1
|
Jaber F, El-Serag HB. HES V2.0 surpasses GALAD for HCC detection: a review of multi-dimensional biomarker scores and studies. Hepat Oncol 2025; 12:2494446. [PMID: 40308043 PMCID: PMC12051611 DOI: 10.1080/20450923.2025.2494446] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
This was a narrative review of select studies published through September of 2024. We review the shift toward multi-dimensional scores such as HCC early detection screening (HES), GALAD, ASAP, and mt-HBT represents a significant advancement in biomarker research for hepatocellular carcinoma (HCC) detection. Unlike single biomarker approaches, these scores integrate various clinical and biochemical factors to enhance predictive accuracy by reflecting different complementary aspects of disease progression and HCC oncogenesis. Proper testing and validation of biomarker scores in phase 3 biomarker studies is essential before wide use can be recommended. We also review the comparative performance of biomarker scores in phase 3 studies. The new version of HES (HES V2.0) which includes AFP, AFP L3, DCP, and changes in their levels the past one year, if available, in addition to age, platelets, albumin, ALT and underlying liver disease etiology outperforms GALAD in detecting early-stage HCC with overall 6.7% higher sensitivity, and ASAP with 13.4%-18.0% higher sensitivity, both at fixed 90% specificity. HES V2.0 is a leading candidate biomarker score for prospective testing in clinical studies of early HCC detection.
Collapse
Affiliation(s)
- Fouad Jaber
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - Hashem B. El-Serag
- Department of Internal Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
2
|
Rodgers SK, Fetzer DT, Seow JH, McGillen K, Burrowes DP, Fung C, Udare AS, Wilson SR, Kamaya A. Optimizing US for HCC surveillance. Abdom Radiol (NY) 2025; 50:2453-2463. [PMID: 39585379 PMCID: PMC12069441 DOI: 10.1007/s00261-024-04631-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 10/02/2024] [Accepted: 10/04/2024] [Indexed: 11/26/2024]
Abstract
Ultrasound is the primary imaging modality used for surveillance of patients at risk for HCC. In 2017, the American College of Radiology Liver Imaging Reporting and Data Systems (ACR LI-RADS) introduced US LI-RADS to standardize the performance, interpretation, and reporting of US for HCC surveillance, with the algorithm recently updated as LI-RADS US Surveillance v2024. The American Association for the Study of Liver Diseases (AASLD) recommends reporting both the examination-level LI-RADS US Category as well as the US Visualization Score. The US Category conveys the overall findings of the exam and primarily determines follow up recommendations. The US Visualization Score conveys the expected sensitivity of the test and stratifies patients into appropriate surveillance pathways. One of the goals of routine surveillance is the detection of HCC at an early, potentially curable stage. Therefore, optimizing US technique is of critical importance. Increasing North American and worldwide utilization of LI-RADS US Surveillance, which includes technical recommendations, through education and outreach will undoubtedly benefit patients undergoing US HCC surveillance.
Collapse
Affiliation(s)
| | - David T Fetzer
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | | | | | | | | | | | | |
Collapse
|
3
|
Lin HY, Jeon AJ, Chen K, Lee CJM, Wu L, Chong SL, Anene-Nzelu CG, Foo RSY, Chow PKH. The epigenetic basis of hepatocellular carcinoma - mechanisms and potential directions for biomarkers and therapeutics. Br J Cancer 2025; 132:869-887. [PMID: 40057667 DOI: 10.1038/s41416-025-02969-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 01/23/2025] [Accepted: 02/20/2025] [Indexed: 05/17/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth leading cancer worldwide and has complex pathogenesis due to its heterogeneity, along with poor prognoses. Diagnosis is often late as current screening methods have limited sensitivity for early HCC. Moreover, current treatment regimens for intermediate-to-advanced HCC have high resistance rates, no robust predictive biomarkers, and limited survival benefits. A deeper understanding of the molecular biology of HCC may enhance tumor characterization and targeting of key carcinogenic signatures. The epigenetic landscape of HCC includes complex hallmarks of 1) global DNA hypomethylation of oncogenes and hypermethylation of tumor suppressors; 2) histone modifications, altering chromatin accessibility to upregulate oncogene expression, and/or suppress tumor suppressor gene expression; 3) genome-wide rearrangement of chromatin loops facilitating distal enhancer-promoter oncogenic interactions; and 4) RNA regulation via translational repression by microRNAs (miRNAs) and RNA modifications. Additionally, it is useful to consider etiology-specific epigenetic aberrancies, especially in viral hepatitis and metabolic dysfunction-associated steatotic liver disease (MASLD), which are the main risk factors of HCC. This article comprehensively explores the epigenetic signatures in HCC, highlighting their potential as biomarkers and therapeutic targets. Additionally, we examine how etiology-specific epigenetic patterns and the integration of epigenetic therapies with immunotherapy could advance personalized HCC treatment strategies.
Collapse
Affiliation(s)
- Hong-Yi Lin
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| | - Ah-Jung Jeon
- Department of Research and Development, Mirxes, Singapore, Singapore
| | - Kaina Chen
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore, Singapore
| | - Chang Jie Mick Lee
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore, Singapore
| | - Lingyan Wu
- Program in Translational and Clinical Research in Liver Cancer, National Cancer Centre Singapore, Singapore, Singapore
| | - Shay-Lee Chong
- Program in Translational and Clinical Research in Liver Cancer, National Cancer Centre Singapore, Singapore, Singapore
| | | | - Roger Sik-Yin Foo
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Cardiovascular Research Institute, National University Heart Centre, Singapore, Singapore
- Department of Cardiology, National University Heart Centre, Singapore, Singapore
| | - Pierce Kah-Hoe Chow
- Program in Translational and Clinical Research in Liver Cancer, National Cancer Centre Singapore, Singapore, Singapore.
- Department of Hepato-pancreato-biliary and Transplant Surgery, Division of Surgery and Surgical Oncology, Singapore General Hospital and National Cancer Centre Singapore, Singapore, Singapore.
- Surgery Academic Clinical Programme, Duke-NUS Medical School, Singapore, Singapore.
| |
Collapse
|
4
|
Fu S, Boers RG, Boers JB, van der Meeren PE, Helmijr J, de Weerd V, Doukas M, Jansen M, Hansen BE, de Wilde RF, Sprengers D, Gribnau J, Wilting SM, Debes JD, Boonstra A. Genome-Wide Methylation Sequencing to Identify DNA Methylation Markers for Early-stage Hepatocellular Carcinoma in Liver and Blood. J Exp Clin Cancer Res 2025; 44:144. [PMID: 40375278 DOI: 10.1186/s13046-025-03412-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Accepted: 05/07/2025] [Indexed: 05/18/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is associated with a poor 5-year survival mainly due to detection at late stages. Better non-invasive surveillance methods are needed to improve early detection and maximize survival. We performed a strict assessment of DNA methylation markers (DMMs) for HCC detection. METHODS A total of 385 samples from liver tissues and blood were analyzed. Genome-wide Methylated DNA sequencing (MeD-seq) was initially performed on 46 liver tissues, followed by the validation using quantitative methylation-specific PCR (qMSP) on 175 liver tissues. The selected DMMs with and without ASAP/GAAD score were further evaluated in 180 blood samples. Additionally, MeD-seq was performed to validate the results on blood. RESULTS MeD-seq revealed a substantial number of differentially methylated regions (DMRs) in HCC tissues compared to non-HCC controls. By qMSP, the top 5 DMMs demonstrated strong performance in distinguishing cirrhotic HCC from cirrhosis controls in tissue (AUC 0.842 to 0.957). However, evaluation of these DMMs in blood showed lower performance in early HCC detection compared to cirrhosis in both the training (sensitivity 26.7-43.3%, 81.3% specificity) and validation cohorts (sensitivity 16.2-43.2%, 85.7% specificity). The addition of DMMs to the ASAP/GAAD score only provided an additional 5.4% sensitivity in the validation cohort compared to the ASAP/GAAD score alone. These findings were confirmed using MeD-seq analysis in blood samples, which revealed no detectable DMRs between cirrhotic HCC and cirrhosis controls. Interestingly, DNA methylation patterns in blood of healthy individuals differed strongly from both groups (cirrhosis and cirrhotic HCC). CONCLUSION DNA methylation patterns in liver tissue were distinctly different between HCC and controls. In blood, DMMs contributed minimally to early-stage HCC detection compared to cirrhosis, whether used alone or in combination with the ASAP/GAAD score. It is likely that high baseline DNA methylation related to cirrhosis and possibly the low input of tumor-related DNA impacts the use of DMMs in early HCC detection in blood.
Collapse
Affiliation(s)
- Siyu Fu
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Ruben G Boers
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Joachim B Boers
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Pam E van der Meeren
- Department of Surgery, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Jean Helmijr
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Vanja de Weerd
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Michail Doukas
- Department of Pathology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Maurice Jansen
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Bettina E Hansen
- Toronto Centre for Liver Disease, University Health Network, University of Toronto, Toronto, Canada
- Department of Epidemiology, Biostatistics, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Roeland F de Wilde
- Department of Surgery, Erasmus MC Transplant Institute, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Dave Sprengers
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
| | - Joost Gribnau
- Department of Developmental Biology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - Saskia M Wilting
- Department of Medical Oncology, Erasmus University Medical Center, Rotterdam, The Netherlands
| | - José D Debes
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands
- Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Andre Boonstra
- Department of Gastroenterology and Hepatology, Erasmus University Medical Center, Wytemaweg 80, 3015 CN, Rotterdam, The Netherlands.
| |
Collapse
|
5
|
Teng W, Li H, Yang H, Chen Y, Xi L, Xin F, Zhang A, Yu L, Zheng L, Wang M, Bai J, Ke F, Wang Y, Sun F, Zhang H, Wu L, Liu J. Discovery and validation of a novel dual-target blood test for the detection of hepatocellular carcinoma across stages from cirrhosis. BMC Med 2025; 23:278. [PMID: 40350453 PMCID: PMC12067762 DOI: 10.1186/s12916-025-04115-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 05/01/2025] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is one of the most common cancers. Early detection of HCC helps improve the patients' 5-year survival rate. Our goal was to identify superior methylation biomarkers to develop a methylation-specific quantitative PCR (MS‒qPCR) assay. METHODS A five-phase case-control study identified HCC methylation biomarkers via capture sequencing, TCGA/RNA-seq filtering, technical (MS-qPCR/Sanger) and biological (quadruplex MS-qPCR) validation. Methylated biomarkers were selected based on differential methylation expression using a tissue discovery cohort (43 HCC, 32 normal) and validated in plasma validation cohorts (Phase 1: 53 HCC, 52 cirrhosis, 20 benign, 50 healthy; Phase 2: 67 HCC, 81 cirrhosis). Then, the final assay's HCC detection performance was compared with existing blood-based surveillance methods. RESULTS Two methylated genes, OSR2 and TSPYL5, and a novel internal reference gene, SDF4, were identified and developed into an MS‒qPCR assay named Qliver. Qliver had an AUC of 0.955 (95% CI: 0.924-0.987) for distinguishing HCC patients from non-HCC patients in the Phase 1 plasma cohort, with a sensitivity of 88.68% (95% CI: 76.97%-95.73%) and a specificity of 89.34% (95% CI: 82.47%-94.20%), and 0.958 (95% CI: 0.927-0.989) for distinguishing HCC patients from cirrhosis patients in the Phase 2 plasma cohort, with a sensitivity of 88.06% (95% CI: 77.82%-94.70%) and a specificity of 92.59% (95% CI: 84.57%-97.23%). For the Phase 1 plus Plasma 2 cohort, Qliver had an AUC of at least 0.958 for detecting HCC in healthy individuals, cirrhosis patients and patients with benign liver diseases, which was superior to that of the GALAD score (AUC: 0.777 to 0.849). For BCLC stage 0 and A HCC patients, the sensitivity of Qliver ranged from 62.50% (95% CI: 24.49%-91.48%) to 72.73% (39.03%-93.98%), with a specificity of 90%. Overall, Qliver was superior to the AFP, AFP-L3, DCP and the GALAD score in terms of cirrhosis history, tumor stage, tumor size and tumor count. CONCLUSIONS Qliver demonstrated superior performance in detecting HCC compared with currently widely used blood biomarkers, suggesting its potential clinical benefit in HCC surveillance in high-risk populations.
Collapse
Grants
- 2023J05234, 2023J011297 Natural Science Foundation of Fujian Province
- 2023J05234, 2023J011297 Natural Science Foundation of Fujian Province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2021Y9232, 2021Y9227, 2024Y9620 Joint Funds for the Innovation of Science and Technology, Fujian province
- 2022ZQNZD009 Young and Middle-aged Scientific Research Major Project of Fujian Provincial Health Commission
- 2022YNG01 high level talents training project of Fujian Cancer Hospital
Collapse
Affiliation(s)
- Wenhao Teng
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
- Fujian Provincial Key Laboratory of Tumor Biotherapy, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Hui Li
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Hao Yang
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
- Genetrix Biotech Corporation, Beijing, China
| | - Yu Chen
- Department of Medical Oncology, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Liying Xi
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fuli Xin
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Aiyuan Zhang
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Lihui Yu
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Lu Zheng
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Ming Wang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Jian Bai
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fayong Ke
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China
| | - Yin Wang
- Berry Oncology Corporation, Beijing, China
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
| | - Fuming Sun
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China
- Genetrix Biotech Corporation, Beijing, China
| | - Hui Zhang
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
| | - Lin Wu
- Berry Oncology Corporation, Beijing, China.
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China.
| | - Jingfeng Liu
- Department of Hepatopancreatobiliary Surgery, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, China.
- Fujian Key Laboratory of Advanced Technology for Cancer Screening and Early Diagnosis, Fuzhou, China.
| |
Collapse
|
6
|
Liou WL, Tan SY, Yamada H, Krishnamoorthy T, Chang JPE, Yeo CP, Tan CK. Performance of the GALAD Model in an Asian Cohort Undergoing Hepatocellular Carcinoma Surveillance: A Prospective Cohort Study. J Gastroenterol Hepatol 2025. [PMID: 40346978 DOI: 10.1111/jgh.16997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 04/05/2025] [Accepted: 04/28/2025] [Indexed: 05/12/2025]
Abstract
BACKGROUND AND AIM Current hepatocellular carcinoma (HCC) surveillance strategy has its limitations, consequently delaying early detection. The GALAD model has been validated in retrospective studies, with two published cut-off values yielding different sensitivities for HCCs of different etiologies. We evaluated the performance of GALAD model in HCC surveillance and determined the ideal cut-off value for our cohort. METHODS Patients undergoing 6-monthly HCC surveillance in Singapore General Hospital were recruited between December 2017-October 2018. Study serum specimens were prospectively collected and retrospectively tested using the μTASWako alpha-fetoprotein (AFP), AFP-L3, and protein induced by vitamin K antagonism-II (PIVKA-II) kits. GALAD score was calculated and compared with individual biomarkers using area under the curve (AUC) analysis. Published GALAD cut-offs of -0.63 and -1.95 were compared for their performance in HCC detection. RESULTS There were 207 patients (median age 59 years, 55.1% males). Hepatitis B was the commonest etiology (72.9%). By February 2023, with a median follow-up of 48.9 months, 20 patients had developed HCC. Eight patients developed HCC within 1 year from specimen collection. For HCC developing within 1 year, GALAD model detected HCC with an AUC of 0.84, greater than AFP (AUC 0.77), AFP-L3 (AUC 0.60), and PIVKA-II (AUC 0.67). GALAD at cut-off -1.95 achieved sensitivity and specificity of 75% and 92.5% for HCCs detected within 1 year, superior to cut-off -0.63 (sensitivity 12.5%, specificity 100%). CONCLUSION In this prospective study of HCC surveillance, the GALAD model performed better than individual biomarkers. The cut-off of -1.95 was more useful in our predominantly chronic hepatitis B cohort.
