiang UniversityHangzhou310029China
- Zhejiang University‐Ordos City Etuoke Banner Joint Research CenterZhejiang UniversityHaining314400China
- The National Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHangzhou310027China
| | - Jiayu Zhou
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- School of MedicineShihezi UniversityShiheziXinjiang832002China
| | - Tangye Zeng
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Jianke Chen
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Lijian Su
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
| | - Angfeng Jiang
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| | - Min Zhou
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Zhejiang University‐University of Edinburgh Institute (ZJU‐UoE Institute)Zhejiang UniversityHaining314400China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
- Institute of Translational MedicineZhejiang UniversityHangzhou310029China
- Zhejiang University‐Ordos City Etuoke Banner Joint Research CenterZhejiang UniversityHaining314400China
- The National Key Laboratory of Biobased Transportation Fuel TechnologyZhejiang UniversityHangzhou310027China
| | - Zhe Tang
- Department of SurgeryCenter for Cancer Medicinethe Fourth Affiliated Hospital of School of MedicineInternational School of MedicineInternational Institutes of MedicineZhejiang UniversityYiwu322000China
- Department of SurgeryThe Second Affiliated HospitalZhejiang University School of MedicineHangzhou310000China
- Zhejiang Key Laboratory of Precision Diagnosis and Treatment for Lung Cancerthe Fourth Affiliated Hospital, Zhejiang University School of MedicineYiwu322000China
| |
Collapse
|
18
|
Zhang F, Wang YS, Li SP, Zhao B, Huang N, Song RP, Meng FZ, Feng ZW, Zhang SY, Song HC, Chen XP, Liu LX, Wang JZ. Alpha-fetoprotein combined with initial tumor shape irregularity in predicting the survival of patients with advanced hepatocellular carcinoma treated with immune-checkpoint inhibitors: a retrospective multi-center cohort study. J Gastroenterol 2025; 60:442-455. [PMID: 39714631 PMCID: PMC11922967 DOI: 10.1007/s00535-024-02202-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Accepted: 12/07/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are playing a significant role in the treatment of hepatocellular carcinoma (HCC). This study aims to explore the prognostic value of alpha-fetoprotein (AFP) and initial tumor shape irregularity in patients treated with ICIs. METHODS In this retrospective, multi-center study, 296 HCC patients were randomly divided into the training set and the validation set in a 3:2 ratio. The training set was used to evaluate prognostic factors and to develop an easily applicable ATSI (AFP and Tumor Shape Irregularity) score, which was verified in the validation set. RESULTS The ATSI score was developed from two independent prognostic risk factors: baseline AFP ≥ 400 ng/ml (HR 1.73, 95% CI 1.01-2.96, P = 0.046) and initial tumor shape irregularity (HR 1.94, 95% CI 1.03-3.65, P = 0.041). The median overall survival (OS) was not reached (95% CI 28.20-NA) in patients who met no criteria (0 points), 25.8 months (95% CI 14.17-NA) in patients who met one criterion (1 point), and 17.03 months (95% CI 11.73-23.83) in patients who met two criteria (2 points) (P = 0.001). The median progression-free survival (PFS) was 10.83 months (95% CI 9.27-14.33) for 0 points, 8.03 months (95% CI 6.77-10.57) for 1 point, and 5.03 months (95% CI 3.83-9.67) for 2 points (P < 0.001). The validation set effectively verified these results (median OS, 37.43/24.27/14.03 months for 0/1/2 points, P = 0.028; median PFS, 13.93/8.30/4.90 months for 0/1/2 points, P < 0.001). CONCLUSIONS The ATSI score can effectively predict prognosis in HCC patients receiving ICIs.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yong-Shuai Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shao-Peng Li
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Bin Zhao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Nan Huang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Rui-Peng Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Fan-Zheng Meng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Zhi-Wen Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China
| | - Shen-Yu Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Hua-Chuan Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xiao-Peng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China.
| | - Lian-Xin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Ji-Zhou Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
19
|
Xu J, Wang X, Jia Z, Sun G. Effectiveness and safety of angiogenesis inhibitors combined with PD-1/PD-L1 blockades in the first-line treatment of patients with advanced hepatocellular carcinoma: A single-center retrospective study. Medicine (Baltimore) 2025; 104:e41814. [PMID: 40101095 PMCID: PMC11922473 DOI: 10.1097/md.0000000000041814] [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: 07/20/2024] [Accepted: 02/21/2025] [Indexed: 03/20/2025] Open
Abstract
The combination of immune checkpoint inhibitors targeting anti-programmed cell death-1 (anti-PD-1) or anti-programmed death ligand-1 (anti-PD-L1) with antiangiogenic agents has emerged as a revolutionary therapy for advanced hepatocellular carcinoma (aHCC). Key antiangiogenic medications encompass monoclonal antibodies targeting vascular endothelial growth factor (anti-VEGF mAbs) and multiple kinase inhibitors (MKIs). The aim of this study is to assess the difference of efficacy and safety between 2 combination therapies. This study retrospectively examined the outcomes of 57 patients with aHCC who underwent first-line treatment with a combination of immune checkpoint inhibitors and antiangiogenic therapy at the First Affiliated Hospital of Anhui Medical University, from September 2018 to July 2023. The analysis, conducted using SPSS software, focused on patient outcomes such as tumor response (assessed according to modified Response Evaluation Criteria in Solid Tumors criteria), objective response rate, disease control rate, progression-free survival, overall survival, and safety. Comparisons among different groups were also made. The anti-PD-1/anti-PD-L1-anti-VEGF mAbs group showed a trend of higher partial response rate (37.50% vs 22.45%), objective response rate (37.50% vs 24.49%), disease control rate (62.50% vs 59.18%), and seemed to achieve longer median progression-free survival (14.93 vs 14.90 months) and median overall survival (15.80 vs 11.10 months) without higher grade 3 or higher adverse events comparing to anti-PD-1/anti-PD-L1-MKIs group. Subgroup analysis showed that the anti-PD-1-lenvatinib group achieved longer median progression-free survival (23.97 months), while the anti-PD-1-regorafenib group achieved longer median overall survival (37.97 months). The anti-PD-1/anti-PD-L1 combined with anti-VEGF mAbs was effective and tolerable compared to anti-PD-1/anti-PD-L1-MKIs in aHCC. The addition of lenvatinib or regorafenib may provide promising incremental benefit for patients with aHCC.
Collapse
Affiliation(s)
- Jing Xu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Xin Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Zhenya Jia
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| | - Guoping Sun
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui Province, China
| |
Collapse
|
20
|
Zhang L, Zhuge Y, Ni J. BUB1 serves as a biomarker for poor prognosis in liver hepatocellular carcinoma. BMC Immunol 2025; 26:20. [PMID: 40069598 PMCID: PMC11895216 DOI: 10.1186/s12865-025-00698-4] [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: 12/01/2024] [Accepted: 02/27/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is the most frequent kind of liver cancer with high morbidity and mortality rates worldwide. Altered expression of BUB1 (budding uninhibited by benzimidazole 1) gene leads to chromosome instability and aneuploidy. This study investigated the expression of BUB1 and its prognostic value as well as its correlation with immune cell infiltration and immune checkpoints in HCC. RESULTS Using the Gene Expression Omnibus (GEO) and The Cancer Genome Atlas (TCGA) databases, we found that BUB1 was up-regulated in HCC, thus prompting us to validate this observation by immunohistochemistry on 57 HCC paraffin embedded tissues from Wuxi No.2 People's Hospital. Kaplan-Meier survival analysis revealed that HCC patients with high BUB1 expression had shorter overall survival (OS) time as well as progression-free interval (PFI), and disease-specific survival (DSS) time compared to the patients with low BUB1 expression. Besides, STRING database showed that the top 10 co-expression genes were mainly involved in the regulation of cell division during the mitosis. Gene Ontology (GO) analysis and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis showed that BUB1 had a connection to cancer related pathways. Lastly, The Tumor Immune Estimation Resource (TIMER) analysis found that BUB1 was positively related to immune cell infiltration and some immune checkpoint gene in HCC. CLINICAL TRIAL NUMBER Not applicable. CONCLUSIONS Our present study demonstrated that BUB1 is a potential prognostic biomarker, and BUB1 may play a role in the tumor immune microenvironment in HCC.
Collapse
Affiliation(s)
- Lili Zhang
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
- Department of Gastroenterology, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, Wuxi, Jiangsu Province, China.
| | - Yuzheng Zhuge
- Department of Gastroenterology, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, Jiangsu Province, China.
| | - Jingbin Ni
- Department of Gastroenterology, Jiangnan University Medical Center, Wuxi No.2 People's Hospital, Wuxi, Jiangsu Province, China
| |
Collapse
|
21
|
Cheng SL, Wu CH, Tsai YJ, Song JS, Chen HM, Yeh TK, Shen CT, Chiang JC, Lee HM, Huang KW, Chen Y, Qiu JT, Yen YT, Shia KS, Chen Y. CXCR4 antagonist-loaded nanoparticles reprogram the tumor microenvironment and enhance immunotherapy in hepatocellular carcinoma. J Control Release 2025; 379:967-981. [PMID: 39863023 DOI: 10.1016/j.jconrel.2025.01.066] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/20/2025] [Accepted: 01/22/2025] [Indexed: 01/27/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer death that has limited treatment options for advanced stages. Although PD-1 inhibitors such as nivolumab and pembrolizumab have been approved for advanced HCC treatment, their effectiveness is often hampered by the immunosuppressive tumor microenvironment (TME), which is due to hypoxia-driven CXCL12/CXCR4 axis activation. In this study, we developed 807-NPs, lipid-coated tannic acid (TA) nanoparticles that encapsulate BPRCX807, a potent CXCR4 antagonist to target HCC. 807-NPs enhance the pharmacokinetics and improve the tumor availability of BPRCX807 without causing systemic toxicity. Our findings show that 807-NPs block the CXCR4/CXCL12 pathway, inhibiting Akt and mTOR activation in HCC cells and M2 macrophages and promoting their repolarization toward the antitumor M1 phenotype. In orthotopic murine HCC models, systemic administration of 807-NPs significantly remodeled the immunosuppressive TME by reprogramming tumor-associated macrophages (TAMs) toward an immunostimulatory phenotype and promoting cytotoxic T-cell infiltration into tumors. This led to suppressed primary tumor growth and metastasis, while enhancing the efficacy of cancer immunotherapies, including PD-1 blockade and whole-cancer cell vaccines, by promoting T-cell activation. Our work demonstrates the potential of using nanotechnology to deliver CXCR4 antagonists for cancer immunotherapy.
Collapse
Affiliation(s)
- Sheng-Liang Cheng
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; International Intercollegiate PhD Program, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Chien-Huang Wu
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Yun-Jen Tsai
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jen-Shin Song
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Hsin-Min Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Teng-Kuang Yeh
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan
| | - Chia-Tung Shen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Jou-Chien Chiang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsin-Mei Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Kuan-Wei Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Yuling Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - J Timothy Qiu
- International PhD Program in Cell Therapy and Regenerative Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Obstetrics and Gynecology, Taipei Medical University Hospital, Taipei 11031, Taiwan; Department of Obstetrics and Gynecology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
| | - Yu-Ting Yen
- Institute of Translational Medicine and New Drug Development, School of Medicine, China Medical University, Taichung, Taiwan.
| | - Kak-Shan Shia
- Institute of Biotechnology and Pharmaceutical Research, National Health Research Institutes, Miaoli County 35053, Taiwan.
| | - Yunching Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu 30013, Taiwan; Department of Chemistry, National Tsing Hua University, Hsinchu 30013, Taiwan.
| |
Collapse
|
22
|
de Zawadzki A, Leeming DJ, Sanyal AJ, Anstee QM, Schattenberg JM, Friedman SL, Schuppan D, Karsdal MA. Hot and cold fibrosis: The role of serum biomarkers to assess immune mechanisms and ECM-cell interactions in human fibrosis. J Hepatol 2025:S0168-8278(25)00148-5. [PMID: 40056933 DOI: 10.1016/j.jhep.2025.02.039] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 05/24/2025]
Abstract
Fibrosis is a pathological condition characterised by excessive accumulation of extracellular matrix (ECM) components, particularly collagens, leading to tissue scarring and organ dysfunction. In fibrosis, an imbalance between collagen synthesis (fibrogenesis) and degradation (fibrolysis) results in the deposition of fibrillar collagens disrupting the structural integrity of the ECM and, consequently, tissue architecture. Fibrosis is associated with a wide range of chronic diseases, including cirrhosis, kidney fibrosis, pulmonary fibrosis, and autoimmune diseases. Recently, the concept of "hot" and "cold" fibrosis has emerged, referring to the immune status within fibrotic tissues and the nature of fibrogenic signalling. Hot fibrosis is characterised by active immune cell infiltration and inflammation, while cold fibrosis is associated with auto- and paracrine myofibroblast activation, immune cell exclusion and quiescence. In this article, we explore the relationship between hot and cold fibrosis, the role of various types of collagens and their biologically active fragments in modulating the immune system, and how serological ECM biomarkers can help improve our understanding of the disease-relevant interactions between immune and mesenchymal cells in fibrotic tissues. Additionally, we draw lessons from immuno-oncology research in solid tumours to shed light on potential strategies for fibrosis treatment and highlight the advantage of having a "hot fibrotic environment" to treat fibrosis by enhancing collagen degradation through modulation of the immune system.
