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
|
Li R, Zhang G, Tao Q, Wu Z, Liu X, Wang R, Liu L, Niu Y, Du K, Wu R, Du F, Zheng X, Li Y, Shi X. Revealing the prognostic potential of natural killer cell-related genes in hepatocellular carcinoma: the key role of NRAS. Discov Oncol 2025; 16:807. [PMID: 40383831 PMCID: PMC12086133 DOI: 10.1007/s12672-025-02200-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 03/21/2025] [Indexed: 05/20/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is a common malignancy associated with high morbidity and mortality rates worldwide. To improve the prognosis of HCC, early diagnosis is crucial. However, to date, little is known about the role of natural killer cell-related genes (NKCRGs) in predicting the prognosis of hepatocellular carcinoma patients. In this study, we identified 24 differentially expressed NKCRGs in HCC specimens from the TCGA dataset, including 22 upregulated genes and 2 downregulated genes. Functional enrichment analysis revealed that these genes were mainly involved in immune response pathways and various cancer-related pathways. Univariate analysis identified 21 prognostic NKCRGs, with eight genes (PAK1, MAP2K2, MAPK3, PLCG1, SHC1, HRAS, NRAS, and MICB) confirmed to be involved in HCC prognosis through Venn diagram analysis. A prognostic model was developed using LASSO-Cox regression, incorporating four genes (MAP2K2, SHC1, HRAS, and NRAS). The model's risk score was significantly associated with overall survival (OS) in both the TCGA and ICGC cohorts. Patients with high-risk scores had poorer OS, as demonstrated by Kaplan-Meier curves and ROC analyses. The risk score was not significantly correlated with gender or age but was higher in patients with advanced tumor grades and stages. Immune status analysis using ssGSEA showed higher enrichment scores for various immune cells and pathways in the high-risk group. Additionally, the risk score was positively correlated with the immune score, indicating its potential role in tumor microenvironment modulation. Expression analysis revealed that HRAS, SHC1, MAP2K2, and NRAS were upregulated in HCC tissues, with higher expressions of HRAS, MAP2K2, and NRAS associated with shorter OS. Knockdown experiments confirmed that silencing NRAS suppressed the proliferation of HCC cells, highlighting its potential as a therapeutic target. Overall, our findings suggest that the identified NKCRGs, particularly NRAS, play crucial roles in HCC progression and could serve as valuable prognostic markers and therapeutic targets.
Collapse
Affiliation(s)
- Ruixi Li
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Guangquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Qiang Tao
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Ziyun Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Xiaoping Liu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Rongrong Wang
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Lei Liu
- Department of Clinical Pharmacy, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yiran Niu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Kaile Du
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Runpeng Wu
- The First Clinical Medical College of Nanchang University, Nanchang, 330031, China
| | - Fei Du
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Xiyan Zheng
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China
| | - Yingliang Li
- Department of Breast Disease Center, The First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, 330006, China.
| | - Xianjie Shi
- Department of Hepatobiliary and Pancreatic Surgery, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen, 518033, China.
| |
Collapse
|
3
|
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] [Download PDF] [Figures] [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
|
4
|
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
|
5
|
Zhuang L, Park SH, Skates SJ, Prosper AE, Aberle DR, Hsu W. Advancing Precision Oncology Through Modeling of Longitudinal and Multimodal Data. ARXIV 2025:arXiv:2502.07836v2. [PMID: 39990791 PMCID: PMC11844620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/25/2025]
Abstract
Cancer evolves continuously over time through a complex interplay of genetic, epigenetic, microenvironmental, and phenotypic changes. This dynamic behavior drives uncontrolled cell growth, metastasis, immune evasion, and therapy resistance, posing challenges for effective monitoring and treatment. However, today's data-driven research in oncology has primarily focused on cross-sectional analysis using data from a single modality, limiting the ability to fully characterize and interpret the disease's dynamic heterogeneity. Advances in multiscale data collection and computational methods now enable the discovery of longitudinal multimodal biomarkers for precision oncology. Longitudinal data reveal patterns of disease progression and treatment response that are not evident from single-timepoint data, enabling timely abnormality detection and dynamic treatment adaptation. Multimodal data integration offers complementary information from diverse sources for more precise risk assessment and targeting of cancer therapy. In this review, we survey methods of longitudinal and multimodal modeling, highlighting their synergy in providing multifaceted insights for personalized care tailored to the unique characteristics of a patient's cancer. We summarize the current challenges and future directions of longitudinal multimodal analysis in advancing precision oncology.
Collapse
Affiliation(s)
- Luoting Zhuang
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Stephen H Park
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Steven J Skates
- Harvard Medical School, Boston, MA 02115 USA, and also with Biostatistics Center, Massachusetts General Hospital, Boston, MA 02114 USA
| | - Ashley E Prosper
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - Denise R Aberle
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| | - William Hsu
- Medical & Imaging Informatics, Department of Radiological Sciences, David Geffen School of Medicine at UCLA, Los Angeles, CA 90024 USA
| |
Collapse
|
6
|
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
|
7
|
Liu Z, Shan D, Han X. GALAD's Impact on HCC Detection: Insights and Future Considerations. Gastroenterology 2025:S0016-5085(25)00629-8. [PMID: 40220969 DOI: 10.1053/j.gastro.2024.09.052] [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: 09/17/2024] [Accepted: 09/18/2024] [Indexed: 04/14/2025]
Affiliation(s)
- Zaoqu Liu
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| | - Dan Shan
- Department of Biobehavioral Sciences, Columbia University, New York, New York
| | - Xinwei Han
- Department of Interventional Radiology, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, Henan, China
| |
Collapse
|
8
|
Spiers J, Li W, Aravinthan AD, Bannaga A, Caddick K, Culver EL, Faulkes RE, Gordon V, Hussain Y, Miller H, Merry J, Saad M, Sheth A, Shah T, Shetty S, Srivastava A, Subhani M, Tahir MN, Than NN, Unitt E, Alazawi W. Current Surveillance Strategy Is Less Effective for Detecting Early-Stage Hepatocellular Carcinoma in Patients with Non-Viral and Non-Cirrhotic Liver Disease. Liver Cancer 2025:1-14. [PMID: 40337094 PMCID: PMC12055015 DOI: 10.1159/000542805] [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: 05/22/2024] [Accepted: 11/21/2024] [Indexed: 05/09/2025] Open
Abstract
Introduction Hepatocellular carcinoma (HCC) is a leading cause of cancer-related deaths. Current international guidelines recommend 6-monthly ultrasound surveillance in all patients with cirrhosis and those with hepatitis B virus-related risk factors to detect early-stage HCC. However, it is unknown whether the benefits of surveillance are comparable across patient groups and underlying disease-related factors. We aimed to evaluate patient- and disease-related factors associated with HCC stage at diagnosis and survival in an ethnically diverse UK population. Methods This was a multicentre retrospective observational study including patients with newly diagnosed HCC between 2007 and 2020 from six UK centres. Cox proportional-hazards regression and multivariate logistic regression models were used. Results Overall, 1,780 HCC patients comprising 20.9% with ArLD, 29.7% with NAFLD, and 31.0% with viral hepatitis were analysed. Surveillance was associated with improved survival in patients with viral hepatitis but not in patients with ArLD and NAFLD. Surveillance was also associated with early-stage disease (BCLC stage 0 or A) at presentation in viral hepatitis but not in patients with ArLD. Females with ArLD were 2.5-fold more likely to present with early-stage HCC than males. Patients with NAFLD were more likely to develop HCC in the absence of cirrhosis. Type 2 diabetes was not associated with mortality, but metformin use did show survival benefit. Patients of white ethnicity had improved survival and were less likely to present with late-stage HCC compared to other ethnicities. Conclusions HCC surveillance as currently delivered was less effective for detecting early-stage HCC in patients with non-viral and non-cirrhotic liver disease. Gender and ethnicity influences stage at presentation and outcomes. HCC surveillance strategies are needed to refine risk stratification particularly in patients with NAFLD or without cirrhosis.
