1
|
Ma J, Wang S, Zhang P, Zheng S, Li X, Li J, Pei H. Emerging roles for fatty acid oxidation in cancer. Genes Dis 2025; 12:101491. [PMID: 40290117 PMCID: PMC12022645 DOI: 10.1016/j.gendis.2024.101491] [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: 06/22/2024] [Accepted: 11/09/2024] [Indexed: 04/30/2025] Open
Abstract
Fatty acid oxidation (FAO) denotes the mitochondrial aerobic process responsible for breaking down fatty acids (FAs) into acetyl-CoA units. This process holds a central position in the cancer metabolic landscape, with certain tumor cells relying primarily on FAO for energy production. Over the past decade, mounting evidence has underscored the critical role of FAO in various cellular processes such as cell growth, epigenetic modifications, tissue-immune homeostasis, cell signal transduction, and more. FAO is tightly regulated by multiple evolutionarily conserved mechanisms, and any dysregulation can predispose to cancer development. In this view, we summarize recent findings to provide an updated understanding of the multifaceted roles of FAO in tumor development, metastasis, and the response to cancer therapy. Additionally, we explore the regulatory mechanisms of FAO, laying the groundwork for potential therapeutic interventions targeting FAO in cancers within the metabolic landscape.
Collapse
Affiliation(s)
- Jialin Ma
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| | - Shuxian Wang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Pingfeng Zhang
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Sihao Zheng
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Xiangpan Li
- Cancer Center, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Juanjuan Li
- Department of Breast and Thyroid Surgery, Renmin Hospital of Wuhan University, Wuhan, Hubei 430060, China
| | - Huadong Pei
- Department of Oncology, Georgetown Lombardi Comprehensive Cancer Center, Georgetown University Medical Center, Washington, DC 20057, USA
| |
Collapse
|
2
|
Yu X, Wu H, Wu Z, Lan Y, Chen W, Wu B, Deng Y, Liu J. Nuclear pore complex protein RANBP2 and related SUMOylation in solid malignancies. Genes Dis 2025; 12:101407. [PMID: 40271196 PMCID: PMC12017851 DOI: 10.1016/j.gendis.2024.101407] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Revised: 04/28/2024] [Accepted: 06/21/2024] [Indexed: 04/25/2025] Open
Abstract
The growing interest in post-translational protein modification, particularly in SUMOylation, is driven by its crucial role in cell cycle regulation. SUMOylation affects various cell cycle regulators, including oncogenes, suggesting its relevance in cancer. SUMO E3 ligases are pivotal in this process, exhibiting diverse functionalities through structural domains and subcellular localizations. A less-explored SUMO E3 ligase, RANBP2, a component of the vertebrate nuclear pore complex, emerges as a central player in cellular cycle processes, as well as in tumorigenesis. The current studies illuminate the importance of RANBP2 and underscore the need for more extensive studies to validate its clinical applicability in neoplastic interventions. Our review elucidates the significance of RANBP2 across various types of malignancies. Additionally, it delves into exploring RANBP2 as a prospective therapeutic target for cancer treatment, offering insights into the avenues that scholars should pursue in their subsequent research endeavors. Thus, further investigation into RANBP2's role in solid tumorigenesis is eagerly awaited.
Collapse
Affiliation(s)
- Xinning Yu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Huatao Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Zheng Wu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yangzheng Lan
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Wenjia Chen
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Bingxuan Wu
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Yu Deng
- Department of General Surgery, The First Affiliated Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
| | - Jing Liu
- The Breast Center, Cancer Hospital of Shantou University Medical College, Shantou, Guangdong 515041, China
- Department of Physiology, Shantou University Medical College, Shantou, Guangdong 515041, China
| |
Collapse
|
3
|
Schnabl SD, Klubien J, O'Rourke CJ, Bull Nordkild S, Kugler JM, Dam Nielsen S, Andersen JB, Pommergaard HC. Validation of Two Prognostic Gene Scores in Patients Undergoing Liver Resection for Hepatocellular Carcinoma. J Clin Exp Hepatol 2025; 15:102544. [PMID: 40248345 PMCID: PMC12002650 DOI: 10.1016/j.jceh.2025.102544] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 03/04/2025] [Indexed: 04/19/2025] Open
Abstract
Background/Aims Several prognostic gene signatures have been proposed as predictors of the prognosis of hepatocellular carcinoma (HCC), yet none are implemented in the clinical setting. We aimed to validate two gene scores previously derived from European cohorts. Methods The patients who underwent liver resection for HCC at Copenhagen University Hospital, Rigshospitalet from 2014 to 2018 were included. RNA sequencing determined the expression of genes in the '5-gene score' (HN1, RAN, RAMP3, KRT19, TAF9B) and 'HepatoPredict' (CLU, DPT, SPRY2, CAPSN1). Univariable Cox regression assessed associations with overall and disease-free survival. These parameters were also analyzed in the The Cancer Genome Atlas Liver Hepatocellular Carcinoma (TCGA-LIHC) (n = 359) and National Institute of Health (NIH) (n = 178) cohorts. Results Among 51 patients (88% male), 59% had no underlying liver disease and 25% had cirrhosis. No individual genes were significantly associated with overall survival in the Danish cohort. In the TCGA-LIHC cohort, CLU was linked to better overall survival, and in the NIH cohort, high expression of SPRY2 was associated with poorer overall survival. In the TCGA-LIHC cohort, HN1, RAN, and TAF9B were associated with poorer overall survival, while RAMP3 was linked to better overall survival. No genes were associated with disease-free survival. Conclusion Few individual genes significantly predicted survival in the larger cohorts, and none in the Danish cohort. However, the clinical implication of this needs further investigation.
Collapse
Affiliation(s)
- Stinna D. Schnabl
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jeanett Klubien
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Colm J. O'Rourke
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Sophie Bull Nordkild
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
| | - Jan-Michael Kugler
- Institute for Molecular and Cellular Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Susanne Dam Nielsen
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Viro-immunology Research Unit, Department of Infectious Diseases, Copenhagen University Hospital, Rigshospitalet, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| | - Jesper B. Andersen
- Biotech Research and Innovation Centre (BRIC), Department of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Hans-Christian Pommergaard
- Department of Surgery and Transplantation, Copenhagen University Hospital, Rigshospitalet, Denmark
- Hepatic Malignancy Surgical Research Unit (HEPSURU), Department of Surgery and Transplantation, Rigshospitalet, Copenhagen University Hospital, Denmark
- Institute for Clinical Medicine, University of Copenhagen, Panum Institute, Copenhagen, Denmark
| |
Collapse
|
4
|
Li YL, He R, Tang M, Lan JY, Liu GY, Jiang LH. Bioinformatics identification of shared signaling pathways and core targets linking Benzo[a]pyrene exposure to HCC progression. Toxicology 2025; 514:154129. [PMID: 40174762 DOI: 10.1016/j.tox.2025.154129] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 03/18/2025] [Accepted: 03/29/2025] [Indexed: 04/04/2025]
Abstract
With the increasing prevalence of environmental pollutants, there is growing concern about the potential effects of these substances in major diseases such as liver cancer. Previous studies have suggested that various chemicals, such as benzo[a]pyrene(BaP), produced by burning carbon containing fuels, may negatively affect liver health, but the exact mechanisms remain unclear. This study aimed to explore the potential molecular mechanisms of BaP in the progression of liver cancer. Through an exhaustive study of databases such as ChEMBL, SwissTargetPrediction, STITCH and TCGA, we identified 169 potential targets that are closely related to BaP and liver cancer. Next, we conducted Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analyses using the clusterProfiler package to study the biological functions and important pathways of potential targets induced by BaP, which showed that these targets were associated with mitochondrial function, cellular energy metabolism and REDOX reactions. The protein interaction (PPI) network was constructed using the STRING database and Cytoscape software to identify the core targets UBA52, NDUFS8, CYP1A2, NDUFS1 and CYP3A4. The interaction between BaP and these core proteins was further analyzed via molecular docking using the CB-Dock2 database, demonstrating high binding stability, which suggests their critical role in BaP-induced hepatocellular carcinoma (HCC) toxicity. Subsequently, we found significant differences in the expression of five core genes (UBA52, NDUFS8, CYP1A2, NDUFS1, CYP3A4) in HCC, and significant correlation between UBA52, NDUFS8 and CYP3A4 and survival of HCC patients. Single-cell sequencing analysis showed that the expression of UBA52 gene was particularly pronounced in the three immune cells.
Collapse
Affiliation(s)
- Yong-Le Li
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China
| | - Rong He
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China
| | - Meng Tang
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China
| | - Jing-Yi Lan
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China
| | - Guo-Yang Liu
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China
| | - Li-He Jiang
- School of Basic Medicine, Youjiang Medical College for Nationalities, Baise 533000, China; Key Laboratory of Environmental Toxicology of Anhui Higher Education Institute, Hefei 230032, China; Guangxi Key Laboratory of Environmental Exposomics and Entire Lifecycle Health, Guilin Medical University, Guilin 541001, China.
| |
Collapse
|
5
|
Sasaki M, Sato Y, Nakanuma Y. Genetic re-classification of combined hepatocellular-cholangiocarcinoma and small duct type intrahepatic cholangiocarcinoma. Pathol Res Pract 2025; 270:155999. [PMID: 40349569 DOI: 10.1016/j.prp.2025.155999] [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: 03/20/2025] [Revised: 05/02/2025] [Accepted: 05/05/2025] [Indexed: 05/14/2025]
Abstract
BACKGROUND Combined hepatocellular-cholangiocarcinoma (cHCC-CCA) shares various features with small duct type intrahepatic cholangiocarcinoma (SmD-iCCA) and sometimes histological diagnosis may be difficult. METHODS We examined genetic alterations such as hTERT promoter (hTERT), p53, and fibroblast growth factor receptor 2 (FGFR2) in 103 PLCs diagnosed as cHCC-CCA or SmD-iCCA. A cluster analysis was performed on the R software for re-classification of PLCs including cHCC-CCA and SmD-iCCA. RESULTS The primary liver carcinomas (PLCs) were divided into 5 clusters; 19 tumors (18 %) in Cluster-1 (with alterations in hTERT and/or p53), 24 (23 %) in Cluster-2 (FGFR2 and/or p53), 13 (13 %) in Cluster-3 (IDH2 or null), 19 (18 %) in Cluster-4 (MTAP and/or FGFR2), 28 (27 %) in Cluster-5 (ARID1A and/or PBRM1), being based on genetic alterations. Cluster-1 and Clusters-2 to- 5 formed distinct 2 groups. Cluster-1 was characterized by significantly bigger size, rich and higher histological grade of HCC component, significantly less cholangiolocellular carcinoma (CLC)-component, ductal plate malformation pattern and bile duct adenoma in the background livers. No SmD-iCCA was included in Cluster-1, whereas SmD-iCCA distributed evenly in Clusters 2-5. Cluster-4 was characterized by higher prevalence of hepatitis B and higher histological diversity scores. CONCLUSION PLCs diagnosed as cHCC-CCA or SmD-iCCAs could be divided into 5 clusters based on genetic alterations. Cluster-1 was HCC-like cluster characterized by hTERT alteration, rich and higher grade of HCC and bigger size. Clusters-2-5 may be iCCA-like clusters characterized by different genetic alterations. cHCC-CCA in Cluster-1 and Clusters-2-5 may be handled separately for further analysis and treatment.
Collapse
Affiliation(s)
- Motoko Sasaki
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan.
| | - Yasunori Sato
- Department of Human Pathology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yasuni Nakanuma
- Division of Pathology, Fukui Saiseikai Hospital, Fukui, Japan
| |
Collapse
|
6
|
Zhang L, Gan L, Lin Y, Mei Z, Liao S. FTO Promotes Hepatocellular Carcinoma Progression by Mediating m6A Modification of BUB1 and Targeting TGF-βR1 to Activate the TGF-β Signaling Pathway. J Clin Transl Hepatol 2025; 13:385-394. [PMID: 40385944 PMCID: PMC12078176 DOI: 10.14218/jcth.2025.00007] [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: 01/03/2025] [Revised: 03/01/2025] [Accepted: 03/17/2025] [Indexed: 05/20/2025] Open
Abstract
Background and Aims Fat mass and obesity-associated protein (FTO) has been linked to various cancers, though its role in hepatocellular carcinoma (HCC) remains unclear. This study aimed to investigate FTO expression, its clinical relevance, functional role in HCC progression, and the underlying molecular mechanisms. Methods Quantitative reverse-transcription polymerase chain reaction and immunohistochemical analysis were used to assess FTO expression in HCC. Functional assays, including proliferation, invasion, and epithelial-mesenchymal transition studies, were conducted using HCC cell lines with FTO knockdown. N6-methyladenosine (m6A) RNA immunoprecipitation and RNA stability assays further elucidated the role of FTO in BUB1 mRNA methylation and stability. Co-immunoprecipitation studies were employed to confirm the interaction between BUB1 and TGF-βR1. In vivo studies in nude mice were conducted to evaluate tumor growth following FTO knockdown. Results FTO was significantly upregulated in HCC tissues compared to normal liver tissues, with higher expression observed in advanced tumor-node-metastasis stages and metastatic HCC. Elevated FTO correlated with poor overall survival in patients. Silencing FTO decreased HCC cell proliferation, colony formation, invasion, epithelial-mesenchymal transition, and tumor growth in nude mice. Mechanistically, FTO downregulation led to increased m6A modification of BUB1 mRNA, thereby promoting its degradation via the YTH domain family 2-dependent pathway and reducing BUB1 protein levels. Additionally, BUB1 physically interacted with TGF-βR1, activating downstream TGF-β signaling. Conclusions FTO is overexpressed in HCC and is associated with poor clinical outcomes. Mechanistically, FTO promotes HCC progression by stabilizing BUB1 mRNA through an m6A-YTH domain family 2-dependent pathway, which activates TGF-β signaling. Targeting the FTO-BUB1-TGF-βR1 regulatory network may offer a promising therapeutic strategy for HCC.
Collapse
Affiliation(s)
- Lin Zhang
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Gastroenterology, Chongqing Jiangjin Central Hospital, Chongqing, China
| | - Li Gan
- Department of Anatomy, and Laboratory of Neuroscience and Tissue Engineering, Basic Medical College, Chongqing Medical University, Chongqing, China
| | - Yuru Lin
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zhechuan Mei
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Shengtao Liao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
7
|
Ellington CN, Lengerich BJ, Watkins TBK, Yang J, Adduri AK, Mahbub S, Xiao H, Kellis M, Xing EP. Learning to estimate sample-specific transcriptional networks for 7,000 tumors. Proc Natl Acad Sci U S A 2025; 122:e2411930122. [PMID: 40408406 DOI: 10.1073/pnas.2411930122] [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: 06/14/2024] [Accepted: 04/06/2025] [Indexed: 05/25/2025] Open
Abstract
Cancers are shaped by somatic mutations, microenvironment, and patient background, each altering gene expression and regulation in complex ways, resulting in heterogeneous cellular states and dynamics. Inferring gene regulatory networks (GRNs) from expression data can help characterize this regulation-driven heterogeneity, but network inference requires many statistical samples, limiting GRNs to cluster-level analyses that ignore intracluster heterogeneity. We propose to move beyond coarse analyses of predefined subgroups by using contextualized learning, a multitask learning paradigm that uses multiview contexts including phenotypic, molecular, and environmental information to infer personalized models. With sample-specific contexts, contextualization enables sample-specific models and even generalizes at test time to predict network models for entirely unseen contexts. We unify three network model classes (Correlation, Markov, and Neighborhood Selection) and estimate context-specific GRNs for 7,997 tumors across 25 tumor types, using copy number and driver mutation profiles, tumor microenvironment, and patient demographics as model context. Our generative modeling approach allows us to predict GRNs for unseen tumor types based on a pan-cancer model of how somatic mutations affect gene regulation. Finally, contextualized networks enable GRN-based precision oncology by providing a structured view of expression dynamics at sample-specific resolution, explaining known biomarkers in terms of network-mediated effects and leading to subtypings that improve survival prognosis. We provide a SKLearn-style Python package https://contextualized.ml for learning and analyzing contextualized models, as well as interactive plotting tools for pan-cancer data exploration at https://github.com/cnellington/CancerContextualized.
