51
|
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
|
52
|
Wang J, Lou Y, Peng X, Ye M, Cao W, Wu J, Yan Z, Zhao X, Zhou Y, Zheng C, Wei X, Chen Q, Hu C, Zhang M, Qu L, Chen Z, Fu Q, Wang W, Li J, Zhang Q, Liang T. Comprehensive analysis of protein post-translational modifications reveals PTPN2-STAT1-AOX axis-mediated tumor progression in hepatocellular carcinomas. Transl Oncol 2025; 53:102275. [PMID: 39837058 PMCID: PMC11788854 DOI: 10.1016/j.tranon.2025.102275] [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: 03/29/2024] [Revised: 12/02/2024] [Accepted: 01/05/2025] [Indexed: 01/23/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignant tumor. Although the proteomics of HCC is well studied, the landscape of post-translational modifications (PTMs) in HCC is poorly understood. The PTMs themselves and their crosstalk might be deeply involved in HCC development and progression. Herein, we investigated nine types of PTMs in paired tumor and normal tissues from nine patients with HCC using the label-free quantitative liquid chromatography with tandem mass spectrometry (LC-MS)-based technique. We identified >60,000 modified sites, and found that phosphorylation and ubiquitination were two most frequently changed PTMs between tumor and normal tissues. Crosstalk between malonylation-ubiquitination, phosphorylation-ubiquitination, and succinylation-propionylation were most significant among all PTMs. Further analysis revealed that Thr-160 of CDK2 regulated EZH2 via H3K27me3, and proposed a PTPN2-STAT1-AOX1 axis for HCC development through driver PTM exploration. In conclusion, our study provides a database of multiple PTMs in HCC, which might help to understand the biology of HCC and reveal novel targets for drug development.
Collapse
Affiliation(s)
- Junli Wang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yu Lou
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaojun Peng
- Cosmos Wisdom Biotechnology Co. Ltd., Hangzhou, China
| | - Mao Ye
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanyue Cao
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Jiangchao Wu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | | | - Yu Zhou
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chenlei Zheng
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Xiaobao Wei
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qitai Chen
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chengyang Hu
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | | | - Lanqing Qu
- Zhejiang University School of Medicine, Hangzhou, China
| | - Zeshe Chen
- Zhejiang University School of Medicine, Hangzhou, China
| | - Qihan Fu
- Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, China; Department of Medical Oncology, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Weixin Wang
- Cosmos Wisdom Biotechnology Co. Ltd., Hangzhou, China
| | - Jingsong Li
- Research Center for Healthcare Data Science, Zhejiang Lab, Hangzhou, China
| | - Qi Zhang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, China; Zhejiang University Cancer Center, Hangzhou, China.
| | - Tingbo Liang
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Department of Hepatobiliary and Pancreatic Surgery, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China; Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Zhejiang Province, China; Zhejiang University Cancer Center, Hangzhou, China.
| |
Collapse
|
53
|
Hu GS, Zheng ZZ, He YH, Wang DC, Nie RC, Liu W. Integrated Analysis of Proteome and Transcriptome Profiling Reveals Pan-Cancer-Associated Pathways and Molecular Biomarkers. Mol Cell Proteomics 2025; 24:100919. [PMID: 39884577 PMCID: PMC11907456 DOI: 10.1016/j.mcpro.2025.100919] [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: 06/12/2024] [Revised: 01/02/2025] [Accepted: 01/24/2025] [Indexed: 02/01/2025] Open
Abstract
Understanding dysregulated genes and pathways in cancer is critical for precision oncology. Integrating mass spectrometry-based proteomic data with transcriptomic data presents unique opportunities for systematic analyses of dysregulated genes and pathways in pan-cancer. Here, we compiled a comprehensive set of datasets, encompassing proteomic data from 2404 samples and transcriptomic data from 7752 samples across 13 cancer types. Comparisons between normal or adjacent normal tissues and tumor tissues identified several dysregulated pathways including mRNA splicing, interferon pathway, fatty acid metabolism, and complement coagulation cascade in pan-cancer. Additionally, pan-cancer upregulated and downregulated genes (PCUGs and PCDGs) were also identified. Notably, RRM2 and ADH1B, two genes which belong to PCUGs and PCDGs, respectively, were identified as robust pan-cancer diagnostic biomarkers. TNM stage-based comparisons revealed dysregulated genes and biological pathways involved in cancer progression, among which the dysregulation of complement coagulation cascade and epithelial-mesenchymal transition are frequent in multiple types of cancers. A group of pan-cancer continuously upregulated and downregulated proteins in different tumor stages (PCCUPs and PCCDPs) were identified. We further constructed prognostic risk stratification models for corresponding cancer types based on dysregulated genes, which effectively predict the prognosis for patients with these cancers. Drug prediction based on PCUGs and PCDGs as well as PCCUPs and PCCDPs revealed that small molecule inhibitors targeting CDK, HDAC, MEK, JAK, PI3K, and others might be effective treatments for pan-cancer, thereby supporting drug repurposing. We also developed web tools for cancer diagnosis, pathologic stage assessment, and risk evaluation. Overall, this study highlights the power of combining proteomic and transcriptomic data to identify valuable diagnostic and prognostic markers as well as drug targets and treatments for cancer.
Collapse
Affiliation(s)
- Guo-Sheng Hu
- Biomedical Research Center of South China, College of Life Sciences, Fujian Normal University, Fuzhou, China; State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Zao-Zao Zheng
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Yao-Hui He
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; MOE Key Lab of Rare Pediatric Diseases, Hengyang Medical School, University of South China, Hengyang, Hunan, China
| | - Du-Chuang Wang
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China
| | - Rui-Chao Nie
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China
| | - Wen Liu
- State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; Xiang An Biomedicine Laboratory, School of Pharmaceutical Sciences, Faculty of Medicine and Life Sciences, Xiamen University, Xiamen, Fujian, China; National Institute for Data Science in Health and Medicine, Xiamen University, Xiamen, Fujian, China.
| |
Collapse
|
54
|
Malone CD, Bajaj S, He A, Mody K, Hickey RM, Sarwar A, Krishnan S, Patel TC, Toskich BB. Combining Radioembolization and Immune Checkpoint Inhibitors for the Treatment of Hepatocellular Carcinoma: The Quest for Synergy. J Vasc Interv Radiol 2025; 36:414-424.e2. [PMID: 39586534 DOI: 10.1016/j.jvir.2024.11.012] [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: 08/05/2024] [Revised: 11/06/2024] [Accepted: 11/13/2024] [Indexed: 11/27/2024] Open
Abstract
Hepatocellular carcinoma is a leading and increasing contributor to cancer-related death worldwide. Recent advancements in both liver-directed therapies in the form of yttrium-90 (90Y) radioembolization (RE) and systemic therapy in the form of immune checkpoint inhibitors (ICI) have expanded treatment options for patients with an otherwise poor prognosis. Despite these gains, ICIs and 90Y-RE each have key limitations with low objective response rates and persistent hazard of out-of-field recurrence, respectively, and overall survival remains low. However, each therapy's strength may mitigate the other's weakness, making them potentially ideal partners for combination treatment strategies. This review discusses the scientific and clinical rationale for combining 90Y-RE with ICIs, highlights early clinical trial data on its safety and effectiveness, and proposes key issues to be addressed in this emerging field. With optimal strategies, combination therapies can potentially result in increasing likelihood of durable and curative outcomes in later stage patients.
Collapse
Affiliation(s)
- Christopher D Malone
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St Louis, Missouri.
| | - Suryansh Bajaj
- Department of Radiology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Aiwu He
- Division of Gastroenterology and Medical Oncology, MedStar Health, Washington, DC
| | | | - Ryan M Hickey
- Department of Radiology, NYU Langone Health, New York, New York
| | - Ammar Sarwar
- Department of Radiology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Sunil Krishnan
- Vivian L. Smith Department of Neurosurgery, The University of Texas Health Science Center, Houston, Texas
| | - Tushar C Patel
- Department of Transplant, Mayo Clinic, Jacksonville, Florida
| | - Beau B Toskich
- Division of Vascular and Interventional Radiology, Mayo Clinic, Jacksonville, Florida
| |
Collapse
|
55
|
Suzuki H, Mishra S, Paul S, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma for therapeutic development. JOURNAL OF LIVER CANCER 2025; 25:9-18. [PMID: 39639434 PMCID: PMC7617546 DOI: 10.17998/jlc.2024.12.02] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 11/26/2024] [Accepted: 12/02/2024] [Indexed: 12/07/2024]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide, with an estimated 750,000 deaths in 2022. Recent emergence of molecular targeted agents and immune checkpoint inhibitors and their combination therapies have been transforming HCC care, but their prognostic impact in advanced-stage disease remains unsatisfactory. In addition, their application to early-stage disease is still an unmet need. Omics profiling studies have elucidated recurrent and heterogeneously present molecular aberrations involved in pro-cancer tumor (immune) microenvironment that may guide therapeutic strategies. Recurrent aberrations such somatic mutations in TERT promoter and TP53 have been regarded undruggable, but recent studies have suggested that these may serve as new classes of therapeutic targets. HCC markers such as alpha-fetoprotein, glypican-3, and epithelial cell adhesion molecule have also been explored as therapeutic targets. These molecular features may be utilized as biomarkers to guide the application of new approaches as companion biomarkers to maximize therapeutic benefits in patients who are likely to benefit from the therapies, while minimizing unnecessary harm in patients who will not respond. The explosive number of new agents in the pipelines have posed challenges in their clinical testing. Novel clinical trial designs guided by predictive biomarkers have been proposed to enable their efficient and cost-effective evaluation. These new developments collectively facilitate clinical translation of personalized molecular-targeted therapies in HCC and substantially improve prognosis of HCC patients.
Collapse
Affiliation(s)
- Hiroyuki Suzuki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sumit Mishra
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Subhojit Paul
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
56
|
Putatunda V, Jusakul A, Roberts L, Wang XW. Genetic, Epigenetic, and Microenvironmental Drivers of Cholangiocarcinoma. THE AMERICAN JOURNAL OF PATHOLOGY 2025; 195:362-377. [PMID: 39532242 PMCID: PMC11841490 DOI: 10.1016/j.ajpath.2024.10.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Revised: 10/07/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024]
Abstract
Cholangiocarcinoma (CCA) is an aggressive and heterogeneous malignancy of the biliary tree that carries a poor prognosis. Multiple features at the genetic, epigenetic, and microenvironmental levels have been identified to better characterize CCA carcinogenesis. Genetic alterations, such as mutations in IDH1/2, BAP1, ARID1A, and FGFR2, play significant roles in CCA pathogenesis, with variations across different subtypes, races/ethnicities, and causes. Epigenetic dysregulation, characterized by DNA methylation and histone modifications, further contributes to the complexity of CCA, influencing gene expression and tumor behavior. Furthermore, CCA cells exchange autocrine and paracrine signals with other cancer cells and the infiltrating cell types that populate the microenvironment, including cancer-associated fibroblasts and tumor-associated macrophages, further contributing to an immunosuppressive niche that supports tumorigenesis. This review explores the multifaceted genetic, epigenetic, and microenvironmental drivers of CCA. Understanding these diverse mechanisms is essential for characterizing the complex pathways of CCA carcinogenesis and developing targeted therapies to improve patient outcomes.
Collapse
Affiliation(s)
- Vijay Putatunda
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Surgical Oncology Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland.
| | - Apinya Jusakul
- Cholangiocarcinoma Research Institute, Khon Kaen University, Khon Kaen, Thailand; Faculty of Associated Medical Sciences, Khon Kaen University, Khon Kaen, Thailand
| | - Lewis Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Xin Wei Wang
- Liver Cancer Program, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland; Laboratory of Human Carcinogenesis, Center for Cancer Research, National Cancer Institute, Bethesda, Maryland
| |
Collapse
|
57
|
Dhanasekaran R, Suzuki H, Lemaitre L, Kubota N, Hoshida Y. Molecular and immune landscape of hepatocellular carcinoma to guide therapeutic decision-making. Hepatology 2025; 81:1038-1057. [PMID: 37300379 PMCID: PMC10713867 DOI: 10.1097/hep.0000000000000513] [Citation(s) in RCA: 25] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 05/12/2023] [Indexed: 06/12/2023]
Abstract
Liver cancer, primarily HCC, exhibits highly heterogeneous histological and molecular aberrations across tumors and within individual tumor nodules. Such intertumor and intratumor heterogeneities may lead to diversity in the natural history of disease progression and various clinical disparities across the patients. Recently developed multimodality, single-cell, and spatial omics profiling technologies have enabled interrogation of the intertumor/intratumor heterogeneity in the cancer cells and the tumor immune microenvironment. These features may influence the natural history and efficacy of emerging therapies targeting novel molecular and immune pathways, some of which had been deemed undruggable. Thus, comprehensive characterization of the heterogeneities at various levels may facilitate the discovery of biomarkers that enable personalized and rational treatment decisions, and optimize treatment efficacy while minimizing the risk of adverse effects. Such companion biomarkers will also refine HCC treatment algorithms across disease stages for cost-effective patient management by optimizing the allocation of limited medical resources. Despite this promise, the complexity of the intertumor/intratumor heterogeneity and ever-expanding inventory of therapeutic agents and regimens have made clinical evaluation and translation of biomarkers increasingly challenging. To address this issue, novel clinical trial designs have been proposed and incorporated into recent studies. In this review, we discuss the latest findings in the molecular and immune landscape of HCC for their potential and utility as biomarkers, the framework of evaluation and clinical application of predictive/prognostic biomarkers, and ongoing biomarker-guided therapeutic clinical trials. These new developments may revolutionize patient care and substantially impact the still dismal HCC mortality.
Collapse
Affiliation(s)
| | - Hiroyuki Suzuki
- Division of Gastroenterology, Department of Medicine, Kurume University School of Medicine, Kurume, Fukuoka
| | - Lea Lemaitre
- Division of Gastroenterology and Hepatology, Stanford University, Stanford, California
| | - Naoto Kubota
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Yujin Hoshida
- Liver Tumor Translational Research Program, Simmons Comprehensive Cancer Center, Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| |
Collapse
|
58
|
Zhou W, Chi H, Zhao X, Tao G, Gan J. A mutational signature and ARID1A mutation associated with outcome in hepatocellular carcinoma. Clin Transl Oncol 2025; 27:1166-1175. [PMID: 39179940 DOI: 10.1007/s12094-024-03669-0] [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: 06/12/2024] [Accepted: 08/09/2024] [Indexed: 08/26/2024]
Abstract
OBJECTIVE The prognosis of hepatocellular carcinoma (HCC) is poor and there is no stable and reliable molecular biomarker for evaluation. This study attempted to find reliable prognostic markers from tumor mutational profiles. METHODS A total of 362 HCC samples with whole-exome sequencing were collected as discovery datasets, and 200 samples with targeted sequencing were used for validation of the relevant results. All HCC samples were obtained from previously published studies. Bayesian non-negative matrix factorization was used to extract mutational signatures, and multivariate Cox regression models were utilized to identify the prognostic role of mutational factors. Gene set enrichment analysis was employed to discover potential signaling pathways associated with specific mutational groups. RESULTS In the HCC discovery dataset, a total of four mutational signatures (i.e., signatures 4, 6, 16, and 22) were extracted, of which signature 16 characterized by T>C mutations was observed to be associated with favorable HCC prognosis, and this correlation was also found in the validation dataset. Further analysis showed that patients with ARID1A mutations exhibited inferior survival outcomes in both discovery and validation datasets. Mechanistic exploration revealed that the presence of signature 16 was associated with better immune infiltration and tumor immunogenicity, while patients with ARID1A mutations were away from these favorable immunological features. CONCLUSION By integrating somatic mutation data and clinical information of HCC, this study identified that signature 16 and ARID1A mutations were associated with better and worse outcomes respectively, providing a basis for prognosis prediction and clinical treatment strategies of HCC.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China
| | - Hao Chi
- Department of Medical Laboratory, Huaian Hospital of Huaian City, Huaian, China
| | - Xiaohu Zhao
- Department of Infectious Diseases, Huaian Hospital of Huaian City, Huaian, China
| | - Guangrong Tao
- Department of Infectious Diseases, Huaian Hospital of Huaian City, Huaian, China
| | - Jianhe Gan
- Department of Infectious Diseases, The First Affiliated Hospital of Soochow University, Suzhou, 215006, Jiangsu, China.
| |
Collapse
|
59
|
Müller M, May S, Hall H, Kendall TJ, McGarry L, Blukacz L, Nuciforo S, Georgakopoulou A, Jamieson T, Phinichkusolchit N, Dhayade S, Suzuki T, Huguet-Pradell J, Powley IR, Officer-Jones L, Pennie RL, Esteban-Fabró R, Gris-Oliver A, Pinyol R, Skalka GL, Leslie J, Hoare M, Sprangers J, Malviya G, Mackintosh A, Johnson E, McCain M, Halpin J, Kiourtis C, Nixon C, Clark G, Clark W, Shaw R, Hedley A, Drake TM, Tan EH, Neilson M, Murphy DJ, Lewis DY, Reeves HL, Le Quesne J, Mann DA, Carlin LM, Blyth K, Llovet JM, Heim MH, Sansom OJ, Miller CJ, Bird TG. Human-correlated genetic models identify precision therapy for liver cancer. Nature 2025; 639:754-764. [PMID: 39972137 PMCID: PMC11922762 DOI: 10.1038/s41586-025-08585-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Accepted: 01/02/2025] [Indexed: 02/21/2025]
Abstract
Hepatocellular carcinoma (HCC), the most common form of primary liver cancer, is a leading cause of cancer-related mortality worldwide1,2. HCC occurs typically from a background of chronic liver disease, caused by a spectrum of predisposing conditions. Tumour development is driven by the expansion of clones that accumulate progressive driver mutations3, with hepatocytes the most likely cell of origin2. However, the landscape of driver mutations in HCC is broadly independent of the underlying aetiologies4. Despite an increasing range of systemic treatment options for advanced HCC, outcomes remain heterogeneous and typically poor. Emerging data suggest that drug efficacies depend on disease aetiology and genetic alterations5,6. Exploring subtypes in preclinical models with human relevance will therefore be essential to advance precision medicine in HCC7. Here we generated a suite of genetically driven immunocompetent in vivo and matched in vitro HCC models. Our models represent multiple features of human HCC, including clonal origin, histopathological appearance and metastasis. We integrated transcriptomic data from the mouse models with human HCC data and identified four common human-mouse subtype clusters. The subtype clusters had distinct transcriptomic characteristics that aligned with the human histopathology. In a proof-of-principle analysis, we verified response to standard-of-care treatment and used a linked in vitro-in vivo pipeline to identify a promising therapeutic candidate, cladribine, that has not previously been linked to HCC treatment. Cladribine acts in a highly effective subtype-specific manner in combination with standard-of-care therapy.