Collapse
Affiliation(s)
- Wei-Lun Liou
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Si-Yu Tan
- Department of Clinical Pathology, Singapore General Hospital, Singapore
| | - Hiroyuki Yamada
- Medical Systems Business Division, FUJIFILM Corporation, Osaka, Japan
| | | | - Jason Pik-Eu Chang
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| | - Chin-Pin Yeo
- Department of Clinical Pathology, Singapore General Hospital, Singapore
| | - Chee-Kiat Tan
- Department of Gastroenterology and Hepatology, Singapore General Hospital, Singapore
| |
Collapse
|
7
|
Li H, Jiao J, Gu Y, Zeng Y, Sheng Y. Risk factors and clinical outcomes in patients with HCV eradication by direct-acting antivirals: a systematic review and meta-analysis. Infect Dis (Lond) 2025:1-31. [PMID: 40333300 DOI: 10.1080/23744235.2025.2493370] [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: 01/09/2025] [Revised: 03/09/2025] [Accepted: 04/07/2025] [Indexed: 05/09/2025] Open
Abstract
BACKGROUND In hepatitis C patients with sustained virologic response (SVR) achieved after direct-acting antivirals (DAAs), the incidence of adverse clinical outcomes can be reduced but not completely eliminated. This meta-analysis aims at estimating the incidence of clinical outcomes in hepatitis C patients after achieving SVR with DAAs. METHODS Literature search was carried out in PubMed, Cochrane Library database, Web of Science, and Embase. The primary endpoint was the incidence of hepatocellular carcinoma (HCC) occurrence, HCC recurrence, decompensated cirrhosis, and liver-related mortality, following DAA-induced elimination of hepatitis C virus (HCV). Subgroup analyses were performed according to age, gender, comorbidities, region, fibrosis stage, presence of decompensation, duration of follow-up, start point of follow-up, and HCC treatment modality. Furthermore, meta-regression was performed to explore sources of high heterogeneity. RESULTS Finally, 132 articles were included in our study. The pooled HCC occurrence rate was 1.50/100 person-years (95% CI, 1.35-1.65), HCC recurrence rate was 17.00/100 person-years (95% CI, 13.83-20.42), decompensation rate was 0.30/100 person-years (95% CI, 0.16-0.48), and liver-related mortality was 0.32/100 person-years (95% CI, 0.14-0.56). Meta-regression showed that duration of follow-up and fibrosis grade were important contributors to HCC occurrence. Age, start point of follow-up, and duration of follow-up were important contributors to HCC recurrence rate. CONCLUSION Patients with DAA-induced HCV elimination remain at risk for adverse outcomes, particularly those with cirrhosis and HCC history. The exposure to adverse outcomes tended to decrease over time, and the frequency and intensity of follow-up might be reduced in the future, which will require new scoring models to identify these individuals.
Collapse
Affiliation(s)
- Hualing Li
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jiahuan Jiao
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yuyi Gu
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yu Zeng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Yunjian Sheng
- Department of Infectious Diseases, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| |
Collapse
|
8
|
Seif El Dahan K, Yokoo T, Daher D, Davenport MS, Fetzer DT, Mendiratta-Lala M, Rich NE, Yang E, Parikh ND, Singal AG. Multicenter evaluation of abbreviated MRI and ultrasound for detecting early-stage hepatocellular carcinoma. JHEP Rep 2025; 7:101357. [PMID: 40321196 PMCID: PMC12048809 DOI: 10.1016/j.jhepr.2025.101357] [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: 06/06/2024] [Revised: 02/06/2025] [Accepted: 02/10/2025] [Indexed: 05/08/2025] Open
Abstract
Background & aims Abbreviated MRI (AMRI) has been proposed as an alternative to ultrasound for hepatocellular carcinoma (HCC) surveillance; however, comparative data for AMRI and ultrasound are needed. Thus, we evaluated the sensitivity and specificity of dynamic contrast-enhanced (DCE)-AMRI and ultrasound for early-stage HCC detection in patients with cirrhosis. Methods We conducted a multicenter retrospective case-control study among patients with cirrhosis (cases with early-stage HCC as per Milan Criteria; controls without HCC) who underwent an ultrasound and a DCE-MRI within a 6-month period between 2012 and 2019. HCC diagnosis was confirmed by imaging alone in 85% and by histopathology in 15% of patients. Dynamic AMRI examinations were simulated from the full MRI by selecting relevant sequences. Independent, blinded interpretations of ultrasounds and AMRI results were performed using Liver Imaging Reporting and Data System algorithms. Ultrasounds were considered positive if US-3 observations were detected. AMRI was considered positive if LR-4, LR-5, or LR-M were detected. Per-patient sensitivity and specificity for early-stage HCC detection were estimated, and cross-modality differences were tested. Results We included 216 cases and 432 controls. Patient-level sensitivity and specificity of AMRI were significantly higher compared with ultrasound: 80.1% (95% CI 76.1-83.6) vs. 71.1% (95% CI 66.6-75.2), p <0.001, and 91.9% (95% CI 89.9-93.5) vs. 72.3% (95% CI 69.3-75.2), p <0.001, respectively. AMRI sensitivity was significantly higher compared with ultrasound among patients with Child-Pugh B cirrhosis (80.8% vs. 57.4%, p <0.001) but not among those with Child-Pugh A (84.7% vs. 78.6%, p = 0.07) or Child-Pugh C cirrhosis (52.6% vs. 68.4%, p = 0.18). Conclusions Dynamic AMRI may be more sensitive and specific for early-stage HCC detection in patients with cirrhosis compared with ultrasound, although its relative benefit might be smaller in patients with Child-Pugh A cirrhosis. Larger direct comparative data sets are needed, particularly among patients with Child-Pugh C cirrhosis who may benefit from alternative surveillance strategies. Impact and implications Abbreviated MRI (AMRI) is increasingly recognized as an alternative to ultrasound for hepatocellular carcinoma (HCC) surveillance. However, existing data are limited by single-center samples, spectrum bias, and lack of comparative data for AMRI vs. ultrasound. We found that AMRI had significantly higher per-patient sensitivity and specificity compared with ultrasound for the detection of early-stage HCC, although its relative benefit might be smaller in patients with Child-Pugh A cirrhosis, and both modalities underperformed in patients with Child-Pugh C cirrhosis. If sufficiently validated, AMRI could be adopted into practice guidelines for HCC surveillance and serve as a preferred alternative in select subgroups of patients.
Collapse
Affiliation(s)
- Karim Seif El Dahan
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Takeshi Yokoo
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | - Darine Daher
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Matthew S. Davenport
- Department of Radiology, University of Michigan, Ann Arbor, MI, USA
- Department of Urology, University of Michigan, Ann Arbor, MI, USA
| | - David T. Fetzer
- Department of Radiology, UT Southwestern Medical Center, Dallas, TX, USA
| | | | - Nicole E. Rich
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Edward Yang
- Division of Gastroenterology, Kaiser Permanente Medical Group, Riverside, CA, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
9
|
Vogel A, Chan SL, Dawson LA, Kelley RK, Llovet JM, Meyer T, Ricke J, Rimassa L, Sapisochin G, Vilgrain V, Zucman-Rossi J, Ducreux M. Hepatocellular carcinoma: ESMO Clinical Practice Guideline for diagnosis, treatment and follow-up. Ann Oncol 2025; 36:491-506. [PMID: 39986353 DOI: 10.1016/j.annonc.2025.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/24/2025] Open
Affiliation(s)
- A Vogel
- Department of Gastroenterology, Hepatology and Endocrinology, Hannover Medical School, Hannover, Germany; Division of Hepatology, Toronto General Hospital, Toronto, Canada; Division of Medical Oncology, Princess Margaret Cancer Centre, Toronto, Canada
| | - S L Chan
- State Key Laboratory of Translational Oncology, Department of Clinical Oncology, Sir YK Pao Centre for Cancer, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - L A Dawson
- Radiation Medicine Program, Princess Margaret Cancer Centre, University Health Network, Toronto, Canada; Department of Radiation Oncology, University of Toronto, Toronto, Canada
| | - R K Kelley
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, USA
| | - J M Llovet
- Mount Sinai Liver Cancer Program, Division of Liver Diseases, Icahn School of Medicine at Mount Sinai, New York, USA; Liver Cancer Translational Research Group, Liver Unit, Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), Hospital Clínic, Universitat de Barcelona, Barcelona, Spain; Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - T Meyer
- Department of Oncology, Royal Free Hospital, London, UK; UCL Cancer Institute, University College London, London, UK
| | - J Ricke
- Klinik und Poliklinik für Radiologie, Ludwig-Maximilians-Universität München, Munich, Germany
| | - L Rimassa
- Department of Biomedical Sciences, Humanitas University, Pieve Emanuele, Milan, Italy; Medical Oncology and Hematology Unit, Humanitas Cancer Center, IRCCS Humanitas Research Hospital, Rozzano, Milan, Italy
| | - G Sapisochin
- Department of Surgery, University of Toronto, Toronto, Canada
| | - V Vilgrain
- Centre de Recherche sur l'Inflammation U 1149, Université Paris Cité, Paris, France; Department of Radiology, Beaujon Hospital, APHP Nord, Clichy, France
| | - J Zucman-Rossi
- Centre de Recherche des Cordeliers, Université Paris Cité, Sorbonne Université, INSERM, Paris, France
| | - M Ducreux
- INSERM U1279, Université Paris-Saclay, Villejuif, France; Department of Cancer Medicine, Gustave Roussy, Villejuif, France
| |
Collapse
|
10
|
Rafati I, Destrempes F, Yazdani L, Barat M, Karam E, Fohlen A, Nguyen BN, Castel H, Tang A, Cloutier G. Enhancing Liver Nodule Visibility and Diagnostic Classification Using Ultrasound Local Attenuation Coefficient Slope Imaging. ULTRASOUND IN MEDICINE & BIOLOGY 2025; 51:807-814. [PMID: 39890529 DOI: 10.1016/j.ultrasmedbio.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 01/03/2025] [Accepted: 01/12/2025] [Indexed: 02/03/2025]
Abstract
OBJECTIVE B-mode ultrasound (US) presents challenges in accurately detecting and distinguishing between benign and malignant liver nodules. This study utilized quantitative US local attenuation coefficient slope (LACS) imaging to address these limitations. MATERIALS AND METHODS This is a prospective, cross-sectional study in adult patients with definable solid liver nodules at US conducted from March 2021 to December 2023. The composite reference standard included histopathology when available or magnetic resonance imaging. LACS images were obtained using a phantom-free method. Nodule visibility was assessed by computing the contrast-to-noise ratio (CNR). Classification accuracy for differentiating benign and malignant lesions was assessed with the area under the receiver operating characteristic curve (AUC), along with sensitivity and specificity. RESULTS The study enrolled 97 patients (age: 62 y ± 13 [standard deviation]), with 57.0% malignant and 43.0% benign observations (size: 26.3 ± 18.9 mm). LACS images demonstrated higher CNR (12.3 dB) compared to B-mode (p < 0.0001). The AUC for differentiating nodules and liver parenchyma was 0.85 (95% confidence interval [CI]: 0.79-0.90), with higher values for malignant (0.93, CI: 0.88-0.97) than benign nodules (0.76, CI: 0.66-0.87). A LACS threshold of 0.94 dB/cm/MHz provided a sensitivity of 0.83 (CI: 0.74-0.89) and a specificity of 0.82 (CI: 0.73-0.88). LACS mean values were higher (p < 0.0001) in malignant (1.28 ± 0.27 dB/cm/MHz) than benign nodules (0.98 ± 0.19 dB/cm/MHz). CONCLUSION LACS imaging improves nodule visibility and provides better differentiation between benign and malignant liver nodules, showing promise as a diagnostic tool.
Collapse
Affiliation(s)
- Iman Rafati
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - François Destrempes
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada
| | - Ladan Yazdani
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada
| | - Maxime Barat
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Elige Karam
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Audrey Fohlen
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Bich N Nguyen
- Department of Pathology, University of Montreal Hospital, Montréal, Québec, Canada
| | - Hélène Castel
- Departments of Hepatology and Liver Transplantation, University of Montreal Hospital, Montréal, Québec, Canada
| | - An Tang
- Department of Radiology, University of Montreal Hospital, Montréal, Québec, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada; Laboratory of Clinical Image Processing, University of Montreal Hospital Research Center, Montréal, Québec, Canada.
| | - Guy Cloutier
- Laboratory of Biorheology and Medical Ultrasonics, University of Montreal Hospital Research Center, Montréal, Québec, Canada; Institute of Biomedical Engineering, University of Montreal, Montréal, Québec, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, University of Montreal, Montréal, Québec, Canada.
| |
Collapse
|
11
|
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
|
12
|
Seif El Dahan K, Yokoo T, Mendiratta-Lala M, Fetzer D, Davenport M, Daher D, Rich NE, Yang E, Parikh ND, Singal AG. Exam quality of ultrasound and dynamic contrast-enhanced abbreviated MRI and impact on early-stage HCC detection. Abdom Radiol (NY) 2025; 50:2097-2109. [PMID: 39542949 DOI: 10.1007/s00261-024-04674-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/02/2024] [Indexed: 11/17/2024]
Abstract
PURPOSE MRI is a potential alternative to ultrasound for hepatocellular carcinoma (HCC) detection. We evaluated the impact of ultrasound and dynamic abbreviated MRI (AMRI) exam quality on early-stage HCC detection. METHODS We conducted a multicenter case-control study among patients with cirrhosis (cases with early-stage HCC per Milan Criteria; controls without HCC) who underwent both a liver ultrasound and dynamic contrast-enhanced (DCE) AMRI within 6 months in 2012-2019. Two radiologists performed independent, blinded interpretations of both exams for HCC detection and scored exam quality as no/mild, moderate, or severe limitations. Associations between exam quality, patient characteristics, and HCC detection were assessed by odds ratios (OR). RESULTS Of 216 cases and 432 controls, severe limitations were reported in 7% and 8% of ultrasounds and DCE-AMRIs, respectively. Severe limitations at ultrasound were associated with obesity (OR 2.08, 95%CI [1.32-3.32]) and metabolic dysfunction-associated steatotic liver disease (MASLD) (OR 1.98 [1.12-3.44]) but not for DCE-AMRI. Decompensated cirrhosis (Child-Pugh C) was associated with severe limitations for both ultrasound (OR 2.54 [1.37-4.58]) and DCE-AMRI (OR 3.96 [2.36-6.58]). Compared to exams with no/mild limitations, exams with severe limitations had lower sensitivity for ultrasound (79.6% vs. 21.7%, P < 0.001) and AMRI (86.1% vs. 50.0%, P = 0.001). In patients in whom ultrasound was severely limited, DCE-AMRI had significantly higher odds of early-stage HCC detection than ultrasound (OR 8.23 [1.25-54.02]). CONCLUSIONS HCC detection by DCE-AMRI may be preferred in patients with severe limitations at ultrasound due to obesity and MASLD. Both modalities remain limited for patients with decompensated cirrhosis, for whom alternative strategies may be needed.