Collapse
Affiliation(s)
- Caleb N Ellington
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Benjamin J Lengerich
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142
| | - Thomas B K Watkins
- Cancer Institute, University College London, London WC1E 6DD, United Kingdom
| | - Jiekun Yang
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142
| | - Abhinav K Adduri
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Sazan Mahbub
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213
| | - Hanxi Xiao
- Department of Computational and Systems Biology, University of Pittsburgh, Pittsburgh, PA 15260
| | - Manolis Kellis
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA 02139
- Broad Institute, Massachusetts Institute of Technology and Harvard University, Cambridge, MA 02142
| | - Eric P Xing
- Computational Biology Department, Carnegie Mellon University, Pittsburgh, PA 15213
- Machine Learning Department, Mohamed bin Zayed University of Artificial Intelligence, Masdar City SE45 05, Abu Dhabi, United Arab Emirates
- GenBio AI Inc., Palo Alto, CA 94301
| |
Collapse
|
8
|
Kawai-Kitahata F, Asahina Y, Kakinuma S, Inada K, Mochida T, Watakabe K, Nobusawa T, Shimizu T, Tsuchiya J, Miyoshi M, Kaneko S, Murakawa M, Nitta S, Nakagawa M, Kinowaki Y, Ban D, Tanaka S, Anzai T, Takano S, Maekawa S, Enomoto N, Okamoto R. Genetic alterations in hepatocellular carcinoma after sustained virological response in relation to the molecular characterization of metabolic diseases. Hepatol Res 2025. [PMID: 40423574 DOI: 10.1111/hepr.14214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/09/2025] [Revised: 05/09/2025] [Accepted: 05/19/2025] [Indexed: 05/28/2025]
Abstract
AIM The mechanism of hepatocarcinogenesis after sustained virological response (SVR) in hepatitis C virus (HCV) patients is unclear. We compared gene profiles of hepatocellular carcinoma (HCC) between HCV-SVR, steatotic liver disease (SLD), and HCV-non-SVR patients. METHODS This study analyzed 126 resected HCCs from patients with HCV and SLD, classifying them as HCV-SVR (n = 22), HCV-non-SVR (n = 56), and SLD (n = 48). Deep sequencing of 2910 hotspots in 55 cancer-related genes was conducted to examine mutations and copy number variations in both cancerous and background liver tissues. RESULTS The HCV-SVR group comprised more patients who consumed alcohol (45.5% vs. 15.7%, p = 0.008), were obese (54.5% vs. 17.9%, p = 0.002), and had dyslipidemia (18.2% vs. 3.6%, p = 0.029) and hyperuricemia (18.2% vs. 3.6%, p = 0.029) than the HCV-non-SVR group. Mutational profiling of the HCV-SVR HCC showed significantly lower alteration rates of AXIN1 (13.6% vs. 42.9%, p = 0.016), ARID2 (9.1% vs. 39.3%, p = 0.013), and TP53 (9.1% vs. 32.1%, p = 0.030) than HCV-non-SVR patients. Compared with HCV-non-SVR-HCC, SLD-HCCs showed significantly lower rates of TERT promoter mutations (62.5% vs. 85.7%, p = 0.004), ARID2 alterations (12.5% vs. 39.3%, p = 0.003), and AXIN1 alterations (12.5% vs. 42.9%, p = 0.002). HCV-SVR/MASH/MASLD/ALD-HCC had significantly lower alteration rates of the Wnt/β-catenin (41.4% vs. 60.7%, p = 0.048) and chromatin remodeling pathways (27.1% vs. 48.2%, p = 0.026) than HCV-non-SVR-HCC. CONCLUSIONS HCV-SVR HCC is linked to alcohol use and metabolic diseases, showing a mutational profile similar to SLD-HCC.
Collapse
Affiliation(s)
- Fukiko Kawai-Kitahata
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Yasuhiro Asahina
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Sei Kakinuma
- Department of Clinical and Diagnostic Laboratory Science, Institute of Science Tokyo, Tokyo, Japan
| | - Kento Inada
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Tomohiro Mochida
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Keiya Watakabe
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Tsubasa Nobusawa
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Taro Shimizu
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Jun Tsuchiya
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Masato Miyoshi
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Shun Kaneko
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Miyako Murakawa
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Sayuri Nitta
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Mina Nakagawa
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| | - Yuko Kinowaki
- Department of Comprehensive Pathology, Graduate School of Medical and Dental Sciences, Advanced Therapeutic Sciences, Institute of Science Tokyo, Tokyo, Japan
| | - Daisuke Ban
- Department of Hepato-Biliary-Pancreatic Surgery, Institute of Science Tokyo, Tokyo, Japan
| | - Shinji Tanaka
- Department of Molecular Oncology, Institute of Science Tokyo, Tokyo, Japan
| | - Tatsuhiko Anzai
- Department of Biostatistics, M&D Data Science Center, Institute of Integrated Research, Institute of Science Tokyo, Tokyo, Japan
| | - Shinichi Takano
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Shinya Maekawa
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Nobuyuki Enomoto
- Department of Gastroenterology and Hepatology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, Tokyo, Japan
| |
Collapse
|
9
|
Yang C, Zhang Y, Liu Y, Wu X, Sun F. Study on the molecular mechanism of UBA52 and BARD1 regulating hepatocellular carcinoma through the PI3 K/AKT signaling pathway. Discov Oncol 2025; 16:840. [PMID: 40397202 PMCID: PMC12095107 DOI: 10.1007/s12672-025-02600-5] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Accepted: 05/06/2025] [Indexed: 05/22/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the leading causes of cancer-related deaths globally, with its development closely related to complex molecular mechanisms such as gene mutations and abnormal signaling pathways. However, the specific roles of many key genes remain unclear. UBA52 and BARD1 are important genes associated with protein degradation, DNA repair, and cell cycle regulation, but their mechanisms in liver cancer are not well understood. METHODS This study integrated HCC datasets (GSE135631, GSE184733, GSE202853) from the gene expression omnibus (GEO) database to screen for differentially expressed genes (DEGs), perform functional enrichment analysis, weighted gene co-expression network analysis (WGCNA), construct protein-protein interaction (PPI) networks, and conduct survival analysis. Western Blot (WB) and RT-qPCR experiments were used to verify the expression of UBA52 and BARD1 in liver cancer cells and their association with the PI3K/AKT signaling pathway. RESULTS Bioinformatics analysis identified UBA52 and BARD1 as core genes, showing high expression in HCC tissues and correlation with poor prognosis. Western Blot and RT-qPCR results further confirmed the high expression of UBA52 and BARD1 in HCC cell lines (HepG2 and Hep3b). PI3K inhibitors significantly downregulated the expression of UBA52 and BARD1, restored the levels of apoptosis-related factors (Fas, BAX, Caspase-3), and inhibited the expression of cell cycle-related proteins (Cyclin-D1, c-Myc). These findings suggest that UBA52 and BARD1 may regulate HCC cell proliferation, apoptosis, and metastasis through the PI3K/AKT signaling pathway. Furthermore, the molecular mechanism of hepatocellular carcinoma can be modulated by knocking out BARD1 or UBA52. CONCLUSION UBA52 and BARD1 are highly expressed in HCC, and their abnormal expression may promote the occurrence and development of liver cancer by regulating the PI3K/AKT signaling pathway and mechanisms related to apoptosis and cell cycle. The high expression of UBA52 and BARD1 is closely associated with poor prognosis, indicating their potential value as early diagnostic and targeted therapeutic biomarkers for HCC.
Collapse
Affiliation(s)
- Chenrui Yang
- Department of General Surgery, Danzhou People's Hospital (Danzhou People's Hospital Medical Group), 21-1 Datong Road, Nada Town, Danzhou City, 571700, Hainan, China
| | - Yanzhong Zhang
- Department of General Surgery, Danzhou People's Hospital (Danzhou People's Hospital Medical Group), 21-1 Datong Road, Nada Town, Danzhou City, 571700, Hainan, China.
| | - Yajuan Liu
- Department of Clinical Pharmacy, Danzhou People's Hospital (Danzhou People's Hospital Medical Group), 21-1 Datong Road, Nada Town, Danzhou City, 571700, Hainan, China
| | - Xiaoyong Wu
- Department of General Surgery, Danzhou People's Hospital (Danzhou People's Hospital Medical Group), 21-1 Datong Road, Nada Town, Danzhou City, 571700, Hainan, China
| | - Fangyuan Sun
- Department of General Surgery, Danzhou People's Hospital (Danzhou People's Hospital Medical Group), 21-1 Datong Road, Nada Town, Danzhou City, 571700, Hainan, China
| |
Collapse
|
10
|
Vij M, Raju LP, Jothimani D, Simon E, Radhakrishnan S, Martin CA, Gowrishankar G, Subbiah K, Rajalingam R, Kaliamoorthy I, Rammohan A, Rela M. Granuloma rich hepatocellular carcinoma (GrHCC): Clinicopathologic and genomic characterization. Ann Diagn Pathol 2025; 79:152504. [PMID: 40413956 DOI: 10.1016/j.anndiagpath.2025.152504] [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: 03/21/2025] [Revised: 05/16/2025] [Accepted: 05/19/2025] [Indexed: 05/27/2025]
Abstract
There is limited literature on sarcoid like granuloma (SLG) associated with hepatocellular carcinoma (HCC). Here in, we studied clinicopathological characteristics, and explored the potential significance of SLG in HCC. We termed these tumors as granuloma rich HCC (GrHCC). We reviewed clinicopathologic features in 30 GrHCC tumors that were diagnosed in 21 patients during a period of 68-month at a single institution. The study included 17 males and 4 females, with ages ranging from 43 to 71 years in males and 20 to 69 years in females. Tumor downstaging was done in 4 patients. Tumor sizes ranged from 0.6 to 23 cm, with a mean size of 2.41 cm. Majority of tumors showed well to moderate cellular differentiation. A solitary well-formed epithelioid granuloma sufficed to classify the tumor as GrHCC. The intratumoral granulomas were compact, well-formed, and discrete, consisting of collections of epithelioid histiocytes and multinucleate histiocytic giant cells. Mild lymphocytic inflammation was also noted. Single to several granulomas were identified in the tumor. Size of granuloma ranged from 170 to 650 μm. Only one tumor showed necrotizing granulomas. Genomic analysis of 4 tumors revealed TP53 mutation. Two tumors also exhibited a TERT promoter mutation. All patients were alive till last follow-up, except for one, who died due to septic shock, unrelated to the HCC. This study provides valuable insights into the clinical findings, histopathological features and molecular characteristics of GrHCC.
Collapse
Affiliation(s)
- Mukul Vij
- Department of Pathology, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India.
| | - Lexmi Priya Raju
- Department of Pathology, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Dinesh Jothimani
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India.
| | - Evangeline Simon
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India.
| | - Subathra Radhakrishnan
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Catherine Ann Martin
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Gowripriya Gowrishankar
- Department of Pathology, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India.
| | - Komalavalli Subbiah
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Rajesh Rajalingam
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Ilankumaran Kaliamoorthy
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India.
| | - Ashwin Rammohan
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| | - Mohamed Rela
- The Institute of Liver Disease & Transplantation, Dr. Rela Institute & Medical Centre, No. 7 CLC Works road Chromepet, Chennai 44, Tamil Nadu, India
| |
Collapse
|
11
|
Wong K, Bishop JA, Weinreb I, Motta M, Del Castillo Velasco-Herrera M, Bellacchio E, Ferreira I, van der Weyden L, Boccacino JM, Lauri A, Rotundo G, Ciolfi A, Cheema S, Olvera-León R, Offord V, Droop A, Vermes I, Allgäuer M, Hyrcza M, Anderson E, Smith K, de Saint Aubain N, Mogler C, Stenzinger A, Arends MJ, Brenn T, Tartaglia M, Adams DJ. Wnt/β-catenin activation by mutually exclusive FBXW11 and CTNNB1 hotspot mutations drives salivary basal cell adenoma. Nat Commun 2025; 16:4657. [PMID: 40389436 PMCID: PMC12089348 DOI: 10.1038/s41467-025-59871-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 05/07/2025] [Indexed: 05/21/2025] Open
Abstract
Basal cell adenoma (BCA) and basal cell adenocarcinoma (BCAC) of the salivary gland are rare tumours that can be difficult to distinguish from each other and other salivary gland tumour subtypes. Using next-generation sequencing, we identify a recurrent FBXW11 missense mutation (p.F517S) in BCA that is mutually exclusive with the previously reported CTNNB1 p.I35T gain-of-function (GoF) mutation with these mutations collectively accounting for 94% of BCAs. In vitro, mutant FBXW11 is characterised by defective binding to β-catenin and higher protein levels within the nucleus. This is consistent with the increased nuclear expression of β-catenin and activation of the Wnt/β-catenin pathway. The genomic profiles of BCAC are distinct from BCA, with hotspot DICER1 and HRAS mutations and putative driver mutations affecting PI3K/AKT and NF-κB signalling pathway genes. These findings have important implications for the diagnosis and treatment of BCA and BCAC, which, despite histopathologic overlap, may be unrelated entities.
Collapse
Affiliation(s)
- Kim Wong
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Justin A Bishop
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Ilan Weinreb
- Laboratory Medicine Program, University Health Network, Toronto General Hospital, Toronto, ON, Canada
- Department of Pathobiology and Laboratory Medicine, University of Toronto, Toronto, ON, Canada
| | - Marialetizia Motta
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | | | - Emanuele Bellacchio
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Ingrid Ferreira
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | | | | | - Antonella Lauri
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Giovannina Rotundo
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Andrea Ciolfi
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - Saamin Cheema
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Rebeca Olvera-León
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Victoria Offord
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Alastair Droop
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Ian Vermes
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Michael Allgäuer
- Institute of Pathology, University Hospital Heidelberg, Heidelberg, Germany
| | - Martin Hyrcza
- Department of Pathology and Laboratory Medicine, University of Calgary, Arnie Charboneau Cancer Institute, Calgary, AB, Canada
| | - Elizabeth Anderson
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Katie Smith
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK
| | - Nicolas de Saint Aubain
- Department of Pathology, Hôpital Universitaire de Bruxelles (HUB), Université Libre de Bruxelles, Brussels, Belgium
| | - Carolin Mogler
- School of Medicine and Health, Technical University Munich, Munich, Germany
| | | | - Mark J Arends
- Edinburgh Pathology, Cancer Research UK Scotland Centre, The University of Edinburgh, Institute of Genetics and Cancer, Edinburgh, UK
| | - Thomas Brenn
- Departments of Pathology and Dermatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco Tartaglia
- Molecular Genetics and Functional Genomics, Ospedale Pediatrico Bambino Gesù, IRCCS, Rome, Italy
| | - David J Adams
- Experimental Cancer Genetics, Wellcome Sanger Institute, Hinxton, Cambridge, UK.
| |
Collapse
|
12
|
Xiao Q, Liu Y, Li T, Wang C, He S, Zhai L, Yang Z, Zhang X, Wu Y, Liu Y. Viral oncogenesis in cancer: from mechanisms to therapeutics. Signal Transduct Target Ther 2025; 10:151. [PMID: 40350456 PMCID: PMC12066790 DOI: 10.1038/s41392-025-02197-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 01/22/2025] [Accepted: 03/03/2025] [Indexed: 05/14/2025] Open
Abstract
The year 2024 marks the 60th anniversary of the discovery of the Epstein-Barr virus (EBV), the first virus confirmed to cause human cancer. Viral infections significantly contribute to the global cancer burden, with seven known Group 1 oncogenic viruses, including hepatitis B virus (HBV), human papillomavirus (HPV), EBV, Kaposi sarcoma-associated herpesvirus (KSHV), hepatitis C virus (HCV), human T-cell leukemia virus type 1 (HTLV-1), and human immunodeficiency virus (HIV). These oncogenic viruses induce cellular transformation and cancer development by altering various biological processes within host cells, particularly under immunosuppression or co-carcinogenic exposures. These viruses are primarily associated with hepatocellular carcinoma, gastric cancer, cervical cancer, nasopharyngeal carcinoma, Kaposi sarcoma, lymphoma, and adult T-cell leukemia/lymphoma. Understanding the mechanisms of viral oncogenesis is crucial for identifying and characterizing the early biological processes of virus-related cancers, providing new targets and strategies for treatment or prevention. This review first outlines the global epidemiology of virus-related tumors, milestone events in research, and the process by which oncogenic viruses infect target cells. It then focuses on the molecular mechanisms by which these viruses induce tumors directly or indirectly, including the regulation of oncogenes or tumor suppressor genes, induction of genomic instability, disruption of regular life cycle of cells, immune suppression, chronic inflammation, and inducing angiogenesis. Finally, current therapeutic strategies for virus-related tumors and recent advances in preclinical and clinical research are discussed.
