1
|
Yan Q, Sun X, Wang Y, Duan S, Wang B. The effect of continuous infusion chemotherapy through femoral artery catheterization on GP73, AFP-L3, and safety efficacy in liver cancer patients. Clin Exp Med 2025; 25:148. [PMID: 40347395 PMCID: PMC12065681 DOI: 10.1007/s10238-025-01560-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Accepted: 01/06/2025] [Indexed: 05/12/2025]
Abstract
This study examines the impact of continuous infusion chemotherapy via femoral artery catheterization on Golgi protein 73 (GP73) and alpha fetoprotein heterogeneity (AFP-L3) in liver cancer patients. A retrospective analysis was conducted on 108 liver cancer patients treated from January 2020 to December 2022, divided into two groups: transarterial chemoembolization (TACE) and continuous infusion regional arterial chemotherapy via femoral artery catheterization (CIFAC), with 54 patients in each group. Serum tumor markers, liver function, adverse reactions, quality of life, and 1-year survival rate were analyzed and compared between the two groups of patients. Prior to treatment, no significant differences were observed in tumor markers, liver function, and quality of life between groups (P > 0.05). After 60 and 90 days, the CIFAC group exhibited significantly lower levels of GP73, AFP, and AFP-L3 compared to TACE (P < 0.05). Additionally, CIFAC patients had lower levels of alanine aminotransferase (ALT), aspartate transaminase (AST), indocyanine green (ICG15) (P < 0.05), reduced adverse reactions (nausea, vomiting, etc.), and higher Karnofsky scores (P < 0.05). The one-year survival rate of the CIFAC group was significantly higher than that of the TACE group (P < 0.05). Continuous infusion chemotherapy through femoral artery catheterization can help reduce serum tumor marker levels, improve liver function, and reduce adverse reactions in liver cancer patients.
Collapse
Affiliation(s)
- Qiong Yan
- Interventional Catheterization Room, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Xinguo Sun
- Interventional Catheterization Room, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Yubo Wang
- Production Department, Guhan Traditional Chinese Medicine Co., Ltd, Hengyang, 421000, China
| | - Shijiao Duan
- Interventional Catheterization Room, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China
| | - Bo Wang
- Physical examination center, Affiliated Nanhua Hospital, University of South China, Hengyang, 421002, China.
| |
Collapse
|
2
|
Schulze K, Rose TD, Adlung L, Peschka M, Pagani F, Gorgulho J, Fründt TW, Labgaa I, Haber PK, Zimpel C, Castven D, Weinmann A, Garzia-Lezana T, Waldmann M, Renné T, Voß H, Moritz M, Orlikowski D, Schlüter H, Baumbach J, Schwartz M, Lohse AW, Huber S, Sangro B, Macias RI, Izquierdo-Sanchez L, Banales JM, Wege H, Marquardt JU, Villanueva A, Pauling JK, von Felden J. Metabolomic liquid biopsy dynamics predict early-stage HCC and actionable candidates of human hepatocarcinogenesis. JHEP Rep 2025; 7:101340. [PMID: 40290517 PMCID: PMC12023797 DOI: 10.1016/j.jhepr.2025.101340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 01/16/2025] [Accepted: 01/22/2025] [Indexed: 04/30/2025] Open
Abstract
Background & Aims Actionable candidates of hepatocarcinogenesis remain elusive, and tools for early detection are suboptimal. Our aim was to demonstrate that serum metabolome profiles reflect the initiation of hepatocellular carcinoma (HCC) and enable the identification of biomarkers for early HCC detection and actionable candidates for chemoprevention. Methods This global cohort study included 654 patients and 801 biospecimens. Following serum metabolome profiling across the spectrum of hepatocarcinogenesis, we conducted a phase II biomarker case-control study for early HCC detection. Findings were independently validated through in silico analysis, mRNA sequencing, and proteome profiling of primary HCC and non-tumoral tissue, and in vitro experiments. Results Aspartic acid, glutamic acid, taurine, and hypoxanthine were differentially abundant in the serum across chronic liver disease, cirrhosis, initial HCC, and progressed HCC, independent of sex, age, and etiology. In a phase II biomarker case-control study, a blood-based metabolite signature yielded an AUC of 94% to discriminate between patients with early-stage HCC and controls with cirrhosis, including independent validation. Unsupervised biclustering (MoSBi), lipid network analysis (LINEX2), and pathway enrichment analysis confirmed alterations in amino acid-, lipid-, and nucleotide-related pathways. In tumor tissue, these pathways were significantly deregulated regarding gene and protein expression in two independent datasets, including actionable targets RRM2, GMPS, BCAT1, PYCR2, and NEU1. In vitro knockdown confirmed a functional role in proliferation and migration, as exemplified for PYCR2. Conclusions These findings demonstrate that serum metabolome profiling indicates deregulated metabolites and pathways during hepatocarcinogenesis. Our liquid biopsy approach accurately detects early-stage HCC outperforming currently recommended surveillance tools and facilitates identification of actionable candidates for chemoprevention. Impact and implications Deregulated cellular metabolism is a hallmark of cancer. In smaller studies, circulating metabolite profiles have been associated with HCC, although mainly in the context of fatty liver disease. Translation strategies for primary prevention or early detection are lacking. In this global study, we present an unsupervised landscape of the altered serum metabolome profile during hepatocarcinogenesis, independent of age, sex, and etiology. We provide a blood-based metabolite signature that accurately identifies early-stage HCC in a phase II biomarker study including independent validation. Further RRM2, GMPS, BCAT1, PYCR2, and NEU1 are identified in tumor tissue as actionable candidates for prevention. Our data provide the rationale for clinical trials testing liquid biopsy metabolome-based signatures for early HCC detection and the development of chemoprevention strategies.
Collapse
Affiliation(s)
- Kornelius Schulze
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Tim Daniel Rose
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Lorenz Adlung
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Hamburg Center for Translational Immunology (HCTI), and Center for Biomedical AI (bAIome), Hamburg, Germany
- Mildred Scheel Cancer Career Center HaTriCS4, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Peschka
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Newborn Screening and Metabolic Laboratory, Department of Pediatrics, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Francesca Pagani
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Joao Gorgulho
- Department of Oncology, Hematology and Bone Marrow Transplantation with Section of Pneumology, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
- University Cancer Center Hamburg–Hubertus Wald Tumorzentrum, University Medical Centre Hamburg-Eppendorf, Hamburg, Germany
| | - Thorben W. Fründt
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Ismail Labgaa
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Department of Visceral Surgery, Lausanne University Hospital (CHUV), University of Lausanne (UNIL), Lausanne, Switzerland
| | - Philipp K. Haber
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Carolin Zimpel
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Darko Castven
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
| | - Arndt Weinmann
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Teresa Garzia-Lezana
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Moritz Waldmann
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Thomas Renné
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- Irish Centre for Vascular Biology, School of Pharmacy and Biomolecular Sciences, Royal College of Surgeons in Ireland, Dublin, Ireland
- Center for Thrombosis and Hemostasis (CTH), Johannes Gutenberg University Medical Center, Mainz, Germany
| | - Hannah Voß
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Manuela Moritz
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Dorian Orlikowski
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Hartmut Schlüter
- Institute of Clinical Chemistry and Laboratory Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
| | - Jan Baumbach
- Chair of Computational Systems Biology, University of Hamburg, 22607 Hamburg, Germany
| | - Myron Schwartz
- Recanati Miller Transplant Institute, The Icahn School of Medicine at Mount Sinai Hospital, New York, NY, USA
| | - Ansgar W. Lohse
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Samuel Huber
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Bruno Sangro
- Liver Unit, Clinica Universidad de Navarra-IDISNA and CIBEREHD, Pamplona, Spain
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
| | - Rocio I.R. Macias
- Center for the Study of Liver and Gastrointestinal Diseases (CIBEREHD), Carlos III National Institute of Health, Madrid, Spain
- Experimental Hepatology and Drug Targeting (HEVEPHARM), University of Salamanca, IBSAL, Salamanca, Spain
| | - Laura Izquierdo-Sanchez
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
| | - Jesus M. Banales
- Department of Liver and Gastrointestinal Diseases, Biogipuzkoa Health Research Institute–Donostia University Hospital, University of the Basque Country (UPV/EHU), CIBEREHD, Donostia-San Sebastian, Spain
- IKERBASQUE, Basque Foundation for Science, Bilbao, Spain
- Department of Biochemistry and Genetics, School of Sciences, University of Navarra, Pamplona, Spain
| | - Henning Wege
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| | - Jens U. Marquardt
- Department of Medicine I, University Medical Center Schleswig-Holstein-Campus Lübeck, Germany
- I. Department of Medicine, University Medical Center Mainz, Germany
| | - Augusto Villanueva
- Division of Liver Diseases, Liver Cancer Program, Tisch Cancer Institute, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Division of Hematology and Medical Oncology, Department of Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Josch Konstantin Pauling
- LipiTUM, Chair of Experimental Bioinformatics, TUM School of Life Sciences, Technical University of Munich, Munich, Germany
- Institute for Clinical Chemistry and Laboratory Medicine, University Hospital and Faculty of Medicine Carl Gustav Carus, Dresden University of Technology, Dresden, Germany
| | - Johann von Felden
- I. Department of Medicine, University Medical Center Hamburg-Eppendorf, Hamburg, Germany
- ERN-RARE-LIVER, Hamburg, Germany
| |
Collapse
|
3
|
He H, Wu Y, Jia Z, Zhang Y, Pan Y, Zhang Y, Su K, Cui Y, Sun Y, Li D, Lv H, Yi J, Wang Y, Kou C, Sun X, Jiang J. A stratified precision screening strategy for enhancing hepatitis B- and C-associated liver cancer detection: a prospective study. Sci Rep 2025; 15:11396. [PMID: 40181083 PMCID: PMC11968812 DOI: 10.1038/s41598-025-95795-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2024] [Accepted: 03/24/2025] [Indexed: 04/05/2025] Open
Abstract
This study explores new screening strategies to enhance liver cancer screening effectiveness. In a prospective study, 2605 participants underwent baseline, 6-months self-reported, and 1-year follow-up screenings using abdominal ultrasonography, AFP, AFP-L3%, and DCP. The results demonstrated the GALADUS protocol exhibited superior performance with higher AUC (0.935 vs. 0.836; DeLong P < 0.001), sensitivity (91.0% vs. 70.8%; P < 0.001), detection (3.1% vs. 2.4%; P < 0.001), and early diagnosis rates (64.2% vs. 58.7%) compared to the AFP/US protocol. Notably, among individuals with an aMAP score ≥ 60, GALADUS had significantly outperformed AFP/US in AUC (0.923 vs. 0.826; DeLong P < 0.001), sensitivity (94.2% vs. 69.6%; P < 0.001), detection (9.7% vs. 7.2%; P < 0.001), and early diagnosis rates (63.1% vs. 54.2%). However, for those with an aMAP score < 60, GALADUS offered no significant advantages. Introducing the "aMAP triage" protocol, combining GALADUS for aMAP ≥ 60 and AFP/US for aMAP < 60, further enhanced AUC to 0.925 (DeLong P < 0.001), improved sensitivity by 19.1% (89.9% vs. 70.8%; P < 0.001), and increased detection (3.1% vs. 2.4%; P < 0.001) and early diagnosis rates (65.0% vs. 58.7%), being cost-effective compared to GALADUS. In conclusion, this study highlights the potential of a stratified precision screening strategy in identifying high-risk individuals, applying tailored early detection protocols to improve liver cancer screening efficacy.
Collapse
Affiliation(s)
- Hua He
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Cancer Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yanhua Wu
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Zhifang Jia
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yangyu Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuchen Pan
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Yuzheng Zhang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Kaisheng Su
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yingnan Cui
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuanlin Sun
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Dongming Li
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Haiyong Lv
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Jiaxin Yi
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China
| | - Yuehui Wang
- Department of Geriatrics, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Changgui Kou
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China
| | - Xiaofeng Sun
- Department of Cadre's Wards Ultrasound Diagnostics, Ultrasound Diagnostic Center, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China
| | - Jing Jiang
- Department of Clinical Epidemiology, the First Hospital of Jilin University, No. 1, Xinmin Street, Changchun, 130021, Jilin Province, China.
- Department of Epidemiology and Biostatistics, School of Public Health, Jilin University, Changchun, 130021, Jilin Province, China.