Collapse
Affiliation(s)
| | - Takeshi Yokoo
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - David Fetzer
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - Darine Daher
- The University of Texas Southwestern Medical Center, Dallas, USA
| | - Nicole E Rich
- The University of Texas Southwestern Medical Center, Dallas, USA
| | - Edward Yang
- The University of Texas Southwestern Medical Center, Dallas, USA
| | | | - Amit G Singal
- The University of Texas Southwestern Medical Center, Dallas, USA.
| |
Collapse
|
13
|
Rui H, Yueqin N, Wei W, Bangtao L, Li X. Combining AFP, PIVKA-II, and GP73 has diagnostic utility for hepatitis B-associated hepatocellular carcinoma and is consistent with liver pathology results. Sci Rep 2025; 15:14869. [PMID: 40295542 PMCID: PMC12037889 DOI: 10.1038/s41598-025-92067-9] [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: 07/21/2024] [Accepted: 02/25/2025] [Indexed: 04/30/2025] Open
Abstract
Although liquid biopsy has garnered increasing attention in recent years for diagnosing hepatocellular carcinoma (HCC), serum biomarkers continue to hold significant value for HCC diagnosis due to their simple operation, cost-effectiveness, and high efficiency. This study aimed to screen for the optimal diagnostic combinations of alpha fetoprotein (AFP), a protein induced by vitamin K deficiency or antagonist II (PIVKA-II), golgi glycoprotein 73 (GP73), and routine clinical indicators for diagnosing hepatitis B-associated HCC (HBV-HCC). A retrospective analysis was conducted on 358 HBV-HCC patients treated at Taizhou People's Hospital from August 2015 to October 2021; 124 patients with chronic hepatitis B (CHB) and 241 patients with hepatitis B cirrhosis composed the control group. With liver pathology as the gold standard, the concordance between the screened indicators and liver pathology for HCC diagnosis was analyzed by Cohen's kappa coefficient. In the CHB group, AFP, PIVKA-II, and GP73 were statistical significance, and the triple biomarker combination achieved the highest AUC (0.908) for HCC diagnosis, surpassing the efficacy of both individual indicators and two biomarker combinations. In both the Child‒Pugh A and Child‒Pugh B&C cirrhosis groups, AFP and PIVKA-II were significantly different between patients with and without HCC, and the AUC values of AFP combined with PIVKA-II for HCC diagnosis were 0.969 and 0.956, respectively. Using liver pathology as the gold standard, the Kappa values of the above combinations in the three groups were 0.866, 0.780, and 0.800, respectively. The triple combination of AFP, PIVKA-II, and GP73 in the CHB group and the combination of AFP and PIVKA-II in both the Child‒Pugh A and Child‒Pugh B&C cirrhosis groups had excellent diagnostic accuracy for HCC, consistent with liver pathology, and were superior to the diagnostic ability of individual biomarkers.
Collapse
Affiliation(s)
- Hu Rui
- Department of Hepatology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou City, 225300, Jiangsu Province, China
- The First People's Hospital of Shaoguan, Shaoguan City, 512099, Guangdong Province, China
| | - Ni Yueqin
- Department of Hepatology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou City, 225300, Jiangsu Province, China
| | - Wang Wei
- Department of Hepatology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou City, 225300, Jiangsu Province, China
| | - Li Bangtao
- Department of Hepatology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou City, 225300, Jiangsu Province, China.
| | - Xiao Li
- Department of Hepatology, The Affiliated Taizhou People's Hospital of Nanjing Medical University, Taizhou City, 225300, Jiangsu Province, China.
| |
Collapse
|
14
|
Nedrud M, Wolfson T, Allen B, Aslam A, Burke L, Chernyak V, Fowler K, Fraum TJ, Ha HI, Hecht EM, Jaffe T, Kalisz K, Siobhan Kierans A, Ludwig DR, Makkar JS, McGinty K, McInnes M, Mendiratta-Lala M, Oloruntoba O, Ranathunga D, Wildman-Tobriner B, Gamst AC, Cardona DM, Muir A, Bashir M. Predictors of hepatocellular carcinoma in LR-M category lesions, a multi-institutional analysis. Abdom Radiol (NY) 2025:10.1007/s00261-025-04960-6. [PMID: 40293522 DOI: 10.1007/s00261-025-04960-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 04/15/2025] [Accepted: 04/16/2025] [Indexed: 04/30/2025]
Abstract
PURPOSE The Liver Imaging Reporting and Data System (LI-RADS, LR) provides a framework for diagnosing hepatocellular carcinoma (HCC, LR-5). However, not all HCCs meet LR-5 criteria and are instead categorized as LR-M, probably or definitely malignant but not specific for HCC, necessitating biopsy for diagnosis. The purpose is to identify factors associated with HCC in LR-M observations. METHODS This is an IRB-approved, retrospective analysis of participants from 8 institutions that had a LR-M observation on CT or MRI with corresponding histopathologic diagnosis. Demographics and biochemical data were examined. Central review using the LI-RADS v2018 algorithm was performed. Kappa statistics defined inter-reader agreement. Random forest and logistic regression analyses generated a model for HCC diagnosis. RESULTS 162 participants with 162 LR-M observations were included. 46% of observations (74/162) were HCC and 37% were cholangiocarcinoma (60/162). Two of 34 imaging features- observation size and intra-lesion iron- showed moderate to strong inter-reader agreement (Kappa ≥ 0.60) while the remainder showed weak or no agreement (Kappa < 0.60). Random forest analysis showed biochemical features to be more predictive of HCC than imaging features. Logistic regression analysis of a model based on INR and AFP provided a 72% sensitivity and 61% specificity for HCC by Youden's index and a 90% specificity threshold yielded 38% sensitivity, 75% positive predictive value, and 66% negative predictive value. CONCLUSIONS Our results show INR and AFP are associated with HCC in LR-M observations. A high-specificity threshold may assist in the non-invasive diagnosis of HCC in the appropriate setting. In certain at-risk patients with a LR-M observation on diagnostic imaging, serum AFP and INR maybe useful tools for the non-invasive diagnosis of HCC.
Collapse
Affiliation(s)
| | | | | | - Anum Aslam
- University of Michigan-Ann Arbor, Ann Arbor, USA
| | - Lauren Burke
- University of North Carolina at Chapel Hill, Chapel Hill, USA
| | | | | | | | | | | | | | | | | | | | | | - Katrina McGinty
- University of North Carolina at Chapel Hill, Chapel Hill, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Sequeira LM, Ozturk NB, Sierra L, Gurakar M, Toruner MD, Zheng M, Simsek C, Gurakar A, Kim AK. Hepatocellular Carcinoma and the Role of Liver Transplantation: An Update and Review. J Clin Transl Hepatol 2025; 13:327-338. [PMID: 40206277 PMCID: PMC11976436 DOI: 10.14218/jcth.2024.00432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 01/25/2025] [Accepted: 02/08/2025] [Indexed: 04/11/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the fourth leading cause of cancer-related death worldwide. Multiple treatment modalities are available for the management of HCC, depending on its stage as determined by the Barcelona Clinic Liver Cancer staging system. Because liver transplantation (LT) theoretically removes the cancer and replaces the organ at risk for future malignancy, LT is often considered the most definitive and one of the most efficacious treatment options for HCC. Nevertheless, the success and efficacy of liver transplantation depend on various tumor characteristics. As a result, multiple criteria have been developed to assess the appropriateness of a case of HCC for LT, with the pioneering Milan Criteria established in 1996. Over the past 20 to 30 years, these criteria have been critically evaluated, expanded, and often liberalized to make LT for patients with HCC a more universally applicable option. Furthermore, the development of other treatment modalities has enabled downstaging and bridging strategies for HCC prior to LT. In this narrative and comprehensive review, we provided an update on recent trends in the epidemiology of HCC, selection criteria for LT, implementation of LT across different regions, treatment modalities available as bridges, downstaging strategies, alternatives to LT, and, finally, post-LT surveillance.
Collapse
Affiliation(s)
- Lynette M. Sequeira
- Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - N. Begum Ozturk
- Department of Internal Medicine, Beaumont Hospital, Royal Oak, MI, USA
| | - Leandro Sierra
- Department of Internal Medicine, Cleveland Clinic, Cleveland, OH, USA
| | - Merve Gurakar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Melanie Zheng
- Department of Internal Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Cem Simsek
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Ahmet Gurakar
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Amy K. Kim
- Division of Gastroenterology and Hepatology, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| |
Collapse
|
16
|
Grimm LJ, Kruse DE, Tailor TD, Johnson KS, Allen BC, Ryser MD. Current Challenges in Imaging-Based Cancer Screening, From the AJR Special Series on Screening. AJR Am J Roentgenol 2025. [PMID: 40266702 DOI: 10.2214/ajr.25.32808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/25/2025]
Abstract
The early detection of cancer confers many significant benefits for patients, primarily by enabling less invasive and more effective treatments and thus lowering disease mortality. Radiology is integral to early cancer detection, playing either a primary or complementary role in screening programs. Imaging-based screening is often performed in conjunction with other screening tests and may involve multiple modalities depending on patient demographics and cancer type. When developing a screening program for cancer early detection, both its potential benefits and harms need to be assessed. These harms, although specific to the modality and cancer, often include overdiagnosis, overtreatment, and false-positive examinations. As radiology technology improves and new tools become available, the ratios of risk to harm of imaging-based screening will shift, and screening recommendations will need to adapt accordingly. Radiologists must be major partners in the development and execution of screening guidelines to ensure the highest quality of care for their patients. This review discusses the major challenges of cancer screening programs and guidelines, exploring sources of evidence as well as harms of overdiagnosis and overtreatment. The article focuses on the most common cancer types that incorporate imaging-based screening including lung cancer, breast cancer, colon cancer, prostate cancer, and hepatocellcular carcinoma.
Collapse
|
17
|
Li K, Mathew B, Saldanha E, Ghosh P, Krainer AR, Dasarathy S, Huang H, Xiang X, Mishra L. New insights into biomarkers and risk stratification to predict hepatocellular cancer. Mol Med 2025; 31:152. [PMID: 40269686 PMCID: PMC12020275 DOI: 10.1186/s10020-025-01194-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 04/01/2025] [Indexed: 04/25/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the third major cause of cancer death worldwide, with more than a doubling of incidence over the past two decades in the United States. Yet, the survival rate remains less than 20%, often due to late diagnosis at advanced stages. Current HCC screening approaches are serum alpha-fetoprotein (AFP) testing and ultrasound (US) of cirrhotic patients. However, these remain suboptimal, particularly in the setting of underlying obesity and metabolic dysfunction-associated steatotic liver disease/steatohepatitis (MASLD/MASH), which are also rising in incidence. Therefore, there is an urgent need for novel biomarkers that can stratify risk and predict early diagnosis of HCC, which is curable. Advances in liver cancer biology, multi-omics technologies, artificial intelligence, and precision algorithms have facilitated the development of promising candidates, with several emerging from completed phase 2 and 3 clinical trials. This review highlights the performance of these novel biomarkers and algorithms from a mechanistic perspective and provides new insight into how pathological processes can be detected through blood-based biomarkers. Through human studies compiled with animal models and mechanistic insight in pathways such as the TGF-β pathway, the biological progression from chronic liver disease to cirrhosis and HCC can be delineated. This integrated approach with new biomarkers merit further validation to refine HCC screening and improve early detection and risk stratification.
Collapse
Affiliation(s)
- Katrina Li
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Brandon Mathew
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Ethan Saldanha
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Puja Ghosh
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA
| | - Adrian R Krainer
- Cold Spring Harbor Laboratory, Cold Spring Harbor, NY, 11724, USA
| | - Srinivasan Dasarathy
- Division of Gastroenterology and Hepatology, Cleveland Clinic, Cleveland, OH, 44106, USA
| | - Hai Huang
- Center for Immunology and Inflammation, Feinstein Institutes for Medical Research, Donald and Barbara Zucker School of Medicine at Hofstra, Northwell Health, Manhasset, NY, 11030, USA
| | - Xiyan Xiang
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA.
| | - Lopa Mishra
- The Institute for Bioelectronic Medicine, Feinstein Institutes for Medical Research & Cold Spring Harbor Laboratory, Department of Medicine, Division of Gastroenterology and Hepatology, Northwell Health, NY, 11030, USA.
- Department of Surgery, George Washington University, Washington, DC, 20037, USA.
| |
Collapse
|
18
|
Xie L, Song D, Lan J, Liu P, Qin S, Ning Y, Liu Q. Plasma protein levels and hepatocellular carcinoma: a Mendelian randomization study with drug screening implications. Discov Oncol 2025; 16:567. [PMID: 40252200 PMCID: PMC12009266 DOI: 10.1007/s12672-025-02307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Accepted: 04/03/2025] [Indexed: 04/21/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a significant cause of cancer-related mortality, highlighting the need for novel therapeutic strategies. Identifying key proteins and potential therapeutic agents is critical for improving treatment outcomes. METHODS We employed Mendelian randomization to identify proteins associated with HCC risk and utilized drug enrichment and molecular docking analyses to discover potential therapeutic agents. The efficacy of identified drugs was evaluated in vitro using immune-tumor co-culture systems and in vivo in a murine HCC model. Single-cell expression profiling and clinical sample analyses were conducted to explore expression patterns. RESULTS Our analyses identified 16 proteins linked to HCC pathogenesis. Among the therapeutic agents tested, Belinostat significantly enhanced T cell-mediated cytotoxicity against HCC cells and effectively reduced tumor growth in vivo. Single-cell analysis revealed significant modulation of immune cells within the tumor microenvironment, suggesting potential mechanisms for the observed therapeutic effects. CONCLUSION This study highlights the potential of Belinostat as a promising therapeutic agent for HCC. By modulating immune responses and tumor growth, Belinostat offers a novel approach to HCC treatment, warranting further clinical investigation to validate its efficacy and therapeutic potential.
