1
|
Akabane M, Imaoka Y, Kawashima J, Pawlik TM. Advancing precision medicine in hepatocellular carcinoma: current challenges and future directions in liquid biopsy, immune microenvironment, single nucleotide polymorphisms, and conversion therapy. Hepat Oncol 2025; 12:2493457. [PMID: 40260687 PMCID: PMC12026093 DOI: 10.1080/20450923.2025.2493457] [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: 01/31/2025] [Accepted: 04/11/2025] [Indexed: 04/24/2025] Open
Abstract
Hepatocellular carcinoma (HCC) remains a health concern characterized by heterogeneity and high mortality. Surgical resection, radiofrequency ablation, trans-arterial chemoembolization, and liver transplantation offer potentially curative treatments for early-stage disease, but recurrence remains high. Most patients present with advanced-stage HCC, where locoregional therapies are less effective, and systemic treatments-primarily multi-kinase inhibitors and immune checkpoint inhibitors-often yield limited responses. Precision medicine aims to tailor therapy to molecular and genetic profiles, yet its adoption in HCC is hindered by inter-/intra-tumoral heterogeneity and limited biopsy availability. Advances in molecular diagnostics support reintroducing tissue sampling to better characterize genetic, epigenetic, and immunological features. Liquid biopsy offers a minimally invasive method for capturing real-time tumor evolution, overcoming spatial and temporal heterogeneity. Artificial intelligence and machine learning are revolutionizing biomarker discovery, risk stratification, and treatment planning by integrating multi-omics data. Immunological factors such as tumor-infiltrating lymphocytes, natural killer cells, macrophages, and fibroblasts have emerged as determinants of HCC progression and treatment response. Conversion therapy-combining systemic agents with locoregional treatments-has showndemonstrated promise in downstaging unresectable HCC. Ongoing efforts to refine biomarker-driven approaches and optimize multi-modality regimens underscore precision medicine's potential to improve outcomes. PubMed (January 2002-February 2025) was searched for relevant studies.
Collapse
Affiliation(s)
- Miho Akabane
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Yuki Imaoka
- Division of Abdominal Transplant, Department of Surgery, Stanford University, CA, USA
| | - Jun Kawashima
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| | - Timothy M. Pawlik
- Department of Surgery, The Ohio State University Wexner Medical Center and James Comprehensive Cancer Center, Columbus, OH, USA
| |
Collapse
|
2
|
Malayaperumal S, Sriramulu S, Jothimani G, Banerjee A, Zhang H, Mohammed Rafi ST, Ramachandran I, NR RK, Sun XF, Pathak S. MicroRNA-122 overexpression suppresses the colon cancer cell proliferation by downregulating the astrocyte elevated gene-1/metadherin oncoprotein. Ann Med 2025; 57:2478311. [PMID: 40208016 PMCID: PMC11986857 DOI: 10.1080/07853890.2025.2478311] [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: 10/26/2023] [Revised: 11/09/2024] [Accepted: 02/17/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND MicroRNAs (miRNAs) are small non-coding RNAs that regulate essential cellular functions, such as cell adhesion, proliferation, migration, invasion, and programmed cell death, and therefore, alterations in miRNAs can contribute to carcinogenesis. Previous studies have shown that miRNA-122 is abundant in the liver and regulates cell proliferation, migration, and apoptosis. However, the expression pattern and mechanism of actions of miR-122 remain primarily unknown in colon cancer. METHODS In this study, we analyzed The Cancer Genome Atlas Colon Adenocarcinoma (TCGA-COAD) database to assess the clinical significance of astrocyte elevated gene-1 (AEG-1)/metadherin (MTDH) and miR-122 in colon cancer. MiR-122 overexpression studies were performed in HCT116, SW480, and SW620 cell lines. Dual-luciferase assay was carried out to confirm the interaction between AEG-1 and miR-122. In vivo-JetPEI-transfection reagent was used for in-vivo transient transfection of miR-122 in the AOM/DSS-induced colon tumor mouse model. RESULTS Our results demonstrate that miR-122 was downregulated in colon cancer cells, and it influences the expressions of apoptotic factors and inflammatory cytokines. MiR-122 overexpression in HCT116, SW480, and SW620 cells showed upregulation of Caspase 3, Caspase 9, and BAX and decreased expression of BCL2, which are pro-apoptotic and anti-apoptotic members that maintain a ratio between cellular survival and cell death. In vivo transient transfection of miR-122 mimic in AOM/DSS induced colon tumor mouse model showed less inflammation and disease activity. The TCGA-COAD data indicated that AEG-1 expression was higher in patients with low expression of miR-122 and lower AEG-1 expression in patients with higher expression miR-122. CONCLUSION Our findings highlight the key role of miR-122 in the high grade of colonic inflammation, and possibly in colon cancer, and the use of miR-122 mimic might be a therapeutic option.
Collapse
Affiliation(s)
- Sarubala Malayaperumal
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Sushmitha Sriramulu
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Ganesan Jothimani
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Antara Banerjee
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Hong Zhang
- Department of Medical Sciences, School of Medicine, Orebro University, Örebro, Sweden
| | - Shabana Thabassum Mohammed Rafi
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Ilangovan Ramachandran
- Department of Endocrinology, Dr. ALM PG Institute of Basic Medical Sciences, University of Madras, Taramani Campus, Chennai, India
| | - Rajesh Kanna NR
- Department of Pathology, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| | - Xiao-Feng Sun
- Division of Oncology, Department of Biomedical and Clinical Sciences, Linköping University, Linköping, Sweden
| | - Surajit Pathak
- Faculty of Allied Health Sciences, Chettinad Academy of Research and Education, Chettinad Hospital and Research Institute, Chennai, India
| |
Collapse
|
3
|
Zhou S, Qin Z, Cai S, Ma T, Lin L, Feng L, Gao X, Ma D. Prognostic value of immune infiltration in colorectal cancer: Development of a histopathology-related immunoscore via multiplexed immunohistochemistry. Surgery 2025; 182:109350. [PMID: 40233469 DOI: 10.1016/j.surg.2025.109350] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 02/27/2025] [Accepted: 03/10/2025] [Indexed: 04/17/2025]
Abstract
BACKGROUND Our objective was to evaluate the prognostic value of immune infiltration within the intratumoral and peritumoral tissues and to establish a novel histopathology-related immunoscore associated with postoperative colorectal cancer prognosis. METHODS In the tissue microarrays, a total of 104 patients with colorectal cancer were enrolled and randomly assigned to the derivation cohort (n = 61) or the validation cohort (n = 43). Eighteen prognostic immune biomarkers in both intratumoral and peritumoral tissues were examined by the multiplexed immunohistochemistry method, with quantification performed through digital pathology. The histopathology-related immunoscore score was constructed using least absolute shrinkage and selection operator Cox analysis by selected immune features. On the basis of the Cox regression analysis, 3 predictive models were established. Harrell C-statistics were used to assess the performance of those models. RESULTS The area under the curve was 0.743 (confidence interval, 0.457-1.000) in the derivation cohort and 0.739 (confidence interval, 0.538-0.940) in the validation cohort. Subsequently, the groups were classified on the basis of the optimal cutoff value, with the high-risk group exhibiting a poorer prognosis. Furthermore, 3 predictive clinical models were constructed, incorporating the significant risk factors and histopathology-related immunoscore score. The first model incorporating both histopathology-related immunoscore score and statistically significant factors identified through univariate analysis demonstrated superior predictive capability for survival across all 3 models, with an area under the curve of 0.852 and C-index of 0.837. CONCLUSION The histopathology-related immunoscore score offers a novel means of estimating of survival in patients with colorectal cancer. These findings indicated that the immunoscore and the clinical factors might serve as complementary tools to TNM staging to improve the accuracy of patient survival prediction.
Collapse
Affiliation(s)
- Shiqi Zhou
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China; Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Zhaofu Qin
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shunv Cai
- Department of Anesthesiology, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Luyi Lin
- Department of Radiology, Shanghai Cancer Center, Fudan University, Shanghai, China
| | - Longhai Feng
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinyi Gao
- Department of Radiology, Zhejiang Cancer Hospital, Hangzhou, China.
| | - Dening Ma
- Department of Colorectal Surgery, Zhejiang Cancer Hospital, Hangzhou, China; Postgraduate Training Base Alliance of Wenzhou Medical University (Zhejiang Cancer Hospital), Hangzhou, Zhejiang, China.
| |
Collapse
|
4
|
Zhou M, Zhang Y, Song W. Single-cell transcriptome analysis identifies subclusters and signature with N-glycosylation in endometrial cancer. Clin Transl Oncol 2025; 27:2467-2483. [PMID: 39589706 DOI: 10.1007/s12094-024-03802-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 11/18/2024] [Indexed: 11/27/2024]
Abstract
INTRODUCTION Endometrial cancer (EC) is a prevalent gynecologic cancer, with worldwide increasing incidence and disease-associated mortality. N-glycosylation, a critical post-translational modification, has been implicated in cancer progression and immune response modulation. We aimed to elucidate the role of N-glycosylation-related genes on EC cell heterogeneity, prognosis, and immunotherapy response. METHODS Data from single-cell RNA sequencing (scRNA) of five patients with EC were acquired from the Gene Expression Omnibus (GEO) database. Nonnegative matrix factorization (NMF) was used to identify cell subtypes related to N-glycosylation from a scRNA matrix. Subsequently, a consensus prognostic signature by integrating 101 combinations of 10 machine learning algorithms. The response to immunotherapy in EC was further examined by multiple algorithms. RESULTS Our findings identified 11,020 differentially expressed genes (DEGs), of which 34 N-glycosylation-related DEGs were remarkably associated with overall survival (OS) in EC. Single-cell RNA sequencing analysis revealed 30,233 cells divided into eight clusters, with T cells and epithelial cells showing distinct functional characteristics. NMF clustering further classified malignant cells into four subtypes: N-glycosylation-C0, Glycosphingolipid-C1, O-GalNAc-C2, and Elongation-C3. The O-GalNAc-C2 subtype exhibited the highest metabolic pathway activity and activation of transcription factors SOX4, JUND, and FOS. Additionally, cell-cell interaction networks highlighted the MK signaling pathway as a critical mediator of intercellular communication. An integrated machine learning framework generated a prognostic model comprising eight DEGs (LAMC2, KRT7, IL32, KRT18, SERPINA1, PGR, AKAP12, EDN2), achieving an average C-index of 0.712 in training and validation cohorts. A low-risk score implies more significant immune cell infiltration and better response to immunotherapy. CONCLUSIONS Our study underscores the role of N-glycosylation-related genes in EC prognosis and immunotherapy response prediction, and may provide a basis for the development of targeted therapies and personalized treatment strategies.
Collapse
Affiliation(s)
- Min Zhou
- Department of Gynecology and Obstetrics, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Yuefeng Zhang
- Department of Hepatobiliary Surgery, Renmin Hospital of Wuhan University, Wuhan, 430060, Hubei, China
| | - Wei Song
- Department of Gastrointestinal Surgery II, Renmin Hospital of Wuhan University, No. 238, Jiefang Road, Wuhan, 430060, China.
| |
Collapse
|
5
|
Qi Y, Sun D, Zhai X, Chen F, Niu J, Zhu H. Macrophages in the premetastatic and metastatic niche: key functions and therapeutic directions. J Transl Med 2025; 23:602. [PMID: 40448239 DOI: 10.1186/s12967-025-06556-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Accepted: 04/30/2025] [Indexed: 06/02/2025] Open
Abstract
Metastasis plays a significant role in the high mortality rates associated with cancer and is usually the endpoint of a series of sequential and dynamic events. A crucial step in metastasis development and progression is the formation of a premetastatic niche (PMN), which provides a conducive microenvironment for the settlement and colonization of disseminated tumor cells at distant metastatic sites. Extensive research has demonstrated the significance of macrophage populations within primary tumors in promoting metastatic progression. Nevertheless, the contribution of macrophages at secondary sites to the regulation of PMN formation is frequently overlooked. This review systematically explores the role of macrophages in priming the PMN to facilitate cancer metastasis. Additionally, we provide a compendium of existing strategies to target macrophages in cancer therapy.
Collapse
Affiliation(s)
- Yana Qi
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China
| | - Dongmei Sun
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China
| | - Xiaoyang Zhai
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China
| | - Feihu Chen
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China
| | - Jiling Niu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China
| | - Hui Zhu
- Department of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, 440 Jiyan Road, Jinan, Shandong Province, 250117, China.
| |
Collapse
|
6
|
Ghebremedhin A, Varner JA. PI3Kγ in Tumour Inflammation: Bridging Immune Response and Cancer Progression-A Mini-Review. Immunology 2025. [PMID: 40434054 DOI: 10.1111/imm.13959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 05/04/2025] [Accepted: 05/16/2025] [Indexed: 05/29/2025] Open
Abstract
Phosphatidylinositol 3-kinase gamma (PI3Kγ), a class I PI3K family member, plays a critical role in modulating inflammation and immune responses within the tumour microenvironment. Emerging evidence suggests that PI3Kγ promotes myeloid cell trafficking and transcription, leading to tumour progression and metastasis. This review explores the multifaceted roles of PI3Kγ in tumour-associated inflammation, highlighting its involvement in immune cell polarisation, cytokine production, and the dynamic interaction between tumour cells and the surrounding stromal environment. We also discuss the potential therapeutic implications of targeting PI3Kγ to modulate inflammation and inhibit tumour growth. Given its pivotal role in immune response and tumour progression, PI3Kγ represents a promising target for future cancer therapies to reduce inflammation-driven tumorigenesis.
Collapse
Affiliation(s)
- Anghesom Ghebremedhin
- Moores Cancer Center, University of California, San Diego, California, USA
- Department of Pathology, University of California, San Diego, California, USA
| | - Judith A Varner
- Moores Cancer Center, University of California, San Diego, California, USA
- Department of Pathology, University of California, San Diego, California, USA
| |
Collapse
|
7
|
Zhang S, Du K, Gao S, Liu Z, Chen L, Wu X, Li L. APM-Related gene signature model to predict prognosis and immunotherapy response in hepatocellular carcinoma. Hum Genet 2025:10.1007/s00439-025-02753-x. [PMID: 40423787 DOI: 10.1007/s00439-025-02753-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2025] [Accepted: 05/09/2025] [Indexed: 05/28/2025]
Abstract
Hepatocellular carcinoma (HCC) is a primary liver malignancy with a dismal prognosis. This study established and validated a prognostic model based on antigen-processing and presenting machinery (APM)-related genes through Mendelian randomization and publicly available datasets. Systematic analysis revealed CXCL5, SGPP2, and GLP1R as critical prognostic biomarkers, which were subsequently integrated into a risk model. The model demonstrated significant associations with pathways linked to bile acid, fatty acid, and amino acid metabolism, alongside variations in immune cell infiltration and genomic mutations, including TTN, TP53, and MUC16. Patient stratification into high- and low-risk groups indicated that low-risk individuals exhibited reduced immune infiltration, potentially correlating with enhanced immunotherapy sensitivity. These findings offer a robust gene signature for HCC prognosis and a framework for evaluating responses to immunotherapy.
Collapse
Affiliation(s)
- Shangdi Zhang
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Kewei Du
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Shan Gao
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Zejing Liu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Linmei Chen
- Operating Theatre, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Xue Wu
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
- Department of Cardiovascular Medicine, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China
| | - Linjing Li
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China.
- Cuiying Biomedical Research Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou, 730030, China.
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, No. 82, Cuiyingmen Street, Chengguan District, Lanzhou, Gansu Province, 730030, China.
| |
Collapse
|
8
|
Jia H, Wei J, Zheng W, Li Z. The dual role of autophagy in cancer stem cells: implications for tumor progression and therapy resistance. J Transl Med 2025; 23:583. [PMID: 40414839 DOI: 10.1186/s12967-025-06595-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Accepted: 05/08/2025] [Indexed: 05/27/2025] Open
Abstract
Cancer stem cells (CSCs) constitute a small yet crucial subgroup in tumors, known for their capacity to self-renew, differentiate, and promote tumor growth, metastasis, and resistance to therapy. These characteristics position CSCs as significant factors in tumor recurrence and unfavorable clinical results, emphasizing their role as targets for therapy. Autophagy, an evolutionarily preserved cellular mechanism for degradation and recycling, has a complex function in cancer by aiding cell survival during stress and preserving balance by eliminating damaged organelles and proteins. Although autophagy can hinder tumor growth by reducing genomic instability, it also aids tumor advancement, particularly in harsh microenvironments, highlighting its dual characteristics. Recent research has highlighted the complex interactions between autophagy and CSCs, showing that autophagy governs CSC maintenance, boosts survival, and aids in resistance to chemotherapy and radiotherapy. On the other hand, in specific situations, autophagy may restrict CSC growth by increasing differentiation or inducing cell death. These intricate interactions offer both obstacles and possibilities for therapeutic intervention. Pharmacological modulation of autophagy, via inhibitors like chloroquine or by enhancing autophagy when advantageous, has demonstrated potential in making CSCs more responsive to standard treatments. Nonetheless, applying these strategies in clinical settings necessitates a better understanding of context-dependent autophagy dynamics and the discovery of dependable biomarkers indicating autophagic activity in CSCs. Progressing in this area might unveil novel, accurate strategies to tackle therapy resistance, lessen tumor recurrence, and ultimately enhance patient outcomes.