Collapse
Affiliation(s)
- Qing Xiao
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Yi Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Tingting Li
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Chaoyu Wang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Sanxiu He
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Liuyue Zhai
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Zailin Yang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Xiaomei Zhang
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yongzhong Wu
- Department of Radiation Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| | - Yao Liu
- Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Department of Hematology-Oncology, Chongqing University Cancer Hospital, Chongqing, China.
| |
Collapse
|
13
|
Lei W, Zhou K, Lei Y, Li Q, Zhu H. Pathogenesis and Systemic Treatment of Hepatocellular Carcinoma: Current Status and Prospects. Mol Cancer Ther 2025; 24:692-708. [PMID: 39417575 DOI: 10.1158/1535-7163.mct-24-0403] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2024] [Revised: 08/14/2024] [Accepted: 10/08/2024] [Indexed: 10/19/2024]
Abstract
Hepatocellular carcinoma (HCC) remains one of the major threats to human health worldwide. The emergence of systemic therapeutic options has greatly improved the prognosis of patients with HCC, particularly those with advanced stages of the disease. In this review, we discussed the pathogenesis of HCC, genetic alterations associated with the development of HCC, and alterations in the tumor immune microenvironment. Then, important indicators and emerging technologies related to the diagnosis of HCC are summarized. Also, we reviewed the major advances in treatments for HCC, offering insights into future prospects for next-generation managements.
Collapse
Affiliation(s)
- Wanting Lei
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Kexun Zhou
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Ye Lei
- College of Liberal Arts, Neijiang Normal University, Neijiang, China
| | - Qiu Li
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| | - Hong Zhu
- Department of Medical Oncology, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
- Division of Abdominal Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
14
|
Yang T, Wang M, Wang N, Pan M, Xu Y, You Q, Yao L, Xu J, Gu L, Sun X, Zhang L, Xu J, Li B, Wang G, Cai S, Lv G, Shen F. Cost-Effective Identification of Hepatocellular Carcinoma from Cirrhosis or Chronic Hepatitis Virus Infection Using Eight Methylated Plasma DNA Markers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411945. [PMID: 40135830 PMCID: PMC12097027 DOI: 10.1002/advs.202411945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/20/2025] [Indexed: 03/27/2025]
Abstract
Early detection of hepatocellular carcinoma (HCC) in patients with liver cirrhosis (LC) and/or hepatitis virus B/C infection (HVI) improves survival, highlighting the need for accurate, affordable diagnostic tools. Here, 11 methylated DNA markers (MDMs) are identified during marker discovery. In phase I, each selected MDM is validated in 175 plasma samples (HCC, n = 85; LC/HVI, n = 72) by the CO-methylation aMplification rEal-Time PCR (COMET) assay. Of these, 8 MDMs are qualified for phase II study, where a logistic regression model (COMET-LR) is trained and validated with 336 plasma samples (HCC, n = 211; LC/HVI, n = 113; training vs validation, 2:1). In the validation, the COMET-LR achieved 90.0% sensitivity at 97.4% specificity. Notably, sensitivity in patients with TNM stage I, diameter<3 cm, AFP-negative (<20 ng mL-1), PIVKA-II-negative (<40 mAU mL-1) is 82.4%, 77.8%, 88.6%, and 85.7%, respectively. The COMET-LR outperformed multiple protein markers (AFP, AFP-L3, and PIVKA-II) and published scores for HCC screening (GALAD, Doylestown, and ASAP), in terms of both sensitivity and specificity. The assay represents a significant advancement in addressing the unmet need for accurate, non-invasive, accessible, and cost-effective early detection tools for LC/HVI individuals. Further validation in a prospective cohort is warranted.
Collapse
Affiliation(s)
- Tian Yang
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| | - Mingda Wang
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| | - Nanya Wang
- Department of Hepatobiliary and Pancreatic SurgeryGeneral Surgery CenterFirst Hospital of Jilin UniversityChangchunJilin130021China
- Phase I clinical trials unitFirst Hospital of Jilin UniversityChangchunJilin130021China
| | - Mingxin Pan
- Department of Hepatobiliary Surgery IIGeneral Surgery CenterZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Yu Xu
- Burning Rock BiotechGuangzhou510300China
| | | | - Lanqing Yao
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| | - Jiahao Xu
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| | - Lihui Gu
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic SurgeryGeneral Surgery CenterFirst Hospital of Jilin UniversityChangchunJilin130021China
| | - Lei Zhang
- Burning Rock BiotechGuangzhou510300China
| | - Jiayue Xu
- Burning Rock BiotechGuangzhou510300China
| | - Bingsi Li
- Burning Rock BiotechGuangzhou510300China
| | | | | | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic SurgeryGeneral Surgery CenterFirst Hospital of Jilin UniversityChangchunJilin130021China
| | - Feng Shen
- Department of Hepatobiliary SurgeryEastern Hepatobiliary Surgery HospitalNaval Medical UniversityShanghai200438China
| |
Collapse
|
15
|
Yost KE, Zhao Y, Hung KL, Zhu K, Xu D, Corces MR, Shams S, Louie BH, Sarmashghi S, Sundaram L, Luebeck J, Clarke S, Doane AS, Granja JM, Choudhry H, Imieliński M, Cherniack AD, Khurana E, Bafna V, Felau I, Zenklusen JC, Laird PW, Curtis C, Greenleaf WJ, Chang HY. Three-dimensional genome landscape of primary human cancers. Nat Genet 2025; 57:1189-1200. [PMID: 40355593 DOI: 10.1038/s41588-025-02188-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/02/2025] [Indexed: 05/14/2025]
Abstract
Genome conformation underlies transcriptional regulation by distal enhancers, and genomic rearrangements in cancer can alter critical regulatory interactions. Here we profiled the three-dimensional genome architecture and enhancer connectome of 69 tumor samples spanning 15 primary human cancer types from The Cancer Genome Atlas. We discovered the following three archetypes of enhancer usage for over 100 oncogenes across human cancers: static, selective gain or dynamic rewiring. Integrative analyses revealed the enhancer landscape of noncancer cells in the tumor microenvironment for genes related to immune escape. Deep whole-genome sequencing and enhancer connectome mapping provided accurate detection and validation of diverse structural variants across cancer genomes and revealed distinct enhancer rewiring consequences from noncoding point mutations, genomic inversions, translocations and focal amplifications. Extrachromosomal DNA promoted more extensive enhancer rewiring among several types of focal amplification mechanisms. These results suggest a systematic approach to understanding genome topology in cancer etiology and therapy.
Collapse
Affiliation(s)
- Kathryn E Yost
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Whitehead Institute for Biomedical Research, Cambridge, MA, USA
| | - Yanding Zhao
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - King L Hung
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kaiyuan Zhu
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Duo Xu
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Pathos AI, Chicago, IL, USA
| | - M Ryan Corces
- Gladstone Institute of Neurological Disease, San Francisco, CA, USA
- Gladstone Institute of Data Science and Biotechnology, San Francisco, CA, USA
- Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Shadi Shams
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | - Bryan H Louie
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
| | | | - Laksshman Sundaram
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Illumina AI laboratory, Illumina Inc, Foster City, CA, USA
- NVIDIA Bio Research, NVIDIA, Santa Clara, CA, USA
| | - Jens Luebeck
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Stanley Clarke
- Vilcek Institute of Graduate Biomedical Sciences, NYU Grossman School of Medicine, New York City, NY, USA
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Ashley S Doane
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Jeffrey M Granja
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
| | - Hani Choudhry
- Department of Biochemistry, Faculty of Science, Cancer and Mutagenesis Unit, King Fahd Center for Medical Research, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Marcin Imieliński
- Laura and Isaac Perlmutter Cancer Center, New York University Langone Health, New York City, NY, USA
- Department of Pathology, New York University Langone Health, New York City, NY, USA
- New York Genome Center, New York City, NY, USA
| | - Andrew D Cherniack
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, MA, USA
- Harvard Medical School, Boston, MA, USA
| | - Ekta Khurana
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York City, NY, USA
- Department of Physiology and Biophysics, Weill Cornell Medicine, New York City, NY, USA
- Institute for Computational Biomedicine, Weill Cornell Medicine, New York City, NY, USA
- Englander Institute for Precision Medicine, Weill Cornell Medicine, New York City, NY, USA
| | - Vineet Bafna
- Department of Computer Science and Engineering, University of California, San Diego, La Jolla, CA, USA
| | - Ina Felau
- National Cancer Institute, NIH, Bethesda, MD, USA
| | | | - Peter W Laird
- Center for Epigenetics, Van Andel Research Institute, Grand Rapids, MI, USA
| | - Christina Curtis
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - William J Greenleaf
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Applied Physics, Stanford University, Stanford, CA, USA.
| | - Howard Y Chang
- Center for Personal Dynamic Regulomes, Stanford University School of Medicine, Stanford, CA, USA.
- Department of Dermatology, Stanford University School of Medicine, Stanford, CA, USA.
- Howard Hughes Medical Institute, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
16
|
Liu X, Ma Q, Jia Z, Zhou Y, Zou C, Xiao Y, Chen Y, Ma C, Song L, Yang J, Wang C, Xu H, Chen H, Shi J, Yue J, Sun Y, Hu D, Petersen RB, Li Y, Peng A, Huang K, Zheng L. ISG15 Enhances the Activity of γ-Glutamate Cysteine Ligase to Suppress Apoptosis in High Fat Diet-Promoted Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2416401. [PMID: 40126377 PMCID: PMC12097128 DOI: 10.1002/advs.202416401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/07/2024] [Revised: 03/07/2025] [Indexed: 03/25/2025]
Abstract
Obesity is a leading risk factor for development of hepatocellular carcinoma (HCC). High-fat intake produces cytotoxic effects in liver cells, such as excessive reactive oxygen species (ROS) accumulation and apoptosis. How HCC cells regulate ROS level and escape the cytotoxic effects of high fat diet (HFD) stress remains unclear. Herein, this work reports a critical anti-ROS/apoptotic role of the ubiquitin-like protein interferon stimulated gene 15 (ISG15) in HFD-promoted HCC. In mouse models and clinical HCC samples, upregulation of ISG15 is associated with hepatic steatosis. Notably, upregulated ISG15 elevates cellular glutathione levels, which subsequently reduces ROS accumulation and confers resistance to apoptosis in HCC cells. In diethylnitrosamine-induced HCC mouse model, HFD-feeding promotes HCC progression in wildtype mice, while tumor growth is significantly suppressed accompanied by apoptosis of HCC cells in Isg15-KO mice. Mechanistically, ISG15 promotes the activity of γ-glutamate cysteine ligase (γ-GCL), a rate-limiting heterodimeric holoenzyme of glutathione synthesis consisting of glutamate-cysteine ligase catalytic subunit (GCLC) and glutamate-cysteine ligase modifier subunit (GCLM). Independent of ISGylation, ISG15 forms an ISG15/GCLM/GCLC complex that promotes GCLM-GCLC interaction, increases glutathione generation and inhibits HFD-induced apoptosis in HCC cells. Together, an anti-apoptotic ISG15-γ-GCL-glutothione axis is suggested in HFD-promoted HCC.
Collapse
Affiliation(s)
- Xinran Liu
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Qiujin Ma
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Zhao Jia
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Yihao Zhou
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, TaiKang Center for Life and Medical Sciences; Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhan430072China
| | - Churong Zou
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Yushuo Xiao
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Yuchen Chen
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Chuyao Ma
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Liangliang Song
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Jing Yang
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Chen Wang
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Huidie Xu
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Hong Chen
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Jiajian Shi
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Junqiu Yue
- Department of PathologyHubei Cancer HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430079China
| | - Yu Sun
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, TaiKang Center for Life and Medical Sciences; Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhan430072China
| | - Desheng Hu
- Department of Integrated Traditional Chinese and Western MedicineUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
- China‐Russia Medical Research Center for Stress ImmunologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430000China
| | - Robert B Petersen
- Foundational SciencesCentral Michigan University College of MedicineMt. PleasantMI48859USA
| | - Yangkai Li
- Department of Thoracic SurgeryTongji HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430030China
| | - Anlin Peng
- Department of PharmacyThe Third Hospital of WuhanTongren Hospital of Wuhan UniversityWuhan430070China
| | - Kun Huang
- School of PharmacyTongji Medical College and State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious DiseasesHuazhong University of Science & TechnologyWuhan430030China
| | - Ling Zheng
- State Key Laboratory of Metabolism and Regulation in Complex Organisms, TaiKang Center for Life and Medical Sciences; Frontier Science Center for Immunology and Metabolism, College of Life SciencesWuhan UniversityWuhan430072China
| |
Collapse
|
17
|
Cheng Z, Ren Y, Wang X, Zhang Y, Hua Y, Zhao H, Lu H. A novel prognostic framework for HBV-infected hepatocellular carcinoma: insights from ferroptosis and iron metabolism proteomics. Brief Bioinform 2025; 26:bbaf216. [PMID: 40381315 PMCID: PMC12085197 DOI: 10.1093/bib/bbaf216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 04/15/2025] [Accepted: 04/21/2025] [Indexed: 05/20/2025] Open
Abstract
Effective classification methods and prognostic models enable more accurate classification and treatment of hepatocellular carcinoma (HCC) patients. However, the weak correlation between RNA and protein data has limited the clinical utility of previous RNA-based prognostic models for HCC. In this work, we constructed a novel prognostic framework for HCC patients using seven differentially expressed proteins associated with ferroptosis and iron metabolism. Furthermore, this prognostic model robustly classifies HCC patients into three clinically relevant risk groups. Significant differences in overall survival, age, tumor differentiation, microvascular invasion, distant metastasis, and alpha-fetoprotein levels were observed among the risk groups. Based on the prognostic model and known biological pathways, we explored the potential mechanisms underlying the inconsistent differential expression patterns of FTH1 (Ferritin heavy chain 1) mRNA and protein. Our findings demonstrated that tumor tissues in HCC patients promote liver cancer progression by downregulating FTH1 protein expression, rather than upregulating FTH1 mRNA expression, ultimately leading to poor prognosis. Subsequently, based on risk score and tumor size, we developed a nomogram for predicting the prognosis of HCC patients, which demonstrated superior predictive performance in both the training and validation cohorts (C-index: 0.774; AUC for 1-5 years: 0.783-0.964). Additionally, our findings demonstrated that the adverse prognosis of high-risk HCC patients was closely correlated with ferroptosis in liver cancer tissues, alterations in iron metabolism, and changes in the tumor immune microenvironment. In conclusion, our prognostic model and predictive nomogram offer novel insights and tools for the effective classification of HCC patients, potentially enhancing clinical decision-making and outcomes.