Collapse
Affiliation(s)
- Longhui Xie
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Yongzhou Central Hospital, Yongzhou, Hunan, China
| | - Dekun Song
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
- Binzhou People's Hospital Affiliated to Shandong First Medical University, Binzhou, Shandong, China
| | - Jianwei Lan
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Pengpeng Liu
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Shuang Qin
- Yongzhou Central Hospital, Yongzhou, Hunan, China
| | - Yinkuan Ning
- Department of Interventional Vascular Surgery, Shaoyang Central Hospital Shaoyang, Shaoyang, Hunan, China.
| | - Quanyan Liu
- Department of Hepatobiliary Surgery, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
19
|
Shen H, Liu M, Hu D, Xie F, Jin Q, Xiao D, Peng Z, Huang D. Application of immunofluorescence-based detection of AFP-L3 in the diagnosis of hepatocellular carcinoma. Medicine (Baltimore) 2025; 104:e42194. [PMID: 40258775 PMCID: PMC12014041 DOI: 10.1097/md.0000000000042194] [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: 06/07/2024] [Accepted: 04/03/2025] [Indexed: 04/23/2025] Open
Abstract
Alpha-fetoprotein (AFP) together with Lens culinaris-agglutinin-reactive fraction of AFP (AFP-L3) serve as the preferred tumor markers for hepatocellular carcinoma (HCC) diagnosis. The authors performed diagnostic value analysis on AFP along with AFP-L3 as individual biomarkers and together to determine the optimal biomarker option. Researchers evaluated 149 HCC patient sera and 70 healthy control sera using automatic microchip capillary electrophoresis and liquid phase combination-based analysis. They found the area under the curve value for the conventional marker AFP reached 0.844 and the diagnostic value of AFP-L3 was 0.923. The diagnostic effectiveness did not enhance as both biomarkers were combined for testing. The performance metrics of AFP-L3 increase during disease progression which leads to complete diagnostic sensitivity and perfect specificity and an area under the curve value of 1 during Barcelona clinical liver cancer and D stage.
Collapse
Affiliation(s)
- Haibin Shen
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Meijin Liu
- Laboratory Medicine, People’s Hospital of Ganzhou Economic Development Zone, Ganzhou, China
| | - Die Hu
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Fangfang Xie
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Qing Jin
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| | - Dewang Xiao
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Zongbo Peng
- The First School of Clinical Medicine, Gannan Medical University, Ganzhou, China
| | - Defa Huang
- Laboratory Medicine, First Affiliated Hospital of Gannan Medical University, Ganzhou, China
| |
Collapse
|
20
|
Wang Q, Cui H, Zhu Y, Zhu Y, He C. Serum Golgi protein 73 (GP73) is a diagnostic and prognostic marker of hepatocellular carcinoma. Front Med (Lausanne) 2025; 12:1571761. [PMID: 40297156 PMCID: PMC12034698 DOI: 10.3389/fmed.2025.1571761] [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: 02/06/2025] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) constitutes a significant global health burden, and is characterized by limited early detection methods and poor survival rates. Golgi protein 73 (GP73), previously associated with liver-related diseases, has a controversial diagnostic value for HCC. The present study aimed to determine the diagnostic efficacy of serum GP73 (sGP73) levels in HCC and to explore their potential correlations with the development of HCC. Methods The levels of sGP73 and serum alpha-fetoprotein (sAFP) were measured in 134 HCC patients, 200 healthy controls (HCs), and 45 non-HCC patients with various liver diseases. Additionally, immunohistochemical staining was conducted on paraffin-embedded tissue samples obtained from 30 HCC patients to examine the expression of CD4+ T cells, CD8+ T cells, Foxp3+ Treg cells, Ki-67, and interferon-gamma (IFN-γ) in the tissue specimens. Results sGP73 and sAFP were markedly higher in HCC patients than in HCs and non-HCC patients. However, sGP73 showed significantly higher sensitivity as a diagnostic marker for HCC than sAFP. The combination of sGP73 and sAFP further improved the accuracy (AUROC: 0.830). Besides, in the immunohistochemical staining analyses, sGP73-positive patients had lower expression of CD4+ and CD8+ T cells, higher expression of Foxp3+ Treg cells, higher expression of nuclear Ki67, and lower expression of IFN-γ than GP73-negative patients. In addition, sGP73-positive patients tended to have higher mortality rate, higher rate of metastasis, higher AFP levels, and more pronounced liver inflammation and damage than GP73-negative patients. Conclusions sGP73 could be utilized as a marker for the diagnosis of HCC, and may be implicated in the development of HCC through its interactions with the tumor microenvironment.
Collapse
Affiliation(s)
- Qian Wang
- Department of Clinical Laboratory, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Hongxia Cui
- Department of Pathology, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu, China
| | - Yaping Zhu
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Yichun Zhu
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| | - Chunyan He
- Department of Clinical Laboratory, Kunshan Hospital of Chinese Medicine, Kunshan, Jiangsu, China
| |
Collapse
|
21
|
Zhu W, Wang H, Cai Y, Lei J, Yu J, Li A, Yu Z. Plasma methylated HIST1H3G as a non-invasive biomarker for diagnostic modeling of hepatocellular carcinoma. Front Med (Lausanne) 2025; 12:1571737. [PMID: 40241895 PMCID: PMC12000021 DOI: 10.3389/fmed.2025.1571737] [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: 02/06/2025] [Accepted: 03/20/2025] [Indexed: 04/18/2025] Open
Abstract
Background DNA methylation carrying epigenetic aberrations could potentially serve as a non-invasive tool for revolutionizing cancer diagnosis and monitoring. Here, we comprehensively evaluated the diagnostic value of plasma methylated HIST1H3G, and constructed diagnostic and prognostic models aimed at facilitating early detection and improving the prognosis of hepatocellular carcinoma (HCC). Methods The level of HIST1H3G promoter methylation in HCC tissues was evaluated based on the UALCAN database, followed by validation through serum samples collected from HCC patients. We recruited 205 participants, encompassing 70 HCC patients, 79 liver cirrhosis (LC) patients, 46 hepatitis patients and 10 HCC patients before and after treatment with either transarterial chemoembolization (TACE) or radiofrequency ablation (RFA). Analysis of plasma HIST1H3G was performed using methylation-specific quantitative polymerase chain reaction (qPCR). Diagnostic and prognostic prediction models were formulated using the random forest algorithm, and the performance of these models was rigorously evaluated through receiver operating characteristics curve (ROC) analysis. Results The methylation level of HIST1H3G was markedly elevated in both HCC tissues and plasma samples derived from HCC patients. HIST1H3G, PIVKA-II, total bilirubin (TBIL) and age were selected as the optimal markers and were included in the development of a diagnostic model. This model demonstrated superior accuracy in distinguishing HCC from high-risk populations, outperforming alpha-fetoprotein (AFP) in both the training cohort consisting of LC patients and the validation cohort comprising hepatitis patients. Additionally, HIST1H3G and albumin (Alb) were chosen to establish a prediction model for early HCC diagnosis, and this model exhibited a remarkable ability to identify early HCC. Furthermore, our prognostic prediction model proved effective in predicting the prognosis and survival outcomes of HCC patients. Conclusion Together, we identified and validated a diagnostic model that incorporated methylated HIST1H3G and clinically applicable serological indicators in HCC. The findings of our study established a pivotal foundation for the development of a non-invasive approach to identification and management in HCC.
Collapse
Affiliation(s)
- Weiwei Zhu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Huifen Wang
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Yudie Cai
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jun Lei
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Jia Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Ang Li
- Precision Medicine Center, Gene Hospital of Henan Province, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Zujiang Yu
- Department of Infectious Diseases, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| |
Collapse
|
22
|
Soliman N, Maqsood A, Connor AA. Role of genomics in liver transplantation for cholangiocarcinoma. Curr Opin Organ Transplant 2025; 30:158-170. [PMID: 39917813 DOI: 10.1097/mot.0000000000001209] [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: 03/06/2025]
Abstract
PURPOSE OF REVIEW The purpose of this review is to summarize the current knowledge of cholangiocarcinoma molecular biology and to suggest a framework for implementation of next-generation sequencing in all stages of liver transplantation. This is timely as recent guidelines recommend increased use of these technologies with promising results. RECENT FINDINGS The main themes covered here address germline and somatic genetic alterations recently discovered in cholangiocarcinoma, particularly those associated with prognosis and treatment responses, and nascent efforts to translate these into contemporary practice in the peri-liver transplantation period. SUMMARY Early efforts to translate molecular profiling to cholangiocarcinoma care demonstrate a growing number of potentially actionable alterations. Still lacking is a consensus on what biomarkers and technologies to adopt, at what scale and cost, and how to integrate them most effectively into care with the ambition of increasing the number of patients eligible for liver transplantation and improving their long-term outcomes.
Collapse
Affiliation(s)
- Nadine Soliman
- Department of Surgery
- J. C. Walter Jr. Transplant Center, Houston Methodist Hospital
- Houston Methodist Academic Institute
| | - Anaum Maqsood
- Department of Medicine
- Neill Cancer Center, Houston Methodist Hospital, Houston, Texas
| | - Ashton A Connor
- Department of Surgery
- J. C. Walter Jr. Transplant Center, Houston Methodist Hospital
- Houston Methodist Academic Institute
- Neill Cancer Center, Houston Methodist Hospital, Houston, Texas
- Department of Surgery, Weill Cornell Medicine, Cornell University, New York, New York, USA
| |
Collapse
|
23
|
Adugna A, Azanaw Amare G, Jemal M. Current Advancements in Serum Protein Biomarkers for Hepatitis B Virus-Associated Hepatocyte Remodeling and Hepatocellular Carcinoma. Immun Inflamm Dis 2025; 13:e70171. [PMID: 40192058 PMCID: PMC11973733 DOI: 10.1002/iid3.70171] [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: 04/01/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Hepatitis B virus (HBV)-related liver cancer is the third most common cause of cancer-related death globally. Hepatocyte remodeling, also known as hepatocyte transformation and immortalization, and hepatocellular carcinoma (HCC), are brought on by persistent inflammation caused by HBV in the host hepatocytes. One of the main concerns in the perspective of HBV-induced hepatocyte remodeling and liver cancer is accurately identifying cancer stages to maximize early screening and detection. Biological signatures have a significant impact on solving this problem. OBJECTIVE This review article aimed to discuss the novel serum protein biomarkers for HBV-induced hepatocyte remodeling and HCC. METHODS The information was collected from various peer-reviewed journals through electronic searches utilizing various search engines, including PubMed, Google Scholar, HINARI, and Cochrane Library from 2017 to 2024. Keywords for searches included "serum protein biomarkers in HBV-HCC," "blood-based biomarkers in HBV-HCC," and "viral biomarkers for HBV-HCC." RESULTS Recently, novel protein signatures have been discovered for the early diagnosis, treatment, and prognosis of HBV-induced hepatic cell remodeling and HCC from proteomic data sets. We have discussed the recent literature on the clinical utility of the protein signatures for the diagnosis and forecasting of HBV-associated hepatocyte remodeling and HCC, including golgi protein 73 (GP73), glypican-3 (GPC3), midkine (MDK), des-γ-carboxy-prothrombin (DCP), von Willebrand factor (vWF), pentraxin 3 (PTX3), pseudouridine synthases 7 (PUSs 7), squamous cell carcinoma antigen (SCCA), and osteopontin (OPN). CONCLUSION All these protein markers also exhibit the survival of HBV-related HCC patients, the proliferation, migration, antiapoptosis, mitogenesis, transformation, and angiogenesis of HBV-infected hepatocytes.
Collapse
Affiliation(s)
- Adane Adugna
- Medical Laboratory SciencesCollege of Health SciencesDebre Markos UniversityDebre MarkosEthiopia
| | - Gashaw Azanaw Amare
- Medical Laboratory SciencesCollege of Health SciencesDebre Markos UniversityDebre MarkosEthiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of MedicineDebre Markos UniversityDebre MarkosEthiopia
| |
Collapse
|
24
|
Heo S, Kim SY, Lee SJ, Lee SS, Byun JH, Won HJ, Shin YM, Choi SH, Sirlin CB. LI-RADS Ultrasound Surveillance Version 2024: Comparison With Version 2017 for Hepatocellular Carcinoma Detection and Risk Factors for Visualization Score C. AJR Am J Roentgenol 2025; 224:e2432433. [PMID: 39840963 DOI: 10.2214/ajr.24.32433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2025]
Abstract
BACKGROUND. The LI-RADS Ultrasound Surveillance algorithm was updated in 2024, incorporating α-fetoprotein (AFP) and a visualization score of VIS-C into management recommendations after nonpositive results. OBJECTIVE. The purpose of this study was to compare the diagnostic performance of LI-RADS Ultrasound Surveillance version 2017 (v2017) and version 2024 (v2024) for the detection of hepatocellular carcinoma (HCC) in at-risk patients and to identify predictors of VIS-C on follow-up surveillance examinations. METHODS. This retrospective analysis included 407 patients (230 men and 177 women; median age, 56 years) with cirrhosis who underwent rounds of semiannual surveillance ultrasound as part of a prospective trial from November 2011 to August 2014. Two radiologists independently assigned ultrasound categories to round 1 examinations and visualization scores to round 1 and round 2 examinations; a third radiologist adjudicated disagreements. The AFP level was considered positive if elevated or increasing from preenrollment values, per v2024 criteria. The reference standard for HCC was positive biopsy or an LR-5 observation on MRI. Diagnostic performance was compared between v2017 and v2024. Logistic regression analyses were performed to identify predictors of a round 2 VIS-C result, with attention given to risk factors for VIS-C described in v2024. RESULTS. HCC was diagnosed in 28 patients (6.9%). LI-RADS Ultrasound Surveillance v2024, in comparison with v2017, showed greater sensitivity for reader 1 (64.3% vs 42.9%, p = .03) and reader 2 (64.3% vs 39.3%, p = .02) and lower specificity for reader 1 (82.1% vs 92.6%, p < .001) and reader 2 (82.3% vs 92.9%, p < .001). All seven patients with HCC detected by v2024 but not v2017 by means of consensus assessments had increasing AFP; two also had elevated AFP. Among 299 patients who underwent round 2 ultrasound after negative round 1 v2024 surveillance results, the only independent predictor of a round 2 VIS-C result was a round 1 VIS-C result (adjusted OR = 21.04 [95% CI, 10.84-40.83], p < .001). For 88 of these patients with round 1 VIS-C, no v2024 risk factor showed a significant univariable association with repeat VIS-C. CONCLUSION. Compared with v2017, LI-RADS Ultrasound Surveillance v2024 had higher sensitivity but lower specificity for HCC detection, which was primarily related to increasing, rather than elevated, AFP. The only independent predictor of VIS-C on subsequent ultrasound was the initial VIS-C result. CLINICAL IMPACT. The findings support the use of LI-RADS Ultrasound Surveillance v2024 to improve HCC detection in at-risk patients.