Collapse
Affiliation(s)
- Jessica Spiers
- Barts Liver Centre, Blizard Institute, Queen Mary Unversity of London, London, UK
| | - Wenhao Li
- Barts Liver Centre, Blizard Institute, Queen Mary Unversity of London, London, UK
| | - Aloysious D. Aravinthan
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Ayman Bannaga
- Liver Unit, Queen Elizabeth Hospital, Birmingham, UK
| | | | - Emma L. Culver
- Translational Gastroenterology Unit, John Radcliffe Hospital, and Oxford NIHR and BRC Oxford, Oxford, UK
| | | | - Victoria Gordon
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Yaqza Hussain
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Hamish Miller
- Barts Liver Centre, Blizard Institute, Queen Mary Unversity of London, London, UK
- Translational Gastroenterology Unit, John Radcliffe Hospital, and Oxford NIHR and BRC Oxford, Oxford, UK
| | - Jenny Merry
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Muhammad Saad
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Abhishek Sheth
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | - Tahir Shah
- Liver Unit, Queen Elizabeth Hospital, Birmingham, UK
| | | | | | - Mohsan Subhani
- NIHR Nottingham Biomedical Research Centre, Nottingham University Hospitals NHS Trust and University of Nottingham, Nottingham, UK
- Nottingham Digestive Diseases Centre, Translational Medical Sciences, School of Medicine, University of Nottingham, Nottingham, UK
| | | | - Nwe Ni Than
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - Esther Unitt
- University Hospitals Coventry and Warwickshire NHS Trust, Coventry, UK
| | - William Alazawi
- Barts Liver Centre, Blizard Institute, Queen Mary Unversity of London, London, UK
| |
Collapse
|
9
|
Ni Y, Liu B, Zhang W, Pang Y, Tian Y, Lv Q, Shi S, Zheng Y, Fan H. Evaluation of PDZD11 in hepatocellular carcinoma: prognostic value and diagnostic potential in combination with AFP. Front Oncol 2025; 15:1533865. [PMID: 40201341 PMCID: PMC11975663 DOI: 10.3389/fonc.2025.1533865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2024] [Accepted: 03/06/2025] [Indexed: 04/10/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most prevalent liver cancer, with a 5-year survival rate below 20% and an average survival time of 3-6 months. Identifying new biomarkers is crucial for early diagnosis and prognosis. The function of PDZ domain protein 11 (PDZD11) in HCC remains unclear. Methods In this study, PDZD11 was investigated as a potential biomarker for HCC using bioinformatic analysis of the TCGA and ICGC datasets. Furthermore, we assessed the potential of serum PDZD11 as a clinical diagnostic marker by enrolling a cohort comprising 78 HCC patients and 62 healthy controls (HC) using the ELISA analysis and combining its expression with common tumor markers. Results Our research found significantly higher PDZD11 mRNA expression in HCC tissues compared to tumor-adjacent tissues (p < 0.001), which was associated with lower overall survival (OS) rates (p < 0.01). Multivariate evaluation methods established PDZD11 as a standalone predictor of prognosis. A nomogram incorporating PDZD11 expression and clinicopathological factors predicted OS rates for HCC patients over various years. Patients with HCC exhibited notably elevated serum PDZD11 levels compared to HC, with these levels rising further in advanced disease stages and deteriorating performance status (PS). ROC analysis showed high diagnostic accuracy when PDZD11 is combined with AFP (AUC = 0.958). Conclusion PDZD11 is more sensitive than AFP in assessing HCC prognosis. In conclusion, PDZD11 is a promising supplementary biomarker for HCC diagnosis and prognosis alongside AFP.
Collapse
Affiliation(s)
- Yiyun Ni
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Bin Liu
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Weizhen Zhang
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yilin Pang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yaling Tian
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Qingsong Lv
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Shengwen Shi
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Yang Zheng
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| | - Huihui Fan
- The Central Hospital of Yongzhou, Yongzhou Clinical College, University of South China, Yongzhou, Hunan, China
| |
Collapse
|
10
|
Schäfer H, Lajmi N, Valente P, Pedrioli A, Cigoianu D, Hoehne B, Schenk M, Guo C, Singhrao R, Gmuer D, Ahmed R, Silchmüller M, Ekinci O. The Value of Clinical Decision Support in Healthcare: A Focus on Screening and Early Detection. Diagnostics (Basel) 2025; 15:648. [PMID: 40075895 PMCID: PMC11899545 DOI: 10.3390/diagnostics15050648] [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: 11/15/2024] [Revised: 02/18/2025] [Accepted: 02/24/2025] [Indexed: 03/14/2025] Open
Abstract
In a rapidly changing technology landscape, "Clinical Decision Support" (CDS) has become an important tool to improve patient management. CDS systems offer medical professionals new insights to improve diagnostic accuracy, therapy planning, and personalized treatment. In addition, CDS systems provide cost-effective options to augment conventional screening for secondary prevention. This review aims to (i) describe the purpose and mechanisms of CDS systems, (ii) discuss different entities of algorithms, (iii) highlight quality features, and (iv) discuss challenges and limitations of CDS in clinical practice. Furthermore, we (v) describe contemporary algorithms in oncology, acute care, cardiology, and nephrology. In particular, we consolidate research on algorithms across diseases that imply a significant disease and economic burden, such as lung cancer, colorectal cancer, hepatocellular cancer, coronary artery disease, traumatic brain injury, sepsis, and chronic kidney disease.
Collapse
Affiliation(s)
- Hendrik Schäfer
- Clinical Development & Medical Affairs, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland (R.S.)
- Medical Faculty, Friedrich Schiller University Jena, 07737 Jena, Germany
| | - Nesrine Lajmi
- Clinical Value & Validation, Roche Information Solutions, 2881 Scott Blvd, Santa Clara, CA 95050, USA
| | - Paolo Valente
- Clinical Development & Medical Affairs, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland (R.S.)
| | - Alessandro Pedrioli
- Clinical Value & Validation, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Daniel Cigoianu
- Clinical Development & Medical Affairs, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland (R.S.)
| | - Bernhard Hoehne
- Clinical Development & Medical Affairs, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland (R.S.)
| | - Michaela Schenk
- Quality & Regulatory Roche Information Solutions, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland
| | - Chaohui Guo
- Clinical Value & Validation, Roche Information Solutions, F. Hoffmann-La Roche Ltd., Grenzacherstrasse 124, 4070 Basel, Switzerland
| | - Ruby Singhrao
- Clinical Development & Medical Affairs, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland (R.S.)
| | - Deniz Gmuer
- Healthcare Insights, Roche Information Solutions, Roche Diagnostics International Ltd., Forrenstrasse 2, 6343 Rotkreuz, Switzerland
| | - Rezwan Ahmed
- Data, Analytics & Research, Roche Information Solutions, 2881 Scott Blvd, Santa Clara, CA 95050, USA
| | - Maximilian Silchmüller
- Medical Faculty, Friedrich Schiller University Jena, 07737 Jena, Germany
- Wiener Gesundheitsverbund, Klinik Landstraße, Juchgasse 25, 1030 Vienna, Austria
| | - Okan Ekinci
- Digital Technology & Health Information, Roche Information Solutions, 2841 Scott Blvd, Santa Clara, CA 95050, USA
- School of Medicine, University College Dublin, D04 C1P1 Dublin, Ireland
| |
Collapse
|
11
|
Mysko C, Landi S, Purssell H, Allen AJ, Prince M, Lindsay G, Rodrigues S, Irvine J, Street O, Gahloth D, MacLennan S, Piper Hanley K, Hanley N, Athwal VS. Health inequalities in hepatocellular carcinoma surveillance, diagnosis, treatment, and survival in the United Kingdom: a scoping review. BJC REPORTS 2025; 3:13. [PMID: 40033086 DOI: 10.1038/s44276-025-00126-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 12/13/2024] [Accepted: 01/31/2025] [Indexed: 03/05/2025]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a deadly cancer in the UK despite advancements in curative therapies. Societal conditions and health inequalities influence the development of chronic liver disease and outcomes from complications including HCC. Scoping this emergent evidence-base is required to inform research and solutions for the NHS. METHODS A PRISMA scoping review was performed up to September 2023. Articles exploring health inequalities in HCC involving the UK population were included. RESULTS This review has characterised axes of health inequality and their impact across the HCC care continuum in the UK. Studies predominantly employed a cohort design or population-based analyses, with meta-analyses of surveillance utilisation including only a single UK study. These methodologies provided an appropriate lens to understand longitudinal trends and identify disadvantaged groups. However, important evidence gaps remain, including exploration of patient perspectives, intersectional analyses, and statistical measures of socioeconomic inequity in HCC. CONCLUSIONS HCC is a rapidly growing cause of cancer mortality and disproportionally affects underserved groups, presenting a major public health concern. Further research is required to innovate and evaluate surveillance and management pathways to reduce systemic inequities. Direction is needed at the national level to improve prevention, early diagnosis and access to curative treatment.