- Center of Infectious Diseases and Pathogen Biology, the First Hospital of Jilin University, Changchun, 130021, Jilin Province, China.
| |
Collapse
|
4
|
Sanduzzi-Zamparelli M, Cabibbo G. Surveillance in HCC: Making the Most of What We Have Today. Liver Int 2025; 45:e70057. [PMID: 40083260 DOI: 10.1111/liv.70057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/16/2025]
Affiliation(s)
- Marco Sanduzzi-Zamparelli
- Liver Oncology Unit, Liver Unit, ICMDM, Hospital Clinic Barcelona, Barcelona, Spain
- Barcelona Clinic Liver Cancer (BCLC) Group, IDIBAPS, Barcelona, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Giuseppe Cabibbo
- Section of Gastroenterology and Hepatology, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties PROMISE, University of Palermo, Piazza delle Cliniche n 2, Palermo, Italy
- Gastroenterology Unit, Azienda Ospedaliera Universitaria Policlinico "Paolo Giaccone", Palermo, Italy
| |
Collapse
|
5
|
Cheraghpour M, Hatami B, Singal AG. Lifestyle and Pharmacologic Approaches to Prevention of Metabolic Dysfunction-associated Steatotic Liver Disease-related Hepatocellular Carcinoma. Clin Gastroenterol Hepatol 2025; 23:685-694.e6. [PMID: 39800201 DOI: 10.1016/j.cgh.2024.09.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 09/19/2024] [Accepted: 09/30/2024] [Indexed: 01/15/2025]
Abstract
Hepatocellular carcinoma (HCC) is a major concern for public health. Fatty liver disease, related to alcohol misuse or metabolic syndrome, has become the leading cause of chronic liver disease and HCC. The strong association between type 2 diabetes mellitus and HCC can be partly attributed to the development of metabolic dysfunction-associated steatotic liver disease (MASLD). There is a strong interest in strategies that may mitigate HCC risk and reduce HCC incidence in this growing population of at-risk individuals. In this review, we describe the pathogenesis of HCC in patients with MASLD and discuss potential emerging pharmacological and lifestyle interventions for MASLD-related HCC. HCC risk has been observed to be lower with healthy lifestyle behaviors, such as healthy dietary patterns (eg, high consumption of vegetables, whole grains, fish and poultry, yogurt, and olive oil, and low consumption of red and processed meats and dietary sugar) and increased physical activity. Selecting an appropriate pharmacologic approach for individuals with MASLD may also decrease the occurrence of HCC. Metformin, PPAR activators, sodium-glucose cotransporter 2 inhibitors, dipeptidyl peptidase-4 inhibitors, glucagon-like peptide-1 receptor agonists, aspirin, and statins have all shown promise to reduce the risk of HCC, although guidelines do not recommend their use for the sole purpose of chemoprevention at this time, given a dearth of data defining their risk-benefit ratio.
Collapse
Affiliation(s)
- Makan Cheraghpour
- Basic and Molecular Epidemiology of Gastrointestinal Disorders Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Behzad Hatami
- Gastroenterology and Liver Diseases Research Center, Research Institute for Gastroenterology and Liver Diseases, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
6
|
Al-Hasan M, Mehta N, Yang JD, Singal AG. Role of biomarkers in the diagnosis and management of HCC. Liver Transpl 2025; 31:384-394. [PMID: 38738964 DOI: 10.1097/lvt.0000000000000398] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/06/2024] [Indexed: 05/14/2024]
Abstract
For many cancers, biomarkers have served as an important tool across the cancer care continuum from risk stratification and early detection to diagnosis and treatment. Alpha-fetoprotein (AFP) remains one of the few validated biomarkers for patients with HCC. Although AFP has shown potential for each of these steps, its performance, when used alone, has often been suboptimal. There continue to be discordant recommendations about AFP's value when combined with ultrasound for surveillance, as well as its role in diagnostic algorithms. Conversely, high AFP levels are associated with aggressive tumor biology and survival, so it remains a key factor for the selection of candidates for liver transplant. There have been immense efforts to identify and validate additional biomarkers for each of these steps in the HCC care continuum. Indeed, biomarker panels have shown promising data for HCC risk stratification and surveillance among patients with cirrhosis, as well as prognostication and detection of minimal residual disease in patients undergoing HCC treatment. Several large prospective studies are currently ongoing to evaluate the role of these emerging biomarkers in clinical practice.
Collapse
Affiliation(s)
- Mohammed Al-Hasan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Neil Mehta
- Department of Internal Medicine, University of California San Francisco, San Francisco, California, USA
| | - Ju Dong Yang
- Department of Internal Medicine, Cedars Sinai, Los Angeles, California, USA
| | - Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| |
Collapse
|
7
|
Gurley T, Hernaez R, Cerda V, Thomas T, Narasimman M, Mittal S, Al-Hasan M, Daher D, Singal AG. Cost-effectiveness of an outreach program for HCC screening in patients with cirrhosis: a microsimulation modeling study. EClinicalMedicine 2025; 81:103113. [PMID: 40040860 PMCID: PMC11876903 DOI: 10.1016/j.eclinm.2025.103113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/26/2024] [Revised: 01/29/2025] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Background Patients with cirrhosis are at high risk for hepatocellular carcinoma (HCC), but few undergo guideline-recommended semi-annual screening. Randomized clinical trials (RCTs) demonstrate that mailed outreach can increase screening versus visit-based screening. We estimated the costs and cost-effectiveness of an outreach strategy versus usual care. Methods We built a 10-year Markov chain Monte Carlo microsimulation model to conduct a cost-effectiveness analysis comparing a mailed outreach program versus usual care for HCC screening in a cohort of 10,000 patients with cirrhosis. Model inputs were based on literature review (2005-current), and costs were based on inflation-adjusted estimates from Surveillance, Epidemiology, and End Results (SEER)-Medicare claims data. We conducted one-way sensitivity analyses for HCC incidence, outreach costs, efficacy of the outreach strategy to increase screening, and efficacy of curative (versus palliative) HCC treatments. Findings Mailed outreach was estimated to cost $32.45 per patient in the first year and $21.90 per patient in subsequent years. The outreach program increased the number of HCC patients detected at an early stage by 48.4% and increased quality-adjusted life years (QALYs) by 300. Cost savings from these increases offset the costs of mailed outreach. Mailed outreach remained cost-effective across a wide range of HCC incidence rates, outreach costs, efficacy of the outreach strategy to increase screening, and the efficacy of curative HCC treatments. Annual out-of-pocket patient costs in the outreach arm were low at $13 per year. Interpretation Mailed outreach to encourage HCC screening in patients with cirrhosis dominates usual care and should be considered for implementation in routine practice. Funding National Cancer Institute and Cancer Prevention Research Institute of Texas.
Collapse
Affiliation(s)
- Tami Gurley
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ruben Hernaez
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX, USA
| | - Vanessa Cerda
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Tynaje Thomas
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Manasa Narasimman
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Sukul Mittal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Mohammed Al-Hasan
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Darine Daher
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Amit G. Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
8
|
Shin H, Yu SJ. A concise review of updated global guidelines for the management of hepatocellular carcinoma: 2017-2024. JOURNAL OF LIVER CANCER 2025; 25:19-30. [PMID: 39925090 PMCID: PMC12010826 DOI: 10.17998/jlc.2025.02.03] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 02/11/2025]
Abstract
Many guidelines for hepatocellular carcinoma (HCC) have been published and are regularly updated worldwide. HCC management involves a broad range of treatment options and requires multidisciplinary care, resulting in significant heterogeneity in management practices across international communities. To support standardized care for HCC, we systematically appraised 13 globally recognized guidelines and expert consensus statements, including five from Asia, four from Europe, and four from the United States. These guidelines share similarities but reveal notable discrepancies in surveillance strategies, treatment allocation, and other recommendations. Geographic differences in tumor biology (e.g., prevalence of viral hepatitis, alcohol-related liver disease, or metabolic dysfunction-associated steatotic liver disease) and disparities in available medical resources (e.g., organ availability, healthcare infrastructure, and treatment accessibility) complicate the creation of universally applicable guidelines. Previously, significant gaps existed between Asian and Western guidelines, particularly regarding treatment strategies. However, these differences have diminished over the years. Presently, variations are often more attributable to publication dates than to regional differences. Nonetheless, Asia-Pacific experts continue to diverge from the Barcelona Clinic Liver Cancer system, particularly with respect to surgical resection and locoregional therapies, which are viewed as overly conservative in Western guidelines. Advancements in systemic therapies have prompted ongoing updates to these guidelines. Given that each set of guidelines reflects distinct regional characteristics, strengths, and limitations, fostering collaboration and mutual complementarity is essential for addressing discrepancies and advancing global HCC care.
Collapse
Affiliation(s)
- Hyunjae Shin
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Liver Research Institute, Seoul National University, Seoul, Korea
| | - Su Jong Yu
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, Korea
- Liver Research Institute, Seoul National University, Seoul, Korea
| |
Collapse
|
9
|
Fujiwara N, Lopez C, Marsh TL, Raman I, Marquez CA, Paul S, Mishra SK, Kubota N, Katz C, Kanzaki H, Gonzalez M, Quirk L, Deodhar S, Selvakumar P, Raj P, Parikh ND, Roberts LR, Schwartz ME, Nguyen MH, Befeler AS, Page-Lester S, Srivastava S, Feng Z, Reddy KR, Khaderi S, Asrani SK, Kanwal F, El-Serag HB, Marrero JA, Singal AG, Hoshida Y. Phase 3 Validation of PAaM for Hepatocellular Carcinoma Risk Stratification in Cirrhosis. Gastroenterology 2025; 168:556-567.e7. [PMID: 39521255 PMCID: PMC7617545 DOI: 10.1053/j.gastro.2024.10.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Revised: 10/08/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) risk stratification is an urgent unmet need for cost-effective HCC screening and early detection in patients with cirrhosis to improve poor HCC prognosis. METHODS Molecular (prognostic liver secretome signature with α-fetoprotein) and clinical (aMAP [age, male sex, albumin-bilirubin, and platelets] score) variable-based scores were integrated into PAaM (prognostic liver secretome signature with α-fetoprotein plus age, male sex, albumin-bilirubin, and platelets), which was subsequently validated in 2 phase 3 biomarker validation studies: the statewide Texas HCC Consortium and nationwide HCC Early Detection Strategy prospective cohorts, following the prospective specimen collection, retrospective blinded evaluation design. The associations between baseline PAaM and incident HCC were assessed using Fine-Gray regression, with overall death and liver transplantation as competing events. RESULTS Of 2156 patients with cirrhosis in the Texas HCC Consortium, PAaM identified 404 (19%) high-risk, 903 (42%) intermediate-risk, and 849 (39%) low-risk patients with annual HCC incidence rates of 5.3%, 2.7%, and 0.6%, respectively. Compared with low-risk patients, high- and intermediate-risk groups had sub-distribution hazard ratios for incident HCC of 7.51 (95% CI, 4.42-12.8) and 4.20 (95% CI, 2.52-7.01), respectively. Of 1328 patients with cirrhosis in the HCC early detection strategy, PAaM identified 201 high-risk (15%), 540 intermediate-risk (41%), and 587 low-risk (44%) patients, with annual HCC incidence rates of 6.2%, 1.8%, and 0.8%, respectively. High- and intermediate-risk groups were associated with sub-distribution hazard ratios for incident HCC of 6.54 (95% CI, 3.85-11.1) and 1.77 (95% CI, 1.02-3.08), respectively. Subgroup analysis showed robust risk stratification across HCC etiologies, including metabolic dysfunction-associated steatotic liver disease and cured hepatitis C infection. CONCLUSIONS PAaM enables accurate HCC risk stratification in patients with cirrhosis from contemporary etiologies.
Collapse
Affiliation(s)
- Naoto Fujiwara
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas; Department of Gastroenterology and Hepatology, Graduate School of Medicine, Mie University, Mie, Japan
| | - Camden Lopez
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Tracey L Marsh
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Indu Raman
- BioCenter, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Cesia A Marquez
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Subhojit Paul
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sumit K Mishra
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Naoto Kubota
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Courtney Katz
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Hiroaki Kanzaki
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Michael Gonzalez
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Lisa Quirk
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sneha Deodhar
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas
| | | | - Prithvi Raj
- BioCenter, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, Minnesota
| | - Myron E Schwartz
- Department of Surgery, Icahn School of Medicine at Mount Sinai, New York, New York
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, Palo Alto, California
| | - Alex S Befeler
- Division of Gastroenterology and Hepatology, Saint Louis University School of Medicine, St Louis, Missouri
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Sudhir Srivastava
- Cancer Biomarker Research Group, Division of Cancer Prevention, National Cancer Institute, National Institutes of Health, Rockville, Maryland
| | - Ziding Feng
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - K Rajender Reddy
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Saira Khaderi
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | - Sumeet K Asrani
- Baylor University Medical Center, Baylor Scott and White, Dallas, Texas
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, Houston, Texas
| | | | - Jorge A Marrero
- Division of Gastroenterology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Amit G Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| | - Yujin Hoshida
- Division of Digestive and Liver Diseases, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
10
|
Yang L, Tan W, Wang M, Wei Y, Xie Z, Wang Q, Zhang Z, Zhuang H, Ma X, Wang B, Jiang J, Chen Y, Shang C. circCCNY enhances lenvatinib sensitivity and suppresses immune evasion in hepatocellular carcinoma by serving as a scaffold for SMURF1 mediated HSP60 degradation. Cancer Lett 2025; 612:217470. [PMID: 39826668 DOI: 10.1016/j.canlet.2025.217470] [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: 07/11/2024] [Revised: 01/11/2025] [Accepted: 01/16/2025] [Indexed: 01/22/2025]
Abstract
Lenvatinib is the standard first-line therapy for advanced hepatocellular carcinoma (HCC), but drug resistance significantly hampers its efficacy. Increasing evidence has shown that circular RNAs (circRNAs) play critical roles in HCC pathogenesis. However, the underlying mechanisms of lenvatinib sensitivity regulated by circRNAs remain largely unclear. The present study aims to identify circRNAs involved in lenvatinib resistance, as well as to elucidate the underlying mechanisms. High-throughput sequencing revealed that hsa_circ_0000235 (circCCNY) was downregulated in matched HCC tumor tissues and lenvatinib-resistant cells. Both in vitro and in vivo experiments revealed that downregulation of circCCNY could induce lenvatinib resistance in HCC cells. Subsequently, RNA pull-down, mass spectrometry, and RNA immunoprecipitation techniques were employed to investigate the interactions between circCCNY, HSP60, and the E3 ubiquitin ligase SMURF1. Briefly, circCCNY bounds to HSP60, subsequently leading to HSP60 ubiquitination and degradation through its interaction with the E3 ubiquitin ligase SMURF1. As a result, HSP60 degradation released Raf kinase inhibitor protein (RKIP), leading to the inactivation of the MAPK signaling pathway, and subsequently enhanced the anti-tumor effect of lenvatinib against HCC. Moreover, we also demonstrated that circCCNY could enhance CD8+ T-cell infiltration and suppress immune evasion through inhibiting the MAPK/c-Myc/PD-L1 signaling pathway. Our findings revealed that circCCNY enhances HCC sensitivity to lenvatinib and suppresses immune evasion by inhibiting the MAPK signaling pathway in HCC. This suggests that circCCNY could serve as a promising therapeutic target in HCC treatment and a potential biomarker for predicting HCC sensitivity to lenvatinib.