Collapse
Affiliation(s)
- Haiqing Jia
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Jing Wei
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China
| | - Wei Zheng
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| | - Zhuo Li
- Department of Gynecology, Cancer Hospital of Dalian University of Technology, Liaoning Cancer Hospital & Institute, No.44 xiaoheyan road, Shenyang, 110042, China.
| |
Collapse
|
9
|
Pingen M, Hughes CE, Medina-Ruiz L, Mathie H, Barrie JA, Hansell CA, Bartolini R, MacLeod MK, Graham GJ. Inflammatory chemokine receptors CCR1, CCR2, CCR3 and CCR5 are essential for an optimal T cell response to influenza. Mucosal Immunol 2025:S1933-0219(25)00052-2. [PMID: 40414601 DOI: 10.1016/j.mucimm.2025.05.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 05/09/2025] [Accepted: 05/14/2025] [Indexed: 05/27/2025]
Abstract
Inflammatory chemokine receptors CCR1/2/3/5 (iCCRs) play an important role in the recruitment of immune cells involved in innate immune functions and orchestrating the adaptive immune response. Here we utilise an influenza A virus (IAV) challenge to investigate the combinatorial roles of the iCCRs in the anti-IAV immune response. We did not observe any gross differences in infection-driven pathology in the absence of iCCRs. iCCR deletion resulted in decreased numbers of some antigen-presenting cell types in the lung (B cells, DC1s, monocytes and inflammatory macrophages), though cell numbers in the draining lymph node were not affected. Whilst the total number of T cells was similar in lungs of iCCR-deficient mice, the number of IAV-specific CD4 but not CD8 T cells in the lung was strongly reduced in the absence of iCCRs. Furthermore, fewer CD4, but not CD8, T cells produced IFN-γ. This CD4 T cell phenotype persisted into the memory stage of infection, with fewer IAV-specific and IFN-γ+ CD4 but not CD8 T cells at 29 days post infection. In conclusion, despite having limited impact on antigen-presenting cell migration between the lung and the draining lymph node, iCCR deletion is associated with an altered CD4 T cell response to IAV infection.
Collapse
Affiliation(s)
- Marieke Pingen
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom.
| | - Catherine E Hughes
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Laura Medina-Ruiz
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Heather Mathie
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Jennifer A Barrie
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Chris Ah Hansell
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Robin Bartolini
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Megan Kl MacLeod
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom
| | - Gerard J Graham
- School of Infection and Immunity, College of Medical, Veterinary and Life Sciences, University of Glasgow, United Kingdom.
| |
Collapse
|
10
|
Peng Y, Liu H, Miao M, Cheng X, Chen S, Yan K, Mu J, Cheng H, Liu G. Micro-Nano Convergence-Driven Radiotheranostic Revolution in Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:29047-29081. [PMID: 40347149 DOI: 10.1021/acsami.5c05525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/12/2025]
Abstract
Radiotherapy, as an important means of treating hepatocellular carcinoma (HCC), has shown unique therapeutic advantages, especially in patients who are unable to undergo surgery or transplantation. It mainly includes external radiotherapy, transarterial radioembolization and intratumoral radioactive particle implantation. However, under the influence of factors such as the hypoxic characteristics of the liver tumor microenvironment and the radioresistance of tumor cells, the effect of radiotherapy may be unstable and may cause side effects, affecting the quality of life of patients. In recent years, with the development of nanotechnology, drug delivery systems based on micro-nanomaterials have provided new solutions for improving the effect of radiotherapy for HCC. Despite this, the application of micro-nano drug delivery systems in the treatment of HCC still faces some challenges, mainly including the in vivo safety and in vivo metabolism of micro-nano materials. This article reviews the latest progress of micro-nano materials in the treatment of HCC, especially their application in radiosensitization and their clinical translation potential. This article systematically analyzes the role of micro-nanomaterials in external or internal radiotherapy sensitization and radioimmunotherapy and explores the advantages of micro-nanomaterials in improving the treatment effect of HCC.
Collapse
Affiliation(s)
- Yisheng Peng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Hui Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Mengmeng Miao
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Xu Cheng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Shangqing Chen
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Kaifei Yan
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| | - Jing Mu
- Institute of Precision Medicine, Peking University Shenzhen Hospital, Shenzhen 518036, China
| | - Hongwei Cheng
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
- Zhuhai UM Science & Technology Research Institute, University of Macau, Macau SAR 999078, China
| | - Gang Liu
- State Key Laboratory of Vaccine for Infectious Diseases, Xiang An Biomedicine Laboratory, National Innovation Platform for Industry-Education Integration in Vaccine Research, State Key Laboratory of Molecular Vaccinology and Molecular Diagnostics, School of Public Health, Xiamen University, Xiamen 361102, China
| |
Collapse
|
11
|
Jiang Y, Dong X, Zhang Y, Su F, Zhao L, Shi X, Zhong J. Navigating the complexities: challenges and opportunities in conversion therapy for advanced hepatocellular carcinoma. Clin Exp Med 2025; 25:169. [PMID: 40382739 PMCID: PMC12086121 DOI: 10.1007/s10238-025-01698-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Accepted: 04/14/2025] [Indexed: 05/20/2025]
Abstract
Primary liver cancer ranks as the sixth most prevalent malignant tumor and stands as the second leading cause of cancer-related mortality globally, posing a significant threat to public health. Hepatocellular carcinoma (HCC) is the most common type of liver cancer worldwide. Surgical resection remains the cornerstone treatment for achieving radical cure and prolonged survival in HCC patients. Contrary to Western countries, the majority of HCC patients in China present with hepatitis B virus infection and consequent liver cirrhosis, with most cases diagnosed at an intermediate or advanced stage. This complexity results in a poor prognosis. Recent advancements in local therapeutic techniques and the introduction of systemic therapies, including targeted and immunotherapy agents, have provided new avenues for both clinical and basic conversion therapy for advanced HCC. Integrating multi-dimensional local and systemic therapies, multi-modal sequential, and comprehensive multidisciplinary approaches into the management of HCC patients has demonstrated promising conversion success rates. This holistic management strategy involves combining multiple treatment modalities vertically and coordinating various disciplines horizontally. However, significant challenges remain, including the precise selection of patients eligible for conversion therapy, the optimal choice of conversion therapy regimens, and the accurate determination of surgical timing post-conversion therapy. Addressing these challenges is crucial for hepatobiliary surgeons. High-quality, randomized controlled trials are urgently needed to generate robust evidence for clinical practice. This review aims to synthesize the latest research developments both in China and internationally and examines key issues in the realm of HCC conversion therapy.
Collapse
Affiliation(s)
- Yubo Jiang
- Department of Gastroenterology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Science, Jinan, Shandong Province, China
| | - Xiaofeng Dong
- Department of Hepatobiliary, Pancreas and Spleen Surgery, the People's Hospital of Guangxi Zhuang Autonomous Region (Guangxi Academy of Medical Sciences), Nanning, Guangxi Zhuang Autonomous Region, China
| | - Yingying Zhang
- Department of Oncology, Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong Province, China
| | - Feiyan Su
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Lei Zhao
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong Province, China.
| |
Collapse
|
12
|
Patysheva MR, Fedorenko AA, Khozyainova AA, Denisov EV, Gerashchenko TS. Immune Evasion in Cancer Metastasis: An Unappreciated Role of Monocytes. Cancers (Basel) 2025; 17:1638. [PMID: 40427136 PMCID: PMC12110646 DOI: 10.3390/cancers17101638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2025] [Revised: 05/02/2025] [Accepted: 05/10/2025] [Indexed: 05/29/2025] Open
Abstract
Metastasis is the leading cause of cancer-related deaths. During the metastatic cascade, cancer cells tightly interact with immune cells influencing each other in the tumor microenvironment and systemically. Monocytes are important components of immune evasion and critical regulators of cancer progression. They circulate through the bloodstream and contribute to the formation of a pro-tumor microenvironment both in the tumor and pre-metastatic niche. Whereas monocyte participation in cancer development and response to therapy has been described extensively, its impact on metastasis remains a completely uncovered area. This review first summarizes data concerning the influence of monocytes on metastasis formation during their presence in the circulation, primary tumor, and pre-metastatic niche. We also highlight the latest examinations into the clinical relevance of targeting monocytes to prevent metastasis.
Collapse
Affiliation(s)
- Marina R. Patysheva
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk 634009, Russia (E.V.D.); (T.S.G.)
| | - Anastasya A. Fedorenko
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk 634009, Russia (E.V.D.); (T.S.G.)
| | - Anna A. Khozyainova
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk 634009, Russia (E.V.D.); (T.S.G.)
| | - Evgeny V. Denisov
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk 634009, Russia (E.V.D.); (T.S.G.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya Str. 6, Moscow 115093, Russia
| | - Tatiana S. Gerashchenko
- Cancer Research Institute, Tomsk National Research Medical Center, Russian Academy of Sciences, Kooperativny Str. 5, Tomsk 634009, Russia (E.V.D.); (T.S.G.)
- Research Institute of Molecular and Cellular Medicine, Peoples’ Friendship University of Russia (RUDN University), Miklukho-Maklaya Str. 6, Moscow 115093, Russia
| |
Collapse
|
13
|
Yalikun K, Li Z, Zhang J, Chang Z, Li M, Sun Z, Liu Z, Yang Y, Xu L, Li L, Zhang C, Sun P, Zhong J, Cui K, Shi X, Zhang B, Zhao L. Hepatic artery infusion chemotherapy combined with camrelizumab and apatinib as conversion therapy for patients with unresectable hepatocellular carcinoma: a single-arm exploratory trial. BMC Cancer 2025; 25:838. [PMID: 40335980 PMCID: PMC12056981 DOI: 10.1186/s12885-025-14250-5] [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: 10/25/2024] [Accepted: 04/29/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND The development of systemic therapy, including targeted drugs and immune checkpoint inhibitors, has significantly improved the prognosis of patients with advanced unresectable hepatocellular carcinoma (uHCC). Hepatic arterial infusion chemotherapy (HAIC) has been gradually applied to the treatment of advanced uHCC, showing good potential as conversion therapy. We aimed to investigate the efficacy and safety of HAIC combined with camrelizumab and apatinib as conversion therapy for uHCC. METHODS This study was a single-arm exploratory trial (NCT05099848) in patients with uHCC. Eligible patients received apatinib 250 mg once daily, camrelizumab 200 mg on day 3, and HAIC with FOLFOX regimen (oxaliplatin 85 mg/m2 at hours 0-2, leucovorin 400 mg/m2 at hours 2-3, and fluorouracil 400 mg/m2 at hour 3, followed by fluorouracil 2400 mg/m2 for 46 h) on days 4-5 of each 21-day cycle for up to 8 cycles. Primary endpoints were conversion rate and margin-free (R0) resection rate. RESULTS Between March 2021 and July 2023, 19 patients were enrolled. Median follow-up was 14.9 months (interquartile range, 10.9-21.1). Disease became resectable in 14 (73.7%) of 19 patients; nine (47.4%) patients received R0 resection, while five (26.3%) refused surgery and opted for observation. Three (33.3%) of nine patients with surgery achieved major pathological response, including two (22.2%) with pathological complete response. Objective response and disease control rates were 47.4% (9/19) and 89.5% (17/19) per Response Evaluation Criteria In Solid Tumors (RECIST) version 1.1 and both 89.5% (17/19) per modified RECIST. Survival data were immature. Fourteen (73.7%) of 19 patients had grade 3 or higher treatment-related adverse events, with the most common being increased alanine aminotransferase or aspartate aminotransferase (seven [36.8%]) and increased lymphocyte count (six [31.6%]). No treatment-related deaths occurred. CONCLUSIONS The combination of HAIC, camrelizumab, and apatinib as conversion therapy shows promising clinical benefits and a manageable safety profile in patients with uHCC. Future randomized controlled trials are warranted. TRIAL REGISTRATION ClinicalTrials.gov NCT05099848. Registered on October 13, 2021.
Collapse
Affiliation(s)
- Kugeluke Yalikun
- The Affiliated Cancer Hospital of Xinjiang Medical University, 789 Suzhou East Road, Xinshi District, Urumqi, China
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Zhongchao Li
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China.
| | - Jianxin Zhang
- Department of Intervention Oncology, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, China
| | - Zhibin Chang
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China
| | - Mingming Li
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China
| | - Zhicheng Sun
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China
| | - Zhaogang Liu
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Yue Yang
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China
| | - Lei Xu
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China
| | - Lei Li
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Chengsheng Zhang
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Pengfei Sun
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Jingtao Zhong
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Kai Cui
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Xuetao Shi
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Bo Zhang
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China
| | - Lei Zhao
- The Affiliated Cancer Hospital of Xinjiang Medical University, 789 Suzhou East Road, Xinshi District, Urumqi, China.
- Department of Hepatobiliary Surgery, Shandong First Medical University and Shandong Academy of Medical Sciences, Shandong Cancer Hospital and Institute, 440 Jiyan Road, Huaiyin District, Jinan, 250117, China.
- Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, 6699 Qingdao Road, Huaiyin District, China.
| |
Collapse
|
14
|
Aquino A, Franzese O. Reciprocal Modulation of Tumour and Immune Cell Motility: Uncovering Dynamic Interplays and Therapeutic Approaches. Cancers (Basel) 2025; 17:1547. [PMID: 40361472 PMCID: PMC12072109 DOI: 10.3390/cancers17091547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2025] [Revised: 04/28/2025] [Accepted: 04/30/2025] [Indexed: 05/15/2025] Open
Abstract
Dysregulated cell movement is a hallmark of cancer progression and metastasis, the leading cause of cancer-related mortality. The metastatic cascade involves tumour cell migration, invasion, intravasation, dissemination, and colonisation of distant organs. These processes are influenced by reciprocal interactions between cancer cells and the tumour microenvironment (TME), including immune cells, stromal components, and extracellular matrix proteins. The epithelial-mesenchymal transition (EMT) plays a crucial role in providing cancer cells with invasive and stem-like properties, promoting dissemination and resistance to apoptosis. Conversely, the mesenchymal-epithelial transition (MET) facilitates metastatic colonisation and tumour re-initiation. Immune cells within the TME contribute to either anti-tumour response or immune evasion. These cells secrete cytokines, chemokines, and growth factors that shape the immune landscape and influence responses to immunotherapy. Notably, immune checkpoint blockade (ICB) has transformed cancer treatment, yet its efficacy is often dictated by the immune composition of the tumour site. Elucidating the molecular cross-talk between immune and cancer cells, identifying predictive biomarkers for ICB response, and developing strategies to convert cold tumours into immune-active environments is critical to overcoming resistance to immunotherapy and improving patient survival.
Collapse
Affiliation(s)
| | - Ornella Franzese
- Department of Systems Medicine, University of Rome Tor Vergata, Via Montpellier 1, 00133 Rome, Italy;
| |
Collapse
|
15
|
Zhou S, Song C, Liu P, Ju S, Wang YC. A nationwide investigation on imaging follow-up after Locoregional therapy for hepatocellular carcinoma in China: Current practices and challenges. Eur J Radiol 2025; 186:112057. [PMID: 40132470 DOI: 10.1016/j.ejrad.2025.112057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2025] [Revised: 02/22/2025] [Accepted: 03/17/2025] [Indexed: 03/27/2025]
Abstract
PURPOSE To investigate the perspectives of interventional radiologists in China on imaging follow-up protocols and adherence to treatment response criteria for hepatocellular carcinoma (HCC) following locoregional therapies (LRT), with a particular focus on identifying gaps and proposing strategies to bridge the discrepancy between clinical guidelines and real-world practice. MATERIALS AND METHODS The web-based survey was conducted among members of Chinese Society of Radiology and Zhongda Radiology Alliance between 1st October 2024 to 30th November 2024, via WPS Office survey tool. The frequencies and percentages of responses were summarized as n (%), and a chi-squared test was employed to compare the responses among diagnostic radiologists, interventional radiologists, and interventional physicians. RESULTS A total of 604 respondents from 325 hospitals in 31 provinces were analyzed. Of the respondents, 72.7 % (439/604) believed that the same imaging modality should be used for follow-up after conventional transarterial chemoembolization (cTACE) and other LRT for HCC. Among these respondents, contrast-enhanced computed tomography (CE-CT) (57.2 %, 251/439) was the most preferred imaging modality for initial follow-up, and hepatobiliary contrast-enhanced MRI (63.1 %, 277/439) for subsequent follow-up examinations. For respondents (27.3 %, 165/604) who believed that follow-up strategies should be tailored to the type of LRT, CE-CT was most commonly recommended for post-cTACE HCC, both for initial (64.2 %, 106/165) and subsequent (60.6 %, 100/165) follow-up. For HCC treated with other LRT, the majority of respondents preferred extracellular contrast-enhanced MRI for initial follow-up (55.8 %, 92/165) and hepatobiliary contrast-enhanced MRI for subsequent follow-up (61.2 %, 101/165). The most recommended time frame for initial follow-up was "within 1-2 months" among all respondents. However, significant differences in the recommended time frame were observed among diagnostic radiologists, interventional radiologists, and interventional physicians (P < 0.001). Notably, more than 95 % of the respondents who selected 'unclear' were diagnostic radiologists. The most recommended monitoring frequency was every 3-4 months (46.0 %, 278/604) for viable lesions and every 5-6 months (32.9 %, 199/604) for nonviable lesions. Regarding adherence to treatment response criteria, mRECIST (32.0 %, 193/604) and LR-TRA (v2017/v2024) (24.2 %, 146/604) were commonly adopted in clinical practice. Nevertheless, a significant proportion of respondents (25.2 %, 152/604) indicated that none of these criteria were applied in their clinical practice. CONCLUSION CE-CT performed within 1-2 months was the most preferred modality and time frame for initial follow-up. Significant variability remains in follow-up frequency and treatment response criteria for post-LRT HCC, highlighting the need for further standardization of imaging follow-up protocols and structured treatment response assessment to optimize post-LRT management in clinical practice.