Collapse
Affiliation(s)
- Zhiwei Cheng
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Hongkou District, Shanghai 200080, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yongyong Ren
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Institute of Bioinformatics, Shanghai Academy of Experimental Medicine, 528 Hongshan Road, Pudong New District, Shanghai 200126, China
| | - Xinbo Wang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yuening Zhang
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
| | - Yingqi Hua
- Department of Orthopedic Oncology, Shanghai Bone Tumor Institute, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, 100 Haining Road, Hongkou District, Shanghai 200080, China
| | - Hongyu Zhao
- Department of Biostatistics, Yale University, 300 George Street, New Haven, CT 06511, United States
| | - Hui Lu
- Department of Bioinformatics and Biostatistics, School of Life Sciences and Biotechnology, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- SJTU-Yale Joint Center of Biostatistics and Data Science, National Center for Translational Medicine, Shanghai Jiao Tong University, 800 Dongchuan Road, Minhang District, Shanghai 200240, China
- Institute of Bioinformatics, Shanghai Academy of Experimental Medicine, 528 Hongshan Road, Pudong New District, Shanghai 200126, China
- Shanghai Engineering Research Center for Big Data in Pediatric Precision Medicine, Center for Biomedical Informatics, Shanghai Children’s Hospital, School of Medicine, Shanghai Jiao Tong University, 1400 Beijing West Road, Jing'an District, Shanghai 200040, China
| |
Collapse
|
18
|
Han Y, Zeng A, Liang X, Jiang Y, Wang F, Song L. Multi-omics analyses develop and validate the optimal prognostic model on overall survival prediction for resectable hepatocellular carcinoma. J Gastrointest Oncol 2025; 16:628-649. [PMID: 40386602 PMCID: PMC12078830 DOI: 10.21037/jgo-24-710] [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: 09/20/2024] [Accepted: 02/21/2025] [Indexed: 05/20/2025] Open
Abstract
Background Prediction of prognosis in patients with hepatocellular carcinoma (HCC) by single-omics profiling has been widely studied. However, the prognosis related to biomarkers of multiple omics has not been investigated. We aimed to establish and validate a prediction model for prognosis prediction of resectable HCC combining multi-omics and clinicopathological factors. Methods The training cohort involved multi-omics data of 330 patients with resectable HCC (stage I-IIIA) at mutational, copy number variation (CNV), transcriptional, and methylation levels from The Cancer Genome Atlas (TCGA) database, along with clinicopathological information. The validation cohort involved samples from 40 HCC patients of Beijing Youan Hospital. Univariate and multivariate analyses were performed in single-omics with clinicopathological variables regarding patient prognosis, and independent risk factors were combined to establish the multi-omics model. The predictive accuracy was assessed by the receiver operating characteristic (ROC) method. Results The mutational, copy number, transcriptional, and methylation alterations in HCC were characterized. TP53, CTNNB1, and TTN were among the genes with the top mutational frequency, and FBN1 and MAP1B mutations were independent risk factors for patient overall survival (OS). 1q21.3 and 1q23.3 ranked the highest in copy number amplifications, and 8p12 and 8p23.3 ranked the highest in deletions, and CSMD1, TP53, and RB1 were genes with the most frequent CNVs. AFP, GPC3, and TERT were among genes with the most significant aberrant transcription, and the transcription of CCNJL, FRMD1, and GRPEL2 were independent risk factors for OS. Both hypermethylation and hypomethylation can be observed. The aberrant methylation of CXorf15, DACT2, GP6, KIAA1522, and PDIA3 were independent risk factors. Single-omics models were established with independent risk factors, and were validated by internal and external datasets. A prognostic model for OS with multi-omics independent risk factors and clinicopathlogical information was established. Internal and external validation achieved an optimal maximal area under the curve (AUC) of 0.98 at 1 year and 0.88 at 2 years, respectively. Conclusions A multi-omics model combining molecular aberrancies and clinicopathological information was established and proved to be optimal for prognosis prediction of resectable HCC. This model may be helpful for therapeutic strategy selection and survival assessment.
Collapse
Affiliation(s)
- Ying Han
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ajuan Zeng
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xueying Liang
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingying Jiang
- Department of Hepatology and Gastroenterology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Fenglin Wang
- College of Life Sciences, Nankai University, Tianjin, China
| | - Lele Song
- Department of Radiotherapy, the Eighth Medical Center of the Chinese PLA General Hospital, Beijing, China
| |
Collapse
|
19
|
Shao Q, Gao HY, Wang ZY, Qian YL, Chen WX. Construction of a novel five programmed cell death-related gene signature as a promising prognostic model for triple negative breast cancer. PeerJ 2025; 13:e19359. [PMID: 40313394 PMCID: PMC12045267 DOI: 10.7717/peerj.19359] [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: 11/20/2024] [Accepted: 04/02/2025] [Indexed: 05/03/2025] Open
Abstract
Background Triple negative breast cancer (TNBC) is a more aggressive subtype of breast cancer that usually progresses rapidly, develops drug resistance, metastasis, and relapses, and remains a challenge for clinicians to treat. Programmed cell death (PCD), a conserved mechanism of cell suicide controlled by various pathways, contributed to carcinogenesis and cancer progression. Nevertheless, the prognostic significance of PCD-related genes in TNBC remains largely unclear, and more accurate prognostic models are urgently needed. Methods Gene expression profiles and clinical information of TNBC patients were obtained from The Cancer Genome Atlas (TCGA) and Gene Expression Omnibus (GEO) database. Least absolute shrinkage and selection operator (LASSO) and multivariate Cox regression analysis were used to establish the PCD-related gene signature. Kaplan-Meier plotter, receiver operating characteristic curves, and nomogram were applied to validate the prognostic value of the gene signature. Gene set enrichment analysis was carried out to investigate the pathways and molecular functions. Results Five PCD-related genes including SEPTIN3, SCARB1, CHML, SYNM, and COL5A3 were identified to establish the PCD-related risk score for TNBC patients. Patients stratified into high-risk or low-risk group showed significantly different survival outcome, immune infiltration, and drug susceptibility. Kaplan-Meier and receiver operating characteristic curves showed a good performance for survival prediction in different cohorts. Gene set enrichment analysis revealed that the five-gene signature was associated with tumor metabolism, cancer cell proliferation, invasion and metastasis, and tumor microenvironment. Nomogram including the five-gene signature was established. Conclusion A novel five PCD-related gene signature and nomogram could be used for prognostic prediction in TNBC. The present work might offer useful insights in digging sensitive and effective biomarkers for TNBC prognosis prediction and establishing accurate prognostic model in clinical management.
Collapse
Affiliation(s)
- Quanfeng Shao
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Hai-yan Gao
- Department of Breast Surgery, Changzhou Cancer Hospital, Changzhou, Jiangsu, China
| | - Zi-ying Wang
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| | - Yu-ling Qian
- Kangda Clinical College, Kangda College of Nanjing Medical University, Lianyungang, Jiangsu, China
| | - Wei-xian Chen
- Department of Breast Surgery, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
- Post-doctoral Working Station, Changzhou No. 2 People’s Hospital, The Third Affiliated Hospital of Nanjing Medical University, Changzhou, Jiangsu, China
| |
Collapse
|
20
|
Shao F, Wang R, Li X, Hu Y, Zhang Z, Cai J, Yang J, Feng X, Ren S, Huang Z, Xie Y. TTC36 promotes proliferation and drug resistance in hepatocellular carcinoma cells by inhibiting c-Myc degradation. Cell Death Dis 2025; 16:332. [PMID: 40274799 PMCID: PMC12022016 DOI: 10.1038/s41419-025-07663-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 04/08/2025] [Accepted: 04/11/2025] [Indexed: 04/26/2025]
Abstract
High c-Myc protein accumulation contributes to the proliferation, invasion, and drug resistance in multiple cancer cells, but the underlying mechanism about c-Myc accumulation remains not to be elucidated. Here, we demonstrate that TTC36 promotes c-Myc protein accumulation in hepatocellular carcinoma cells, thereby driving the proliferation and sorafenib resistance in hepatocellular carcinoma cells. Ttc36 depletion disrupts the interaction between SET and PPP2R1A, consequently activating PP2A. Activated PP2A directly dephosphorylates p-c-MycS62 and activates GSK3β, relying on AKT, leading increased phosphorylation of p-c-MycT58, finally promotes FBXW7-mediated polyubiquitination and degradation of c-Myc. Inhibitors targeting GSK3β and PP2A effectively reverse the sorafenib resistance promoted by TTC36. These findings highlight the crucial role of TTC36 in c-Myc accumulation-caused proliferation and sorafenib resistance in HCC, providing a promising combination strategy for treating patients with c-Myc protein accumulation in advanced HCC.
Collapse
Affiliation(s)
- Fengling Shao
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Runzhi Wang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xinyi Li
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Yanxia Hu
- School of Life and Health Sciences, Hainan University, Haikou, China
| | - Zaikuan Zhang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Jing Cai
- College of Basic Medical Sciences, Harbin Medical University, Harbin, China
| | - Jieru Yang
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Xiaosong Feng
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China
| | - Suxia Ren
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
| | - Zengyi Huang
- Department of Cell Biology and Genetics, School of Basic Medical Sciences, Chongqing Medical University, Chongqing, China.
- Mitomedical laboratory of Children's Hospital of Chongqing Medical University, National Clinical Research Center for Child Health and Disorders, Ministry of Education Key Laboratory of Child Development and Disorders, Chongqing Key Laboratory of Child Rare Diseases in Infection and Immunity, Chongqing, China.
| | - Yajun Xie
- The Ministry of Education Key Laboratory of Laboratory Medical Diagnostics, the College of Laboratory Medicine, Chongqing Medical University, Chongqing, China.
| |
Collapse
|
21
|
Li K, Mathew B, Saldanha E, Ghosh P, Krainer AR, Dasarathy S, Huang H, Xiang X, Mishra L. New insights into biomarkers and risk stratification to predict hepatocellular cancer. Mol Med 2025; 31:152. [PMID: 40269686 PMCID: PMC12020275 DOI: 10.1186/s10020-025-01194-6] [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: 12/09/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the third major cause of cancer death worldwide, with more than a doubling of incidence over the past two decades in the United States. Yet, the survival rate remains less than 20%, often due to late diagnosis at advanced stages. Current HCC screening approaches are serum alpha-fetoprotein (AFP) testing and ultrasound (US) of cirrhotic patients. However, these remain suboptimal, particularly in the setting of underlying obesity and metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH), which are also rising in incidence. Therefore, there is an urgent need for novel biomarkers that can stratify risk and predict early diagnosis of HCC, which is curable. Advances in liver cancer biology, multi-omics technologies, artificial intelligence, and precision algorithms have facilitated the development of promising candidates, with several emerging from completed phase 2 and 3 clinical trials. This review highlights the performance of these novel biomarkers and algorithms from a mechanistic perspective and provides new insight into how pathological processes can be detected through blood-based biomarkers. Through human studies compiled with animal models and mechanistic insight in pathways such as the TGF-β pathway, the biological progression from chronic liver disease to cirrhosis and HCC can be delineated. This integrated approach with new biomarkers merit further validation to refine HCC screening and improve early detection and risk stratification.
Collapse
Affiliation(s)
- Katrina Li
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Brandon Mathew
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Ethan Saldanha
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Puja Ghosh
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Adrian R Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Hai Huang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY, 11030, USA
| | - Xiyan Xiang
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA.
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA.
- Department of Surgery, George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
22
|
Meaddough EL, Sarasua SM, Kunkel D, Boccuto L, Ganakammal SR, Moersen M, Farrell CL. Assessment of CYP2D6 gene expression in liver tissue: Variability in CYP2D6 mRNA levels within genotype-predicted metabolizer phenotype groups. Chem Biol Interact 2025; 416:111526. [PMID: 40280382 DOI: 10.1016/j.cbi.2025.111526] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 04/29/2025]
Abstract
Pharmacogenetic (PGx) testing can be used to help guide drug therapy and decrease or avoid the risk of adverse drug reactions. CYP2D6 is an important pharmacogene in pharmacogenomics testing panels. However, phenoconversion, whereby an individual's ability to metabolize a drug does not match the genotype-predicted metabolizer status, is a confounding factor to the accurate application of PGx testing results to patient care. To address this issue, CYP2D6 expression between and within genotype-predicted CYP2D6 metabolizer phenotype groups was compared using WGS and RNA-Seq data from 134 normal liver tissue donors obtained from the GTEx program. Wide variability in CYP2D6 mRNA levels was observed within metabolizer phenotype groups. The median expression level for ultrarapid metabolizers (UMs) was 738.9 TPM (transcripts per million; 196.8-778.9 TPM), 212.5 TPM (32.1-666.5 TPM) for normal metabolizers (NMs), 219.6 TPM for intermediate metabolizers (IMs) (22-389.8 TPM), and 121.2 TPM for poor metabolizers (PMs) (9.3-298.2 TPM). The PM and UM phenotypes were significant predictors of CYP2D6 expression (p = 0.0004 and p = 0.019, respectively). Interestingly, expression of the gene encoding human serum albumin (ALB) was also a significant predictor of CYP2D6 expression (p = 0.0003). Data from 50 patients with hepatocellular carcinoma obtained from the TCGA program showed no significant difference in expression between tumor tissue (median = 119.7 TPM, range 0.16-817.7 TPM) and normal matched tissue (median = 143.3 TPM, range 26.2-810.7 TPM). Transcriptional regulation of CYP2D6 expression may contribute to differences in drug response and risk for CYP2D6 phenoconversion. Efforts to understand the role of gene expression to predict CYP2D6 phenoconversion may inform the use of PGx testing in the clinical setting.
Collapse
Affiliation(s)
- Erika L Meaddough
- School of Nursing, Healthcare Genetics & Genomics Program, College of Behavioral, Social, and Health Sciences, Clemson University, Clemson, SC, USA.
| | - Sara M Sarasua
- School of Nursing, Healthcare Genetics & Genomics Program, College of Behavioral, Social, and Health Sciences, Clemson University, Clemson, SC, USA
| | - Deborah Kunkel
- School of Mathematical and Statistical Sciences, College of Science, Clemson University, Clemson, SC, USA
| | - Luigi Boccuto
- School of Nursing, Healthcare Genetics & Genomics Program, College of Behavioral, Social, and Health Sciences, Clemson University, Clemson, SC, USA
| | - Satishkumar R Ganakammal
- School of Nursing, Healthcare Genetics & Genomics Program, College of Behavioral, Social, and Health Sciences, Clemson University, Clemson, SC, USA
| | - Matt Moersen
- Research Computing & Data, Clemson Computing and Information Technology, Clemson University, Clemson, SC, USA
| | - Christopher L Farrell
- School of Nursing, Healthcare Genetics & Genomics Program, College of Behavioral, Social, and Health Sciences, Clemson University, Clemson, SC, USA
| |
Collapse
|
23
|
Do LK, Lee HM, Ha YS, Lee CH, Kim J. Amino acids in cancer: Understanding metabolic plasticity and divergence for better therapeutic approaches. Cell Rep 2025; 44:115529. [PMID: 40193251 PMCID: PMC12038367 DOI: 10.1016/j.celrep.2025.115529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 02/24/2025] [Accepted: 03/17/2025] [Indexed: 04/09/2025] Open
Abstract
Metabolic reprogramming is a hallmark of malignant transformation. While initial studies in the field of cancer metabolism focused on central carbon metabolism, the field has expanded to metabolism beyond glucose and glutamine and uncovered the important role of amino acids in tumorigenesis and tumor immunity as energy sources, signaling molecules, and precursors for (epi)genetic modification. As a result of the development and application of new technologies, a multifaceted picture has emerged, showing that context-dependent heterogeneity in amino acid metabolism exists between tumors and even within distinct regions of solid tumors. Understanding the complexity and flexibility of amino acid metabolism in cancer is critical because it can influence therapeutic responses and predict clinical outcomes. This overview discusses the current findings on the heterogeneity in amino acid metabolism in cancer and how understanding the metabolic diversity of amino acids can be translated into more clinically relevant therapeutic interventions.
Collapse
Affiliation(s)
- Linda K Do
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Hyun Min Lee
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Yun-Sok Ha
- Department of Urology, School of Medicine, Kyungpook National University, Kyungpook National University Chilgok Hospital, Daegu 41404, Korea
| | - Chan-Hyeong Lee
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA
| | - Jiyeon Kim
- Department of Urology, Yale School of Medicine, New Haven, CT 06519, USA; Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, CT 06519, USA.
| |
Collapse
|
24
|
Ananthakrishnan AI, Mahin A, Prasad TSK, Abhinand CS. Transcriptome Profiling and Viral-Human Interactome Insights Into HBV-Driven Oncogenic Alterations in Hepatocellular Carcinoma. Microbiol Immunol 2025. [PMID: 40243270 DOI: 10.1111/1348-0421.13219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 03/21/2025] [Accepted: 03/27/2025] [Indexed: 04/18/2025]
Abstract
Hepatocellular carcinoma (HCC) is the primary form of liver cancer that poses a significant global health concern due to its increasing incidence rates and diverse etiology. Chronic infection induced by hepatitis B virus (HBV) is a prominent etiological factor influencing the development of HCC. Although recent advances in multi-omics approaches have facilitated extensive exploration of HCC molecular characteristics, translating the characteristics of subtypes into clinical applications has been challenging due to parameters like limited sample size and complex classifiers for early detection. In the present study, we performed transcriptomics profiling of HBV-infected HCC patient tissue data to gather comprehensive insights into the intricate molecular mechanisms underlying HBV-associated HCC, specifically, viral protein interactions that influence the expression of oncogenes. The 1059 differentially expressed genes (DEGs) identified across two GEO data sets revealed upregulation of cell cycle and mitosis-related genes, alongside downregulation of genes involved in fatty acid degradation and cytochrome P450 activity. CDK1 and CDC20 which are part of the top cluster and hub gene from interactome analysis were identified as potential markers for HBV-positive HCC through gene expression pattern and overall survival analysis. Additionally, 19 DEGs showing significance in HCC development were identified as interacting partners with HBV proteins. Among them, the interaction of HBsAg with ALB and SHBG and their downregulation correlates to the lower testosterone levels identified in HBV and HCC patients. Together, the study enhances the understanding of the heterogeneity and molecular pathogenesis of HBV-positive HCC.