Collapse
MESH Headings
- Humans
- Carcinoma, Hepatocellular/drug therapy
- Carcinoma, Hepatocellular/genetics
- Carcinoma, Hepatocellular/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/metabolism
- Liver Neoplasms/drug therapy
- Liver Neoplasms/genetics
- Liver Neoplasms/immunology
- Liver Neoplasms/pathology
- Liver Neoplasms/metabolism
- Phenylurea Compounds/pharmacology
- Phenylurea Compounds/therapeutic use
- Quinolines/pharmacology
- Quinolines/therapeutic use
- Ubiquitin-Protein Ligases/metabolism
- Ubiquitin-Protein Ligases/genetics
- RNA, Circular/genetics
- RNA, Circular/metabolism
- Animals
- Drug Resistance, Neoplasm/genetics
- Mice
- Cell Line, Tumor
- Tumor Escape/drug effects
- Gene Expression Regulation, Neoplastic
- Xenograft Model Antitumor Assays
- Male
- Mice, Nude
- Female
- Ubiquitination
Collapse
Affiliation(s)
- Lei Yang
- Department of General Surgery, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230000, Anhui, China
| | - Wenliang Tan
- Center of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Min Wang
- Department of Pharmacy, Hainan General Hospital, Hainan Medical University, Haikou, 570311, Hai Nan, China
| | - Yingcheng Wei
- Department of Hepatopancreatobiliary Surgery, Shenshan Medical Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Shanwei, 516621, Guangdong, China
| | - Zhiqin Xie
- Center of Hepatobiliary and Pancreatic Surgery, Zhuzhou Hospital Affiliated to Xiangya School of Medicine, Central South University, Zhuzhou, 412000, Hunan, China
| | - Qingbin Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Ziyu Zhang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Hongkai Zhuang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Xiaowu Ma
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Bingkun Wang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Jiahao Jiang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China
| | - Yajin Chen
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| | - Changzhen Shang
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China; Department of Hepatobiliary Surgery, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou, 510120, Guangdong, China.
| |
Collapse
|
11
|
Nahon P, Lusivika-Nzinga C, Merle P, Zoulim F, Decaens T, Ganne-Carrié N, Pageaux GP, Leroy V, Alric L, Bronowicki JP, Bourlière M, Gournay J, Tran A, Pol S, Mathurin P, Loustaud-Ratti V, Métivier S, De Ledinghen V, Abergel A, Thabut D, D'Alteroche L, Bouattour M, Asselah T, Ouzan D, Cales P, Chazouillères O, Gelu-Simeon M, Roulot D, Boursier J, Cagnot C, Tamazirt S, Pascale A, Nilusmas S, Lewin M, Ziol M, Carrat F, Duclos-Vallée JC. Value of non-invasive test dynamics in guiding HCC surveillance decisions after HCV cure in patients with cirrhosis. J Hepatol 2025:S0168-8278(25)00084-4. [PMID: 40020929 DOI: 10.1016/j.jhep.2025.02.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/02/2024] [Revised: 01/30/2025] [Accepted: 02/04/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND & AIMS Whether the dynamics of non-invasive tests (NITs) correlate with hepatocellular carcinoma (HCC) risk in patients with cirrhosis following sustained virological response (SVR) remains unknown. Thus, we aimed to describe NIT dynamics and assess their correlation with HCC risk. METHODS The dynamics of NITs (fibrosis-4 index [FIB-4], aspartate aminotransferase-to-platelet ratio index [APRI] and liver stiffness measurement) were described in patients with cirrhosis after SVR included in two prospective French multicenter cohorts (ANRS CO22 Hepather and CO12 CirVir) between 2006 and 2015. To assess their relationship with the risk of HCC, a joint modeling approach was employed using both standard and flexible models adjusted for age and sex. The impacts of NIT current value and slope during follow-up on HCC risk were assessed, considering competing risks of death. RESULTS A total of 3,067 patients with cirrhosis who achieved SVR were analyzed, among whom 228 (7.4%) developed HCC and 210 (6.9%) died during a 26-month follow-up. All NITs were increased at baseline in patients who ultimately developed HCC, whereas platelet counts were lower. All NITs improved in patients who did not develop HCC. More varied changes were observed during the follow-up of patients who ultimately developed HCC. Joint model analyses showed that current values of FIB-4, APRI and platelet count at any time impacted HCC risk. Only FIB-4 and APRI slopes influenced the same outcome. When considering NIT current value and slope simultaneously, only the current value of NITs impacted HCC risk while the slopes were not informative. CONCLUSIONS The dynamics of NITs following SVR do not identify patients with cirrhosis who could be safely excluded from surveillance programs. NIT current value is more informative than slope, which will necessitate regularly re-assessment of HCC risk to design individualized surveillance strategies. IMPACT AND IMPLICATIONS It has been postulated that monitoring non-invasive test (NIT) dynamics following HCV cure may provide information on the residual risk of hepatocellular carcinoma (HCC) in patients with cirrhosis and may allow for the discontinuation of surveillance in certain patient subsets. We analyzed data from over 3,000 patients and found that while all NITs improved in patients with cirrhosis who did not develop HCC, those who eventually developed liver cancer showed more varied changes in these tests. Specifically, the current values of tests like FIB-4 (fibrosis-4 index) and APRI (aspartate aminotransferase-to-platelet ratio index) were linked to an increased risk of HCC, while their slopes did not provide additional useful information, suggesting that dedicated prospective studies are warranted to define how repeated measurement of NITs could be combined with other variables into HCC risk stratification algorithms. Until then, HCC surveillance should be maintained in all patients with cirrhosis following HCV eradication, even in case of decreased NIT values.
Collapse
Affiliation(s)
- Pierre Nahon
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Liver Unit, Bobigny; Université Sorbonne Paris Nord, F-93000 Bobigny; Inserm, UMR-1138 "Functional Genomics of Solid Tumors", Centre de Recherche des Cordeliers, Université de Paris, France.
| | - Clovis Lusivika-Nzinga
- Sorbonne Université, Inserm, Institut Pierre Louis d'Epidémiologie et de Santé Publique, AP-HP, Hôpital Saint-Antoine, Unité de Santé Publique, Paris, France
| | - Philippe Merle
- Hepatology Department, Hospices Civils de Lyon; INSERM Unit 1052; Université Claude Bernard Lyon 1; Lyon Hepatology Institute, Lyon, France
| | - Fabien Zoulim
- Hepatology Department, Hospices Civils de Lyon; INSERM Unit 1052; Université Claude Bernard Lyon 1; Lyon Hepatology Institute, Lyon, France
| | - Thomas Decaens
- Univ. Grenoble Alpes, CHU Grenoble Alpes, Inserm U 1209, CNRS UMR 5309, Institute for Advanced Biosciences, Grenoble, France
| | - Nathalie Ganne-Carrié
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Liver Unit, Bobigny; Université Sorbonne Paris Nord, F-93000 Bobigny; Inserm, UMR-1138 "Functional Genomics of Solid Tumors", Centre de Recherche des Cordeliers, Université de Paris, France
| | | | - Vincent Leroy
- AP-HP, Hôpital Henri Mondor, Service d'Hépatologie, Créteil, France
| | - Laurent Alric
- Internal Medicine-Digestive disease Department, CHU Rangueil, Toulouse 3 University, Toulouse, France
| | | | - Marc Bourlière
- Hôpital Saint Joseph, Service d'Hépatologie, Marseille, France
| | | | - Albert Tran
- Université Côte d'Azur, Nice; CHU de Nice, Digestive Center, Nice; INSERM, U1065, C3M, Team 8 « Chronic liver diseases associated with obesity and alcohol », Nice, France
| | - Stanislas Pol
- AP-HP.Centre Université Paris Centre, Groupe Hospitalier Cochin Port Royal, DMU Cancérologie et spécialités médico-chirurgicales, Service des Maladies du foie, Paris; Université Paris Cité, F-75006, Paris, France
| | | | | | | | | | - Armand Abergel
- Hôpital Hôtel Dieu, Service d'Hépatologie, Clermont-Ferrand, France
| | - Dominique Thabut
- AP-HP, Groupe Hospitalier de La Pitié-Salpêtrière, Service d'Hépatologie, Paris, France
| | | | - Mohammed Bouattour
- AP-HP, Hôpital Beaujon, Service d'Hépatologie, and Université Paris Cité, Sorbonne Paris Cité, CRI, UMR 1149, France
| | - Tarik Asselah
- AP-HP, Hôpital Beaujon, Service d'Hépatologie, and Université Paris Cité, Sorbonne Paris Cité, CRI, UMR 1149, France
| | - Denis Ouzan
- Institut Arnaud Tzanck, Service d'Hépatologie, St Laurent du Var, France
| | - Paul Cales
- CHU d'Angers, Service d'Hépatologie, Angers, France
| | - Olivier Chazouillères
- AP-HP, Hôpital Saint-Antoine, Service d'Hépatologie, Sorbonne Université, Paris, France
| | | | - Dominique Roulot
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Unité d'Hépatologie, service de Médecine Interne, Bobigny; Université Sorbonne Paris Nord, F-93000 Bobigny, France
| | | | - Carole Cagnot
- Clinical Research Department, ANRS | Emerging Infectious Diseases, Paris, France
| | - Sonia Tamazirt
- AP-HP, Hôpital Paul Brousse, Unité d'Hépatologie, UMR-1193, FHU Hepatinov, Villejuif, France
| | - Alina Pascale
- AP-HP, Hôpital Paul Brousse, Unité d'Hépatologie, UMR-1193, FHU Hepatinov, Villejuif, France
| | - Samuel Nilusmas
- Sorbonne Université, Inserm, Institut Pierre Louis d'Epidémiologie et de Santé Publique, AP-HP, Hôpital Saint-Antoine, Unité de Santé Publique, Paris, France
| | - Maïté Lewin
- AP-HP, Hôpital Paul Brousse, Service de Radiologie, Villejuif, France
| | - Marianne Ziol
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Pathology Unit and Centre de Ressources biologiques, Bobigny; Université Sorbonne Paris Nord, F-93000 Bobigny; Inserm, UMR-1138 "Functional Genomics of Solid Tumors", Centre de Recherche des Cordeliers, Université de Paris, France
| | - Fabrice Carrat
- Sorbonne Université, Inserm, Institut Pierre Louis d'Epidémiologie et de Santé Publique, AP-HP, Hôpital Saint-Antoine, Unité de Santé Publique, Paris, France
| | | |
Collapse
|
12
|
Arvind A, Redmon K, Singal AG. Persisting challenges in the early detection of hepatocellular carcinoma. Expert Rev Anticancer Ther 2025:1-12. [PMID: 39943795 DOI: 10.1080/14737140.2025.2467184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Prognosis in patients with HCC is largely determined by stage at diagnosis, highlighting the importance of effective early detection strategies. HCC surveillance is associated with increased early detection and reduced HCC-related mortality and is currently recommended in patients with cirrhosis or chronic HBV infection. AREAS COVERED We performed a targeted literature review to identify limitations of current HCC surveillance practices and strategies for improvement. EXPERT OPINION Semi-annual ultrasound continues as the cornerstone modality for HCC surveillance but has limited sensitivity for detecting early-stage HCC, particularly in patients with obesity and non-viral etiologies. Although sensitivity for early-stage HCC can be improved by using ultrasound with alpha fetoprotein, this strategy misses over one-third of HCC at an early stage. Emerging imaging and biomarker-based surveillance strategies currently remain in varying stages of validation and are not yet ready for routine use in practice. The cost-effectiveness of surveillance in patients with non-cirrhotic liver disease related to hepatitis C or metabolic dysfunction-associated steatotic liver disease continues to be debated, although subgroups with advanced fibrosis may warrant surveillance. Finally, the effectiveness of surveillance is diminished by underuse in clinical practice, particularly in racial minority and low-income groups, highlighting a need for interventions to increase utilization.
Collapse
Affiliation(s)
- Ashwini Arvind
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Kennedy Redmon
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Amit G Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
13
|
Chen J, Wang X, Bai Y, Li Z, Li H, Wang B, Gong Q, Luo K. An ASGP-R-targeting magnetic resonance imaging contrast agent for liver cancer diagnosis. J Mater Chem B 2025; 13:2549-2558. [PMID: 39838934 DOI: 10.1039/d4tb02708f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
Development of novel Gd-based contrast agents for targeted magnetic resonance imaging (MRI) of liver cancer remains a great challenge. Herein we reported a novel Gd-based MRI contrast agent with improved relaxivity for specifically diagnosing liver cancer. This GSH-responsive macromolecular contrast agent (mCA), POLDGd, was prepared by RAFT polymerization, and its lactic acid moiety could precisely target the ASGP-R surface protein on liver cancer cells, whereas PODGd without the lactic acid moiety was prepared as a control. POLDGd had a high molecular weight of 45 kDa and a particle size of 103 nm. Its longitudinal relaxivity (11.39 mM-1 s-1) measured via a 3.0 T MR scanner was three times that of the clinically used contrast agent DTPA-Gd. In comparison with the PODGd-treated group, the signal enhancement at the tumor site was significantly prolonged, with a maximum enhancement peak of about 190% after intravenous injection of POLDGd into tumor-bearing mice. A high accumulation level of POLDGd in the liver tumors observed via MRI was also confirmed by fluorescence imaging. POLDGd showed minimal side effects, which may be ascribed to its metabolism through the kidneys. Therefore, POLDGd may be used as a highly effective biosafe nanoscale contrast agent for targeted MRI of liver cancer.