Collapse
Affiliation(s)
- Subin Heo
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - So Yeon Kim
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - So Jung Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Seung Soo Lee
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Jae Ho Byun
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Hyung Jin Won
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Yong Moon Shin
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Sang Hyun Choi
- Department of Radiology and Research Institute of Radiology, University of Ulsan College of Medicine, Asan Medical Center, 88 Olympic-ro 43-gil Songpa-gu, 05505 Seoul, Republic of Korea
| | - Claude B Sirlin
- Department of Radiology, Liver Imaging Group, UC San Diego, San Diego, CA
| |
Collapse
|
25
|
Yang T, Wang M, Wang N, Pan M, Xu Y, You Q, Yao L, Xu J, Gu L, Sun X, Zhang L, Xu J, Li B, Wang G, Cai S, Lv G, Shen F. Cost-Effective Identification of Hepatocellular Carcinoma from Cirrhosis or Chronic Hepatitis Virus Infection Using Eight Methylated Plasma DNA Markers. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2411945. [PMID: 40135830 DOI: 10.1002/advs.202411945] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 02/20/2025] [Indexed: 03/27/2025]
Abstract
Early detection of hepatocellular carcinoma (HCC) in patients with liver cirrhosis (LC) and/or hepatitis virus B/C infection (HVI) improves survival, highlighting the need for accurate, affordable diagnostic tools. Here, 11 methylated DNA markers (MDMs) are identified during marker discovery. In phase I, each selected MDM is validated in 175 plasma samples (HCC, n = 85; LC/HVI, n = 72) by the CO-methylation aMplification rEal-Time PCR (COMET) assay. Of these, 8 MDMs are qualified for phase II study, where a logistic regression model (COMET-LR) is trained and validated with 336 plasma samples (HCC, n = 211; LC/HVI, n = 113; training vs validation, 2:1). In the validation, the COMET-LR achieved 90.0% sensitivity at 97.4% specificity. Notably, sensitivity in patients with TNM stage I, diameter<3 cm, AFP-negative (<20 ng mL-1), PIVKA-II-negative (<40 mAU mL-1) is 82.4%, 77.8%, 88.6%, and 85.7%, respectively. The COMET-LR outperformed multiple protein markers (AFP, AFP-L3, and PIVKA-II) and published scores for HCC screening (GALAD, Doylestown, and ASAP), in terms of both sensitivity and specificity. The assay represents a significant advancement in addressing the unmet need for accurate, non-invasive, accessible, and cost-effective early detection tools for LC/HVI individuals. Further validation in a prospective cohort is warranted.
Collapse
Affiliation(s)
- Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Mingda Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Nanya Wang
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
- Phase I clinical trials unit, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Mingxin Pan
- Department of Hepatobiliary Surgery II, General Surgery Center, Zhujiang Hospital, Southern Medical University, Guangzhou, 510280, China
| | - Yu Xu
- Burning Rock Biotech, Guangzhou, 510300, China
| | | | - Lanqing Yao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Jiahao Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Lihui Gu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Lei Zhang
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Jiayue Xu
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Bingsi Li
- Burning Rock Biotech, Guangzhou, 510300, China
| | | | - Shangli Cai
- Burning Rock Biotech, Guangzhou, 510300, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, 130021, China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Naval Medical University, Shanghai, 200438, China
| |
Collapse
|
26
|
Yang HK, Lee S, Lee MY, Kim MJ. Effectiveness of noncontrast-abbreviated magnetic resonance imaging in a real-world hepatocellular carcinoma surveillance. Eur Radiol 2025:10.1007/s00330-025-11517-0. [PMID: 40111496 DOI: 10.1007/s00330-025-11517-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 03/22/2025]
Abstract
OBJECTIVES Noncontrast-abbreviated magnetic resonance imaging (NC-AMRI) is emerging as a promising alternative to ultrasound (US) for surveillance of hepatocellular carcinoma (HCC) in at-risk patients. We aimed to assess the effectiveness of NC-AMRI in a real-world surveillance population, and to evaluate the appropriateness of NC-AMRI in selected patients with inadequate prior US. MATERIALS AND METHODS This retrospective study included Child-Pugh class A or B adults with chronic hepatitis B or cirrhosis from any cause who underwent NC-AMRI between December 2018 and August 2022. Early- and very early-stage detection, receipt of curative treatment, and false referral were evaluated. Subgroup analysis was performed for patients with inadequate prior US examinations. Descriptive statistics were used. RESULTS Among the 1853 patients (mean age, 58.8 years; 1045 males), 68 HCCs developed in 61 (61/1853, 3.3%, 95% confidence interval: 2.5-4.2) patients. The proportions of early- and very early-stage detection were 95.1% (58/61, 72.2-100.0) and 70.5% (43/61, 51.0-95.0); receipt of curative treatment, 67.2% (41/61, 48.2-91.2); and proportion of false referral, 12.9% (9/70, 5.9-24.4). Among the 375 patients with inadequate prior US, the proportions of early- and very early-stage detection were 94.7% (18/19, 56.2-100.0) and 57.9% (11/19, 28.9-100.0); receipt of curative treatment, 52.6% (10/19, 25.2-96.8); and proportion of false referrals, 17.4% (4/23, 4.7-44.5). CONCLUSION NC-AMRI may be an effective HCC surveillance modality given the results related to early- and very early-stage detection, receipt of curative treatment, and false referral. NC-AMRI can be an alternative HCC surveillance strategy, especially for patients with inadequate prior US examinations. KEY POINTS Question There is insufficient evidence to support the use of noncontrast-abbreviated MRI as an effective surveillance tool in large real-life populations under surveillance. Findings Using noncontrast-abbreviated MRI, most patients who developed HCCs during surveillance were diagnosed at an early stage, with an acceptable false referral rate of 12.9%. Clinical relevance Noncontrast-abbreviated MRI is an effective HCC surveillance modality, especially for patients with inadequate prior ultrasound examinations.
Collapse
Affiliation(s)
- Hyun Kyung Yang
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| | - Sunyoung Lee
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea.
| | - Min Young Lee
- Office of Research Affairs/University Industry Foundation, Yonsei University, Seoul, Republic of Korea
| | - Myeong-Jin Kim
- Department of Radiology and Research Institute of Radiological Science, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
- Yonsei Liver Center, Severance Hospital, Seoul, Republic of Korea
| |
Collapse
|
27
|
Fassio E, Colombato L, Gualano G, Perez S, Puga-Tejada M, Landeira G. Hepatocellular Carcinoma After HCV Eradication with Direct-Acting Antivirals: A Reappraisal Based on New Parameters to Assess the Persistence of Risk. Cancers (Basel) 2025; 17:1018. [PMID: 40149352 PMCID: PMC11940336 DOI: 10.3390/cancers17061018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Revised: 03/12/2025] [Accepted: 03/16/2025] [Indexed: 03/29/2025] Open
Abstract
Approximately 95% of patients with chronic hepatitis C achieve viral eradication through direct-acting antiviral (DAA) treatment. Ensuing clinical benefits include halting liver fibrosis, thereby reducing the need for liver transplantation, and decreasing both liver-related and overall mortality. It is well established that, although ameliorated, the risk of developing hepatocellular carcinoma (HCC) persists, particularly among patients with pre-treatment advanced fibrosis/cirrhosis. Current guidelines recommend indefinite HCC surveillance in these patients. However, a recent Markov model evaluation shows that HCC surveillance is cost-effective only for patients with cirrhosis but not so for those with F3 fibrosis, a finding which points out the need to better define the risk of HCC in hepatitis C patients after cure and further characterize pre- and post-treatment factors that might affect the incidence of HCC in this setting. We reviewed the literature analyzing this aspect. Here we summarize the main findings: male gender and older age are independent predictors of increased risk of post-cure HCC development. Moreover, non-invasive tests for hepatic fibrosis, namely FIB4, APRI, and liver stiffness, measured before and after treatment and their post-therapy change, contribute to better stratifying the risk of HCC occurrence. Furthermore, low serum albumin, as well as an AFP above 7 ng/mL prior to and after DAA therapy, also constitute independent predictors of HCC development. Considering these findings, we propose to classify patients with HCV viral eradication and advanced fibrosis/cirrhosis into groups of low, medium, or high risk of HCC and to adopt adequate surveillance strategies for each group, including protocols for abbreviated magnetic resonance imaging (MRI) for those at the highest risk.
Collapse
Affiliation(s)
- Eduardo Fassio
- Liver Section, Gastroenterology Service, Hospital Nacional Profesor Alejandro Posadas, El Palomar, Buenos Aires 1684, Argentina; (S.P.); (G.L.)
| | - Luis Colombato
- Hospital Británico de Buenos Aires, Buenos Aires 1280, Argentina;
| | - Gisela Gualano
- Hospital Regional Dr. Ramón Carrillo, Santiago del Estero 4200, Argentina;
| | - Soledad Perez
- Liver Section, Gastroenterology Service, Hospital Nacional Profesor Alejandro Posadas, El Palomar, Buenos Aires 1684, Argentina; (S.P.); (G.L.)
| | - Miguel Puga-Tejada
- Instituto Ecuatoriano de Enfermedades Digestivas, Guayaquil 090505, Ecuador;
| | - Graciela Landeira
- Liver Section, Gastroenterology Service, Hospital Nacional Profesor Alejandro Posadas, El Palomar, Buenos Aires 1684, Argentina; (S.P.); (G.L.)
| |
Collapse
|
28
|
Lee DH. Recent advances and issues in imaging modalities for hepatocellular carcinoma surveillance. JOURNAL OF LIVER CANCER 2025; 25:31-40. [PMID: 40007309 PMCID: PMC12010830 DOI: 10.17998/jlc.2025.02.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 02/05/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related mortality worldwide. Early detection via surveillance plays a crucial role in enabling curative treatment and improving survival rates. Since the initial randomized controlled trial, biannual ultrasound (US) has been established as the standard surveillance method because of its accessibility, safety, and low cost. However, US has some limitations, including operator dependency, suboptimal sensitivity for early-stage HCC, and challenges such as a limited sonic window that may result in inadequate examination. Alternative imaging modalities, including contrast-enhanced computed tomography (CT) and magnetic resonance imaging (MRI), have demonstrated higher sensitivity for detecting very early-stage HCC. Recent advancements, such as low-dose CT with deep learning-based reconstruction, have enhanced the safety and feasibility of CT-based surveillance by reducing radiation exposure and amount of contrast media. MRI, particularly with gadoxetic acid or abbreviated protocols, offers superior tissue contrast and sensitivity, although its accessibility and cost remain challenges. Tailored surveillance strategies based on individual risk profiles and integration of advanced imaging technologies have the potential to enhance the detection performance and cost-effectiveness. This review highlights the recent developments in imaging technologies for HCC surveillance, focusing on their respective strengths and limitations.
Collapse
Affiliation(s)
- Dong Ho Lee
- Department of Radiology, Seoul National University Hospital, Seoul, Korea
- Department of Radiology, Seoul National University College of Medicine, Seoul, Korea
| |
Collapse
|
29
|
An S, Liu F, Shi Y. Identification of serum phosphatidylinositol glycan anchor biosynthesis class U protein as diagnostic biomarker for breast cancer. Clin Chim Acta 2025; 569:120183. [PMID: 39922250 DOI: 10.1016/j.cca.2025.120183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2024] [Revised: 01/26/2025] [Accepted: 02/05/2025] [Indexed: 02/10/2025]
Abstract
BACKGROUND Finding markers of breast cancer that are strong specificity and high. Sensitivity is important. The expression of phosphatidylinositol U(PIGU) protein is increased in a variety of tumor cells, but the role of its secreted fragment in breast cancer is lacking. METHODS Differential expression analysis was performed in breast cancer patients by bioinformatics method. Univariate survival analysis and ROC curve plotting were used to explore the correlation between serum PIGU and the prognosis of breast cancer patients. ELISA was used to detect serum PIGU level. Electrochemiluminescence immunoassay was used to determine serum carcinoembryonic antigen (CEA), carbohydrate antigen-153(CA153) and carbohydrate antigen-125 (CA125) levels. RESULTS The expression level of PIGU protein in breast cancer tumor tissues was higher than that in normal tissues, and PIGU expression level was a prognostic risk factor for breast cancer patients (HR > 1, p < 0.05) and had good predictive power (AUC = 0.8941). Compared with healthy individuals, the serum PIGU level of breast cancer patients was significantly highly expressed (p < 0.01), and the serum PIGU expression level was weakly positively correlated with CEA (r = 0.3270), but not significantly correlated with CA153 and CA125 (p > 0.05). CONCLUSION PIGU has the potential to be a predictive prognostic marker for breast cancer.
Collapse
Affiliation(s)
- Shuo An
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| | - Feifei Liu
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| | - Yue Shi
- Department of Clinical Laboratory, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center for Cancer, Tianjin's Clinical Research Center for Cancer, Key Laboratory of Cancer Prevention and Therapy, Tianjin 300060, China.
| |
Collapse
|
30
|
Eilard A, Ringlander J, Andersson ME, Nilsson S, Norkrans G, Lindh M. Long-Term Outcome of Chronic Hepatitis B-Histological Score and Viral Genotype Are Important Predictors of Hepatocellular Carcinoma. J Viral Hepat 2025; 32:e70008. [PMID: 39878776 PMCID: PMC11777188 DOI: 10.1111/jvh.70008] [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: 10/01/2024] [Revised: 01/13/2025] [Accepted: 01/21/2025] [Indexed: 01/31/2025]
Abstract
Current guidelines to prevent hepatocellular carcinoma (HCC) by chronic hepatitis B virus (HBV) infection are based on risk assessments that include age, sex, and virological and biochemical parameters. The study aim was to investigate the impact of predictive markers on long-term outcomes. The clinical outcomes of 100 patients with chronic hepatitis B were investigated 30 years after a baseline assessment that included liver biopsy. A favourable outcome-HBsAg loss or HBeAg-negative infection (ENI; previously termed 'inactive carrier')-was observed in 74% of all patients, whereas 7% developed HCC. HBsAg loss was observed in 75% of patients with genotype A, compared with 42%, 33% and 0% with genotypes D, B and C, respectively (p < 0.0001). HCC developed in 3 patients (33%) with genotype C as compared with 3 (17%), 1 (2%) and 0 patients with genotypes B, D and A, respectively (p < 0.0001). In multiple logistic regression analysis, both HBsAg loss and HCC were associated with HBV genotype and baseline HBV DNA level, and HCC also with histological score. The results suggest that genotyping and histological assessment may improve outcome prediction and help decisions about HCC screening, particularly in populations with HBV-infected individuals of mixed geographic origin.