Collapse
Affiliation(s)
- Shuwei Zhou
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Chenxin Song
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Pei Liu
- Department of Epidemiology and Biostatistics, School of Public Health, Southeast University, Nanjing 210009, China.
| | - Shenghong Ju
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| | - Yuan-Cheng Wang
- Department of Radiology, Zhongda Hospital, Nurturing Center of Jiangsu Province for State Laboratory of AI Imaging & Interventional Radiology, School of Medicine, Southeast University, Nanjing 210009, China.
| |
Collapse
|
16
|
Shen W, Li Z, Wang L, Liu Q, Zhang R, Yao Y, Zhao Z, Ji L. Tumor-resident Malassezia can promote hepatocellular carcinoma development by downregulating bile acid synthesis and modulating tumor microenvironment. Sci Rep 2025; 15:15020. [PMID: 40301518 PMCID: PMC12041395 DOI: 10.1038/s41598-025-99973-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2025] [Accepted: 04/24/2025] [Indexed: 05/01/2025] Open
Abstract
Bacterial dysbiosis coincides with the carcinogenesis in malignancies such as lung and colon cancer, and has recently been suggested to involve in the pathogenesis of hepatocellular carcinoma (HCC). However, the mycobiome has not yet been definitively linked to liver tumorigenesis. Here we showed that the microbiota composition of HCC tumors was distinct from that of the normal adjacent to tumor (NAT) on the basis of richness and beta-diversity indices. Specifically, the fungal community that infiltrated HCC tumors was markedly enriched for Malassezia spp. and genus Malassezia in tumors was substantially more abundant than that in NAT. We also discovered that the relative abundance of genus Malassezia was strongly correlated with the tumor microenvironment (TME) signatures, including stromal and immune components. In addition, tumor-resident Malassezia could inhibit bile acid synthesis by downregulating the expression level of CYP7 A1 and CYP27 A1. To improve clinical usability, we developed a set of Malassezia-related genes, called Malassezia.Sig, which could accurately predict patient survival. Collectively, our work shows that tumor-resident Malasseiza may promote HCC progression by downregulating bile acid synthesis and modulating the TME, although more studies are needed.
Collapse
Affiliation(s)
- Weixi Shen
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Zhihong Li
- Ganzi Tibetan Autonomous Prefecture People's Hospital, Tibet, 850002, China
| | - Lei Wang
- Women's Health Section, Harbin Red Cross Central Hospital, Harbin, 150076, China
| | - Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Renjie Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150086, China
| | - Yuhua Yao
- School of Mathematics and Statistics, Hainan Normal University, Haikou, 571158, China
| | - Zhicheng Zhao
- The Fourth Affiliated Hospital of Heilongjiang University of Traditional Chinese Medicine, Harbin, 150018, China.
| | - Lei Ji
- Geneis Beijing Co., Ltd, Beijing, 100102, China.
| |
Collapse
|
17
|
Ang ZH, Wong SW. Management of the Malignant Rectal Polyp-A Narrative Review. Cancers (Basel) 2025; 17:1464. [PMID: 40361391 PMCID: PMC12071011 DOI: 10.3390/cancers17091464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
PURPOSE The aim of this review is to provide a contemporary update on the current management approaches and options with specific considerations in malignant rectal polyps. METHODS A literature review was carried out in PubMed, Embase and Cochrane databases using the keywords "malignant" and "polyp*". Only publications in English language were included. RESULTS Histopathological features including margins, depth of invasion, tumour grade, LVI and tumour budding determines the risk of lymph node metastasis in malignant polyps. Rectal malignant polyps should be considered differently compared to their colonic counterpart. A low threshold should be considered for utilising transrectal excision to fully excise the polyp and to assess the margins. The rates of complete pathological response associated with total neoadjuvant therapy as well as the advent of "watch and wait" adds to the complexity of managing malignant rectal polyps. CONCLUSIONS The management of malignant colorectal polyps lies in risk-stratifying patients who will benefit from an oncological resection.
Collapse
Affiliation(s)
- Zhen Hao Ang
- Department of Colorectal Surgery, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
- Randwick Campus, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2033, Australia
| | - Shing Wai Wong
- Department of Colorectal Surgery, Prince of Wales Hospital, Sydney, NSW 2031, Australia;
- Randwick Campus, School of Clinical Medicine, University of New South Wales, Sydney, NSW 2033, Australia
| |
Collapse
|
18
|
Zheng Q, Zhou T, Yao D. The roles of immune cells and non-immune cells in Pre-Metastatic Niche of Breast Cancer. Crit Rev Oncol Hematol 2025; 211:104744. [PMID: 40274245 DOI: 10.1016/j.critrevonc.2025.104744] [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: 03/20/2025] [Revised: 04/18/2025] [Accepted: 04/19/2025] [Indexed: 04/26/2025] Open
Abstract
Distant metastasis is a pivotal and important event in patients with breast cancer, and inhibition of metastasis has always been the focus of clinical research. Recent advances have established that the metastasis of breast cancer is exacerbated not only by cancer cells and the tumor microenvironment but also by the pre-metastatic niche (PMN). Primary tumor secretory factors, immune cells including bone marrow-derived cells mobilized by tumors and non-immune cells within the local matrix microenvironment of the host are three key factors for PMN formation. This article reviews the roles of bone marrow-derived cells, lymphocytes, fibroblasts, endothelial cells, epithelial cells and cancer stem cells in the establishment of PMN before metastasis to further understand the metastasis mechanism of breast cancer and to explore clues for the inhibition of distant metastasis. Different cells play distinct but important roles in the establishment of the PMN and the induction of breast cancer metastasis. The interaction between different cells and tumor cells determines whether CTCs can be attached, survive and proliferate to promote distant metastasis.
Collapse
Affiliation(s)
- Qiao Zheng
- Department of Oncology, Hospital of Chengdu Uiversity of Traditional Chinese Medicine, 610075 Chengdu, China
| | - Tiecheng Zhou
- Department of Oncology, Sichuan Integrative Medicine Hospital, 610042 Chengdu, China,.
| | - Dejiao Yao
- Department of Oncology, Hospital of Chengdu Uiversity of Traditional Chinese Medicine, 610075 Chengdu, China
| |
Collapse
|
19
|
Wang J, Cui J, Hao T, Zhang Q, Chen Y, Guo L, Tong Y, Dong D. Regulation of cyclophosphamide induced hepatotoxicity by REV-ERBα modifiers. Expert Opin Drug Metab Toxicol 2025:1-11. [PMID: 40211567 DOI: 10.1080/17425255.2025.2490741] [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: 07/26/2024] [Accepted: 03/28/2025] [Indexed: 04/14/2025]
Abstract
INTRODUCTION Cyclophosphamide (CPA) is a widely used broad-spectrum antitumor drug with severe hepatotoxicity. Finding an effective way to mitigate the hepatotoxicity caused by CPA is a challenge in its clinical application. METHODS In Rev-erbα knockout and wild-type mice, hepatotoxicity was evaluated by ALT, AST, and histopathological scores 4-h post dose of CPA (i.p. 300 mg/kg). CYP2B10 expression and pharmacokinetic behavior of CPA were also detected. SR9009 (i.p. 10 mg/kg) and Berberine (BBR, i.p. 50 mg/kg) were pre-administered to mice. Then, the measurements were carried out following the same procedures as previous. The regulation effects of SR9009 and BBR on CYP2B10 were validated using Hepa-1c1c7 cells. RESULTS Firstly, REV-ERBα negatively regulated CPA-induced hepatotoxicity by altering the expression of CYP2B10 and CPA pharmacokinetics. Secondly, REV-ERBα agonists, SR9009 and BBR, increased REV-ERBα expression and alleviated hepatic toxicity of CPA. Furthermore, both SR9009 and BBR reduced expression of CYP2B10 and REV-ERBα target gene Bmal1 both in vivo and in vitro. CONCLUSIONS REV-ERBα agonists can significantly attenuate the hepatotoxicity of CPA by regulating CYP2B10. The discovery of REV-ERBα as novel regulator for CYP2B10 will help to establish new targets to improve drug efficacy or reduce toxicity.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Jialu Cui
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| | - Tingying Hao
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Qi Zhang
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Yutong Chen
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Lianxia Guo
- Institute of Molecular Rhythm and Metabolism, Guangzhou University of Chinese Medicine, Guangzhou, China
| | - Yongbin Tong
- School of Pharmacy, Guangdong Medical University, Dongguan, China
| | - Dong Dong
- Department of Public Health and Preventive Medicine, School of Medicine, Jinan University, Guangzhou, China
| |
Collapse
|
20
|
Liu GM, Guo R, Xu JW. A bibliometric and visual analysis based on immune checkpoint inhibitors for hepatocellular carcinoma: 2014 - 2024. Front Pharmacol 2025; 16:1520055. [PMID: 40260385 PMCID: PMC12009821 DOI: 10.3389/fphar.2025.1520055] [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: 10/30/2024] [Accepted: 03/24/2025] [Indexed: 04/23/2025] Open
Abstract
Background Immune checkpoint inhibitors (ICIs) have changed the treatment landscape of hepatocellular carcinoma (HCC), especially those with unresectable advanced stages. The field has progressed rapidly, and the research hotspots have significantly changed compared to previous years. The study aims to comprehensively review and analyze the development history, knowledge structure, current research focus, and emerging trends in ICIs for HCC. Materials and methods Reviews and articles published in English from The Web of Science Core Collection (WoSCC) database from 2014 to 2024 were systemically retrieved. Citespace, VOSviewer, and Bibliometrix R package were used for further bibliometric analysis and visualization for countries, institutions, authors, references, and keywords. Results 2,941 records were included for analysis. The literature on ICIs for HCC has continued to grow steadily over the past decade. Three major research centers have emerged: North America, Europe, and East Asia. The Chinese institution has the highest publication volume, but Kudo Masatoshi from Japan has the highest number of publications. At the same time, Richard S. Finn from the United States leads in citations and co-citations. The most prolific journal is "Cancers". The clustering and Timeline view of critical literature and keywords indicated that research on ICIs for HCC is rapidly advancing toward a more evidence-based, personalized, and multimodal approach. Immune evasion mechanisms, predictive biomarkers, and high-quality clinical trials focusing on Novel combination, conversion, and perioperative therapies, including ICIs, are emerging hotspots. Conclusion This study highlights the groundbreaking advancements of ICIs in treating HCC and shows a trend rapidly advancing towards a more evidence-based, personalized, and multimodal approach. The study updated the current understanding of ICIs in hepatocellular carcinoma and identified vital future directions for research, such as the exploration of mechanisms of immune evasion, developing predictive biomarkers, and combining therapy strategies.
Collapse
Affiliation(s)
- Gao-Min Liu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Rui Guo
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| | - Ji-Wei Xu
- Meizhou Clinical Medical College of Shantou University Medical College, Meizhou, China
- Department of Hepatobiliary Surgery, Meizhou People’s Hospital, Meizhou, China
| |
Collapse
|
21
|
Pan Z, Liu D, Cao J, Fu L, Zhang X, Zhu X, Pan Y, Liu J, Han C, Jin R, Shen S, Zhang X, Liu H, Yang X, Hu K, Shi X, Wang D, Zhao Y, Zhong J, Xiang B, Gu S, Li T, Zhang S, Zhou L, Zhao H, Zeng Y, Wen T, Kuang M, Liang X, Peng T, Wang K, Xu L, Li H, Song T, Sun H, Zhang W, China Liver Cancer Study Group Young Investigators (CLEAP). Comparison of Efficacy between Lenvatinib and Bevacizumab in Combination of Immune Checkpoint Inhibitor and Interventional Triple Therapy in Chinese Advanced Hepatocellular Carcinoma: The CLEAP 2302 Study. Liver Cancer 2025:1-19. [PMID: 40417458 PMCID: PMC12097796 DOI: 10.1159/000545545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 02/10/2025] [Indexed: 05/27/2025] Open
Abstract
Background The conversion therapy for advanced hepatocellular carcinoma (HCC) shows promise with a triple therapy approach that combines interventional therapy, immune checkpoint inhibitors, and molecular targeted therapy (primarily small-molecule TKIs and the large-molecule bevacizumab). This combination has achieved the highest objective response rates (ORR) along with acceptable safety profiles. The aim of this study was to compare the clinical efficacy of lenvatinib versus bevacizumab, when combined with immune checkpoint inhibitors and interventional triple therapy, as first-line treatments for Chinese patients with unresectable HCC (uHCC). Method This retrospective multicenter study involved 371 consecutive patients from 21 centers in China, observed between April 2017 and December 2023. The study focused on patients with uHCC at Chinese liver cancer stages IIb to IIIb (Barcelona Clinic Liver Cancer stage B or C) who received lenvatinib or bevacizumab combined with anti-PD-1/L1 and interventional therapy (including TACE and/or HAIC) as first-line treatment. Of the 371 patients, 258 received lenvatinib-based triple therapy, while 113 received bevacizumab-based triple therapy. The primary endpoints were overall survival (OS) and progression-free survival (PFS). To balance baseline clinical characteristics, propensity score matching (PSM) and inverse probability of treatment weighting (IPTW) were applied. Subgroup analysis was also performed based on different clinicopathological characteristics of the enrolled uHCC patients. Results The median OS in the lenvatinib group was significantly longer than in the bevacizumab group, both before (36.0 vs. 27.9 months; hazard ratio [HR]: 0.536; 95% confidence interval [CI]: 0.344-0.835; p = 0.0016) and after PSM (HR: 0.524; 95% CI: 0.305-0.900; p = 0.01), as well as after IPTW (HR: 0.549; 95% CI: 0.331-0.908; p = 0.01). Before adjustment, PFS in the lenvatinib group was also significantly longer than in the bevacizumab group (20.0 vs. 12.1 months; HR: 0.649; 95% CI: 0.457-0.922; p = 0.0078). However, after PSM (HR: 0.808; 95% CI: 0.535-1.222; p = 0.33) and IPTW, there was no significant difference in PFS between the two groups. Multivariate analysis showed that lenvatinib-based triple therapy was independently associated with improved OS compared to bevacizumab-based triple therapy. Subgroup analysis indicated that patients with age ≤65 years, no history of hepatitis B virus infection, Barcelona Clinic Liver Cancer stage C (BCLC-C), ALT levels ≤40 U/L, platelets ≥100 × 109/L, or log 10 AFP ≥1.40 benefited more from lenvatinib-based triple therapy. Conclusion Lenvatinib-based triple therapy tends to prolong OS compared to bevacizumab, although the PFS was similar between the two groups. Patients aged ≤65 years, without a history of hepatitis B virus infection, with BCLC-C stage, ALT ≤40 U/L, platelets ≥100 × 109/L, or log 10 AFP ≥1.40 are likely to benefit more from lenvatinib-based triple therapy.