Collapse
Affiliation(s)
- Anilkumar I Ananthakrishnan
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Althaf Mahin
- Centre for Integrative Omics Data Science (CIODS), Yenepoya (Deemed to be University), Mangalore, India
| | | | - Chandran S Abhinand
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
- Department of Virus Genomics, Bioinformatics, and Statistics, Institute of Advanced Virology, Thiruvananthapuram, India
| |
Collapse
|
25
|
Igarashi R, Oda M, Okada R, Yano T, Takahashi S, Pastuhov S, Matano M, Masuda N, Togasaki K, Ohta Y, Sato S, Hishiki T, Suematsu M, Itoh M, Fujii M, Sato T. Generation of human adult hepatocyte organoids with metabolic functions. Nature 2025:10.1038/s41586-025-08861-y. [PMID: 40240606 DOI: 10.1038/s41586-025-08861-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 03/04/2025] [Indexed: 04/18/2025]
Abstract
Proliferating hepatocytes often undergo ductal metaplasia to balance the energy trade-off between cellular functions and replication, hindering the expansion of human adult hepatocytes with functional competency1. Here we demonstrate that the combined activation of Wnt and STAT3 signalling enables long-term self-renewal of human adult hepatocyte organoids. YAP activation facilitates hepatocyte proliferation but commits it towards the biliary duct lineage. By contrast, STAT3 activation by oncostatin M induces hepatocyte proliferation while counteracting ductal metaplasia and maintaining the hepatic identity. Xenotransplanted hepatocyte organoids repopulate the recipient mouse liver and reconstitute the metabolic zonation structure. Upon niche factor removal and hormone supplementation, hepatocyte organoids form cord-like structures with bile canalicular networks and exhibit major liver metabolic functions comparable to those of in vivo hepatocytes. Hepatocyte organoids are amenable to gene editing, prompting functional modelling of inherent metabolic liver diseases. The new culture system offers a promising avenue for developing therapeutic strategies against human liver diseases.
Collapse
Affiliation(s)
- Ryo Igarashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mayumi Oda
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Ryo Okada
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Tomoki Yano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Sirirat Takahashi
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Strahil Pastuhov
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Mami Matano
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Norio Masuda
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Kazuhiro Togasaki
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Ohta
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Saeko Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Takako Hishiki
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Makoto Suematsu
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
- Department of Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Itoh
- JSR-Keio University Medical and Chemical Innovation Center (JKiC), JSR Corporation, Tokyo, Japan
| | - Masayuki Fujii
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan
| | - Toshiro Sato
- Department of Organoid Medicine, Sakaguchi Laboratory, Keio University School of Medicine, Tokyo, Japan.
- Department of Integrated Medicine and Biochemistry, Keio University School of Medicine, Tokyo, Japan.
| |
Collapse
|
26
|
Zhang G, Zhang G, Zhao Y, Wan Y, Jiang B, Wang H. Unveiling the nexus of p53 and PD-L1: insights into immunotherapy resistance mechanisms in hepatocellular carcinoma. Am J Cancer Res 2025; 15:1410-1435. [PMID: 40371157 PMCID: PMC12070102 DOI: 10.62347/brto3272] [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: 01/26/2025] [Accepted: 03/25/2025] [Indexed: 05/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the predominant form of primary liver cancer worldwide, continues to pose a substantial health challenge with limited treatment options for advanced stages. Despite progress in therapies such as surgery, transplantation, and targeted treatments, prognosis remains bleak for many patients. The advent of immunotherapy has revolutionized the landscape of advanced HCC treatment, offering hope for improved outcomes. However, its efficacy is limited, with a modest response rate of approximately 20% as a single-agent therapy, underscoring the urgent need to decipher mechanisms of immunotherapy resistance. Tumor protein 53 gene (TP53), a pivotal tumor suppressor gene, and Programmed death ligand 1 (PD-L1), a crucial immune checkpoint ligand, play central roles in HCC's evasion of immune responses. Understanding how tumor protein 53 (p53) influences PD-L1 expression and immune system interactions is essential for unraveling the complexities of immunotherapy resistance mechanisms. Elucidating these molecular interactions not only enhances our understanding of HCC's underlying mechanisms but also lays the foundation for developing targeted treatments that may improve outcomes for patients with advanced-stage liver cancer. Ultimately, deciphering the nexus of p53 and PD-L1 in immunotherapy resistance promises to advance treatment strategies and outcomes in the challenging landscape of HCC. This review delves into the intricate relationship between p53 and PD-L1 concerning immunotherapy resistance in HCC, offering insights that could pave the way for novel therapeutic strategies aimed at enhancing treatment efficacy and overcoming resistance in advanced stages of the disease.
Collapse
Affiliation(s)
- Guoyuan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Gan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yixuan Zhao
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Yunyan Wan
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Bin Jiang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| | - Huaxiang Wang
- Department of Hepatobiliary and Pancreatic Surgery, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei Province, China
- Department of Hepatobiliary and Pancreatic Surgery, Hubei Provincial Clinical Research Center for Precision Diagnosis and Treatment of Liver Cancer, Taihe Hospital, Hubei University of MedicineShiyan 442000, Hubei, China
| |
Collapse
|
27
|
Yang H, Chen Y, Zhou Z, Wang Y, Li P, Li Y. Integrating multi-omics and experimental techniques to decode ubiquitinated protein modifications in hepatocellular carcinoma. Front Pharmacol 2025; 16:1545472. [PMID: 40290433 PMCID: PMC12022440 DOI: 10.3389/fphar.2025.1545472] [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: 12/15/2024] [Accepted: 03/21/2025] [Indexed: 04/30/2025] Open
Abstract
Background Ubiquitination, a critical post-translational modification, plays a pivotal role in regulating protein stability and activity, influencing various aspects of cancer development, including metabolic reprogramming, immune evasion, and tumor progression. However, the specific role of ubiquitination in hepatocellular carcinoma (HCC), particularly in relation to the tumor microenvironment (TME), remains poorly understood. This study aims to systematically explore the role of ubiquitination in shaping the TME of HCC, with a focus on its impact on cancer progression and immune modulation. Methods We performed bioinformatics analysis by integrating multiple publicly available HCC datasets to assess the ubiquitination status across various cell types in the TME, including plasma cells, fibroblasts, endothelial cells, and epithelial-mesenchymal transition (EMT) cells. Ubiquitination scores were calculated to categorize these cell types, and survival data, along with spatial transcriptomics, were employed to evaluate how different levels of ubiquitination influence HCC progression. In vitro experiments, such as transwell, CCK8, and wound healing assays, were used to further investigate the role of the key ubiquitination gene UBE2C in HCC phenotypes. Results Our study revealed that ubiquitination-related genes are significantly upregulated in HCC tissues, with high expression levels correlating with poor prognosis in patients. Pathway analysis showed that these genes are enriched in key processes such as cell cycle regulation, DNA repair, metabolic reprogramming, and p53 signaling. These pathways contribute to the TME by promoting tumor cell proliferation, facilitating matrix remodeling, and enhancing angiogenesis. Notably, UBE2C, a critical ubiquitination enzyme, appears to play a key role in immune evasion, potentially by inhibiting anti-tumor immune responses and reducing the immune system's ability to recognize and eliminate tumor cells. Furthermore, experimental data confirmed that UBE2C overexpression promotes HCC cell proliferation, invasion, and metastasis, further supporting its role in tumor progression and TME remodeling. Conclusion This study reveals the multifaceted regulatory roles of ubiquitination in HCC. Ubiquitination not only supports proliferation and anti-apoptotic functions within tumor cells but also promotes tumor progression by modulating the activity of immune and stromal cells. Among all ubiquitination-related genes, UBE2C emerges as a potential prognostic biomarker and therapeutic target in HCC, offering new directions for precision treatment of HCC in the future.
Collapse
Affiliation(s)
- Haikun Yang
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Yuan Chen
- Department of Geriatric Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
- Department of Geriatrics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Zheng Zhou
- The Hepatobiliary and Pancreatic Disease Area Department of Shanxi Provincial People Hospital, Taiyuan, China
| | - Yanjing Wang
- Department of Gastroenterology, Shanxi Provincial People’s Hospital, Taiyuan, China
| | - Peng Li
- The Hepatobiliary and Pancreatic Disease Area Department of Shanxi Provincial People Hospital, Taiyuan, China
| | - Yang Li
- Department of Geriatric Medicine, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Third Hospital of Shanxi Medical University, Tongji Shanxi Hospital, Taiyuan, China
| |
Collapse
|
28
|
Yan F, G. Telonis A, Yang Q, Jiang L, E. Garrett-Bakelman F, Sekeres MA, Santini V, Ceccarelli M, Goel N, Garcia-Martinez L, Morey L, Figueroa ME, Guo Y. Genome-wide methylome modeling via generative AI incorporating long- and short-range interactions. SCIENCE ADVANCES 2025; 11:eadt4152. [PMID: 40215314 PMCID: PMC11988400 DOI: 10.1126/sciadv.adt4152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/05/2025] [Indexed: 04/14/2025]
Abstract
Using millions of methylation segments, we developed DiffuCpG, a generative artificial intelligence (AI) diffusion model designed to solve the critical challenge of missing data in high-throughput methylation technologies. DiffuCpG goes beyond conventional methods by leveraging both short-range interactions including nearby CpGs from both latitude and longitude of the dataset, local DNA sequences, and long-range interactions, including three-dimensional genome architecture and long-distance correlations, to comprehensively model the methylome. Compared to previous methods, through extensive independent validations across different tissue types, cancers, and technologies (whole-genome bisulfite sequencing, enhanced reduced representation bisulfite sequencing, single-cell bisulfite sequencing, and methylation arrays), DiffuCpG has demonstrated superior performance in accuracy, scalability, and versatility. On average, bisulfite sequencing dataset, DiffuCpG can extend the original dataset by millions of additional CpGs. As an alternative application of generative AI, DiffuCpG addresses a key bottleneck in epigenetic research and will substantially benefit studies relying on high-throughput methylation data.
Collapse
Affiliation(s)
- Fengyao Yan
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Aristeidis G. Telonis
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Qin Yang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Limin Jiang
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Francine E. Garrett-Bakelman
- Department of Biochemistry and Molecular Genetics, University of Virginia, Charlottesville, VA 22908, USA
- Department of Medicine, University of Virginia, Charlottesville, VA 22908, USA
- Comprehensive Cancer Center, University of Virginia, Charlottesville, VA 22908, USA
| | - Mikkael A. Sekeres
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Hematology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Valeria Santini
- MDS Unit, DMSC, University of Florence, AOU Careggi, Florence 50134, Italy
| | - Michele Ceccarelli
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Neha Goel
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Surgery, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Liliana Garcia-Martinez
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Lluis Morey
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Human Genetics, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Maria E. Figueroa
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Biochemistry and Molecular Biology, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Division of Hematology, Department of Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA
| | - Yan Guo
- Sylvester Comprehensive Cancer Center, University of Miami Miller School of Medicine, Miami, FL 33136, USA
- Department of Public Health and Sciences, University of Miami, Miami, FL 33136, USA
| |
Collapse
|
29
|
Lu J, Liu X, Fan K, Lin X. Traditional Chinese Medicine as a Tool for the Treatment of Hepatocellular Carcinoma by Targeting Pathophysiological Mechanism. Cancer Manag Res 2025; 17:779-792. [PMID: 40225699 PMCID: PMC11993174 DOI: 10.2147/cmar.s513729] [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: 12/22/2024] [Accepted: 03/20/2025] [Indexed: 04/15/2025] Open
Abstract
Liver cancer is a significant global health concern, with projections indicating that the incidence of morbidity may surpass one million cases by 2025. Hepatocellular carcinoma (HCC) is the predominant subtype of liver cancer, constituting approximately 90% of all liver cancer diagnoses. Infections caused by the hepatitis B virus (HBV) and hepatitis C virus (HCV) are recognized as primary risk factors for the development of HCC. However, non-alcoholic steatohepatitis (NASH), which is often linked to metabolic syndrome or diabetes, is increasingly being recognized as a prevalent risk factor in Western populations. Furthermore, HCC associated with NASH exhibits distinct molecular pathogenesis. Patients diagnosed with HCC have access to a range of therapeutic interventions, including liver transplantation, surgical resection, percutaneous ablation, radiation therapy, and transarterial and systemic therapies. Consequently, effective clinical decision-making requires a multidisciplinary approach to adapt individualized treatment plans based on the patient's tumor stage, liver function, and overall performance status. The approval of new first- and second-line pharmacological agents, along with the establishment of immune checkpoint inhibitor therapies as standard treatment modalities, has contributed to an improved prognosis for patients with HCC. Nevertheless, the optimal sequencing of these therapeutic agents remains to be elucidated, highlighting the urgent need for predictive biomarkers to inform treatment selections. Traditional Chinese Medicine (TCM) has demonstrated potential as a complementary and alternative therapeutic approach for liver cancer, warranting further investigation. This review aimed to examine the comprehensive treatment of HCC through the lens of TCM, informed by the current understanding of its epidemiology, diagnosis, and pathophysiology.
Collapse
Affiliation(s)
- Jialin Lu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Xiaoyu Liu
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Kaiyan Fan
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| | - Xiaofeng Lin
- Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, People’s Republic of China
| |
Collapse
|
30
|
Zezulinski D, Hoteit MA, Kaplan DE, Simeone A, Zhan T, Doria C, Ahmed FY, Roberts LR, Block TM, Sayeed A. Detection of Circulating mRNA Variants in Hepatocellular Carcinoma Patients Using Targeted RNAseq. Liver Cancer 2025:1-32. [PMID: 40331063 PMCID: PMC12052365 DOI: 10.1159/000545366] [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: 09/06/2024] [Accepted: 03/02/2025] [Indexed: 05/08/2025] Open
Abstract
Introduction Mutations in circulating nucleic acids can be used as biomarkers for the early detection and management of hepatocellular carcinoma (HCC). However, while circulating tumor DNA and microRNA have been extensively explored, circulating tumor mRNA and circulating mRNA mutants (ctmutRNA), which may provide advantages over other analytes, remain less well described. We previously reported the identification of 288 HCC selective ctmutRNA variants, called "candidates," from a small cohort of HCC patients using total RNAseq. The objective of the current study was to use targeted RNAseq to validate the specificity and sensitivity of these HCC selective variants in an independent cohort of patients with liver cirrhosis (LC). Methods Several methods to isolate small extracellular vesicles and amplify mRNA from the circulation were compared. RNA was isolated, and the primers and probes selective for the 288 regions of interest were used with RNA from HCC (N = 50) and LC and no HCC (N = 35) patients. HCC tumor tissues (N = 11), a normal liver tissue and 3 cell lines were also studied. cDNA synthesis was followed by library construction using QIAseq RNA Fusion XP panel. QC analysis was carried out with an Agilent Bioanalyzer before sequencing on a NextSeq 550 instrument. A GATK HaplotypeCaller was used for variant calling and annotation carried out using snpEff. Results Among the test panel of 288 ctmutRNA candidates in the original cohort, 75 were detected in the new cohort of plasma samples. Moreover, 388 other variants in proximity to the original lesions were also found in multiple HCC but not LC plasma samples. A subset of 36 HCC selective variants was able to identify all HCC patients. The most common tumor specific variants were Indels and SNPs. Novel mRNA fusion variants, corresponding to SENP7, HYI, SAR1A, RASA2, TUBA transcripts, etc., were identified in HCC and LC patients. Conclusion Circulating RNA could be a robust analyte for noninvasive early detection of HCC and circulating RNA panels could be powerful tools in the entire spectrum of clinical management.