Collapse
Affiliation(s)
- Anders Eilard
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Infectious DiseasesSahlgrenska University HospitalGothenburgSweden
| | - Johan Ringlander
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Maria E. Andersson
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Staffan Nilsson
- Department of Laboratory Medicine, Institute of BiomedicineUniversity of GothenburgSweden
| | - Gunnar Norkrans
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Magnus Lindh
- Department of Infectious Diseases, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
31
|
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
|
32
|
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
|
33
|
Mohnasky M, Gad S, Moon A, Barritt AS, Charalel RA, Eckblad C, Caddell A, Xing M, Kokabi N. Hepatocellular Carcinoma Screening: From Current Standard of Care to Future Directions. J Am Coll Radiol 2025; 22:260-268. [PMID: 40044304 DOI: 10.1016/j.jacr.2024.10.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 10/09/2024] [Accepted: 10/23/2024] [Indexed: 05/13/2025]
Abstract
Hepatocellular carcinoma (HCC) represents a significant portion of global cancer incidence and mortality. Screening with ultrasound with or without alpha-fetoprotein is recommended for those at high-risk. Although screening can lead to earlier treatment and better outcomes, existing screening paradigms have several flaws. Ultrasound does not capture all early lesions and has lower efficacy in specific populations such as patients with obesity or those with metabolic dysfunction-associated steatotic liver disease (MASLD). Additionally, individuals with noncirrhotic MASLD and chronic hepatitis C also develop HCC, although not at high enough rates to justify screening based on current standards. These individuals, however, represent a substantial proportion of new HCC cases given rising MASLD rates and the endemic nature of hepatitis C in certain regions. Risk-stratifying these populations may reveal subsets that are higher risk and warrant screening. Several imaging advances, including contrast-enhanced ultrasound and abbreviated MRI protocols, may improve detection compared with the current approach. Evaluation of risk stratification and validation of these new imaging methods via clinical trials would likely lead to adjusting screening guidelines. This narrative review provides a diagnostic and interventional radiology-focused summary of the HCC screening guidelines and their recent evolution and highlights emerging imaging methods as potential screening tools of the future.
Collapse
Affiliation(s)
- Michael Mohnasky
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina.
| | - Sandra Gad
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina; Saint George's University, School of Medicine, West Indies, Grenada
| | - Andrew Moon
- Assistant Professor of Medicine, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - A Sidney Barritt
- Professor of Medicine, Director of Hepatology, Transplant Hepatology Program Director, Division of Gastroenterology and Hepatology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Resmi A Charalel
- Assistant Professor of Population Health Science, Assistant Professor of Radiology, Division of Interventional Radiology, Department of Radiology, Weill Cornell Medicine, New York, New York
| | - Caroline Eckblad
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Andrew Caddell
- University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| | - Minzhi Xing
- Assistant Professor of Public Health Leadership and Practice, Gillings School of Global Public Health; Adjunct Assistant Professor of Radiology, University of North Carolina School of Medicine, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina
| | - Nima Kokabi
- Associate Professor of Radiology, Vice Chair of Clinical Research, Director of Interventional Oncology, and Director of Cancer Imaging, Department of Radiology, University of North Carolina at Chapel Hill, School of Medicine, Chapel Hill, North Carolina
| |
Collapse
|
34
|
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
|
35
|
Xu K, Zhang H, Dai H, Mao W. Machine learning and multi-omics characterization of SLC2A1 as a prognostic factor in hepatocellular carcinoma: SLC2A1 is a prognostic factor in HCC. Gene 2025; 938:149178. [PMID: 39681148 DOI: 10.1016/j.gene.2024.149178] [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: 07/31/2024] [Revised: 12/12/2024] [Accepted: 12/13/2024] [Indexed: 12/18/2024]
Abstract
Hepatocellular carcinoma (HCC) is characterized by high incidence, significant mortality, and marked heterogeneity, making accurate molecular subtyping essential for effective treatment. Using multi-omics data from HCC patients, we applied diverse clustering algorithms to identify three HCC subtypes (HSs) with distinct prognostic characteristics. Among these, HS1 emerged as an immune-compromised subtype associated with the poorest prognosis. Additionally, we developed a novel, robust, and highly accurate machine learning-guided prognostic signature (MLPS) by integrating multiple machine learning algorithms and their combinations. Our study also identified SLC2A1, the core gene of MLPS, as being highly expressed during advanced stages of tumor progression. Knockdown experiments demonstrated that reducing SLC2A1 expression significantly suppressed the malignant behavior of HCC cells. Furthermore, SLC2A1 expression was linked to responsiveness to dasatinib and vincristine, suggesting potential therapeutic relevance. MLPS and SLC2A1 offer promising tools for individualized prognosis prediction and targeted therapy in HCC, providing new opportunities to improve patient outcomes.
Collapse
Affiliation(s)
- Kangjie Xu
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Binhai County People's Hospital, Jiangsu Province, Yancheng 224000, PR China
| | - Houliang Zhang
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China
| | - Hua Dai
- Yangzhou University Clinical Medical College, Jiangsu Key Laboratory of Experimental & Translational Non-coding RNA Research, Jiangsu Province, Yangzhou 225009, PR China.
| | - Weipu Mao
- Zhongda Hospital, Southeast University, Jiangsu Province, Nanjing 210009, PR China; Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
36
|
Nwude VN, Lesi OA, Onyekwere C, Charpentier E, Hübschen JM. Clinical Characteristics of Hepatitis B Virus-Associated Hepatocellular Carcinoma Patients in Southwest Nigeria. Pathogens 2025; 14:169. [PMID: 40005544 PMCID: PMC11858220 DOI: 10.3390/pathogens14020169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 02/03/2025] [Accepted: 02/04/2025] [Indexed: 02/27/2025] Open
Abstract
Hepatitis B virus (HBV)-associated hepatocellular carcinoma (HCC) is a major cause of morbidity and mortality in West Africa, but its presentation is poorly understood. In this study, we describe the clinical characteristics of HBV-associated HCC patients in Lagos, Nigeria. Data for all cases were collected at the emergency and gastroenterology units (2017-2019), considering chronic carriers as controls. Clinical data and routine biochemical and radiologic test results were extracted from the files. The serum biomarkers (Osteopontin, AFP-L3, DCP) were investigated. For some cases, the hepatitis B viral load was determined. The mean age of the cases (n = 92) was 41.4 years, compared to 39.9 years for the controls (n = 100). Clinically, 69.5% of cases presented with ascites, 66.3% had nodules occupying >50% of the liver, and 67.4% had moderate hepatic encephalopathy. The mean viral load and the median values of Osteopontin, AFP-L3, and DCP for the cases were significantly higher than for the controls (p < 0.001). The area under the curve, sensitivity, and specificity were significantly higher for Osteopontin, compared with DCP and AFP-L3 (p < 0.001). Most HCC patients presented at a late disease stage, when the prognosis is usually poor. Especially Osteopontin seems to have potential for early HCC detection and could possibly complement AFP and abdominal ultrasound scan for risk-group screening.
Collapse
Affiliation(s)
- Vivian N. Nwude
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, 200 Hawkins Drive, Iowa City, IA 52242, USA
| | - Olufunmilayo A. Lesi
- Department of Internal Medicine, College of Medicine, University of Lagos, Lagos 12003, Lagos State, Nigeria
| | - Charles Onyekwere
- Lagos State University Teaching Hospital, 1-5 Oba Akinjobi Way, Ikeja 101233, Lagos State, Nigeria;
| | - Emilie Charpentier
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg;
| | - Judith M. Hübschen
- Clinical and Applied Virology, Department of Infection and Immunity, Luxembourg Institute of Health, 4354 Esch-sur-Alzette, Luxembourg;
| |
Collapse
|
37
|
Fadlallah H, El Masri D, Bahmad HF, Abou-Kheir W, El Masri J. Update on the Complications and Management of Liver Cirrhosis. Med Sci (Basel) 2025; 13:13. [PMID: 39982238 PMCID: PMC11843904 DOI: 10.3390/medsci13010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 02/01/2025] [Accepted: 02/02/2025] [Indexed: 02/22/2025] Open
Abstract
Liver cirrhosis represents the advanced pathological stage of chronic liver disease, characterized by the progressive destruction and regeneration of the hepatic parenchyma over years, culminating in fibrosis and disruption of the vascular architecture. As a leading global cause of morbidity and mortality, it continues to affect millions worldwide, imposing a substantial burden on healthcare systems. Alcoholic/nonalcoholic fatty liver disease and chronic viral hepatitis infection, hepatitis C (HCV) in particular, remain leading causes of cirrhosis. Despite significant advances in understanding the pathogenesis of cirrhosis, its management is still complex due to the multifaceted complications, including ascites, hepatic encephalopathy, variceal bleeding, and hepatocellular carcinoma, all of which severely compromise the patient outcomes and quality of life. This review aims at filling a critical gap by providing a comprehensive summary of the latest evidence on the complications and management of liver cirrhosis. Evidence-based therapies targeting both the etiologies and complications of cirrhosis are essential for improving outcomes. While liver transplantation is considered a definitive cure, advancements in pharmacological therapies offer promising avenues for halting and potentially reversing disease progression. This review summarizes the latest management strategies for cirrhosis and its associated complications, emphasizing the importance of early intervention and novel therapeutic options for improving outcomes and quality of life in affected individuals.
Collapse
Affiliation(s)
- Hiba Fadlallah
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (H.F.); (J.E.M.)
| | - Diala El Masri
- Faculty of Medicine, University of Balamand, Al-Kurah, Tripoli P.O. Box 100, Lebanon;
| | - Hisham F. Bahmad
- Department of Pathology and Laboratory Medicine, University of Miami Miller School of Medicine, Miami, FL 33136, USA;
| | - Wassim Abou-Kheir
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (H.F.); (J.E.M.)
| | - Jad El Masri
- Department of Anatomy, Cell Biology, and Physiological Sciences, American University of Beirut, Beirut 1107-2020, Lebanon; (H.F.); (J.E.M.)
- Faculty of Medical Sciences, Lebanese University, Beirut 1107-2020, Lebanon
| |
Collapse
|
38
|
Daher D, Gopal P, Coignet MV, Xiao V, Kurtzman KN, Singal AG. Performance of a Multi-cancer Early Detection Test for Detection of Hepatocellular Carcinoma in Patients With Cirrhosis. Clin Gastroenterol Hepatol 2025; 23:369-370. [PMID: 39074522 DOI: 10.1016/j.cgh.2024.07.016] [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: 05/18/2024] [Revised: 06/11/2024] [Accepted: 07/16/2024] [Indexed: 07/31/2024]
Affiliation(s)
- Darine Daher
- University of Texas Southwestern Medical Center, Dallas, Texas
| | - Purva Gopal
- University of Texas Southwestern Medical Center, Dallas, Texas
| | | | | | | | - Amit G Singal
- University of Texas Southwestern Medical Center, Dallas, Texas.
| |
Collapse
|
39
|
Groß S, Bitzer M, Albert J, Blödt S, Boda-Heggemann J, Borucki K, Brunner T, Caspari R, Dombrowski F, Evert M, Follmann M, Freudenberger P, Gani C, Gebert J, Geier A, Gkika E, Götz M, Helmberger T, Hoffmann RT, Huppert P, Krug D, Fougère CL, Lang H, Langer T, Lenz P, Lüdde T, Mahnken A, Nadalin S, Nguyen HHP, Nothacker M, Ockenga J, Oldhafer K, Ott J, Paprottka P, Pereira P, Persigehl T, Plentz R, Pohl J, Recken H, Reimer P, Riemer J, Ringe K, Roeb E, Rüssel J, Schellhaas B, Schirmacher P, Schlitt HJ, Schmid I, Schütte K, Schuler A, Seehofer D, Sinn M, Stengel A, Steubesand N, Stoll C, Tannapfel A, Taubert A, Trojan J, van Thiel I, Utzig M, Vogel A, Vogl T, Wacker F, Waidmann O, Wedemeyer H, Wege H, Wenzel G, Wildner D, Wörns MA, Galle P, Malek N. [Not Available]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:e82-e158. [PMID: 39919781 DOI: 10.1055/a-2460-6347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Affiliation(s)
- Sabrina Groß
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Michael Bitzer
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Jörg Albert
- Katharinenhospital, Klinik für Allgemeine Innere Medizin, Gastroenterologie, Hepatologie, Infektiologie und Pneumologie, Stuttgart
| | - Susanne Blödt
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | | | - Katrin Borucki
- Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Institut für Klinische Chemie und Pathobiochemie
| | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz
| | - Reiner Caspari
- Klinik Niederrhein Erkrankungen des Stoffwechsels der Verdauungsorgane und Tumorerkrankungen, Bad Neuenahr-Ahrweiler
| | | | | | - Markus Follmann
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | | | - Cihan Gani
- Klinik für Radioonkologie, Universitätsklinikum Tübingen
| | - Jamila Gebert
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Andreas Geier
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg
| | - Eleni Gkika
- Klinik für Strahlenheilkunde, Department für Radiologische Diagnostik und Therapie, Universitätsklinikum Freiburg
| | - Martin Götz
- Medizinische Klinik IV - Gastroenterologie/Onkologie, Klinikverbund Südwest, Böblingen
| | - Thomas Helmberger
- Institut für Radiologie, Neuroradiologie und minimal invasive Therapie, München Klinik Bogenhausen
| | - Ralf-Thorsten Hoffmann
- Institut und Poliklinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Dresden
| | - Peter Huppert
- Radiologisches Zentrum, Max Grundig Klinik, Bühlerhöhe
| | - David Krug
- Strahlentherapie Campus Kiel, Universitätsklinikum Schleswig-Holstein
| | - Christian La Fougère
- Nuklearmedizin und Klinische Molekulare Bildgebung, Eberhard-Karls Universität, Tübingen
| | - Hauke Lang
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Johannes Gutenberg-Universität, Mainz
| | - Thomas Langer
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | - Philipp Lenz
- Zentrale Einrichtung Palliativmedizin, Universitätsklinikum Münster
| | - Tom Lüdde
- Medizinische Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Düsseldorf
| | - Andreas Mahnken
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Marburg
| | - Silvio Nadalin
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Eberhard-Karls Universität, Tübingen
| | | | - Monika Nothacker
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | - Johann Ockenga
- Medizinische Klinik II, Gesundheit Nord, Klinikverbund Bremen
| | - Karl Oldhafer
- Klinik für Leber-, Gallenwegs- und Pankreaschirurgie, Asklepios Klinik Barmbek
| | - Julia Ott
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Philipp Paprottka
- Sektion für Interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München
| | - Philippe Pereira
- Zentrum für Radiologie, Minimal-invasive Therapien und Nuklearmedizin, SLK-Klinken Heilbronn
| | - Thorsten Persigehl
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln
| | - Ruben Plentz
- Digestive Diseases and Nutrition, Gastroenterology, University of Kentucky
| | - Jürgen Pohl
- Abteilung für Gastroenterologie, Asklepios Klinik Altona
| | | | - Peter Reimer
- Institut für Diagnostische und Interventionelle Radiologie, Städtisches Klinikum Karlsruhe
| | | | - Kristina Ringe
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | - Elke Roeb
- Medizinische Klinik II Pneumologie, Nephrologie und Gastroenterologie, Universitätsklinikum Gießen
| | - Jörn Rüssel
- Medizinische Klinik IV Hämatologie und Onkologie, Universitätsklinikum Halle (Saale)
| | - Barbara Schellhaas
- Medizinische Klinik I Gastroenterologie, Pneumologie und Endokrinologie, Friedrich-Alexander-Universität, Erlangen
| | - Peter Schirmacher
- Allgemeine Pathologie und pathologische Anatomie, Universitätsklinikum Heidelberg
| | | | - Irene Schmid
- Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU München
| | - Kerstin Schütte
- Klinik für Innere Medizin und Gastroenterologie, Niels-Stensen-Kliniken, Marienhospital Osnabrück
| | - Andreas Schuler
- Medizinische Klinik, Gastroenterologie, Alb-Fils-Kliniken, Geislingen an der Steige
| | - Daniel Seehofer
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig
| | - Marianne Sinn
- II. Medizinische Klinik und Poliklinik (Onkologie, Hämatologie, Knochenmarktransplantation mit Abteilung für Pneumologie), Universitätsklinikum Hamburg-Eppendorf
| | - Andreas Stengel
- Innere Medizin VI - Psychosomatische Medizin und Psychotherapie, Eberhard-Karls Universität, Tübingen
| | | | | | | | - Anne Taubert
- Klinische Sozialarbeit, Universitätsklinikum Heidelberg
| | - Jörg Trojan
- Medizinische Klinik 1: Gastroenterologie und Hepatologie, Pneumologie und Allergologie, Endokrinologie und Diabetologie sowie Ernährungsmedizin, Goethe-Universität, Frankfurt
| | | | - Martin Utzig
- Abteilung Zertifizierung, Deutsche Krebsgesellschaft e.V., Berlin
| | - Arndt Vogel
- Institute of Medical Science, University of Toronto
| | - Thomas Vogl
- Institut für Diagnostische und Interventionelle Radiologie, Goethe-Universität, Frankfurt
| | - Frank Wacker
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | | | - Heiner Wedemeyer
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover
| | - Henning Wege
- Klinik für Allgemeine Innere Medizin, Onkologie/Hämatologie, Gastroenterologie und Infektiologie, Klinikum Esslingen
| | - Gregor Wenzel
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e.V., Berlin
| | - Dane Wildner
- Innere Medizin, Krankenhäuser Nürnberger Land GmbH, Standort Lauf
| | - Marcus-Alexander Wörns
- Klinik für Gastroenterologie, Hämatologie und internistische Onkologie und Endokrinologie, Klinikum Dortmund
| | - Peter Galle
- 1. Medizinische Klinik und Poliklinik, Gastroenterologie, Hepatologie, Nephrologie, Rheumatologie, Infektiologie, Johannes Gutenberg-Universität, Mainz
| | - Nisar Malek
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| |
Collapse
|
40
|
Vutien P, Kim NJ, Nguyen MH. The Diagnosis and Staging of Hepatocellular Carcinoma: A Review of Current Practices. Clin Liver Dis 2025; 29:33-48. [PMID: 39608956 DOI: 10.1016/j.cld.2024.08.007] [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] [Indexed: 11/30/2024]
Abstract
Promoting the early detection and diagnosis of hepatocellular carcinoma (HCC) is a critical strategy to improve patient outcomes as this can lead to greater access to curative treatments. This review highlights the diagnostic tests for HCC, including the use of the Liver Imaging Reporting and Data System systems and histopathology. Staging is essential for informing prognosis and guiding treatment decisions; this review also covers a widely used and well-validated staging system called the Barcelona-Clinic Liver Cancer (BCLC) algorithm. The BCLC incorporates tumor status, liver function, and patient performance to stage patients with newly diagnosed HCC.