Collapse
Affiliation(s)
- Christopher Mysko
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - Stephanie Landi
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - Huw Purssell
- Manchester University NHS Foundation Trust, Manchester, UK
- University of Manchester, Manchester, UK
| | - A Joy Allen
- Roche Diagnostics Limited, Welwyn Garden City, UK
| | - Martin Prince
- Manchester University NHS Foundation Trust, Manchester, UK
| | | | | | | | | | | | | | | | - Neil Hanley
- University of Birmingham, Birmingham, UK
- University Hospitals Birmingham NHS Foundation Trust, Birmingham, UK
| | - Varinder Singh Athwal
- Manchester University NHS Foundation Trust, Manchester, UK.
- University of Manchester, Manchester, UK.
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Ma Q, Ye J, Luo L, Sun Y, Wang W, Feng S, Liao B, Zhong B. Effect of potent nucleos(t)ide analog on alpha fetoprotein changes and occurrence of hepatocellular carcinoma in patients with chronic hepatitis B. Infect Agent Cancer 2025; 20:8. [PMID: 39920817 PMCID: PMC11804019 DOI: 10.1186/s13027-025-00639-1] [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: 10/07/2024] [Accepted: 01/17/2025] [Indexed: 02/09/2025] Open
Abstract
BACKGROUND Successful antiviral therapy significantly decreases the incidence of hepatocellular carcinoma (HCC) in patients with chronic hepatitis B (CHB). Alpha-fetoprotein (AFP) in the serum is a valuable early indicator of HCC. However, it is unclear whether different antiviral medications have varying effects on AFP levels. The purpose of this study was to evaluate this issue in those treated with entecavir (ETV) versus tenofovir disoproxil fumarate (TDF). METHODS We prospectively enrolled treatment-naive CHB adults who commenced treatment with ETV or TDF. Their changes in biochemical, virological, and fibrosis parameters and the elevation of AFP or development of HCC during follow-up were analyzed. RESULTS A total of 1942 CHB patients were included (10-90% follow-up time 3-60 months), and 104 patients with elevated AFP (5.3%) and 27 patients with HCC development (1.4%) were identified during the follow-up. The difference in the cumulative incidence of AFP abnormalities and HCC was statistically significant between patients who received ETV or TDF therapy. Multivariate Cox regression showed that elevated liver stiffness with shear wave elastography (Hazard ratio (HR) = 1.05, 95% Confidence interval (CI) 1.03-1.08, P < 0.001) and abnormal AFP at baseline (HR = 1.00, 95% CI 1.00-1.00, P < 0.001) were independent risk factors for abnormal AFP in CHB patients, while shear wave elastography (HR = 1.07, 95% CI 1.02-1.12, P < 0.001) was also independent risk factor for HCC. Similar results were obtained after propensity score matching (PSM) analysis. The combination of shear wave elastography (SWE), mPage-B score, age and type 2 diabetes mellitus had an area under the curve of 0.838 (P < 0.001) in predicting the occurrence of HCC. CONCLUSIONS Similar AFP elevation and HCC development rates were observed in CHB patients treated with ETV or TDF. Elevated SWE and abnormal AFP at baseline were independent risk factors for abnormal AFP in CHB patients.
Collapse
Affiliation(s)
- Qianqian Ma
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
- Department of Infectious Diseases, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510080, China
| | - Junzhao Ye
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Ling Luo
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Yanhong Sun
- Department of Clinical Laboratories, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Wei Wang
- Department of Ultrasonography, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Shiting Feng
- Department of Radiology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Bing Liao
- Department of Pathology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China
| | - Bihui Zhong
- Department of Gastroenterology, The First Affiliated Hospital of Sun Yat-Sen University, No. 58 Zhongshan II Road, Yuexiu District, Guangzhou, 510080, China.
| |
Collapse
|
15
|
El-Serag HB, Jin Q, Tayob N, Salem E, Luster M, Alsarraj A, Khaderi S, Singal AG, Marrero JA, Asrani SK, Kanwal F. HES V2.0 outperforms GALAD for detection of HCC: A phase 3 biomarker study in the United States. Hepatology 2025; 81:465-475. [PMID: 38899967 PMCID: PMC11655698 DOI: 10.1097/hep.0000000000000953] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 05/20/2024] [Indexed: 06/21/2024]
Abstract
BACKGROUND AND AIMS The original hepatocellular carcinoma early detection screening (HES) score, which combines alpha-fetoprotein (AFP) with age, alanine aminotransferase, and platelets, has better performance than AFP alone for early HCC detection. We have developed HES V2.0 by adding AFP-L3 and des-gamma-carboxy prothrombin to the score and compared its performance to GALAD and ASAP scores among patients with cirrhosis. APPROACH AND RESULTS We conducted a prospective-specimen collection, retrospective-blinded-evaluation phase 3 biomarker cohort study in patients with cirrhosis enrolled in imaging and AFP surveillance. True-positive rate (TPR)/sensitivity and false-positive rate for any or early HCC were calculated for GALAD, ASAP, and HES V2.0 scores within 6, 12, and 24 months of HCC diagnosis. We calculated the AUROC curve and estimated TPR based on an optimal threshold at a fixed false-positive rate of 10%. We analyzed 2331 patients, of whom 125 developed HCC (71% in the early stages). For any HCC, HES V2.0 had higher TPR than GALAD overall (+7.2%), at 6 months (+3.6%), at 12 months (+7.2%), and 24 months (+13.0%) before HCC diagnosis. HES V2.0 had higher TPR than ASAP for all time points (+5.9% to +12.0%). For early HCC, HES V2.0 had higher sensitivity/TPR than GALAD overall (+6.7%), at 12 months (+6.3%), and 24 months (+14.6%) but not at 6 months (+0.0%) and higher than ASAP for all time points (+13.4% to +18.0%). CONCLUSIONS In a prospective cohort study, HES V2.0 had a significantly higher performance for identifying new HCC, including early stage, than GALAD or ASAP.