Collapse
Affiliation(s)
- Jie Chen
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Xiaoming Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Yinan Bai
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Zhiqian Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haonan Li
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Bing Wang
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Qiyong Gong
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
- Xiamen Key Lab of Psychoradiology and Neuromodulation, Department of Radiology, West China Xiamen Hospital of Sichuan University, Xiamen 361021, China
| | - Kui Luo
- Department of Radiology, Huaxi MR Research Center (HMRRC), Institution of Radiology and Medical Imaging, Breast Center, Institute of Breast Health Medicine, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
- Functional and Molecular Imaging Key Laboratory of Sichuan Province, Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu 610041, China
| |
Collapse
|
14
|
Marsh TL, Parikh ND, Roberts LR, Schwartz ME, Nguyen MH, Befeler A, Page-Lester S, Tayob N, Srivastava S, Rinaudo JA, Singal AG, Reddy KR, Marrero JA. A Phase 3 Biomarker Validation of GALAD for the Detection of Hepatocellular Carcinoma in Cirrhosis. Gastroenterology 2025; 168:316-326.e6. [PMID: 39293548 DOI: 10.1053/j.gastro.2024.09.008] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 08/12/2024] [Accepted: 09/02/2024] [Indexed: 09/20/2024]
Abstract
BACKGROUND & AIMS Better surveillance tests for hepatocellular carcinoma (HCC) are needed. The GALAD score (gender, age, α-fetoprotein [AFP] L3, AFP, and des-γ carboxyprothrombin) has been shown to have excellent sensitivity and specificity for HCC in phase 2 studies. We performed a phase 3 biomarker validation study to compare GALAD with AFP in detecting HCC. METHODS This is a prospective study of patients with cirrhosis enrolled at 7 centers. Surveillance for HCC was performed every 6 months at each site, and HCC diagnosis was confirmed per American Association for the Study of Liver Diseases guidelines. Blood for biomarker research was obtained at each follow-up visit and stored in a biorepository. Measurements of AFP, AFP-L3, and des-γ carboxyprothrombin) were performed in a FujiFilm laboratory by staff blinded to clinical data. The performance of GALAD in detecting HCC was retrospectively evaluated within 12 months before the clinical diagnosis. All analyses were conducted by an unblinded statistician in the Early Detection Research Network data management and coordinating center. RESULTS A total of 1,558 patients with cirrhosis were enrolled and followed for a median of 2.2 years. A total of 109 patients developed HCC (76 very early or early stage), with an annual incident rate of 2.4%. The areas under the curve for AFP and GALAD within 12 months before HCC were 0.66 and 0.78 (P < .001), respectively. Using a cutoff for GALAD of -1.36, the specificity was 82%, and the sensitivity at 12 months before HCC diagnosis was 62%. For comparison, performance of AFP at 82% specificity showed 41% sensitivity at 12 months before HCC diagnosis (P = .001). CONCLUSIONS GALAD score, compared to AFP, improves the detection of HCC within 12 months before the actual diagnosis.
Collapse
Affiliation(s)
- Tracey L Marsh
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan
| | - Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, Rochester, Minnesota
| | - Myron E Schwartz
- Recanati/Miller Transplant Institute, Mount Sinai Medical Center, New York, New York
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology and Department of Epidemiology and Population Health, Stanford University, Palo Alto, California
| | - Alex Befeler
- Division of Gastroenterology, Saint Louis University, St. Louis, Missouri
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Sciences Division, Fred Hutchinson Cancer Research Center, Seattle, Washington
| | - Nabihah Tayob
- Department of Data Science, Dana Farber Cancer Institute, Boston, Massachusetts
| | - Sudhir Srivastava
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Jo Ann Rinaudo
- Cancer Biomarkers Research Group, Division of Cancer Prevention, National Cancer Institute, Bethesda, Maryland
| | - Amit G Singal
- Division of Digestive and Liver Disease, University of Texas Southwestern, Dallas, Texas
| | - K Rajender Reddy
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Jorge A Marrero
- Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania.
| |
Collapse
|
15
|
Romeo M, Dallio M, Napolitano C, Basile C, Di Nardo F, Vaia P, Iodice P, Federico A. Clinical Applications of Artificial Intelligence (AI) in Human Cancer: Is It Time to Update the Diagnostic and Predictive Models in Managing Hepatocellular Carcinoma (HCC)? Diagnostics (Basel) 2025; 15:252. [PMID: 39941182 PMCID: PMC11817573 DOI: 10.3390/diagnostics15030252] [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: 12/23/2024] [Revised: 01/20/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
In recent years, novel findings have progressively and promisingly supported the potential role of Artificial intelligence (AI) in transforming the management of various neoplasms, including hepatocellular carcinoma (HCC). HCC represents the most common primary liver cancer. Alarmingly, the HCC incidence is dramatically increasing worldwide due to the simultaneous "pandemic" spreading of metabolic dysfunction-associated steatotic liver disease (MASLD). MASLD currently constitutes the leading cause of chronic hepatic damage (steatosis and steatohepatitis), fibrosis, and liver cirrhosis, configuring a scenario where an HCC onset has been reported even in the early disease stage. On the other hand, HCC represents a serious plague, significantly burdening the outcomes of chronic hepatitis B (HBV) and hepatitis C (HCV) virus-infected patients. Despite the recent progress in the management of this cancer, the overall prognosis for advanced-stage HCC patients continues to be poor, suggesting the absolute need to develop personalized healthcare strategies further. In this "cold war", machine learning techniques and neural networks are emerging as weapons, able to identify the patterns and biomarkers that would have normally escaped human observation. Using advanced algorithms, AI can analyze large volumes of clinical data and medical images (including routinely obtained ultrasound data) with an elevated accuracy, facilitating early diagnosis, improving the performance of predictive models, and supporting the multidisciplinary (oncologist, gastroenterologist, surgeon, radiologist) team in opting for the best "tailored" individual treatment. Additionally, AI can significantly contribute to enhancing the effectiveness of metabolomics-radiomics-based models, promoting the identification of specific HCC-pathogenetic molecules as new targets for realizing novel therapeutic regimens. In the era of precision medicine, integrating AI into routine clinical practice appears as a promising frontier, opening new avenues for liver cancer research and treatment.
Collapse
Affiliation(s)
- Mario Romeo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Marcello Dallio
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Carmine Napolitano
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Claudio Basile
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Fiammetta Di Nardo
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | - Paolo Vaia
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| | | | - Alessandro Federico
- Hepatogastroenterology Division, Department of Precision Medicine, University of Campania Luigi Vanvitelli, 80138 Naples, Italy; (M.R.); (C.N.); (C.B.); (F.D.N.); (P.V.); (A.F.)
| |
Collapse
|
16
|
Rhee H, Kim MJ, Kim DY, An C, Kang W, Han K, Roh YH, Han KH, Ahn SH, Choi JY, Park JY, Chung YE, Kim SU, Kim BK, Lee S, Lee HW, Lee JS. Noncontrast Magnetic Resonance Imaging vs Ultrasonography for Hepatocellular Carcinoma Surveillance: A Randomized, Single-Center Trial. Gastroenterology 2025:S0016-5085(25)00049-6. [PMID: 39855314 DOI: 10.1053/j.gastro.2024.12.035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 01/27/2025]
Abstract
BACKGROUND & AIMS This study aimed to compare ultrasonography (US) and noncontrast magnetic resonance imaging (MRI) in the surveillance of hepatic malignancy. METHODS We conducted a randomized, nonblinded trial at a single center in South Korea. Eligible individuals were aged 20 to 70 years with liver cirrhosis, Child-Pugh class A, and no history of liver cancer or other recent malignancy. Participants were randomized 1:1 to receive up to 10 semiannual surveillance using US or noncontrast MRI with serum alpha-fetoprotein testing. The primary endpoints were the detection rates of Barcelona Clinic Liver Cancer (BCLC) stage 0 or A tumors, stage distribution at initial diagnosis, and false-positive referral rates. RESULTS From June 2015 to November 2017, 416 patients were screened, and 414 were enrolled and assigned to the US (n = 207) or MRI (n = 207) group. In total, 23 participants in the US group and 25 in the MRI group were diagnosed with liver cancer by November 2022. The detection rates of BCLC stage 0 or A tumors were not different between the US and MRI groups (7% [95% confidence interval (CI), 4%-11%] vs 12% [8%-17%]). BCLC stage 0 tumors were more frequently detected in the MRI group than in the US group (8% vs 2%). The MRI group had earlier BCLC stage (P = .014) and lower false-positive referral rate (0.7% [95% CI, 0.4%-1.2%] vs 3.1% [2.3%-4.1%], P < .001) compared with the US group. CONCLUSIONS Noncontrast MRI is a better alternative to US for the surveillance of cirrhotic patients offering earlier stage at initial diagnosis and lower false-positive referral rate. (ClincalTrials.gov, Number: NCT02514434.).
Collapse
Affiliation(s)
- Hyungjin Rhee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Myeong-Jin Kim
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| | - Do Young Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea.
| | - Chansik An
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Wonseok Kang
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Kyunghwa Han
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yun Ho Roh
- Biostatistics Collaboration Unit, Medical Research Center, Yonsei University College of Medicine, Seoul, South Korea
| | - Kwang-Hyub Han
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sang Hoon Ahn
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jin-Young Choi
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jun Yong Park
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Yong Eun Chung
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Seung Up Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Beom Kyung Kim
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Sunyoung Lee
- Department of Radiology, Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Hye Won Lee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| | - Jae Seung Lee
- Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, South Korea
| |
Collapse
|
17
|
Jia J, Zhou X, Chu Q. Mechanisms and therapeutic prospect of the JAK-STAT signaling pathway in liver cancer. Mol Cell Biochem 2025; 480:1-17. [PMID: 38519710 DOI: 10.1007/s11010-024-04983-5] [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: 12/12/2023] [Accepted: 02/29/2024] [Indexed: 03/25/2024]
Abstract
Liver cancer (LC) poses a significant global health challenge due to its high incidence and poor prognosis. Current systemic treatment options, such as surgery, chemotherapy, radiofrequency ablation, and immunotherapy, have shown limited effectiveness for advanced LC patients. Moreover, owing to the heterogeneous nature of LC, it is crucial to uncover more in-depth pathogenic mechanisms and develop effective treatments to address the limitations of the existing therapeutic modalities. Increasing evidence has revealed the crucial role of the Janus kinase (JAK)-signal transducer and activator of transcription (STAT) pathway in the pathogenesis of LC. The specific mechanisms driving the JAK-STAT pathway activation in LC, participate in a variety of malignant biological processes, including cell differentiation, evasion, anti-apoptosis, immune escape, and treatment resistance. Both preclinical and clinical investigations on the JAK-STAT pathway inhibitors have exhibited potential in LC treatment, thereby opening up avenues for the development of more targeted therapeutic strategies for LC. In this study, we provide an overview of the JAK-STAT pathway, delving into the composition, activation, and dynamic interplay within the pathway. Additionally, we focus on the molecular mechanisms driving the aberrant activation of the JAK-STAT pathway in LC. Furthermore, we summarize the latest advancements in targeting the JAK-STAT pathway for LC treatment. The insights presented in this review aim to underscore the necessity of research into the JAK-STAT signaling pathway as a promising avenue for LC therapy.
Collapse
Affiliation(s)
- JunJun Jia
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, No. 79 Qingchun Road, Shangcheng District, Hangzhou, 310003, Zhejiang, China.
| | - Xuelian Zhou
- Division of Endocrinology, National Clinical Research Center for Child Health, Children's Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Qingfei Chu
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| |
Collapse
|
18
|
Galvanin C, Buch S, Nahon P, Trépo E. PNPLA3 in Alcohol-Related Liver Disease. Liver Int 2025; 45:e16211. [PMID: 39679853 DOI: 10.1111/liv.16211] [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: 11/18/2024] [Revised: 11/28/2024] [Accepted: 11/29/2024] [Indexed: 12/17/2024]
Abstract
The discovery of PNPLA3 as a genetic risk factor for liver disease has transformed our understanding of the pathogenesis of alcohol-related liver disease (ALD). The recent reclassification of fatty liver disease as steatotic liver disease (SLD), introducing metabolic dysfunction and alcohol-related liver disease (MetALD), has highlighted how genetic and environmental factors synergistically drive liver damage. The PNPLA3 rs738409 variant stands as a paradigmatic example of gene-environment interaction, where its effect on liver disease is dramatically amplified by alcohol consumption, obesity and type 2 diabetes. Understanding these interactions has revealed novel pathogenic mechanisms. The robust genetic evidence and a growing understanding of molecular mechanisms have made PNPLA3 an attractive therapeutic target. Several compounds targeting PNPLA3 are now in clinical development. While initial trials have focused on metabolic dysfunction-associated SLD, the recognition that almost all individuals with excessive alcohol consumption have metabolic comorbidities provides an unprecedented opportunity to evaluate these genetically informed therapies in MetALD. In this review, we examine the role of PNPLA3 in ALD, focusing on gene-environment interactions and therapeutic implications in the context of the new disease classification framework.