Collapse
Affiliation(s)
| | - Stephanie May
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Holly Hall
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Timothy J Kendall
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK
| | - Lynn McGarry
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Lauriane Blukacz
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Sandro Nuciforo
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
| | - Anastasia Georgakopoulou
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | - Narisa Phinichkusolchit
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | | | | | - Júlia Huguet-Pradell
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Ian R Powley
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | | | - Roger Esteban-Fabró
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Albert Gris-Oliver
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | - Roser Pinyol
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
| | | | - Jack Leslie
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - Matthew Hoare
- Early Cancer Institute, University of Cambridge, Cambridge, UK
- Department of Medicine, University of Cambridge, Addenbrooke's Hospital, Cambridge, UK
| | | | | | | | - Emma Johnson
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Misti McCain
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
| | - John Halpin
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Christos Kiourtis
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Colin Nixon
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Graeme Clark
- Cancer Research UK Scotland Institute, Glasgow, UK
| | | | - Robin Shaw
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Ann Hedley
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Thomas M Drake
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Centre for Medical Informatics, Usher Institute, University of Edinburgh, Edinburgh, UK
| | - Ee Hong Tan
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Matt Neilson
- Cancer Research UK Scotland Institute, Glasgow, UK
| | - Daniel J Murphy
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - David Y Lewis
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Helen L Reeves
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
- Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- Liver Group, Newcastle-upon-Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, UK
| | - John Le Quesne
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Department of Histopathology, Queen Elizabeth University Hospital, Glasgow, UK
| | - Derek A Mann
- Newcastle Fibrosis Research Group, Biosciences Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
- The Newcastle University Centre for Cancer, Newcastle University, Newcastle upon Tyne, UK
- Department of Gastroenterology and Hepatology, School of Medicine, Koç University, Istanbul, Turkey
| | - Leo M Carlin
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Karen Blyth
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Josep M Llovet
- Liver Cancer Translational Research Group, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Institució Catalana de Recerca i Estudis Avançats, Barcelona, Spain
| | - Markus H Heim
- Department of Biomedicine, University Hospital and University of Basel, Basel, Switzerland
- University Digestive Health Care Center Basel-Clarunis, Basel, Switzerland
| | - Owen J Sansom
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
- Cancer Research UK Scotland Centre, Edinburgh, UK
- Cancer Research UK Scotland Centre, Glasgow, UK
| | - Crispin J Miller
- Cancer Research UK Scotland Institute, Glasgow, UK
- School of Cancer Sciences, University of Glasgow, Glasgow, UK
| | - Thomas G Bird
- Cancer Research UK Scotland Institute, Glasgow, UK.
- School of Cancer Sciences, University of Glasgow, Glasgow, UK.
- Centre for Inflammation Research, University of Edinburgh, Edinburgh, UK.
- Cancer Research UK Scotland Centre, Edinburgh, UK.
- Cancer Research UK Scotland Centre, Glasgow, UK.
| |
Collapse
|
60
|
Barcena-Varela M, Monga SP, Lujambio A. Precision models in hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2025; 22:191-205. [PMID: 39663463 DOI: 10.1038/s41575-024-01024-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/11/2024] [Indexed: 12/13/2024]
Abstract
Hepatocellular carcinoma (HCC) represents a global health challenge, and ranks among one of the most prevalent and deadliest cancers worldwide. Therapeutic advances have expanded the treatment armamentarium for patients with advanced HCC, but obstacles remain. Precision oncology, which aims to match specific therapies to patients who have tumours with particular features, holds great promise. However, its implementation has been hindered by the existence of numerous 'HCC influencers' that contribute to the high inter-patient heterogeneity. HCC influencers include tumour-related characteristics, such as genetic alterations, immune infiltration, stromal composition and aetiology, and patient-specific factors, such as sex, age, germline variants and the microbiome. This Review delves into the intricate world of HCC, describing the most innovative model systems that can be harnessed to identify precision and/or personalized therapies. We provide examples of how different models have been used to nominate candidate biomarkers, their limitations and strategies to optimize such models. We also highlight the importance of reproducing distinct HCC influencers in a flexible and modular way, with the aim of dissecting their relative contribution to therapy response. Next-generation HCC models will pave the way for faster discovery of precision therapies for patients with advanced HCC.
Collapse
Affiliation(s)
- Marina Barcena-Varela
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Satdarshan P Monga
- Department of Pharmacology and Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Pittsburgh Liver Research Center, University of Pittsburgh Medical Center and University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Pathology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Division of Gastroenterology, Hepatology and Nutrition, Department of Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Amaia Lujambio
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Liver Cancer Program, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA.
- Graduate School of Biomedical Sciences at Icahn School of Medicine at Mount Sinai, New York, NY, USA.
| |
Collapse
|
61
|
Wu ST, Zhu L, Feng XL, Wang HY, Li F. Strategies for discovering novel hepatocellular carcinoma biomarkers. World J Hepatol 2025; 17:101201. [PMID: 40027561 PMCID: PMC11866143 DOI: 10.4254/wjh.v17.i2.101201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/13/2024] [Accepted: 12/23/2024] [Indexed: 02/20/2025] Open
Abstract
Liver cancer, particularly hepatocellular carcinoma (HCC), remains a significant global health challenge due to its high mortality rate and late-stage diagnosis. The discovery of reliable biomarkers is crucial for improving early detection and patient outcomes. This review provides a comprehensive overview of current and emerging biomarkers for HCC, including alpha-fetoprotein, des-gamma-carboxy prothrombin, glypican-3, Golgi protein 73, osteopontin, and microRNAs. Despite advancements, the diagnostic limitations of existing biomarkers underscore the urgent need for novel markers that can detect HCC in its early stages. The review emphasizes the importance of integrating multi-omics approaches, combining genomics, proteomics, and metabolomics, to develop more robust biomarker panels. Such integrative methods have the potential to capture the complex molecular landscape of HCC, offering insights into disease mechanisms and identifying targets for personalized therapies. The significance of large-scale validation studies, collaboration between research institutions and clinical settings, and consideration of regulatory pathways for clinical implementation is also discussed. In conclusion, while substantial progress has been made in biomarker discovery, continued research and innovation are essential to address the remaining challenges. The successful translation of these discoveries into clinical practice will require rigorous validation, standardization of protocols, and cross-disciplinary collaboration. By advancing the development and application of novel biomarkers, we can improve the early detection and management of HCC, ultimately enhancing patient survival and quality of life.
Collapse
Affiliation(s)
- Shi-Tao Wu
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Li Zhu
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Xiao-Ling Feng
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Hao-Yu Wang
- Department of Hepatopancreatobiliary Surgery, Chongqing General Hospital, Chongqing 401147, China
| | - Fang Li
- Department of General Surgery, Chongqing General Hospital, Chongqing 401147, China.
| |
Collapse
|
62
|
Lu L, Zhou X, Zheng J, Li D. Coilin Affects the Prognosis of Hepatocellular Carcinoma Through Cell Cycle and Apoptosis. J Hepatocell Carcinoma 2025; 12:367-382. [PMID: 40008396 PMCID: PMC11853879 DOI: 10.2147/jhc.s500119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Purpose Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality with a challenging prognosis. HCC lacks effective prognostic biomarkers. We investigated the diagnostic and prognostic value of COIL expression in HCC. Patients and Methods This study evaluated the expression and prognostic significance of COIL using data from the TCGA and local hospital samples, with 374 and 118 liver cancer patients in the TCGA database and local hospital, respectively. The techniques include bioinformatics analysis, qRT-PCR, immunohistochemistry (IHC), and in vitro cell experiments, which encompass CCK-8 assays, wound healing assays, and Transwell invasion assays. The relationship between COIL expression and clinical outcomes was assessed, and COIL's biological function in HCC was investigated through cellular assays. Results Analysis of cell lines and HCC tissue samples revealed that COIL mRNA or protein expression levels were significantly higher in HCC cell lines/tissues compared to normal liver cells/tissues. Univariate and multivariate analyses indicated that COIL is an independent prognostic factor for overall survival (OS) in HCC. Additionally, 14% of HCC patients had alterations in the COIL gene, and patients with COIL gene alterations had significantly lower OS (p<0.001) and disease-free survival (DFS) (p<0.001) compared to those without gene alterations. Knockdown of COIL expression inhibited the proliferation, migration, and invasion of Hep3B, HepG2, and Huh7. Compared to the control group, COIL knockdown cells showed a marked reduction in CDC25C and CCNB1 protein levels, suggesting that COIL knockdown leads to G2/M phase cell cycle arrest. After COIL knockdown, caspase-3 and BCL-2 protein levels were downregulated, while cleaved caspase and BAX protein levels were upregulated, indicating that COIL knockdown promotes apoptosis in liver cancer cells. Conclusion COIL is an independent predictor of prognosis. COIL's association with poor OS and its role in enhancing cancer cell proliferation and invasion highlight its potential as a therapeutic target.
Collapse
Affiliation(s)
- Lingling Lu
- Department of Infection Disease, Fujian Medical University Union Hospital, Fuzhou, People’s Republic of China
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
| | - Xiaoling Zhou
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Jiaolong Zheng
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| | - Dongliang Li
- Fuzong Clinical Medical College of Fujian Medical University, Fuzhou, People’s Republic of China
- Department of Hepatobiliary Disease, The 900th Hospital of Joint Logistic Support Force, PLA, Fuzhou, People’s Republic of China
| |
Collapse
|
63
|
Turnham RE, Pitea A, Jang GM, Xu Z, Lim HC, Choi AL, Von Dollen J, Levin RS, Webber JT, McCarthy E, Hu J, Li X, Che L, Singh A, Yoon A, Chan G, Kelley RK, Swaney DL, Zhang W, Bandyopadhyay S, Theis FJ, Eckhardt M, Chen X, Shokat KM, Ideker T, Krogan NJ, Gordan JD. HBV Remodels PP2A Complexes to Rewire Kinase Signaling in Hepatocellular Carcinoma. Cancer Res 2025; 85:660-674. [PMID: 39652575 PMCID: PMC11949624 DOI: 10.1158/0008-5472.can-24-0456] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 09/09/2024] [Accepted: 12/04/2024] [Indexed: 02/18/2025]
Abstract
Hepatitis B virus (HBV) infections promote liver cancer initiation by inducing inflammation and cellular stress. Despite a primarily indirect effect on oncogenesis, HBV is associated with a recurrent genomic phenotype in hepatocellular carcinoma (HCC), suggesting that it impacts the biology of established HCC. Characterization of the interaction of HBV with host proteins and the mechanistic contributions of HBV to HCC initiation and maintenance could provide insights into HCC biology and uncover therapeutic vulnerabilities. In this study, we used affinity purification mass spectrometry to comprehensively map a network of 145 physical interactions between HBV and human proteins in HCC. A subset of the host factors targeted by HBV proteins were preferentially mutated in non-HBV-associated HCC, suggesting that their interaction with HBV influences HCC biology. HBV interacted with proteins involved in mRNA splicing, mitogenic signaling, and DNA repair, with the latter set interacting with the HBV oncoprotein X (HBx). HBx remodeled the PP2A phosphatase complex by excluding striatin regulatory subunits from the PP2A holoenzyme, and the HBx effects on PP2A caused Hippo kinase activation. In parallel, HBx activated mTOR complex 2, which can prevent YAP degradation. mTOR complex 2-mediated upregulation of YAP was observed in human HCC specimens and mouse HCC models and could be targeted with mTOR kinase inhibitors. Thus, HBV interaction with host proteins rewires HCC signaling rather than directly activating mitogenic pathways, providing an alternative paradigm for the cellular effects of a tumor-promoting virus. Significance: Integrative proteomic and genomic analysis of HBV/host interactions illuminated modifiers of hepatocellular carcinoma behavior and key signaling mechanisms in advanced disease, which suggested that HBV may have therapeutically actionable effects.
Collapse
Affiliation(s)
- Rigney E Turnham
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Adriana Pitea
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Gwendolyn M Jang
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Zhong Xu
- Department of Bioengineering, University of California, San Francisco CA
| | - Huat Chye Lim
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Alex L Choi
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - John Von Dollen
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Rebecca S. Levin
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - James T Webber
- Department of Bioengineering, University of California, San Francisco CA
| | - Elizabeth McCarthy
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Junjie Hu
- Department of Bioengineering, University of California, San Francisco CA
| | - Xiaolei Li
- Department of Bioengineering, University of California, San Francisco CA
| | - Li Che
- Department of Bioengineering, University of California, San Francisco CA
| | - Ananya Singh
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Alex Yoon
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Gary Chan
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| | - Robin K Kelley
- Division of Hematology/Oncology, University of California, San Francisco CA
| | - Danielle L Swaney
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
| | - Wei Zhang
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | | | - Fabian J Theis
- TUM School of Life Sciences, Technical University of Munich, Munich, Germany
| | - Manon Eckhardt
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA
| | - Xin Chen
- Department of Bioengineering, University of California, San Francisco CA
| | - Kevan M Shokat
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Howard Hughes Medical Institute, University of California, San Francisco CA
| | - Trey Ideker
- Department of Medicine, University of California, San Diego, La Jolla, CA
| | - Nevan J Krogan
- Quantitative Biosciences Institute, University of California, San Francisco CA
- Department of Cellular and Molecular Pharmacology, University of California, San Francisco CA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco
- Gladstone Institute of Data Science and Biotechnology, J. David Gladstone Institutes, San Francisco, CA
| | - John D Gordan
- Division of Hematology/Oncology, University of California, San Francisco CA
- Quantitative Biosciences Institute, University of California, San Francisco CA
| |
Collapse
|
64
|
Zheng J, Wang S, Xia L, Sun Z, Chan KM, Bernards R, Qin W, Chen J, Xia Q, Jin H. Hepatocellular carcinoma: signaling pathways and therapeutic advances. Signal Transduct Target Ther 2025; 10:35. [PMID: 39915447 PMCID: PMC11802921 DOI: 10.1038/s41392-024-02075-w] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 09/18/2024] [Accepted: 11/14/2024] [Indexed: 02/09/2025] Open
Abstract
Liver cancer represents a major global health concern, with projections indicating that the number of new cases could surpass 1 million annually by 2025. Hepatocellular carcinoma (HCC) constitutes around 90% of liver cancer cases and is primarily linked to factors incluidng aflatoxin, hepatitis B (HBV) and C (HCV), and metabolic disorders. There are no obvious symptoms in the early stage of HCC, which often leads to delays in diagnosis. Therefore, HCC patients usually present with tumors in advanced and incurable stages. Several signaling pathways are dis-regulated in HCC and cause uncontrolled cell propagation, metastasis, and recurrence of HCC. Beyond the frequently altered and therapeutically targeted receptor tyrosine kinase (RTK) pathways in HCC, pathways involved in cell differentiation, telomere regulation, epigenetic modification and stress response also provide therapeutic potential. Investigating the key signaling pathways and their inhibitors is pivotal for achieving therapeutic advancements in the management of HCC. At present, the primary therapeutic approaches for advanced HCC are tyrosine kinase inhibitors (TKI), immune checkpoint inhibitors (ICI), and combination regimens. New trials are investigating combination therapies involving ICIs and TKIs or anti-VEGF (endothelial growth factor) therapies, as well as combinations of two immunotherapy regimens. The outcomes of these trials are expected to revolutionize HCC management across all stages. Here, we provide here a comprehensive review of cellular signaling pathways, their therapeutic potential, evidence derived from late-stage clinical trials in HCC and discuss the concepts underlying earlier clinical trials, biomarker identification, and the development of more effective therapeutics for HCC.