Collapse
Affiliation(s)
- Hashem B. El-Serag
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Qingchun Jin
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Nabihah Tayob
- Department of Data Science, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | - Emad Salem
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Michelle Luster
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Abeer Alsarraj
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Saira Khaderi
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| | - Amit G. Singal
- Department of Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Jorge A. Marrero
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sumeet K. Asrani
- Department of Medicine, Baylor University Medical Center, Dallas, Texas, USA
| | - Fasiha Kanwal
- Section of Gastroenterology and Hepatology, Department of Medicine, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
- Section of Health Services Research, Michael E. DeBakey VA Medical Center and Baylor College of Medicine, Houston, Texas, USA
| |
Collapse
|
16
|
Singal AG, Daher D, Narasimman M, Yekkaluri S, Liu Y, Cerda V, Banala C, Khan A, Lee M, Seif El Dahan K, Murphy CC, Kramer JR, Hernaez R. Benefits and harms of hepatocellular carcinoma screening outreach in patients with cirrhosis: a multicenter randomized clinical trial. J Natl Cancer Inst 2025; 117:262-269. [PMID: 39288308 PMCID: PMC11807434 DOI: 10.1093/jnci/djae228] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/06/2024] [Accepted: 08/29/2024] [Indexed: 09/19/2024] Open
Abstract
BACKGROUND The value of hepatocellular carcinoma screening is defined by the balance of benefits from early tumor detection vs harms because of false-positive results. We evaluated the value of a mailed outreach strategy for hepatocellular carcinoma screening in patients with cirrhosis. METHODS We conducted a multicenter pragmatic randomized clinical trial comparing mailed outreach for hepatocellular carcinoma screening (n = 1436) and usual care with visit-based screening (n = 1436) among patients with cirrhosis at 3 health systems from March 2018 to September 2021. Outcomes of interest were early stage hepatocellular carcinoma detection (ie, screening benefit) and diagnostic evaluation for false-positive or indeterminate results (ie, screening harm). Screening harm was categorized as mild, moderate, and severe based on number and type of diagnostic exams. All patients were included in intention-to-screen analyses. RESULTS Of 125 patients diagnosed with hepatocellular carcinoma (67 outreach and 58 usual care), 71.2% were found at an early stage per the Milan criteria. Early tumor detection did not statistically significantly differ between the outreach and usual care arms (64.2% vs 79.3%; P = .06). The proportion of patients with physical harms also did not differ between the outreach and usual care arms (10.8% vs 10.7%; P = .95) with 5.9% in both arms having mild harms; 4.0% and 3.8%, respectively, with moderate harms; and 0.9% and 1.0%, respectively, with severe harms. CONCLUSION Most patients enrolled in hepatocellular carcinoma screening were detected at an early stage, and a minority experienced physical harms. A mailed outreach strategy did not increase early hepatocellular carcinoma detection or physical harms compared with usual care. CLINICAL TRIALS NUMBER NCT02582918 and NCT03756051.
Collapse
Affiliation(s)
- Amit G Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Darine Daher
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Manasa Narasimman
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Sruthi Yekkaluri
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Yan Liu
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - Vanessa Cerda
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Chaitra Banala
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX, USA
| | - Aisha Khan
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
| | - MinJae Lee
- O’Donnell School of Public Health, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Karim Seif El Dahan
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Caitlin C Murphy
- Department of Internal Medicine, University of Texas Southwestern Medical Center and Parkland Health Hospital System, Dallas, TX, USA
| | - Jennifer R Kramer
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX, USA
| | - Ruben Hernaez
- Center for Innovations in Quality, Effectiveness and Safety, Michael E. DeBakey Veterans Affairs Medical Center, Houston, TX, USA
- Department of Medicine, Michael E. DeBakey Veterans Affairs Medical Center and Baylor College of Medicine, Houston, TX, USA
| |
Collapse
|
17
|
Goldberg D, Wilder J, Terrault N. Health disparities in cirrhosis care and liver transplantation. Nat Rev Gastroenterol Hepatol 2025; 22:98-111. [PMID: 39482363 DOI: 10.1038/s41575-024-01003-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/02/2024] [Indexed: 11/03/2024]
Abstract
Morbidity and mortality from cirrhosis are substantial and increasing. Health disparities in cirrhosis and liver transplantation are reflective of inequities along the entire spectrum of chronic liver disease care, from screening and diagnosis to prevention and treatment of liver-related complications. The key populations experiencing disparities in health status and healthcare delivery include racial and ethnic minority groups, sexual and gender minorities, people of lower socioeconomic status and underserved rural communities. These disparities lead to delayed diagnosis of chronic liver disease and complications of cirrhosis (for example, hepatocellular carcinoma), to differences in treatment of chronic liver disease and its complications, and ultimately to unequal access to transplantation for those with end-stage liver disease. Calling out these disparities is only the first step towards implementing solutions that can improve health equity and clinical outcomes for everyone. Multi-level interventions along the care continuum for chronic liver disease are needed to mitigate these disparities and provide equitable access to care.
Collapse
Affiliation(s)
- David Goldberg
- Division of Digestive Health and Liver Diseases, University of Miami, Miami, FL, USA
| | - Julius Wilder
- Division of Gastroenterology, Duke University, Durham, NC, USA
| | - Norah Terrault
- Division of GI and Liver Diseases, University of Southern California, Los Angeles, CA, USA.
| |
Collapse
|
18
|
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
|
19
|
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
|
20
|
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
|
21
|
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
|
22
|
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
|
23
|
Xu C, Liang L, Liu G, Feng Y, Xu B, Zhu D, Jia W, Wang J, Zhao W, Ling X, Zhou Y, Ding W, Kong L. Predicting hepatocellular carcinoma outcomes and immune therapy response with ATP-dependent chromatin remodeling-related genes, highlighting MORF4L1 as a promising target. Cancer Cell Int 2025; 25:4. [PMID: 39757177 DOI: 10.1186/s12935-024-03629-2] [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: 07/04/2024] [Accepted: 12/29/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) continues to be a major cause of cancer-related death worldwide, primarily due to delays in diagnosis and resistance to existing treatments. Recent research has identified ATP-dependent chromatin remodeling-related genes (ACRRGs) as promising targets for therapeutic intervention across various types of cancer. This development offers potential new avenues for addressing the challenges in HCC management. METHODS This study integrated bioinformatics analyses and experimental approaches to explore the role of ACRRGs in HCC. We utilized data from The Cancer Genome Atlas (TCGA) and the Gene Expression Omnibus (GEO), applying machine learning algorithms to develop a prognostic model based on ACRRGs' expression. Experimental validation was conducted using quantitative real-time Polymerase Chain Reaction (qRT-PCR), Western blotting, and functional assays in HCC cell lines and xenograft models. RESULTS Our bioinformatics analysis identified four key ACRRGs-MORF4L1, HDAC1, VPS72, and RUVBL2-that serve as prognostic markers for HCC. The developed risk prediction model effectively distinguished between high-risk and low-risk patients, showing significant differences in survival outcomes and predicting responses to immunotherapy in HCC patients. Experimentally, MORF4L1 was demonstrated to enhance cancer stemness by activating the Hedgehog signaling pathway, as supported by both in vitro and in vivo assays. CONCLUSION ACRRGs, particularly MORF4L1, play crucial roles in modulating HCC progression, offering new insights into the molecular mechanisms driving HCC and potential therapeutic targets. Our findings advocate for the inclusion of chromatin remodeling dynamics in the strategic development of precision therapies for HCC.
Collapse
Affiliation(s)
- Chao Xu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Litao Liang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Guoqing Liu
- Children's Hospital of Nanjing Medical University, No. 72, Guangzhou Road, Nanjing, 210008, Jiangsu, China
| | - Yanzhi Feng
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Bin Xu
- Department of Hepatobiliary and Pancreatic Surgery, Affiliated Hospital of Qingdao University, Qingdao, China
| | - Deming Zhu
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Wenbo Jia
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Jinyi Wang
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Wenhu Zhao
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Xiangyu Ling
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China
| | - Yongping Zhou
- Department of Hepatobiliary Surgery, Wuxi No.2 People's Hospital, No. 68 Zhongshan Road, Wuxi, China.
| | - Wenzhou Ding
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| | - Lianbao Kong
- Hepatobiliary Center, The First Affiliated Hospital of Nanjing Medical University, Key Laboratory of Liver Transplantation, Chinese Academy of Medical Sciences, NHC Key Laboratory of Hepatobiliary Cancers, Nanjing, Jiangsu, China.
| |
Collapse
|
24
|
Shiha G, Hassan A, Mousa N, El-Domiaty N, Mikhail N, Gameaa R, Kobtan A, El Bassat H, Sharaf-Eldin M, Waked I, Eslam M, Soliman R. Individualized HCC surveillance using risk stratification scores in advanced fibrosis and cirrhotic HCV patients who achieved SVR: Prospective study. Aliment Pharmacol Ther 2025; 61:99-108. [PMID: 39313490 DOI: 10.1111/apt.18291] [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: 07/10/2024] [Revised: 08/09/2024] [Accepted: 09/09/2024] [Indexed: 09/25/2024]
Abstract
BACKGROUND Several HCC risk stratification scores were developed; however, none has been prospectively validated. The primary aim is to validate the clinical utility of six HCC risk scores in large prospective study of F3-4 patients achieving SVR following DAAs according to EASL guidelines. The secondary aim is to explore whether individualized risk stratification improves detection of HCC at early stages amenable to curative treatment. METHODS This prospective study included two cohorts: Egyptian Liver Research Institute and Hospital (ELRIAH) cohort of 463 chronic HCV patients with advanced liver disease (F3 and F4) achieved SVR with a follow-up every 6 months according to EASL guidelines using 6 simple HCC risk scores and Tanta cohort of 492 comparable patients where individualized surveillance intervals were tailored based on HCC risk assessments using GES score as follows: low-risk patients were followed yearly, intermediate-risk every 6 months and high-risk every 2-3 months. RESULTS All scores, except Watanabe post, successfully stratified patients into low-, intermediate- and high-risk groups, with log-rank p-value of 0.001 and Harrell's C ranging from 0.669 to 0.728. Clinical utility of these scores revealed that the highest percentage of patients classified as low risk was 42.5% using the GES, while the lowest was 8.9% using the aMAP. ELRIAH cohort, 25 patients developed HCC with 52% diagnosed at BCLC 0 and A. Tanta cohort, 35 patients developed HCC, with 80% diagnosed at BCLC 0 and A. CONCLUSION Individualized risk stratification using HCC risk scores was associated with improved early-stage detection and receipt of curative treatment.