Collapse
Affiliation(s)
| | - Maarouf A. Hoteit
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
| | - David E. Kaplan
- Division of Gastroenterology and Hepatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA, USA
- The Corporal Michael J. Crescenz Veterans Administration Hospital, Philadelphia, PA, USA
| | | | - Tingting Zhan
- Division of Biostatistics, Department of Pharmacology and Experimental Therapeutics, Thomas Jefferson University, Philadelphia, PA, USA
| | | | - Fowsiyo Y. Ahmed
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | - Lewis R. Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, MN, USA
| | | | - Aejaz Sayeed
- Baruch S. Blumberg Institute, Doylestown, PA, USA
| |
Collapse
|
31
|
Luo Q, Teschendorff AE. Cell-type-specific subtyping of epigenomes improves prognostic stratification of cancer. Genome Med 2025; 17:34. [PMID: 40181447 PMCID: PMC11967111 DOI: 10.1186/s13073-025-01453-5] [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: 08/15/2024] [Accepted: 03/10/2025] [Indexed: 04/05/2025] Open
Abstract
BACKGROUND Most molecular classifications of cancer are based on bulk-tissue profiles that measure an average over many distinct cell types. As such, cancer subtypes inferred from transcriptomic or epigenetic data are strongly influenced by cell-type composition and do not necessarily reflect subtypes defined by cell-type-specific cancer-associated alterations, which could lead to suboptimal cancer classifications. METHODS To address this problem, we here propose the novel concept of cell-type-specific combinatorial clustering (CELTYC), which aims to group cancer samples by the molecular alterations they display in specific cell types. We illustrate this concept in the context of DNA methylation data of liver and kidney cancer, deriving in each case novel cancer subtypes and assessing their prognostic relevance against current state-of-the-art prognostic models. RESULTS In both liver and kidney cancer, we reveal improved cell-type-specific prognostic models, not discoverable using standard methods. In the case of kidney cancer, we show how combinatorial indexing of epithelial and immune-cell clusters define improved prognostic models driven by synergy of high mitotic age and altered cytokine signaling. We validate the improved prognostic models in independent datasets and identify underlying cytokine-immune-cell signatures driving poor outcome. CONCLUSIONS In summary, cell-type-specific combinatorial clustering is a valuable strategy to help dissect and improve current prognostic classifications of cancer in terms of the underlying cell-type-specific epigenetic and transcriptomic alterations.
Collapse
Affiliation(s)
- Qi Luo
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China
| | - Andrew E Teschendorff
- CAS Key Laboratory of Computational Biology, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, 320 Yue Yang Road, Shanghai, 200031, China.
| |
Collapse
|
32
|
Hu H, Ning S, Liu F, Zhang Z, Zeng W, Liu Y, Liao Z, Zhang H, Zhang Z. Hafnium Metal-Organic Framework-Based Glutamine Metabolism Disruptor For Potentiating Radio-Immunotherapy in MYC-Amplified Hepatocellular Carcinoma. ACS APPLIED MATERIALS & INTERFACES 2025; 17:19367-19381. [PMID: 40116395 DOI: 10.1021/acsami.4c21998] [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: 03/23/2025]
Abstract
Hepatocellular carcinoma (HCC) with MYC oncogene amplification remains a serious challenge in clinical practice. Recent advances in comprehensive treatment strategies, particularly the combination of radiotherapy and immunotherapy, offer new hope. To further improve efficacy while lowering radiation doses, nanopharmaceuticals based on high-Z elements have been extensively studied in radio-immunotherapy. In this work, a hafnium-based metal-organic framework (Hf-MOF), UiO-66-Hf(2OH)-CB-839/BSO@HA (UiO-66-Hf(2OH)-C/B@HA), was designed to codeliver telaglenastat (CB-839) and buthionine sulfoximine (BSO), which synergistically inhibited glutamine metabolism and alleviated tumor hypoxia. Further modification with hyaluronic acid (HA) enhanced tumor targeting, ultimately strengthening the efficacy of radiotherapy in MYC-amplified HCC. Beyond increasing reactive oxygen species (ROS) generation, promoting DNA damage, and inducing tumor apoptosis, more importantly, UiO66-Hf(2OH)-C/B@HA triggered immunogenic cell death (ICD), driving the antitumor immune response. Combination with immune checkpoint blockade (ICB) further enhanced the efficacy, accompanied by increased infiltration of T cells with high granzyme B expression (GZMB+ T cells) within the tumor microenvironment (TME). In the orthotopic HCC model, established with MYC-amplified tumor cells, intravenous administration of UiO66-Hf(2OH)-C/B@HA significantly potentiated the efficacy of radio-immunotherapy, resulting in superior tumor regression. In summary, our study provides insights into the design of Hf-MOF for radio-immunotherapy and proposes a promising therapeutic approach for MYC-amplified HCC.
Collapse
Affiliation(s)
- Haofan Hu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Shangwu Ning
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Furong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Ze Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Weifeng Zeng
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Yachong Liu
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Zhibin Liao
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Hongwei Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| | - Zhanguo Zhang
- Hepatic Surgery Center, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei, China
- Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Wuhan 430030, Hubei, China
- Key Laboratory of Organ Transplantation, Ministry of Education, Chinese Academy of Medical Sciences; NHC Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, Wuhan 430030, China
| |
Collapse
|
33
|
Chen YZ, Meng ZS, Zhang YN, Xiang ZL. Natural Killer Cell-Associated Radiogenomics Model for Hepatocellular Carcinoma: Integrating CD2 and Enhanced CT-Derived Radiomics Signatures. Acad Radiol 2025; 32:1981-1992. [PMID: 39542805 DOI: 10.1016/j.acra.2024.10.043] [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: 09/14/2024] [Revised: 10/13/2024] [Accepted: 10/24/2024] [Indexed: 11/17/2024]
Abstract
RATIONALE AND OBJECTIVES Hepatocellular carcinoma (HCC) is a leading cause of cancer mortality. Natural Killer (NK) cells play a crucial role in immune defense against HCC, but their activity is often impaired by the tumor microenvironment (TME). This study aims to integrate radiomics and transcriptomics to develop a prognostic model linking NK cell characteristics to clinical outcomes in HCC. METHODS Transcriptomic data from five cohorts (734 HCC patients) from the Gene Expression Omnibus and The Cancer Genome Atlas databases were analyzed using the Microenvironment Cell Populations-counter algorithm. NK cell-related prognostic biomarkers were identified via weighted gene co-expression network analysis and LASSO-Cox regression. Radiomics models were established using CT imaging features from 239 patients in three datasets from The Cancer Imaging Archive and Shanghai East Hospital. HCC radiogenomic subtypes were proposed by integrating genetic biomarkers and radiomics models. RESULTS CD2 expression was identified as an independent NK cell-related prognostic biomarker, with a positive impact on prognosis and a strong correlation with NK cell-associated biological processes in HCC. A robust radiomics model was constructed, and the integration of CD2 expression with radioscore identified potential radiogenomic subtypes of HCC. CONCLUSION Radiomics has potential to link TME immune phenotypes with HCC prognosis. CD2 is a key biomarker connecting NK cells with radiomic features, offering a new classification of HCC into radiogenomic subtypes. This approach supports the use of radiogenomics in personalized HCC treatment.
Collapse
Affiliation(s)
- Yan-Zhu Chen
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.Z.C., Z.L.X.)
| | - Zhi-Shang Meng
- Department of Ophthalmology, The Second Xiangya Hospital, Central South University, Changsha, China (Z.S.M.)
| | - Yan-Nan Zhang
- Department of Radiology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.N.Z.)
| | - Zuo-Lin Xiang
- Department of Radiation Oncology, Shanghai East Hospital, School of Medicine, Tongji University, Shanghai, China (Y.Z.C., Z.L.X.); Department of Radiation Oncology, Shanghai East Hospital Ji'an Hospital, Ji'an, China (Z.L.X.).
| |
Collapse
|
34
|
Gao F, Shah R, Xin G, Wang R. Metabolic Dialogue Shapes Immune Response in the Tumor Microenvironment. Eur J Immunol 2025; 55:e202451102. [PMID: 40223597 PMCID: PMC11995254 DOI: 10.1002/eji.202451102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/20/2025] [Accepted: 03/24/2025] [Indexed: 04/15/2025]
Abstract
The fate of immune cells is fundamentally linked to their metabolic program, which is also influenced by the metabolic landscape of their environment. The tumor microenvironment represents a unique system for intercellular metabolic interactions, where tumor-derived metabolites suppress effector CD8+ T cells and promote tumor-promoting macrophages, reinforcing an immune-suppressive niche. This review will discuss recent advancements in metabolism research, exploring the interplay between various metabolites and their effects on immune cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Fengxia Gao
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Rushil Shah
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| | - Gang Xin
- Department of Microbial Infection and ImmunityPelotonia Institute for Immuno‐OncologyThe Ohio State UniversityColumbusOhioUSA
| | - Ruoning Wang
- Center for Childhood Cancer ResearchHematology/Oncology & BMTAbigail Wexner Research Institute at Nationwide Children's HospitalDepartment of PediatricsThe Ohio State UniversityColumbusOhioUSA
| |
Collapse
|
35
|
Lai YJ, Wang LJ, Yasaka TM, Shin Y, Ning M, Tan Y, Shih CH, Guo Y, Chen PY, Galloway H, Liu Z, Das A, Tseng GC, Monga SP, Huang Y, Chiu YC. Inferring Drug-Gene Relationships in Cancer Using Literature-Augmented Large Language Models. CANCER RESEARCH COMMUNICATIONS 2025; 5:706-718. [PMID: 40293950 PMCID: PMC12036822 DOI: 10.1158/2767-9764.crc-25-0030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Revised: 03/17/2025] [Accepted: 04/15/2025] [Indexed: 04/30/2025]
Abstract
SIGNIFICANCE This study presents a novel approach that integrates LLMs with real-time biomedical literature to uncover drug-gene relationships, transforming how cancer researchers identify therapeutic targets, repurpose drugs, and interpret complex molecular interactions. GeneRxGPT, our user-friendly tool, enables researchers to leverage this approach without requiring computational expertise.
Collapse
Affiliation(s)
- Ying-Ju Lai
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Li-Ju Wang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Tyler M. Yasaka
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Medical Scientist Training Program, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yuna Shin
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Electrical and Computer Engineering, Carnegie Mellon University, Pittsburgh, Pennsylvania
| | - Michael Ning
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Computer Science, The University of Texas at Austin, Austin, Texas
| | - Yanhao Tan
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Chien-Hung Shih
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yibing Guo
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Human Genetics, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Po-Yuan Chen
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Hugh Galloway
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Zhentao Liu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Arun Das
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - George C. Tseng
- Department of Biostatistics and Health Data Science, School of Public Health, University of Pittsburgh, Pittsburgh, Pennsylvania
| | - Satdarshan P. Monga
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Organ Pathobiology and Therapeutics Institute, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yufei Huang
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Electrical and Computer Engineering, Swanson School of Engineering, University of Pittsburgh, Pittsburgh, Pennsylvania
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| | - Yu-Chiao Chiu
- UPMC Hillman Cancer Center, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
- Department of Computational and Systems Biology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania
| |
Collapse
|
36
|
Su H, Zhou X, Lin G, Luo C, Meng W, Lv C, Chen Y, Wen Z, Li X, Wu Y, Xiao C, Yang J, Lu J, Luo X, Chen Y, Tam PKH, Li C, Sun H, Pan X. Deciphering the Oncogenic Landscape of Hepatocytes Through Integrated Single-Nucleus and Bulk RNA-Seq of Hepatocellular Carcinoma. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412944. [PMID: 39960344 PMCID: PMC11984907 DOI: 10.1002/advs.202412944] [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: 10/14/2024] [Revised: 01/01/2025] [Indexed: 04/12/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major cause of cancer-related mortality, while the hepatocyte mechanisms driving oncogenesis remains poorly understood. In this study, single-nucleus RNA sequencing of samples from 22 HCC patients revealed 10 distinct hepatocyte subtypes, including beneficial Hep0, predominantly malignant Hep2, and immunosuppressive Hep9. These subtypes were strongly associated with patient prognosis, confirmed in TCGA-LIHC and Fudan HCC cohorts through hepatocyte composition deconvolution. A quantile-based scoring method is developed to integrate data from 29 public HCC datasets, creating a Quantile Distribution Model (QDM) with excellent diagnostic accuracy (Area Under the Curve, AUC = 0.968-0.982). QDM was employed to screen potential biomarkers, revealing that PDE7B functions as a key gene whose suppression promotes HCC progression. Guided by the genes specific to Hep0/2/9 subtypes, HCC is categorized into metabolic, inflammatory, and matrix classes, which are distinguishable in gene mutation frequencies, survival times, enriched pathways, and immune infiltration. Meanwhile, the sensitive drugs of the three HCC classes are identified, namely ouabain, teniposide, and TG-101348. This study presents the largest single-cell hepatocyte dataset to date, offering transformative insights into hepatocarcinogenesis and a comprehensive framework for advancing HCC diagnostics, prognostics, and personalized treatment strategies.
Collapse
Affiliation(s)
- Huanhou Su
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xuewen Zhou
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Guanchuan Lin
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Chaochao Luo
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- College of Life SciencesShihezi UniversityShiheziXinjiang832003China
| | - Wei Meng
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Cui Lv
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Yuting Chen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Zebin Wen
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Xu Li
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Yongzhang Wu
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Changtai Xiao
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
| | - Jian Yang
- Department of Hepatobiliary Surgery IGeneral Surgery Center and Guangdong Provincial Clinical and Engineering Center of Digital MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Jiameng Lu
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Xingguang Luo
- Department of PsychiatryYale University School of MedicineNew HavenCT06510USA
| | - Yan Chen
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Paul KH Tam
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
| | - Chuanjiang Li
- Division of Hepatobiliopancreatic SurgeryDepartment of General SurgeryNanfang HospitalSouthern Medical UniversityGuangzhouGuangdong510515China
| | - Haitao Sun
- Clinical Biobank CenterMicrobiome Medicine CenterDepartment of Laboratory MedicineGuangdong Provincial Clinical Research Center for Laboratory MedicineZhujiang HospitalSouthern Medical UniversityGuangzhou510280China
| | - Xinghua Pan
- Department of Biochemistry and Molecular BiologySchool of Basic Medical SciencesSouthern Medical University and Guangdong Provincial Key Laboratory of Single Cell Technology and ApplicationGuangzhou510515China
- Precision Regenerative Medicine Research CentreMedical Science Divisionand State Key Laboratory of Quality Research in Chinese MedicineMacau University of Science and TechnologyMacao999078China
- Key Laboratory of Infectious Diseases Research in South China (China Ministry Education)Southern Medical UniversityGuangzhouGuangdong510515China
| |
Collapse
|
37
|
Ajouaou Y, Sadler KC. Uncovering epigenetic heterogeneity in HCC. Hepatology 2025; 81:1120-1122. [PMID: 39292862 DOI: 10.1097/hep.0000000000001097] [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: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/20/2024]
Affiliation(s)
- Yousra Ajouaou
- Program in Biology, Center for Genomics and Systems Biology, NYU Abu Dhabi, Abu Dhabi, UAE
| | | |
Collapse
|
38
|
Brzozowska N, Wu LYD, Khodzhaeva V, Griffiths WJ, Duckworth A, Jung H, Coorens THH, Hooks Y, Chambers JE, Campbell PJ, Marciniak SJ, Hoare M. Selection for somatic escape variants in SERPINA1 in the liver of patients with alpha-1 antitrypsin deficiency. Nat Genet 2025; 57:875-883. [PMID: 40065168 PMCID: PMC11985350 DOI: 10.1038/s41588-025-02125-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Accepted: 02/11/2025] [Indexed: 03/28/2025]
Abstract
Somatic variants accumulate in non-malignant tissues with age. Functional variants, leading to clonal advantage of hepatocytes, accumulate in the liver of patients with acquired chronic liver disease (CLD). Whether somatic variants are common to CLD from differing etiologies is unknown. We analyzed liver somatic variants in patients with genetic CLD from alpha-1 antitrypsin (A1AT) deficiency or hemochromatosis. We show that somatic variants in SERPINA1, the gene encoding A1AT, are strongly selected for in A1AT deficiency, with evidence of convergent evolution. Acquired SERPINA1 variants are clustered at the carboxyl terminus of A1AT, leading to truncation. In vitro and in vivo, C-terminal truncation variants reduce disease-associated Z-A1AT polymer accumulation and disruption of the endoplasmic reticulum, supporting the C-terminal domain swap mechanism. Therefore, somatic escape variants from a deleterious germline variant are selected for in A1AT deficiency, suggesting that functional somatic variants are disease-specific in CLD and point to disease-associated mechanisms.