Collapse
Affiliation(s)
- Philip Vutien
- Division of Gastroenterology and Hepatology, University of Washington Medical Center, 1536 North 115th Street, Suite 105, Box 358811, Seattle, WA 98133, USA.
| | - Nicole J Kim
- Division of Gastroenterology and Hepatology, University of Washington Medical Center, 1536 North 115th Street, Suite 105, Box 358811, Seattle, WA 98133, USA
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, University of Washington Medical Center, 325 9th Avenue, Box 359773, Seattle, WA 98104, USA; Stanford University Medical Center, 780 Welch Road, Suite CJ250K, Palo Alto, CA 94304, USA
| |
Collapse
|
41
|
Sangro B, Argemi J, Ronot M, Paradis V, Meyer T, Mazzaferro V, Jepsen P, Golfieri R, Galle P, Dawson L, Reig M. EASL Clinical Practice Guidelines on the management of hepatocellular carcinoma. J Hepatol 2025; 82:315-374. [PMID: 39690085 DOI: 10.1016/j.jhep.2024.08.028] [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: 08/29/2024] [Accepted: 08/29/2024] [Indexed: 12/19/2024]
Abstract
Liver cancer is the third leading cause of cancer-related deaths worldwide, with hepatocellular carcinoma (HCC) accounting for approximately 90% of primary liver cancers. Advances in diagnostic and therapeutic tools, along with improved understanding of their application, are transforming patient treatment. Integrating these innovations into clinical practice presents challenges and necessitates guidance. These clinical practice guidelines offer updated advice for managing patients with HCC and provide a comprehensive review of pertinent data. Key updates from the 2018 EASL guidelines include personalised surveillance based on individual risk assessment and the use of new tools, standardisation of liver imaging procedures and diagnostic criteria, use of minimally invasive surgery in complex cases together with updates on the integrated role of liver transplantation, transitions between surgical, locoregional, and systemic therapies, the role of radiation therapies, and the use of combination immunotherapies at various stages of disease. Above all, there is an absolute need for a multiparametric assessment of individual risks and benefits, considering the patient's perspective, by a multidisciplinary team encompassing various specialties.
Collapse
|
42
|
Roberts LR. Surveillance for Hepatocellular Carcinoma. Clin Liver Dis 2025; 29:17-31. [PMID: 39608955 DOI: 10.1016/j.cld.2024.09.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2024]
Abstract
This article reviews surveillance for the detection of early stage hepatocellular carcinoma, covering the rationale for surveillance, optimal selection of persons needing surveillance, methods and frequency of screening, strategies for addressing barriers to surveillance, and trends for future improvement in surveillance leading to more effective cancer control and improved patient outcomes. The importance of integrating liver cancer surveillance as a core component of national public health programs is emphasized. The impact of emerging technologies for identifying persons at risk, stratifying individual risk to improve the cost-effectiveness of surveillance programs, and improving the performance, accessibility, and convenience of surveillance are discussed.
Collapse
Affiliation(s)
- Lewis R Roberts
- Division of Gastroenterology and Hepatology, Mayo Clinic College of Medicine and Science, 200 First Street Southwest, Rochester, MN 55905, USA.
| |
Collapse
|
43
|
Hwang SY, Danpanichkul P, Agopian V, Mehta N, Parikh ND, Abou-Alfa GK, Singal AG, Yang JD. Hepatocellular carcinoma: updates on epidemiology, surveillance, diagnosis and treatment. Clin Mol Hepatol 2025; 31:S228-S254. [PMID: 39722614 PMCID: PMC11925437 DOI: 10.3350/cmh.2024.0824] [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: 09/22/2024] [Revised: 11/08/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is a major global burden, ranking as the third leading cause of cancer-related mortality. HCC due to chronic hepatitis B virus (HBV) or C virus (HCV) infection has decreased due to universal vaccination for HBV and effective antiviral therapy for both HBV and HCV, but HCC related to metabolic dysfunction-associated steatotic liver disease and alcohol-associated liver disease is increasing. Biannual liver ultrasonography and serum α-fetoprotein are the primary surveillance tools for early HCC detection among high-risk patients (e.g., cirrhosis, chronic HBV). Alternative surveillance tools such as blood-based biomarker panels and abbreviated magnetic resonance imaging (MRI) are being investigated. Multiphasic computed tomography or MRI is the standard for HCC diagnosis, but histological confirmation should be considered, especially when inconclusive findings are seen on cross-sectional imaging. Staging and treatment decisions are complex and should be made in multidisciplinary settings, incorporating multiple factors including tumor burden, degree of liver dysfunction, patient performance status, available expertise, and patient preferences. Early-stage HCC is best treated with curative options such as resection, ablation, or transplantation. For intermediate-stage disease, locoregional therapies are primarily recommended although systemic therapies may be preferred for patients with large intrahepatic tumor burden. In advanced-stage disease, immune checkpoint inhibitor-based therapy is the preferred treatment regimen. In this review article, we discuss the recent global epidemiology, risk factors, and HCC care continuum encompassing surveillance, diagnosis, staging, and treatments.
Collapse
Affiliation(s)
- Soo Young Hwang
- Department of Internal Medicine, University of Maryland Medical Center, Midtown Campus, Baltimore, Maryland, USA
| | - Pojsakorn Danpanichkul
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, Texas, USA
| | - Vatche Agopian
- Dumont-UCLA Transplant and Liver Cancer Centers, Department of Surgery, David Geffen School of Medicine at UCLA, Los Angeles, California, USA
| | - Neil Mehta
- Division of Gastroenterology, Department of Medicine, University of California, San Francisco, California, USA
| | - Neehar D. Parikh
- Division of Gastroenterology and Hepatology, University of Michigan, Ann Arbor, Michigan, USA
| | - Ghassan K. Abou-Alfa
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, USA
- Department of Medicine, Weill Medical College at Cornell University, New York, USA
- Trinity College Dublin, Dublin, Ireland
| | - Amit G. Singal
- Division of Digestive and Liver Diseases, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Comprehensive Transplant Center, Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
44
|
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
|
45
|
Mak LY, Yuen MF. Surveillance for Hepatocellular Carcinoma in Cirrhosis: End of Monopoly for Serum Alpha Fetoprotein. Gastroenterology 2025; 168:217-219. [PMID: 39349108 DOI: 10.1053/j.gastro.2024.09.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/02/2024]
Affiliation(s)
- Lung-Yi Mak
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong
| | - Man-Fung Yuen
- Department of Medicine, Queen Mary Hospital, The University of Hong Kong, Hong Kong; State Key Laboratory of Liver Research, The University of Hong Kong, Hong Kong.
| |
Collapse
|
46
|
He Y, Gong L, Wu J, Wen B, Kong W. The value of contrast-enhanced ultrasound fusion imaging in percutaneous liver biopsy for liver lesions invisible on conventional B-mode ultrasound. Quant Imaging Med Surg 2025; 15:1528-1542. [PMID: 39995703 PMCID: PMC11847198 DOI: 10.21037/qims-24-1104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Accepted: 12/23/2024] [Indexed: 02/26/2025]
Abstract
Background Ultrasound (US) is the most commonly used imaging method for guiding percutaneous liver biopsies. For lesions that are invisible on B-mode ultrasound (BMUS), contrast-enhanced ultrasound (CEUS) improves the contrast between neoplasms and liver parenchyma, while emerging fusion imaging techniques can enhance the localization of lesions. Therefore, our study aimed to assess the value of CEUS fusion imaging in detecting and guiding percutaneous liver biopsy for lesions invisible on BMUS. Methods Patients with focal liver lesions (FLLs) identified on computed tomography (CT) or magnetic resonance imaging (MRI) but not visible on BMUS were retrospectively included at Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University from September 2019 to December 2023. All patients underwent BMUS fusion imaging and CEUS fusion imaging. We evaluated the lesion visibility, detection rate of BMUS fusion, and CEUS fusion before liver biopsy and the rate of technical success and diagnostic success after CEUS fusion-guided biopsy. Results This study included 70 FLLs from 61 patients. The mean visibility score of the 70 lesions on BMUS was 1.51±0.52. BMUS fusion detected 31 (31/70, 44.3%) lesions, yielding a mean visibility score of 1.81±1.03. CEUS fusion detected 63 (63/70, 90%) lesions, and achieved a mean visibility score of 2.90±0.30, which was significantly higher than that of BMUS fusion. Subgroup analysis corroborated the superiority of CEUS fusion in detecting infiltrative lesions and lesions <2 cm. Finally, CEUS fusion-guided liver biopsy achieved a technical success rate of 98.4% and a diagnostic success rate of 82.0% (50/61). Conclusions Compared with conventional BMUS or BMUS fusion imaging, the combination of CEUS and fusion imaging enabled an increase in the detection rate and lesion visibility of BMUS-invisible lesions, particularly for infiltrative masses and small-sized lesions, thereby increasing operators' confidence and success rate during the percutaneous liver biopsy.