Collapse
Affiliation(s)
- Gamal Shiha
- Hepatology and Gastroenterology Unit, Internal Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
| | - Ayman Hassan
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
- Higher Institute of Applied Medical Sciences, Mansoura, Egypt
| | - Nasser Mousa
- Tropical Medicine Department, Faculty of Medicine, Mansoura University, Mansoura, Egypt
| | - Nada El-Domiaty
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
- Endemic Medicine Department, Faculty of Medicine, Helwan University, Cairo, Egypt
| | - Nabiel Mikhail
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
- Biostatistics and Cancer Epidemiology Department, South Egypt Cancer Institute, Assiut University, Assiut, Egypt
| | - Reham Gameaa
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Abdelrahman Kobtan
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Hanan El Bassat
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Mohamed Sharaf-Eldin
- Tropical Medicine and Infectious Diseases Department, Faculty of Medicine, Tanta University, Tanta, Egypt
| | - Imam Waked
- National Liver Institute, Menofia University, Menofia, Egypt
| | - Mohamed Eslam
- Storr Liver Centre, Westmead Institute for Medical Research, Westmead Hospital and University of Sydney, Sydney, New South Wales, Australia
| | - Riham Soliman
- Egyptian Liver Research Institute and Hospital (ELRIAH), Mansoura, Egypt
- Tropical Medicine Department, Faculty of Medicine, Port Said University, Port Said, Egypt
| |
Collapse
|
25
|
Wang M, Grauzam S, Bayram MF, Dressman J, DelaCourt A, Blaschke C, Liang H, Scott D, Huffman G, Black A, Ochoa-Rios S, Lewin D, Angel PM, Drake RR, Ball L, Bethard J, Castellino S, Kono Y, Kubota N, Hoshida Y, Quirk L, Yopp A, Gopal P, Singal A, Mehta AS. Spatial omics-based machine learning algorithms for the early detection of hepatocellular carcinoma. COMMUNICATIONS MEDICINE 2024; 4:258. [PMID: 39627514 PMCID: PMC11614901 DOI: 10.1038/s43856-024-00677-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 11/12/2024] [Indexed: 12/06/2024] Open
Abstract
BACKGROUND Worldwide, hepatocellular carcinoma (HCC) is the second most lethal cancer, although early-stage HCC is amenable to curative treatment and can facilitate long-term survival. Early detection has proved difficult, as proteomics, transcriptomics, and genomics have been unable to discover suitable biomarkers. METHODS To find new biomarkers of HCC, we utilized a spatial omics N-glycan imaging method to identify altered glycosylation in cancer tissue (n = 53) and in paired serum of individuals with HCC (n = 23). Glycoproteomics identified the glycoproteins carrying these N-glycan structures, and we utilized an antibody array-based glycan imaging method to examine all the N-glycans associated with the identified glycoproteins. N-glycans from the examined glycoproteins were used to create machine learning algorithms, which were tested in a case-control sample set of 100 patients with cirrhosis and HCC and 101 matched patients with cirrhosis alone. RESULTS Spatial glycan imaging identifies thirteen branched, fucosylated, and high mannose glycans as altered in HCC tissue and in matched patient serum. Glycoproteomics identifies over 50 proteins containing these changes, of which sixteen glycoproteins were selected for further testing in an independent patient set. Algorithms using a combination of glycan and glycoproteins accurately differentiate early-stage and all HCC from cirrhosis with AUROC values of 0.88-0.97. CONCLUSIONS In conclusion, we present the development and application of a new biomarker platform, which can identify effective biomarkers for the early detection of HCC. This platform may also apply to other diseases, in which changes in N-linked glycosylation are known to occur.
Collapse
Affiliation(s)
- Mengjun Wang
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Stephane Grauzam
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
- GlycoPath, Inc, 22 WestEdge St - Suite 400, Charleston, SC, 29403, USA
| | - Muhammed Furkan Bayram
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - James Dressman
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Andrew DelaCourt
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Calvin Blaschke
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Hongyan Liang
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Danielle Scott
- GlycoPath, Inc, 22 WestEdge St - Suite 400, Charleston, SC, 29403, USA
| | - Gray Huffman
- GlycoPath, Inc, 22 WestEdge St - Suite 400, Charleston, SC, 29403, USA
| | - Alyson Black
- HTX Technologies, LLC, Chapel Hill, Chapel Hill, NC, 27516, USA
| | - Shaaron Ochoa-Rios
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - David Lewin
- Medical University of South Carolina, Department of Pathology and Laboratory Medicine, Charleston, SC, 29425, USA
| | - Peggi M Angel
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Richard R Drake
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Lauren Ball
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | - Jennifer Bethard
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA
| | | | - Yuko Kono
- University of California San Diego, Department of Medicine, Gastroenterology and Hepatology, San Diego, CA, 92103, USA
| | - Naoto Kubota
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yujin Hoshida
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Lisa Quirk
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Adam Yopp
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Purva Gopal
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Amit Singal
- University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Anand S Mehta
- Medical University of South Carolina, Department of Cell and Molecular Pharmacology, Charleston, SC, 29425, USA.
| |
Collapse
|
26
|
Kamaya A, Fetzer DT, Seow JH, Burrowes DP, Choi HH, Dawkins AA, Fung C, Gabriel H, Hong CW, Khurana A, McGillen KL, Morgan TA, Sirlin CB, Tse JR, Rodgers SK. LI-RADS US Surveillance Version 2024 for Surveillance of Hepatocellular Carcinoma: An Update to the American College of Radiology US LI-RADS. Radiology 2024; 313:e240169. [PMID: 39625378 DOI: 10.1148/radiol.240169] [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: 12/17/2024]
Abstract
In 2017, the American College of Radiology introduced the US Liver Imaging Reporting and Data System (LI-RADS) as a framework for US surveillance of patients at risk for developing hepatocellular carcinoma. This has aided in the standardization of technique, clinical reporting, patient management, data collection, and research. Emerging evidence has helped inform changes to the algorithm, now released as LI-RADS US Surveillance version 2024. The updated algorithm, the rationale for changes, and its alignment with the 2023 American Association for the Study of Liver Diseases Practice Guidance are presented.