Collapse
Affiliation(s)
- Jiaojiao Zheng
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Siying Wang
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Lei Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Zhen Sun
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Kui Ming Chan
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, PR China
| | - René Bernards
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
- Division of Molecular Carcinogenesis, Oncode Institute, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Wenxin Qin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China
| | - Jinhong Chen
- Department of General Surgery, Huashan Hospital, Fudan University, Shanghai, PR China.
| | - Qiang Xia
- Department of Liver Surgery, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| | - Haojie Jin
- State Key Laboratory of Systems Medicine for Cancer, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, PR China.
| |
Collapse
|
65
|
Gritti I, Wan J, Weeresekara V, Vaz JM, Tarantino G, Bryde TH, Vijay V, Kammula AV, Kattel P, Zhu S, Vu P, Chan M, Wu MJ, Gordan JD, Patra KC, Silveira VS, Manguso RT, Wein MN, Ott CJ, Qi J, Liu D, Sakamoto K, Gujral TS, Bardeesy N. DNAJB1-PRKACA Fusion Drives Fibrolamellar Liver Cancer through Impaired SIK Signaling and CRTC2/p300-Mediated Transcriptional Reprogramming. Cancer Discov 2025; 15:382-400. [PMID: 39326063 PMCID: PMC11803398 DOI: 10.1158/2159-8290.cd-24-0634] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 08/16/2024] [Accepted: 09/25/2024] [Indexed: 09/28/2024]
Abstract
SIGNIFICANCE This work combines functional studies in model systems and examination of human tumor specimens to define a central oncogenic pathway driven by DNAJB1-PRKACA fusions in FLC. DNAJB1-PRKACA-mediated inactivation of the SIK stimulates CRTC2-p300-mediated transcription to drive tumor growth. The findings illuminate pathogenic mechanisms and inform therapeutic development.
Collapse
Affiliation(s)
- Ilaria Gritti
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Jinkai Wan
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Vajira Weeresekara
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Joel M. Vaz
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Giuseppe Tarantino
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Tenna Holgersen Bryde
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Vindhya Vijay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Ashwin V. Kammula
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Prabhat Kattel
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Songli Zhu
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Phuong Vu
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
| | - Marina Chan
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Meng-Ju Wu
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - John D. Gordan
- Division of Hematology and Oncology, University of California, San Francisco, San Francisco, California
| | | | - Vanessa S. Silveira
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Robert T. Manguso
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Marc N. Wein
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Endocrine Unit, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts
| | - Christopher J. Ott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| | - Jun Qi
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - David Liu
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts
| | - Kei Sakamoto
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Taranjit S. Gujral
- Division of Human Biology, Fred Hutchinson Cancer Center, Seattle, Washington
| | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital, Boston, Massachusetts
- Center for Cancer Research, Center for Regenerative Medicine, Massachusetts General, Boston, Massachusetts
- Department of Medicine, Harvard Medical School, Boston, Massachusetts
- Broad Institute of Massachusetts Institute of Technology and Harvard, Cambridge, Massachusetts
| |
Collapse
|
66
|
Karin M, Kim JY. MASH as an emerging cause of hepatocellular carcinoma: current knowledge and future perspectives. Mol Oncol 2025; 19:275-294. [PMID: 38874196 PMCID: PMC11793012 DOI: 10.1002/1878-0261.13685] [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: 07/17/2023] [Revised: 04/15/2024] [Accepted: 06/04/2024] [Indexed: 06/15/2024] Open
Abstract
Hepatocellular carcinoma is one of the deadliest and fastest-growing cancers. Among HCC etiologies, metabolic dysfunction-associated fatty liver disease (MAFLD) has served as a major HCC driver due to its great potential for increasing cirrhosis. The obesogenic environment fosters a positive energy balance and results in a continuous rise of obesity and metabolic syndrome. However, it is difficult to understand how metabolic complications lead to the poor prognosis of liver diseases and which molecular mechanisms are underpinning MAFLD-driven HCC development. Thus, suitable preclinical models that recapitulate human etiologies are essentially required. Numerous preclinical models have been created but not many mimicked anthropometric measures and the course of disease progression shown in the patients. Here we review the literature on adipose tissues, liver-related HCC etiologies and recently discovered genetic mutation signatures found in MAFLD-driven HCC patients. We also critically review current rodent models suggested for MAFLD-driven HCC study.
Collapse
Affiliation(s)
- Michael Karin
- Laboratory of Gene Regulation and Signal Transduction, Departments of Pharmacology and Pathology, School of MedicineUniversity of California San DiegoLa JollaCAUSA
| | - Ju Youn Kim
- Department of Molecular and Life ScienceHanyang University ERICAAnsanKorea
| |
Collapse
|
67
|
Young EP, O’Neill AF, Rangaswami AA. Pediatric Hepatocellular Carcinoma: A Review of Predisposing Conditions, Molecular Mechanisms, and Clinical Considerations. Int J Mol Sci 2025; 26:1252. [PMID: 39941018 PMCID: PMC11818592 DOI: 10.3390/ijms26031252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 01/21/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
Pediatric hepatocellular carcinoma (HCC) is a rare malignant liver tumor affecting children and adolescents and occurring either sporadically or in the context of underlying liver disease. In this review, we detail the epidemiology of pediatric HCC with a focus on predisposing factors including hepatic or systemic disease, genetic disorders, and familial cancer syndromes. We summarize existing research on the pathophysiology of pediatric HCC, including molecular mechanisms of oncogenesis, highlighting unique disease features differentiating pediatric HCC from adult HCC. We then survey the landscape of therapeutic options for pediatric HCC, including novel therapeutics. Lastly, we discuss the pathologic spectrum upon which pediatric HCC is postulated to exist, ranging from hepatoblastoma to HCC and including the hybrid entity hepatocellular neoplasm not otherwise specifed (HCN-NOS). In summary, we highlight the key clinical and molecular features of pediatric HCC that may inform future research and novel approaches to the clinical care of these patients.
Collapse
Affiliation(s)
- Elizabeth P. Young
- Department of Pediatrics, Division of Oncology, University of California San Francisco, San Francisco, CA 94158, USA;
| | - Allison F. O’Neill
- Department of Pediatric Oncology, Dana-Farber Cancer Institute, Boston Children’s Hospital and Harvard Medical School, Boston, MA 02215, USA;
| | - Arun A. Rangaswami
- Department of Pediatrics, Division of Oncology, University of California San Francisco, San Francisco, CA 94158, USA;
| |
Collapse
|
68
|
Mano Y, Igarashi Y, Komori K, Hashimoto I, Watanabe H, Takahashi K, Kano K, Fujikawa H, Yamada T, Himuro H, Kouro T, Wei F, Tsuji K, Horaguchi S, Komahashi M, Oshima T, Sasada T. Characteristics and clinical significance of immune cells in omental milky spots of patients with gastric cancer. Front Immunol 2025; 16:1521278. [PMID: 39949777 PMCID: PMC11821591 DOI: 10.3389/fimmu.2025.1521278] [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: 11/01/2024] [Accepted: 01/02/2025] [Indexed: 02/16/2025] Open
Abstract
The omentum is a common site of peritoneal metastasis in various cancers, including gastric cancer. It contains immune cell aggregates known as milky spots, which provide a microenvironment for peritoneal immunity by regulating innate and adaptive immune responses. In this study, we investigated gene expression profiles in cells from omental milky spots of patients with gastric cancer (n = 37) by RNA sequencing analysis and classified the patients into four groups (G1-4). Notably, significant differences were observed between the groups in terms of macroscopic type, lymphatic invasion, venous invasion, and pathological stage (pStage). G3, which was enriched in genes related to acquired immunity, showed earlier tumor stages (macroscopic type 0, Ly0, V0, and pStage I) and a better prognosis. In contrast, G4 showed enrichment of genes related to neutrophils and innate immunity; G1 and G2 showed no enrichment of innate or adaptive immune-related genes, suggesting an immune desert microenvironment. Cytometric analysis revealed significantly more T and B cells and fewer neutrophils in G3. Accordingly, the immune microenvironment in omental milky spots may vary depending on the stage of gastric cancer progression. When univariate Cox proportional hazards regression models were used to search for prognostically relevant genes specific to G3, 23 potential prognostic genes were identified as common genes associated with relapse-free survival and overall survival. In addition, the multivariate Cox proportional hazards model using these prognostic genes and clinicopathological information showed that combining the B cell marker CD19 and Ly had a high predictive accuracy for prognosis. Based on this study's results, it is possible that tumor progression, such as lymphatic and/or venous infiltration of tumor cells, may affect the immune cell composition and proportions in omental milky spots of patients with gastric cancer and analysis of gene expression in omental milky spots may help to predict gastric cancer prognosis.
Collapse
Affiliation(s)
- Yasunobu Mano
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Yuka Igarashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Keisuke Komori
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
- Department of Surgery, Yokohama City University, Yokohama, Japan
| | - Itaru Hashimoto
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
- Department of Surgery, Yokohama City University, Yokohama, Japan
| | - Hayato Watanabe
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kosuke Takahashi
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Kazuki Kano
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hirohito Fujikawa
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Takanobu Yamada
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Hidetomo Himuro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Taku Kouro
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Feifei Wei
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Kayoko Tsuji
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| | - Shun Horaguchi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Mitsuru Komahashi
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo, Japan
| | - Takashi Oshima
- Department of Gastrointestinal Surgery, Kanagawa Cancer Center, Yokohama, Japan
| | - Tetsuro Sasada
- Division of Cancer Immunotherapy, Kanagawa Cancer Center Research Institute, Yokohama, Japan
- Cancer Vaccine and Immunotherapy Center, Kanagawa Cancer Center Research Institute, Yokohama, Japan
| |
Collapse
|
69
|
Hapaer G, Che F, Xu Q, Li Q, Liang A, Wang Z, Ziluo J, Zhang X, Wei Y, Yuan Y, Song B. Radiomics-based biomarker for PD-1 status and prognosis analysis in patients with HCC. Front Immunol 2025; 16:1435668. [PMID: 39944703 PMCID: PMC11813882 DOI: 10.3389/fimmu.2025.1435668] [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: 05/20/2024] [Accepted: 01/13/2025] [Indexed: 03/17/2025] Open
Abstract
Purpose To investigate the impact of preoperative contrast-enhanced CT-based radiomics model on PD-1 prediction in hepatocellular carcinoma (HCC) patients. Methods The study included 105 HCC patients (training cohort: 72; validation cohort: 33) who underwent preoperative contrast-enhanced CT and received systemic sorafenib treatment after surgery. Radiomics score was built for each patient and was integrated with independent clinic radiologic predictors into the radiomics model using multivariable logistic regression analysis. Results Seventeen radiomics features were finally selected to construct the radiomics score. In multivariate analysis, serum creatine and peritumoral enhancement were significant independent factors for PD-1 prediction. The radiomics model integrated radiomics signature with serum creatine and peritumoral enhancement showed good discriminative performance (AUC of 0.897 and 0.794 in the training and validation cohort). Overall survival (OS) was significantly different between the radiomics-predicted PD-1-positive and PD-1-negative groups (OS: 29.66 months, CI:16.03-44.40 vs. 31.04 months, CI: 17.10-44.07, P<0.001). Radiomics-predicted PD-1 was an independent predictor of OS of patients treated with sorafenib after surgery. (Hazard ratio [HR]: 1.61 [1.23-2.1], P<0.001). Conclusion The proposed model based on radiomic signature helps to evaluate PD-1 status of HCC patients and may be used for evaluating patients most likely to benefit from sorafenib as a potentially combination therapy regimen with immune checkpoint therapies.
Collapse
Affiliation(s)
- Gulizaina Hapaer
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Feng Che
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qing Xu
- Institute of Clinical Pathology, Key Laboratory of Transplant Engineering and Immunology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Qian Li
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ailin Liang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Zhou Wang
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jituome Ziluo
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Xin Zhang
- Pharmaceutical Diagnostics, General Electric (GE) Healthcare, Shanghai, China
| | - Yi Wei
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yuan Yuan
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Bin Song
- Department of Radiology, West China Hospital, Sichuan University, Chengdu, Sichuan, China
- Department of Radiology, Sanya People’s Hospital, Sanya, China
| |
Collapse
|
70
|
Park SJ, Shin K, Kim IH, Lee M, Hong TH, Kim H. Exploring the efficacy of fluorouracil and platinum based chemotherapy in advanced hepatocellular carcinoma to bridge the treatment gap in resource limited settings. Sci Rep 2025; 15:3386. [PMID: 39870720 PMCID: PMC11772854 DOI: 10.1038/s41598-025-86523-9] [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: 05/28/2024] [Accepted: 01/13/2025] [Indexed: 01/29/2025] Open
Abstract
Advanced hepatocellular carcinoma (HCC) poses treatment challenges, especially where access to multi-kinase inhibitors and ICIs is limited by high costs and lack of insurance. This study evaluates the effectiveness of 5-fluorouracil (5-FU) plus platinum-based chemotherapy as an alternative systemic treatment for advanced HCC. A retrospective analysis of advanced HCC patients treated with 5-FU plus platinum-based chemotherapy was conducted. The Kaplan-Meier method determined median progression-free survival (PFS) and overall survival (OS). From April 2009 to October 2023, 48 patients with advanced HCC were included in the study. Nearly all patients (97.9%) had extrahepatic metastasis and stable liver function, with three-quarters previously treated with sorafenib. At a median follow-up of 7.8 months, the median PFS was 4.2 months (95% CI, 1.3-7.1), and the median OS was 8.2 months (95% CI, 2.5-13.9). A high pretreatment neutrophil-to-lymphocyte ratio (≥ 3.0) adversely affected both PFS (HR = 1.79; 95% CI, 0.99-3.25; p = 0.034) and OS (HR = 2.02; 95% CI, 1.10-3.69; p = 0.011). Hematologic toxicities related to the treatment were substantial, with 62.5% of patients experiencing grade 3 or 4 events, primarily neutropenia, which affected 60.4% of them. Our findings suggest that 5-FU combined with platinum-based chemotherapy is a viable, cost-effective alternative for advanced HCC treatment in resource-limited settings, particularly compared to ICIs and multi-kinase inhibitors, with significant implications for developing countries.
Collapse
Affiliation(s)
- Se Jun Park
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
| | - Kabsoo Shin
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
| | - In-Ho Kim
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
| | - MyungAh Lee
- Division of Medical Oncology, Department of Internal Medicine, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
- Cancer Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
| | - Tae Ho Hong
- Department of General Surgery, Seoul St. Mary's Hospital, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Secho-gu, Seoul, Korea
| | - Hyunho Kim
- Division of Medical Oncology, Department of Internal Medicine, College of Medicine, St. Vincent's Hospital, The Catholic University of Korea, 93 Jungbu-daero, Paldal-gu, Suwon, 16247, Korea.
| |
Collapse
|
71
|
Aggeletopoulou I, Pantzios S, Triantos C. Personalized Immunity: Neoantigen-Based Vaccines Revolutionizing Hepatocellular Carcinoma Treatment. Cancers (Basel) 2025; 17:376. [PMID: 39941745 PMCID: PMC11815775 DOI: 10.3390/cancers17030376] [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: 12/20/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 02/16/2025] Open
Abstract
Hepatocellular carcinoma (HCC), the most prevalent form of primary liver cancer, presents significant therapeutic challenges due to its molecular complexity, late-stage diagnosis, and inherent resistance to conventional treatments. The intermediate to low mutational burden in HCC and its ability to evade the immune system through multiple mechanisms complicate the development of effective therapies. Recent advancements in immunotherapy, particularly neoantigen-based vaccines, offer a promising, personalized approach to HCC treatment. Neoantigens are tumor-specific peptides derived from somatic mutations in tumor cells. Unlike normal cellular antigens, neoantigens are foreign to the immune system, making them highly specific targets for immunotherapy. Neoantigens arise from genetic alterations such as point mutations, insertions, deletions, and gene fusions, which are expressed as neoepitopes that are not present in healthy tissues, thus evading the immune tolerance mechanisms that typically protect normal cells. Preclinical and early-phase clinical studies of neoantigen-based vaccines have shown promising results, demonstrating the ability of these vaccines to elicit robust T cell responses against HCC. The aim of the current review is to provide an in-depth exploration of the therapeutic potential of neoantigen-based vaccines in HCC, focusing on neoantigen identification, vaccine platforms, and their integration with immune checkpoint inhibitors to enhance immunogenicity. It also evaluates preclinical and clinical data on efficacy and safety while addressing challenges in clinical translation. By taking advantage of the unique antigenic profile of each patient's tumor, neoantigen-based vaccines represent a promising approach in the treatment of HCC, offering the potential for improved patient outcomes, long-term remission, and a shift towards personalized, precision medicine in liver cancer therapy.