Collapse
Affiliation(s)
| | - Diana J Leeming
- Nordic Bioscience A/S, Biomarkers & Research, Herlev, Denmark
| | - Arun J Sanyal
- Stravitz-Sanyal Institute for Liver Disease and Metabolic Health and Division of Gastroenterology, Hepatology and Nutrition, Virginia Commonwealth University School of Medicine, Richmond, VA, USA
| | - Quentin M Anstee
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle Upon Tyne, UK; Newcastle NIHR Biomedical Research Center, Newcastle Upon Tyne Hospitals NHS Trust, Newcastle Upon Tyne, UK
| | - Jörn M Schattenberg
- Department of Internal Medicine II, Saarland University Medical Centre, Homburg, Germany
| | - Scott L Friedman
- Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Detlef Schuppan
- Institute of Translational Immunology and Research Center for Immune Therapy, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany; Division of Gastroenterology, Beth Israel Deaconess Medical Center, Boston, Harvard Medical School, MA, USA
| | | |
Collapse
|
23
|
Zhang X, Lou Y, Zheng S, Chang X. HCC-derived CX3CL1 affects hepatocellular carcinoma prognosis and CX3CR1 + MDSC infiltration. Eur J Med Res 2025; 30:153. [PMID: 40051011 PMCID: PMC11884201 DOI: 10.1186/s40001-025-02410-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] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 02/26/2025] [Indexed: 03/10/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related mortality worldwide, largely because of its ability to reshape the tumor microenvironment and evade immune surveillance. METHODS CX3CL1 expression in HCC tumor tissues was assessed via immunohistochemistry, while plasma levels were quantified using enzyme-linked immunosorbent assays (ELISAs). CX3CR1-positive immune cell infiltration was analyzed by immunofluorescence. The associations among CX3CL1 expression, CX3CR1-positive cell infiltration, and patient prognosis were examined. Additionally, cell-based assays were conducted to evaluate the impact of CX3CL1 amplification on the expression of myeloid-derived suppressor cell (MDSC)-recruiting factors. RESULTS Elevated CX3CL1 levels were significantly correlated with increased MDSC infiltration in the tumor microenvironment and poorer patient prognosis. CX3CL1 amplification led to the upregulation of MDSC-recruiting factors, suggesting a potential mechanism for immune evasion. CONCLUSIONS These findings highlight the possible involvement of CX3CL1 in HCC progression via MDSC recruitment, suggesting that it is a promising therapeutic target for promoting antitumor immunity. Further studies are needed to confirm these findings and explore potential therapeutic strategies.
Collapse
Affiliation(s)
- Xiaoling Zhang
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China
| | - Yidan Lou
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China
- Zhejiang University School of Medicine, Hangzhou, 310006, China
| | - Song Zheng
- Department of Medical Oncology, Hangzhou First People's Hospital, Hangzhou, 310006, China.
- Key Laboratory of Clinical Cancer Pharmacology and Toxicology Research of Zhejiang Province, Hangzhou First People's Hospital, Hangzhou, 310006, China.
| | - Xu Chang
- Department of Interventional Therapy II, Shandong Cancer Hospital and Institute, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong, China.
| |
Collapse
|
24
|
Liu Y, Zhang J, Lai C, Wang W, Huang Y, Bao X, Yan H, Sun X, Liu Q, Chen D, Dai X, Qian X, Zhao P. Injectable celastrol-loading emulsion hydrogel for immunotherapy of low-immunogenic cancer. J Nanobiotechnology 2025; 23:183. [PMID: 40050985 PMCID: PMC11887069 DOI: 10.1186/s12951-025-03154-y] [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: 09/29/2024] [Accepted: 01/22/2025] [Indexed: 03/09/2025] Open
Abstract
Immunotherapy, exemplified by immune checkpoint blockade (ICB), has been extensively employed in antitumor treatments. Nevertheless, its efficacy in addressing low-immunogenic tumors has not yielded satisfactory results, primarily due to the depletion and inadequate infiltration of effector T cells within the tumor microenvironment (TME). Here, we construct an injectable water-in-oil emulsion hydrogel to load clinically used Celastrol (Gel@Cel), which addresses the limitations of Cel's hydrophobicity. Cel can both inhibit tumor cell proliferation and promote tumor cell apoptosis, while simultaneously inducing immunogenic cell death, through activation of the AKT and MAPK pathways. In a model of clinically refractory hepatocellular carcinoma with malignant ascites, intraperitoneal administration of Gel@Cel significantly inhibits tumor progression and activates antitumor immune effects through lipase-controlled release of Cel, as compared to free Cel. Intriguingly, the Gel@Cel induces the activation of dendritic cells, resulting in the infiltration of cytotoxic T cells in the TME of ascites. Furthermore, the administration of Cel increases the expression of programmed cell death protein ligand-1 (PD-L1) in tumor cells. Moreover, combining the PD-1 antibody (αPD-1) with Gel@Cel further enhances the antitumor effect and amplifies the immune activation. In conclusion, Gel@Cel exhibits promising therapeutic potential in the treatment of low-immunogenic tumors, especially when combined with ICB therapy.
Collapse
Affiliation(s)
- Yu Liu
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
| | - Jia Zhang
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
- College of Energy Engineering, State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou, 310027, China
| | - Chunyu Lai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
| | - Wenjun Wang
- Department of Plastic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Yangyue Huang
- Department of Hepatobiliary Pancreatic Oncology, Cancer Center, Integrated Hospital of Traditional Chinese Medicine, Southern Medical University, Guangzhou, 510000, China
| | - Xuanwen Bao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China
| | - Haimeng Yan
- College of Medicine, Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University, Qingchun Road 79, Hangzhou, 310003, China
| | - Xuqi Sun
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China
| | - Qiqi Liu
- Department of Radiation Oncology, The First Affiliated Hospital, Zhejiang University, Hangzhou, 310003, China
| | - Dong Chen
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China.
- College of Energy Engineering, State Key Laboratory of Clean Energy Utilization, Zhejiang University, Hangzhou, 310027, China.
| | - Xiaomeng Dai
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
- Hubei Key Laboratory of Precision Radiation Oncology, Wuhan, 430022, China.
| | - Xinyu Qian
- Department of Medical Oncology, Hangzhou Cancer Hospital, Hangzhou, 310002, China.
| | - Peng Zhao
- Department of Medical Oncology, The First Affiliated Hospital, School of Medicine, Zhejiang University, #79 Qingchun Road, Hangzhou, 310003, China.
- National Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
25
|
Liu L, Wang L, Ding Y, Zhang Q, Shu Y. Cost-effectiveness of atezolizumab plus bevacizumab versus sorafenib as first-line therapy in unresectable hepatocellular carcinoma in the US and Chinese setting: a modelling comparison study. BMJ Open 2025; 15:e094804. [PMID: 40050065 PMCID: PMC11887288 DOI: 10.1136/bmjopen-2024-094804] [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: 10/09/2024] [Accepted: 02/05/2025] [Indexed: 03/09/2025] Open
Abstract
OBJECTIVE Atezolizumab plus bevacizumab demonstrates a significant improvement in overall survival and progression-free survival compared with sorafenib in patients with unresectable hepatocellular carcinoma (HCC). The combined usage of these two medications could result in substantial consumption of resources, primarily due to their exceptionally high costs. The current study aims to evaluate the cost-effectiveness of atezolizumab plus bevacizumab as a first-line treatment for advanced HCC from the perspective of payers in developed and developing countries. DESIGN A partitioned survival model was constructed to evaluate the cost-effectiveness of atezolizumab plus bevacizumab versus sorafenib as a first-line treatment for advanced HCC. The efficacy and safety data incorporated within the model were derived from the IMbrave150 trial. Costs and utilities were extracted from published sources. INTERVENTIONS Atezolizumab plus bevacizumab versus sorafenib. OUTCOME MEASURES Estimates were calculated for costs, life-years, quality-adjusted life-years (QALYs), incremental cost-effectiveness ratio (ICER) for both treatment strategies. One-way sensitivity, probabilistic sensitivity, expected value of perfect information (EVPI), subgroup and scenario analyses were conducted. RESULTS The combination therapy of atezolizumab and bevacizumab results in an additional 0.72 life-years/0.57 QALYs in the USA and 0.64 life-years/0.47 QALYs in China compared with standard sorafenib treatment, although with a significant increase in costs, yielding an average ICER of US$253 247.07/QALY in the USA and US$181 552.71/QALY in China. The probability sensitivity analysis indicated that atezolizumab plus bevacizumab demonstrated a 13.60% likelihood of cost-effectiveness in the USA, whereas this likelihood is negligible (0%) in China. The expected value of uncertainty, as quantified by the EVPI, was estimated at approximately US$3658.41/patient in the USA and US$0/patient in China. The ICER was most sensitive to the cost of subsequent treatment in the USA, and most sensitive to the cost of atezolizumab in China. In scenario analyses, the atezolizumab plus bevacizumab treatment becomes favourable when the cost of atezolizumab decreases to 67.85% and 18.45% of its original price in the USA and China, respectively. CONCLUSIONS The atezolizumab plus bevacizumab is unlikely to be cost-effective compared with sorafenib for patients with unresectable HCC in the context of the USA and China. The implementation of significant reductions in drug prices may render the treatment economically viable.
Collapse
Affiliation(s)
- Lulu Liu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
- Department of Pharmacy, The Second Affiliated Hospital of Army Medical University, Chongqing, China
| | - Lei Wang
- Outpatient Department Office, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiling Ding
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Qilin Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Yamin Shu
- Department of Pharmacy, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
26
|
Yu J, Li Y, Yu J, Yang Y, Chen Y, Yi P. Hepatic arterial infusion chemotherapy enhances the efficacy of lenvatinib and PD-1 inhibitors for advanced hepatocellular carcinoma: A meta-analysis and trial sequential analysis. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109573. [PMID: 39793379 DOI: 10.1016/j.ejso.2025.109573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 01/02/2025] [Indexed: 01/13/2025]
Abstract
BACKGROUND Hepatic arterial infusion chemotherapy (HAIC) was an effective treatment for advanced hepatocellular carcinoma (HCC), and its effectiveness in combination with targeted immunotherapy regimens was controversial. This meta-analysis was performed to evaluate the efficacy of adding HAIC to lenvatinib in combination with programmed death-1 (PD-1) inhibitors. METHODS Literature related to the efficacy of HAIC in combination with lenvatinib plus PD-1 inhibitors in the treatment of advanced HCC was searched through PubMed, Cochrane Library, Embase, and Web of Science databases. TSA was used to control for the risk of random error and assess whether the meta-analysis evidence was conclusive. RESULTS Eight relevant papers with a total of 1244 patients. Compared with the L-P treatment group, the H-L-P treatment group significantly prolonged OS (hazard ratio [HR] 2.11 [95 % confidence interval (CI) 1.82-2.44]; p < 0.001) and PFS (HR 1.91 [95 % CI 1.67-2.17]; p < 0.001) and improved ORR (risk ratio [RR] 2.20 [95 % CI 1.74-2.78]; p < 0.001) and DCR (RR 1.28 [95 % CI 1.15-1.42]; p < 0.001) in patients with advanced HCC. TSA analysis indicated that further trials were unnecessary, preliminary positive results were promptly obtained. Prognostic factor analysis demonstrated that extrahepatic metastasis were common independent risk factor for OS and PFS. The rate of adverse events (AEs) was higher in the H-L-P treatment group than in the L-P treatment group. CONCLUSION HAIC combined with lenvatinib plus PD-1 inhibitors markedly extended OS and PFS, particularly in patients without extrahepatic metastases. Furthermore, it markedly enhanced ORR and DCR in patients with HCC.