Collapse
Affiliation(s)
- Zhaolong Pan
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Dongming Liu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Junbo Cao
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Linlin Fu
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xihao Zhang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Xiaodong Zhu
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Yangxun Pan
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Jianwei Liu
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Chuangye Han
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Renan Jin
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
| | - Shunli Shen
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiaoyun Zhang
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Hongzhi Liu
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Xiaobo Yang
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Kuan Hu
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoyi Shi
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Dongxu Wang
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Yang Zhao
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Jianhong Zhong
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Bangde Xiang
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| | - Shanzhi Gu
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Tao Li
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
| | - Shuijun Zhang
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ledu Zhou
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Haitao Zhao
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
| | - Yongyi Zeng
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Tianfu Wen
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
| | - Ming Kuang
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Xiao Liang
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
| | - Kui Wang
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Li Xu
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
| | - Huikai Li
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Tianqiang Song
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - Huichuan Sun
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
| | - Wei Zhang
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
| | - China Liver Cancer Study Group Young Investigators (CLEAP)
- Tianjin Medical University Cancer Institute & Hospital, National Clinical Research Center for Cancer, Tianjin, China
- Tianjin’s Clinical Research Center for Cancer, Tianjin, China
- Tianjin Key Laboratory of Digestive Cancer, Tianjin, China
- Department of Liver Surgery and Transplantation, Liver Cancer Institute and Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Liver Surgery, Sun Yat-Sen University Cancer Center, Sun Yat-Sen University, Guangzhou, China
- Department of Hepatic Surgery II, The Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, China
- Department of General Surgery, Sir Run Run Shaw Hospital, College of Medicine, Institute of Minimally Invasive Surgery, Zhejiang University, Hangzhou, China
- Department of Hepatic Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
- Department of Liver Surgery, West China Hospital of Sichuan University, Chengdu, China
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatobiliary Surgery, Peking Union Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Department of Hepatobiliary Surgery, Xiangya Hospital, Central South University, Changsha, China
- Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Department of General Surgery, Qilu Hospital, Shandong University, Jinan, China
- Department of Interventional Therapy, Hunan Cancer Hospital, The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
- Hepatobiliary Surgery Department, Guangxi Medical University Cancer Hospital, Nanning, China
| |
Collapse
|
22
|
Gao CH, Cao LP, Cheng SQ, Wang L, Wang JZ, Yan ML, Zheng SS, Yang Z. Radiologic complete response after conversion therapy for hepatocellular carcinoma: is liver transplantation necessary? Hepatobiliary Surg Nutr 2025; 14:351-354. [PMID: 40342783 PMCID: PMC12057504 DOI: 10.21037/hbsn-2024-702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 05/11/2025]
Affiliation(s)
- Chen-Hao Gao
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
| | - Lin Ping Cao
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shu-Qun Cheng
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Lei Wang
- Department of Hepatic Surgery VI, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Ji-Zhou Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of the University of Science and Technology of China, Hefei, China
| | - Mao-Lin Yan
- Department of Hepatobiliary Surgery, Fujian Provincial Hospital of Fujian Medical University, Fuzhou, China
| | - Shu-Sen Zheng
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Boao) Hospital, Boao, China
| | - Zhe Yang
- Department of Hepatobiliary and Pancreatic Surgery, Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Shulan (Hangzhou) Hospital, Shulan International Medical College, Zhejiang Shuren University, Hangzhou, China
- Department of Hepatobiliary and Pancreatic Surgery, Shulan (Boao) Hospital, Boao, China
| |
Collapse
|
23
|
Yan H, Xu J, Li Z, Li N, Guo X, Wu M, Wang D, Lin N, Dong J, Xu X. Efficacy of radiotherapy combined with targeted therapy and immunotherapy for lymph node metastasis of liver cancer. J Cancer Res Clin Oncol 2025; 151:129. [PMID: 40169437 PMCID: PMC11961524 DOI: 10.1007/s00432-025-06182-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025]
Abstract
PURPOSE To investigate the efficacy and safety of radiotherapy combined with targeted therapy and immunotherapy for liver cancer with lymph node metastasis (LNM). METHODS We analysed patients who received radiotherapy for liver cancer with LNM in our hospital from June 2020 to June 2023. 62 patients were enrolled in this study, who received radiotherapy with a median radiation dose of 60.0 Gy, combined with targeted therapy and/or immunotherapy. The objective response rate (ORR), overall survival (OS), progression free survival (PFS), and adverse events were observed to evaluate treatment efficacy and safety. RESULTS With a median follow-up of 18.5 months, the best ORR was 90.3%. The median OS was 26.0 months. The 1-year and 2-year OS rates were 78.93% and 57.37%, respectively. The median PFS was 17.0 months, and the 1-year and 2-year PFS rates were 59.06% and 49.22%, respectively. Multivariate analysis showed that alanine aminotransferase (HR = 2.34, 95% CI 1.07-5.11, P = 0.033), prothrombin time (HR = 4.51, 95% CI 1.76-11.57, P = 0.002), alpha fetal protein (HR = 2.94, 95% CI 1.34-6.45, P = 0.007), and the volume of LNM (HR = 3.05, 95% CI 1.25-7.46, P = 0.014) were independent predictors for OS, while non-regional LNM (HR = 3.19, 95% CI 1.24-8.16, P = 0.016) was an independent predictor for PFS. Toxicity was generally mild and moderate. CONCLUSIONS Radiotherapy combined with targeted therapy and immunotherapy is an effective treatment option, and expected to become new treatment strategy for liver cancer with LNM.
Collapse
Affiliation(s)
- Huamei Yan
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Jianliang Xu
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Zhenghuan Li
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Nuoya Li
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Xianyu Guo
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Manya Wu
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Donghui Wang
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China
| | - Nan Lin
- Department of Hepatobiliary Surgery, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Jie Dong
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| | - Xiangying Xu
- Department of Radiation Oncology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510630, China.
| |
Collapse
|
24
|
Ma X, Lu G. Could the TAE score predicting the prognosis of unresectable HCC patients treated with TACE plus lenvatinib with PD-1 inhibitors be better if certain revisions are made? Hepatol Int 2025; 19:484-485. [PMID: 39976826 DOI: 10.1007/s12072-025-10789-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/31/2025] [Indexed: 04/17/2025]
Affiliation(s)
- Xijing Ma
- Department of Hepatobiliary Pancreatic Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Nine One North Road 105, LongYan, 364000, Fujian, China
| | - Geng Lu
- Department of Hepatobiliary Pancreatic Surgery, Longyan First Affiliated Hospital of Fujian Medical University, Nine One North Road 105, LongYan, 364000, Fujian, China.
| |
Collapse
|
25
|
Bi X, Zhao H, Zhao H, Li G, Wang X, Chen B, Zhang W, Che X, Huang Z, Han Y, Jiang L, Sun Y, Yang Z, Zhou J, Zhang Y, Zhu Z, Chen M, Cheng S, Cai J. Consensus of Chinese Experts on Neoadjuvant and Conversion Therapies for Hepatocellular Carcinoma: 2023 Update. Liver Cancer 2025; 14:223-238. [PMID: 40255878 PMCID: PMC12005702 DOI: 10.1159/000541249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 08/06/2024] [Indexed: 11/25/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy in China, with high recurrence rate and low resection rate among patients first diagnosed. Preoperative treatments including neoadjuvant and conversion therapy have the potential to overcome these challenges. In December 2021, Chinese expert consensus on neoadjuvant and conversion therapies for hepatocellular carcinoma was published. With the emersion of new evidence regarding the neoadjuvant and conversion therapies for HCC, the cooperative group brought together multidisciplinary researchers and scholars with experience in related fields to update the new edition (2023 Edition) for reference in China, including principle of the treatment strategies, the potential populations selection, treatment methods, multidisciplinary team, and future research for preoperative treatments. The new consensus aims to provide guidance for clinical application. Through the use of neoadjuvant therapy and conversion therapy, we can enhance the resection rate and reduce the recurrence of intermediate-to-advanced HCC patients, thereby improving survival outcomes.
Collapse
Affiliation(s)
- Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital/PUMC/Chinese Academy of Medical Sciences, Beijing, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Guangming Li
- Department of Hepatobiliary Surgery, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xiaodong Wang
- Departments of Interventional Oncology, Peking University Cancer Hospital and Institute, Beijing, China
| | - Bo Chen
- Department of Radiation Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Wen Zhang
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Xu Che
- Department of Hepatobiliary and Pancreatic Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital and Shenzhen Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Shenzhen, China
| | - Zhen Huang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue Han
- Department of Interventional Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Liming Jiang
- Department of Diagnostic Imaging, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yongkun Sun
- Department of Medical Oncology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhengqiang Yang
- Department of Interventional Radiology, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jianguo Zhou
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yefan Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Zhenyu Zhu
- Department of Hepatology, Fifth Medical Center of Chinese PLA General Hospital/ Chinese PLA Medical School, Beijing, China
| | - Minshan Chen
- Department of Hepatobiliary Surgery, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Shuqun Cheng
- The Third Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University, Shanghai, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
26
|
Zhang XD, Zhang LY, Luo JL, Yu KH, Zhu KL. Neoadjuvant therapy: Dawn of reducing the high post-surgery recurrence rate of hepatocellular carcinoma. World J Gastrointest Surg 2025; 17:103740. [PMID: 40162404 PMCID: PMC11948117 DOI: 10.4240/wjgs.v17.i3.103740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/01/2025] [Accepted: 01/14/2025] [Indexed: 02/24/2025] Open
Abstract
The high postoperative recurrence rate remains a major challenge in the treatment of hepatocellular carcinoma (HCC) following resection. Increasing research has been delved into investigating the role of neoadjuvant therapy on the prognosis of resectable HCC. Recent trends in combination therapy with molecularly targeted agents and immune checkpoint inhibitors have significantly improved the efficacy of systemic antitumor treatments, yielding survival benefits exceeding 40%. Neoadjuvant therapy for HCC, whether based on systemic antitumor treatments, locoregional therapies, or their combination, has emerged as a promising research direction. However, there remains a matter of debate on neoadjuvant therapy. In this review, we summarize and discuss the research progress and challenges of neoadjuvant therapy for HCC over the past five years from the perspective of Chinese guidelines to provide new insights and future directions in this field.
Collapse
Affiliation(s)
- Xiao-Dong Zhang
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Lu-Yi Zhang
- Zhejiang Provincial Key Laboratory of Pathophysiology, Health Science Center, Ningbo University, Ningbo 315211, Zhejiang Province, China
| | - Jia-Liang Luo
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Ke-Heng Yu
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| | - Ke-Lei Zhu
- Department of Hepatopancreatobiliary Surgery, The Affiliated People's Hospital of Ningbo University, Ningbo 315040, Zhejiang Province, China
| |
Collapse
|
27
|
He H, Feng Z, Duan J, Deng W, Wu Z, He Y, Liang Q, Xie Y. Radiomic features at contrast-enhanced CT predict proliferative hepatocellular carcinoma and its prognosis after transarterial chemoembolization. Sci Rep 2025; 15:10533. [PMID: 40148399 PMCID: PMC11950328 DOI: 10.1038/s41598-025-94684-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/17/2025] [Indexed: 03/29/2025] Open
Abstract
Proliferative hepatocellular carcinoma (HCC) is an aggressive phenotype associated with unfavorable clinical outcomes. Predicting the preoperative subtype of HCC can aid in the development of individualized treatment. We retrospectively recruited 180 HCC patients who underwent hepatic resection and established a CT-based radiomics model for predicting proliferative HCCs. The evaluation of tumor response to transarterial chemoembolization therapy and progression-free survival (PFS) according to the radiomics model was further performed in internal (n = 54) and external (n = 80) outcome cohorts. In our study, 98 of 180 (54%) patients were confirmed to have proliferative HCCs. The radiomics model comprising 9 radiomic features and exhibited good performance for predicting proliferative HCCs. The nomogram integrated radiomics and serum α-fetoprotein level showed good calibration and discrimination in both the training cohort (AUC = 0.848) and the validation cohort (AUC = 0.825). Predicted proliferative HCCs (high radiomics scores) were associated with lower response rate (P < 0.05) and worse PFS (P < 0.05) compared to predicted non-proliferative HCCs in outcomes cohorts. We linked radiomics model to gene expression, unveiling that activated/immature B cells and tertiary lymphoid structures were downregulated in the high radiomics group. The proposed CT radiomics model exhibited good performance for identifying proliferative HCCs, which may facilitate clinical decision-making. Our findings suggest a potential correlation between proliferative HCC and immunosuppressive tumor microenvironment.
Collapse
Affiliation(s)
- Haifeng He
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China
- Department of PET-CT Center, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Zhichao Feng
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China
| | - Junhong Duan
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China
| | - Wenzhi Deng
- Department of Pathology, The Third Xinagya Hospital Central South University, Changsha, China
| | - Zuowei Wu
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China
| | - Yizi He
- Department of Lymphoma and Hematology, Hunan Cancer Hospital/The Affiliated Cancer Hospital of Xiangya School of Medicine, Central South University, Changsha, China
| | - Qi Liang
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China.
| | - Yongzhi Xie
- Department of Radiology, The Third Xinagya Hospital Central South University, Changsha, China.
| |
Collapse
|
28
|
WU BIAO, GUO XIANLIN, WU ZHISHI, CHEN LIANG, ZHANG SUQING. COPB2 promotes hepatocellular carcinoma progression through regulation of YAP1 nuclear translocation. Oncol Res 2025; 33:975-988. [PMID: 40191726 PMCID: PMC11964868 DOI: 10.32604/or.2025.058085] [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: 09/04/2024] [Accepted: 12/13/2024] [Indexed: 04/09/2025] Open
Abstract
Objectives Although Yes-associated protein 1 (YAP1) is an important oncogene in hepatocellular carcinoma (HCC) progression, its nuclear localization prevents it from being considered a potential therapeutic target. Recently, studies have reported that coatomer protein complex subunit beta 2 (COPB2) also plays a critical role in HCC development; however its mechanism of action is unclear. This study aimed to investigate the role of COPB2 and YAP1 in the progression of HCC and to elucidate the underlying mechanisms. Methods COPB2 and YAP1 expression in HCC tissues were first analyzed by database searches and immunohistochemistry. Nomogram and artificial neural network models were established based on COPB2 and YAP1 expression. Cell proliferation was detected by cell counting kit-8 and clone formation assay, while cell migration and invasion were assessed using Transwell assays. Finally, the potential mechanisms underlying COPB2 regulation of YAP1 nuclear translocation were explored by immunofluorescence assay and Western blot. Results COPB2 combined with YAP1 expression was associated with overall postoperative survival in HCC patients and was an independent prognostic factor. High expression of both COPB2 and YAP1 in patients may reduce the efficacy of postoperative transarterial chemoembolization therapy. In vitro experiments revealed that COPB2 affected the sensitivity of HCC cells to Cisplatin (DDP) by regulating YAP1 nuclear translocation. Conclusions Our findings suggest that COPB2/YAP1 affects the drug sensitivity of HCC cells to DDP and that targeting COPB2/YAP1 may be a promising strategy for the precision treatment of HCC.
Collapse
Affiliation(s)
- BIAO WU
- Department of General Surgery, Changhai Hospital, Second (Navy) Military Medical University, Shanghai, 200433, China
| | - XIANLIN GUO
- Department of General Surgery, Zhengzhou First People’s Hospital, Zhengzhou, 450000, China
| | - ZHISHI WU
- Department of General Surgery, Changhai Hospital, Second (Navy) Military Medical University, Shanghai, 200433, China
| | - LIANG CHEN
- Department of General Surgery, Changhai Hospital, Second (Navy) Military Medical University, Shanghai, 200433, China
| | - SUQING ZHANG
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Tumor Hospital of Nantong University, Nantong, 226361, China
| |
Collapse
|
29
|
Villagrán-Silva F, Loren P, Sandoval C, Lanas F, Salazar LA. Circulating microRNAs as Potential Biomarkers of Overweight and Obesity in Adults: A Narrative Review. Genes (Basel) 2025; 16:349. [PMID: 40149500 PMCID: PMC11942292 DOI: 10.3390/genes16030349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/29/2025] Open
Abstract
In an obesogenic environment, such as the one we have been experiencing in recent decades, epigenetics provides answers to the relationship between hereditary and environmentally acquired patterns that have significantly contributed to the global rise in obesity prevalence. MicroRNA (miRNA) constitutes a diminutive non-coding small RNA molecule, 20 to 24 nucleotides in length, that functions as a regulator of gene regulation at the post-translational level. Circulating miRNAs (c-miRNAs) have been detected in multiple body fluids, including blood, plasma, serum, saliva, milk from breastfeeding mothers, and urine. These molecules hold significant therapeutic value and serve as extracellular biomarkers in metabolic diseases. They aid in the diagnosis and tracking of therapy responses, as well as dietary and physical habit modifications. Researchers have studied c-miRNAs as potential biomarkers for diagnosing and characterizing systemic diseases in people of all ages and backgrounds since then. These conditions encompass dyslipidemia, type 2 diabetes mellitus (T2DM), cardiovascular risk, metabolic syndrome, cardiovascular diseases, and obesity. This review therefore analyzes the usefulness of c-miRNAs as therapeutic markers over the past decades. It also provides an update on c-miRNAs associated with general obesity and overweight, as well as with the most prevalent pathologies in the adult population. It also examines the effect of different nutritional approaches and physical activity regarding the activity of miRNAs in circulation in adults with overweight or general obesity. All of this is done with the aim of evaluating their potential use as biomarkers in various research contexts related to overweight and obesity in adults.
Collapse
Affiliation(s)
- Francisca Villagrán-Silva
- Doctoral Program in Morphological Sciences, Faculty of Medicine, Universidad de la Frontera, Temuco 4811230, Chile;
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Pía Loren
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| | - Cristian Sandoval
- Escuela de Tecnología Médica, Facultad de Salud, Universidad Santo Tomás, Los Carreras 753, Osorno 5310431, Chile;
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Fernando Lanas
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
- Department of Internal Medicine, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile
| | - Luis A. Salazar
- Center of Molecular Biology and Pharmacogenetics, Department of Basic Sciences, Faculty of Medicine, Universidad de La Frontera, Temuco 4811230, Chile; (P.L.); (F.L.)
| |
Collapse
|
30
|
Uzun M, Gokcek S, Kaya E, Semiz HS. The prognostic role of systemic immune-inflammation index, SII, in Metastatic Castration-Resistant Prostate Cancer patients. Discov Oncol 2025; 16:317. [PMID: 40085163 PMCID: PMC11908992 DOI: 10.1007/s12672-025-02084-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 03/06/2025] [Indexed: 03/16/2025] Open
Abstract
Our study aimed to examine the predictive relevance of the Systemic Immune-Inflammation Index (SII) in patients with metastatic castration-resistant prostate cancer (mCRPC). A total of 113 mCRPC patients were assessed. In this descriptive study, SII was calculated using the formula (neutrophil count × platelet count)/lymphocyte count. The optimal threshold for SII, determined via the ROC curve, was 700. Patients with SII ≤ 700 were classified as SII-low, while those with SII > 700 were categorized as SII-high. The median overall survival (mOS) was significantly longer in the low SII group compared to the high SII group (*P = 0.015). In multivariate analysis, Gleason score, albumin levels, CHAARTED volume, and SII were identified as significant prognostic factors. Our findings indicate that SII has a strong correlation with survival and can serve as an independent prognostic marker in mCRPC patients.