Collapse
Affiliation(s)
- Ioanna Aggeletopoulou
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece;
| | - Spyridon Pantzios
- Hepatogastroenterology Unit, Academic Department of Internal Medicine, General Oncology Hospital of Kifissia “Agioi Anargyroi”, National and Kapodistrian University of Athens, 14564 Athens, Greece
| | - Christos Triantos
- Division of Gastroenterology, Department of Internal Medicine, University Hospital of Patras, 26504 Patras, Greece;
| |
Collapse
|
72
|
Zhao K, Karimi A, Kelly L, Petre E, Marinelli B, Alexander ES, Sotirchos VS, Erinjeri JP, Covey A, Sofocleous CT, Harding JJ, Jarnagin W, Sigel C, Vakiani E, Ziv E, Yarmohammadi H. TP53 Mutation Predicts Worse Survival and Earlier Local Progression in Patients with Hepatocellular Carcinoma Treated with Transarterial Embolization. Curr Oncol 2025; 32:51. [PMID: 39851967 PMCID: PMC11764326 DOI: 10.3390/curroncol32010051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 12/30/2024] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
The aim of this study was to evaluate associations between TP53 status and outcomes after transarterial embolization (TAE) for the treatment of patients with hepatocellular carcinoma (HCC). This single-institution study included patients from 1/2014 to 6/2022 who underwent TAE of HCC and genomic analysis of tumoral tissue. The primary outcome was overall survival (OS) with relation to TP53 status, and the secondary outcome was the time to progression. Survival analysis was performed using the Kaplan-Meier method. The time to progression with death or the last patient contact without progression as competing risks were used to obtain a cumulative incidence function, and the association with TP53 status was evaluated using the Gray test. In total, 75 patients (63 men) with a median age of 70.0 (IQR 62.0-76.3) years were included. Of these, 26/75 (34.7%) patients had TP53-mutant HCC. Patients with TP53-mutant HCC had a significantly worse median OS of 15.2 (95% CI, 9.5-29.3) months, versus 31.2 (95% CI, 21.2-52.4) months as the median OS (p = 0.023) for TP53 wild-type HCC. Competing risk analysis showed a shorter time to local hepatic progression (at the site of the previously treated tumor) after TAE in patients with TP53-mutant HCC. The cumulative incidences of local progression at 6 and 12 months for TP53-mutant HCC were 65.4% and 84.6%, versus 40.8% and 55.1% for TP53 wild-type HCC (p = 0.0072). A TP53 mutation may predict a worse overall survival and a shorter time to local progression in HCC patients treated with TAE.
Collapse
Affiliation(s)
- Ken Zhao
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Anita Karimi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Luke Kelly
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Elena Petre
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Brett Marinelli
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Erica S. Alexander
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Vlasios S. Sotirchos
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Joseph P. Erinjeri
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Anne Covey
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Constantinos T. Sofocleous
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - James J. Harding
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - William Jarnagin
- Department of Surgery, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA;
| | - Carlie Sigel
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.S.); (E.V.)
| | - Efsevia Vakiani
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA; (C.S.); (E.V.)
| | - Etay Ziv
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| | - Hooman Yarmohammadi
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA (E.P.); (B.M.); (E.S.A.); (V.S.S.); (J.P.E.); (A.C.); (C.T.S.); (E.Z.); (H.Y.)
| |
Collapse
|
73
|
Hermán-Sánchez N, Del Rio-Moreno M, Ciria R, Sánchez-Frias ME, Fernández-Barrena MG, Uriarte I, Chicano-Galvez E, Ortea I, Peralbo-Molina Á, Briceño J, Avila MA, Rodríguez-Perálvarez M, Luque RM, López-Cánovas JL, Gahete MD. Quantitative proteomic analysis unveils a critical role of VARS1 in hepatocellular carcinoma aggressiveness through the modulation of MAGI1 expression. Mol Cancer 2025; 24:15. [PMID: 39810176 PMCID: PMC11731432 DOI: 10.1186/s12943-024-02206-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 12/24/2024] [Indexed: 01/16/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) genetic/transcriptomic signatures have been widely described. However, its proteomic characterization is incomplete. We performed non-targeted quantitative proteomics of HCC samples and explored its clinical, functional, and molecular consequences. METHODS Non-targeted quantitative proteomics were performed on cytosolic and nuclear fractions of liver samples [HCC vs. non-tumour adjacent tissue (NTAT), n = 42 patients]. Changes were confirmed in 7 in silico HCC cohorts. Functional and molecular implications were evaluated on HCC-derived cell lines after silencing/overexpressing VARS1 and/or MAGI1. VARS1-overexpressing Hep3B cells were used for in vivo studies [Extreme Limiting Dilution Assay (ELDA) and orthotopic tumour formation]. Quantitative proteomics were performed on VARS1-overexpressing HCC cell lines. RESULTS Quantitative proteomics revealed the dysregulation of the cytosolic and nuclear proteomes in HCC, and defined two proteomic HCC subgroups, the most aggressive associated to the dysregulation of the aminoacyl-tRNA synthetases (ARSs). ARSs dysregulation was corroborated in in silico HCC cohorts and associated to poor prognosis. Patients with ARSs upregulation had genomic/transcriptomic characteristics of the proliferative HCC. Valine tRNA-aminoacyl synthetase (VARS1) was the ARSs most consistently overexpressed and associated to aggressiveness. VARS1 modulation (silencing/overexpression) altered tumour establishment-associated parameters in vitro and/or in vivo. Quantitative proteomics on cells overexpressing VARS1 and rescue experiments identified the downregulation of MAGI1, a tumour suppressor in HCC, as a mediator of VARS1 function. CONCLUSIONS Quantitative proteomics defines two prognosis-related proteomic HCC subgroups. ARSs machinery is dysregulated in the aggressive subgroup, bearing potential as prognostic biomarkers. VARS1 promotes aggressiveness through the modulation of MAGI1, representing a novel targetable vulnerability in HCC.
Collapse
Affiliation(s)
- Natalia Hermán-Sánchez
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Mercedes Del Rio-Moreno
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Rubén Ciria
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Marina E Sánchez-Frias
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Anatomical Pathology Department, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Maite G Fernández-Barrena
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Iker Uriarte
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
| | - Eduardo Chicano-Galvez
- IMIBIC Mass Spectrometry and Molecular Imaging Unit (IMSMI), Reina Sofía University Hospital, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba (UCO), Cordoba, 14004, Spain
| | - Ignacio Ortea
- Centro de Investigación en Nanomateriales y Nanotecnología (CINN-CSIC), Instituto de Investigación Sanitaria del Principado de Asturias (ISPA), Oviedo, 33011, Spain
| | - Ángela Peralbo-Molina
- IMIBIC Mass Spectrometry and Molecular Imaging Unit (IMSMI), Reina Sofía University Hospital, Maimónides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba (UCO), Cordoba, 14004, Spain
| | - Javier Briceño
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Unit of Hepatobiliary Surgery and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Matías A Avila
- Hepatology Laboratory, Solid Tumors Program, CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), CIMA, University of Navarra, Instituto de Salud Carlos III, Pamplona, Spain
- Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Manuel Rodríguez-Perálvarez
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
- Department of Hepatology and Liver Transplantation, Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Raúl M Luque
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Juan L López-Cánovas
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain
| | - Manuel D Gahete
- Department of Cell Biology, Physiology, and Immunology, University of Córdoba, CIBER Pathophysiology of Obesity and Nutrition (CIBERobn), Córdoba, 14004, Spain.
- Maimónides Institute of Biomedical Research of Córdoba (IMIBIC), Reina Sofía University Hospital, Córdoba, 14004, Spain.
| |
Collapse
|
74
|
Qiao J, Yu Z, Zhou H, Wang W, Wu H, Ye J. The Pentose Phosphate Pathway: From Mechanisms to Implications for Gastrointestinal Cancers. Int J Mol Sci 2025; 26:610. [PMID: 39859324 PMCID: PMC11765532 DOI: 10.3390/ijms26020610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 12/27/2024] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
The pentose phosphate pathway (PPP), traditionally recognized for its role in generating nicotinamide adenine dinucleotide phosphate (NADPH) and ribose-5-phosphate (R5P), has emerged as a critical metabolic hub with involvements in various gastrointestinal (GI) cancers. The PPP plays crucial roles in the initiation, development, and tumor microenvironment (TME) of GI cancers by modulating redox homeostasis and providing precursors for nucleotide biosynthesis. Targeting PPP enzymes and their regulatory axis has been a potential strategy in anti-GI cancer therapies. In this review, we summarize the regulatory mechanisms of PPP enzymes, elucidate the relationships between the PPP and TME's elements, and discuss the therapeutic potential of targeting the PPP in GI cancers.
Collapse
Affiliation(s)
- Jincheng Qiao
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (J.Q.); (Z.Y.)
- Cancer Institute (A Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Zhengchen Yu
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (J.Q.); (Z.Y.)
- Cancer Institute (A Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Han Zhou
- Cancer Institute (A Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Wankun Wang
- Department of Surgical Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310016, China;
| | - Hao Wu
- Cancer Institute (A Key Laboratory for Cancer Prevention & Intervention, China National Ministry of Education), The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China;
| | - Jun Ye
- Department of Gastroenterology, The Second Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310009, China; (J.Q.); (Z.Y.)
| |
Collapse
|
75
|
Varanasi SK, Chen D, Liu Y, Johnson MA, Miller CM, Ganguly S, Lande K, LaPorta MA, Hoffmann FA, Mann TH, Teneche MG, Casillas E, Mangalhara KC, Mathew V, Sun M, Jensen IJ, Farsakoglu Y, Chen T, Parisi B, Deota S, Havas A, Lee J, Chung HK, Schietinger A, Panda S, Williams AE, Farber DL, Dhar D, Adams PD, Feng GS, Shadel GS, Sundrud MS, Kaech SM. Bile acid synthesis impedes tumor-specific T cell responses during liver cancer. Science 2025; 387:192-201. [PMID: 39787217 DOI: 10.1126/science.adl4100] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Revised: 08/26/2024] [Accepted: 10/30/2024] [Indexed: 01/12/2025]
Abstract
The metabolic landscape of cancer greatly influences antitumor immunity, yet it remains unclear how organ-specific metabolites in the tumor microenvironment influence immunosurveillance. We found that accumulation of primary conjugated and secondary bile acids (BAs) are metabolic features of human hepatocellular carcinoma and experimental liver cancer models. Inhibiting conjugated BA synthesis in hepatocytes through deletion of the BA-conjugating enzyme bile acid-CoA:amino acid N-acyltransferase (BAAT) enhanced tumor-specific T cell responses, reduced tumor growth, and sensitized tumors to anti-programmed cell death protein 1 (anti-PD-1) immunotherapy. Furthermore, different BAs regulated CD8+ T cells differently; primary BAs induced oxidative stress, whereas the secondary BA lithocholic acid inhibited T cell function through endoplasmic reticulum stress, which was countered by ursodeoxycholic acid. We demonstrate that modifying BA synthesis or dietary intake of ursodeoxycholic acid could improve tumor immunotherapy in liver cancer model systems.
Collapse
Affiliation(s)
- Siva Karthik Varanasi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Dan Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Yingluo Liu
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Melissa A Johnson
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Cayla M Miller
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Souradipta Ganguly
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Kathryn Lande
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Michael A LaPorta
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Filipe Araujo Hoffmann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Thomas H Mann
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Marcos G Teneche
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Eduardo Casillas
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Kailash C Mangalhara
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Varsha Mathew
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Ming Sun
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Isaac J Jensen
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Yagmur Farsakoglu
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Timothy Chen
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Bianca Parisi
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Shaunak Deota
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Aaron Havas
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Jin Lee
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - H Kay Chung
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Andrea Schietinger
- Immunology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, New York, NY, USA
- Parker Institute for Cancer Immunotherapy, New York, NY, USA
| | - Satchidananda Panda
- Regulatory Biology Laboratory, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - April E Williams
- The Razavi Newman Integrative Genomics and Bioinformatics Core Facility, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Donna L Farber
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, NY, USA
- Department of Surgery, Columbia University Irving Medical Center, New York, NY, USA
| | - Debanjan Dhar
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
- Department of Medicine, School of Medicine, University of California, San Diego, CA, USA
| | - Peter D Adams
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Gen-Sheng Feng
- Department of Pathology, School of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Gerald S Shadel
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| | - Mark S Sundrud
- Department of Microbiology and Immunology, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Department of Medicine, Geisel School of Medicine at Dartmouth, Hanover, NH, USA
- Center for Digestive Health, Dartmouth Health, Lebanon, NH, USA
- Dartmouth Cancer Center, Dartmouth Health, Lebanon, NH, USA
| | - Susan M Kaech
- NOMIS Center for Immunobiology and Microbial Pathogenesis, Salk Institute for Biological Studies, La Jolla, CA, USA
| |
Collapse
|
76
|
Muchtar A, Onomura D, Ding D, Nishitsuji H, Shimotohno K, Okada S, Ueda K, Watashi K, Wakita T, Iida K, Yoshiyama H, Iizasa H. MicroRNA-3145 as a potential therapeutic target for hepatitis B virus: inhibition of viral replication via downregulation of HBS and HBX. Front Microbiol 2025; 15:1499216. [PMID: 39834379 PMCID: PMC11743939 DOI: 10.3389/fmicb.2024.1499216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 12/02/2024] [Indexed: 01/22/2025] Open
Abstract
Current treatments for hepatitis B virus (HBV), such as interferons and nucleic acid analogs, have limitations due to side effects like depression and the development of drug-resistant mutants, highlighting the need for new therapeutic approaches. In this study, we identified microRNA-3145 (miR-3145) as a host-derived miRNA with antiviral activity that is upregulated in primary hepatocytes during HBV infection. The expression of its precursor, pri-miR-3145, increased in response to the the virus infection, and miR-3145 downregulated the hepatitis B virus S (HBS) antigen and hepatitis B virus X (HBX), thereby inhibiting viral replication. The binding site for miR-3145 was located in the HBV polymerase (pol) region, as experimentally confirmed. Moreover, overexpression of HBS and HBX induced pri-miR-3145 expression through endoplasmic reticulum stress. The expression of pri-miR-3145 showed a strong correlation with the Nance-Horan syndrome-like 1 (NHSL1) gene, as it is encoded within an intron of NHSL1, and higher NHSL1 expression in hepatocellular carcinoma patients with HBV infection was associated with better prognosis. These findings suggest that miR-3145-3p, along with small molecules targeting its binding sites, holds promise as a potential therapeutic candidate for HBV treatment.
Collapse
Affiliation(s)
- Amrizal Muchtar
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
- Faculty of Medicine, Universitas Muslim Indonesia, Makassar, Indonesia
| | - Daichi Onomura
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
- Division of Virology, Department of Infection and Immunity, Jichi Medical University, Shimotsuke, Japan
| | - Dan Ding
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
- Department of Pathology, Changhai Hospital, Naval Medical University, Shanghai, China
| | - Hironori Nishitsuji
- Department of Virology and Parasitology, Fujita Health University School of Medicine, Toyoake, Japan
| | - Kunitada Shimotohno
- Genome Medical Sciences Project, National Center for Global Health and Medicine, Ichikawa, Japan
| | - Shunpei Okada
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Keiji Ueda
- Division of Virology, Department of Microbiology and Immunology, Osaka University Graduate School of Medicine, Suita, Japan
| | - Koichi Watashi
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Takaji Wakita
- Department of Virology II, National Institute of Infectious Diseases, Tokyo, Japan
| | - Kei Iida
- Faculty of Science and Engineering, Kindai University, Higashiōsaka, Japan
- Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hironori Yoshiyama
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
| | - Hisashi Iizasa
- Department of Microbiology, Faculty of Medicine, Shimane University, Izumo, Japan
| |
Collapse
|
77
|
Mishra A, Huang SB, Dubash T, Burr R, Edd JF, Wittner BS, Cunneely QE, Putaturo VR, Deshpande A, Antmen E, Gopinathan KA, Otani K, Miyazawa Y, Kwak JE, Guay SY, Kelly J, Walsh J, Nieman LT, Galler I, Chan P, Lawrence MS, Sullivan RJ, Bardia A, Micalizzi DS, Sequist LV, Lee RJ, Franses JW, Ting DT, Brunker PAR, Maheswaran S, Miyamoto DT, Haber DA, Toner M. Tumor cell-based liquid biopsy using high-throughput microfluidic enrichment of entire leukapheresis product. Nat Commun 2025; 16:32. [PMID: 39746954 PMCID: PMC11696112 DOI: 10.1038/s41467-024-55140-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 11/28/2024] [Indexed: 01/04/2025] Open
Abstract
Circulating Tumor Cells (CTCs) in blood encompass DNA, RNA, and protein biomarkers, but clinical utility is limited by their rarity. To enable tumor epitope-agnostic interrogation of large blood volumes, we developed a high-throughput microfluidic device, depleting hematopoietic cells through high-flow channels and force-amplifying magnetic lenses. Here, we apply this technology to analyze patient-derived leukapheresis products, interrogating a mean blood volume of 5.83 liters from seven patients with metastatic cancer. High CTC yields (mean 10,057 CTCs per patient; range 100 to 58,125) reveal considerable intra-patient heterogeneity. CTC size varies within patients, with 67% overlapping in diameter with WBCs. Paired single-cell DNA and RNA sequencing identifies subclonal patterns of aneuploidy and distinct signaling pathways within CTCs. In prostate cancers, a subpopulation of small aneuploid cells lacking epithelial markers is enriched for neuroendocrine signatures. Pooling of CNV-confirmed CTCs enables whole exome sequencing with high mutant allele fractions. High-throughput CTC enrichment thus enables cell-based liquid biopsy for comprehensive monitoring of cancer.