Collapse
Affiliation(s)
- Clélia Galvanin
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Stephan Buch
- Department of Medicine I, Dresden University Hospital, Dresden, Germany
- Center for Regenerative Therapies Dresden (CRTD), Technische Universität Dresden (TU Dresden), Dresden, Germany
| | - Pierre Nahon
- Service d'Hépatologie, AP-HP Avicenne, Bobigny, France
- Unité de Formation et de Recherche Santé Médecine et Biologie Humaine, Université Paris 13, Communauté d'Universités et Etablissements Sorbonne Paris Cité, Paris, France
- Centre de Recherche des Cordeliers, Sorbonne Université, Inserm, Université de Paris, Team «Functional Genomics of Solid Tumors», Paris, France
| | - Eric Trépo
- Department of Gastroenterology, Hepatopancreatology and Digestive Oncology, Hôpital Universitaire de Bruxelles, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
19
|
Singh M, Alka, Shukla P, Wen ZH, Ko CY, Vinayagam R. TPGS-modified Chitosan Nanoparticles of EGFR Inhibitor: Physicochemical and In vitro Evaluation against HepG2 Cell Lines. Curr Drug Deliv 2025; 22:465-478. [PMID: 38204256 DOI: 10.2174/0115672018268315231206045504] [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: 07/11/2023] [Revised: 10/12/2023] [Accepted: 10/23/2023] [Indexed: 01/12/2024]
Abstract
BACKGROUND Gefitinib (GFN) is an Epithelial Growth Factor Receptor (EGFR) inhibitor, and Food and Drug Administration (FDA) has approved medication to treat lung cancer. However, this investigation aimed to produce and characterize Gefitinib (GFN)-loaded chitosan and soy lecithin nanoparticles (NPs) modified with D-α-tocopheryl polyethylene glycol 1000 succinate mono ester (TPGS) and assess their therapeutic potential against HepG2 liver cell lines. METHODS Chitosan, a cationic polymer with biocompatible and biodegradable properties, was combined with soy lecithin to develop the NPs loaded with GFN using a self-organizing ionic interaction methodology. RESULTS The entrapment efficiency and drug loading were found to be 59.04±4.63 to 87.37±3.82% and 33.46±3.76 to 49.50±4.35%, respectively, and results indicated the encapsulation of GEN in NPs. The pH of the formulations was observed between 4.48-4.62. Additionally, all the prepared NPs showed the size and PDI range of 89.2±15.9 nm to 799.2±35.8 nm and 0.179±0.065 to 0.455±0.097, respectively. The FTIR bands in optimized formulation (GFN-NP1) indicated that the drug might be contained within the NP's core. The SEM photograph revealed the spherical shape of NPs. The kinetic release model demonstrated the combination of diffusion and erosion mechanisms. The IC50 value of GFN and GFN-NP1 formulation against the HepG2 cell lines were determined and found to be 63.22±3.36 μg/ml and 45.80±2.53 μg/ml, respectively. DAPI and PI staining agents were used to detect nuclear morphology. CONCLUSION It was observed that the optimized GFN-NP1 formulation successfully internalized and inhibited the growth of HepG2 cells. Hence, it can be concluded that the prepared NPs can be a new therapeutic option for treating liver cancer.
Collapse
Affiliation(s)
- Mahendra Singh
- Department of Biotechnology, Institute of Biotechnology, Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| | - Alka
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India
| | - Prashant Shukla
- Department of Pharmaceutical Sciences, School of Health Sciences and Technology, UPES, Dehradun 248007, Uttarakhand, India
| | - Zhi-Hong Wen
- Department of Marine Biotechnology and Resources, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
- Institute of BioPharmaceutical Sciences, National Sun Yat-sen University, Kaohsiung 804201, Taiwan
| | - Chou-Yuan Ko
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Kaohsiung Armed Forces General Hospital, Kaohsiung 80284, Taiwan
- Institute of Medical Science and Technology, National Sun Yat-sen University, Kaohsiung 80424, Taiwan
| | - Ramachandran Vinayagam
- Department of Biotechnology, Institute of Biotechnology, Life and Applied Sciences, Yeungnam University, Gyeongsan, Gyeongbuk 38541, Republic of Korea
| |
Collapse
|
20
|
He X, Zhang X, Li Z, Niu X, Li L, Liu Z, Ren H, Zhang D. Total bile acid as a preoperative risk factor for post-hepatectomy liver failure in patients with hepatocellular carcinoma and normal bilirubin. Cir Esp 2024; 102:642-648. [PMID: 39393491 DOI: 10.1016/j.cireng.2024.09.007] [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: 06/17/2024] [Accepted: 09/13/2024] [Indexed: 10/13/2024]
Abstract
BACKGROUND AND AIMS Total bile acid (TBA) is associated with portal hypertension, a risk factor for post-hepatectomy liver failure (PHLF). We conducted this study to clarify whether TBA is also associated with PHLF in patients with hepatocellular carcinoma (HCC). METHODS We recruited patients with HCC and Child-Pugh class A, who underwent liver resection, and applied multivariate analyses to identify risk factors for PHLF. RESULTS We analyzed data from 154 patients. The prevalence of PHLF was 14.3%. The median maximum tumor diameter was 5.1 cm (2.9-6.9 cm). The proportions of patients with elevated TBA levels (P = 0.001), severe albumin-bilirubin (AIBL) grades (P = 0.033), and low platelet counts (P = 0.031) were significantly higher within the subgroup of patients with PHLF than in the subgroup without PHLF. The multivariate analysis results suggest that TBA level (OR, 1.08; 951.03-1.14; P = 0.003) and MRI tumor diameter (OR, 1.17; 95% CI, 1.01-1.35; P = 0.038) are independent preoperative risk factors for PHLF. The TBA levels correlated with the indocyanine green retention rate at 15 minutes (P = 0.001) and the effective hepatic blood flow (P < 0.001), two markers of portal hypertension. However, TBA levels did not correlate with tumor diameter (P = 0.536). CONCLUSIONS Compared to ICG R15 and AIBL score, preoperative TBA was risk factor for PHLF in Chinese patients with HCC, and it may impact PHLF through its potential role as a marker of portal hypertension.
Collapse
Affiliation(s)
- Xi He
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaofeng Zhang
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhijie Li
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Xiaofeng Niu
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Lixin Li
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Zhenwen Liu
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China
| | - Hui Ren
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| | - Dali Zhang
- Senior Department of Hepatology, The Fifth Medical Center of Chinese People's Liberation Army General Hospital, Beijing, China.
| |
Collapse
|
21
|
Wu G, Bajestani N, Pracha N, Chen C, Makary MS. Hepatocellular Carcinoma Surveillance Strategies: Major Guidelines and Screening Advances. Cancers (Basel) 2024; 16:3933. [PMID: 39682122 DOI: 10.3390/cancers16233933] [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: 10/10/2024] [Revised: 11/14/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
BACKGROUND/OBJECTIVES Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths globally, with prognosis and treatment outcomes that are significantly influenced by the stage at diagnosis. Early detection through regular surveillance is crucial for improving patient outcomes, especially in high-risk groups such as those with cirrhosis or chronic hepatitis B. Geographic variations in HCC risk factors, including viral hepatitis and non-alcoholic fatty liver disease (NAFLD), have led to the development of different international surveillance guidelines. This review aims to compare and evaluate the surveillance strategies proposed by the Asian Pacific Association for the Study of the Liver (APASL), the American Association for the Study of Liver Diseases (AASLD), and the European Association for the Study of the Liver and European Organization for Research and Treatment of Cancer (EASL-EORTC). METHODS The review examined and compared major international guidelines on HCC surveillance, focusing on patient selection, imaging modalities, and the integration of biomarkers. We also explored recent advancements in screening techniques, including artificial intelligence and emerging biomarkers, to identify future directions for improving surveillance strategies. RESULTS Our analysis identified key differences in the guidelines, particularly in imaging modality preferences and the use of biomarkers for early detection. While all guidelines place emphasis on high-risk populations, the inclusion criteria and surveillance intervals vary. Additionally, novel technologies such as artificial intelligence show potential to enhance the accuracy and efficiency of HCC detection. CONCLUSIONS This review highlights the need to harmonize the international guidelines, particularly in regard to patients with non-cirrhotic NAFLD who remain under-represented in current surveillance protocols. Future research should focus on integrating emerging technologies and biomarkers to improve early detection and overall patient outcomes.
Collapse
Affiliation(s)
- Gavin Wu
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nojan Bajestani
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Nooruddin Pracha
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Cindy Chen
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| | - Mina S Makary
- Department of Radiology, The Ohio State University Wexner Medical Center, Columbus, OH 43210, USA
| |
Collapse
|
22
|
Singal AG, Parikh ND, Kanwal F, Marrero JA, Deodhar S, Page-Lester S, Lopez C, Feng Z, Tayob N. National Liver Cancer Screening Trial (TRACER) study protocol. Hepatol Commun 2024; 8:e0565. [PMID: 39495136 PMCID: PMC11537583 DOI: 10.1097/hc9.0000000000000565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Professional guidelines recommend HCC screening in at-risk patients using semi-annual ultrasound with or without alpha-fetoprotein (AFP); however, this strategy has limited effectiveness due to low adherence and sensitivity. Increasing data support the potential role of blood-based biomarker panels, which could improve both aspects. The biomarker panel GALAD, comprised of sex, age, and 3 blood biomarkers (AFP, AFP-L3, and des-carboxy prothrombin des-carboxy prothrombin), has shown high sensitivity and specificity in biomarker phase II (case-control) and phase III (retrospective cohort) validation studies. However, prospective validation in a large phase IV biomarker clinical utility trial is necessary before its adoption in practice. METHODS The National Liver Cancer Screening Trial is an adaptive pragmatic randomized phase IV trial, which began enrollment in January 2024, comparing ultrasound-based versus biomarker-based screening in 5500 patients with chronic hepatitis B infection or cirrhosis from any etiology. Eligible patients are randomly assigned in a 1:1 ratio to semi-annual screening with ultrasound ± alpha-fetoprotein (arm A) or semi-annual screening with GALAD (arm B). Randomization is stratified by enrollment site, liver disease severity (per Child-Pugh class), liver disease etiology (viral, nonviral, and noncirrhotic HBV), and sex. Patients are being recruited from 15 sites (a mix of tertiary care academic referral centers, safety-net health systems, and large community health systems) over a 3-year period, and the primary endpoint, reduction in late-stage HCC, will be assessed at the end of year 5.5. DISCUSSION The results of this trial will inform the best strategy for HCC screening and early-stage detection in patients with chronic liver diseases. If GALAD shows superiority, HCC screening would primarily shift from an ultrasound-based strategy to the adoption of the biomarker panel. TRIAL REGISTRATION NCT06084234. TRIAL STATUS The TRACER Study is actively enrolling.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jorge A. Marrero
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sneha Deodhar
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Camden Lopez
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ziding Feng
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nabihah Tayob
- Department of Data Science, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
23
|
Singal AG, Parikh ND, Shetty K, Han SH, Xie C, Ning J, Rinaudo JA, Arvind A, Lok AS, Kanwal F. Natural History of Indeterminate Liver Nodules in Patients With Advanced Liver Disease: A Multicenter Retrospective Cohort Study. Am J Gastroenterol 2024; 119:2251-2258. [PMID: 38686922 PMCID: PMC11534566 DOI: 10.14309/ajg.0000000000002827] [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: 06/26/2023] [Accepted: 03/11/2024] [Indexed: 05/02/2024]
Abstract
INTRODUCTION Indeterminate liver nodules (ILNs) are frequently encountered on diagnostic imaging after positive hepatocellular carcinoma (HCC) surveillance results, but their natural history remains unclear. METHODS We conducted a multicenter retrospective cohort study among patients with ≥1 newly detected LI-RADS 3 (LR-3) lesion ≥1 cm or LI-RADS 4 (LR-4) lesion of any size (per LI-RADS v2018) between January 2018 and December 2019. Patients were followed with repeat imaging at each site per institutional standard of care. Multivariable Fine-Gray models were used to evaluate associations between potential risk factors and patient-level time-to-HCC diagnosis, with death and liver transplantation as competing risks. RESULTS Of 307 patients with ILNs, 208 had LR-3 lesions, 83 had LR-4 lesions, and 16 had both LR-3 and LR-4 lesions. HCC incidence rates for patients with LR-3 and LR-4 lesions were 110 (95% CI 70-150) and 420 (95% CI 310-560) per 1,000 person-year, respectively. In multivariable analysis, incident HCC among patients with LR-3 lesions was associated with older age, thrombocytopenia (platelet count ≤150 ×10 9 /L), and elevated serum alpha-fetoprotein levels. Among those with LR-4 lesions, incident HCC was associated with a maximum lesion diameter >1 cm. Although most patients had follow-up computed tomography or magnetic resonance imaging, 13.7% had no follow-up imaging and another 14.3% had follow-up ultrasound only. DISCUSSION ILNs have a high but variable risk of HCC, with 4-fold higher risk in patients with LR-4 lesions than those with LR-3 lesions, highlighting a need for accurate risk stratification tools and close follow-up in this population.