Collapse
Affiliation(s)
- Aya Kamaya
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - David T Fetzer
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - James H Seow
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - David P Burrowes
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Hailey H Choi
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Adrian A Dawkins
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Christopher Fung
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Helena Gabriel
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Cheng William Hong
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Aman Khurana
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Kathryn L McGillen
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Tara A Morgan
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Claude B Sirlin
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Justin R Tse
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| | - Shuchi K Rodgers
- From the Department of Radiology, Stanford University, 300 Pasteur Dr, Palo Alto, CA 94304 (A. Kamaya, J.R.T.); The University of Texas Southwestern Medical Center, Dallas, Tex (D.T.F.); Department of Radiology, Royal Perth Hospital, Perth, Western Australia, Australia (J.H.S.); Department of Radiology, University of Calgary, Calgary, Alberta, Canada (D.P.B.); Department of Radiology and Biomedical Imaging, University of California at San Francisco, San Francisco, Calif (H.H.C., C.W.H.); Department of Radiology, University of Kentucky, Lexington, Ky (A.A.D.); MIC Medical Imaging, Edmonton, Alberta, Canada (C.F.); Department of Radiology, Northwestern University, Chicago, Ill (H.G.); Department of Radiology, University of California at San Diego, UC San Diego Medical Center, San Diego, Calif (A. Khurana); Liver Imaging Group, Department of Radiology, UC San Diego, San Diego, Calif (C.B.S.); Penn State Health Milton S. Hershey Medical Center, Hershey, Pa (K.L.M.); Department of Radiology, Mayo Clinic Arizona, Phoenix, Ariz (T.A.M.); and Department of Radiology, Thomas Jefferson University, Cherry Hill, NJ (S.K.R.)
| |
Collapse
|
27
|
Singal AG, Chhatwal J, Parikh N, Tapper E. Cost-Effectiveness of a Biomarker-Based Screening Strategy for Hepatocellular Carcinoma in Patients with Cirrhosis. Liver Cancer 2024; 13:643-654. [PMID: 39687038 PMCID: PMC11649260 DOI: 10.1159/000539895] [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: 02/18/2024] [Accepted: 06/16/2024] [Indexed: 12/18/2024] Open
Abstract
Introduction Given suboptimal performance of ultrasound-based surveillance for early hepatocellular carcinoma (HCC) detection in patients with cirrhosis, there is interest in alternative surveillance strategies, including blood-based biomarkers. We aimed to evaluate the cost-effectiveness of biomarker-based surveillance in patients with cirrhosis. Methods We constructed a decision-analytic model to compare ultrasound/alpha-fetoprotein (AFP) and biomarker-based surveillance strategies in 1,000,000 simulated patients with compensated cirrhosis. Model inputs for adherence, benefits, and harms of each strategy were based on literature review, and costs were derived from the Medicare fee schedule. Primary outcomes were quality-adjusted life-years (QALY) and incremental cost-effectiveness ratio (ICER) of the surveillance strategies, with cost-effectiveness assessed at a threshold of USD 150,000 per QALY. We performed sensitivity analyses for HCC incidence, test performance characteristics, surveillance adherence, and biomarker costs. Results In the base case, both ultrasound/AFP and biomarker-based surveillance were cost-effective versus no surveillance, with ICERs of USD 105,620, and USD 101,295, per QALY, respectively. Biomarker-based surveillance was also cost-effective versus ultrasound/AFP, with an ICER of USD 14,800 per QALY. Biomarker sensitivity exceeding 80%, cost below USD 210, or adherence exceeding 58% were necessary for biomarker-based screening to be cost-effective versus ultrasound/AFP. In two-way sensitivity analyses, biomarker costs were directly related with test sensitivity and adherence, whereas sensitivity and adherence were inversely related. In a probabilistic sensitivity analysis, biomarker-based screening was the most cost-effective strategy in most (65%) simulations. Conclusion Biomarker-based screening appears cost-effective for HCC screening, but results are sensitive to test sensitivity, adherence, and costs.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Jagpreet Chhatwal
- Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Neehar Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Elliot Tapper
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
28
|
Chan HLY, Hu Y, Malinowsky K, Madin K, Kroeniger K, Hou J, Sharma A. Prospective appraisal of clinical diagnostic algorithms for hepatocellular carcinoma surveillance in Chinese patients with chronic hepatitis B infection. Sci Rep 2024; 14:28996. [PMID: 39578653 PMCID: PMC11584881 DOI: 10.1038/s41598-024-80257-w] [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: 08/21/2024] [Accepted: 11/18/2024] [Indexed: 11/24/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is often detected at advanced stages among patients with hepatitis B virus (HBV), underscoring the urgency for more precise surveillance tests. Here, we compare the clinical performance of the novel - GAAD (gender [biological sex], age, alpha-fetoprotein [AFP], protein-induced by vitamin K absence-II [PIVKA-II]) and GALAD (gender [biological sex], age, AFP, Lens-culinaris AFP [AFP-L3]), PIVKA-II) algorithms to assess the utility of AFP-L3 for distinguishing HCC from benign chronic liver disease (CLD) in Chinese patients with predominantly chronic HBV infection. Eligible adults were enrolled, and biomarkers were measured using Elecsys (Cobas) or µTASWAKO assays. In total, 411 participants provided serum samples (HCC, n = 176 [early-stage, n = 110]; CLD, n = 136; specificity n = 101). HBV was the underlying disease etiology for most participants (HCC, 95%; benign CLD, 72%). For GAAD (Cobas), GALAD (Cobas), and GALAD (µTASWAKO), AUCs were 93.1% (95% CI: 90.0-96.2), 93.2% (90.0-96.3), and 92.7% (88.4-96.9) for early-stage, and 95.6% (93.6-97.6), 95.6% (93.6-97.7), and 95.8% (93.2-98.3) for all-stage HCC, versus CLD, respectively. Interestingly, both GAAD and GALAD algorithms demonstrated comparable diagnostic performance regardless of disease etiology (HBV vs. non-HBV), presence of cirrhosis, geographic region, and within pan-tumor specificity panels (p < 0.001), indicating AFP-L3 may have a negligible role in HCC surveillance.
Collapse
Affiliation(s)
- Henry L Y Chan
- The Chinese University of Hong Kong, Hong Kong, Hong Kong Special Administrative Region, China
| | - Yao Hu
- Department of Laboratory Medicine, Huashan Hospital, Shanghai Medical College, Fudan University, Shanghai, China
| | | | | | | | - Jinlin Hou
- Southern Medical University, Guangzhou, China
| | - Ashish Sharma
- Clinical Development and Medical Affairs, Roche Diagnostics International AG, Rotkreuz, Switzerland.
| |
Collapse
|
29
|
Singal AG, Narasimman M, Daher D, Yekkaluri S, Liu Y, Lee M, Cerda V, Khan A, Seif El Dahan K, Kramer J, Gopal P, Murphy C, Hernaez R. Effectiveness of mailed outreach and patient navigation to promote HCC screening process completion: a multicentre pragmatic randomised clinical trial. Gut 2024; 73:2037-2044. [PMID: 38839269 PMCID: PMC11560624 DOI: 10.1136/gutjnl-2024-332508] [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: 03/25/2024] [Accepted: 05/22/2024] [Indexed: 06/07/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) is plagued by failures across the cancer care continuum, leading to frequent late-stage diagnoses and high mortality. We evaluated the effectiveness of mailed outreach invitations plus patient navigation to promote HCC screening process completion in patients with cirrhosis. METHODS Between April 2018 and September 2021, we conducted a multicentre pragmatic randomised clinical trial comparing mailed outreach plus patient navigation for HCC screening (n=1436) versus usual care with visit-based screening (n=1436) among patients with cirrhosis at three US health systems. Our primary outcome was screening process completion over a 36-month period, and our secondary outcome was the proportion of time covered (PTC) by screening. All patients were included in intention-to-screen analyses. RESULTS All 2872 participants (median age 61.3 years; 32.3% women) were included in intention-to-screen analyses. Screening process completion was observed in 6.6% (95% CI: 5.3% to 7.9%) of patients randomised to outreach and 3.3% (95% CI: 2.4% to 4.3%) of those randomised to usual care (OR 2.05, 95% CI: 1.44 to 2.92). The intervention increased HCC screening process completion across most subgroups including age, sex, race and ethnicity, Child-Turcotte-Pugh class and health system. PTC was also significantly higher in the outreach arm than usual care (mean 37.5% vs 28.2%; RR 1.33, 95% CI: 1.31 to 1.35). Despite screening underuse, most HCC in both arms were detected at an early stage. CONCLUSION Mailed outreach plus navigation significantly increased HCC screening process completion versus usual care in patients with cirrhosis, with a consistent effect across most examined subgroups. However, screening completion remained suboptimal in both arms, underscoring a need for more intensive interventions. TRIAL REGISTRATION NUMBER NCT02582918.