Collapse
Affiliation(s)
- Mehmet Uzun
- Department of Medical Oncology, Necip Fazıl City Hospital, Kahramanmaras, Türkiye.
| | - Savas Gokcek
- Department of Medical Oncology, Necip Fazıl City Hospital, Kahramanmaras, Türkiye
| | - Erhan Kaya
- Department of Public Health, Sütçü Imam University, Kahramanmaraş, Türkiye
| | | |
Collapse
|
31
|
Xue Y, Wang Y, Ren Z, Yu K. Tissue factor promotes TREX1 protein stability to evade cGAS-STING innate immune response in pancreatic ductal adenocarcinoma. Oncogene 2025; 44:739-752. [PMID: 39658648 PMCID: PMC11888988 DOI: 10.1038/s41388-024-03248-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/12/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains the most challenging human malignancy that urgently needs effective therapy. Tissue factor (TF) is expressed in ~80% of PDAC and represents a potential therapeutic target. While a novel TF-ADC (MRG004A) demonstrated efficacy for PDAC and TNBC in a Phase I/II trial [Ref. 18], the functional role of TF in PDAC remains incompletely understood. We investigated the relationship between TF and the innate STING pathway. We found that patients with TF-overexpression had poor survival, very low levels of P-STING/P-TBK1, reduced amounts of ISGs and chemokines as well as low numbers of cytotoxic immunocytes in their tumor. In experimental models of mouse and human PDAC, tumor cell-intrinsic TF expression played a major role in silencing the cytosolic micronuclei sensing and cGAS-STING activation. This process involved a TREX1 exonuclease-dependent clearance of micronucleus-DNA accumulated in tumor cells. Treatment of tumors with TF-KO/shRNA or anti-TF antibody HuSC1-39 (parent antibody of MRG004A) triggered a rapid and proteasome-dependent degradation of TREX1 thereby restoring the STING/TBK1 cascade phosphorylation. TF-inhibition therapy promoted a robust STING/IRF3-dependent IFN/CCL5/CXCL9-11 production, immune effector cell infiltration and antitumor efficacy. Moreover, in the PBMC and cancer cell co-culture, TF-inhibition synergized with a STING agonist compound. A covalently conjugated TF antibody-STING agonist ADC strongly increased the efficacy of tumor-targeted STING agonism on chemokine secretion and tumor inhibition in vitro and in vivo. Thus, TF-inhibition reshapes an "immune hot" tumor environment. TF-targeted therapy warrants clinical investigation as a single agent or in combination with immunotherapy for treating TF-positive PDAC and TNBC.
Collapse
Affiliation(s)
- Yinyin Xue
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Yue Wang
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Zhiqiang Ren
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China
| | - Ker Yu
- Department of Pharmacology, Fudan University School of Pharmacy, Shanghai, China.
| |
Collapse
|
32
|
Zhang Y, Li Z, Zhang J, Mafa T, Zhang J, Zhu H, Chen L, Zong Z, Yang L. Fibrinogen: A new player and target on the formation of pre-metastatic niche in tumor metastasis. Crit Rev Oncol Hematol 2025; 207:104625. [PMID: 39826884 DOI: 10.1016/j.critrevonc.2025.104625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 01/05/2025] [Accepted: 01/15/2025] [Indexed: 01/22/2025] Open
Abstract
Tumor metastasis involves a series of complex and coordinated processes, which is the main cause of patient death and still a significant challenge in cancer treatment. Pre-metastatic niches (PMN), a specialized microenvironment that develops in distant organs prior to the arrival of metastatic cancer cells, plays a crucial role in driving tumor metastasis. The development of PMN depends on a complex series of cellular and molecular components including tumor-derived factors, bone marrow-derived cells, resident immune cells, and extracellular matrix. Fibrinogen, a key factor in the typical blood clotting process, is related to tumor metastasis and prognosis, according to a growing body of evidence in recent years. Fibrinogen has emerged as an important factor in mediating the formation of tumor microenvironment. Nevertheless, a clear and detailed mechanism by which fibrinogen promotes tumor metastasis remains unknown. In this review, we first explore the roles of fibrinogen in the development of PMN from four perspectives: immunosuppression, inflammation, angiogenesis, and extracellular matrix remodeling. We highlight the significance of fibrinogen in shaping PMN and discuss its potential therapeutic values, opening new avenues for targeting fibrinogen to prevent or treat metastasis.
Collapse
Affiliation(s)
- Yuxin Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Zelin Li
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The First Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Jiamao Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; The Second Clinical Medical College, Nanchang University, No. 1299 Xuefu Ave, Nanchang, Jiangxi 330031, China
| | - Tatenda Mafa
- Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA
| | - Jingyu Zhang
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang, Jiangxi 330006, China
| | - Hui Zhu
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China
| | - Lifang Chen
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China
| | - Zhen Zong
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital of Nanchang University, No.1 MinDe Road, Nanchang, Jiangxi 330006, China
| | - Lingling Yang
- Department of Gastroenterology, The Second Affiliated Hospital of Nanchang University, No. 1 Minde Rd, Nanchang, Jiangxi 330006, China; Department of Molecular Biosciences, The University of Kansas, Lawrence, KS 66045, USA.
| |
Collapse
|
33
|
Xu W, Xu J, Liu J, Wang N, Zhou L, Guo J. Liver Metastasis in Cancer: Molecular Mechanisms and Management. MedComm (Beijing) 2025; 6:e70119. [PMID: 40027151 PMCID: PMC11868442 DOI: 10.1002/mco2.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/15/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Liver metastasis is a leading cause of mortality from malignant tumors and significantly impairs the efficacy of therapeutic interventions. In recent years, both preclinical and clinical research have made significant progress in understanding the molecular mechanisms and therapeutic strategies of liver metastasis. Metastatic tumor cells from different primary sites undergo highly similar biological processes, ultimately achieving ectopic colonization and growth in the liver. In this review, we begin by introducing the inherent metastatic-friendly features of the liver. We then explore the panorama of liver metastasis and conclude the three continuous, yet distinct phases based on the liver's response to metastasis. This includes metastatic sensing stage, metastatic stress stage, and metastasis support stage. We discuss the intricate interactions between metastatic tumor cells and various resident and recruited cells. In addition, we emphasize the critical role of spatial remodeling of immune cells in liver metastasis. Finally, we review the recent advancements and the challenges faced in the clinical management of liver metastasis. Future precise antimetastatic treatments should fully consider individual heterogeneity and implement different targeted interventions based on stages of liver metastasis.
Collapse
Affiliation(s)
- Wenchao Xu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Jia Xu
- State Key Laboratory of Fine ChemicalsDepartment of Pharmaceutical SciencesSchool of Chemical EngineeringDalian University of TechnologyDalianChina
| | - Jianzhou Liu
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Nanzhou Wang
- Department of Colorectal SurgeryState Key Laboratory of Oncology in South ChinaSun Yat‐sen University Cancer CenterGuangdong Provincial Clinical Research Center for CancerGuangzhouChina
| | - Li Zhou
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| | - Junchao Guo
- Department of General SurgeryPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
- Key Laboratory of Research in Pancreatic TumorChinese Academy of Medical SciencesBeijingChina
- National Infrastructures for Translational MedicinePeking Union Medical College HospitalBeijingChina
- State Key Laboratory of ComplexSevere, and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical Sciences and Peking Union Medical CollegeBeijingChina
| |
Collapse
|
34
|
Sun Y, Li T, Ding L, Wang J, Chen C, Liu T, Liu Y, Li Q, Wang C, Huo R, Wang H, Tian T, Zhang C, Pan B, Zhou J, Fan J, Yang X, Yang W, Wang B, Guo W. Platelet-mediated circulating tumor cell evasion from natural killer cell killing through immune checkpoint CD155-TIGIT. Hepatology 2025; 81:791-807. [PMID: 38779918 DOI: 10.1097/hep.0000000000000934] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Accepted: 04/23/2024] [Indexed: 05/25/2024]
Abstract
BACKGROUND AND AIMS Circulating tumor cells (CTCs) are precursors of cancer metastasis. However, how CTCs evade immunosurveillance during hematogenous dissemination remains unclear. APPROACH AND RESULTS We identified CTC-platelet adhesions by single-cell RNA sequencing and multiplex immunofluorescence of blood samples from multiple cancer types. Clinically, CTC-platelet aggregates were associated with significantly shorter progression-free survival and overall survival in patients with HCC. In vitro, ex vivo, and in vivo assays demonstrated direct platelet adhesions gifted cancer cells with an evasive ability from NK cell killing by upregulating inhibitory checkpoint CD155 (PVR cell adhesion molecule), therefore facilitating distant metastasis. Mechanistically, CD155 was transcriptionally regulated by the FAK/JNK/c-Jun cascade in a platelet contact-dependent manner. Further competition assays and cytotoxicity experiments revealed that CD155 on CTCs inhibited NK-cell cytotoxicity only by engaging with immune receptor TIGIT, but not CD96 and DNAM1, another 2 receptors for CD155. Interrupting the CD155-TIGIT interactions with a TIGIT antibody restored NK-cell immunosurveillance on CTCs and markedly attenuated tumor metastasis. CONCLUSIONS Our results demonstrated CTC evasion from NK-cell-mediated innate immunosurveillance mainly through immune checkpoint CD155-TIGIT, potentially offering an immunotherapeutic strategy for eradicating CTCs.
Collapse
Affiliation(s)
- Yunfan Sun
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Tong Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Lin Ding
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jiyan Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chen Chen
- Department of Cell Biology, Shanghai Dunwill Medical Technology Company, Shanghai, China
| | - Te Liu
- Shanghai Geriatric Institute of Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yu Liu
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Qian Li
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chuyu Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Ran Huo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hao Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Tongtong Tian
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Chunyan Zhang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
| | - Baishen Pan
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Jian Zhou
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Xinrong Yang
- Department of Liver Surgery & Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University; Key Laboratory of Carcinogenesis and Cancer Invasion, Ministry of Education, Shanghai, China
| | - Wenjing Yang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Beili Wang
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
| | - Wei Guo
- Department of Laboratory Medicine, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Wusong Branch, Zhongshan Hospital, Fudan University, Shanghai, China
- Department of Laboratory Medicine, Xiamen Branch, Zhongshan Hospital, Fudan University, Xiamen, China
- Department of Laboratory Medicine, Shanghai Geriatric Medical Center, Shanghai, China
- Cancer Center, Zhongshan Hospital, Fudan University, Shanghai, China
| |
Collapse
|
35
|
Xiong X, Guo JJ. Cost Effectiveness of Tremelimumab Plus Durvalumab for Unresectable Hepatocellular Carcinoma in the USA. PHARMACOECONOMICS 2025; 43:271-282. [PMID: 39546248 DOI: 10.1007/s40273-024-01453-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 10/29/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUND Treating unresectable hepatocellular carcinoma (uHCC) is challenging. Clinical trials have shown that Single Tremelimumab Regular Interval Durvalumab (STRIDE) offers clinical benefits as a first-line treatment for uHCC, but its cost effectiveness remains unknown in the USA. OBJECTIVE We aimed to assess the cost effectiveness of STRIDE (tremelimumab plus durvalumab) versus sorafenib and durvalumab monotherapy as the first-line treatment for uHCC in the USA. METHODS A partitioned survival model was constructed to assess the cost effectiveness of STRIDE compared to sorafenib and durvalumab monotherapy as the first-line treatment for uHCC from the US societal perspective. The time horizon was 48 months with 1-month cycles. Seven parametric survival functions replicated survival curves from clinical trials, with the best-fitting model used to calculate survival probabilities. Costs, health utilities, and adverse events were included, with quality-adjusted life-years (QALYs) as the primary effectiveness measure. Both costs and effectiveness were discounted at 3%. In the base-case analysis, the incremental cost-effectiveness ratio was compared to a willingness-to-pay threshold of $150,000 per QALY gained. Deterministic and probabilistic sensitivity analyses were conducted to examine the uncertainty of the model. RESULTS In the base-case analysis, STRIDE was cost effective compared to sorafenib, with an incremental cost-effectiveness ratio of $97,995.51 per QALY gained, based on a willingness-to-pay threshold of $150,000 per QALY gained. However, STRIDE was not cost effective compared to durvalumab monotherapy at the same threshold, with an incremental cost-effectiveness ratio of $754,408.92 per QALY gained. Deterministic sensitivity analyses were consistent with the base-case analysis. A probabilistic sensitivity analysis indicated that STRIDE was more likely to be cost effective than sorafenib and durvalumab monotherapy when the willingness-to-pay exceeded $101,000 and $713,000, respectively. CONCLUSIONS The STRIDE regimen appears to be cost effective compared to sorafenib but not compared to durvalumab for first-line uHCC treatment in the USA. However, durvalumab has not yet been approved for uHCC in the USA. Future research should focus on long-term data and economic evaluations of other recommended biologics.
Collapse
MESH Headings
- Humans
- Cost-Benefit Analysis
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/economics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Liver Neoplasms/drug therapy
- Liver Neoplasms/economics
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- United States
- Quality-Adjusted Life Years
- Antibodies, Monoclonal, Humanized/economics
- Antibodies, Monoclonal, Humanized/administration & dosage
- Sorafenib/economics
- Sorafenib/administration & dosage
- Antibodies, Monoclonal/administration & dosage
- Antibodies, Monoclonal/economics
- Antineoplastic Combined Chemotherapy Protocols/economics
- Antineoplastic Combined Chemotherapy Protocols/administration & dosage
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Antineoplastic Combined Chemotherapy Protocols/adverse effects
- Models, Economic
- Cost-Effectiveness Analysis
Collapse
Affiliation(s)
- Xiaomo Xiong
- Division of Pharmacy Practice and Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, 3255 Eden Ave, Cincinnati, OH, 45267, USA.
| | - Jeff Jianfei Guo
- Division of Pharmacy Practice and Administrative Sciences, James L. Winkle College of Pharmacy, University of Cincinnati Academic Health Center, 3255 Eden Ave, Cincinnati, OH, 45267, USA
| |
Collapse
|
36
|
Li W, Xu G, Chai GW, Ball A, Zhang Q, Kutryk MJB. The MiR-139-5p and CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration. Sci Rep 2025; 15:6738. [PMID: 40000897 PMCID: PMC11861593 DOI: 10.1038/s41598-025-91100-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Accepted: 02/18/2025] [Indexed: 02/27/2025] Open
Abstract
MicroRNAs, a class of small non-coding RNA molecules that regulate gene expression post-transcriptionally, are implicated in various pathological conditions including diabetes mellitus (DM). DM has been increasingly recognized as an inflammatory disease and monocytes play a key role in propagating inflammation under hyperglycemic conditions. We hypothesize that high glucose dysregulates microRNAs to promote monocyte inflammatory activity, which may contribute to the pathogenesis of DM. THP-1 monocytes were cultured in normal (5 mM) and high (25 mM) glucose conditions. RT-qPCR and Western blotting were performed to assay microRNAs and proteins, respectively. Monocytes were transfected with microRNA mimics using Lipofectamine RNAiMAX reagent. THP-1 monocyte growth was assessed using Calcein-AM dye and a Boyden chamber assay was applied to measure monocyte migration. The results showed that high glucose downregulated miR-139-5p associated with increased protein expression of CXCR4, an experimentally validated target of miR-139-5p. Correspondingly, treatment with high glucose resulted in a significant increase in THP-1 cell migration towards SDF-1, a cognate ligand for CXCR4. MiR-139-5p overexpression inhibited high glucose-induced CXCR4 expression, leading to reduced cell migration towards SDF-1. High glucose did not affect THP-1 monocyte growth. In conclusion, the miR-139-5p-CXCR4 axis may play a role in high glucose-induced inflammation by regulating monocyte migration.