Collapse
Affiliation(s)
- Avanish Mishra
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Shih-Bo Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA
| | - Taronish Dubash
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Risa Burr
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Jon F Edd
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ben S Wittner
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Quinn E Cunneely
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Victor R Putaturo
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Akansha Deshpande
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Ezgi Antmen
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Kaustav A Gopinathan
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Keisuke Otani
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Yoshiyuki Miyazawa
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Ji Eun Kwak
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sara Y Guay
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Justin Kelly
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - John Walsh
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Linda T Nieman
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - Isabella Galler
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - PuiYee Chan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Michael S Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA
| | - Ryan J Sullivan
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Aditya Bardia
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
- Hematology/Oncology, University of California, Los Angeles, USA
| | - Douglas S Micalizzi
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Lecia V Sequist
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Richard J Lee
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Joseph W Franses
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - David T Ting
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA
| | - Patricia A R Brunker
- Department of Pathology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA
| | - Shyamala Maheswaran
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA
| | - David T Miyamoto
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Department of Radiation Oncology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Broad Institute of MIT and Harvard, Cambridge, MA, 02142, USA.
| | - Daniel A Haber
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center and Harvard Medical School, Charlestown, MA, 02129, USA.
- Howard Hughes Medical Institute, Bethesda, MD, 20815, USA.
- Division of Hematology Oncology, Massachusetts General Hospital Cancer Center and Harvard Medical School, Boston, MA, 02114, USA.
| | - Mehmet Toner
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital and Harvard Medical School, Boston, MA, 02114, USA.
- Shriners Children's Boston, Boston, MA, 02114, USA.
| |
Collapse
|
78
|
Guo Y, Li T, Gong B, Hu Y, Wang S, Yang L, Zheng C. From Images to Genes: Radiogenomics Based on Artificial Intelligence to Achieve Non-Invasive Precision Medicine in Cancer Patients. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408069. [PMID: 39535476 PMCID: PMC11727298 DOI: 10.1002/advs.202408069] [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: 07/15/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
With the increasing demand for precision medicine in cancer patients, radiogenomics emerges as a promising frontier. Radiogenomics is originally defined as a methodology for associating gene expression information from high-throughput technologies with imaging phenotypes. However, with advancements in medical imaging, high-throughput omics technologies, and artificial intelligence, both the concept and application of radiogenomics have significantly broadened. In this review, the history of radiogenomics is enumerated, related omics technologies, the five basic workflows and their applications across tumors, the role of AI in radiogenomics, the opportunities and challenges from tumor heterogeneity, and the applications of radiogenomics in tumor immune microenvironment. The application of radiogenomics in positron emission tomography and the role of radiogenomics in multi-omics studies is also discussed. Finally, the challenges faced by clinical transformation, along with future trends in this field is discussed.
Collapse
Affiliation(s)
- Yusheng Guo
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Tianxiang Li
- Department of UltrasoundState Key Laboratory of Complex Severe and Rare DiseasesPeking Union Medical College HospitalChinese Academy of Medical. SciencesPeking Union Medical CollegeBeijing100730China
| | - Bingxin Gong
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Yan Hu
- Research Institute of Trustworthy Autonomous Systems and Department of Computer Science and EngineeringSouthern University of Science and TechnologyShenzhen518055China
| | - Sichen Wang
- School of Life Science and TechnologyComputational Biology Research CenterHarbin Institute of TechnologyHarbin150001China
| | - Lian Yang
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| | - Chuansheng Zheng
- Department of RadiologyUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhan430022China
- Hubei Key Laboratory of Molecular ImagingWuhan430022China
| |
Collapse
|
79
|
Li YT, Zeng XZ. Establishment and Validation of the Novel Necroptosis-related Genes for Predicting Stemness and Immunity of Hepatocellular Carcinoma via Machine-learning Algorithm. Comb Chem High Throughput Screen 2025; 28:146-165. [PMID: 39641162 DOI: 10.2174/0113862073271292231108113547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 12/07/2024]
Abstract
BACKGROUND Necroptosis, a recently identified mechanism of programmed cell death, exerts significant influence on various aspects of cancer biology, including tumor cell proliferation, stemness, metastasis, and immunosuppression. However, the role of necroptosis-related genes (NRGs) in Hepatocellular Carcinoma (HCC) remains elusive. METHODS In this study, we assessed the mutation signature, copy number variation, and expression of 37 NRGs in HCC using the TCGA-LIHC dataset. We further validated our results using the ICGC-LIRI-JP dataset. To construct our prognostic model, we utilized the least absolute shrinkage and selection operator (LASSO), and evaluated the predictive efficacy of the NRGs-score using various machine learning algorithms, including K-M curves, time-ROC curves, univariate and multivariate Cox regression, and nomogram. In addition, we analyzed immune infiltration using the CIBERSOFT and ssGSEA algorithms, calculated the stemness index through the one-class logistic regression (OCLR) algorithm, and performed anti-cancer stem cells (CSCs) drug sensitivity analysis using oncoPredict. Finally, we validated the expression of the prognostic NRGs through qPCR both in vitro and in vivo. RESULTS About 18 out of 37 NRGs were found to be differentially expressed in HCC and correlated with clinical outcomes. To construct a prognostic model, six signature genes (ALDH2, EZH2, PGAM5, PLK1, SQSTM1, and TARDBP) were selected using LASSO analysis. These genes were then employed to categorize HCC patients into two subgroups based on NRGs-score (low vs. high). A high NRGs score was associated with a worse prognosis. Furthermore, univariate and multivariate Cox regression analyses were performed to confirm the NRGs-score as an independent risk factor. These analyses revealed strong associations between NRGs-score and critical factors, such as AFP, disease stage, and tumor grade in the HCC cohort. NRGs-score effectively predicted the 1-, 3-, and 5-year survival of HCC patients. Immune infiltration analysis further revealed that the expression of immune checkpoint molecules was significantly enhanced in the high NRGs-score group. Stemness analysis in the HCC cohort showed that NRGs-score was positively correlated with mRNA stemness index, and patients with high NRGs-score were sensitive to CSCs inhibitors. The findings from the external validation cohort provided confirmation that the NRGs-score presented a trait with universal applicability in accurately predicting the survival of HCC. Additionally, the six prognostic genes were consistently differentially expressed in both the HCC cell line and the mouse HCC model. CONCLUSION Our study demonstrated the pivotal role of NRGs in promoting stemness and immune suppression in HCC and established a robust model which could successfully predict HCC prognosis.
Collapse
Affiliation(s)
- Yao-Ting Li
- Department of Forensic Science, Guangdong Police College, 500 Binjiang East Road, Guangzhou 510230, Guangdong, China
| | - Xue-Zhen Zeng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
80
|
Elkhadragy L, Carlino MJ, Jordan LR, Pennix T, Ismail N, Guzman G, Samuelson JP, Schook LB, Schachtschneider KM, Gaba RC. Development of a genetically tailored implantation hepatocellular carcinoma model in Oncopigs by somatic cell CRISPR editing. Dis Model Mech 2025; 18:dmm052079. [PMID: 39780710 PMCID: PMC11810043 DOI: 10.1242/dmm.052079] [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: 08/26/2024] [Accepted: 12/27/2024] [Indexed: 01/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is an aggressive disease with poor prognosis, necessitating preclinical models for evaluating novel therapies. Large-animal models are particularly valuable for assessing locoregional therapies, which are widely employed across HCC stages. This study aimed to develop a large-animal HCC model with tailored tumor mutations. The Oncopig, a genetically engineered pig with inducible TP53R167H and KRASG12D, was used in the study. Hepatocytes were isolated from Oncopigs and exposed to Cre recombinase in vitro to create HCC cells, and additional mutations were introduced by CRISPR/Cas9 knockout of PTEN and CDKN2A. These edits increased Oncopig HCC cell proliferation and migration. Autologous HCC cells with these CRISPR edits were implanted into Oncopigs using two approaches: ultrasound-guided percutaneous liver injections, which resulted in the development of localized intrahepatic masses, and portal vein injections, which led to multifocal tumors that regressed over time. Tumors developed by both approaches harbored PTEN and CDKN2A knockout mutations. This study demonstrates the feasibility of developing genetically tailored HCC tumors in Oncopigs using somatic cell CRISPR editing and autologous implantation, providing a valuable large-animal model for in vivo therapeutic assessment.
Collapse
Affiliation(s)
- Lobna Elkhadragy
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | | | - Luke R. Jordan
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Thomas Pennix
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Nahed Ismail
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Grace Guzman
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jonathan P. Samuelson
- Department of Veterinary Clinical Medicine, University of Illinois at Urbana-Champaign, Champaign, IL 61802, USA
| | - Lawrence B. Schook
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Champaign, IL 61801, USA
| | | | - Ron C. Gaba
- Department of Radiology, University of Illinois at Chicago, Chicago, IL 60612, USA
- Department of Pathology, University of Illinois at Chicago, Chicago, IL 60612, USA
| |
Collapse
|
81
|
Lu X, Zhang Y, Xue J, Evert M, Calvisi D, Chen X, Wang X. MAD2L1 supports MYC-driven liver carcinogenesis in mice and predicts poor prognosis in human hepatocarcinoma. Toxicol Sci 2025; 203:41-51. [PMID: 39316419 DOI: 10.1093/toxsci/kfae126] [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] [Indexed: 09/25/2024] Open
Abstract
Mitotic arrest-deficient 2 like 1 (MAD2L1) is a component of the mitotic spindle assembly checkpoint implicated in cancer cell proliferation and tumorigenesis. The functional role of MAD2L1 in hepatocellular carcinoma (HCC) has not been adequately investigated, especially in vivo. In the current manuscript, we sought to address the function of MAD2L1 in hepatocarcinogenesis. We found that MAD2L1 expression is upregulated in human HCCs, where its expression is associated with higher aggressive tumor grade, elevated proliferative activity, and poor prognosis. In human HCC cell lines, MAD2L1 knockdown led to decreased cell growth. Moreover, RNA-seq results demonstrated that MAD2L1 silencing induces the expression of genes associated with cell cycle, DNA replication, and various cancer-related pathways, supporting the critical role of MAD2L1 during HCC growth and differentiation. In a c-MYC-induced mouse HCC model, we revealed an increased expression of Mad2l1. Furthermore, Mad2l1 CRIPSR-mediated silencing prevented c-MYC-driven mouse liver development. Altogether, our study suggests that MAD2L1 plays a crucial role in hepatocarcinogenesis, and that its suppression could be a promising therapeutic strategy for treating human HCC. MAD2L1 plays a critical role in liver cancer development, silencing MAD2L1 reduced cell growth in vitro and inhibited c-MYC-driven liver cancer development in vivo. MAD2L1 suppression might be a promising therapeutic approach for treating human liver cancer.
Collapse
Affiliation(s)
- Xinjun Lu
- Department of Biliary-Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Ya Zhang
- Cancer Biology Program, University of Hawai'i Cancer Center, University of Hawai'i, Honolulu, HA 96813, United States
| | - Jiahao Xue
- Department of Biliary-Pancreatic Surgery, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510120, China
| | - Matthias Evert
- Institute of Pathology, University of Regensburg, Regensburg 93053, Germany
| | - Diego Calvisi
- Institute of Pathology, University of Regensburg, Regensburg 93053, Germany
| | - Xin Chen
- Cancer Biology Program, University of Hawai'i Cancer Center, University of Hawai'i, Honolulu, HA 96813, United States
| | - Xue Wang
- Cancer Biology Program, University of Hawai'i Cancer Center, University of Hawai'i, Honolulu, HA 96813, United States
| |
Collapse
|
82
|
Xiao J, Cao Y, Li X, Xu L, Wang Z, Huang Z, Mu X, Qu Y, Xu Y. Elucidation of Factors Affecting the Age-Dependent Cancer Occurrence Rates. Int J Mol Sci 2024; 26:275. [PMID: 39796131 PMCID: PMC11720044 DOI: 10.3390/ijms26010275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2024] [Revised: 12/24/2024] [Accepted: 12/29/2024] [Indexed: 01/13/2025] Open
Abstract
Cancer occurrence rates exhibit diverse age-related patterns, and understanding them may shed new and important light on the drivers of cancer evolution. This study systematically analyzes the age-dependent occurrence rates of 23 carcinoma types, focusing on their age-dependent distribution patterns, the determinants of peak occurrence ages, and the significant difference between the two genders. According to the SEER reports, these cancer types have two types of age-dependent occurrence rate (ADOR) distributions, with most having a unimodal distribution and a few having a bimodal distribution. Our modeling analyses have revealed that (1) the first type can be naturally and simply explained using two age-dependent parameters: the total number of stem cell divisions in an organ from birth to the current age and the availability levels of bloodborne growth factors specifically needed by the cancer (sub)type, and (2) for the second type, the first peak is due to viral infection, while the second peak can be explained as in (1) for each cancer type. Further analyses indicate that (i) the iron level in an organ makes the difference between the male and female cancer occurrence rates, and (ii) the levels of sex hormones are the key determinants in the onset age of multiple cancer types. This analysis deepens our understanding of the dynamics of cancer evolution shared by diverse cancer types and provides new insights that are useful for cancer prevention and therapeutic strategies, thereby addressing critical gaps in the current paradigm of oncological research.
Collapse
Affiliation(s)
- Jun Xiao
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Yangkun Cao
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Artificial Intelligence, Jilin University, Changchun 130012, China
| | - Xuan Li
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Long Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhihang Wang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Zhenyu Huang
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Xuechen Mu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
- School of Mathematics, Jilin University, Changchun 130012, China
| | - Yinwei Qu
- College of Computer Science and Technology, Jilin University, Changchun 130012, China; (J.X.); (X.L.); (Z.W.); (Z.H.)
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| | - Ying Xu
- Systems Biology Laboratory for Metabolic Reprogramming, School of Medicine, Southern University of Science and Technology, Shenzhen 518055, China; (Y.C.); (L.X.); (X.M.)
| |
Collapse
|
83
|
Requena D, Medico JA, Soto-Ugaldi LF, Shirani M, Saltsman JA, Torbenson MS, Coffino P, Simon SM. Liver cancer multiomics reveals diverse protein kinase A disruptions convergently produce fibrolamellar hepatocellular carcinoma. Nat Commun 2024; 15:10887. [PMID: 39738196 PMCID: PMC11685927 DOI: 10.1038/s41467-024-55238-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 12/03/2024] [Indexed: 01/01/2025] Open
Abstract
Fibrolamellar Hepatocellular Carcinoma (FLC) is a rare liver cancer characterized by a fusion oncokinase of the genes DNAJB1 and PRKACA, the catalytic subunit of protein kinase A (PKA). A few FLC-like tumors have been reported showing other alterations involving PKA. To better understand FLC pathogenesis and the relationships among FLC, FLC-like, and other liver tumors, we performed a massive multi-omics analysis. RNA-seq data of 1412 liver tumors from FLC, hepatocellular carcinoma, hepatoblastoma and intrahepatic cholangiocarcinoma are analyzed, obtaining transcriptomic signatures unrestricted by experimental processing methods. These signatures reveal which dysregulations are unique to specific tumors and which are common to all liver cancers. Moreover, the transcriptomic FLC signature identifies a unifying phenotype for all FLC tumors regardless of how PKA was activated. We study this signature at multi-omics and single-cell levels in the first spatial transcriptomic characterization of FLC, identifying the contribution of tumor, normal, stromal, and infiltrating immune cells. Additionally, we study FLC metastases, finding small differences from the primary tumors.
Collapse
Affiliation(s)
- David Requena
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Jack A Medico
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Luis F Soto-Ugaldi
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Mahsa Shirani
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - James A Saltsman
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | | | - Philip Coffino
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA
| | - Sanford M Simon
- Laboratory of Cellular Biophysics, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
84
|
Yu X, Sarabia S, Urbicain M, Somvanshi S, Patel R, Tran TM, Yeh YP, Chang KS, Lo YT, Epps J, Scorsone KA, Chiu HS, Hollingsworth EF, Perez CR, Najaf Panah MJ, Zorman B, Finegold M, Goss JA, Alaggio R, Roy A, Fisher KE, Heczey A, Woodfield S, Vasudevan S, Patel K, Chen TW, Lopez-Terrada D, Sumazin P. Asynchronous Transitions from Hepatoblastoma to Carcinoma in High-Risk Pediatric Tumors. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.12.24.630261. [PMID: 39763896 PMCID: PMC11703271 DOI: 10.1101/2024.12.24.630261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Most malignant hepatocellular tumors in children are classified as either hepatoblastoma (HB) or hepatocellular carcinoma (HCC), but some tumors demonstrate features of both HB and HCC1-3. These tumors have been recognized under a provisional diagnostic category by the World Health Organization and are distinguished from HB and HCC by a combination of histological, immunohistochemical, and molecular features4-6. Their outcomes and cellular composition remain an open question7-9. The heterogeneous histological and molecular profiles of hepatoblastomas with carcinoma features (HBCs)4 may result from cells with combined HB and HCC characteristics (HBC cells) or from mixtures of cells displaying either HB or HCC signatures. We used multiomics profiling to show that HBCs are mixtures of HB, HBC, and HCC cell types. HBC cells are more chemoresistant than HB cells, and their chemoresistance-a driver of poor outcomes10-12-is determined by their cell types, genetic alterations, and embryonic differentiation stages. We showed that the prognosis of HBCs is significantly worse than that of HBs. We also showed that HBC cells are derived from HB cells at early hepatoblast differentiation stages, that aberrant activation of WNT-signaling initiates HBC transformation, and that WNT inhibition promotes differentiation and increases sensitivity to chemotherapy. Furthermore, our analysis revealed that each HBC is the product of multiple HB-to-HBC and HBC-to-HCC transitions. Thus, multiomics profiling of HBCs provided key insights into their biology and resolved major questions regarding the etiology of these childhood liver tumors.