Collapse
Affiliation(s)
- Jiahui Yu
- Department of Hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Yong Li
- Department of Hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Jinxin Yu
- North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Yuting Yang
- Department of Educational Technology, Institute of Education, China West Normal University, Nanchong, Sichuan, 637000, PR China; Nanchong Gaoping District Wangcheng Primary School, Nanchong, Sichuan, 637100, PR China
| | - Yimiao Chen
- Department of Hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China
| | - Pengsheng Yi
- Department of Hepato-biliary-pancrease II, Affiliated Hospital of North Sichuan Medical College, Nanchong, Sichuan, 637000, PR China.
| |
Collapse
|
27
|
Du F, Wang G, Dai Q, Huang J, Li J, Liu C, Du K, Tian H, Deng Q, Xie L, Zhao X, Zhang Q, Yang L, Li Y, Wu Z, Zhang Z. Targeting novel regulated cell death: disulfidptosis in cancer immunotherapy with immune checkpoint inhibitors. Biomark Res 2025; 13:35. [PMID: 40012016 DOI: 10.1186/s40364-025-00748-4] [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: 01/07/2025] [Accepted: 02/11/2025] [Indexed: 02/28/2025] Open
Abstract
The battle against cancer has evolved over centuries, from the early stages of surgical resection to contemporary treatments including chemotherapy, radiation, targeted therapies, and immunotherapies. Despite significant advances in cancer treatment over recent decades, these therapies remain limited by various challenges. Immune checkpoint inhibitors (ICIs), a cornerstone of tumor immunotherapy, have emerged as one of the most promising advancements in cancer treatment. Although ICIs, such as CTLA-4 and PD-1/PD-L1 inhibitors, have demonstrated clinical efficacy, their therapeutic impact remains suboptimal due to patient-specific variability and tumor immune resistance. Cell death is a fundamental process for maintaining tissue homeostasis and function. Recent research highlights that the combination of induced regulatory cell death (RCD) and ICIs can substantially enhance anti-tumor responses across multiple cancer types. In cells exhibiting high levels of recombinant solute carrier family 7 member 11 (SLC7A11) protein, glucose deprivation triggers a programmed cell death (PCD) pathway characterized by disulfide bond formation and REDOX (reduction-oxidation) reactions, termed "disulfidptosis." Studies suggest that disulfidptosis plays a critical role in the therapeutic efficacy of SLC7A11high cancers. Therefore, to investigate the potential synergy between disulfidptosis and ICIs, this study will explore the mechanisms of both processes in tumor progression, with the goal of enhancing the anti-tumor immune response of ICIs by targeting the intracellular disulfidptosis pathway.
Collapse
Affiliation(s)
- Fei Du
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| | - Guojun Wang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qian Dai
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Jiang Huang
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pharmacy, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Junxin Li
- Department of pharmacy, Zigong Fourth People's Hospital, Zigong, 643000, China
| | - Congxing Liu
- Department of Pharmacy, Chengfei Hospital, Chengdu, 610000, China
| | - Ke Du
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China
- Department of Pediatrics, Luzhou Maternal and Child Health Hospital, Luzhou Second People's Hospital, Luzhou, 646000, Sichuan, China
| | - Hua Tian
- School of Nursing, Chongqing College of Humanities, Science & Technology, Chongqing, 401520, China
| | - Qiwei Deng
- Heruida Pharmaceutical Co.,ltd, Haikou, Hainan, 570100, China
| | - Longxiang Xie
- The TCM Hospital of Longquanyi District, Chengdu, 610100, Sichuan, China
| | - Xin Zhao
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Qimin Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Lan Yang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China
| | - Yaling Li
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhigui Wu
- Department of Pharmacy, The Affiliated Hospital of Southwest Medical University, Luzhou, 646000, Sichuan, China
| | - Zhuo Zhang
- Department of Pharmacy, The Fourth Affiliated Hospital Of Southwest Medical University, Meishan, 620000, Sichuan, China.
- Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou, 646000, Sichuan, China.
| |
Collapse
|
28
|
Liu M, Li L, Cao L, Li W, Gu X, Yang M, Wu D, Li Y, Deng Y, Zhang J, Yang C, Liang Q, Liu H, Rong P, Ma X, Wang W. Targeted delivery of CCL3 reprograms macrophage antigen presentation and enhances the efficacy of immune checkpoint blockade therapy in hepatocellular carcinoma. J Immunother Cancer 2025; 13:e010947. [PMID: 39988347 PMCID: PMC11848677 DOI: 10.1136/jitc-2024-010947] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Accepted: 02/05/2025] [Indexed: 02/25/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a leading cause of cancer-related deaths worldwide, especially in advanced stages where limited treatment options result in poor prognosis. The immunosuppressive tumor immune microenvironment (TIME), characterized by low immune cell infiltration and exhaustion, limits immunotherapy efficacy. To address this, our study investigates the role of C-C motif chemokine ligand 3 (CCL3) in modulating the HCC TIME. METHODS We analyzed CCL3 expression in human HCC samples from The Cancer Genome Atlas database, focusing on its correlation with inflammatory gene signatures and immune cell infiltration. High-dimensional single-cell RNA sequencing (scRNA-seq), flow cytometry, and multiplex immunofluorescence were used to investigate CCL3's effects on macrophage function and T cell activation. The biological impact of CCL3 on macrophages was assessed using co-culture systems, confocal imaging, metabolite detection, and inhibition assays. Preclinical HCC models and ex vivo tumor fragment assays further explored how CCL3 modulates immune responses and enhances immune checkpoint blockade efficacy. RESULTS Our study shows that CCL3 is suppressed in the tumor microenvironment and positively correlates with immune infiltration and inflammatory responses. Targeted liver delivery of rAAV-Ccl3 reprograms the immune microenvironment in HCC, promoting immune cell recruitment and tertiary lymphoid structure formation, thus suppressing tumor growth via immune engagement. Through scRNA-seq, flow cytometry, and multiplex immunofluorescence, we found that CCL3 enhances macrophage antigen uptake and activates cytotoxic T cells. In vivo and in vitro experiments confirmed that CCL3 facilitates T cell infiltration and upregulates MHC II expression on macrophages, enhancing antigen presentation. The CCL3-CCR5 pathway also boosts macrophage metabolism, increasing lysosomal activity and antigen uptake, thereby strengthening adaptive immune responses and increasing sensitivity to immune checkpoint blockade therapies in preclinical models. CONCLUSIONS This study highlights the pivotal role of CCL3 in reshaping the TIME and enhancing antitumor immunity in HCC. By promoting immune cell recruitment and enhancing antigen presentation, CCL3 demonstrates significant potential to improve the efficacy of immunotherapy, particularly in combination with immune checkpoint inhibitors. Targeting CCL3 may help to overcome the immunosuppressive TIME in HCC and improve patient outcomes.
Collapse
Affiliation(s)
- Muqi Liu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Linzhe Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Lu Cao
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Wei Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Xingshi Gu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Min Yang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Di Wu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Yanan Li
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Yao Deng
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Juan Zhang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Cejun Yang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Qi Liang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Huaping Liu
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Pengfei Rong
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Xiaoqian Ma
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| | - Wei Wang
- Institute for Cell Transplantation and Gene Therapy, Third Xiangya Hospital of Central South University, Changsha, Hunan, China
- Clinical Research Center for Minimally Invasive Diagnosis and Therapy Under Image Navigation, Changsha, Hunan, China
| |
Collapse
|
29
|
Piao Y, Zhai N, Zhang X, Zhao W, Li M. Post-translational modifications in hepatocellular carcinoma: unlocking new frontiers in immunotherapy. Front Immunol 2025; 16:1554372. [PMID: 40040703 PMCID: PMC11876159 DOI: 10.3389/fimmu.2025.1554372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Accepted: 01/31/2025] [Indexed: 03/06/2025] Open
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), is one of the most common and aggressive malignancies worldwide. Immunotherapy has shown promising results in treating HCC, but its efficacy is often limited by complex mechanisms of immune evasion. Post-translational modifications (PTMs) of proteins play a critical role in regulating the immune responses within the tumor microenvironment (TME). These modifications influence protein function, stability, and interactions, which either promote or inhibit immune cell activity in cancer. In this mini-review, we explore the diverse PTMs that impact immune evasion in liver cancer, including glycosylation, phosphorylation, acetylation, and ubiquitination. We focus on how these PTMs regulate key immune checkpoint molecules such as PD-L1, CTLA-4, and the TCR complex. Furthermore, we discuss the potential of targeting PTMs in combination with existing immunotherapies to enhance the effectiveness of treatment in HCC. Understanding the role of PTMs in immune regulation may lead to the development of novel therapeutic strategies to overcome resistance to immunotherapy in liver cancer.
Collapse
Affiliation(s)
- Yuexian Piao
- Department of Interventional Therapy, First Hospital of Jilin University, Changchun, China
| | - Naicui Zhai
- Core Facility of First Hospital of Jilin University, Changchun, China
| | - Xiaoling Zhang
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Wenjie Zhao
- Key Laboratory of Organ Regeneration and Transplantation of Ministry of Education, First Hospital of Jilin University, Changchun, China
- National-Local Joint Engineering Laboratory of Animal Models for Human Disease, First Hospital of Jilin University, Changchun, China
| | - Min Li
- Department of Interventional Therapy, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
30
|
Hong X, Guo Y, Shi W, Zhu K, Liang L, Lin L, Chen Y, Zhou J, Huang J, Huang J, Wu Y, Huang W, Cai M. Donafenib combined with sintilimab for advanced hepatocellular carcinoma: a single arm phase II trial. BMC Cancer 2025; 25:205. [PMID: 39910472 PMCID: PMC11796178 DOI: 10.1186/s12885-025-13605-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 01/29/2025] [Indexed: 02/07/2025] Open
Abstract
BACKGROUND Previous studies evaluating antiangiogenic agents plus immune checkpoint inhibitors for unresectable hepatocellular carcinoma (HCC) have shown encouraging results. This study was conducted to investigate the efficacy and safety of donafenib combined with sintilimab (Don-Sin) for advanced HCC. METHODS This was a single-center, single-arm phase II trial recruiting patients with BCLC stage C HCC. A safety run-in cohort was planned with the first 6 patients receiving oral donafenib 200 mg twice daily and intravenous sintilimab 200 mg once every 3 weeks. Dose-limiting toxicities (DLTs) were evaluated to determine the recommended dose of donafenib for those enrolled thereafter. The primary endpoint of this study was progression-free survival (PFS) per mRECIST. RESULTS 30 patients were enrolled. As 3 patients (50.0%) experienced DLTs during safety run-in, the initial dose of donafenib was adjusted to 200 mg once daily for subsequent patients. The primary endpoint was met with a median PFS of 6.2 (95% confidence interval [CI], 4.4-8.0) months per mRECIST (6.3 [95% CI, 5.4-7.2] months per RECIST 1.1). The objective response rate was 23.3% per mRECIST and 16.7% per RECIST 1.1, while the disease control rate reached 76.7% per mRECIST/RECIST 1.1. The median overall survival was 16.0 (95% CI, 13.5-18.5) months. Treatment-related adverse events (TRAEs) occurred in 28 patients (93.3%) and grade 3 TRAEs were observed in 9 patients (30.0%). CONCLUSIONS Don-Sin showed promising antitumor effects with an acceptable safety profile in patients with advanced stage HCC. The preliminary findings need to be further evaluated in phase III randomized controlled trials. TRIAL REGISTRATION ClinicalTrials.gov (identifier: NCT05162352; date of registration: December 4, 2021).