Collapse
Affiliation(s)
| | - Lily Y D Wu
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | - Vera Khodzhaeva
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Cambridge, UK
| | | | - Adam Duckworth
- Department of Pathology, Addenbrooke's Hospital, Cambridge, UK
| | | | - Tim H H Coorens
- Wellcome Trust Sanger Institute, Hinxton, UK
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | | | - Joseph E Chambers
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | | | - Stefan J Marciniak
- Cambridge Institute for Medical Research, Keith Peters Building, Cambridge Biomedical Campus, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Matthew Hoare
- Department of Medicine, University of Cambridge, Cambridge, UK.
- Early Cancer Institute, University of Cambridge, Cambridge, UK.
| |
Collapse
|
39
|
Akarca FG, Grenert JP, Kakar S. Role of genomic analysis in the classification of well differentiated hepatocellular lesions. Hum Pathol 2025; 158:105794. [PMID: 40374146 DOI: 10.1016/j.humpath.2025.105794] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/05/2025] [Accepted: 05/12/2025] [Indexed: 05/17/2025]
Abstract
BACKGROUND The distinction of focal nodular hyperplasia (FNH) and hepatocellular adenoma (HCA) from well-differentiated hepatocellular carcinoma (WD-HCC) in noncirrhotic liver can be challenging. High-grade dysplastic nodule (HGDN) in cirrhosis can have overlapping features with WD-HCC. In some cases, HCA diagnosis is evident but glutamine synthetase (GS) staining is indeterminate for β-catenin activation, which does not allow reliable risk assessment. This study examines the role of genomic analysis in better categorization of WD hepatocellular lesions (WDHL). DESIGN Genomic analysis using capture-based NGS assay was done in 23 WDHLs that could not be definitely classified based on morphology, reticulin stain and IHC, and were designated as 'atypical hepatocellular neoplasms' (AHNs). GS staining was classified as diffuse homogeneous (moderate to strong staining in >90 % of tumor cells), diffuse heterogeneous (50-90 %), not diffuse (<50 %) and borderline (not clear if more or less than 50 %). RESULTS The genomic profile provided additional information for the diagnosis and/or risk assessment enabling a benign diagnosis in 15/23 cases (66 %) and HCC in 4/23 cases (17 %), while the diagnosis remained as atypical in the remaining 4 cases. Of the 4 cases with final HCC diagnosis, findings were suspicious but not diagnostic based on morphology/IHC; additional changes like TERT promoter mutation (n = 2), AXIN mutation (n = 1), CDKN2A loss (n = 2) and copy number alterations (n = 3) helped to support HCC. Of the 15 cases with a final benign diagnosis, the status of β-catenin activation was unclear based on GS stain in 8 cases, 2 of which showed CTNNB1 exon 7 mutation, 1 showed CTNNB1 exon 8 mutation, while genomic changes in 5 cases did not show any evidence of Wnt activation. FNH-like features were seen in 2 cases, but the genomic changes excluded FNH (CTNNB1 and ARID1A mutation). The final diagnosis was unchanged from the initial diagnosis of AHN in 4/23 cases (17 %) as the molecular findings did not favor HCC. CONCLUSION Genomic changes were helpful in characterization of WDHLs, supporting HCC in 17 % of cases and clarifying β-catenin activation status in all 7 cases with borderline GS staining. Genomic changes are not specific but can provide diagnostic clues in selected challenging cases that cannot be classified on morphology and IHC. Given the significant treatment implications of distinguishing between HCC and benign/premalignant entities, routine use of genomic analysis in diagnostically challenging settings should be considered.
Collapse
MESH Headings
- Humans
- Liver Neoplasms/genetics
- Liver Neoplasms/pathology
- Liver Neoplasms/classification
- Liver Neoplasms/diagnosis
- Liver Neoplasms/chemistry
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/classification
- Carcinoma, Hepatocellular/chemistry
- Carcinoma, Hepatocellular/diagnosis
- Adenoma, Liver Cell/genetics
- Adenoma, Liver Cell/pathology
- Adenoma, Liver Cell/classification
- Adenoma, Liver Cell/diagnosis
- Adenoma, Liver Cell/chemistry
- Male
- Middle Aged
- Female
- Aged
- Focal Nodular Hyperplasia/genetics
- Focal Nodular Hyperplasia/pathology
- Focal Nodular Hyperplasia/diagnosis
- Focal Nodular Hyperplasia/classification
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/analysis
- Adult
- Genomics/methods
- Diagnosis, Differential
- Glutamate-Ammonia Ligase/analysis
- beta Catenin/genetics
- Mutation
- High-Throughput Nucleotide Sequencing
- Predictive Value of Tests
- Cell Differentiation
Collapse
|
40
|
Pinto E, Lazzarini E, Pelizzaro F, Gambato M, Santarelli L, Potente S, Zanaga P, Zappitelli T, Cardin R, Burra P, Farinati F, Romualdi C, Boscarino D, Tosello V, Indraccolo S, Russo FP. Somatic Copy Number Alterations in Circulating Cell-Free DNA as a Prognostic Biomarker for Hepatocellular Carcinoma: Insights from a Proof-of-Concept Study. Cancers (Basel) 2025; 17:1115. [PMID: 40227625 PMCID: PMC11988118 DOI: 10.3390/cancers17071115] [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: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite advances in hepatocellular carcinoma (HCC) management, prognosis remains poor. Advanced-stage diagnosis often excludes curative treatments, and current biomarkers (e.g., alpha-fetoprotein [AFP]) have limited utility in early detection. Liquid biopsy has emerged as a promising cancer detection tool, with circulating cell-free DNA (ccfDNA) showing significant diagnostic potential. This proof-of-concept study aimed to investigate the potential role of tumor fraction (TF) within ccfDNA as a biomarker in HCC patients. METHODS A total of sixty patients were recruited, including thirteen with chronic liver disease (CLD), twenty-four with cirrhosis, and twenty-three with HCC. Plasma samples were collected, and ccfDNA was extracted for shallow whole genome sequencing (sWGS) analysis. The TF was calculated by focusing on somatic copy number alterations (SCNAs) within the ccfDNA. RESULTS Among patients with CLD and cirrhosis (n = 37), ctDNA was undetectable in all but one cirrhotic patient who exhibited a significant tumor fraction (TF) of 17% and subsequently developed HCC. Conversely, five out of twenty-three HCC patients (21.7%) displayed detectable ctDNA with TF levels ranging from 3.0% to 32.6%. Patients with detectable ctDNA were characterized by more aggressive oncological features, including a higher number of nodules (p = 0.005), advanced-stage disease (60% BCLC C, p = 0.010), and poorer response to therapy (80% PD, p = 0.001). Moreover, the overall survival (OS) was significantly reduced in patients with detectable ctDNA (median OS: 17 months; CI 95% 4.5-26.5) compared to those without (median OS: 24.0 months; CI 95% 7.0-66.0; log-rank p = 0.002). CONCLUSIONS Our results suggest that the analysis of TF by sWGS is a promising non-invasive tool for the identification of HCC with aggressive clinical behavior, whereas it is not sensitive enough for early HCC detection. This molecular assay can improve prognostic stratification in HCC patients.
Collapse
Affiliation(s)
- Elisa Pinto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Elisabetta Lazzarini
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Martina Gambato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Laura Santarelli
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Sara Potente
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | - Paola Zanaga
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Teresa Zappitelli
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Romilda Cardin
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Patrizia Burra
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Chiara Romualdi
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | | | - Valeria Tosello
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Stefano Indraccolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| |
Collapse
|
41
|
Ramirez CFA, Akkari L. Myeloid cell path to malignancy: insights into liver cancer. Trends Cancer 2025:S2405-8033(25)00054-8. [PMID: 40140328 DOI: 10.1016/j.trecan.2025.02.006] [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: 12/03/2024] [Revised: 02/12/2025] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
Clinically approved treatments for advanced liver cancer often lack potency because of the heterogeneous characteristics of hepatocellular carcinoma (HCC). This complexity is largely driven by context-dependent inflammatory responses brought on by diverse etiologies, such as metabolic dysfunction-associated steatohepatitis (MASH), the genetic makeup of cancer cells, and the versatile adaptability of immune cells, such as myeloid cells. In this review, we discuss the evolutionary dynamics of the immune landscape, particularly that of liver-resident Kupffer cells (KCs), TREM2+, and SPP1+ macrophages with an active role during liver disease progression, which eventually fuels hepatocarcinogenesis. We highlight exploitable immunomodulatory avenues amenable to mitigate both the inherent pathological characteristics of liver cancers and the associated external factors that favor malignancy, paving a roadmap toward improving the management and therapeutic outcome for patients with HCC.
Collapse
Affiliation(s)
- Christel F A Ramirez
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands
| | - Leila Akkari
- Division of Tumor Biology and Immunology, Oncode Institute, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, The Netherlands.
| |
Collapse
|
42
|
Wang B, Huang C, Liu X, Liu Z, Zhang Y, Zhao W, Xu Q, Ho PC, Xiao Z. iMetAct: An integrated systematic inference of metabolic activity for dissecting tumor metabolic preference and tumor-immune microenvironment. Cell Rep 2025; 44:115375. [PMID: 40053454 DOI: 10.1016/j.celrep.2025.115375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Revised: 12/03/2024] [Accepted: 02/10/2025] [Indexed: 03/09/2025] Open
Abstract
Metabolic enzymes play a central role in cancer metabolic reprogramming, and their dysregulation creates vulnerabilities that can be exploited for therapy. However, accurately measuring metabolic enzyme activity in a high-throughput manner remains challenging due to the complex, multi-layered regulatory mechanisms involved. Here, we present iMetAct, a framework that integrates metabolic-transcription networks with an information propagation strategy to infer enzyme activity from gene expression data. iMetAct outperforms expression-based methods in predicting metabolite conversion rates by accounting for the effects of post-translational modifications. With iMetAct, we identify clinically significant subtypes of hepatocellular carcinoma with distinct metabolic preferences driven by dysregulated enzymes and metabolic regulators acting at both the transcriptional and non-transcriptional levels. Moreover, applying iMetAct to single-cell RNA sequencing data allows for the exploration of cancer cell metabolism and its interplay with immune regulation in the tumor microenvironment. An accompanying online platform further facilitates tumor metabolic analysis, patient stratification, and immune microenvironment characterization.
Collapse
Affiliation(s)
- Binxian Wang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Chao Huang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Xuan Liu
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Zhenni Liu
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Yilei Zhang
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Wei Zhao
- Department of General Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China
| | - Qiuran Xu
- Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang 310014, China
| | - Ping-Chih Ho
- Department of Oncology, University of Lausanne, Epalinges, Switzerland; Ludwig Institute for Cancer Research, University of Lausanne, Epalinges, Switzerland.
| | - Zhengtao Xiao
- Institute of Molecular and Translational Medicine, Department of Biochemistry and Molecular Biology, Xi'an Jiaotong University Health Science Center, Xi'an, Shaanxi 710061, China; Key Laboratory of Environment and Disease-Related Genes, Ministry of Education, Xi'an Jiaotong University, Xi'an, Shaanxi 710061, China.
| |
Collapse
|
43
|
Jiang X, Liu J, Wang K, Sun J, Yin H, Jiang Y, Liu Y, Wang N, Ding X, Gao P, Li L, Zhang X, Li J, Zhang R. ASPM mediates nuclear entrapment of FOXM1 via liquid-liquid phase separation to promote progression of hepatocarcinoma. Genome Biol 2025; 26:68. [PMID: 40122889 PMCID: PMC11929996 DOI: 10.1186/s13059-025-03526-5] [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: 04/07/2024] [Accepted: 03/05/2025] [Indexed: 03/25/2025] Open
Abstract
BACKGROUND Fork-head box protein M1 (FOXM1) plays critical roles in development and progression of multiple cancers, including hepatocellular carcinoma (HCC). However, the exact regulatory hierarchy of FOXM1 remains unclear. Here, a genome-wide screen is performed to identify intranuclear proteins that promote FOXM1 transcription activity via liquid-liquid phase separation (LLPS). RESULTS Abnormal spindle-like microcephaly associated (ASPM) is identified to interact with FOXM1 protein via LLPS and enhance its stability by preventing proteasome-mediated degradation. ChIP-sequencing data show ASPM and FOXM1 co-occupy the promoters of multiple genes to promote their transcription, enhancing FOXM1-driven oncogenic progression. In functional experiments, inhibition of ASPM suppresses tumor growth of HCC cells in vivo and in vitro, while overexpression of ASPM has opposite effects. Importantly, reconstitution of FOXM1 partially compensates for the weakened proliferative capacity of HCC cells caused by ASPM silencing. Intriguingly, FOXM1 binds to the promoter region of ASPM and transcriptionally activates ASPM expression in HCC cells. Furthermore, we find that a higher co-expression of ASPM and FOXM1 significantly correlates with poor prognosis in HCC patients. It indicates a double positive feedback loop between ASPM and FOXM1 which coordinately promotes the aggressive progression of HCC. CONCLUSIONS Collectively, we demonstrate that LLPS and transcriptional regulation form an oncogenic double positive feedback loop between ASPM and FOXM1. This provides a rationale strategy to treat HCC by targeting this mechanism.
Collapse
Affiliation(s)
- Xunliang Jiang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Jun Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ke Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
- Department of General Surgery, Central Theater Command General Hospital of the Chinese People's Liberation Army, Wuhan, 430070, China
| | - Jianyong Sun
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
- Department of Thoracic Surgery, Tangdu Hospital, Fourth Military Medical University, Xi'an, 710000, China
| | - Huilong Yin
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
- Henan Key Laboratory of Immunology and Targeted Therapy, School of Laboratory Medicine, Xinxiang Medical University, Xinxiang, 453003, China
| | - Yu Jiang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
- School of Clinical Medicine, Xi'an Medical University, Xi'an, 710000, China
| | - Yongkang Liu
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Ningbo Wang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiaochen Ding
- Department of Experimental Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China
| | - Pu Gao
- CAS Key Laboratory of Infection and Immunity, Institute of Biophysics, National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Chinese Academy of Sciences, Beijing, 100101, China
| | - Lin Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China
| | - Xiang Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
- The Ministry of Education Key Lab of Hazard Assessment and Control in Special Operational Environment, Fourth Military Medical University, Xi'an, 710032, China.
| | - Jipeng Li
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, National Clinical Research Center for Digestive Diseases and Xijing Hospital of Digestive Diseases, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Gastrointestinal Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
- Department of Experimental Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, China.
| | - Rui Zhang
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, 710032, China.
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Immunology, Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
44
|
Li Y, Xiong J, Hu Z, Chang Q, Ren N, Zhong F, Dong Q, Liu L. Denoised recurrence label-based deep learning for prediction of postoperative recurrence risk and sorafenib response in HCC. BMC Med 2025; 23:162. [PMID: 40102873 PMCID: PMC11921616 DOI: 10.1186/s12916-025-03977-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 02/27/2025] [Indexed: 03/20/2025] Open
Abstract
BACKGROUND Pathological images of hepatocellular carcinoma (HCC) contain abundant tumor information that can be used to stratify patients. However, the links between histology images and the treatment response have not been fully unveiled. METHODS We trained and evaluated a model by predicting the prognosis of 287 non-treated HCC patients postoperatively, and further explored the model's treatment response predictive ability in 79 sorafenib-treated patients. Based on prognostic relevant pathological signatures (PPS) extracted from CNN-SASM, which was trained by denoised recurrence label (DRL) under different thresholds, the PPS-based prognostic model was formulated. A total of 78 HCC patients from TCGA-LIHC were used for the external validation. RESULTS We proposed the CNN-SASM based on tumor pathology and extracted PPS. Survival analysis revealed that the PPS-based prognostic model yielded the AUROC of 0.818 and 0.811 for predicting recurrence at 1 and 2 years after surgery, with an external validation reaching 0.713 and 0.707. Furthermore, the predictive ability of the PPS-based prognostic model was superior to clinical risk indicators, and it could stratify patients with significantly different prognoses. Importantly, our model can also stratify sorafenib-treated patients into two groups associated with significantly different survival situations, which could effectively predict survival benefits from sorafenib. CONCLUSIONS Our prognostic model based on pathology deep learning provided a valuable means for predicting HCC patient recurrence condition, and it could also improve patient stratification to sorafenib treatment, which help clinical decision-making in HCC.