Collapse
Affiliation(s)
- Xinjian Yu
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Stephen Sarabia
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Martin Urbicain
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Sonal Somvanshi
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Roma Patel
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Tuan M Tran
- Department of Systems Biology, Department of Genetics, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Yen-Ping Yeh
- Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-Devices, and Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Keng-Shih Chang
- Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-Devices, and Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Yi-Tzu Lo
- Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-Devices, and Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Jessica Epps
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Kathleen A. Scorsone
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Hua-Sheng Chiu
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Emporia Faith Hollingsworth
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Cintia R. Perez
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | | | - Barry Zorman
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Milton Finegold
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - John A. Goss
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Rita Alaggio
- Department of Pathology, Bambino Gesù Children’s Hospital, IRCCS, Rome, Italy
| | - Angshumoy Roy
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Kevin E. Fisher
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Andras Heczey
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| | - Sarah Woodfield
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Sanjeev Vasudevan
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Divisions of Pediatric Surgery and Surgical Research, Michael E. DeBakey Department of Surgery, Baylor College of Medicine, Houston, TX, USA
| | - Kalyani Patel
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Ting-Wen Chen
- Biological Science and Technology, Center for Intelligent Drug Systems and Smart Bio-Devices, and Institute of Bioinformatics and Systems Biology, National Yang Ming Chiao Tung University, Hsinchu, Taiwan
| | - Dolores Lopez-Terrada
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
- Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Pavel Sumazin
- Department of Pediatrics, Texas Children’s Cancer Center, Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
85
|
Tian H, Tian Y, Li D, Zhao M, Luo Q, Kong L, Qin T. Artificial intelligence model predicts M2 macrophage levels and HCC prognosis with only globally labeled pathological images. Front Oncol 2024; 14:1474155. [PMID: 39759153 PMCID: PMC11695232 DOI: 10.3389/fonc.2024.1474155] [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: 08/22/2024] [Accepted: 12/09/2024] [Indexed: 01/07/2025] Open
Abstract
Background and aims The levels of M2 macrophages are significantly associated with the prognosis of hepatocellular carcinoma (HCC), however, current detection methods in clinical settings remain challenging. Our study aims to develop a weakly supervised artificial intelligence model using globally labeled histological images, to predict M2 macrophage levels and forecast the prognosis of HCC patients by integrating clinical features. Methods CIBERSORTx was used to calculate M2 macrophage abundance. We developed a slide-level, weakly-supervised clustering method for Whole Slide Images (WSIs) by integrating Masked Autoencoders (MAE) with ResNet-32t to predict M2 macrophage abundance. Results We developed an MAE-ResNet model to predict M2 macrophage levels using WSIs. In the testing dataset, the area under the curve (AUC) (95% CI) was 0.73 (0.59-0.87). We constructed a Cox regression model showing that the predicted probabilities of M2 macrophage abundance were negatively associated with the prognosis of HCC (HR=1.89, p=0.031). Furthermore, we incorporated clinical data, screened variables using Lasso regression, and built the comprehensive prediction model that better predicted prognosis. (HR=2.359, p=0.001). Conclusion Our models effectively predicted M2 macrophage levels and HCC prognosis. The findings suggest that our models offer a novel method for determining biomarker levels and forecasting prognosis, eliminating additional clinical tests, thereby delivering substantial clinical benefits.
Collapse
Affiliation(s)
- Huiyuan Tian
- Department of Scientific Research and Foreign Affairs, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, China
| | - Yongshao Tian
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Dujuan Li
- Department of Pathology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, China
| | - Minfan Zhao
- School of Computer Science and Technology, University of Science and Technology of China, Hefei, Anhui, China
| | - Qiankun Luo
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, China
| | - Lingfei Kong
- Department of Pathology, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, China
| | - Tao Qin
- Department of Hepatobiliary and Pancreatic Surgery, Henan Provincial People’s Hospital, Zhengzhou University People’s Hospital, Zhengzhou, Henan, China
| |
Collapse
|
86
|
Li S, Lin Y, Gao X, Zeng D, Cen W, Su Y, Su J, Zeng C, Huang Z, Zeng H, Huang S, Tang M, Li X, Luo M, Huang Z, Liang R, Ye J. Integrative multi-omics analysis reveals a novel subtype of hepatocellular carcinoma with biological and clinical relevance. Front Immunol 2024; 15:1517312. [PMID: 39712016 PMCID: PMC11659151 DOI: 10.3389/fimmu.2024.1517312] [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: 10/25/2024] [Accepted: 11/18/2024] [Indexed: 12/24/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) is a highly heterogeneous tumor, and the development of accurate predictive models for prognosis and drug sensitivity remains challenging. Methods We integrated laboratory data and public cohorts to conduct a multi-omics analysis of HCC, which included bulk RNA sequencing, proteomic analysis, single-cell RNA sequencing (scRNA-seq), spatial transcriptomics sequencing (ST-seq), and genome sequencing. We constructed a tumor purity (TP) and tumor microenvironment (TME) prognostic risk model. Proteomic analysis validated the TP-TME-related signatures. Joint analysis of scRNA-seq and ST-seq revealed characteristic clusters associated with TP high-risk subtypes, and immunohistochemistry confirmed the expression of key genes. We conducted functional enrichment analysis, transcription factor activity inference, cell-cell interaction, drug efficacy analysis, and mutation information analysis to identify a novel subtype of HCC. Results Our analyses constructed a robust HCC prognostic risk prediction model. The patients with TP-TME high-risk subtypes predominantly exhibit hypoxia and activation of the Wnt/beta-catenin, Notch, and TGF-beta signaling pathways. Furthermore, we identified a novel subtype, XPO1+Epithelial. This subtype expresses signatures of the TP risk subtype and aligns with the biological behavior of high-risk patients. Additional analyses revealed that XPO1+Epithelial is influenced primarily by fibroblasts via ligand-receptor interactions, such as FN1-(ITGAV+ITGB1), and constitute a significant component of the TP-TME subtype. Moreover, XPO1+Epithelial interact with monocytes/macrophages, T/NK cells, and endothelial cells through ligand-receptor pairs, including MIF-(CD74+CXCR4), MIF-(CD74+CD44), and VEGFA-VEGFR1R2, respectively, thereby promoting the recruitment of immune-suppressive cells and angiogenesis. The ST-seq cohort treated with Tyrosine Kinase Inhibitors (TKIs) and Programmed Cell Death Protein 1 (PD-1) presented elevated levels of TP and TME risk subtype signature genes, as well as XPO1+Epithelial, T-cell, and endothelial cell infiltration in the treatment response group. Drug sensitivity analyses indicated that TP-TME high-risk subtypes, including sorafenib and pembrolizumab, were associated with sensitivity to multiple drugs. Further exploratory analyses revealed that CTLA4, PDCD1, and the cancer antigens MSLN, MUC1, EPCAM, and PROM1 presented significantly increase expression levels in the high-risk subtype group. Conclusions This study constructed a robust prognostic model for HCC and identified novel subgroups at the single-cell level, potentially assisting in the assessment of prognostic risk for HCC patients and facilitating personalized drug therapy.
Collapse
Affiliation(s)
- Shizhou Li
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yan Lin
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xing Gao
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Dandan Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Weijie Cen
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Yuejiao Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jingting Su
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Can Zeng
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhenbo Huang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Haoyu Zeng
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Shilin Huang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Minchao Tang
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Xiaoqing Li
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Min Luo
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Zhihu Huang
- Department of Clinical Laboratory, Minzu Hospital Guangxi Zhuang Autonomous Region, Affiliated Minzu Hospital of Guangxi Medical University, Nanning, Guangxi, ;China
| | - Rong Liang
- Department of Medical Oncology, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| | - Jiazhou Ye
- Department of Hepatobiliary Surgery, Guangxi Medical University Cancer Hospital, Nanning, Guangxi, ;China
| |
Collapse
|
87
|
Wang X, Gao L, Li H, Ma Y, Wang B, Gu B, Li X, Xiang L, Bai Y, Ma C, Chen H. Integrative analysis of multi-omics data identified PLG as key gene related to Anoikis resistance and immune phenotypes in hepatocellular carcinoma. J Transl Med 2024; 22:1104. [PMID: 39633373 PMCID: PMC11616313 DOI: 10.1186/s12967-024-05858-5] [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: 07/25/2024] [Accepted: 11/06/2024] [Indexed: 12/07/2024] Open
Abstract
PURPOSE The extracellular matrix (ECM) plays a pivotal role in the initiation and progression of hepatocellular carcinoma (HCC) by facilitating the proliferation of HCC cells and enabling resistance to Anoikis. ECM also provide structural support that aids in the invasion of HCC cells, thereby influencing the tumor microenvironment. Due to genetic variations and molecular heterogeneity, significant challenges exist in the treatment of HCC, particularly with immunotherapy, which frequently leads to immune tolerance and suboptimal immune responses. Therefore, there is an urgent need for a multi-omics-based classification system for HCC that clarifies the molecular mechanisms underlying the establishment of immune phenotypes and Anoikis resistance in HCC cells. In this study, we employed advanced clustering algorithms to analyze and integrate multi-omics data from HCC patients, with the objective of identifying key genes that possess prognostic potential associated with the Anoikis resistance phenotype. This methodology resulted in the development of a consensus machine learning-driven signature (CMLS), which demonstrates robust predictive capabilities by examining variations in epigenetics, transcription, and immune metabolism, as well as their effects on the core differential gene, plasminogen (PLG). RESULTS The integrated multi-omics approach has identified PLG as a critical node within the gene regulatory network associated with Anoikis resistance and immunometabolic phenotypes. As an independent risk factor for poor prognosis in patients with HCC, PLG facilitates Anoikis resistance and enhances the migration of HCC cells. This study provides novel insights into the molecular subtypes of HCC through the application of robust clustering algorithms based on multi-omics data. The constructed CMLS serves as a valuable tool for early prognostic prediction and for screening potential drug candidates that may enhance the efficacy of immunotherapy, thereby establishing a foundation for personalized treatment strategies in HCC. CONCLUSIONS Our data underscore the pivotal role of PLG in the development of Anoikis resistance and the immunometabolic phenotype in HCC cells. Furthermore, we present compelling experimental evidence that PLG functions as a significant tumor promoter, suggesting its potential as a target for the formulation of tailored therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Xueyan Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lei Gao
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Haiyuan Li
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yanling Ma
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Bofang Wang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Baohong Gu
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Xuemei Li
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Lin Xiang
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Department of Pathology, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yuping Bai
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
- Department of Nuclear Magnetic, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Chenhui Ma
- Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Hao Chen
- Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Department of Surgical Oncology, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
- Key Laboratory of Digestive System Tumors of Gansu Province, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
88
|
Destefanis E, Sighel D, Dalfovo D, Gilmozzi R, Broso F, Cappannini A, Bujnicki J, Romanel A, Dassi E, Quattrone A. The three YTHDF paralogs and VIRMA are strong cross-histotype tumor driver candidates among m 6A core genes. NAR Cancer 2024; 6:zcae040. [PMID: 39411658 PMCID: PMC11474903 DOI: 10.1093/narcan/zcae040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Revised: 09/04/2024] [Accepted: 10/09/2024] [Indexed: 10/19/2024] Open
Abstract
N6-Methyladenosine (m6A) is the most abundant internal modification in mRNAs. Despite accumulating evidence for the profound impact of m6A on cancer biology, there are conflicting reports that alterations in genes encoding the m6A machinery proteins can either promote or suppress cancer, even in the same tumor type. Using data from The Cancer Genome Atlas, we performed a pan-cancer investigation of 15 m6A core factors in nearly 10000 samples from 31 tumor types to reveal underlying cross-tumor patterns. Altered expression, largely driven by copy number variations at the chromosome arm level, results in the most common mode of dysregulation of these factors. YTHDF1, YTHDF2, YTHDF3 and VIRMA are the most frequently altered factors and the only ones to be uniquely altered when tumors are grouped according to the expression pattern of the m6A factors. These genes are also the only ones with coherent, pan-cancer predictive power for progression-free survival. On the contrary, METTL3, the most intensively studied m6A factor as a cancer target, shows much lower levels of alteration and no predictive power for patient survival. Therefore, we propose the non-enzymatic YTHDF and VIRMA genes as preferred subjects to dissect the role of m6A in cancer and as priority cancer targets.
Collapse
Affiliation(s)
- Eliana Destefanis
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Denise Sighel
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Davide Dalfovo
- Laboratory of Bioinformatics and Computational Biology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Riccardo Gilmozzi
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Francesca Broso
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Andrea Cappannini
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, PL-02-109 Warsaw, Poland
| | - Janusz M Bujnicki
- Laboratory of Bioinformatics and Protein Engineering, International Institute of Molecular and Cell Biology in Warsaw, PL-02-109 Warsaw, Poland
| | - Alessandro Romanel
- Laboratory of Bioinformatics and Computational Biology, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Erik Dassi
- Laboratory of RNA Regulatory Networks, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| | - Alessandro Quattrone
- Laboratory of Translational Genomics, Department of Cellular, Computational and Integrative Biology - CIBIO, University of Trento, Trento 38123, Italy
| |
Collapse
|
89
|
Dantzer C, Dif L, Vaché J, Basbous S, Billottet C, Moreau V. Specific features of ß-catenin-mutated hepatocellular carcinomas. Br J Cancer 2024; 131:1871-1880. [PMID: 39261716 PMCID: PMC11628615 DOI: 10.1038/s41416-024-02849-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024] Open
Abstract
CTNNB1, encoding the ß-catenin protein, is a key oncogene contributing to liver carcinogenesis. Hepatocellular carcinoma (HCC) is the most common form of primary liver cancer in adult, representing the third leading cause of cancer-related death. Aberrant activation of the Wnt/ß-catenin pathway, mainly due to mutations of the CTNNB1 gene, is observed in a significant subset of HCC. In this review, we first resume the major recent advances in HCC classification with a focus on CTNNB1-mutated HCC subclass. We present the regulatory mechanisms involved in β-catenin stabilisation, transcriptional activity and binding to partner proteins. We then describe specific phenotypic characteristics of CTNNB1-mutated HCC thanks to their unique gene expression patterns. CTNNB1-mutated HCC constitute a full-fledged subclass of HCC with distinct pathological features such as well-differentiated cells with low proliferation rate, association to cholestasis, metabolic alterations, immune exclusion and invasion. Finally, we discuss therapeutic approaches to target ß-catenin-mutated liver tumours and innovative perspectives for future drug developments.
Collapse
Affiliation(s)
| | - Lydia Dif
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | - Justine Vaché
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | - Sara Basbous
- University Bordeaux, INSERM, BRIC, U1312, Bordeaux, France
| | | | | |
Collapse
|
90
|
Song Y, Chen H, Li J, Zhong F, Liu Y, Liu H, Wan S. Lower Serum Albumin Level: A Prospective Risk Predictor of Hepatocellular Carcinoma in Patients With Chronic Hepatitis B Virus Infection. J Viral Hepat 2024; 31:857-865. [PMID: 39283028 DOI: 10.1111/jvh.14008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 08/26/2024] [Accepted: 09/01/2024] [Indexed: 11/21/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumours in China, at high annual incidence and mortality. Chronic hepatitis B virus infection (CHB) is considered as a leading cause to bring about HCC in China. Serum albumin (ALB) level has been adopted to verify its risk with HCC development as a combination variable with other factors. However, the predictive value of a single ALB level on HBV-related HCC risk remained unclear. The aim of this study was to evaluate the prediction ability of serum ALB concentration on the risk of HBV-related HCC development. A prospectively enrolled clinical cohort compromising 2932 cases of CHB patients with at least 1-year exclusion window was selected to explore the predictive role of serum ALB level on incident HCC risk. Baseline clinical data including host characters and laboratory test were collected at the initial period of hospitalisation. The hazard ratio of ALB level associated with HCC development was assessed by Cox proportional hazards regression model using univariate and multivariate analyses. We evaluated the discrimination accuracy of ALB level in predicting HCC development by receiver operating characteristic (ROC) curves. Dose-dependent and time-dependent effects of ALB level on HCC risk prediction were demonstrated, respectively, using a restricted cubic spline and a Fine and Grey competing risk model. Referred to patients with higher ALB level, those with lower ALB level exhibited significantly increased risk of HCC development after adjustment for host variables (dichotomised analyses: hazard ratio = 3.12, 95% confidence interval 1.63-5.97, p = 8.23 × 10-4, plog-rank = 5.97 × 10-4; tertile analyses: hazard ratio = 2.07, 95% confidence interval 1.63-2.64, p = 3.77 × 10-9, plog-rank < 2.00 × 10-16; quartile analyses: hazard ratio = 2.10, 95% confidence interval 1.56-2.84, p = 9.87 × 10-7, plog-rank < 2.00 × 10-16). There was a statistically increasing trend on HCC risk which was found following by the decrease of ALB level (ptrend < 0.0001). Similar findings were present by the Kaplan-Meier analysis, cumulative incidences of HCC development were significantly higher in patients with lower ALB levels, with the p value obtained from log-rank test were all < 0.0001. The result of dose-dependent effect showed hazard ratio (HR) value of HCC risk was gradually decreasing as the increasing of ALB level, with non-linear correlation being statistically significant (Wald χ2 = 20.59, p = 0.000). HR value in lower ALB level remained persistently prominent by fluctuating around 2.73 in the whole follow-up time by adjusting for host variables. Sub-cohort analysis by ROC revealed that the discrimination ability of the ALB model was performed better than Child-Pugh (C-P) model in both cohort of patients with 1-year (area under curve [AUC] 0.762 vs. 0.720) and 2-year exclusion window (AUC 0.768 vs. 0.728). The AUC added by ALB level was demonstrated significantly from host model to full model. Lower ALB level was significantly associated with an increased risk of HBV-related HCC and could provide extra useful clinical utility to other host features, which might be a promising non-invasive indicator for surveillance on HCC development.