Collapse
Affiliation(s)
- Xiaoyang Hong
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Department of Radiology, Yuebei People's Hospital, Shaoguan, China
| | - Yongjian Guo
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Wenbo Shi
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- Shanghai Clinical Research Ward (SCRW), Shanghai Sixth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Licong Liang
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Liteng Lin
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Ye Chen
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingwen Zhou
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jingjun Huang
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Jiabai Huang
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
| | - Yaozhu Wu
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China
| | - Wensou Huang
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
| | - Mingyue Cai
- Department of Minimally Invasive Interventional Radiology, the Second Affiliated Hospital, Guangzhou Medical University, Guangzhou, China.
- The Second School of Clinical Medicine, Guangzhou Medical University, Guangzhou, China.
| |
Collapse
|
31
|
Yao CY, Tao HT, He JJ, Zhu FY, Xie CQ, Cheng YN, Li JQ, Liu ZZ, Hou CY, Liu XL, Fan YL, Fang D, Lv XR. NUAK1 acts as a novel regulator of PD-L1 via activating GSK-3β/β-catenin pathway in hepatocellular carcinoma. Mol Med 2025; 31:38. [PMID: 39901136 PMCID: PMC11789290 DOI: 10.1186/s10020-025-01088-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Accepted: 01/16/2025] [Indexed: 02/05/2025] Open
Abstract
BACKGROUND NUAK1 is associated with metastasis and drug resistance in hepatocellular carcinoma (HCC). However, little is known about the immune functions of NUAK1 in HCC. Therefore, the aim of this study was to elucidate the novel role of NUAK1 in facilitating immune evasion in HCC and to investigate the mechanisms underpinning this process. METHOD The levels of NUAK1 expression and the infiltration of CD8+ T cells were assessed in tumor tissues from HCC patients and mice xenograft model. HCC cell lines were used to validate the role of NUAK1 in regulating the transcription of PD-L1, the diethylnitrosamine-induced HCC model was established and the expression levels of NUAK1 and PD-L1 proteins in the rat livers were detected. Western blotting, immunofluorescence, real time PCR, and immunohistochemical staining were used to investigate the underlying mechanisms by which NUAK1 regulates PD-L1 expression in hepatocellular carcinoma. RESULTS NUAK1 expression was negatively correlated with CD8+ T cell infiltration in tumor tissues from HCC patients and mice xenograft model. Both gain and loss of functions have identified NUAK1 promoted PD-L1 expression at transcriptional level in HCC cells. The increased expression of NUAK1 and PD-L1 proteins were observed in the rat livers of diethylnitrosamine-induced HCC model. Moreover, overexpression of NUAK1 promotes GSK3β Ser9 phosphorylation, β-catenin expression and nuclear accumulation in HCC cells. By contrast, knockdown of NUAK1 has opposite effects. Inhibition of GSK3β activity significantly promoted β-catenin expression and PD-L1 expression in HCC cells. IHC analyses of tumor tissues from HCC patients suggested that the levels of p-GSK3β and β-catenin were positively correlated with NUAK1 expression. Knockdown of β-catenin also reversed NUAK1-mediated PD-L1 expression in HCC cells. CONCLUSIONS This study revealed a novel role for NUAK1, which promotes the transcriptional expression of PD-L1 by activating GSK3β/β-catenin signaling pathway, leading to immune escape of hepatocellular carcinoma. Registry and the registration no. of the study/trial: Not applicable.
Collapse
Affiliation(s)
- Chao-Yan Yao
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Hang-Tian Tao
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Jin-Jin He
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China
| | - Feng-Yi Zhu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Cui-Qing Xie
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yu-Na Cheng
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Ji-Qin Li
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Zhuang-Zhuang Liu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Chun-Yu Hou
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Xue-Li Liu
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China
| | - Yong-Li Fan
- Department of Oncology, The First Affiliated Hospital of Henan University, Kaifeng, 475000, China.
| | - Dong Fang
- Department of Pharmacy, The First Affiliated Hospital of Henan University, Kaifeng, 475004, China.
- Institute of Chemical Biology, School of Pharmacy, Henan University, N. Jinming Ave, Kaifeng, 475004, China.
| | - Xin-Rui Lv
- Kaifeng Key Laboratory for Infectious Diseases and Biosafety, The First Affiliated Hospital of Henan University, Ximen Ave, Kaifeng, 475000, China.
| |
Collapse
|
32
|
Ren T, Huang Y. Recent advancements in improving the efficacy and safety of chimeric antigen receptor (CAR)-T cell therapy for hepatocellular carcinoma. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:1433-1446. [PMID: 39316087 DOI: 10.1007/s00210-024-03443-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 09/08/2024] [Indexed: 09/25/2024]
Abstract
The liver is one of the most frequent sites of primary malignancies in humans. Hepatocellular carcinoma (HCC) is one of the most prevalent solid tumors with poor prognosis. Current treatments showed limited efficacy in some patients, and, therefore, alternative strategies, such as immunotherapy, cancer vaccines, adoptive cell therapy (ACT), and recently chimeric antigen receptors (CAR)-T cells, are developed to offer better efficacy and safety profile in patients with HCC. Unlike other ACTs like tumor-infiltrating lymphocytes (TILs), CAR-T cells are equipped with engineered CAR receptors that effectively identify tumor antigens and eliminate cancer cells without major histocompatibility complex (MHC) restriction. This process induces intracellular signaling, leading to T lymphocyte recruitment and subsequent activation of other effector cells in the tumor microenvironment (TME). Until today, novel approaches have been used to develop more potent CAR-T cells with robust persistence, specificity, trafficking, and safety. However, the clinical application of CAR-T cells in solid tumors is still challenging. Therefore, this study aims to review the advancement, prospects, and possible avenues of CAR-T cell application in HCC following an outline of the CAR structure and function.
Collapse
Affiliation(s)
- Tuo Ren
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongsahn 2nd Road, Guangzhou, Guangdong, 510080, China
| | - Yonghui Huang
- Department of Interventional Radiology, The First Affiliated Hospital, Sun Yat-Sen University, No.58 Zhongsahn 2nd Road, Guangzhou, Guangdong, 510080, China.
| |
Collapse
|
33
|
Sun Y, Shi G, Yang J, Zhou CZ, Peng C, Luo YH, Pan Y, Wang RQ. Deciphering the heterogeneity and plasticity of the tumor microenvironment in liver cancer provides insights for prognosis. Front Pharmacol 2025; 16:1495280. [PMID: 39950116 PMCID: PMC11821625 DOI: 10.3389/fphar.2025.1495280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025] Open
Abstract
Liver cancer exhibits diverse molecular characteristics and distinct immune cell infiltration patterns, which significantly influence patient outcomes. In this study, we thoroughly examined the liver cancer tumor environment by analyzing data from 419,866 individual cells across nine datasets involving 99 patients. By categorizing patients into different groups based on their immune cell profiles, including immune deficiency, B cells-enriched, T cells-enriched and macrophages-enriched, we better understood how these cells change in various patient subgroups. Our investigation of liver metastases from intestinal cancer uncovered a group of mast cells that might promote metastasis through pathways like inositol phosphate metabolism. Using genomic and clinical data from The Cancer Genome Atlas, we identified specific cell components linked to tumor characteristics and genetics. Our detailed study of cancer-associated fibroblasts (CAFs) revealed how they adapt and acquire new functions in the tissue environment, highlighting their flexibility. Additionally, we found a significant connection between CAF-related genes and the prognosis of hepatocellular carcinoma patients. This research provides valuable insights into the makeup of the liver cancer tumor environment and its profound impact on patient outcomes, offering fresh perspectives for managing this challenging disease.
Collapse
Affiliation(s)
- Yihao Sun
- Department of Pharmacy, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
- Guangdong Provincial Key Laboratory of Tumor Interventional Diagnosis and Treatment, Zhuhai Institute of Translational Medicine, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Guojuan Shi
- Department of Nephrology, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Jian Yang
- Department of Respiratory and Critical Care Medicine, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Chun-Zhong Zhou
- Department of Pharmacy, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Chuhan Peng
- Canyon Crest Academy, San Diego, CA, United States
| | - Yu-Hong Luo
- Department of Pharmacy, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Ying Pan
- Department of Oncology, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| | - Rui-Qi Wang
- Department of Pharmacy, Zhuhai People’s Hospital (The Affiliated Hospital of Beijing Institute of Technology, Zhuhai Clinical Medical College of Jinan University), Zhuhai, China
| |
Collapse
|
34
|
Maravelia P, Yao H, Cai C, Nascimento Silva D, Fransson J, Nilsson OB, Lu YCW, Micke P, Botling J, Gatto F, Rovesti G, Carlsten M, Sallberg M, Stål P, Jorns C, Buggert M, Pasetto A. Unlocking novel T cell-based immunotherapy for hepatocellular carcinoma through neoantigen-driven T cell receptor isolation. Gut 2025:gutjnl-2024-334148. [PMID: 39832892 DOI: 10.1136/gutjnl-2024-334148] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Accepted: 12/23/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Tumour-infiltrating T cells can mediate both antitumour immunity and promote tumour progression by creating an immunosuppressive environment. This dual role is especially relevant in hepatocellular carcinoma (HCC), characterised by a unique microenvironment and limited success with current immunotherapy. OBJECTIVE We evaluated T cell responses in patients with advanced HCC by analysing tumours, liver flushes and liver-draining lymph nodes, to understand whether reactive T cell populations could be identified despite the immunosuppressive environment. DESIGN T cells isolated from clinical samples were tested for reactivity against predicted neoantigens. Single-cell RNA sequencing was employed to evaluate the transcriptomic and proteomic profiles of antigen-experienced T cells. Neoantigen-reactive T cells expressing 4-1BB were isolated and characterised through T-cell receptor (TCR)-sequencing. RESULTS Bioinformatic analysis identified 542 candidate neoantigens from seven patients. Of these, 78 neoantigens, along with 11 hotspot targets from HCC driver oncogenes, were selected for ex vivo T cell stimulation. Reactivity was confirmed in co-culture assays for 14 targets, with most reactive T cells derived from liver flushes and lymph nodes. Liver flush-derived T cells exhibited central memory and effector memory CD4+ with cytotoxic effector profiles. In contrast, tissue-resident memory CD4+ and CD8+ T cells with an exhausted profile were primarily identified in the draining lymph nodes. CONCLUSION These findings offer valuable insights into the functional profiles of neoantigen-reactive T cells within and surrounding the HCC microenvironment. T cells isolated from liver flushes and tumour-draining lymph nodes may serve as a promising source of reactive T cells and TCRs for further use in immunotherapy for HCC.