Collapse
Affiliation(s)
- Amit G Singal
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Manasa Narasimman
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Darine Daher
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Sruthi Yekkaluri
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Yan Liu
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
| | - MinJae Lee
- Population and Data Sciences, UT Southwestern Medical, Dallas, Texas, USA
| | - Vanessa Cerda
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Aisha Khan
- Michael E. DeBakey Veterans Affairs Medical Center, Center for Innovations in Quality, Effectiveness and Safety and Baylor College of Medicine, Houston, Texas, USA
| | - Karim Seif El Dahan
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Jennifer Kramer
- Michael E. DeBakey Veterans Affairs Medical Center, Center for Innovations in Quality, Effectiveness and Safety and Baylor College of Medicine, Houston, Texas, USA
| | - Purva Gopal
- Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas, USA
| | - Caitlin Murphy
- School of Public Health, The University of Texas Health Science Center at Houston, Houston, Texas, USA
| | - Ruben Hernaez
- Gastroenterology and Hepatology, Baylor College of Medicine, Houston, Texas, USA
- Gastroenterology and Hepatology, Michael E. DeBakey Veterans Affairs Medical Center, Houston, Texas, USA
| |
Collapse
|
30
|
Singal AG, Kilgore KM, Shvets E, Parikh ND, Mehta N, Burak Ozbay A, Teigland C, Hafez O, Schroeder A, Yang A, Schinkel J. Impact of social determinants of health on hepatocellular carcinoma surveillance, treatment, and health care costs. Hepatol Commun 2024; 8:e0517. [PMID: 39392769 PMCID: PMC11469853 DOI: 10.1097/hc9.0000000000000517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Accepted: 07/06/2024] [Indexed: 10/13/2024] Open
Abstract
BACKGROUND The impact of clinical factors and social determinants of health on treatment patterns and health care costs among patients with HCC is unknown. METHODS Using 100% Medicare Fee-For-Service claims and a commercial multipayor claims database, we identified patients diagnosed with HCC from January 1, 2017, to December 31, 2020. Surveillance receipt was defined 12 months prior to HCC diagnosis, whereas treatment and health care costs were assessed post-HCC diagnosis. Multinomial logistic regression was used to assess the association between demographics, social determinants of health, and surveillance or HCC treatment. Multivariable generalized linear regression was used to identify factors associated with total health care costs. RESULTS Of the 32,239 patients with HCC (mean age 68 y, 67% male, 73% White), 70% received surveillance and only half (51%) received any treatment. Curative treatment receipt was higher among those with prior surveillance (24% with CT/MRI and 18% with ultrasound vs. 9% with no surveillance). Curative treatment was independently associated with HCC surveillance and inversely associated with Black race, lower education level, and diagnosis in the year 2020 (COVID-19 year). Higher health care costs were independently associated with Black race, low English proficiency, living alone, and diagnosis in 2018-2020, and inversely associated with CT/MRI-based surveillance. CONCLUSIONS Race and social determinants of health were independently associated with curative treatment receipt and health care costs. Increasing access to high-quality HCC surveillance may improve treatment receipt and reduce health disparities among patients with HCC.
Collapse
Affiliation(s)
- Amit G. Singal
- Division of Digestive and Liver Disease, UT Southwestern Medical Center, Dallas, Texas, USA
| | | | | | - Neehar D. Parikh
- Department of Internal Medicine, Division of Gastroenterology and Hepatology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Neil Mehta
- Department of Medicine, University of California San Francisco, San Francisco, California, USA
| | | | | | - Omar Hafez
- Avalere Health, Washington, District of Columbia, USA
| | - Amy Schroeder
- Avalere Health, Washington, District of Columbia, USA
| | - Audrey Yang
- Avalere Health, Washington, District of Columbia, USA
| | | |
Collapse
|
31
|
Singal AG, Parikh ND, Kanwal F, Marrero JA, Deodhar S, Page-Lester S, Lopez C, Feng Z, Tayob N. National Liver Cancer Screening Trial (TRACER) study protocol. Hepatol Commun 2024; 8:e0565. [PMID: 39495136 PMCID: PMC11537583 DOI: 10.1097/hc9.0000000000000565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Accepted: 09/11/2024] [Indexed: 11/05/2024] Open
Abstract
BACKGROUND Professional guidelines recommend HCC screening in at-risk patients using semi-annual ultrasound with or without alpha-fetoprotein (AFP); however, this strategy has limited effectiveness due to low adherence and sensitivity. Increasing data support the potential role of blood-based biomarker panels, which could improve both aspects. The biomarker panel GALAD, comprised of sex, age, and 3 blood biomarkers (AFP, AFP-L3, and des-carboxy prothrombin des-carboxy prothrombin), has shown high sensitivity and specificity in biomarker phase II (case-control) and phase III (retrospective cohort) validation studies. However, prospective validation in a large phase IV biomarker clinical utility trial is necessary before its adoption in practice. METHODS The National Liver Cancer Screening Trial is an adaptive pragmatic randomized phase IV trial, which began enrollment in January 2024, comparing ultrasound-based versus biomarker-based screening in 5500 patients with chronic hepatitis B infection or cirrhosis from any etiology. Eligible patients are randomly assigned in a 1:1 ratio to semi-annual screening with ultrasound ± alpha-fetoprotein (arm A) or semi-annual screening with GALAD (arm B). Randomization is stratified by enrollment site, liver disease severity (per Child-Pugh class), liver disease etiology (viral, nonviral, and noncirrhotic HBV), and sex. Patients are being recruited from 15 sites (a mix of tertiary care academic referral centers, safety-net health systems, and large community health systems) over a 3-year period, and the primary endpoint, reduction in late-stage HCC, will be assessed at the end of year 5.5. DISCUSSION The results of this trial will inform the best strategy for HCC screening and early-stage detection in patients with chronic liver diseases. If GALAD shows superiority, HCC screening would primarily shift from an ultrasound-based strategy to the adoption of the biomarker panel. TRIAL REGISTRATION NCT06084234. TRIAL STATUS The TRACER Study is actively enrolling.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, Michigan, USA
| | - Fasiha Kanwal
- Department of Medicine, Baylor College of Medicine, Houston, Texas, USA
| | - Jorge A. Marrero
- Department of Internal Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Sneha Deodhar
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, Texas, USA
| | - Stephanie Page-Lester
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Camden Lopez
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Ziding Feng
- Biostatistics Program, Public Health Science Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Nabihah Tayob
- Department of Data Science, Dana Farber Cancer Institute, Boston, Massachusetts, USA
| |
Collapse
|
32
|
Singal AG, Ng M, Kulkarni A. Advancing Surveillance Strategies for Hepatocellular Carcinoma: A New Era of Efficacy and Precision. J Clin Exp Hepatol 2024; 14:101448. [PMID: 38946864 PMCID: PMC11214318 DOI: 10.1016/j.jceh.2024.101448] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/01/2024] [Accepted: 05/13/2024] [Indexed: 07/02/2024] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the few cancers with a 5-year survival that has remained below 20%; however, prognosis differs by tumor stage at diagnosis. Curative treatment options among patients with early-stage HCC afford a median survival of 5-10 years. Accordingly, international society guidelines recommend semi-annual HCC surveillance in at-risk patients, including those with cirrhosis or high-risk chronic hepatitis B infection. Surveillance is associated with increased early-stage HCC detection and curative treatments, leading to reduced HCC-related mortality. Abdominal ultrasound has been the cornerstone for HCC surveillance for the past two decades, but recent data have highlighted its suboptimal sensitivity for early-stage HCC detection, particularly in patients with obesity and those with non-viral etiologies of liver disease. The combination of ultrasound plus alpha fetoprotein (AFP) has higher sensitivity for early-stage HCC detection than ultrasound alone, although the combination still misses over one-third of HCC at an early stage. Emerging imaging and blood-based biomarker strategies have promising data in biomarker phase 2 (case-control) and phase 3 (cohort) studies. Beyond ultrasound, Magnetic resonance imaging (MRI) is the best-studied imaging strategy, with superior sensitivity and specificity compared to ultrasound in a cohort study. Abbreviated MRI protocols have been proposed to address concerns about MRI radiological capacity, costs, and patient acceptance. Of biomarker strategies, GALAD (a panel including gender, age, AFP, AFP-L3, and DCP) is the best validated, with promising sensitivity for early-stage HCC detection in a national multi-center cohort study. Liquid biopsy biomarkers, including methylated DNA markers, have also shown promising accuracy in case-control studies. Abbreviated MRI and GALAD are now entering prospective trials that examine clinical outcomes such as early-stage HCC detection and screening-related harms, which are essential data to understand for adoption in clinical practice. As additional surveillance strategies become available, it will allow an era of precision surveillance in which optimal surveillance modalities are tailored to individual patient risk and expected test performance.