Collapse
Affiliation(s)
- Yusheng Song
- Department of Interventional Radiology, The People's Hospital of Ganzhou City, Ganzhou, China
| | - Haiyan Chen
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jinlong Li
- Institute of Hepatology, Department of Hepatology, The Affiliated Fifth People's Hospital of Ganzhou, Gannan Medical University, Ganzhou, China
| | - Feifei Zhong
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yunbing Liu
- Department of Medical Oncology, Ganxian District People's Hospital, Ganzhou, China
| | - Hui Liu
- Gannan Healthcare Vocational College, Ganzhou, China
| | - Shaogui Wan
- Center for Molecular Pathology, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| |
Collapse
|
91
|
Pathak S, Singh V, Kumar G N, Jayandharan GR. AAV-mediated combination gene therapy of inducible Caspase 9 and miR-199a-5p is therapeutic in hepatocellular carcinoma. Cancer Gene Ther 2024; 31:1796-1803. [PMID: 39385010 DOI: 10.1038/s41417-024-00844-x] [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: 06/13/2024] [Revised: 09/27/2024] [Accepted: 10/01/2024] [Indexed: 10/11/2024]
Abstract
Advanced-stage hepatocellular carcinoma (HCC) remains an untreatable disease with an overall survival of less than one year. One of the critical molecular mediators contributing to increased resistance to therapy and relapse, is increased hypoxia-inducible factor 1α (HIF-1α) levels, leading to metastasis of tumor cells. Several microRNAs are known to be dysregulated and impact HIF-1α expression in HCC. An in silico analysis demonstrated that hsa-miR-199a-5p is downregulated at various stages of HCC and is known to repress HIF-1α expression. Based on this analysis, we developed a combinatorial suicide gene therapy by employing hepatotropic Adeno-associated virus-based vectors encoding an inducible caspase 9 (iCasp9) and miR-199a. The overexpression of miR-199a-5p alone significantly decreased ( ~ 2-fold vs. Mock treated cells, p < 0.05) HIF-1α mRNA levels, with a concomitant increase in cancer cell cytotoxicity in Huh7 cells in vitro and in xenograft models in vivo. To further enhance the efficacy of gene therapy, we evaluated the synergistic therapeutic effect of AAV8-miR-199a and AAV6-iCasp9 in a xenograft model of HCC. Our data revealed that mice receiving combination suicide gene therapy exhibited reduced expression of HIF-1α ( ~ 4-fold vs. Mock, p < 0.001), with a significant reduction in tumor growth when compared to mock-treated animals. These findings underscore the therapeutic potential of downregulating HIF-1α during suicide gene therapy for HCC.
Collapse
Affiliation(s)
- Subhajit Pathak
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Vijayata Singh
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Narendra Kumar G
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India
| | - Giridhara R Jayandharan
- Laurus Center for Gene Therapy, Department of Biological Sciences and Bioengineering and Mehta Family Center for Engineering in Medicine and Gangwal School of Medical Sciences and Technology, Indian Institute of Technology Kanpur, Uttar Pradesh, Kanpur, 208016, India.
| |
Collapse
|
92
|
Tu J, Wang B, Wang X, Huo K, Hu W, Zhang R, Li J, Zhu S, Liang Q, Han S. Current status and new directions for hepatocellular carcinoma diagnosis. LIVER RESEARCH 2024; 8:218-236. [PMID: 39958920 PMCID: PMC11771281 DOI: 10.1016/j.livres.2024.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/17/2024] [Accepted: 12/01/2024] [Indexed: 02/18/2025]
Abstract
Liver cancer ranks as the sixth most common cancer globally, with hepatocellular carcinoma (HCC) accounting for approximately 75%-85% of cases. Most patients present with moderately advanced disease, while those with advanced HCC face limited and ineffective treatment options. Despite diagnostic efforts, no ideal tumor marker exists to date, highlighting the urgent clinical need for improved early detection of HCC. A key research objective is the development of assays that target specific pathways involved in HCC progression. This review explores the pathological origin and development of HCC, providing insights into the mechanistic rationale, clinical statistics, and the advantages and limitations of commonly used diagnostic tumor markers. Additionally, it discusses the potential of emerging biomarkers for early diagnosis and offers a brief overview of relevant assay methodologies. This review aims to summarize existing markers and investigate new ones, providing a basis for subsequent research.
Collapse
Affiliation(s)
- Jinqi Tu
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Bo Wang
- Animal Experimental Center, Xinjiang Medical University, Urumqi, Xinjiang, China
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, China
| | - Kugeng Huo
- Cyagen Biosciences (Guangzhou) Inc., Guangzhou, Guangdong, China
| | - Wanting Hu
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Rongli Zhang
- Department of Medicine, Institute for Transformative Molecular Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
- Cardiovascular Research Institute, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Jinyao Li
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| | - Shijie Zhu
- Department of Oncology, Wangjing Hospital, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qionglin Liang
- MOE Key Laboratory of Bioorganic Phosphorus Chemistry & Chemical Biology, Beijing Key Lab of Microanalytical Methods & Instrumentation, Center for Synthetic and Systems Biology, Department of Chemistry, Tsinghua University, Beijing, China
| | - Shuxin Han
- Xinjiang Key Laboratory of Biological Resources and Genetic Engineering, College of Life Science and Technology, Xinjiang University, Urumqi, Xinjiang, China
| |
Collapse
|
93
|
Ainiwaer A, Cheng J, Lang R, Peng T, Bi X, Lu Y, Chinese Research Hospital Association Society for Molecular Diagnosis, Translational Medicine Branch, China Association of Gerontology and Geriatrics. Chinese expert consensus on the clinical application of molecular diagnostics in hepatobiliary cancers (2024 edition). LIVER RESEARCH (BEIJING, CHINA) 2024; 8:195-206. [PMID: 39958921 PMCID: PMC11771259 DOI: 10.1016/j.livres.2024.11.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 11/23/2024] [Accepted: 11/26/2024] [Indexed: 01/04/2025]
Abstract
Hepatocellular carcinoma (HCC) and biliary tract cancer (BTC) are significant health challenges in China because of their high prevalence and mortality rates. Advances in molecular diagnostics have opened new avenues for personalized treatment strategies. This consensus provides a comprehensive update on the clinical applications of molecular diagnostics in HCC and BTC and addresses the urgent need for personalized treatment strategies tailored to the Chinese population, emphasizing the importance of molecular markers in guiding targeted therapies and immunotherapies. By employing the Delphi method and the Grading of Recommendations Assessment, Development, and Evaluation system, the expert panel formulated evidence-based recommendations for the use of molecular diagnostics in the clinical management of HCC and BTC. Key molecular markers, such as isocitrate dehydrogenase (IDH) 1 and 2, fibroblast growth factor receptor 2 (FGFR2), BRAF V600E, human epidermal growth factor receptor 2 (HER2), rearranged during transfection (RET), and neurotrophic tyrosine receptor kinase (NTRK), are highlighted for their roles in optimizing treatment regimens. The consensus also explores the significance of emerging biomarkers, such as tumor mutation burden and microsatellite instability, in predicting responses to immune checkpoint inhibitors. The recommendations aim to enhance precision medicine approaches, improve patient outcomes, and foster the integration of molecular diagnostics into routine clinical practice.
Collapse
Affiliation(s)
- Aizier Ainiwaer
- Comprehensive Liver Cancer Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Jiamin Cheng
- Comprehensive Liver Cancer Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Ren Lang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
| | - Tao Peng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
| | - Xinyu Bi
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yinying Lu
- Comprehensive Liver Cancer Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
| | - Chinese Research Hospital Association Society for Molecular Diagnosis
- Comprehensive Liver Cancer Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Translational Medicine Branch, China Association of Gerontology and Geriatrics
- Comprehensive Liver Cancer Center, The Fifth Medical Center of PLA General Hospital, Beijing, China
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi, China
- Department of Hepatobiliary Surgery, National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| |
Collapse
|
94
|
Gao YF, Liu YQ, Zhang H, Zhang MY. Proteo-genomic characterization of cirrhosis-associated liver cancers reveals potential subtypes and therapeutic targets. Clin Transl Oncol 2024; 26:3085-3099. [PMID: 38806996 DOI: 10.1007/s12094-024-03517-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Accepted: 05/06/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND This study aimed to identify potential subtypes of hepatocellular carcinoma (HCC) associated with cirrhosis and to investigate key markers using bioinformatic analysis of gene expression datasets-0. METHODS Three data sets (GSE17548, GSE56140, and GSE87630) were extracted from the Gene Expression Omnibus (GEO) database and normalized using the Limma package in R. Principal component analysis (PCA) and cluster analysis was performed to examine data distribution and identify subtypes. Differential gene expression analysis was performed using the Limma software package. Protein-protein interaction analysis and functional annotation were performed using the STRING database and Cytoscape software. Important signaling pathways and processes were identified using Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) Pathway Analysis. RESULTS The analysis revealed different subtypes of HCC associated with cirrhosis and identified several key genes, including CCNB2, MCM4, and CDC20, with strong binding power and prognostic value. Functional annotation indicated involvement in cell cycle regulation and metabolic pathways. ROC analysis showed high sensitivity and specificity of these genes in predicting HCC prognosis. CONCLUSION These results suggest that CCNB2, MCM4, and CDC20 may serve as potential biomarkers for predicting HCC prognosis in patients with cirrhosis and provide insights into the molecular mechanisms of HCC progression.
Collapse
Affiliation(s)
- Yi-Fan Gao
- Department of Research and Discipline Construction, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Weiwu Road No. 7, Zhengzhou City, 450003, Henan, China.
| | - Yang-Qing Liu
- Department of Research and Discipline Construction, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Weiwu Road No. 7, Zhengzhou City, 450003, Henan, China
| | - Hui Zhang
- Department of Research and Discipline Construction, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Weiwu Road No. 7, Zhengzhou City, 450003, Henan, China
| | - Meng-Yi Zhang
- Department of Oncology, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Zhengzhou, 450003, Henan, China
| |
Collapse
|
95
|
Das D, Wang X, Chiu YC, Bouamar H, Sharkey FE, Lopera JE, Lai Z, Weintraub ST, Han X, Zou Y, Chen HIH, Zeballos Torrez CR, Gu X, Cserhati M, Michalek JE, Halff GA, Chen Y, Zheng S, Cigarroa FG, Sun LZ. Integrative multi-omics characterization of hepatocellular carcinoma in Hispanic patients. J Natl Cancer Inst 2024; 116:1961-1978. [PMID: 39189979 PMCID: PMC11630563 DOI: 10.1093/jnci/djae207] [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: 05/01/2024] [Revised: 07/23/2024] [Accepted: 08/20/2024] [Indexed: 08/28/2024] Open
Abstract
BACKGROUND The incidence and mortality rates of hepatocellular carcinoma among Hispanic individuals in the United States are much higher than in non-Hispanic White people. We conducted multi-omics analyses to elucidate molecular alterations in hepatocellular carcinoma among Hispanic patients. METHODS Paired tumor and adjacent nontumor samples were collected from 31 Hispanic hepatocellular carcinomas in South Texas for genomic, transcriptomic, proteomic, and metabolomic profiling. Serum lipids were profiled in 40 Hispanic and non-Hispanic patients with or without clinically diagnosed hepatocellular carcinoma. RESULTS Exome sequencing revealed high mutation frequencies of AXIN2 and CTNNB1 in South Texas Hispanic hepatocellular carcinoma patients, suggesting a predominant activation of the Wnt/β-catenin pathway. TERT promoter mutations were also statistically significantly more frequent in the Hispanic cohort (Fisher exact test, P < .05). Cell cycles and liver function were positively and negatively enriched, respectively, with gene set enrichment analysis. Gene sets representing specific liver metabolic pathways were associated with dysregulation of corresponding metabolites. Negative enrichment of liver adipogenesis and lipid metabolism corroborated with a significant reduction in most lipids in serum samples of hepatocellular carcinoma patients (paired t test, P < .0001). Two hepatocellular carcinoma subtypes from our Hispanic cohort were identified and validated with the Cancer Genome Atlas liver cancer cohort. Patients with better overall survival showed higher activity of immune and angiogenesis signatures and lower activity of liver function-related gene signatures. They also had higher levels of immune checkpoint and immune exhaustion markers. CONCLUSIONS Our study revealed specific molecular features of Hispanic hepatocellular carcinoma and potential biomarkers for therapeutic management. It provides a unique resource for studying Hispanic hepatocellular carcinoma.
Collapse
Affiliation(s)
- Debodipta Das
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiaojing Wang
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yu-Chiao Chiu
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hakim Bouamar
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Francis E Sharkey
- Department of Pathology and Laboratory Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Jorge E Lopera
- Department of Radiology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Zhao Lai
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Molecular Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Susan T Weintraub
- Department of Biochemistry & Structural Biology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xianlin Han
- Sam and Ann Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yi Zou
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Hung-I H Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Carla R Zeballos Torrez
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Xiang Gu
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Matyas Cserhati
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Joel E Michalek
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Glenn A Halff
- Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Yidong Chen
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Siyuan Zheng
- Greehey Children’s Cancer Research Institute, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
- Department of Population Health Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Francisco G Cigarroa
- Transplant Center, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Lu-Zhe Sun
- Department of Cell Systems and Anatomy, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| |
Collapse
|
96
|
Alvarado-Tapias E, Maya-Miles D, Albillos A, Aller R, Ampuero J, Andrade RJ, Arechederra M, Aspichueta P, Banales JM, Blas-García A, Caparros E, Cardoso Delgado T, Carrillo-Vico A, Claria J, Cubero FJ, Díaz-Ruiz A, Fernández-Barrena MG, Fernández-Iglesias A, Fernández-Veledo S, Francés R, Gallego-Durán R, Gracia-Sancho J, Irimia M, Lens S, Martínez-Chantar ML, Mínguez B, Muñoz-Hernández R, Nogueiras R, Ramos-Molina B, Riveiro-Barciela M, Rodríguez-Perálvarez ML, Romero-Gómez M, Sabio G, Sancho-Bru P, Ventura-Cots M, Vidal S, Gahete MD. Proceedings of the 5th Meeting of Translational Hepatology, organized by the Spanish Association for the Study of the Liver (AEEH). GASTROENTEROLOGIA Y HEPATOLOGIA 2024; 47:502207. [PMID: 38723772 DOI: 10.1016/j.gastrohep.2024.502207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 05/02/2024] [Indexed: 11/30/2024]
Abstract
This is the summary report of the 5th Translational Hepatology Meeting, endorsed by the Spanish Association for the Study of the Liver (AEEH) and held in Seville, Spain, in October 2023. The meeting aimed to provide an update on the latest advances in the field of basic and translational hepatology, covering different molecular, cellular, and pathophysiological aspects of the most relevant clinical challenges in liver pathologies. This includes the identification of novel biomarkers and diagnostic tools, the understanding of the relevance of immune response and inflammation in liver diseases, the characterization of current medical approaches to reverse liver diseases, the incorporation of novel molecular insights through omics techniques, or the characterization of the impact of toxic and metabolic insults, as well as other organ crosstalk, in liver pathophysiology.