Collapse
Affiliation(s)
- Yixin Li
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Ji Xiong
- Department of Pathology, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Zhiqiu Hu
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Qimeng Chang
- Department of Hepatobiliary and Pancreatic Surgery, Minhang Hospital, Fudan University, Shanghai, 201199, China
| | - Ning Ren
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, No. 170 XinSong Road, Minhang, Shanghai, 201199, China.
- Department of Liver Surgery and Transplantation, Liver Cancer Institute, Zhongshan Hospital, Fudan University, No. 180 FengLin Road, Xuhui, Shanghai, 200032, China.
| | - Fan Zhong
- Intelligent Medicine Institute, Fudan University, No.131 DongAn Road, Xuhui, Shanghai, 200032, China.
| | - Qiongzhu Dong
- Key Laboratory of Whole-Period Monitoring and Precise Intervention of Digestive Cancer, Shanghai Municipal Health Commission, Minhang Hospital, Fudan University, No. 170 XinSong Road, Minhang, Shanghai, 201199, China.
| | - Lei Liu
- Intelligent Medicine Institute, Fudan University, No.131 DongAn Road, Xuhui, Shanghai, 200032, China.
| |
Collapse
|
45
|
Tang Y, Hu H, Chen S, Hao B, Xu X, Zhu H, Zhan W, Zhang T, Hu H, Chen G. Multi-omics analysis revealed the novel role of NQO1 in microenvironment, prognosis and immunotherapy of hepatocellular carcinoma. Sci Rep 2025; 15:8591. [PMID: 40074806 PMCID: PMC11903666 DOI: 10.1038/s41598-025-92700-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/03/2025] [Indexed: 03/14/2025] Open
Abstract
NAD(P)H dehydrogenase quinone 1 (NQO1) is overexpressed in various cancers and is strongly associated with an immunosuppressive microenvironment and poor prognosis. In this study, we explored the role of NQO1 in the microenvironment, prognosis and immunotherapy of Hepatocellular carcinoma (HCC) using multi-omics analysis and machine learning. The results revealed that NQO1 was significantly overexpressed in HCC cells. NQO1+HCC cells were correlated with poor prognosis and facilitated tumor-associated macrophages (TAMs) polarization to M2 macrophages. We identified core NQO1-related genes (NRGs) and developed the NRGs-related risk-scores in hepatocellular carcinoma (NRSHC). The comprehensive nomogram integrating NRSHC, age, and pathological tumor-node-metastasis (pTNM) Stage achieved an area under the curve (AUC) above 0.7, demonstrating its accuracy in predicting survival outcomes and immunotherapy responses of HCC patients. High-risk patients exhibited worse prognoses but greater sensitivity to immunotherapy. Additionally, a web-based prediction tool was designed to enhance clinical utility. In conclusion, NQO1 may play a critical role in M2 polarization and accelerates HCC progression. The NRSHC model and accompanying tools offer valuable insights for personalized HCC treatment.
Collapse
Affiliation(s)
- Ya Tang
- School of Public Health, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Haihong Hu
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Siyuan Chen
- Department of Breast and Thyroid Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Bo Hao
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Xuefeng Xu
- Department of Function, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
| | - Hongxia Zhu
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
| | - Wendi Zhan
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China
| | - Taolan Zhang
- Department of Pharmacy, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- School of Pharmacy, Hengyang Medical College, University of South China, 28 Western Changsheng Road, Hengyang, 421001, Hunan, China.
- Research Center for Clinical Trial, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
| | - Hongjuan Hu
- Department of Public Health Service, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421000, Hunan, China.
| | - Guodong Chen
- Department of Hepatobiliary Pancreatic Surgery, The First Affiliated Hospital, Hengyang Medical School, University of South China, Hengyang, 421001, Hunan, China.
- Department of General Surgery, Turpan City People's Hospital, Turpan, 838000, China.
| |
Collapse
|
46
|
Naghdi S, Mishra P, Roy SS, Weaver D, Walter L, Davies E, Antony AN, Lin X, Moehren G, Feitelson MA, Reed CA, Lindsten T, Thompson CB, Dang HT, Hoek JB, Knudsen ES, Hajnóczky G. VDAC2 and Bak scarcity in liver mitochondria enables targeting hepatocarcinoma while sparing hepatocytes. Nat Commun 2025; 16:2416. [PMID: 40069152 PMCID: PMC11897174 DOI: 10.1038/s41467-025-56898-4] [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: 11/12/2023] [Accepted: 02/05/2025] [Indexed: 03/15/2025] Open
Abstract
Differences between normal tissues and invading tumors that allow tumor targeting while saving normal tissue are much sought after. Here we show that scarcity of VDAC2, and the consequent lack of Bak recruitment to mitochondria, renders hepatocyte mitochondria resistant to permeabilization by truncated Bid (tBid), a Bcl-2 Homology 3 (BH3)-only, Bcl-2 family protein. Increased VDAC2 and Bak is found in most human liver cancers and mitochondria from tumors and hepatic cancer cell lines exhibit VDAC2- and Bak-dependent tBid sensitivity. Exploring potential therapeutic targeting, we find that combinations of activators of the tBid pathway with inhibitors of the Bcl-2 family proteins that suppress Bak activation enhance VDAC2-dependent death of hepatocarcinoma cells with little effect on normal hepatocytes. Furthermore, in vivo, combination of S63845, a selective Mcl-1 inhibitor, with tumor-nectrosis factor-related, apoptosis-induncing ligand (TRAIL) peptide reduces tumor growth, but only in tumors expressing VDAC2. Thus, we describe mitochondrial molecular fingerprint that discriminates liver from hepatocarcinoma and allows sparing normal tissue while targeting tumors.
Collapse
Affiliation(s)
- Shamim Naghdi
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Piyush Mishra
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Soumya Sinha Roy
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - David Weaver
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Ludivine Walter
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Erika Davies
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Anil Noronha Antony
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Xuena Lin
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Gisela Moehren
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Mark A Feitelson
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Christopher A Reed
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - Tullia Lindsten
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Craig B Thompson
- Department of Cancer Biology, Abramson Family Cancer Research Institute, University of Pennsylvania, Philadelphia, PA, USA
- Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, NY, 10065, USA
| | - Hien T Dang
- Department of Surgery, Thomas Jefferson University, Philadelphia, PA, USA
| | - Jan B Hoek
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA
| | - Erik S Knudsen
- Department of Cancer Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology and Genomic Medicine and Thomas Jefferson University, Philadelphia, PA, USA.
| |
Collapse
|
47
|
Talubo NDD, Dela Cruz EWB, Fowler PMPT, Tsai PW, Tayo LL. QSAR-Based Drug Repurposing and RNA-Seq Metabolic Networks Highlight Treatment Opportunities for Hepatocellular Carcinoma Through Pyrimidine Starvation. Cancers (Basel) 2025; 17:903. [PMID: 40075750 PMCID: PMC11898721 DOI: 10.3390/cancers17050903] [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: 01/27/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: The molecular heterogeneity and metabolic flexibility of Hepatocellular Carcinoma (HCC) pose significant challenges to the efficacy of systemic therapy for advanced cases. Early screening difficulties often delay diagnosis, leading to more advanced stages at presentation. Combined with the inconsistent responses to current systemic therapies, HCC continues to have one of the highest mortality rates among cancers. Thus, this paper seeks to contribute to the development of systemic therapy options through the consideration of HCC's metabolic vulnerabilities and lay the groundwork for future in vitro studies. Methods: Transcriptomic data were used to calculate single and double knockout options for HCC using genetic Minimal Cut Sets. Furthermore, using QSAR modeling, drug repositioning opportunities were assessed to inhibit the selected genes. Results: Two single knockout options that were also annotated as essential pairs were found within the pyrimidine metabolism pathway of HCC, wherein the knockout of either DHODH or TYMS is potentially disruptive to proliferation. The result of the flux balance analysis and gene knockout simulation indicated a significant decrease in biomass production. Three machine learning algorithms were assessed for their performance in predicting the pIC50 of a given compound for the selected genes. SVM-rbf performed the best on unseen data achieving an R2 of 0.82 for DHODH and 0.81 for TYMS. For DHODH, the drugs Oteseconazole, Tipranavir, and Lusutrombopag were identified as potential inhibitors. For TYMS, the drugs Tadalafil, Dabigatran, Baloxavir Marboxil, and Candesartan Cilexetil showed promise as inhibitors. Conclusions: Overall, this study suggests in vitro testing of the identified drugs to assess their capabilities in inducing pyrimidine starvation on HCC.
Collapse
Affiliation(s)
- Nicholas Dale D. Talubo
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (N.D.D.T.); (E.W.B.D.C.); (P.M.P.T.F.)
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Emery Wayne B. Dela Cruz
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (N.D.D.T.); (E.W.B.D.C.); (P.M.P.T.F.)
- School of Graduate Studies, Mapúa University, Manila 1002, Philippines
| | - Peter Matthew Paul T. Fowler
- School of Chemical, Biological, and Materials Engineering and Sciences, Mapúa University, Manila 1002, Philippines; (N.D.D.T.); (E.W.B.D.C.); (P.M.P.T.F.)
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1203, Philippines
| | - Po-Wei Tsai
- Department of Food Science, National Taiwan Ocean University, Keelung 202, Taiwan;
| | - Lemmuel L. Tayo
- Department of Biology, School of Health Sciences, Mapúa University, Makati 1203, Philippines
| |
Collapse
|
48
|
Shiode Y, Kodama T, Sato Y, Takahashi R, Matsumae T, Shirai K, Doi A, Tahata Y, Hikita H, Tatsumi T, Fukai M, Taketomi A, Ruchirawat M, Wang XW, Takehara T. Folate receptor 1 is a stemness trait-associated diagnostic and prognostic marker for hepatocellular carcinoma. Biomark Res 2025; 13:37. [PMID: 40038575 DOI: 10.1186/s40364-025-00752-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Accepted: 02/25/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) can be classified into several subtypes based on molecular traits, aiding in prognostic stratification. The subtype with a poor prognosis is often associated with stem/progenitor features. This study focused on identifying circulating biomarkers for aggressive HCC. METHODS We searched for secretory proteins whose expression was positively associated with the stem/progenitor markers KRT19, EPCAM, and PROM1 in 2 independent HCC cohorts. Serum folate receptor 1 (FOLR1) levels were measured in 238 chronic liver disease and 247 HCC patients, evaluating their diagnostic and prognostic capabilities. RESULTS FOLR1 was identified as a secretory protein that was positively correlated with all 3 stem/progenitor markers and a poor prognosis in both the discovery and validation cohorts. Higher FOLR1 expression was detected in tumor than nontumor tissues and was associated with aggressive subtypes, and activation of p53, DNA repair, Myc, E2F, and PI3K/AKT/mTOR pathways. Serum FOLR1 levels correlated with tumoral FOLR1 expression in HCC patients and were significantly elevated compared with those in patients with chronic hepatitis or nonliver disease. Serum FOLR1 levels demonstrated diagnostic performance for HCC comparable to that of alpha-fetoprotein (AFP), and their combination increased the diagnostic accuracy. Elevated serum FOLR1 levels were associated with poor prognosis in HCC patients, regardless of treatment, especially in patients with early-stage disease. The multivariate analysis revealed that the serum FOLR1 level and the Gender, Age, AFP-L3, AFP, and Des-gamma-carboxy prothrombin (GALAD) score were independent predictors of a poor prognosis with their combination further stratifying prognosis. CONCLUSIONS FOLR1 is a stemness-associated biomarker for HCC, with serum levels serving as a diagnostic marker for HCC and a prognostic indicator for early-stage disease.
Collapse
Affiliation(s)
- Yuto Shiode
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Yu Sato
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Ryo Takahashi
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Takayuki Matsumae
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Kumiko Shirai
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Akira Doi
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Yuki Tahata
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Hayato Hikita
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Tomohide Tatsumi
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan
| | - Moto Fukai
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Akinobu Taketomi
- Department of Gastroenterological Surgery I, Graduate School of Medicine, Hokkaido University, Sapporo, Japan
| | - Mathuros Ruchirawat
- Laboratory of Chemical Carcinogenesis, Chulabhorn Research Institute, Bangkok, 10210, Thailand
- Center of Excellence On Environmental Health and Toxicology (EHT), OPS, MHESI, Bangkok, Thailand
| | - Xin Wei Wang
- Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, MD, USA
| | - Tetsuo Takehara
- Department of Gastroenterology and Hepatology, Osaka University, Graduate School of Medicine, 2-2 Yamadaoka Suita, Osaka, 565-0871, Japan.
| |
Collapse
|
49
|
Lindblad KE, Donne R, Liebling I, Barcena-Varela M, Lozano A, de Galarreta MR, Dhainaut M, Param NJ, Giotti B, Cappuyns S, Kodama T, Wang Y, Kamphorst AO, Tsankov AM, Lujambio A. NOTCH1 Drives Sexually Dimorphic Immune Responses in Hepatocellular Carcinoma. Cancer Discov 2025; 15:495-510. [PMID: 39560425 PMCID: PMC11875915 DOI: 10.1158/2159-8290.cd-24-1215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 10/28/2024] [Accepted: 11/15/2024] [Indexed: 11/20/2024]
Abstract
Hepatocellular carcinoma presents strong sexual dimorphism, being two to three times more frequent in males than in females; however, the role of sex in response to immunotherapies in HCC remains unknown. We demonstrate that NOTCH1, an understudied oncogene in HCC, elicits sexually dimorphic antitumor immunity and response to FDA-approved immunotherapies. Surprisingly, males harboring NOTCH1-driven tumors displayed enhanced antitumor immune responses, which, in mice, were mediated by dendritic and T cells. Conversely, females harboring NOTCH1-driven tumors presented immune evasion and resistance to immunotherapies through a defect in dendritic cell (DC)-mediated priming and activation of CD8+ T cells in mice, which was restored therapeutically with CD40 agonism. Mechanistically, the sexually dimorphic immunity was mediated by genes in the sex chromosomes but not by sex hormones. Together, our study unravels an unexpected association between NOTCH1 and sex in cancer immunity and highlights the potential of restoring the DC-CD8+ T-cell axis with CD40 agonism to improve outcomes. Significance: Although HCC presents strong sexual dimorphism, the role of sex in response to immunotherapies remains elusive. With a novel HCC mouse model and validation in patients with HCC, we demonstrate that NOTCH1 disrupts antitumor immunity specifically in females through a mechanism mediated by sex chromosome genes, which is reversed with CD40 agonism. See related commentary by Zhu and Koltsova, p. 452.
Collapse
Affiliation(s)
- Katherine E. Lindblad
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, USA
| | - Romain Donne
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Ian Liebling
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Marina Barcena-Varela
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Anthony Lozano
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Marina Ruiz de Galarreta
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Maxime Dhainaut
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
| | - Nesteene J. Param
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, USA
| | - Bruno Giotti
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
| | - Sarah Cappuyns
- Laboratory of Clinical Digestive Oncology, Department of Oncology, KU Leuven, Leuven, Belgium
| | - Takahiro Kodama
- Department of Gastroenterology and Hepatology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Yulei Wang
- Department of Oncology Biomarker Development, Genentech, Inc., South San Francisco, CA, USA
| | - Alice O. Kamphorst
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
| | - Alexander M. Tsankov
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York
| | - Amaia Lujambio
- The Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, USA
- Liver Cancer Program, Division of Liver Diseases, Department of Medicine, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, USA
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, USA
| |
Collapse
|
50
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|