Collapse
Affiliation(s)
- Amit G Singal
- Division of Digestive and Liver Diseases, University of Texas Southwestern, Dallas, Texas, USA
| | - Neehar D Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Kirti Shetty
- Division of Gastroenterology and Hepatology, University of Maryland, Baltimore, Maryland, USA
| | - Steven-Huy Han
- Pfleger Liver Institute, Vatche and Tamar Manoukian Division of Digestive Diseases, UCLA, Los Angeles, California, USA
| | - Cassie Xie
- Department of Biostatistics, Fred Hutchinson Cancer Center, Seattle, Washington, USA
| | - Jing Ning
- Department of Biostatistics, University of Texas MD Anderson Cancer Center, Houston, Texas, USA
| | | | - Ashwini Arvind
- Division of Digestive and Liver Diseases, University of Texas Southwestern, Dallas, Texas, USA
| | - Anna S Lok
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Fasiha Kanwal
- Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- VA HSR'D Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
24
|
Campani C, Imbeaud S, Couchy G, Ziol M, Hirsch TZ, Rebouissou S, Noblet B, Nahon P, Hormigos K, Sidali S, Seror O, Taly V, Ganne Carrie N, Laurent-Puig P, Zucman-Rossi J, Nault JC. Circulating tumour DNA in patients with hepatocellular carcinoma across tumour stages and treatments. Gut 2024; 73:1870-1882. [PMID: 39054058 DOI: 10.1136/gutjnl-2024-331956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024]
Abstract
OBJECTIVE Circulating tumour DNA (ctDNA) is a promising non-invasive biomarker in cancer. We aim to assess the dynamic of ctDNA in patients with hepatocellular carcinoma (HCC). DESIGN We analysed 772 plasmas from 173 patients with HCC collected at the time of diagnosis or treatment (n=502), 24 hours after locoregional treatment (n=154) and during follow-up (n=116). For controls, 56 plasmas from patients with chronic liver disease without HCC were analysed. All samples were analysed for cell free DNA (cfDNA) concentration, and for mutations in TERT promoter, CTNNB1, TP53, PIK3CA and NFE2L2 by sequencing and droplet-based digital PCR. Results were compared with 232 corresponding tumour samples. RESULTS In patients with active HCC, 40.2% of the ctDNA was mutated vs 14.6% in patients with inactive HCC and 1.8% in controls (p<0.001). In active HCC, we identified 27.5% of mutations in TERT promoter, 21.3% in TP53, 13.1% in CTNNB1, 0.4% in PIK3CA and 0.2% in NFE2L2, most of the times similar to those identified in the corresponding tumour. CtDNA mutation rate increased with advanced tumour stages (p<0.001). In 103 patients treated by percutaneous ablation, the presence and number of mutations in the ctDNA before treatment were associated with higher risk of death (p=0.001) and recurrence (p<0.001). Interestingly, cfDNA concentration and detectable mutations increased 24 hours after a locoregional treatment. Among 356 plasmas collected in 53 patients treated by systemic treatments, we detected mutations at baseline in 60.4% of the cases. In patients treated by atezolizumab-bevacizumab, persistence of mutation in ctDNA was associated with radiological progression (63.6% vs 36.4% for disappearance, p=0.019). In two patients progressing under systemic treatments, we detected the occurrence of mutations in CTNNB1 in the plasma that was subclonal in the tumour for one patient and not detectable in the tumour for the other one. CONCLUSION ctDNA offers dynamic information reflecting tumour biology. It represents a non-invasive tool useful to guide HCC clinical management.
Collapse
Affiliation(s)
- Claudia Campani
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Internal Medicine and Hepatology Unit, Department of Experimental and Clinical Medicine, University of Firenze, Florence, Italy
| | - Sandrine Imbeaud
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Gabrielle Couchy
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Marianne Ziol
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Pathology Department and Biological Resource Center Center (BB-0033-00027), Paris-Seine-Saint-Denis, University Hospital, Avicenne Hospital, APHP, Sorbonne Paris Nord University, Bobugny, France
| | - Theo Z Hirsch
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Sandra Rebouissou
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Bénédicte Noblet
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
| | - Pierre Nahon
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| | - Katia Hormigos
- Cordeliers Research Center, INSERM, CNRS SNC 5096, Sorbonne University, Paris Cité University, Paris, France
| | - Sabrina Sidali
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver unit, Paris Cité University, Beaujon Hospital, APHP, DMU DIGEST, Clichy, France
| | - Olivier Seror
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Interventional Radiology Unit, Avicenne Hospital, APHP, Bobigny, Paris, France
| | - Valerie Taly
- Cordeliers Research Center, INSERM, CNRS SNC 5096, Sorbonne University, Paris Cité University, Paris, France
| | - Nathalie Ganne Carrie
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| | - Pierre Laurent-Puig
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Institut of Cancer Paris CARPEM, AP-HP-Hôpital Européen Georges Pompidou, Paris, France
| | - Jessica Zucman-Rossi
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Cordeliers Research Center, INSERM, Sorbonne University, Paris Cité University, Institut of Cancer Paris CARPEM, AP-HP-Hôpital Européen Georges Pompidou, Paris, France
| | - Jean-Charles Nault
- Cordeliers Research Center, INSERM, Paris Cité University, "Functional Genomics of Solid Tumors" Team, Ligue Nationale Contre le Cancer Accredited Team, Labex OncoImmunology, Sorbonne Université, Université Paris Cité, Paris, France
- Liver Unit, Avicenne Hospital, APHP, University Sorbonne Paris Nord, Bobigny, France
| |
Collapse
|
25
|
Teng W, Wang HW, Lin SM. Management Consensus Guidelines for Hepatocellular Carcinoma: 2023 Update on Surveillance, Diagnosis, Systemic Treatment, and Posttreatment Monitoring by the Taiwan Liver Cancer Association and the Gastroenterological Society of Taiwan. Liver Cancer 2024; 13:468-486. [PMID: 39435274 PMCID: PMC11493393 DOI: 10.1159/000537686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 02/02/2024] [Indexed: 10/08/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is the leading cause of cancer-related mortality in Taiwan. The Taiwan Liver Cancer Association and the Gastroenterological Society of Taiwan established HCC management consensus guidelines in 2016 and updated them in 2023. Current recommendations focus on addressing critical issues in HCC management, including surveillance, diagnosis, systemic treatment, and posttreatment monitoring. For surveillance and diagnosis, we updated the guidelines to include the role of protein induced by vitamin K absence or antagonist II (PIVKA-II) and gadoxetic acid (Gd-EOB-DTPA)-enhanced magnetic resonance imaging (MRI) in detecting HCCs. For systemic treatment, the updated guidelines summarize the multiple choices available for targeted therapy, immune checkpoint inhibitors, and a combination of both, especially for those carcinomas refractory to or unsuitable for transarterial chemoembolization. We have added a new section, posttreatment monitoring, that describes the important roles of PIVKA-II and EOB-MRI after HCC therapy, including surgery, locoregional therapy, and systemic treatment. Through this update of the management consensus guidelines, patients with HCC may benefit from optimal diagnosis, therapeutic modalities, and posttreatment monitoring.
Collapse
Affiliation(s)
- Wei Teng
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Wei Wang
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| | - Shi-Ming Lin
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - On Behalf of Diagnosis Group and Systemic Therapy Group of TLCA
- Department of Gastroenterology and Hepatology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan, Taiwan
- College of Medicine, Chang Gung University, Taoyuan, Taiwan
- Center for Digestive Medicine, Department of Internal Medicine, China Medical University Hospital, Taichung, Taiwan
- School of Medicine, China Medical University, Taichung, Taiwan
| |
Collapse
|
26
|
Li H, Yang B, Wang C, Li B, Han L, Jiang Y, Song Y, Wen L, Rao M, Zhang J, Li X, He K, Han Y. Construction of an interpretable model for predicting survival outcomes in patients with middle to advanced hepatocellular carcinoma (≥5 cm) using lasso-cox regression. Front Pharmacol 2024; 15:1452201. [PMID: 39372198 PMCID: PMC11450703 DOI: 10.3389/fphar.2024.1452201] [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: 06/20/2024] [Accepted: 09/04/2024] [Indexed: 10/08/2024] Open
Abstract
Background In this retrospective study, we aimed to identify key risk factors and establish an interpretable model for HCC with a diameter ≥ 5 cm using Lasso regression for effective risk stratification and clinical decision-making. Methods In this study, 843 patients with advanced hepatocellular carcinoma (HCC) and tumor diameter ≥ 5 cm were included. Using Lasso regression to screen multiple characteristic variables, cox proportional hazard regression and random survival forest models (RSF) were established. By comparing the area under the curve (AUC), the optimal model was selected. The model was visualized, and the order of interpretable importance was determined. Finally, risk stratification was established to identify patients at high risk. Result Lasso regression identified 8 factors as characteristic risk factors. Subsequent analysis revealed that the lasso-cox model had AUC values of 0.773, 0.758, and 0.799, while the lasso-RSF model had AUC values of 0.734, 0.695, and 0.741, respectively. Based on these results, the lasso-cox model was chosen as the superior model. Interpretability assessments using SHAP values indicated that the most significant characteristic risk factors, in descending order of importance, were tumor number, BCLC stage, alkaline phosphatase (ALP), ascites, albumin (ALB), and aspartate aminotransferase (AST). Additionally, through risk score stratification and subgroup analysis, it was observed that the median OS of the low-risk group was significantly better than that of the middle- and high-risk groups. Conclusion We have developed an interpretable predictive model for middle and late HCC with tumor diameter ≥ 5 cm using lasso-cox regression analysis. This model demonstrates excellent prediction performance and can be utilized for risk stratification.
Collapse
Affiliation(s)
- Han Li
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Bo Yang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Chenjie Wang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Bo Li
- Department of General Surgery (Hepatobiliary Surgery), The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Lei Han
- Department of Oncology, Affiliated Hospital of Jining Medical University, Jining, China
| | - Yi Jiang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yanqiong Song
- Sichuan Cancer Hospital & Institute, Sichuan Cancer Center, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lianbin Wen
- Sichuan Provincial People’s Hospital, Chengdu, China
| | - Mingyue Rao
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jianwen Zhang
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xueting Li
- Department of Oncology, 363 Hospital, Chengdu, China
| | - Kun He
- Clinical Medical College, Southwest Medical University, Luzhou, China
| | - Yunwei Han
- Department of Oncology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
27
|
Liu Z, Yuan H, Suo C, Zhao R, Jin L, Zhang X, Zhang T, Chen X. Point-based risk score for the risk stratification and prediction of hepatocellular carcinoma: a population-based random survival forest modeling study. EClinicalMedicine 2024; 75:102796. [PMID: 39263676 PMCID: PMC11388332 DOI: 10.1016/j.eclinm.2024.102796] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/03/2024] [Accepted: 08/06/2024] [Indexed: 09/13/2024] Open
Abstract
Background The precise associations between common clinical biomarkers and hepatocellular carcinoma (HCC) risk remain unclear but hold valuable insights for HCC risk stratification and prediction. Methods We examined the linear and nonlinear associations between the baseline levels of 32 circulating biomarkers and HCC risk in the England cohort of UK Biobank (UKBB) (n = 397,702). The participants were enrolled between 2006 and 2010 and followed up to 31st October 2022. The primary outcome is incident HCC cases. We then employed random survival forests (RSF) to select the top ten most informative biomarkers, considering their association with HCC, and developed a point-based risk score to predict HCC. The performance of the risk score was evaluated in three validation sets including UKBB Scotland and Wales cohort (n = 52,721), UKBB non-White-British cohort (n = 29,315), and the Taizhou Longitudinal Study in China (n = 17,269). Findings Twenty-five biomarkers were significantly associated with HCC risk, either linearly or nonlinearly. Based on the RSF model selected biomarkers, our point-based risk score showed a concordance index of 0.866 in the England cohort and varied between 0.814 and 0.849 in the three validation sets. HCC incidence rates ranged from 0.95 to 30.82 per 100,000 from the lowest to the highest quintiles of the risk score in the England cohort. Individuals in the highest risk quintile had a 32-73 times greater risk of HCC compared to those in the lowest quintile. Moreover, over 70% of HCC cases were detected in individuals within the top risk score quintile across all cohorts. Interpretation Our simple risk score enables the identification of high-risk individuals of HCC in the general population. However, including some biomarkers, such as insulin-like growth factor 1, not routinely measured in clinical practice may increase the model's complexity, highlighting the need for more accessible biomarkers that can maintain or improve the predictive accuracy of the risk score. Funding This work was supported by the National Natural Science Foundation of China (grant numbers: 82204125) and the Science and Technology Support Program of Taizhou (TS202224).
Collapse
Affiliation(s)
- Zhenqiu Liu
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Huangbo Yuan
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Chen Suo
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Renjia Zhao
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Li Jin
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
| | - Xuehong Zhang
- Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
- Yale University School of Nursing, Orange, CT, USA
| | - Tiejun Zhang
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- Key Laboratory of Public Health Safety, Fudan University, Ministry of Education, Shanghai, China
- Department of Epidemiology, School of Public Health, Fudan University, Shanghai, China
| | - Xingdong Chen
- State Key Laboratory of Genetic Engineering, Human Phenome Institute, and School of Life Sciences, Fudan University, Shanghai, China
- Fudan University Taizhou Institute of Health Sciences, Taizhou, China
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, China
| |
Collapse
|
28
|
Sanchez JI, Fontillas AC, Kwan SY, Sanchez CI, Calderone TL, Lee JL, Elsaiey A, Cleere DW, Wei P, Vierling JM, Victor DW, Beretta L. Metabolomics biomarkers of hepatocellular carcinoma in a prospective cohort of patients with cirrhosis. JHEP Rep 2024; 6:101119. [PMID: 39139459 PMCID: PMC11321296 DOI: 10.1016/j.jhepr.2024.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 08/15/2024] Open
Abstract
Background & Aims The effectiveness of surveillance for hepatocellular carcinoma (HCC) in patients with cirrhosis is limited, due to inadequate risk stratification and suboptimal performance of current screening modalities. Methods We developed a multicenter prospective cohort of patients with cirrhosis undergoing surveillance with MRI and applied global untargeted metabolomics to 612 longitudinal serum samples from 203 patients. Among them, 37 developed HCC during follow-up. Results We identified 150 metabolites with significant abundance changes in samples collected prior to HCC (Cases) compared to samples from patients who did not develop HCC (Controls). Tauro-conjugated bile acids and gamma-glutamyl amino acids were increased, while acyl-cholines and deoxycholate derivatives were decreased. Seven amino acids including serine and alanine had strong associations with HCC risk, while strong protective effects were observed for N-acetylglycine and glycerophosphorylcholine. Machine learning using the 150 metabolites, age, gender, and PNPLA3 and TMS6SF2 single nucleotide polymorphisms, identified 15 variables giving optimal performance. Among them, N-acetylglycine had the highest AUC in discriminating Cases and Controls. When restricting Cases to samples collected within 1 year prior to HCC (Cases-12M), additional metabolites including microbiota-derived metabolites were identified. The combination of the top six variables identified by machine learning (alpha-fetoprotein, 6-bromotryptophan, N-acetylglycine, salicyluric glucuronide, testosterone sulfate and age) had good performance in discriminating Cases-12M from Controls (AUC 0.88, 95% CI 0.83-0.93). Finally, 23 metabolites distinguished Cases with LI-RADS-3 lesions from Controls with LI-RADS-3 lesions, with reduced abundance of acyl-cholines and glycerophosphorylcholine-related lysophospholipids in Cases. Conclusions This study identified N-acetylglycine, amino acids, bile acids and choline-derived metabolites as biomarkers of HCC risk, and microbiota-derived metabolites as contributors to HCC development. Impact and implications The effectiveness of surveillance for hepatocellular carcinoma (HCC) in patients with cirrhosis is limited. There is an urgent need for improvement in risk stratification and new screening modalities, particularly blood biomarkers. Longitudinal collection of paired blood samples and MRI images from patients with cirrhosis is particularly valuable in assessing how early blood and imaging markers become positive during the period when lesions are observed to obtain a diagnosis of HCC. We generated a multicenter prospective cohort of patients with cirrhosis under surveillance with contrast MRI, applied untargeted metabolomics on 612 serum samples from 203 patients and identified metabolites associated with risk of HCC development. Such biomarkers may significantly improve early-stage HCC detection for patients with cirrhosis undergoing HCC surveillance, a critical step to increasing curative treatment opportunities and reducing mortality.