Collapse
Affiliation(s)
- Panagiota Maravelia
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Karolinska ATMP center, Karolinska Institutet, Stockholm, Sweden
| | - Haidong Yao
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Karolinska ATMP center, Karolinska Institutet, Stockholm, Sweden
| | - Curtis Cai
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institute, Stockholm, Sweden
| | - Daniela Nascimento Silva
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Karolinska ATMP center, Karolinska Institutet, Stockholm, Sweden
| | - Jennifer Fransson
- Dept of Cell and Molecular Biology, National Bioinformatics Infrastructure Sweden, Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | | | - Yong-Chen William Lu
- Department of Pathology, Winthrop P. Rockefeller Cancer Institute, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Patrick Micke
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology, Uppsala University, Uppsala, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Francesca Gatto
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Department of Laboratory Medicine, Division of Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Giulia Rovesti
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Division of Oncology, Department of Medical and Surgical Sciences for Children & Adults, University-Hospital of Modena and Reggio Emilia, Modena, Italy
- Clinical and Experimental Medicine PhD Program, University of Modena and Reggio Emilia, Modena, Italy
| | - Mattias Carlsten
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Center for Cell Therapy and Allogeneic Stem Cell Transplantation, Karolinska Comprehensive Cancer Center, Karolinska University Hospital, Karolinska Institutet, Stockholm, Sweden
| | - Matti Sallberg
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Karolinska ATMP center, Karolinska Institutet, Stockholm, Sweden
| | - Per Stål
- Department of Medicine Huddinge, Karolinska Institutet, Stockholm, Sweden
| | - Carl Jorns
- Department of Transplantation Surgery, Karolinska University Hospital, Division of Transplantation, Clinical Science, Intervention and Technology, Karolinska Institutet, Stockholm, Sweden
| | - Marcus Buggert
- Department of Medicine Huddinge, Center for Infectious Medicine, Karolinska Institute, Stockholm, Sweden
| | - Anna Pasetto
- Division of Clinical Microbiology,Department of Laboratory Medicine, Karolinska Institutet, Stockholm, Sweden, Stockholm, Sweden
- Karolinska ATMP center, Karolinska Institutet, Stockholm, Sweden
- Section for Cell Therapy, Radiumhospitalet, Oslo University Hospital, Oslo, Norway
- Department of Oncology, Institute of Clinical Medicine, University of Oslo, Norway, Oslo, Norway
| |
Collapse
|
35
|
Han H, zhang C, Shi W, Wang J, Zhao W, Du Y, Zhao Z, Wang Y, Lin M, Qin L, Zhao X, Yin Q, Liu Y, Wang Z, Zhang J, You X, Zhou G, Wu H, Ye J, He X, Tian W, Yu H, Yuan Y, Wang Q. NSUN5 Facilitates Hepatocellular Carcinoma Progression by Increasing SMAD3 Expression. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2404083. [PMID: 39531371 PMCID: PMC11727281 DOI: 10.1002/advs.202404083] [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] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 10/06/2024] [Indexed: 11/16/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by frequent intrahepatic and distant metastases, resulting in a poor prognosis for patients. Epithelial-mesenchymal transition (EMT) plays a pivotal role in this process. However, the expression of NOP2/Sun RNA methyltransferase 5 (NSUN5) in HCC and its role in mediating EMT remain poorly understood. In this study, clinicopathological analyses are conducted across multiple independent HCC cohorts and induced tumor formation in Nsun5-knockout mice. The findings reveal an upregulation of NSUN5 expression in tumor tissues; conversely, the absence of Nsun5 hinders the malignant progression of HCC, indicating that NSUN5 may serve as a significant oncogene in HCC. Furthermore, elevated levels of NSUN5 enhance EMT processes within HCC cells. NSUN5-knockout cells exhibit reduced invasion and migration capabilities under both in vivo and in vitro conditions, while overexpression of NSUN5 yields opposing effects. Mechanistically, high levels of NSUN5 promote the enrichment of trimethylated histone H3 at lysine 4 (H3K4me3) at the promoter region of SMAD3 through recruitment of the WDR5, thereby facilitating HCC metastasis via SMAD3-mediated EMT pathways. Collectively, this study identifies NSUN5 as a novel driver of metastasis in HCC and provides a theoretical foundation for potential therapeutic strategies against this malignancy.
Collapse
Affiliation(s)
- Hexu Han
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Chengcheng zhang
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Wenbo Shi
- Oncology majorRuijin‐Hainan HospitalShanghai Jiao Tong University School of MedicineHainan200032China
| | - Jiawei Wang
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
- Department of General SurgeryFirst Affiliated Hospital of Suzhou UniversitySuzhou215000P. R. China
| | - Wenhui Zhao
- Department of Basic MedicineJiangsu College of NursingHuai'anJiangsu223001P. R. China
| | - Yanping Du
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Zhibin Zhao
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Yifan Wang
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Maosong Lin
- Department of GastroenterologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Lei Qin
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
- Department of General SurgeryFirst Affiliated Hospital of Suzhou UniversitySuzhou215000P. R. China
| | - Xiaoxue Zhao
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Qianqian Yin
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Yiyi Liu
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Zhongqi Wang
- Department of Medical Oncology, Longhua Hospital Affiliated to Shanghai University of Traditional Chinese MedicineShanghai University of Traditional Chinese Medicine725 Wanpingnan RoadShanghai200032P. R. China
| | - Jing Zhang
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Xiaomin You
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Guoxiong Zhou
- Department of Gastroenterology, Affiliated Hospital of Nantong UniversityNantong UniversityJiangsu226001P. R. China
| | - Honghui Wu
- Reproduction Medicine CentreThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou225300P. R. China
| | - Jun Ye
- Center for Translational Medicine, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Xianzhong He
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityInnovative Institute of Tumor Immunity and Medicine (ITIM)Anhui Provincial Innovation Institute for Pharmaceutical Basic ResearchAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230000P. R. China
| | - Weizhong Tian
- Department of RadiologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Hong Yu
- Department of PathologyThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Yin Yuan
- Department of Hepatobiliary SurgeryThe Affiliated Taizhou People's Hospital of Nanjing Medical UniversityTaizhou School of Clinical MedicineNanjing Medical UniversityTaizhouJiangsu225300P. R. China
| | - Qiang Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityInnovative Institute of Tumor Immunity and Medicine (ITIM)Anhui Provincial Innovation Institute for Pharmaceutical Basic ResearchAnhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230000P. R. China
| |
Collapse
|
36
|
Wang F, Deng L, Li Z, Cao Q, Jiang R, Xu C, Yang J. Characterization of T-Cell Receptor Profiles Predicts Survival Situation in Patients with Hepatocellular Carcinoma. Technol Cancer Res Treat 2025; 24:15330338251329699. [PMID: 40152632 PMCID: PMC11954561 DOI: 10.1177/15330338251329699] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Revised: 02/27/2025] [Accepted: 03/02/2025] [Indexed: 03/29/2025] Open
Abstract
PurposeHepatocellular carcinoma (HCC) is the most common liver malignancy in the world, and tumor-infiltrating T cells have been shown to be closely related to the prognosis of HCC. This study investigated the potential and efficacy of T cell receptor (TCR) repertoire characterization as a biomarker for predicting survival differences.MethodsIn this study, we used high-throughput sequencing technology to systematically analyze the characteristics of TCR repertoires in tumor tissues obtained from 23 long-survivors and 8 short-survivors diagnosed with HCC.ResultsThe TCR composition in HCC long-survivors was found to be less diverse than in the short-survivors. In addition, in the context of V and J gene segments, long-survivors showed significantly higher usage of TRBJ1-3, TRBV10-1, TRBV15, and TRBV6-5, and lower usage of TRBJ2-2. Both principal component analysis (PCA) and the motif diagram of complementary determination region 3 (CDR3) sequences could clearly discriminate short- and long-survivors. And there were five up-regulated and one down-regulated CDR3 sequences in the LS group compared with the SS group. According to the characteristics of TCR repertoire, we also established the survival-related evaluation system and the prediction model for the survival period of HCC patients.ConclusionOur study adds to the existing knowledge of TCR rearrangement profiles in HCC patients by elucidating the differential TCR rearrangement profiles between long-term and short-term surviving HCC patients. Also, our analysis identified a number of TCR genes that are significantly associated with survival, and these may not only serve as prognostic biomarkers but may also play an important role in antigen-specific immunotherapy.
Collapse
Affiliation(s)
- Fengyan Wang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
- Shandong Provincial Key Laboratory of Neuroprotective Drugs, Zibo, China
| | - Li Deng
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Ziqiang Li
- Department of General Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Qiwei Cao
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Clinical Pathology, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, China
| | - Runze Jiang
- Innovative Institute of Chinese Medicine and Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Changqing Xu
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| | - Jing Yang
- Department of Gastroenterology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, China
| |
Collapse
|
37
|
Wang X, Jing Z, Huang X, Liu X, Zhang Y, Wang Z, Ma P. PD-L1 antibody conjugated dihydrotanshinone I-loaded polymeric nanoparticle for targeted cancer immunotherapy combining PD-L1 blockade with immunogenic cell death. Int J Pharm 2024; 667:125004. [PMID: 39608587 DOI: 10.1016/j.ijpharm.2024.125004] [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: 09/03/2024] [Revised: 11/07/2024] [Accepted: 11/24/2024] [Indexed: 11/30/2024]
Abstract
PURPOSE Combination immune checkpoint inhibitors (ICI) with chemotherapeutic agents has proven to be highly promising in cancer therapy. However, low response rate, immune-related adverse events, and lack of effectively targeted co-delivery strategy are still major hurdles to overcome for this combination therapeutic regimen. Herein, programmed death-L1 (PD-L1) antibody modified and dihydrotanshinone I (DHT) loaded nanoparticle was prepared for tumor targeting drug delivery, thus achieving immune checkpoint blockade (ICB) and immunogenic cell death (ICD) synergistic anti-tumor effects. METHODS The DHT-loaded nanoparticle (DHT NP) was prepared by the emulsion solvent diffusion method. Atezolizumab (ATEZO) was thiolated with 2-iminothiolane and conjugated to the surface of DHT NP to prepare the ATEZO DHT NP. The drug encapsulation efficiency, drug loading, particle size and drug release were determined. The in vitro cellular uptake, cell proliferation inhibition and apoptosis were evaluated on the HGC-27 tumor cell. The in vivo tumor targeting, anti-tumor efficiency and immune regulation were assessed on tumor bearing mice. RESULTS The optimized ATEZO DHT NP was a spherical nanoparticle of about 250 nm with a continuous drug release profile. It was selectively taken up by the tumor cells through PD-L1 receptor-mediated endocytosis, which resulted in enhanced cytotoxicity and cell apoptosis. In vivo imaging further demonstrated its superior tumor tissue targeting ability. When tumor bearing mice were treated with the ATEZO DHT NP, its synergistic anti-tumor effect was much stronger than that of a single drug. Moreover, the tumor targeting delivery of DHT caused tumor necrosis and initiated ICD with release of tumor-associated antigens, which efficiently up-regulated the population of CD4+ and CD8+ T cells. Notably, there were no obvious system toxicity or tissue damage occur during the whole treatment period. CONCLUSION The ATEZO DHT NP could specifically target to tumor and enhance treatment efficiency through combination of PD-L1 blockade with ICD effect.
Collapse
Affiliation(s)
- Xue Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Ziqi Jing
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaobin Huang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoya Liu
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China
| | - Yujie Zhang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| | - Zhijun Wang
- Department of Geriatric Medicine &National Clinical Research Center of Geriatric Disease, The 2nd Medical Center of Chinese PLA General Hospital, Beijing, China; Department of Interventional Radiology, The 1st & 5th Medical Center of Chinese PLA General Hospital, Beijing, China.
| | - Pengkai Ma
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, China.
| |
Collapse
|
38
|
Yin CQ, Song CQ. Tumor Intrinsic Immunogenicity Suppressor SETDB1 Worsens the Prognosis of Patients with Hepatocellular Carcinoma. Cells 2024; 13:2102. [PMID: 39768193 PMCID: PMC11675013 DOI: 10.3390/cells13242102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/24/2024] [Accepted: 12/02/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is clinically distinguished by its covert onset, rapid progression, high recurrence rate, and poor prognosis. Studies have revealed that SETDB1 (SET Domain Bifurcated 1) is a histone H3 methyltransferase located on chromosome 1 and plays a crucial role in carcinogenesis. Therefore, we aimed to evaluate the clinical significance of SETDB1 expression in HCC. In patients with HCC, elevated levels of SETDB1 correlated with a poorer overall survival (OS) rate, marking it as an independent prognostic factor for HCC, as revealed by both univariate and multivariate Cox analyses. Furthermore, we utilized the SangerBox and TISIDB databases to profile the tumor immune microenvironment in HCC, including scoring the tumor microenvironment and assessing immune cell infiltration. The TIDE algorithm was employed to examine the association between SETDB1 expression and immune responses. Our findings indicated that SETDB1 expression negatively correlated with the majority of immune cells, a wide range of immune cell marker genes, and numerous immune pathways, thereby leading to the reduced effectiveness of immune checkpoint inhibitors. Lastly, both in vivo and ex vivo experiments were conducted to substantiate the role of SETDB1 in HCC tumorigenesis. In conclusion, the upregulation of SETDB1 is associated with a poorer prognosis in HCC patients and inversely correlates with immune cell infiltration, potentially serving as a predictive marker for immunotherapy response.