Collapse
Affiliation(s)
- Edilmar Alvarado-Tapias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Gastroenterology, Hospital Santa Creu I Sant Pau, Institut de Recerca Sant Pau, Universidad Autónoma de Barcelona, Barcelona, Spain.
| | - Douglas Maya-Miles
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain.
| | - Agustin Albillos
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Servicio de Gastroenterología y Hepatología, Hospital Universitario Ramón y Cajal/Universidad de Alcalá/Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Rocio Aller
- BioCritic, Group for Biomedical Research in Critical Care Medicine, Spain; Department of Medicine, Dermatology and Toxicology, Universidad de Valladolid, Spain; Gastroenterology Unit, Hospital Clínico Universitario de Valladolid, 47003 Valladolid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Infecciosas (CIBERINFEC), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Javier Ampuero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Raul J Andrade
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigación Biomédica de Málaga-IBIMA, Hospital Universitario Virgen de la Victoria, Universidad de Málaga, Málaga, Spain
| | - Maria Arechederra
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Pamplona, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain
| | - Patricia Aspichueta
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Physiology, Faculty of Medicine and Nursing, University of the Basque Country UPV/EHU, Leioa, Spain; Biobizkaia Health Research Institute, Barakaldo, Spain
| | - Jesus M Banales
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute - Donostia University Hospital - University of the Basque Country (UPV/EHU), Ikerbasque, Donostia-San Sebastian, Spain; Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Ana Blas-García
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Departamento de Fisiología, Universitat de València, Av. Blasco Ibáñez, 15, 46010 Valencia, Spain; FISABIO (Fundación para el Fomento de la Investigación Sanitaria y Biomédica de la Comunidad Valenciana), Av. de Catalunya, 21, 46020 Valencia, Spain
| | - Esther Caparros
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Teresa Cardoso Delgado
- Biobizkaia Health Research Institute, Barakaldo, Spain; IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
| | - Antonio Carrillo-Vico
- Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de Bioquímica Médica y Biología Molecular e Inmunología, Facultad de Medicina, Universidad de Sevilla, Seville, Spain
| | - Joan Claria
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Biochemistry and Molecular Genetics Service, Hospital Clínic, IDIBAPS, Barcelona, Spain; University of Barcelona, Spain
| | - Francisco Javier Cubero
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Health Research Institute Gregorio Marañón (IiSGM), Madrid, Spain
| | - Alberto Díaz-Ruiz
- Laboratory of Cellular and Molecular Gerontology, Precision Nutrition and Aging, Madrid Institute for Advanced Studies - IMDEA Food, CEI UAM+CSIC, Madrid, Spain; CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain
| | - Maite G Fernández-Barrena
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Investigaciones Sanitarias de Navarra IdiSNA, Pamplona, Spain; Hepatology Laboratory, Solid Tumors Program, CIMA, CCUN, University of Navarra, Spain
| | - Anabel Fernández-Iglesias
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Sonia Fernández-Veledo
- Department of Endocrinology and Nutrition and Research Unit, University Hospital of Tarragona Joan XXIII, Institut d'Investigació Sanitària Pere Virgili (IISPV), Universitat Rovira i Virgili (URV), Tarragona, Spain; CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), Instituto de Salud Carlos III, Madrid, Spain
| | - Ruben Francés
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Grupo de Inmunobiología Hepática e Intestinal, Departamento Medicina Clínica, Universidad Miguel Hernández, San Juan, Spain; Instituto de Investigación Sanitaria ISABIAL, Hospital General Universitario de Alicante, Alicante, Spain
| | - Rocío Gallego-Durán
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Jordi Gracia-Sancho
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Vascular Biology Research Group, IDIBAPS, Hospital Clínic de Barcelona, 08036 Barcelona, Spain
| | - Manuel Irimia
- Universitat Pompeu Fabra (UPF), Centre for Genomic Regulation (CRG), The Barcelona Institute of Science and Technology, ICREA, Barcelona, Spain
| | - Sabela Lens
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Immunology, Ophthalmology and ENT, Complutense University School of Medicine, Madrid, Spain; Liver Unit, Hospital Clínic, IDIBAPS, Barcelona, Spain
| | - María Luz Martínez-Chantar
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Disease Lab, Center for Cooperative Research in Biosciences (CIC bioGUNE), Basque Research and Technology Alliance (BRTA), Derio, Bizkaia, Spain
| | - Beatriz Mínguez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Rocío Muñoz-Hernández
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain; Departamento de fisiología, Facultad de Biología, Universidad de Sevilla, Sevilla, Spain
| | - Rubén Nogueiras
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Physiology, CIMUS, University of Santiago de Compostela, Instituto de Investigación Sanitaria, Santiago de Compostela, Spain; Galician Agency of Innovation (GAIN), Xunta de Galicia, Santiago de Compostela, Spain
| | - Bruno Ramos-Molina
- Obesity, Diabetes and Metabolism Laboratory, Biomedical Research Institute of Murcia (IMIB), Murcia, Spain
| | - Mar Riveiro-Barciela
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain
| | - Manuel L Rodríguez-Perálvarez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Department of Hepatology and Liver Transplantation, Reina Sofia University Hospital, Cordoba, Spain; Maimonides Biomedical Research Institute of Córdoba (IMIBIC), University of Córdoba, Cordoba, Spain
| | - Manuel Romero-Gómez
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Instituto de Biomedicina de Sevilla (IBIS), Hospital Universitario Virgen del Rocío (HUVR), CISC, Universidad de Sevilla, Sevilla, Spain
| | - Guadalupe Sabio
- Centro Nacional de Investigaciones Cardiovasculares (CNIC), Stress Kinases in Diabetes, Cancer and Biochemistry, Spain; Centro Nacional de Investigaciones Oncologicas (CNIO), Organ Crosstalk in Metabolic Diseases, Madrid, Spain
| | - Pau Sancho-Bru
- CIBEREHD (Center for Biomedical Network Research in Liver and Digestive Diseases), Instituto de Salud Carlos III, 28029 Madrid, Spain; Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Meritxell Ventura-Cots
- Liver Unit, Hospital Universitari Vall d'Hebron, Vall d'Hebron Institute of Research (VHIR), Universitat Autònoma de Barcelona, Vall d'Hebron Barcelona Hospital Campus, Barcelona, Spain; Center for Liver Diseases, Pittsburgh Liver Research Center, Division of Gastroenterology, Hepatology and Nutrition, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Silvia Vidal
- Group of Inflammatory Diseases, Institut de Recerca de l'Hospital de la Santa Creu i Sant Pau, Biomedical Research Institute Sant Pau (IIB Sant Pau), Barcelona, Spain
| | - Manuel D Gahete
- CIBER Fisiopatología de la Obesidad y Nutrición (CIBERobn), Madrid, Spain; Department of Cell Biology, Physiology and Immunology, University of Córdoba, Spain; Molecular Hepatology Group, Maimonides Biomedical Research Institute of Córdoba (IMIBIC), Spain; Reina Sofia University Hospital, Cordoba, Spain.
| |
Collapse
|
97
|
Backman S, Botling J, Nord H, Ghosal S, Stålberg P, Juhlin CC, Almlöf J, Sundin A, Zhang L, Moens L, Eriksson B, Welin S, Hellman P, Skogseid B, Pacak K, Mollazadegan K, Åkerström T, Crona J. The evolutionary history of metastatic pancreatic neuroendocrine tumours reveals a therapy driven route to high-grade transformation. J Pathol 2024; 264:357-370. [PMID: 39360347 DOI: 10.1002/path.6348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2024] [Revised: 08/08/2024] [Accepted: 08/09/2024] [Indexed: 10/04/2024]
Abstract
Tumour evolution with acquisition of more aggressive disease characteristics is a hallmark of disseminated cancer. Metastatic pancreatic neuroendocrine tumours (PanNETs) in particular may progress from a low/intermediate to a high-grade disease. The aim of this work was to understand the molecular mechanisms underlying metastatic progression as well as PanNET transformation from a low/intermediate to a high-grade disease. We performed multi-omics analysis (genome/exome sequencing, total RNA-sequencing and methylation array) of 32 longitudinal samples from six patients with metastatic low/intermediate grade PanNET. The clonal composition of tumour lesions and underlying phylogeny of each patient were determined with bioinformatics analyses. Findings were validated in post-alkylating chemotherapy samples from 24 patients with PanNET using targeted next generation sequencing. We validate the current PanNET evolutionary model with MEN1 inactivation that occurs very early in tumourigenesis. This was followed by pronounced genetic diversity on both spatial and temporal levels, with parallel and convergent tumour evolution involving the ATRX/DAXX and mechanistic target of the rapamycin (mTOR) pathways. Following alkylating chemotherapy treatment, some PanNETs developed mismatch repair deficiency and acquired a hypermutational phenotype. This was validated among 16 patients with PanNET who had high-grade progression after alkylating chemotherapy, of whom eight had a tumour mutational burden >50 (50%). In comparison, among the eight patients who did not show high-grade progression, 0 had a tumour mutational burden >50 (0%; odds ratio 'infinite', 95% confidence interval 1.8 to 'infinite', p = 0.02). Our findings contribute to broaden the understanding of metastatic/high-grade PanNETs and suggests that therapy driven disease evolution is an important hallmark of this disease. © 2024 The Author(s). The Journal of Pathology published by John Wiley & Sons Ltd on behalf of The Pathological Society of Great Britain and Ireland. This article has been contributed to by U.S. Government employees and their work is in the public domain in the USA.
Collapse
Affiliation(s)
- Samuel Backman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Johan Botling
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Helena Nord
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Suman Ghosal
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Peter Stålberg
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - C Christofer Juhlin
- Department of Oncology - Pathology, Karolinska Institutet, Stockholm, Sweden
| | - Jonas Almlöf
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Anders Sundin
- Section of Radiology, Molecular Imaging, Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Liang Zhang
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Lotte Moens
- Department of Immunology, Genetics and Pathology and Science for Life Laboratory, Uppsala University, Uppsala, Sweden
| | - Barbro Eriksson
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Staffan Welin
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Per Hellman
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Britt Skogseid
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| | - Karel Pacak
- Section on Medical Neuroendocrinology, Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, MD, USA
| | | | - Tobias Åkerström
- Department of Surgical Sciences, Uppsala University, Uppsala, Sweden
| | - Joakim Crona
- Department of Medical Sciences, Uppsala University, Uppsala, Sweden
| |
Collapse
|
98
|
Chen S, Tang Q, Hu M, Song S, Wu X, Zhou Y, Yang Z, Liao S, Zhou L, Wang Q, Liu H, Yang M, Chen Z, Zhao W, He S, Zhou Z. Loss of Carbamoyl Phosphate Synthetase 1 Potentiates Hepatocellular Carcinoma Metastasis by Reducing Aspartate Level. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402703. [PMID: 39387452 PMCID: PMC11615744 DOI: 10.1002/advs.202402703] [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: 03/14/2024] [Revised: 09/01/2024] [Indexed: 10/15/2024]
Abstract
Hepatocellular carcinoma (HCC) is one of the most lethal cancers worldwide. Numerous studies have shown that metabolic reprogramming is crucial for the development of HCC. Carbamoyl phosphate synthase 1 (CPS1), a rate-limiting enzyme in urea cycle, is an abundant protein in normal hepatocytes, however, lacking systemic research in HCC. It is found that CPS1 is low-expressed in HCC tissues and circulating tumor cells, negatively correlated with HCC stage and prognosis. Further study reveals that CPS1 is a double-edged sword. On the one hand, it inhibits the activity of phosphatidylcholine-specific phospholipase C to block the biosynthesis of diacylglycerol (DAG), leading to the downregulation of the DAG/protein kinase C pathway to inhibit invasion and metastasis of cancer cells. On the other hand, CPS1 promotes cell proliferation by increasing intracellular S-adenosylmethionin to enhance the m6A modification of solute carrier family 1 member 3 mRNA, a key transporter for aspartate intake. Finally, CPS1 overexpressing adeno-associated virus can dampen HCC progression. Collectively, this results uncovered that CPS1 is a switch between HCC proliferation and metastasis by increasing intracellular aspartate level.
Collapse
Affiliation(s)
- Siyuan Chen
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Qin Tang
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Manqiu Hu
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Sijie Song
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Xiaohong Wu
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - You Zhou
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zihan Yang
- Department of Biomedical Sciencesand Tung Biomedical Sciences CenterCity University of Hong Kong83 Tat Chee AvenueKowloonHong Kong SAR999077P. R. China
| | - Siqi Liao
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Li Zhou
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Qingliang Wang
- Department of PathologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Hongtao Liu
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Mengsu Yang
- Department of Biomedical Sciencesand Tung Biomedical Sciences CenterCity University of Hong Kong83 Tat Chee AvenueKowloonHong Kong SAR999077P. R. China
| | - Zhe‐Sheng Chen
- Department of Pharmaceutical SciencesInstitute for BiotechnologyCollege of Pharmacy and Health SciencesSt. John's UniversityQueensNY11439USA
| | - Wei Zhao
- School of Clinical MedicineThe First Affiliated HospitalChengdu Medical CollegeSichuan610500P. R. China
- Department of Clinical BiochemistrySchool of Laboratory MedicineChengdu Medical CollegeSichuan610500P. R. China
| | - Song He
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| | - Zhihang Zhou
- Department of GastroenterologyThe Second Affiliated Hospital of Chongqing Medical UniversityChongqing400010China
| |
Collapse
|
99
|
Meng L, Jiang Z, Shen G, Lin S, Gao F, Guo X, Lv B, Hu S, Ni Z, Chen S, Ji Y. Genetic alterations are related to clinicopathological features and risk of recurrence/metastasis of hepatocellular carcinoma. Eur J Cancer Prev 2024:00008469-990000000-00191. [PMID: 39642087 DOI: 10.1097/cej.0000000000000939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/08/2024]
Abstract
Lack of efficient biomarkers and clinical translation of molecular typing impedes the implementation of targeted therapy for hepatocellular carcinoma (HCC). High-throughput sequencing techniques represented by next-generation sequencing (NGS) are tools for detecting targetable genes. The objective of this study is to explore the genetic alterations associated with clinicopathological features and the risk of recurrence/metastasis in HCC. NGS analysis was conducted on formalin-fixed paraffin-embedded tissues from 164 resected liver samples obtained from Chinese patients. Morphologic subtypes were reviewed based on hematoxylin-eosin and immunohistochemistry staining, Correlation to the acquired molecular features were analyzed with clinicopathological information. We also retrieved follow-up information of the 123 transplanted cases from 2017 to 2019 to screen recurrence/metastasis-associated factors by univariate analysis. Generally, the most frequently mutated genes include TP53 and CTNNB1 which showed a trend of mutually exclusive mutation. Copy-number variant with the highest frequency was detected in TAF1 and CCND1 in 11q13.3 loci. Correlation analysis showed that various genetic alterations were associated with morphologic subtypes and other pathologic features. While gene signatures of proliferation/nonproliferation class were correlated with differentiation, satellite foci and other invasive morphological features. Macrotrabecular-massive subtype, TSC2 (tuberous sclerosis complex 2) mutation, Ki-67 expression, and other six factors were found to be associated with recurrence/metastasis after liver transplantation. Genetic alterations detected by NGS show correlation with not only pathological and clinical features, but also with recurrence/metastasis after liver transplantation. Further gene-level molecular typing will be practical for targeted therapy and individual recurrence risk assessment in HCC patients.
Collapse
Affiliation(s)
- Lili Meng
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Zhenjian Jiang
- Department of Pathology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Guangyue Shen
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Shulan Lin
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
- Department of Pathology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Feng Gao
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Xinxin Guo
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Bin Lv
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Shuying Hu
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Zheng Ni
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| | - Shanghua Chen
- Department of Pathology, Zhongshan Hospital, Fudan University (Xiamen Branch), Xiamen, China
| | - Yuan Ji
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai
| |
Collapse
|
100
|
Bakiri L, Wagner EF. c-Jun and Fra-2 pair up to Myc-anistically drive HCC. Cell Cycle 2024:1-9. [PMID: 39581891 DOI: 10.1080/15384101.2024.2429968] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 09/06/2024] [Accepted: 09/11/2024] [Indexed: 11/26/2024] Open
Abstract
Hepatocellular carcinoma (HCC), a leading cause of cancer-related death with limited therapies, is a complex disease developing in a background of Hepatitis Virus infection or systemic conditions, such as the metabolic syndrome. Investigating HCC pathogenesis in model organisms is therefore crucial for developing novel diagnostic and therapeutic tools. Genetically engineered mouse models (GEMMs) have been instrumental in recapitulating the local and systemic features of HCC. Early studies using GEMMs and patient material implicated members of the dimeric Activator Protein-1 (AP-1) transcription factor family, such as c-Jun and c-Fos, in HCC formation. In a recent report, we described how switchable, hepatocyte-restricted expression of a single-chain c-Jun~Fra-2 protein, functionally mimicking the c-Jun/Fra-2 AP-1 dimer, results in spontaneous and largely reversible liver tumors in GEMMs. Dysregulated cell cycle, inflammation, and dyslipidemia are observed at early stages and tumors display molecular HCC signatures. We demonstrate that increased c-Myc expression is an essential molecular determinant of tumor formation that can be therapeutically targeted using the BET inhibitor JQ1. Here, we discuss these findings with additional results illustrating how AP-1 GEMMs can foster preclinical research on liver diseases with novel perspectives offered by the constantly increasing wealth of HCC-related datasets.
Collapse
Affiliation(s)
- Latifa Bakiri
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna (MUW), Vienna, Austria
| | - Erwin F Wagner
- Laboratory Genes and Disease, Department of Laboratory Medicine, Medical University of Vienna (MUW), Vienna, Austria
- Laboratory Genes and Disease, Department of Dermatology, Medical University of Vienna (MUW), Vienna, Austria
| |
Collapse
|