Collapse
Affiliation(s)
- Jessica I. Sanchez
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antoine C. Fontillas
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suet-Ying Kwan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caren I. Sanchez
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tiffany L. Calderone
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jana L. Lee
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Ahmed Elsaiey
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Darrel W. Cleere
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M. Vierling
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - David W. Victor
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
29
|
Qurashi M, Sharma R. Improving hepatocellular carcinoma surveillance in the United Kingdom: challenges and solutions. THE LANCET REGIONAL HEALTH. EUROPE 2024; 43:100963. [PMID: 38946891 PMCID: PMC11211981 DOI: 10.1016/j.lanepe.2024.100963] [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: 03/17/2024] [Revised: 05/27/2024] [Accepted: 05/30/2024] [Indexed: 07/02/2024]
Abstract
Hepatocellular carcinoma (HCC) prognosis remains dismal due to late-stage diagnosis; surveillance has been demonstrated to increase early diagnosis rates and receipt of curative treatment. Acknowledging limitations in the evidence base for HCC surveillance, international professional bodies reiterate the recommendation for biannual HCC surveillance and NHS England supports measures aimed to increase surveillance uptake. The current ad hoc provision of HCC surveillance is prone to failures, evident by low surveillance uptake and high numbers of patients being diagnosed outside of surveillance. We discuss challenges related to HCC surveillance in the UK and potential solutions to addressing them. We highlight the requirements of a consistent and effective national surveillance process, and suggest pathways on how this can be achieved.
Collapse
Affiliation(s)
- Maria Qurashi
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| | - Rohini Sharma
- Department of Surgery & Cancer, Imperial College London, Hammersmith Hospital, Du Cane Road, London W12 0HS, UK
| |
Collapse
|
30
|
Chen XF, Wu HJ, Li T, Liu JB, Zhou WJ, Guo Q. Establishment and validation of a predictive model for peripherally inserted central catheter-related thrombosis in patients with liver cancer. World J Gastrointest Surg 2024; 16:2221-2231. [PMID: 39087116 PMCID: PMC11287695 DOI: 10.4240/wjgs.v16.i7.2221] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 06/03/2024] [Accepted: 06/18/2024] [Indexed: 07/22/2024] Open
Abstract
BACKGROUND Peripherally inserted central catheters (PICCs) are commonly used in hospitalized patients with liver cancer for the administration of chemotherapy, nutrition, and other medications. However, PICC-related thrombosis is a serious complication that can lead to morbidity and mortality in this patient population. Several risk factors have been identified for the development of PICC-related thrombosis, including cancer type, stage, comorbidities, and catheter characteristics. Understanding these risk factors and developing a predictive model can help healthcare providers identify high-risk patients and implement preventive measures to reduce the incidence of thrombosis. AIM To analyze the influencing factors of PICC-related thrombosis in hospitalized patients with liver cancer, construct a predictive model, and validate it. METHODS Clinical data of hospitalized patients with liver cancer admitted from January 2020 to December 2023 were collected. Thirty-five cases of PICC-related thrombosis in hospitalized patients with liver cancer were collected, and 220 patients who underwent PICC placement during the same period but did not develop PICC-related thrombosis were randomly selected as controls. A total of 255 samples were collected and used as the training set, and 77 cases were collected as the validation set in a 7:3 ratio. General patient information, case data, catheterization data, coagulation indicators, and Autar Thrombosis Risk Assessment Scale scores were analyzed. Univariate and multivariate unconditional logistic regression analyses were performed on relevant factors, and the value of combined indicators in predicting PICC-related thrombosis in hospitalized patients with liver cancer was evaluated using receiver operating characteristic (ROC) curve analysis. RESULTS Univariate analysis showed statistically significant differences (P < 0.05) in age, sex, Karnofsky performance status score (KPS), bedridden time, activities of daily living impairment, parenteral nutrition, catheter duration, distant metastasis, and bone marrow suppression between the thrombosis group and the non-thrombosis group. Other aspects had no statistically significant differences (P > 0.05). Multivariate regression analysis showed that age ≥ 60 years, KPS score ≤ 50 points, parenteral nutrition, stage III to IV, distant metastasis, bone marrow suppression, and activities of daily living impairment were independent risk factors for PICC-related thrombosis in hospitalized patients with liver cancer (P < 0.05). Catheter duration of 1-6 months and catheter duration > 6 months were protective factors for PICC-related thrombosis (P < 0.05). The predictive model for PICC-related thrombosis was obtained as follows: P predictive probability = [exp (Logit P)]/[1 + exp (Logit P)], where Logit P = age × 1.907 + KPS score × 2.045 + parenteral nutrition × 9.467 + catheter duration × 0.506 + tumor-node-metastasis (TNM) staging × 2.844 + distant metastasis × 2.065 + bone marrow suppression × 2.082 + activities of daily living impairment × 13.926. ROC curve analysis showed an area under the curve (AUC) of 0.827 (95%CI: 0.724-0.929, P < 0.001), with a corresponding optimal cut-off value of 0.612, sensitivity of 0.755, and specificity of 0.857. Calibration curve analysis showed good consistency between the predicted occurrence of PICC-related thrombosis and actual occurrence (P > 0.05). ROC analysis showed AUCs of 0.888 and 0.729 for the training and validation sets, respectively. CONCLUSION Age, KPS score, parenteral nutrition, TNM staging, distant metastasis, bone marrow suppression, and activities of daily living impairment are independent risk factors for PICC-related thrombosis in hospitalized patients with liver cancer, while catheter duration is a protective factor for the disease. The predictive model has an AUC of 0.827, indicating high predictive accuracy and clinical value.
Collapse
Affiliation(s)
- Xiao-Fei Chen
- Department of Thyroid Surgery, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
- The Second Department of General Surgery, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu 611730, Sichuan Province, China
| | - Hao-Jun Wu
- Department of Biliary Surgery, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Tang Li
- Department of Hepatobiliary and Pancreatic Surgery, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu 611730, Sichuan Province, China
| | - Jia-Bin Liu
- Department of Thyroid Surgery, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| | - Wen-Jie Zhou
- The Second Department of General Surgery, Chengdu Shangjinnanfu Hospital, West China Hospital of Sichuan University, Chengdu 611730, Sichuan Province, China
| | - Qiang Guo
- Department of Vascular Surgery, West China Hospital of Sichuan University, Chengdu 610041, Sichuan Province, China
| |
Collapse
|
31
|
Mendiratta-Lala M, Fetzer D, Kamaya A, Parikh ND, Singal AG. The Future Role of Abdominal US in Hepatocellular Carcinoma Surveillance. Radiology 2024; 311:e232624. [PMID: 38742973 PMCID: PMC11140528 DOI: 10.1148/radiol.232624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 05/16/2024]
Abstract
Abdominal US is currently the best-validated surveillance strategy for hepatocellular carcinoma (HCC) in at-risk patients. It is the only modality shown to have completed all five phases of validation and can achieve high sensitivity and specificity for HCC detection, especially when conducted by expert sonographers in high-volume centers. However, US also has limitations, including operator dependency and varying sensitivity in clinical practice. Further, the sensitivity of US for early-stage HCC detection is lower in patients with obesity or nonviral liver disease, increasingly common populations undergoing surveillance. Imaging-based and blood-based surveillance strategies, including abbreviated MRI and biomarker panels, may overcome some limitations of US-based surveillance. Both strategies have promising test performance in phase II and phase III biomarker studies and are undergoing prospective validation. Considering the variation in HCC risk and test performance between patients, there will likely be a shift away from a one-size-fits-all approach and toward precision screening, in which the "best" test is selected based on individual patient characteristics. In this upcoming era of precision HCC screening among patients with cirrhosis, US will likely continue to have an important, albeit reduced, surveillance role.
Collapse
Affiliation(s)
| | | | - Aya Kamaya
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| | - Neehar D. Parikh
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| | - Amit G. Singal
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| |
Collapse
|
32
|
Nahon P, Layese R, Ganne-Carrié N, Moins C, N'Kontchou G, Chaffaut C, Ronot M, Audureau E, Durand-Zaleski I, Natella PA. The clinical and financial burden of nonhepatocellular carcinoma focal lesions detected during the surveillance of patients with cirrhosis. Hepatology 2024; 79:813-828. [PMID: 37774387 DOI: 10.1097/hep.0000000000000615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 09/01/2023] [Indexed: 10/01/2023]
Abstract
BACKGROUND AND AIMS HCC surveillance is challenged by the detection of hepatic focal lesions (HFLs) of other types. This study aimed to describe the incidence, characteristics, outcomes, and costs of non-HCC HFL detected during surveillance. APPROACH AND RESULTS We retrospectively analyzed nonstandardized workup performed in French patients included in HCC surveillance programs recruited in 57 French tertiary centers (ANRS CirVir and CIRRAL cohorts, HCC 2000 trial). The overall cost of workup was evaluated, with an estimation of an average cost per patient for the entire population and per lesion detected. A total of 3295 patients were followed up for 59.8 months, 391 (11.9%) patients developed HCCs (5-year incidence: 12.1%), and 633 (19.2%) developed non-HCC HFLs (5-year incidence: 21.8%). Characterization of non-HCC HFL required a median additional of 0.7 exams per year. A total of 11.8% of non-HCC HFLs were not confirmed on recall procedures, and 19.6% of non-HCC HFLs remained undetermined. A definite diagnosis of benign liver lesions was made in 65.1%, and malignant tumors were diagnosed in 3.5%. The survival of patients with benign or undetermined non-HCC HFL was similar to that of patients who never developed any HFL (5-year survival 92% vs. 88%, p = 0.07). The average cost of the diagnostic workup was 1087€ for non-HCC HFL and €1572 for HCC. CONCLUSIONS Non-HCC HFLs are frequently detected in patients with cirrhosis, and do not impact prognosis, but trigger substantial costs. This burden must be considered in cost-effectiveness analyses of future personalized surveillance strategies.