Collapse
Affiliation(s)
- Chang-Qing Yin
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Laboratory of Gene Therapeutic Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| | - Chun-Qing Song
- College of Life Sciences, Zhejiang University, Hangzhou 310058, China;
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou 310024, China
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou 310024, China
- Laboratory of Gene Therapeutic Biology, Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou 310024, China
| |
Collapse
|
39
|
Oura K, Morishita A, Tadokoro T, Fujita K, Tani J, Kobara H. Immune Microenvironment and the Effect of Vascular Endothelial Growth Factor Inhibition in Hepatocellular Carcinoma. Int J Mol Sci 2024; 25:13590. [PMID: 39769351 PMCID: PMC11679663 DOI: 10.3390/ijms252413590] [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: 11/04/2024] [Revised: 12/12/2024] [Accepted: 12/17/2024] [Indexed: 01/03/2025] Open
Abstract
Systemic therapy for unresectable hepatocellular carcinoma (HCC) has progressed with the development of multiple kinases, such as vascular endothelial growth factor (VEGF) signaling, targeting cancer growth and angiogenesis. Additionally, the efficacy of sorafenib, regorafenib, lenvatinib, ramucirumab, and cabozantinib has been demonstrated in various clinical trials, and they are now widely used in clinical practice. Furthermore, the development of effective immune checkpoint inhibitors has progressed in systemic therapy for unresectable HCC, and atezolizumab + bevacizumab (atezo/bev) therapy and durvalumab + tremelimumab therapy are now recommended as first-line treatment. Atezo/bev therapy, which combines an anti-programmed cell death 1 ligand 1 antibody with an anti-VEGF antibody, is the first cancer immunotherapy to demonstrate efficacy against unresectable HCC. With the increasing popularity of these treatments, VEGF inhibition is attracting attention from the perspective of its anti-angiogenic effects and impact on the cancer-immune cycle. In this review, we outline the role of VEGF in the tumor immune microenvironment and cancer immune cycle in HCC and outline the potential immune regulatory mechanisms of VEGF. Furthermore, we consider the potential significance of the dual inhibition of angiogenesis and immune-related molecules by VEGF, and ultimately aim to clarify the latest treatment strategies that maximizes efficacy.
Collapse
Affiliation(s)
| | - Asahiro Morishita
- Department of Gastroenterology and Neurology, Faculty of Medicine, Kagawa University, 1750-1 Ikenobe, Miki, Kita 761-0793, Kagawa, Japan; (K.O.)
| | | | | | | | | |
Collapse
|
40
|
Guo K, Wang T, Yin J, Yang S, Cui H, Cao Z, Zhao Q, Xie G, Lu J, Gu G, Wu W. Identification of Cuproptosis-Related Patterns Predict Prognosis and Immunotherapy Response in Hepatocellular Carcinoma. J Cell Mol Med 2024; 28:e70224. [PMID: 39663596 PMCID: PMC11634814 DOI: 10.1111/jcmm.70224] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 12/13/2024] Open
Abstract
A novel copper-dependent mode of death, cuproptosis, has been newly identified. This study developed a cuproptosis score (CS) based on the cuproptosis model to analyse the association of CS with prognosis, immune cell infiltration, drug sensitivity and immunotherapy response in hepatocellular carcinoma (HCC) patients. A typing model of cuproptosis was constructed based on the expression of 19 cuproptosis-related genes (CRGs). A total of 485 samples were divided into high scoring group (HSG) and low scoring group (LSG) according to CS, and the drug sensitivity and responsiveness to immunotherapy were evaluated by combining the immunophenotype score (IPS), oncoPredict, the tumour immune dysfunction and rejection (TIDE). The use of weighted gene coexpression network analysis (WGCNA) identified key prognostic genes for cuproptosis. Western blotting was used to detect the expression level of the key gene. The CRG key gene glutaminase (GLS) is highly expressed in HCC, and patients with high expression of GLS have a poorer prognosis. Furthermore, cell function experiments, such as proliferation, migration and invasion assays, confirmed that GLS knockdown significantly changed the incidence and progression of HCC. This study suggests that new biological markers associated with cuproptosis can be used in the clinical diagnosis of HCC patients to predict prognosis and therapeutic targets.
Collapse
Affiliation(s)
- Kai Guo
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
| | - Tianbing Wang
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
| | - Jimin Yin
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Shoushan Yang
- Department of General SurgeryThe Fourth People's Hospital of Lu'anChina
| | - Haodong Cui
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| | - Zichuan Cao
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Qiang Zhao
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Gongbo Xie
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Jian Lu
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
| | - Guosheng Gu
- Department of General SurgeryAnhui No.2 Provincial People's HospitalHefeiChina
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
| | - Wenyong Wu
- Anhui No.2 Provincial People's Hospital Clinical College of Anhui Medical UniversityHefeiChina
- Anhui No.2 Provincial People's HospitalHefeiChina
- The Fifth Clinical Medical College of Anhui Medical UniversityHefeiChina
| |
Collapse
|
41
|
Wang Q, Yu J, Sun X, Li J, Cao S, Han Y, Wang H, Yang Z, Li J, Hu C, Zhang Y, Jin L. Sequencing of systemic therapy in unresectable hepatocellular carcinoma: A systematic review and Bayesian network meta-analysis of randomized clinical trials. Crit Rev Oncol Hematol 2024; 204:104522. [PMID: 39332750 DOI: 10.1016/j.critrevonc.2024.104522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 09/18/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024] Open
Abstract
PURPOSE For patients with advanced or unresectable hepatocellular carcinoma (HCC), safe and effective therapies are urgently needed to improve their long-term prognosis. Although the guidelines recommend first-line treatments such as sorafenib, lenvatinib, and atezolizumab in combination with bevacizumab (T+A) and second-line treatments such as regorafenib, the efficacy comparison between drugs is lacking, that is, a treatment is not recommended as the optimal or alternative choice for a specific patient population. Therefore, we will conduct a high-quality network meta-analysis based on Phase III randomized controlled trials (RCTs) to systematically evaluate and compare overall survival (OS), progression-free survival (PFS), objective response rate (ORR), and serious adverse events (SAE) of different treatment protocols in the context of first-line and second-line therapies, which are critical for clinical decision making and prognostic improvement in advanced HCC patients. METHODS The studies of interest were Phase III RCTs evaluating the efficacy or safety of first- or second-line therapies in patients with unresectable or advanced HCC. Literature published in English from the four databases of PubMed, Embase, Cochrane Library, and Web of Science was comprehensively searched from the inception to May 23, 2022. Outcomes of interest included OS, PFS, ORR, and SAE. A league table was developed to show the results of the comparison between different treatments. A histogram of cumulative probability was drawn to discuss the ranking probability of treatments based on different outcomes. The effectiveness and safety of various treatments were comprehensively considered and the two-dimensional diagram was plotted to guide clinical practice. The Gemtc package in R Studio was used for network meta-analysis in a Bayesian framework. RESULTS The results showed that HAIC-FO was superior to T+A regimen, regardless of OS, PFS or ORR. TACE combined with lenvatinib performed better than T+A in PFS, and ORR. In addition to the T+A regimen, Sintilimab combined with IBI305 and camrelizumab combined with apatinib were also associated with longer OS, PFS, and ORR, and their SAE incidence was not higher than that of T+A, especially for camrelizumab combined with apatinib, its safety was better than that of T+A regimen. There were no new treatments or combinations that were more effective than regorafenib. It was important to note that for PFS, the efficacy of apatinib and cabozantinib was not statistically different from that of regorafenib, so these two treatments could be used as alternative treatment options in cases where regorafenib was not tolerated or treatment failed. CONCLUSIONS We conducted a network meta-analysis to evaluate the efficacy and safety of multiple treatment modalities by integrating the results of direct and indirect comparisons. This study included high-quality multicenter Phase III RCTs, collated and summarized all treatments involved in advanced or unresectable HCC in first-line and second-line settings, and compared with T+A and regorafenib, respectively, and ranked based on efficacy and safety to support clinical decision making.
Collapse
Affiliation(s)
- Qi Wang
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianan Yu
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Xuedong Sun
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jian Li
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Shasha Cao
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yanjing Han
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Haochen Wang
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Zeran Yang
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Jianjun Li
- Interventional therapy center for oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, China
| | - Caixia Hu
- Interventional therapy center for oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, China
| | - Yonghong Zhang
- Interventional therapy center for oncology, Beijing You'an Hospital, Capital Medical University, Beijing 100069, China.
| | - Long Jin
- Department of interventional radiology, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China.
| |
Collapse
|
42
|
Song G, Yu X, Shi H, Sun B, Amateau S. miRNAs in HCC, pathogenesis, and targets. Hepatology 2024:01515467-990000000-01097. [PMID: 39626210 PMCID: PMC12119976 DOI: 10.1097/hep.0000000000001177] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/22/2024] [Indexed: 12/21/2024]
Abstract
Liver cancer is the third leading cause of cancer-related mortality worldwide. HCC, the most common type of primary liver cancer, is driven by complex genetic, epigenetic, and environmental factors. MicroRNAs, a class of naturally occurring small noncoding RNAs, play crucial roles in HCC by simultaneously modulating the expression of multiple genes in a fine-tuning manner. Significant progress has been made in understanding how miRNAs influence key oncogenic pathways, including cell proliferation, apoptosis, angiogenesis, and epithelial-mesenchymal transition (EMT), as well as their role in modulating the immune microenvironment in HCC. Due to the unexpected stability of miRNAs in the blood and fixed HCC tumors, recent advancements also highlight their potential as noninvasive diagnostic tools. Restoring or inhibiting specific miRNAs has offered promising strategies for targeted HCC treatment by suppressing malignant hepatocyte growth and enhancing antitumor immunity. In this comprehensive review, we consolidate previous research and provide the latest insights into how miRNAs regulate HCC and their therapeutic and diagnostic potential. We delve into the dysregulation of miRNA biogenesis in HCC, the roles of miRNAs in the proliferation and apoptosis of malignant hepatocytes, angiogenesis and metastasis of HCC, the immune microenvironment in HCC, and drug resistance. We also discuss the therapeutic and diagnostic potential of miRNAs and delivery approaches of miRNA drugs to overcome the limitations of current HCC treatment options. By thoroughly summarizing the roles of miRNAs in HCC, our goal is to advance the development of effective therapeutic drugs with minimal adverse effects and to establish precise tools for early diagnosis of HCC.