Collapse
Affiliation(s)
- Yuhong He
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Li Gong
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Jie Wu
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Baojie Wen
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Wentao Kong
- Department of Ultrasound Medicine, Nanjing Drum Tower Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| |
Collapse
|
47
|
Groß S, Bitzer M, Albert J, Blödt S, Boda-Heggemann J, Borucki K, Brunner T, Caspari R, Dombrowski F, Evert M, Follmann M, Freudenberger P, Gani C, Gebert J, Geier A, Gkika E, Götz M, Helmberger T, Hoffmann RT, Huppert P, Krug D, La Fougère C, Lang H, Langer T, Lenz P, Lüdde T, Mahnken A, Nadalin S, Nguyen HHP, Nothacker M, Ockenga J, Oldhafer K, Ott J, Paprottka P, Pereira P, Persigehl T, Plentz R, Pohl J, Recken H, Reimer P, Riemer J, Ringe K, Roeb E, Rüssel J, Schellhaas B, Schirmacher P, Schlitt HJ, Schmid I, Schütte K, Schuler A, Seehofer D, Sinn M, Stengel A, Steubesand N, Stoll C, Tannapfel A, Taubert A, Trojan J, van Thiel I, Utzig M, Vogel A, Vogl T, Wacker F, Waidmann O, Wedemeyer H, Wege H, Wenzel G, Wildner D, Wörns MA, Galle P, Malek N. [Not Available]. ZEITSCHRIFT FUR GASTROENTEROLOGIE 2025; 63:169-203. [PMID: 39919782 DOI: 10.1055/a-2446-2454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/09/2025]
Affiliation(s)
- Sabrina Groß
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Michael Bitzer
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Jörg Albert
- Katharinenhospital, Klinik für Allgemeine Innere Medizin, Gastroenterologie, Hepatologie, Infektiologie und Pneumologie, Stuttgart
| | - Susanne Blödt
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | | | - Katrin Borucki
- Otto-von-Guericke-Universität Magdeburg, Medizinische Fakultät, Institut für Klinische Chemie und Pathobiochemie
| | - Thomas Brunner
- Universitätsklinik für Strahlentherapie-Radioonkologie, Medizinische Universität Graz
| | - Reiner Caspari
- Klinik Niederrhein Erkrankungen des Stoffwechsels der Verdauungsorgane und Tumorerkrankungen, Bad Neuenahr-Ahrweiler
| | | | | | - Markus Follmann
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e. V., Berlin
| | | | - Cihan Gani
- Klinik für Radioonkologie, Universitätsklinikum Tübingen
| | - Jamila Gebert
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Andreas Geier
- Medizinische Klinik und Poliklinik II, Universitätsklinikum Würzburg
| | - Eleni Gkika
- Klinik für Strahlenheilkunde, Department für Radiologische Diagnostik und Therapie, Universitätsklinikum Freiburg
| | - Martin Götz
- Medizinische Klinik IV - Gastroenterologie/Onkologie, Klinikverbund Südwest, Böblingen
| | - Thomas Helmberger
- Institut für Radiologie, Neuroradiologie und minimal invasive Therapie, München Klinik Bogenhausen
| | - Ralf-Thorsten Hoffmann
- Institut und Poliklinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Dresden
| | - Peter Huppert
- Radiologisches Zentrum, Max Grundig Klinik, Bühlerhöhe
| | - David Krug
- Strahlentherapie Campus Kiel, Universitätsklinikum Schleswig-Holstein
| | - Christian La Fougère
- Nuklearmedizin und Klinische Molekulare Bildgebung, Eberhard-Karls Universität, Tübingen
| | - Hauke Lang
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Johannes Gutenberg-Universität, Mainz
| | - Thomas Langer
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e. V., Berlin
| | - Philipp Lenz
- Zentrale Einrichtung Palliativmedizin, Universitätsklinikum Münster
| | - Tom Lüdde
- Medizinische Klinik für Gastroenterologie, Hepatologie und Infektiologie, Universitätsklinikum Düsseldorf
| | - Andreas Mahnken
- Klinik für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Marburg
| | - Silvio Nadalin
- Klinik für Allgemein-, Viszeral- und Transplantationschirurgie, Eberhard-Karls Universität, Tübingen
| | | | - Monika Nothacker
- Arbeitsgemeinschaft der Wissenschaftlichen Medizinischen Fachgesellschaften e. V. (AWMF), Berlin
| | - Johann Ockenga
- Medizinische Klinik II, Gesundheit Nord, Klinikverbund Bremen
| | - Karl Oldhafer
- Klinik für Leber-, Gallenwegs- und Pankreaschirurgie, Asklepios Klinik Barmbek
| | - Julia Ott
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| | - Philipp Paprottka
- Sektion für Interventionelle Radiologie, Klinikum rechts der Isar, Technische Universität München
| | - Philippe Pereira
- Zentrum für Radiologie, Minimal-invasive Therapien und Nuklearmedizin, SLK-Klinken Heilbronn
| | - Thorsten Persigehl
- Institut für Diagnostische und Interventionelle Radiologie, Universitätsklinikum Köln
| | - Ruben Plentz
- Digestive Diseases and Nutrition, Gastroenterology, University of Kentucky
| | - Jürgen Pohl
- Abteilung für Gastroenterologie, Asklepios Klinik Altona
| | | | - Peter Reimer
- Institut für Diagnostische und Interventionelle Radiologie, Städtisches Klinikum Karlsruhe
| | | | - Kristina Ringe
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | - Elke Roeb
- Medizinische Klinik II Pneumologie, Nephrologie und Gastroenterologie, Universitätsklinikum Gießen
| | - Jörn Rüssel
- Medizinische Klinik IV Hämatologie und Onkologie, Universitätsklinikum Halle (Saale)
| | - Barbara Schellhaas
- Medizinische Klinik I Gastroenterologie, Pneumologie und Endokrinologie, Friedrich-Alexander-Universität, Erlangen
| | - Peter Schirmacher
- Allgemeine Pathologie und pathologische Anatomie, Universitätsklinikum Heidelberg
| | - Hans J Schlitt
- Klinik und Poliklinik für Chirurgie, Universitätsklinikum Regensburg
| | - Irene Schmid
- Kinderklinik und Kinderpoliklinik im Dr. von Haunerschen Kinderspital, LMU München
| | - Kerstin Schütte
- Klinik für Innere Medizin und Gastroenterologie, Niels-Stensen-Kliniken, Marienhospital Osnabrück
| | - Andreas Schuler
- Medizinische Klinik, Gastroenterologie, Alb-Fils-Kliniken, Geislingen an der Steige
| | - Daniel Seehofer
- Klinik und Poliklinik für Viszeral-, Transplantations-, Thorax- und Gefäßchirurgie, Universitätsklinikum Leipzig
| | - Marianne Sinn
- II. Medizinische Klinik und Poliklinik (Onkologie, Hämatologie, Knochenmarktransplantation mit Abteilung für Pneumologie), Universitätsklinikum Hamburg-Eppendorf
| | - Andreas Stengel
- Innere Medizin VI - Psychosomatische Medizin und Psychotherapie, Eberhard-Karls Universität, Tübingen
| | | | | | | | - Anne Taubert
- Klinische Sozialarbeit, Universitätsklinikum Heidelberg
| | - Jörg Trojan
- Medizinische Klinik 1: Gastroenterologie und Hepatologie, Pneumologie und Allergologie, Endokrinologie und Diabetologie sowie Ernährungsmedizin, Goethe-Universität, Frankfurt
| | | | - Martin Utzig
- Abteilung Zertifizierung, Deutsche Krebsgesellschaft e. V., Berlin
| | - Arndt Vogel
- Institute of Medical Science, University of Toronto
| | - Thomas Vogl
- Institut für Diagnostische und Interventionelle Radiologie, Goethe-Universität, Frankfurt
| | - Frank Wacker
- Institut für Diagnostische und Interventionelle Radiologie, Medizinische Hochschule Hannover
| | | | - Heiner Wedemeyer
- Klinik für Gastroenterologie, Hepatologie, Infektiologie und Endokrinologie, Medizinische Hochschule Hannover
| | - Henning Wege
- Klinik für Allgemeine Innere Medizin, Onkologie/Hämatologie, Gastroenterologie und Infektiologie, Klinikum Esslingen
| | - Gregor Wenzel
- Office des Leitlinienprogrammes Onkologie, Deutsche Krebsgesellschaft e. V., Berlin
| | - Dane Wildner
- Innere Medizin, Krankenhäuser Nürnberger Land GmbH, Standort Lauf
| | - Marcus-Alexander Wörns
- Klinik für Gastroenterologie, Hämatologie und internistische Onkologie und Endokrinologie, Klinikum Dortmund
| | - Peter Galle
- 1. Medizinische Klinik und Poliklinik, Gastroenterologie, Hepatologie, Nephrologie, Rheumatologie, Infektiologie, Johannes Gutenberg-Universität, Mainz
| | - Nisar Malek
- Abteilung für Gastroenterologie, Gastrointestinale Onkologie, Hepatologie, Infektiologie und Geriatrie, Eberhard-Karls Universität, Tübingen
| |
Collapse
|
48
|
Kuaaroon W, Tiyarattanachai T, Apiparakoon T, Marukatat S, Tanpowpong N, Treeprasertsuk S, Rerknimitr R, Tangkijvanich P, Ananchuensook P, Chotiyaputta W, Samaithongcharoen K, Chaiteerakij R. Machine learning models for predicting hepatocellular carcinoma development in patients with chronic viral hepatitis B infection. ASIAN BIOMED 2025; 19:51-59. [PMID: 40231168 PMCID: PMC11994220 DOI: 10.2478/abm-2025-0007] [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] [Indexed: 04/16/2025]
Abstract
Background Chronic hepatitis B (CHB) infection is the major risk factor for hepatocellular carcinoma (HCC). Objective To develop machine-learning models for predicting an individual risk of HCC development in CHB-infected patients. Methods Machine learning models were constructed using features from follow-up visits of CHB patients to predict the diagnosis of HCC development within 6 months after each index follow-up. We developed 4 model variants using all features, with alpha fetoprotein (AFP) (AF A ) and without AFP (AFN ); and selected features, with AFP (SF A ) and without AFP (SFN ). Performance was evaluated using 10-fold cross-validation on a derivation cohort and further validated on an independent cohort. Results In the derivation cohort of 2,382 patients, of whom 117 developed HCC, AFA achieved higher sensitivity (0.634, 95% confidence interval [CI]: 0.559-0.708) and specificity (0.836; 0.830-0.842) than AF N (sensitivity 0.553; 0.476-0.630 and specificity 0.786; 0.779-0.792). SFA also achieved higher sensitivity (0.683; 0.611-0.755 vs. 0.658; 0.585-0.732) and specificity (0.756; 0.749-0.763 vs. 0.744; 0.737-0.751) than SFN . Performance of SFA and SFN were tested in another cohort of 162 patients in which 57 patients developed HCC. SFA achieved sensitivity and specificity of 0.634 (0.522-0.746) and 0.657 (0.615-0.699), while sensitivity and specificity of SFN were 0.690 (0.583-0.798) and 0.651 (0.609-0.693), respectively. Conclusion The machine learning models demonstrate good performance for predicting short-term risk for HCC development and may potentially be used for tailoring surveillance interval for CHB patients.
Collapse
Affiliation(s)
| | | | - Terapap Apiparakoon
- Department of Computer Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok10330, Thailand
| | - Sanparith Marukatat
- Image Processing and Understanding Team, Artificial Intelligence Research Group, National Electronics and Computer Technology Center,Pathumthani12120, Thailand
| | - Natthaporn Tanpowpong
- Department of Radiology, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
| | - Sombat Treeprasertsuk
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
| | - Rungsun Rerknimitr
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Chulalongkorn University, Bangkok10330, Thailand
| | - Pisit Tangkijvanich
- Center of Excellence in Hepatitis and Liver Cancer, Department of Biochemistry, Faculty of Medicine, Chulalongkorn University, Bangkok10330, Thailand
| | - Prooksa Ananchuensook
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
| | - Watcharasak Chotiyaputta
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | - Kittichai Samaithongcharoen
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok10700, Thailand
| | - Roongruedee Chaiteerakij
- Division of Gastroenterology, Department of Medicine, Faculty of Medicine, Chulalongkorn University and King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok10330, Thailand
- Center of Excellence for Innovation and Endoscopy in Gastrointestinal Oncology, Chulalongkorn University, Bangkok10330, Thailand
| |
Collapse
|
49
|
El-Azab GI, El-Helw GAA, Elsabaawy MMA, Kohla MAS, Omar YAM. Evaluation of screening program for hepatocellular carcinoma at a single center. EGYPTIAN LIVER JOURNAL 2025; 15:2. [DOI: 10.1186/s43066-025-00404-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/16/2025] [Indexed: 03/04/2025] Open
Abstract
Abstract
Background
Hepatocellular carcinoma (HCC) is the seventh most common cancer in the world and is a form of liver cancer that starts in the cells of the liver. The best way to increase the chances of survival for people at high risk for HCC is to detect it early through regular monitoring. For monitoring purposes, it is recommended to conduct ultrasound exams every 4 to 6 months, sometimes in combination with alpha-fetoprotein tests.
Aim of the work
Assess the HCC surveillance and its ability to detect HCC patients early on and improve their management.
Patients and methods
The study involved 300 patients of hepatocellular carcinoma (HCC) investigated at Menoufia University’s Institute of National Liver in Egypt. Patients were evaluated using the Liver Cancer of Barcelona Clinic (BCLC) staging system. Furthermore, the patients were classified into three surveillance categories: no surveillance, routine surveillance, and sporadic surveillance.
Results
A substantial difference statistically among the groups that received and did not receive surveillance with consideration for the stage of hepatocellular carcinoma (HCC) in particular was found. Patients who were observed usually got their diagnoses earlier. Those who were not under surveillance frequently had advanced cases of hepatocellular carcinoma upon diagnosis (HCC).
Conclusion
High-risk patients were regularly investigated for having HCC is necessary for early disease detection, appropriate therapy, and improved survival. Consistent monitoring with AFP and ultrasound allows for early detection of HCC.
Collapse
|
50
|
Toy M, Hutton D, Conners EE, Pham H, Salomon JA, So S. Cost-effectiveness of monitoring and liver cancer surveillance among patients with inactive chronic hepatitis B. PLoS One 2025; 20:e0313898. [PMID: 39841655 PMCID: PMC11753660 DOI: 10.1371/journal.pone.0313898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2024] [Accepted: 01/06/2025] [Indexed: 01/24/2025] Open
Abstract
Patients with chronic hepatitis B infection (CHB) have an increased risk for death from liver cirrhosis and hepatocellular carcinoma (HCC). In the United States, only an estimated 37% of adults with chronic hepatitis B diagnosis without cirrhosis receive monitoring with at least an annual alanine transaminase (ALT) and hepatitis B deoxyribonucleic acid (DNA), and an estimated 59% receive antiviral treatment when they develop active hepatitis or cirrhosis. A Markov model was used to calculate the costs, health impact and cost-effectiveness of increased monitoring of adults with HBeAg negative inactive or HBeAg positive immune tolerant CHB who have no cirrhosis or significant fibrosis and are not recommended by the current American Association for the Study of Liver Diseases (AASLD) clinical practice guidelines to receive antiviral treatment, and to assess whether the addition of HCC surveillance would be cost-effective. For every 100,000 adults with CHB who were initially not recommended for treatment, if the monitoring rate increased from the current 37% to 90% and treatment rate increased from 59% to 80%, 4,600 cases of cirrhosis, 2,450 cases of HCC and 4,700 HBV-related deaths would be averted with a gain of 45,000 QALYs and a savings of $180 million in lifetime health care costs. At a willingness to pay threshold of $100,000/QALY, the addition of HCC surveillance with the standard recommended biannual liver ultrasound and alfa fetoprotein levels is likely cost-effective if the HCC risk ≥ 0.55%/year. Regular monitoring of persons with inactive or immune tolerant CHB who are initially not recommended to receive antiviral treatment in the United States is cost-saving. The addition of HCC surveillance with biannual US and AFP would be cost-effective for individuals with HCC incidence ≥ 0.55%/year.
Collapse
Affiliation(s)
- Mehlika Toy
- Erasmus School of Health Policy & Management, Erasmus University, Rotterdam, The Netherlands
| | - David Hutton
- Department of Health Management and Policy, University of Michigan, Ann Arbor, Michigan, United States of America
| | - Erin E. Conners
- Division of Viral Hepatitis, Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Hang Pham
- Department of Surgery, Asian Liver Center, Stanford University School of Medicine, Stanford, California, United States of America
| | - Joshua A. Salomon
- Department of Health Policy, Stanford University School of Medicine, Stanford, California, United States of America
- Center for Health Policy, Freeman Spogli Institute for International Studies, Stanford University, Stanford, California, United States of America
| | - Samuel So
- Department of Surgery, Asian Liver Center, Stanford University School of Medicine, Stanford, California, United States of America
| |
Collapse
|