Collapse
Affiliation(s)
- Pierre Nahon
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Liver Department, Bobigny; Université Sorbonne Paris Nord, Bobigny, France
- Inserm, UMR-1138 Functional Genomics of Solid Tumors department, Centre de recherche des Cordeliers, Université de Paris, Paris, France
| | - Richard Layese
- Univ Paris Est Créteil, INSERM, IMRB, Equipe CEpiA (Clinical Epidemiology and Ageing), Unité de Recherche Clinique (URC Mondor), Public health department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Nathalie Ganne-Carrié
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Liver Department, Bobigny; Université Sorbonne Paris Nord, Bobigny, France
- Inserm, UMR-1138 Functional Genomics of Solid Tumors department, Centre de recherche des Cordeliers, Université de Paris, Paris, France
| | - Cécile Moins
- Clinical Research Department, ANRS | Emerging Infectious Diseases, Paris, France
| | - Gisèle N'Kontchou
- AP-HP, Hôpitaux Universitaires Paris Seine Saint-Denis, Liver Department, Bobigny; Université Sorbonne Paris Nord, Bobigny, France
- Inserm, UMR-1138 Functional Genomics of Solid Tumors department, Centre de recherche des Cordeliers, Université de Paris, Paris, France
| | - Cendrine Chaffaut
- SBIM, APHP, Hôpital Saint-Louis, Paris, Inserm, UMR-1153, ECSTRA department, Paris, France
| | - Maxime Ronot
- APHP, Hôpital Beaujon, Radiology department, Hôpital Beaujon, APHP. Nord, Clichy-Sous-Bois, & Université Paris Cité, Paris, France
| | - Etienne Audureau
- Univ Paris Est Créteil, INSERM, IMRB, Equipe CEpiA (Clinical Epidemiology and Ageing), Unité de Recherche Clinique (URC Mondor), Public health department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Créteil, France
| | - Isabelle Durand-Zaleski
- Université de Paris, CRESS, INSERM, INRA, URCECo department, AP-HP, Hôpital de l'Hôtel Dieu, Paris, France
| | - Pierre-André Natella
- Univ Paris Est Créteil, INSERM, IMRB, Equipe CEpiA (Clinical Epidemiology and Ageing), Unité de Recherche Clinique (URC Mondor), Public health department, Assistance Publique Hôpitaux de Paris (AP-HP), Hôpitaux Universitaires Henri Mondor, Créteil, France
| |
Collapse
|
33
|
Kim GA, Park Y, Oh SJ, Jung J, Han S, Chang HS, Park SW, Kim TH, Park HW, Choe J, Kim J, Lee HC. A risk prediction model for hepatocellular carcinoma in non-alcoholic fatty liver disease without cirrhosis: HCC prediction for non-cirrhotic NAFLD. Liver Int 2024; 44:738-748. [PMID: 38110797 DOI: 10.1111/liv.15819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 11/21/2023] [Accepted: 12/03/2023] [Indexed: 12/20/2023]
Abstract
BACKGROUND & AIMS Although non-alcoholic fatty liver disease (NAFLD) is becoming a leading cause of hepatocellular carcinoma (HCC), HCC risk in non-cirrhotic NAFLD received little attention. We aimed to develop and validate an HCC risk prediction model for non-cirrhotic NAFLD. METHODS A nationwide cohort of non-cirrhotic NAFLD patients in Korea was recruited to develop a risk prediction model and validate it internally (n = 409 088). A model using a simplified point system was developed by Cox proportional hazard model. K-fold cross-validation assessed the accuracy, discrimination and calibration. The model was validated externally using a hospital cohort from Asan Medical Center (n = 8721). RESULTS An 11-point HCC risk prediction model for non-cirrhotic NAFLD was developed using six independent factors of age, sex, diabetes, obesity, serum alanine aminotransferase level and gamma-glutamyl transferase level (c-index 0.75). The average area under receiver operating curves (AUROCs) of the model was 0.72 at 5 years and 0.75 at 10 years. In the external validation cohort, the AUROCs were 0.79 [95% confidence interval [CI], 0.59-0.95] at 5 years and 0.84 (95% CI, 0.73-0.94) at 10 years. The calibration plots showed the expected risks corresponded well with the observed risks. Risk stratification categorized patients into the low (score 0-6), moderate (7, 8) and high (9-11; estimated incidence rate >0.2%/year) risk groups. CONCLUSIONS A novel HCC risk prediction model for non-cirrhotic NAFLD patients was developed and validated with fair performance. The model is expected to serve as a simple and reliable tool to assess HCC risk and assist precision screening of HCC.
Collapse
Affiliation(s)
- Gi-Ae Kim
- Department of Internal Medicine, College of Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Yewan Park
- Department of Internal Medicine, College of Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Shin Ju Oh
- Department of Internal Medicine, College of Medicine, Kyung Hee University Hospital, Kyung Hee University, Seoul, Republic of Korea
| | - Jooyi Jung
- Department of Biostatistics, Korea University, Seoul, Republic of Korea
| | - Seungbong Han
- Department of Biostatistics, Korea University, Seoul, Republic of Korea
| | - Hye-Sook Chang
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Sung Won Park
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Tae Hyup Kim
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Hye Won Park
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jaewon Choe
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Jaeil Kim
- Health Screening and Promotion Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Han Chu Lee
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| |
Collapse
|
34
|
Daher D, Seif El Dahan K, Cano A, Gonzales M, Ransom C, Jaurez E, Carranza O, Quirk L, Morgan T, Gopal P, Patel MS, Lieber S, Louissaint J, Cotter TG, VanWagner LB, Yang JD, Parikh ND, Yopp A, Rich NE, Singal AG. Hepatocellular Carcinoma Surveillance Patterns and Outcomes in Patients With Cirrhosis. Clin Gastroenterol Hepatol 2024; 22:295-304.e2. [PMID: 37573986 PMCID: PMC11415236 DOI: 10.1016/j.cgh.2023.08.003] [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: 05/19/2023] [Revised: 07/18/2023] [Accepted: 08/02/2023] [Indexed: 08/15/2023]
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) surveillance is associated with improved early detection and reduced mortality, although practice patterns and effectiveness vary in clinical practice. We aimed to characterize HCC surveillance patterns in a large, diverse cohort of patients with HCC. METHODS We conducted a retrospective cohort study of patients diagnosed with HCC between January 2008 and December 2022 at 2 large US health systems. We recorded imaging receipt in the year before HCC diagnosis: ultrasound plus α-fetoprotein (AFP), ultrasound alone, multiphasic contrast-enhanced computed tomography (CT)/magnetic resonance imaging (MRI), and no liver imaging. We used multivariable logistic and Cox regression analysis to compare early tumor detection, curative treatment receipt, and overall survival between surveillance strategies. RESULTS Among 2028 patients with HCC (46.7% Barcelona Clinic Liver Cancer stage A), 703 (34.7%) had ultrasound plus AFP, 293 (14.5%) had ultrasound alone, 326 (16.1%) had multiphasic CT/MRI, and 706 (34.8%) had no imaging in the year before HCC diagnosis. Over the study period, proportions without imaging were stable, whereas use of CT/MRI increased. Compared with no imaging, CT/MRI and ultrasound plus AFP, but not ultrasound alone, were associated with early stage HCC detection and curative treatment. Compared with ultrasound alone, CT/MRI and ultrasound plus AFP were associated with increased early stage detection. CONCLUSIONS HCC surveillance patterns vary in clinical practice and are associated with differing clinical outcomes. While awaiting data to determine if CT or MRI surveillance can be performed in a cost-effective manner in selected patients, AFP has a complementary role to ultrasound-based surveillance, supporting its adoption in practice guidelines.
Collapse
Affiliation(s)
- Darine Daher
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Karim Seif El Dahan
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Alva Cano
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Michael Gonzales
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Crystal Ransom
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Erik Jaurez
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Osiris Carranza
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Lisa Quirk
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Todd Morgan
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Purva Gopal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Madhukar S Patel
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Sarah Lieber
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Jeremy Louissaint
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Thomas G Cotter
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Lisa B VanWagner
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Ju Dong Yang
- Department of Internal Medicine, Cedars Sinai Medical Center, Los Angeles, California
| | - Neehar D Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan
| | - Adam Yopp
- Department of Surgery, UT Southwestern Medical Center, Dallas, Texas
| | - Nicole E Rich
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas
| | - Amit G Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
35
|
Jones-Pauley M, Victor DW, Kodali S. Pushing the limits of treatment for hepatocellular carcinoma. Curr Opin Organ Transplant 2024; 29:3-9. [PMID: 38032256 DOI: 10.1097/mot.0000000000001123] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2023]
Abstract
PURPOSE OF REVIEW We review existing and newer strategies for treatment and surveillance of hepatocellular carcinoma (HCC) both pre and postliver transplantation. SUMMARY HCC is rising in incidence and patients are often diagnosed at later stages. Consequently, there is a need for treatment strategies which include collaboration of multiple specialties. Combinations of locoregional, systemic, and surgical therapies are yielding better postliver transplantation (post-LT) outcomes for patients with HCC than previously seen. Tumor biology (tumor size, number, location, serum markers, response to therapy) can help identify patients who are at high risk for HCC recurrence posttransplantation and may expand transplant eligibility for some patients.
Collapse
Affiliation(s)
| | - David W Victor
- Division of Gastroenterology and Hepatology, Department of Medicine
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston Methodist Hospital, Houston, Texas
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Sudha Kodali
- Division of Gastroenterology and Hepatology, Department of Medicine
- Sherrie and Alan Conover Center for Liver Disease and Transplantation, Houston Methodist Hospital, Houston, Texas
- Department of Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
36
|
Singal AG, Kanwal F, Llovet JM. Global trends in hepatocellular carcinoma epidemiology: implications for screening, prevention and therapy. Nat Rev Clin Oncol 2023; 20:864-884. [PMID: 37884736 DOI: 10.1038/s41571-023-00825-3] [Citation(s) in RCA: 280] [Impact Index Per Article: 140.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2023] [Indexed: 10/28/2023]
Abstract
Hepatocellular carcinoma (HCC) mortality rates are increasing globally, and particularly in the Western world. Cirrhosis remains the predominant risk factor for HCC. However, epidemiological shifts in the incidence of HCC from patients with virus-related liver disease to those with non-viral aetiologies, including alcohol-associated and metabolic dysfunction-associated steatotic liver disease, have important implications for prevention, surveillance and treatment. Hepatitis B vaccination and antiviral therapy for hepatitis B and C are effective for primary prevention of virus-related HCCs, but chemoprevention strategies for non-viral liver disease remain an unmet need. Emerging data suggest associations between aspirin, statins, metformin and coffee and reduced HCC incidence, although none has been proved to be causally related. Secondary prevention of HCC via semi-annual surveillance is associated with improvements in early detection and thus reduced mortality; however, current tools, including abdominal ultrasonography, have suboptimal sensitivity for the detection of early stage HCC, particularly in patients with obesity and/or non-viral liver disease. Promising blood-based or imaging-based surveillance strategies are emerging, although these approaches require further validation before adoption in clinical practice. In the interim, efforts should be focused on maximizing use of the existing surveillance tools given their prevalent underuse globally. Remarkable advances have been made in the treatment of HCC, including expanded eligibility for surgical therapies, improved patient selection for locoregional treatments and increased systemic treatment options, including immune-checkpoint inhibitors. In this Review, we discuss trends in the epidemiology of HCC and their implications for screening, prevention and therapy.
Collapse
Affiliation(s)
- Amit G Singal
- Division of Digestive and Liver Diseases, Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA.
| | - Fasiha Kanwal
- Section of Gastroenterology and Hepatology and Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- Section of Health Services Research, Department of Medicine, Baylor College of Medicine, Houston, TX, USA
- VA Health Services Research & Development Center for Innovations in Quality, Effectiveness, and Safety (IQuESt), Houston, TX, USA
- Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Josep M Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, New York, NY, USA
- Translational Research in Hepatic Oncology, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clinic, University of Barcelona, Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| |
Collapse
|
37
|
Elbahrawy A, Atalla H, Mahmoud AA, Eliwa A, Alsawak A, Alboraie M, Madian A, Alashker A, Mostafa S, Alwassief A, Aly HH. Prediction and surveillance of de novo HCC in patients with compensated advanced chronic liver disease after hepatitis C virus eradication with direct antiviral agents. FRONTIERS IN VIROLOGY 2023; 3. [DOI: 10.3389/fviro.2023.1227317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
Abstract
The risk of hepatocellular carcinoma (HCC) diminishes in patients with hepatitis C virus (HCV)-related advanced chronic liver disease after virological cure. However, despite viral clearance, HCV-induced epigenetic alterations, immune dysregulations, and hepatic parenchymal injuries remain, contributing to de novo HCC occurrence. While HCC incidence is low (0.45 – 0.5%) in patients with advanced fibrosis (F3), the presence of liver cirrhosis and clinically significant portal hypertension increases the HCC risk. The cost-effectiveness of lifelong HCC surveillance in patients with compensated advanced chronic liver disease (cACLD) has sparked debate, raising questions about the most reliable noninvasive tests and stratification models for predicting HCC in patients with sustained virological response (SVR). Furthermore, identifying cACLD patients who may not require long-term HCC surveillance after SVR remains crucial. Several HCC risk stratification scores have been suggested for patients with cACLD, and emerging evidence supports individualized care based on personalized risk assessments. This review focuses on revising the pretreatment and posttreatment predictors of HCC, as well as the indications for HCC surveillance in cACLD patients treated with direct-acting antivirals.
Collapse
|
38
|
Pang M, Sun X, He T, Liang H, Yang H, Chen J. Development of a prognostic model based on anoikis-related genes for predicting clinical prognosis and immunotherapy of hepatocellular carcinoma. Aging (Albany NY) 2023; 15:10253-10271. [PMID: 37787988 PMCID: PMC10599733 DOI: 10.18632/aging.205073] [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: 04/21/2023] [Accepted: 06/19/2023] [Indexed: 10/04/2023]
Abstract
Hepatocellular Carcinoma (HCC) is the predominant cause of cancer-related mortality worldwide. The majority of HCC patients are diagnosed at advanced stages of the disease, with a high likelihood of metastasis and unfavorable prognosis. Anoikis resistance is a crucial factor contributing to tumor invasion and metastasis, although its specific role in HCC remains unclear. Based on the results of univariate Cox regression and least absolute shrink-age and selection operator (LASSO) analysis, a subset of anoikis-related genes (ARGs) significantly associated with overall survival (OS) was identified. A multivariate Cox regression analysis subsequently identified PDK4, STK11, and TFDP1 as three prognostic ARGs, which were then used to establish a prognostic risk model. Differences in OS caused by risk stratification in HCC patients were demonstrated. The nomogram analysis indicated that the ARGs prognostic signature served as an independent prognostic predictor. In vitro experiments further confirmed the abnormal expression of selected ARGs in HCC. The association between risk scores and OS was further examined through Kaplan-Meier analysis, CIBERSORT analysis, and single-sample gene set enrichment analysis (ssGSEA). This study is a pioneering effort to integrate multiple ARGs and establish a risk-predictive model, providing a unique perspective for the development of personalized and precise therapeutic strategies for HCC.
Collapse
Affiliation(s)
- Mu Pang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Xizhe Sun
- Research Center for Drug Safety Evaluation of Hainan, Hainan Medical University, Haikou, Hainan 571199, China
| | - Ting He
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Huichao Liang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Hao Yang
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| | - Jun Chen
- The Fourth Clinical Medical College of Guangzhou University of Chinese Medicine, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong 518000, China
| |
Collapse
|