Collapse
Affiliation(s)
- Guisheng Song
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Genetics, Cell Biology and Development, University of Minnesota, Minneapolis, Minnesota, USA
| | - Xiaofan Yu
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Hongtao Shi
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
- Department of Cardiology, the First Hospital of Shanxi Medical University, Taiyuan City, China
| | - Bo Sun
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| | - Stuart Amateau
- Department of Medicine, University of Minnesota Medical School, Minneapolis, Minnesota, USA
| |
Collapse
|
43
|
Zhuo W, Xia H, Lan B, Chen Y, Wang X, Liu J. Signature of immune-related metabolic genes predicts the prognosis of hepatocellular carcinoma. Front Immunol 2024; 15:1481331. [PMID: 39654885 PMCID: PMC11625796 DOI: 10.3389/fimmu.2024.1481331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 11/08/2024] [Indexed: 12/12/2024] Open
Abstract
Introduction The majority of liver cancer cases (90%) are attributed to hepatocellular carcinoma (HCC), which exhibits significant heterogeneity and an unfavorable prognosis. Modulating the immune response and metabolic processes play a crucial role in both the prevention and treatment of HCC. However, there is still a lack of comprehensive understanding regarding the immune-related metabolic genes that can accurately reflect the prognosis of HCC. Methods In order to address this issue, we developed a prognostic prediction model based on immune and metabolic genes. To evaluate the accuracy of our model, we performed survival analyses including Kaplan-Meier (K-M) curve and time-dependent receiver operating characteristic (ROC) curve. Furthermore, we compared the predictive performance of our risk model with existing models. Finally, we validated the accuracy of our risk model using mouse models with in situ transplanted liver cancer. Results By conducting lasso regression analysis, we identified four independent prognostic genes: fatty acid binding protein 6 (FABP6), phosphoribosyl pyrophosphate amidotransferase (PPAT), spermine synthase (SMS), and dihydrodiol dehydrogenase (DHDH). Based on these findings, we constructed a prognostic model. Survival analysis revealed that the high-risk group had significantly lower overall survival (OS) rates. Besides that, the ROC curve demonstrated the effective prognostic capability of our risk model for hepatocellular carcinoma (HCC) patients. Furthermore, through animal experiments, we validated the accuracy of our model by showing a correlation between high-risk scores and poor prognosis in tumor development. Discussion In conclusion, our prognostic model surpasses those solely based on immune genes or metabolic genes in terms of accuracy. We observed variations in prognosis among different risk groups, accompanied by distinct immune and metabolic characteristics. Therefore, our model provides an original evaluation index for personalized clinical treatment strategies targeting HCC patients.
Collapse
Affiliation(s)
- Weibin Zhuo
- Innovation Center for Cancer Research, Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
| | - Hongmei Xia
- Huzhou Central Hospital, Fifth School of Clinical Medicine, Zhejiang Chinese Medical University, Huzhou, Zhejiang, China
| | - Bin Lan
- Innovation Center for Cancer Research, Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, Fujian, China
- XMU-Fujian Cancer Hospital Research Center of Metabolism and Cancer, Xiamen University, Xiamen, Fujian, China
| | - Yu Chen
- Innovation Center for Cancer Research, Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, Fujian, China
- XMU-Fujian Cancer Hospital Research Center of Metabolism and Cancer, Xiamen University, Xiamen, Fujian, China
| | - Xuefeng Wang
- Innovation Center for Cancer Research, Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, Fujian, China
- XMU-Fujian Cancer Hospital Research Center of Metabolism and Cancer, Xiamen University, Xiamen, Fujian, China
| | - Jingfeng Liu
- Innovation Center for Cancer Research, Laboratory of Radiation Oncology and Radiobiology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, Fujian, China
- Interdisciplinary Institute for Medical Engineering, Fuzhou University, Fuzhou, Fujian, China
- XMU-Fujian Cancer Hospital Research Center of Metabolism and Cancer, Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
44
|
Hu Y, Zhang L, Qi Q, Ren S, Wang S, Yang L, Zhang J, Liu Y, Li X, Cai X, Duan S, Zhang L. Machine learning-based ultrasomics for predicting response to tyrosine kinase inhibitor in combination with anti-PD-1 antibody immunotherapy in hepatocellular carcinoma: a two-center study. Front Oncol 2024; 14:1464735. [PMID: 39610931 PMCID: PMC11602396 DOI: 10.3389/fonc.2024.1464735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Accepted: 10/28/2024] [Indexed: 11/30/2024] Open
Abstract
Objective The objective of this study is to build and verify the performance of machine learning-based ultrasomics in predicting the objective response to combination therapy involving a tyrosine kinase inhibitor (TKI) and anti-PD-1 antibody for individuals with unresectable hepatocellular carcinoma (HCC). Radiomic features can reflect the internal heterogeneity of the tumor and changes in its microenvironment. These features are closely related to pathological changes observed in histology, such as cellular necrosis and fibrosis, providing crucial non-invasive biomarkers to predict patient treatment response and prognosis. Methods Clinical, pathological, and pre-treatment ultrasound image data of 134 patients with recurrent unresectable or advanced HCC who treated with a combination of TKI and anti-PD-1 antibody therapy at Henan Provincial People's Hospital and the First Affiliated Hospital of Zhengzhou University between December 2019 and November 2023 were collected and retrospectively analyzed. Using stratified random sampling, patients from the two hospitals were assigned to training cohort (n = 93) and validation cohort (n = 41) at a 7:3 ratio. After preprocessing the ultrasound images, regions of interest (ROIs) were delineated. Ultrasomic features were extracted from the images for dimensionality reduction and feature selection. By utilizing the extreme gradient boosting (XGBoost) algorithm, three models were developed: a clinical model, an ultrasomic model, and a combined model. By analyzing the area under the receiver operating characteristic (ROC) curve (AUC), specificity, sensitivity, and accuracy, the predicted performance of the models was evaluated. In addition, we identified the optimal cutoff for the radiomic score using the Youden index and applied it to stratify patients. The Kaplan-Meier (KM) survival curves were used to examine differences in progression-free survival (PFS) between the two groups. Results Twenty ultrasomic features were selected for the construction of the ultrasomic model. The AUC of the ultrasomic model for the training cohort and validation cohort were 0.999 (95%CI: 0.997-1.000) and 0.828 (95%CI: 0.690-0.966), which compared significant favorably to those of the clinical model [AUC = 0.876 (95%CI: 0.815-0.936) for the training cohort, 0.766 (95%CI: 0.597-0.935) for the validation cohort]. Compared to the ultrasomic model, the combined model demonstrated comparable performance within the training cohort (AUC = 0.977, 95%CI: 0.957-0.998) but higher performance in the validation cohort (AUC = 0.881, 95%CI: 0.758-1.000). However, there was no statistically significant difference (p > 0.05). Furthermore, ultrasomic features were associated with PFS, which was significantly different between patients with radiomic scores (Rad-score) greater than 0.057 and those with Rad-score less than 0.057 in both the training (HR = 0.488, 95% CI: 0.299-0.796, p = 0.003) and validation cohorts (HR = 0.451, 95% CI: 0.229-0.887, p = 0.02). Conclusion The ultrasomic features demonstrates excellent performance in accurately predicting the objective response to TKI in combination with anti-PD-1 antibody immunotherapy among patients with unresectable or advanced HCC.
Collapse
Affiliation(s)
- Yiwen Hu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Linlin Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Qinghua Qi
- Department of Ultrasound, First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Shanshan Ren
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Simeng Wang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Lanling Yang
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Juan Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Yuanyuan Liu
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiaoxiao Li
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Xiguo Cai
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
| | - Shaobo Duan
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
- Department of Health Management, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
| | - Lianzhong Zhang
- Department of Ultrasound, Henan University People’s Hospital, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, China
- Henan Engineering Technology Research Center of Ultrasonic Molecular Imaging and Nanotechnology, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Rehabilitation Clinical Medical Research Center, Henan Provincial People’s Hospital, Zhengzhou, China
- Henan Key Laboratory of Ultrasound Imaging and Artificial Intelligence in Medicine, Henan Provincial People’s Hospital, Zhengzhou, China
| |
Collapse
|
45
|
Lin P, Xie W, Li Y, Zhang C, Wu H, Wan H, Gao M, Liang F, Han P, Chen R, Cheng G, Liu X, Fan S, Huang X. Intratumoral and peritumoral radiomics of MRIs predicts pathologic complete response to neoadjuvant chemoimmunotherapy in patients with head and neck squamous cell carcinoma. J Immunother Cancer 2024; 12:e009616. [PMID: 39500529 PMCID: PMC11552555 DOI: 10.1136/jitc-2024-009616] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/01/2024] [Indexed: 11/13/2024] Open
Abstract
BACKGROUND For patients with locally advanced head and neck squamous cell carcinoma (HNSCC), combined programmed death receptor-1 inhibitor and chemotherapy improved response rate to neoadjuvant therapy. However, treatment response varies among patients. There is no tool to predict pathologic complete response (pCR) with high accuracy for now. To develop a tool based on radiomics features of MRI to predict pCR to neoadjuvant chemoimmunotherapy (NACI) may provide valuable assistance in treatment regimen determination for HNSCC. METHODS From January 2021 to April 2024, a total of 172 patients with HNSCC from three medical center, who received NACI followed by surgery, were included and allocated into a training set (n=84), an internal validation set (n=37) and an external validation set (n=51). Radiomics features were extracted from intratumoral and different peritumoral areas, and radiomics signature (Rad-score) for each area was constructed. A radiomics-clinical nomogram was developed based on Rad-scores and clinicopathological characteristics, tested in the validation sets, and compared with clinical nomogram and combined positive score (CPS) in predicting pCR. RESULTS The radiomics-clinical nomogram, incorporating peritumoral Rad-score, intratumoral Rad-score and CPS, achieved the highest accuracy with areas under the receiver operating characteristic curve of 0.904 (95% CI, 0.835 to 0.972) in the training cohort, 0.860 (95% CI, 0.722 to 0.998) in the internal validation cohort, and 0.849 (95% CI, 0.739 to 0.959) in the external validation cohort, respectively, which outperformed the clinical nomogram and CPS in predict pCR to NACI for HNSCC. CONCLUSION A nomogram developed based on intratumoral and peritumoral MRI radiomics features outperformed CPS, a widely employed biomarker, in predict pCR to NACI for HNSCC, which would provide incremental value in treatment regimen determination.
Collapse
Affiliation(s)
- Peiliang Lin
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Wenqian Xie
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Yong Li
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Chenjia Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Huiqian Wu
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Pathology Department, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Huan Wan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Cellular & Molecular Diagnostics Center, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ming Gao
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Radiology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Faya Liang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Ping Han
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Renhui Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Gui Cheng
- Department of Otolaryngology, Shenshan Medical Centre, Memorial Hospital of Sun Yat-sen University, Shanwei, China
| | - Xuekui Liu
- Department of Head and Neck Surgery, Sun Yat-sen University Cancer Center, Guangzhou, Guangdong, China
| | - Song Fan
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Oral and Maxillofacial Surgery, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| | - Xiaoming Huang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
- Department of Otolaryngology, Sun Yat-Sen Memorial Hospital, Guangzhou, Guangdong, China
| |
Collapse
|
46
|
Xiang X, Wang K, Zhang H, Mou H, Shi Z, Tao Y, Song H, Lian Z, Wang S, Lu D, Wei X, Xie H, Zheng S, Wang J, Xu X. Blocking CX3CR1+ Tumor-Associated Macrophages Enhances the Efficacy of Anti-PD1 Therapy in Hepatocellular Carcinoma. Cancer Immunol Res 2024; 12:1603-1620. [PMID: 39115356 DOI: 10.1158/2326-6066.cir-23-0627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 04/08/2024] [Accepted: 08/07/2024] [Indexed: 11/05/2024]
Abstract
The efficacy of immune checkpoint inhibitors in the treatment of hepatocellular carcinoma (HCC) remains limited, highlighting the need for further investigation into the mechanisms underlying treatment resistance. Accumulating evidence indicates that tumor-associated macrophages (TAM) within the tumor microenvironment demonstrate a key role in immune evasion and treatment resistance. This study explored the role of TAMs in the HCC tumor microenvironment. Our findings reveal that TAMs expressing CX3C motif chemokine receptor 1 (CX3CR1) induced T-cell exhaustion through IL27 secretion in orthotopic models of HCC following treatment with anti-PD1. Moreover, we identified prostaglandin E2 (PGE2), released by immune-attacked tumor cells, as a key regulator of TAM transition to a CX3CR1+ phenotype. To augment the therapeutic response to anti-PD1 therapy, we propose targeting CX3CR1+ TAMs in addition to anti-PD1 therapy. Our study contributes to the understanding of the role of TAMs in cancer immunotherapy and highlights potential clinical implications for HCC treatment. The combination of targeting CX3CR1+ TAMs with anti-PD1 therapy holds promise for enhancing the efficacy of immunotherapeutic interventions in patients with HCC.