Collapse
Affiliation(s)
- Weifang Li
- Department of Geriatric Endocrinology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gengchen Xu
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Gregory W Chai
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Alexander Ball
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada
| | - Qiuwang Zhang
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| | - Michael J B Kutryk
- Division of Cardiology, Keenan Research Center for Biomedical Science, St. Michael's Hospital, Unity Health Toronto, University of Toronto, Toronto, ON, Canada.
| |
Collapse
|
37
|
Tao C, Liu L, Hu N, Wang H, Zhang K, Liu Y, Wu F, Wang L, Rong W, Wu J. Effect of Narrow-Margin Hepatectomy Combined with Intraoperative Radiotherapy on Long-Term Prognosis of Patients with Centrally Located Hepatocellular Carcinoma: A Propensity Score Matching Analysis. J Hepatocell Carcinoma 2025; 12:261-274. [PMID: 39974613 PMCID: PMC11837753 DOI: 10.2147/jhc.s497998] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2024] [Accepted: 01/25/2025] [Indexed: 02/21/2025] Open
Abstract
Background Radiotherapy offers potential benefits for patients with hepatocellular carcinoma (HCC); however, the distinct role of intraoperative radiotherapy (IORT) during narrow-margin hepatectomy remains inadequately defined. This study aims at assessing the safety and effectiveness of IORT for centrally located HCCs during narrow-margin hepatectomy. Methods This single-center, retrospective research incorporated 659 patients with centrally located HCCs. After applying exclusion criteria, 607 patients remained and were divided into two groups: IORT integrated with liver resection (IORT+LR, 54 patients) and mere liver resection (LR, 553 patients). Propensity score matching (PSM) was performed to balance baseline characteristics. Post PSM, surgical outcomes, long-term recurrence, survival rates and adverse events were analyzed. Results A total of 54 patients were successfully matched, without significant differences upon baseline characteristics (standardized mean difference, SMD <0.15). Post-matching analysis revealed that overall survival (OS) and disease-free survival (DFS) were notably improved in the IORT+LR group (P =0.027 and 0.015, respectively). Multivariate Cox regression identified IORT as an independent prognostic factor for better DFS and OS. Among the 108 patients included after matching, 57 experienced HCC recurrence, 23 in the IORT group and 34 in the LR group, showing a clear difference in recurrence rates (P =0.034). Also, there were no apparent differences in mild/severe complications between IORT and RT groups (96.3% vs 98.2%, P =0.558, respectively). Conclusion IORT is an effective and well-tolerated therapy for HCC patients. The combination of narrow-margin hepatectomy and IORT enhances patient prognosis, with IORT identified as an independent prognostic factor.
Collapse
Affiliation(s)
- Changcheng Tao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Liguo Liu
- Second Department of Hepatopancreatobiliary Surgery, China-Japan Friendship Hospital, Beijing, 100029, People’s Republic of China
| | - Nan Hu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Hongwei Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Kai Zhang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Yue Liu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Fan Wu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Liming Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Weiqi Rong
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| | - Jianxiong Wu
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, 100021, People’s Republic of China
| |
Collapse
|
38
|
Li Z, Chen L, Wei Z, Liu H, Zhang L, Huang F, Wen X, Tian Y. A novel classification method for LUAD that guides personalized immunotherapy on the basis of the cross-talk of coagulation- and macrophage-related genes. Front Immunol 2025; 16:1518102. [PMID: 40018029 PMCID: PMC11866059 DOI: 10.3389/fimmu.2025.1518102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
Purpose The coagulation process and infiltration of macrophages affect the progression and prognosis of lung adenocarcinoma (LUAD) patients. This study was designed to explore novel classification methods that better guide the precise treatment of LUAD patients on the basis of coagulation and macrophages. Methods Weighted gene coexpression network analysis (WGCNA) was applied to identify M2 macrophage-related genes, and TAM marker genes were acquired through the analysis of scRNA-seq data. The MSigDB and KEGG databases were used to obtain coagulation-associated genes. The intersecting genes were defined as coagulation and macrophage-related (COMAR) genes. Unsupervised clustering analysis was used to evaluate distinct COMAR patterns for LUAD patients on the basis of the COMAR genes. The R package "limma" was used to identify differentially expressed genes (DEGs) between COMAR patterns. A prognostic risk score model, which was validated through external data cohorts and clinical samples, was constructed on the basis of the COMAR DEGs. Results In total, 33 COMAR genes were obtained, and three COMAR LUAD subtypes were identified on the basis of the 33 COMAR genes. There were 341 DEGs identified between the three COMAR subtypes, and 60 prognostic genes were selected for constructing the COMAR risk score model. Finally, 15 prognosis-associated genes (CORO1A, EPHA4, FOXM1, HLF, IFIH1, KYNU, LY6D, MUC16, PPARG, S100A8, SPINK1, SPINK5, SPP1, VSIG4, and XIST) were included in the model, which was efficient and robust in predicting LUAD patient prognosis and clinical outcomes in patients receiving anti-PD-1/PD-L1 immunotherapy. Conclusions LUAD can be classified into three subtypes according to COMAR genes, which may provide guidance for precise treatment.
Collapse
Affiliation(s)
- Zhuoqi Li
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Ling Chen
- Department of Oncology, Qingdao Municipal Hospital, Qingdao, China
| | - Zhigang Wei
- Department of Oncology, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Lung Cancer Institute, Jinan, China
| | - Hongtao Liu
- Department of Pathology, The First Affiliated Hospital of Shandong First Medical University and Shandong Provincial Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Clinical Pathology, Shandong Lung Cancer Institute, Shandong Institute of Nephrology, Jinan, China
| | - Lu Zhang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Fujing Huang
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Xiao Wen
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| | - Yuan Tian
- Department of Radiotherapy Oncology, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, China
| |
Collapse
|
39
|
Yu M, He W, Belsham DD. MicroRNA-34a-5p regulates agouti-related peptide via krüppel-like factor 4 and is disrupted by bisphenol A in hypothalamic neurons. Gene 2025; 937:149129. [PMID: 39617277 DOI: 10.1016/j.gene.2024.149129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 10/22/2024] [Accepted: 11/26/2024] [Indexed: 12/08/2024]
Abstract
Obesity is a complex disease marked by increased adiposity and impaired metabolic function. While diet and lifestyle are primary causes, endocrine-disrupting chemicals (EDCs), such as bisphenol A (BPA), significantly contribute to obesity. BPA, found in plastic consumer products, accumulates in the hypothalamus and dysregulates energy homeostasis by disrupting the neuropeptide Y (NPY)/agouti-related peptide (AgRP) and pro-opiomelanocortin (POMC) neurons. However, the precise molecular mechanisms of how BPA disrupts neuropeptide expression remains unclear. We hypothesized that microRNAs (miRNAs), which regulate approximately 60% of the human protein-coding genome and are crucial for hypothalamic energy regulation, may mediate the effects of BPA on Agrp. Using the TargetScanMouse 8.0 and DIANA microT bioinformatics tools, we identified miR-501-5p as a potential miRNA that directly regulates Agrp and the miR-34 family as miRNAs that indirectly regulate Agrp through its transcription factor krüppel-like factor 4 (KLF4). We found that in an immortalized NPY/AgRP-expressing cell line, mHypoE-41, miR-501-5p unexpectedly upregulated Agrp, while miR-34a-5p reduced Klf4 and Agrp mRNA levels. Serum starvation reduced miR-34a-5p levels and elevated Agrp mRNA levels, suggesting a potential role in AgRP regulation. Inhibiting the miR-34a-5p interaction with the Klf4 3'UTR using a specific target site blocker prevented the downregulation of both Klf4 and Agrp, suggesting miR-34a-5p alters Agrp mRNA levels via regulation of KLF4. BPA treatment increased Agrp and Klf4 expression while simultaneously decreasing miR-34a-5p levels, indicating miR-34a-5p may play a role in BPA-mediated dysregulation of Agrp. Overall, this study highlights indirect miRNA-based regulation of Agrp, which can also be dysregulated by obesogens, such as BPA.
Collapse
Affiliation(s)
- Minyi Yu
- Departments of Physiology, University of Toronto, Ontario, Canada
| | - Wenyuan He
- Departments of Physiology, University of Toronto, Ontario, Canada
| | - Denise D Belsham
- Departments of Physiology, University of Toronto, Ontario, Canada; Departments of Medicine, University of Toronto, Ontario, Canada.
| |
Collapse
|
40
|
Verona F, Di Bella S, Schirano R, Manfredi C, Angeloro F, Bozzari G, Todaro M, Giannini G, Stassi G, Veschi V. Cancer stem cells and tumor-associated macrophages as mates in tumor progression: mechanisms of crosstalk and advanced bioinformatic tools to dissect their phenotypes and interaction. Front Immunol 2025; 16:1529847. [PMID: 39981232 PMCID: PMC11839637 DOI: 10.3389/fimmu.2025.1529847] [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: 11/17/2024] [Accepted: 01/17/2025] [Indexed: 02/22/2025] Open
Abstract
Cancer stem cells (CSCs) are a small subset within the tumor mass significantly contributing to cancer progression through dysregulation of various oncogenic pathways, driving tumor growth, chemoresistance and metastasis formation. The aggressive behavior of CSCs is guided by several intracellular signaling pathways such as WNT, NF-kappa-B, NOTCH, Hedgehog, JAK-STAT, PI3K/AKT1/MTOR, TGF/SMAD, PPAR and MAPK kinases, as well as extracellular vesicles such as exosomes, and extracellular signaling molecules such as cytokines, chemokines, pro-angiogenetic and growth factors, which finely regulate CSC phenotype. In this scenario, tumor microenvironment (TME) is a key player in the establishment of a permissive tumor niche, where CSCs engage in intricate communications with diverse immune cells. The "oncogenic" immune cells are mainly represented by B and T lymphocytes, NK cells, and dendritic cells. Among immune cells, macrophages exhibit a more plastic and adaptable phenotype due to their different subpopulations, which are characterized by both immunosuppressive and inflammatory phenotypes. Specifically, tumor-associated macrophages (TAMs) create an immunosuppressive milieu through the production of a plethora of paracrine factors (IL-6, IL-12, TNF-alpha, TGF-beta, CCL1, CCL18) promoting the acquisition by CSCs of a stem-like, invasive and metastatic phenotype. TAMs have demonstrated the ability to communicate with CSCs via direct ligand/receptor (such as CD90/CD11b, LSECtin/BTN3A3, EPHA4/Ephrin) interaction. On the other hand, CSCs exhibited their capacity to influence immune cells, creating a favorable microenvironment for cancer progression. Interestingly, the bidirectional influence of CSCs and TME leads to an epigenetic reprogramming which sustains malignant transformation. Nowadays, the integration of biological and computational data obtained by cutting-edge technologies (single-cell RNA sequencing, spatial transcriptomics, trajectory analysis) has significantly improved the comprehension of the biunivocal multicellular dialogue, providing a comprehensive view of the heterogeneity and dynamics of CSCs, and uncovering alternative mechanisms of immune evasion and therapeutic resistance. Moreover, the combination of biology and computational data will lead to the development of innovative target therapies dampening CSC-TME interaction. Here, we aim to elucidate the most recent insights on CSCs biology and their complex interactions with TME immune cells, specifically TAMs, tracing an exhaustive scenario from the primary tumor to metastasis formation.
Collapse
Affiliation(s)
- Francesco Verona
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Sebastiano Di Bella
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Roberto Schirano
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Camilla Manfredi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| | - Francesca Angeloro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
| | - Giulia Bozzari
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Matilde Todaro
- Department of Health Promotion Sciences, Internal Medicine and Medical Specialties (PROMISE), University of Palermo, Palermo, Italy
- Azienda Ospedaliera Universitaria Policlinico “Paolo Giaccone” (AOUP), Palermo, Italy
| | - Giuseppe Giannini
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
- Istituto Pasteur, Fondazione Cenci-Bolognetti, Sapienza University of Rome, Rome, Italy
| | - Giorgio Stassi
- Department of Precision Medicine in Medical, Surgical and Critical Care, University of Palermo, Palermo, Italy
| | - Veronica Veschi
- Department of Molecular Medicine, University La Sapienza, Rome, Italy
| |
Collapse
|
41
|
Fu CL, Zhao ZW, Zhang QN. The crosstalk between cellular survival pressures and N6-methyladenosine modification in hepatocellular carcinoma. Hepatobiliary Pancreat Dis Int 2025; 24:67-75. [PMID: 39155161 DOI: 10.1016/j.hbpd.2024.08.004] [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: 04/18/2024] [Accepted: 08/09/2024] [Indexed: 08/20/2024]
Abstract
BACKGROUND Within the tumor microenvironment, survival pressures are prevalent with potent drivers of tumor progression, angiogenesis, and therapeutic resistance. N6-methyladenosine (m6A) methylation has been recognized as a critical post-transcriptional mechanism regulating various aspects of mRNA metabolism. Understanding the intricate interplay between survival pressures and m6A modification provides new insights into the molecular mechanisms underlying hepatocellular carcinoma (HCC) progression and highlights the potential for targeting the survival pressures-m6A axis in HCC diagnosis and treatment. DATA SOURCES A literature search was conducted in PubMed, MEDLINE, and Web of Science for relevant articles published up to April 2024. The keywords used for the search included hepatocellular carcinoma, cellular survival, survival pressure, N6-methyladenosine, tumor microenvironment, stress response, and hypoxia. RESULTS This review delves into the multifaceted roles of survival pressures and m6A RNA methylation in HCC, highlighting how survival pressures modulate the m6A landscape, the impact of m6A modification on survival pressure-responsive gene expression, and the consequent effects on HCC cell survival, proliferation, metastasis, and resistance to treatment. Furthermore, we explored the therapeutic potential of targeting this crosstalk, proposing strategies that leverage the understanding of survival pressures and m6A RNA methylation mechanisms to develop novel, and more effective treatments for HCC. CONCLUSIONS The interplay between survival pressures and m6A RNA methylation emerges as a complex regulatory network that influences HCC pathogenesis and progression.
Collapse
Affiliation(s)
- Chu-Li Fu
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou 510623, China
| | - Zheng-Wei Zhao
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China
| | - Qiang-Nu Zhang
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, the Third Affiliated Hospital of Guangzhou Medical University, Guangzhou 510150, China.
| |
Collapse
|
42
|
Zhang Q, Zhang Y, Fu C, He X, Huang Z, Wu G, Wei T, Jin W, Yan L, Wu M, Peng G, Fan L, Li M, Guo Y, Bi J, Bai Y, Roessler S, Yan GR, Liu L. CSTF2 Supports Hypoxia Tolerance in Hepatocellular Carcinoma by Enabling m6A Modification Evasion of PGK1 to Enhance Glycolysis. Cancer Res 2025; 85:515-534. [PMID: 39514400 DOI: 10.1158/0008-5472.can-24-2283] [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: 07/04/2024] [Revised: 08/24/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024]
Abstract
Cleavage stimulation factor subunit 2 (CSTF2) is a fundamental factor in the regulation of 3'-end cleavage and alternative polyadenylation of pre-mRNAs. Previous work has identified a tumor-promoting role of CSTF2, suggesting that it may represent a potential therapeutic target. In this study, we aimed to elucidate the mechanistic function of CSTF2 in hepatocellular carcinoma (HCC). CSTF2 upregulation was frequent in HCC, and elevated levels of CSTF2 correlated with poor patient prognosis. Although CSTF2 inhibition did not suppress HCC growth under nonstress conditions, it supported tolerance and survival of HCC cells under hypoxic conditions. Mechanistically, CSTF2 increased phosphoglycerate kinase 1 (PGK1) protein production to enhance glycolysis, thereby sustaining the energy supply under hypoxic conditions. CSTF2 shortened the 3' untranslated region of PGK1 pre-mRNA by binding near the proximal polyadenylation site. This shortening led to a loss of N6-methyladenosine (m6A) modification sites that are bound by YTH m6A RNA-binding protein F2 and increase degradation of PGK1 mRNA. Concurrently, hypoxia increased m6A modification of PGK1 mRNA near the proximal polyadenylation site that was recognized by the YTH m6A RNA-binding protein C1, which recruited CSTF2 to enhance the shortening of the PGK1 3' untranslated region. A small-molecule screen identified masitinib as an inhibitor of CSTF2. Masitinib counteracted PGK1 upregulation by CSTF2 and suppressed the growth of HCC xenograft and patient-derived organoid models. In conclusion, this study revealed a function of CSTF2 in supporting HCC survival under hypoxia conditions through m6A modification evasion and metabolic reprogramming, indicating that inhibiting CSTF2 may overcome hypoxia tolerance in HCC. Significance: Targeting CSTF2 inhibits hepatocellular carcinoma survival in hypoxic microenvironments, which may be a promising therapeutic strategy for treating liver cancer.
Collapse
Affiliation(s)
- Qiangnu Zhang
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Yusen Zhang
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Chuli Fu
- Department of Hematology and Oncology, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Xiaoyan He
- Department of Pathology, The Fifth Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zuotian Huang
- Department of Hepatobiliary and Pancreatic Tumor Center, Chongqing University Cancer Hospital, Chongqing, China
| | - Geyan Wu
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Teng Wei
- Cytotherapy Laboratory, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Wen Jin
- Department of Neurology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Lesen Yan
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Meilong Wu
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Gongze Peng
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - LinLan Fan
- Experimental and teaching Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China
| | - Mingyue Li
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yuehua Guo
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Jiangang Bi
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Yu Bai
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| | - Stephanie Roessler
- Institute of Pathology, Heidelberg University Hospital, Heidelberg, Germany
| | - Guang-Rong Yan
- Biomedicine Research Center, Guangdong Provincial Key Laboratory of Major Obstetric Disease, Guangdong Provincial Clinical Research Center for Obstetrics and Gynecology, The Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Liping Liu
- Division of Hepatobiliary and Pancreas Surgery, Department of General Surgery, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University, The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, China
| |
Collapse
|
43
|
Liu Q, Zhang R, Shen W. Advancements in locoregional therapy for advanced hepatocellular carcinoma: Emerging perspectives on combined treatment strategies. EUROPEAN JOURNAL OF SURGICAL ONCOLOGY 2025; 51:109502. [PMID: 39615292 DOI: 10.1016/j.ejso.2024.109502] [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: 09/20/2024] [Revised: 11/17/2024] [Accepted: 11/23/2024] [Indexed: 01/03/2025]
Abstract
Hepatocellular carcinoma (HCC) persists as a leading cause of cancer-related mortality, often diagnosed at advanced stages with limited treatment options. Locoregional therapies (LRTs) are crucial in HCC management, playing significant roles in neoadjuvant and palliative treatments, among others. However, the unique disease background of HCC necessitates multidisciplinary and integrated treatment strategies. The therapeutic landscape for advanced HCC has been significantly broadened by the advent of combined therapies, presenting multiple approaches aimed at improving long-term survival, which remains a critical challenge. This review offers a comprehensive overview of major LRTs for HCC, highlighting recent technological advancements and exploring the challenges and limitations in their application, and presents the latest developments in combination therapies, including combinations between different LRTs and their integration with systemic treatments. Additionally, we outline future directions for the development of integrated treatment modalities for advanced HCC.
Collapse
Affiliation(s)
- Qi Liu
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China; The Second Clinical Medical College of Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Renjie Zhang
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China; The Second Clinical Medical College of Harbin Medical University, Harbin, 150081, People's Republic of China
| | - Weixi Shen
- Department of Oncology, The Second Affiliated Hospital of Harbin Medical University, Harbin, 150001, People's Republic of China.
| |
Collapse
|
44
|
Cai H, Chen S, Tang S, Xiao Y, Shi F, Wu Z, Ma P, Chen H, Zhuang W, Guo W. Lenvatinib and tislelizumab versus atezolizumab and bevacizumab in combination with TAE-HAIC for unresectable hepatocellular carcinoma with high tumor burden: a multicenter retrospective cohort study. Cancer Immunol Immunother 2025; 74:88. [PMID: 39891746 PMCID: PMC11787109 DOI: 10.1007/s00262-025-03942-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Accepted: 01/02/2025] [Indexed: 02/03/2025]
Abstract
BACKGROUND Systemic and locoregional combination therapy has demonstrated promising outcomes for unresectable hepatocellular carcinoma (HCC); However, the best combination option remains unknown. This study compared the efficacy and safety of lenvatinib and tislelizumab versus atezolizumab and bevacizumab in combination with transarterial embolization (TAE) plus hepatic artery infusion chemotherapy (HAIC) for unresectable HCC with high tumor burden. METHODS This multicenter retrospective cohort study enrolled treatment-naive patients with unresectable HCC treated with TAE-HAIC plus lenvatinib and tislelizumab (THLP group) or TAE-HAIC plus atezolizumab and bevacizumab (THTA group). The primary endpoint was overall survival (OS). Secondary endpoints included progression-free survival (PFS), tumor response, and adverse events (AEs). Propensity score matching (PSM) was performed to reduce bias. RESULTS Of the 240 patients enrolled, 153 and 51 patients were assigned to the THLP and THTA groups, respectively after PSM (3:1). The THLP group showed a longer median OS (22 months vs. 18.2 months; P = 0.412), whereas the median PFS was longer in the THTA group (8.1 months vs. 7 months; P = 0.723), with statistically insignificant intergroup differences. No statistical differences were observed in objective response rate (RECIST 1.1: 33.9 vs. 31.4%; mRECIST: 77.1% vs. 74.5%; P = 0.635), disease control rate (RECIST 1.1: 88.9% vs. 92.2; mRECIST: 92.2% vs. 94.1%; P = 0.716), and in grade 3/4 AEs. Gastrointestinal hemorrhage rate was significantly higher in the THTA group (9.1% vs. 1.6%; P = 0.007). All AEs were controllable and no treatment-related grade 5 AEs occurred. CONCLUSIONS TAE-HAIC plus lenvatinib and tislelizumab or TAE-HAIC plus atezolizumab and bevacizumab showed similar outcomes for unresectable HCC with high tumor burden, and manageable safety. The results need further validation.
Collapse
MESH Headings
- Humans
- Male
- Female
- Liver Neoplasms/therapy
- Liver Neoplasms/pathology
- Liver Neoplasms/mortality
- Liver Neoplasms/drug therapy
- Retrospective Studies
- Carcinoma, Hepatocellular/therapy
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/mortality
- Carcinoma, Hepatocellular/drug therapy
- Antibodies, Monoclonal, Humanized/therapeutic use
- Antibodies, Monoclonal, Humanized/administration & dosage
- Antibodies, Monoclonal, Humanized/pharmacology
- Middle Aged
- Quinolines/therapeutic use
- Quinolines/administration & dosage
- Quinolines/pharmacology
- Phenylurea Compounds/therapeutic use
- Phenylurea Compounds/administration & dosage
- Phenylurea Compounds/pharmacology
- Bevacizumab/therapeutic use
- Bevacizumab/administration & dosage
- Bevacizumab/pharmacology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- Aged
- Tumor Burden
- Adult
Collapse
Affiliation(s)
- Hongjie Cai
- Department of Interventional Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510062, China
| | - Song Chen
- Department of Minimally Invasive Interventional Therapy, State Key Laboratory of Oncology in South China, Guangdong Provincial Clinical Research Center for Cancer, Sun Yat-Sen University Cancer Center, Guangzhou, 510060, People's Republic of China
| | - Shuangyan Tang
- Department of Radiology, The Eighth Affiliated Hospital of Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yi Xiao
- Center of Hepato-Pancreato-Biliary Surgery, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Feng Shi
- Department of Interventional Radiology, Guangdong Provincial People's Hospital, Guangzhou, 519041, China
| | - Zhiqiang Wu
- Department of Interventional Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510062, China
| | - Ping Ma
- Department of Oncology, The Twelfth People's Hospital of Guangzhou, Guangzhou, 510620, China
| | - Huanwei Chen
- Department of Hepatopancreatic Surgery, The First People's Hospital of Foshan, Foshan, 528010, China
| | - Wenquan Zhuang
- Department of Interventional Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510062, China
| | - Wenbo Guo
- Department of Interventional Radiology, The First Affiliated Hospital of Sun Yat-Sen University, Guangzhou, 510062, China.
| |
Collapse
|
45
|
McBrien C, O’Connell DJ. The Use of Biologics for Targeting GPCRs in Metastatic Cancers. BIOTECH 2025; 14:7. [PMID: 39982274 PMCID: PMC11843943 DOI: 10.3390/biotech14010007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/18/2025] [Accepted: 01/27/2025] [Indexed: 02/22/2025] Open
Abstract
A comprehensive review of studies describing the role of G-protein coupled receptor (GPCR) behaviour contributing to metastasis in cancer, and the developments of biotherapeutic drugs towards targeting them, provides a valuable resource toward improving our understanding of the opportunities to effectively target this malignant tumour cell adaptation. Focusing on the five most common metastatic cancers of lung, breast, colorectal, melanoma, and prostate cancer, we highlight well-studied and characterised GPCRs and some less studied receptors that are also implicated in the development of metastatic cancers. Of the approximately 390 GPCRs relevant to therapeutic targeting, as many as 125 of these have been identified to play a role in promoting metastatic disease in these cancer types. GPCR signalling through the well-characterised pathways of chemokine receptors, to emerging data on signalling by orphan receptors, is integral to many aspects of the metastatic phenotype. Despite having detailed information on many receptors and their ligands, there are only thirteen approved therapeutics specifically for metastatic cancer, of which three are small molecules with the remainder including synthetic and non-synthetic peptides or monoclonal antibodies. This review will cover the existing and potential use of monoclonal antibodies, proteins and peptides, and nanobodies in targeting GPCRs for metastatic cancer therapy.
Collapse
Affiliation(s)
| | - David J. O’Connell
- School of Biomolecular & Biomedical Science, University College Dublin, D04 V1W8 Dublin, Ireland;
| |
Collapse
|
46
|
Shou S, Maolan A, Zhang D, Jiang X, Liu F, Li Y, Zhang X, Geer E, Pu Z, Hua B, Guo Q, Zhang X, Pang B. Telomeres, telomerase, and cancer: mechanisms, biomarkers, and therapeutics. Exp Hematol Oncol 2025; 14:8. [PMID: 39871386 PMCID: PMC11771031 DOI: 10.1186/s40164-025-00597-9] [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: 01/08/2025] [Accepted: 01/15/2025] [Indexed: 01/29/2025] Open
Abstract
Telomeres and telomerase play crucial roles in the initiation and progression of cancer. As biomarkers, they aid in distinguishing benign from malignant tissues. Despite the promising therapeutic potential of targeting telomeres and telomerase for therapy, translating this concept from the laboratory to the clinic remains challenging. Many candidate drugs remain in the experimental stage, with only a few advancing to clinical trials. This review explores the relationship between telomeres, telomerase, and cancer, synthesizing their roles as biomarkers and reviewing the outcomes of completed trials. We propose that changes in telomere length and telomerase activity can be used to stratify cancer stages. Furthermore, we suggest that differential expression of telomere and telomerase components at the subcellular level holds promise as a biomarker. From a therapeutic standpoint, combining telomerase-targeted therapies with drugs that mitigate the adverse effects of telomerase inhibition may offer a viable strategy.
Collapse
Affiliation(s)
- Songting Shou
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Ayidana Maolan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Di Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiaochen Jiang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Fudong Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Yi Li
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Xiyuan Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - En Geer
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Zhenqing Pu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Baojin Hua
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Qiujun Guo
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Xing Zhang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Bo Pang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| |
Collapse
|
47
|
Tang H, Zhang W, Cao J, Cao Y, Bi X, Zhao H, Zhang Z, Liu Z, Wan T, Lang R, Sun W, Du S, Yang Y, Lu Y, Zeng D, Wu J, Duan B, Lin D, Li F, Meng Q, Zhou J, Xing B, Tian X, Zhu J, Gao J, Hao C, Wang Z, Duan F, Wang Z, Wang M, Liang B, Chen Y, Xu Y, Li K, Li C, Hu M, Wang Z, Cai S, Ji W, Xia N, Zheng W, Wang H, Li G, Zhu Z, Huang Z, Zhang W, Tao K, Liang J, Zhang K, Dai C, Li J, Qiu Q, Guo Y, Wu L, Ding W, Zhu Z, Gu W, Cao J, Wang Z, Tian L, Ding H, Li G, Zeng Y, Wang K, Yang N, Jin H, Chen Y, Yang Y, Xiu D, Yan M, Wang X, Han Q, Jiao S, Tan G, Wang J, Liu L, Song J, Liao J, Zhao H, Li P, Song T, Wang Z, Yuan J, Hu B, Yuan Y, Zhang M, Sun S, Zhang J, Wang W, Wen T, Yang J, Du X, Peng T, Xia F, Liu Z, Niu W, Liang P, Xu J, Zhao X, Zhu M, et alTang H, Zhang W, Cao J, Cao Y, Bi X, Zhao H, Zhang Z, Liu Z, Wan T, Lang R, Sun W, Du S, Yang Y, Lu Y, Zeng D, Wu J, Duan B, Lin D, Li F, Meng Q, Zhou J, Xing B, Tian X, Zhu J, Gao J, Hao C, Wang Z, Duan F, Wang Z, Wang M, Liang B, Chen Y, Xu Y, Li K, Li C, Hu M, Wang Z, Cai S, Ji W, Xia N, Zheng W, Wang H, Li G, Zhu Z, Huang Z, Zhang W, Tao K, Liang J, Zhang K, Dai C, Li J, Qiu Q, Guo Y, Wu L, Ding W, Zhu Z, Gu W, Cao J, Wang Z, Tian L, Ding H, Li G, Zeng Y, Wang K, Yang N, Jin H, Chen Y, Yang Y, Xiu D, Yan M, Wang X, Han Q, Jiao S, Tan G, Wang J, Liu L, Song J, Liao J, Zhao H, Li P, Song T, Wang Z, Yuan J, Hu B, Yuan Y, Zhang M, Sun S, Zhang J, Wang W, Wen T, Yang J, Du X, Peng T, Xia F, Liu Z, Niu W, Liang P, Xu J, Zhao X, Zhu M, Wang H, Kuang M, Shen S, Cui X, Zhou J, Liu R, Sun H, Fan J, Chen X, Zhou J, Cai J, Lu S. Chinese expert consensus on sequential surgery following conversion therapy based on combination of immune checkpoint inhibitors and antiangiogenic targeted drugs for advanced hepatocellular carcinoma (2024 edition). Biosci Trends 2025; 18:505-524. [PMID: 39721704 DOI: 10.5582/bst.2024.01394] [Show More Authors] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2024]
Abstract
Up to half of hepatocellular carcinoma (HCC) cases are diagnosed at an advanced stage, for which effective treatment options are lacking, resulting in a poor prognosis. Over the past few years, the combination of immune checkpoint inhibitors and anti-angiogenic targeted therapy has proven highly efficacious in treating advanced HCC, significantly extending patients' survival and providing a potential for sequential curative surgery. After sequential curative hepatectomy or liver transplantation following conversion therapy, patients can receive long-term survival benefits. In order to improve the long-term survival rate of the overall population with liver cancer and achieve the goal of a 15% increase in the overall 5-year survival rate outlined in the Healthy China 2030 blueprint, the Professional Committee for Prevention and Control of Hepatobiliary and Pancreatic Diseases of Chinese Preventive Medicine Association, Chinese Society of Liver Cancer, and the Liver Study Group of Surgery Committee of Beijing Medical Association organized in-depth discussions among relevant domestic experts in the field. These discussions focused on the latest progress since the release of the Chinese expert consensus on conversion therapy of immune checkpoint inhibitors combined antiangiogenic targeted drugs for advanced hepatocellular carcinoma (2021 Edition) and resulted in a new consensus on the modifications and supplements to related key points. This consensus aims to further guide clinical practice, standardize medical care, and promote the development of the discipline.