Collapse
Affiliation(s)
- Xiaonan Xiang
- Zhejiang University School of Medicine, Hangzhou, China
| | - Kai Wang
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Hui Zhang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haibo Mou
- Department of Medical Oncology, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Zhixiong Shi
- Zhejiang University School of Medicine, Hangzhou, China
| | - Yaoye Tao
- Zhejiang University School of Medicine, Hangzhou, China
| | - Hongliang Song
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Zhengxing Lian
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Shuai Wang
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Di Lu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
| | - Xuyong Wei
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Hangzhou, China
| | - Haiyang Xie
- Department of Hepatobiliary & Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
| | - Shusen Zheng
- NHC Key Laboratory of Combined Multi-Organ Transplantation, Hangzhou, China
- Department of Hepatobiliary & Pancreatic Surgery, Shulan (Hangzhou) Hospital, Hangzhou, China
| | - Jianguo Wang
- Department of Hepatobiliary and Pancreatic Surgery, Minimally Invasive Surgery, Zhejiang Provincial People's Hospital (Affiliated People's Hospital), Hangzhou Medical College, Hangzhou, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, China
| |
Collapse
|
47
|
Chen Y, Jia L, Li Y, Cui W, Wang J, Zhang C, Bian C, Wang Z, Lin D, Luo T. Clinical Effectiveness and Safety of Transarterial Chemoembolization: Hepatic Artery Infusion Chemotherapy Plus Tyrosine Kinase Inhibitors With or Without Programmed Cell Death Protein-1 Inhibitors for Unresectable Hepatocellular Carcinoma-A Retrospective Study. Ann Surg Oncol 2024; 31:7860-7869. [PMID: 39090499 DOI: 10.1245/s10434-024-15933-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Accepted: 07/16/2024] [Indexed: 08/04/2024]
Abstract
BACKGROUND Locoregional treatment with transarterial chemoembolization (TACE) or hepatic artery infusion chemotherapy (HAIC) and systemic targeted immunotherapy with tyrosine kinase inhibitors (TKI) and programmed cell death protein-1 (PD-1) inhibitors in the treatment of unresectable hepatocellular carcinoma (uHCC) have achieved promising efficacy. The retrospective study aimed to evaluate the efficacy and safety of TACE and HAIC plus TKI with or without PD-1 for uHCC. PATIENTS AND METHODS From November 2020 to February 2024, the data of 44 patients who received TACE-HAIC + TKI + PD-1 (THKP group) and 34 patients who received TACE-HAIC + TKI (THK group) were retrospectively analyzed. Primary outcomes were overall survival (OS) and progress-free survival (PFS), and secondary outcomes were objective response rate (ORR), disease control rate (DCR), conversion rates, and adverse events (AEs). RESULTS A total of 78 patients were recruited in our single-center study. The patients in THKP group had prolonged median OS [25 months, 95% confidence interval (CI) 24.0-26.0 vs 18 months, 95% CI 16.1-19.9; p = 0.000278], median PFS [16 months, 95% CI 14.1-17.9 vs 12 months 95% CI 9.6-14.4; p = 0.004] and higher ORR (38.6% vs 23.5%, p = 0. 156) and DCR (88.6% vs 64.7%, p = 0.011) compared with those in THK group. Multivariate analysis showed that treatment option and alpha-fetoprotein (AFP) level were independent prognostic factors of OS and PFS. The frequency of AEs were similar between the two groups. CONCLUSIONS The THKP group had better efficacy for uHCC than the THK group, with acceptable safety.
Collapse
Affiliation(s)
- Yue Chen
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Luyao Jia
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Yu Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Wenhao Cui
- Emergency Medicine Department, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Jukun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chao Zhang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Chunjing Bian
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Zhenshun Wang
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Dongdong Lin
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Tao Luo
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
48
|
Yin L, Jiang W, Liu S, Fu Y, Zhou L, Pei X, Ye S, Shen W, Yang H, Shan B. Efficacy and safety of nanoparticle albumin-bound paclitaxel plus carboplatin as neoadjuvant chemotherapy for stages III-IV, unresectable ovarian cancer: a single-arm, open-label, phase Ib/II study. BMC Med 2024; 22:496. [PMID: 39468597 PMCID: PMC11520812 DOI: 10.1186/s12916-024-03697-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 10/09/2024] [Indexed: 10/30/2024] Open
Abstract
BACKGROUND Neoadjuvant chemotherapy may be considered for patients with ovarian cancer (OC) whose tumors are deemed unlikely to be completely cytoreduced to no gross residual disease (R0) or who are poor surgical candidates. This Ib/II study was designed to assess the efficacy and safety of nanoparticle albumin-bound paclitaxel (nab-paclitaxel) plus carboplatin as neoadjuvant chemotherapy for stages III-IV, unresectable OC. METHODS Eligible patients with stage III-IV, unresectable OC were enrolled in this phase Ib/II study. All patients received neoadjuvant nab-paclitaxel (260 mg/m2, day 1, every 3 weeks) plus carboplatin (AUC 5, day 1, every 3 weeks) for 3 cycles before surgery, followed by 3-6 cycles of adjuvant chemotherapy. The phase Ib primary endpoint was safety; the phase II primary endpoint was the R0 resection rate. Secondary endpoints were progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and safety (for all populations). RESULTS Sixty-two patients were enrolled and were given neoadjuvant therapy treated between October 2019 and December 2020, of whom 9 were in the phase Ib portion and 53 in the phase II portion. A total of 53 patients underwent surgery with an R0 resection rate of 73.6% (95% CI, 59.7-84.7%). With a median follow-up of 17.5 (range 0.7-36.7) months, for all patients, the best ORR was 83.9% (95% CI, 71.7-92.4%) with 47 partial responses, the median PFS was 18.6 (95% CI, 13.8-23.3%) months, and median OS was not reached. During the neoadjuvant chemotherapy, treatment-related adverse events (TRAEs) of any grade occurred in 91.9% (57/62) of all patients. The most common hematologic TRAEs were neutropenia (55/62, 88.7%), and non-hematologic toxicity was alopecia (36/62, 58.1%). Forty-nine patients (79.0%) experienced at least one grade 3-4 TRAEs, with the most common was neutropenia (44/62, 71.0%). Besides, delays in neoadjuvant chemotherapy and surgery due to AEs were observed in 9 (1 in phase Ib; 8 in phase II) and 7 (phase II) patients, respectively. CONCLUSIONS The study demonstrated an encouraging efficacy and manageable safety profile of neoadjuvant chemotherapy nab-paclitaxel plus carboplatin in stage III-IV, unresectable OC. In addition, AEs resulting in chemotherapy and surgery delays should be cautiously considered in this clinical setting. TRIAL REGISTRATION ClinicalTrials.gov, ChiCTR1900026893. Registered at 25 October 2019.
Collapse
Affiliation(s)
- Lina Yin
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Wei Jiang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Shuai Liu
- Department of Nuclear Medicine, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Yi Fu
- Department of Radiology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Lin Zhou
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Xuan Pei
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Shuang Ye
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Wenbin Shen
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Huijuan Yang
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| | - Boer Shan
- Department of Gynecological Oncology, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China.
| |
Collapse
|
49
|
Ning J, Wang Y, Tao Z. The complex role of immune cells in antigen presentation and regulation of T-cell responses in hepatocellular carcinoma: progress, challenges, and future directions. Front Immunol 2024; 15:1483834. [PMID: 39502703 PMCID: PMC11534672 DOI: 10.3389/fimmu.2024.1483834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 09/30/2024] [Indexed: 11/08/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a prevalent form of liver cancer that poses significant challenges regarding morbidity and mortality rates. In the context of HCC, immune cells play a vital role, especially concerning the presentation of antigens. This review explores the intricate interactions among immune cells within HCC, focusing on their functions in antigen presentation and the modulation of T-cell responses. We begin by summarizing the strategies that HCC uses to escape immune recognition, emphasizing the delicate equilibrium between immune surveillance and evasion. Next, we investigate the specific functions of various types of immune cells, including dendritic cells, natural killer (NK) cells, and CD8+ T cells, in the process of antigen presentation. We also examine the impact of immune checkpoints, such as cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) and the pathways involving programmed cell death protein 1 (PD-1) and programmed death ligand 1 (PD-L1), on antigen presentation, while taking into account the clinical significance of checkpoint inhibitors. The review further emphasizes the importance of immune-based therapies, including cancer vaccines and CAR-T cell therapy, in improving antigen presentation. In conclusion, we encapsulate the latest advancements in research, propose future avenues for exploration, and stress the importance of innovative technologies and customized treatment strategies. By thoroughly analyzing the interactions of immune cells throughout the antigen presentation process in HCC, this review provides an up-to-date perspective on the field, setting the stage for new therapeutic approaches.
Collapse
Affiliation(s)
- Jianbo Ning
- The Fourth Clinical College, China Medical University, Shenyang, China
| | - Yutao Wang
- Department of Urology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zijia Tao
- Department of Interventional Radiology, the First Hospital of China Medical University, Shenyang, China
| |
Collapse
|
50
|
Hai L, Bai XY, Luo X, Liu SW, Ma ZM, Ma LN, Ding XC. Prognostic modeling of hepatocellular carcinoma based on T-cell proliferation regulators: a bioinformatics approach. Front Immunol 2024; 15:1444091. [PMID: 39445019 PMCID: PMC11496079 DOI: 10.3389/fimmu.2024.1444091] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 09/23/2024] [Indexed: 10/25/2024] Open
Abstract
Background The prognostic value and immune significance of T-cell proliferation regulators (TCRs) in hepatocellular carcinoma (HCC) have not been previously reported. This study aimed to develop a new prognostic model based on TCRs in patients with HCC. Method This study used The Cancer Genome Atlas-Liver Hepatocellular Carcinoma (TCGA-LIHC) and International Cancer Genome Consortium-Liver Cancer-Riken, Japan (ICGC-LIRI-JP) datasets along with TCRs. Differentially expressed TCRs (DE-TCRs) were identified by intersecting TCRs and differentially expressed genes between HCC and non-cancerous samples. Prognostic genes were determined using Cox regression analysis and were used to construct a risk model for HCC. Kaplan-Meier survival analysis was performed to assess the difference in survival between high-risk and low-risk groups. Receiver operating characteristic curve was used to assess the validity of risk model, as well as for testing in the ICGC-LIRI-JP dataset. Additionally, independent prognostic factors were identified using multivariate Cox regression analysis and proportional hazards assumption, and they were used to construct a nomogram model. TCGA-LIHC dataset was subjected to tumor microenvironment analysis, drug sensitivity analysis, gene set variation analysis, and immune correlation analysis. The prognostic genes were analyzed using consensus clustering analysis, mutation analysis, copy number variation analysis, gene set enrichment analysis, and molecular prediction analysis. Results Among the 18 DE-TCRs, six genes (DCLRE1B, RAN, HOMER1, ADA, CDK1, and IL1RN) could predict the prognosis of HCC. A risk model that can accurately predict HCC prognosis was established based on these genes. An efficient nomogram model was also developed using clinical traits and risk scores. Immune-related analyses revealed that 39 immune checkpoints exhibited differential expression between the high-risk and low-risk groups. The rate of immunotherapy response was low in patients belonging to the high-risk group. Patients with HCC were further divided into cluster 1 and cluster 2 based on prognostic genes. Mutation analysis revealed that HOMER1 and CDK1 harbored missense mutations. DCLRE1B exhibited an increased copy number, whereas RAN exhibited a decreased copy number. The prognostic genes were significantly enriched in tryptophan metabolism pathways. Conclusions This bioinformatics analysis identified six TCR genes associated with HCC prognosis that can serve as diagnostic markers and therapeutic targets for HCC.
Collapse
Affiliation(s)
- Long Hai
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiao-Yang Bai
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xia Luo
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Shuai-Wei Liu
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Zi-Min Ma
- Weiluo Microbial Pathogens Monitoring Technology Co., Ltd. of Beijing, Beijing, China
| | - Li-Na Ma
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
| | - Xiang-Chun Ding
- Department of Infectious Disease, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Infectious Disease Clinical Research Center of Ningxia, General Hospital of Ningxia Medical University, Yinchuan, Ningxia, China
- Department of Tropical Disease & Infectious Disease, The Second Affiliated Hospital of Hainan Medical University, Haikou, Hainan, China
| |
Collapse
|