Collapse
Affiliation(s)
- Haowen Tang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Wenwen Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Junning Cao
- Organ Transplantation Center, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Yinbiao Cao
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Haitao Zhao
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Ze Zhang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Zhe Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Tao Wan
- Faculty of Hepato-Pancreato-Biliary Surgery, the Eighth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Ren Lang
- Department of Hepatobiliary Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing, China
| | - Wenbing Sun
- Department of Hepatobiliary Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing, China
| | - Shunda Du
- Department of Liver Surgery, Peking Union Medical College (PUMC) Hospital, PUMC & Chinese Academy of Medical Sciences, Beijing, China
| | - Yongping Yang
- Senior Department of Hepatology, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Yinying Lu
- Comprehensive Liver Cancer Center, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Daobing Zeng
- Department of General Surgery Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jushan Wu
- Department of General Surgery Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Binwei Duan
- Department of General Surgery Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Dongdong Lin
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Fei Li
- Department of General Surgery, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Qinghua Meng
- Department of Medical Oncology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Jun Zhou
- Department of Gastrointestinal Oncology, Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Peking University Cancer Hospital & Institute, Beijing, China
| | - Baocai Xing
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Hepatopancreatobiliary Surgery Department I, Peking University Cancer Hospital & Institute, Beijing, China
| | - Xiaodong Tian
- Department of General Surgery, Peking University First Hospital, Beijing, China
| | - Jiye Zhu
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Jie Gao
- Department of Hepatobiliary Surgery, Peking University People's Hospital, Beijing, China
| | - Chunyi Hao
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Sarcoma Center, Peking University Cancer Hospital & Institute, Beijing, China
| | - Zhiqiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Qinghai Red Cross Hospital, Qinghai, China
| | - Feng Duan
- Department of Interventional Radiology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Zhijun Wang
- Department of Interventional Radiology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Maoqiang Wang
- Department of Interventional Radiology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Bin Liang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Yongwei Chen
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Yinzhe Xu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Kai Li
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Chengang Li
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Minggen Hu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Zhaohai Wang
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Shouwang Cai
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Wenbin Ji
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Nianxin Xia
- Faculty of Hepato-Pancreato-Biliary Surgery, the Sixth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Wenheng Zheng
- Department of Interventional Therapy, Cancer Hospital of China Medical University, Liaoning Cancer Hospital & Institute, Liaoning, China
| | - Hongguang Wang
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Gong Li
- Department of Radiation Oncology, Beijing Tsinghua Changgung Hospital, Tsinghua University, Beijing, China
| | - Ziman Zhu
- Faculty of Hepato-Pancreato-Biliary Surgery, the Fourth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Zhiyong Huang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Wanguang Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Kaishan Tao
- Department of Hepatobiliary Surgery, Xijing Hosptial, the Fourth Military Medical University, Shanxi, China
| | - Jun Liang
- Department of Medical Oncology, Peking University International Hospital, Beijing, China
| | - Keming Zhang
- Department of Hepatobiliary Surgery, Peking University International Hospital, Beijing, China
| | - Chaoliu Dai
- Department of General Surgery, Shengjing Hospital of China Medical University, Liaoning, China
| | - Jiangtao Li
- Department of Surgery, The Second Affiliated Hospital, Zhejiang University School of Medicine, Zhejiang, China
| | - Qiu Qiu
- Department of Gastroenterology, People's Hospital of Chongqing Hechuan, Chongqing, China
| | - Yuan Guo
- Organ Transplantation Center, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Liqun Wu
- Organ Transplantation Center, the Affiliated Hospital of Qingdao University, Shandong, China
| | - Weibao Ding
- Department of Hepatobiliary Surgery, Weifang People's Hospital, Shandong, China
| | - Zhenyu Zhu
- Hepatobiliary Surgery Center, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Wanqing Gu
- Chinese Journal of Hepatobiliary Surgery, Beijing, China
| | - Jingyu Cao
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong China
| | - Zusen Wang
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong China
| | - Lantian Tian
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Hospital of Qingdao University, Shandong China
| | - Huiguo Ding
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Guangming Li
- Department of Liver Transplantation Center, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yongyi Zeng
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fujian, China
| | - Kui Wang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Ning Yang
- Department of Hepatic Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, China
| | - Haosheng Jin
- Department of General Surgery, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangdong, China
| | - Yajin Chen
- Department of Hepatobiliopancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangdong, China
| | - Yinmo Yang
- Department of Hepatobiliary and Pancreatic Surgery, Peking University First Hospital, Beijing, China
| | - Dianrong Xiu
- Department of General Surgery, Peking University Third Hospital, Beijing, China
| | - Maolin Yan
- Department of Hepatobiliary and Pancreatic Surgery, Fujian Provincial Hospital, Fujian, China
| | - Xiaodong Wang
- Key Laboratory of Carcinogenesis and Translational Research (Ministry of Education), Department of Interventional Therapy, Peking UniversityCancer Hospital & Institute, Beijing, China
| | - Quanli Han
- Department of Medical Oncology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Shunchang Jiao
- Department of Medical Oncology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Guang Tan
- Department of General Surgery, the First Affiliated Hospital of Dalian Medical University, Liaoning, China
| | - Jizhou Wang
- Department of Hepatobiliary Surgery and Organ Transplantation Center, The First Affiliated Hospital of USTC, Division of Life Science andMedicine, University of Science and Technology of China, Anhui, China
| | - Lianxin Liu
- Department of Hepatobiliary Surgery and Organ Transplantation Center, The First Affiliated Hospital of USTC, Division of Life Science andMedicine, University of Science and Technology of China, Anhui, China
| | - Jinghai Song
- Department of General Surgery, Beijing Hospital, National Center of Gerontology; Institute of Geriatric Medicine, Chinese Academy of MedicalSciences, Beijing, China
| | - Jiajie Liao
- Zhongshan Hospital, Fudan University, Shanghai, China
| | - Hong Zhao
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Peng Li
- The Department of Hepatobiliary Surgery, the Second Affiliated Hospital of Hainan Medical University, Hainan, China
| | - Tianqiang Song
- Department of Hepatobiliary Cancer, Liver Cancer Center, Tianjin Medical University Cancer Institute & Hospital, National Clinical ResearchCenter for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin's Clinical Research Center for Cancer, Tianjin Key Laboratory ofDigestive Cancer, Tianjin, China
| | - Zhanbo Wang
- Department of Pathology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jing Yuan
- Department of Pathology, the First Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Bingyang Hu
- Department of General Surgery, Beijing Shijingshan Hospital, Beijing, China
| | - Yufeng Yuan
- Department of Hepatobiliary and Pancreatic Surgery, Zhongnan Hospital of Wuhan University, Hubei, China
| | - Meng Zhang
- Department of Hepatobiliary Surgery, the Fourth Hospital of Hebei Medical University, Hebei, China
| | - Shuyang Sun
- Department of Gastroenterology, Beijing Chest Hospital, Capital Medical University, Beijing, China
| | - Jialin Zhang
- Department of Radiology, the First Hospital of China Medical University, Liaoning, China
| | - Wentao Wang
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan, China
| | - Tianfu Wen
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan, China
| | - Jiayin Yang
- Department of Liver Surgery, West China Hospital, Sichuan University, Sichuan, China
| | - Xilin Du
- Department of General Surgery, Tangdu Hospital, the Fourth Military Medical University, Shanxi, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, the First Affiliated Hospital of Guangxi Medical University, Guangxi, China
| | - Feng Xia
- Institute of Hepatobiliary Surgery, Southwest Hospital, Army Medical University, Chongqing, China
| | - Zuojin Liu
- Department of Hepatobiliary Surgery, the Second Affiliated Hospital, Chongqing Medical University, Chongqing, China
| | - Weibo Niu
- Department of Hepatobiliary Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Ping Liang
- Department of Interventional Ultrasound, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Jianming Xu
- Department of Gastrointestinal Oncology, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing, China
| | - Xiao Zhao
- Department of Immunology and National Center for Biomedicine Analysis, the Fifth Medical Center of the Chinese PLA General Hospital, Beijing,China
| | - Min Zhu
- Department of Transplant Surgery, Qilu Hospital of Shandong University, Shandong, China
| | - Huaizhi Wang
- Institute of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing University, Chongqing, China
| | - Ming Kuang
- Center of Hepato Pancreato Biliary Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Shunli Shen
- Center of Hepato Pancreato Biliary Surgery, the First Affiliated Hospital, Sun Yat-sen University, Guangdong, China
| | - Xing Cui
- Department of Oncology and Hematology, the Second Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Shandong,China
| | - Jinxue Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the Affiliated Cancer Hospital of Zhengzhou University & Henan Cancer Hospital, Henan,China
| | - Rong Liu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| | - Huichuan Sun
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry ofEducation, Shanghai Key Laboratory of Organ Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Zhongshan Hospital,Fudan University, Shanghai, China
| | - Jia Fan
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry ofEducation, Shanghai Key Laboratory of Organ Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Zhongshan Hospital,Fudan University, Shanghai, China
| | - Xiaoping Chen
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - Jian Zhou
- Department of Liver Surgery and Transplantation, Key Laboratory of Carcinogenesis and Cancer Invasion (Fudan University), Ministry ofEducation, Shanghai Key Laboratory of Organ Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, Zhongshan Hospital,Fudan University, Shanghai, China
| | - Jianqiang Cai
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Shichun Lu
- Faculty of Hepato-Pancreato-Biliary Surgery, the First Medical Center of the Chinese PLA General Hospital, Institute of Hepatobiliary Surgery of the Chinese PLA, Beijing, China
| |
Collapse
|
48
|
Glaviano A, Lau HSH, Carter LM, Lee EHC, Lam HY, Okina E, Tan DJJ, Tan W, Ang HL, Carbone D, Yee MYH, Shanmugam MK, Huang XZ, Sethi G, Tan TZ, Lim LHK, Huang RYJ, Ungefroren H, Giovannetti E, Tang DG, Bruno TC, Luo P, Andersen MH, Qian BZ, Ishihara J, Radisky DC, Elias S, Yadav S, Kim M, Robert C, Diana P, Schalper KA, Shi T, Merghoub T, Krebs S, Kusumbe AP, Davids MS, Brown JR, Kumar AP. Harnessing the tumor microenvironment: targeted cancer therapies through modulation of epithelial-mesenchymal transition. J Hematol Oncol 2025; 18:6. [PMID: 39806516 PMCID: PMC11733683 DOI: 10.1186/s13045-024-01634-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2024] [Accepted: 11/11/2024] [Indexed: 01/16/2025] Open
Abstract
The tumor microenvironment (TME) is integral to cancer progression, impacting metastasis and treatment response. It consists of diverse cell types, extracellular matrix components, and signaling molecules that interact to promote tumor growth and therapeutic resistance. Elucidating the intricate interactions between cancer cells and the TME is crucial in understanding cancer progression and therapeutic challenges. A critical process induced by TME signaling is the epithelial-mesenchymal transition (EMT), wherein epithelial cells acquire mesenchymal traits, which enhance their motility and invasiveness and promote metastasis and cancer progression. By targeting various components of the TME, novel investigational strategies aim to disrupt the TME's contribution to the EMT, thereby improving treatment efficacy, addressing therapeutic resistance, and offering a nuanced approach to cancer therapy. This review scrutinizes the key players in the TME and the TME's contribution to the EMT, emphasizing avenues to therapeutically disrupt the interactions between the various TME components. Moreover, the article discusses the TME's implications for resistance mechanisms and highlights the current therapeutic strategies toward TME modulation along with potential caveats.
Collapse
Affiliation(s)
- Antonino Glaviano
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Hannah Si-Hui Lau
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117597, Singapore
| | - Lukas M Carter
- Department of Medical Physics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - E Hui Clarissa Lee
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Elena Okina
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Donavan Jia Jie Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Wency Tan
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
- School of Chemical and Life Sciences, Singapore Polytechnic, Singapore, 139651, Singapore
| | - Hui Li Ang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Daniela Carbone
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Michelle Yi-Hui Yee
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
| | - Muthu K Shanmugam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Xiao Zi Huang
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Tuan Zea Tan
- Cancer Science Institute of Singapore, National University of Singapore, Singapore, 117599, Singapore
| | - Lina H K Lim
- Division of Cellular and Molecular Research, Humphrey Oei Institute of Cancer Research, National Cancer Centre Singapore, Singapore, 169610, Singapore
- Immunology Program, Life Sciences Institute, National University of Singapore, Singapore, 117456, Singapore
- Immunology Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore
| | - Ruby Yun-Ju Huang
- School of Medicine and Graduate Institute of Oncology, College of Medicine, National Taiwan University, Taipei, 10051, Taiwan
- Department of Obstetrics & Gynaecology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117456, Singapore
| | - Hendrik Ungefroren
- First Department of Medicine, University Hospital Schleswig-Holstein (UKSH), Campus Lübeck, 23538, Lübeck, Germany
| | - Elisa Giovannetti
- Department of Medical Oncology, Cancer Center Amsterdam, UMC, Vrije Universiteit, HV Amsterdam, 1081, Amsterdam, The Netherlands
- Cancer Pharmacology Lab, Fondazione Pisana Per La Scienza, 56017, San Giuliano, Italy
| | - Dean G Tang
- Department of Pharmacology and Therapeutics, Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
- Experimental Therapeutics (ET) Graduate Program, University at Buffalo & Roswell Park Comprehensive Cancer Center, Buffalo, NY, 14263, USA
| | - Tullia C Bruno
- Department of Immunology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, USA
| | - Peng Luo
- Department of Oncology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Mads Hald Andersen
- National Center for Cancer Immune Therapy, Department of Oncology, Herlev and Gentofte Hospital, Herlev, Denmark
| | - Bin-Zhi Qian
- Fudan University Shanghai Cancer Center, Department of Oncology, Shanghai Medical College, The Human Phenome Institute, Zhangjiang-Fudan International Innovation Center, Fudan University, Shanghai, China
| | - Jun Ishihara
- Department of Bioengineering, Imperial College London, London, W12 0BZ, UK
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Salem Elias
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Saurabh Yadav
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Minah Kim
- Herbert Irving Comprehensive Cancer Center, Columbia University, New York, NY, USA
| | - Caroline Robert
- Department of Cancer Medicine, Inserm U981, Gustave Roussy Cancer Center, Université Paris-Saclay, Villejuif, France
- Faculty of Medicine, University Paris-Saclay, Kremlin Bicêtre, Paris, France
| | - Patrizia Diana
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies, University of Palermo, 90123, Palermo, Italy
| | - Kurt A Schalper
- Department of Pathology, Yale School of Medicine, Yale University, New Haven, CT, USA
| | - Tao Shi
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
| | - Taha Merghoub
- Swim Across America and Ludwig Collaborative Laboratory, Department of Pharmacology, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Department of Medicine, Parker Institute for Cancer Immunotherapy, Weill Cornell Medicine, New York, NY, USA
| | - Simone Krebs
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Anjali P Kusumbe
- Tissue and Tumor Microenvironment Group, MRC Human Immunology Unit, MRC Weatherall Institute of Molecular Medicine, University of Oxford, Oxford, OX3 9DS, UK
| | - Matthew S Davids
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Jennifer R Brown
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA, USA
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 117600, Singapore.
- NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, 119228, Singapore.
| |
Collapse
|
49
|
Al-Mohannadi A, Yahia RM, Bibawi H, Lachica CA, Ahmed W, Pavlovski I, Gentilcore G, Elgaali EE, Ejaz A, Ahmed A, Elanbari M, Awada Z, Al-Kubaisi MJ, Elnaggar M, Saleh A, Cugno C, Deola S. Flow Cytometry Evaluation of Blood-Cell-Bound Surface FVIII in Hemophilia A and Thrombosis. Cells 2025; 14:73. [PMID: 39851501 PMCID: PMC11764290 DOI: 10.3390/cells14020073] [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: 11/20/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/26/2025] Open
Abstract
Hemophilia A (HA) is associated with FVIII coagulation insufficiency or inactivity leading to excessive bleeding. Elevated FVIII, on the contrary, is associated with thrombophilia, thrombosis, myocardial infarctions, and stroke. Active FVIII (aFVIII) uses its C2 domain to bind to blood cells' membranes, consequently carrying out its coagulative function. We developed a reliable flow cytometry (FC) method for FVIII detection that can be utilized for assessing surface-bound FVIII on leukocytes in different coagulation/clinical states; we analyzed 49 pediatric subjects, encompassing patients with HA, other coagulopathies, venous thrombosis, and normal coagulation. Interestingly, the total leukocyte surface FVIII showed a declining trend across thrombosis, normal, and hypo-coagulation states. As expected, the leukocytes of HA patients displayed significantly lower levels of cellular-surface FVIII in comparison to patients with thrombosis. However, no significant correlation was observed between circulating levels of FVIII in plasma and the levels of FVIII bound to leukocytes, indicating that the differences in FVIII surface binding are not directly proportional to the availability of FVIII in the circulation and suggesting a specific binding mechanism governing the interaction between FVIII and leukocytes. Intriguingly, when analyzing the distinct blood subpopulations, we observed that surface FVIII levels were significantly elevated in classical monocytes of thrombosis patients compared to HA patients, healthy controls, and patients with other coagulopathies. Our study highlights the reliability of our FC platform in assessing FVIII abundance on leukocytes' membranes across coagulation states. Monocytes, particularly in cases of thrombosis, exhibit active binding of FVIII on their surface, suggesting a potential role in the pathophysiology of thrombosis that requires further investigation.
Collapse
Affiliation(s)
- Anjud Al-Mohannadi
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
- College of Health and Life Sciences, Hamad Bin Khalifa University, Doha P.O. Box 34110, Qatar
| | | | - Hani Bibawi
- Pathology Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Che-Ann Lachica
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Watfa Ahmed
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Igor Pavlovski
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | | | | | - Anila Ejaz
- Hematology-Oncology Clinic, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Areeg Ahmed
- Hematology-Oncology Clinic, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | | | - Zainab Awada
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | | | | | - Ayman Saleh
- Hematology-Oncology Clinic, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Chiara Cugno
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
- Pathology Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| | - Sara Deola
- Research Department, Sidra Medicine, Doha P.O. Box 26999, Qatar
| |
Collapse
|
50
|
Wang H, Zhang W, Li L, Wang H, Jiang H, Li W, Huang J, Wan Y. Revealing the active ingredients and mechanisms of Xiatianwu against hepatocellular carcinoma: a study based on network pharmacology and bioinformatics. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025; 398:729-746. [PMID: 39052060 DOI: 10.1007/s00210-024-03278-2] [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: 05/26/2024] [Accepted: 07/02/2024] [Indexed: 07/27/2024]
Abstract
Xiatianwu is a traditional Chinese medicine. This study investigates the function of Xiatianwu in treating HCC through database analyses and in vitro experiments. The active ingredients of Xiatianwu were identified from TCMSP and HERB databases and their targets were predicted by Swiss TargetPrediction. The HCC dataset was screened using the GEO database, and the differentially expressed genes between HCC and non-tumor liver tissues were analyzed to identify overlapping targets with Xiatianwu. The intersecting targets underwent enrichment analysis using R software to elucidate the molecular mechanisms of Xiatianwu against HCC. Core targets were identified using the PPI network and MCODE algorithm. Clinical relevance and disease prognosis in HCC were verified using the TCGA database. Meanwhile, binding affinities among components and targets were validated with molecular docking. Finally, the anti-HCC efficacy of the active ingredient was validated in vitro. Our findings revealed that eight active ingredients of Xiatianwu interacted with 11 key targets, providing anti-HCC efficacy. Molecular docking indicated that bicuculline and fumarine exhibited superior binding abilities. Bicuculline, a representative ingredient of Xiatianwu, was chosen for in vitro validation. Results demonstrated that bicuculline, in a dose-dependent manner inhibited HCC cell viability, reduced migration, suppressed the G0/M cell cycle, and decreased core protein expression. Xiatianwu demonstrates significant potential for clinical application in treating HCC. Bicuculline, a key active ingredient of Xiatianwu, exerts anti-HCC effects by inhibiting the cell cycle.
Collapse
Affiliation(s)
- Hui Wang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Weina Zhang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Liling Li
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Hong Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Honglin Jiang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Wenna Li
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China
| | - Jinchang Huang
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| | - Yuxiang Wan
- Beijing University of Chinese Medicine Third Affiliated Hospital, Beijing, 100029, China.
| |
Collapse
|