Collapse
Affiliation(s)
- Amit G. Singal
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Michelle Ng
- Department of Internal Medicine, UT Southwestern Medical Center, Dallas, TX, USA
| | - Anand Kulkarni
- Department of Hepatology, AIG Hospitals, Hyderabad, India
| |
Collapse
|
33
|
Singal A. Emerging Strategies for Hepatocellular Carcinoma Surveillance. Gastroenterol Hepatol (N Y) 2024; 20:560-562. [PMID: 39483999 PMCID: PMC11523088] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Affiliation(s)
- Amit Singal
- Willis C. Maddrey Distinguished Chair in Liver Disease Professor of Internal Medicine Medical Director of the Liver Tumor Program Chief of Hepatology UT Southwestern Medical Center Dallas, Texas
| |
Collapse
|
34
|
Li Y, Wang F, Geng Z, He T, Song Y, Wu J, Wang B. HBx promotes tumorigenicity through RRM2-mediated autophagy in hepatocellular carcinoma. Cell Biosci 2024; 14:116. [PMID: 39256879 PMCID: PMC11389268 DOI: 10.1186/s13578-024-01298-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Accepted: 08/27/2024] [Indexed: 09/12/2024] Open
Abstract
BACKGROUND Hepatitis B virus (HBV) infection can exacerbate liver disease progression through multiple mechanisms, eventually leading to hepatocellular carcinoma (HCC). HBV-encoded oncogene X protein (HBx), a key regulatory protein of HBV infection, serves as a positive regulator of hepatocarcinogenesis. The indispensability of the M2 subunit of ribonucleotide-diphosphate reductase (RRM2) lies in its role in facilitating DNA replication and repair processes. In our previous investigation, it was postulated that the gene RRM2 exhibits elevated expression levels in several categories of malignant tumors, particularly in HBV-related HCC. Additionally, it was observed that RRM2 is present within protein complexes that are centered on HBx. In the present investigation, the objective of this work was to investigate the potential relationship between the elevated expression of RRM2 in HBV-related HCC and the influence of HBx on this expression. The study attempted to determine the specific mechanism by which RRM2 is implicated in the promotion of hepatocarcinogenesis by HBx. There have been multiple scholarly proposals suggesting that the induction of autophagy by HBx is a significant intermediary factor in the development of HCC. However, the precise carcinogenic function of HBx-induced autophagy remains a subject of debate. RESULTS This work initially investigated the impact of suppressing cellular autophagy on the malignant biological behaviors of HBx-promoted cells using an in vitro cellular model. The findings revealed that the suppression of cellular autophagy partially disrupted the oncogenic effects of HBx. In light of this, we proceeded to conduct more investigations into the regulatory association between RRM2 and HBx-induced autophagy in the upstream-downstream context. Our data indicate that HBx proteins increase the expression of RRM2. Suppression of RRM2 expression not only hinders HBx-induced autophagy, but also worsens the cellular G1/S blockage and reduces the HBx-induced malignant growth of hepatocellular carcinoma tumors, while stimulating apoptosis. CONCLUSIONS Therefore, we hypothesised that RRM2 is a potential downstream target of HBx-induced hepatocarcinogenesis, and mining the oncogenic mechanism of RRM2 is significant in exploring the preventive treatment of HBV-related HCC.
Collapse
Affiliation(s)
- Yaqun Li
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
- Department of Pharmacy, Department of Oncology, Shanghai Medical College, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Furan Wang
- Pfizer Research China, Shanghai, 200000, China
| | - Zikai Geng
- Pharmacy School, Binzhou Medical University, Yantai, Shandong Province, 264003, China
| | - Tianye He
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Yun Song
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China
| | - Jian Wu
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| | - Bin Wang
- Department of Pharmacy, Huashan Hospital, Fudan University, Shanghai, 200040, China.
- National Clinical Research Center for Aging and Medicine, Huashan Hospital, Fudan University, Shanghai, 200040, China.
| |
Collapse
|
35
|
Chan YT, Zhang C, Wu J, Lu P, Xu L, Yuan H, Feng Y, Chen ZS, Wang N. Biomarkers for diagnosis and therapeutic options in hepatocellular carcinoma. Mol Cancer 2024; 23:189. [PMID: 39242496 PMCID: PMC11378508 DOI: 10.1186/s12943-024-02101-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 08/23/2024] [Indexed: 09/09/2024] Open
Abstract
Liver cancer is a global health challenge, causing a significant social-economic burden. Hepatocellular carcinoma (HCC) is the predominant type of primary liver cancer, which is highly heterogeneous in terms of molecular and cellular signatures. Early-stage or small tumors are typically treated with surgery or ablation. Currently, chemotherapies and immunotherapies are the best treatments for unresectable tumors or advanced HCC. However, drug response and acquired resistance are not predictable with the existing systematic guidelines regarding mutation patterns and molecular biomarkers, resulting in sub-optimal treatment outcomes for many patients with atypical molecular profiles. With advanced technological platforms, valuable information such as tumor genetic alterations, epigenetic data, and tumor microenvironments can be obtained from liquid biopsy. The inter- and intra-tumoral heterogeneity of HCC are illustrated, and these collective data provide solid evidence in the decision-making process of treatment regimens. This article reviews the current understanding of HCC detection methods and aims to update the development of HCC surveillance using liquid biopsy. Recent critical findings on the molecular basis, epigenetic profiles, circulating tumor cells, circulating DNAs, and omics studies are elaborated for HCC diagnosis. Besides, biomarkers related to the choice of therapeutic options are discussed. Some notable recent clinical trials working on targeted therapies are also highlighted. Insights are provided to translate the knowledge into potential biomarkers for detection and diagnosis, prognosis, treatment response, and drug resistance indicators in clinical practice.
Collapse
Affiliation(s)
- Yau-Tuen Chan
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Cheng Zhang
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Junyu Wu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Pengde Lu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Lin Xu
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Hongchao Yuan
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Yibin Feng
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong
| | - Zhe-Sheng Chen
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, 8000 Utopia Parkway, Queens, NY, 11439, USA.
| | - Ning Wang
- School of Chinese Medicine, The University of Hong Kong, Pok Fu Lam, Hong Kong.
| |
Collapse
|
36
|
Kao SYZ, Sangha K, Fujiwara N, Hoshida Y, Parikh ND, Singal AG. Cost-effectiveness of a precision hepatocellular carcinoma surveillance strategy in patients with cirrhosis. EClinicalMedicine 2024; 75:102755. [PMID: 39234558 PMCID: PMC11372615 DOI: 10.1016/j.eclinm.2024.102755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 09/06/2024] Open
Abstract
Background Hepatocellular carcinoma (HCC) surveillance is currently performed using a one-size-fits-all strategy with ultrasound plus AFP (US + AFP). There is increasing interest in risk-stratified and precision surveillance strategies incorporating individual risk and variance in surveillance test performance; however, the cost-effectiveness of these approaches has not been evaluated. Methods We conducted a cost-effectiveness analysis to evaluate four surveillance strategies (no surveillance, universal US + AFP surveillance, risk-stratified surveillance, and precision surveillance) in a simulated cohort of 50-year-old patients with compensated cirrhosis. The most cost-effective strategy was that with the highest incremental cost-effectiveness ratio (ICER) and below the willingness-to-pay (WTP) threshold of $150,000/QALY gained. Model inputs were based on literature review, and costs were derived from the Medicare fee schedule. Findings The precision surveillance strategy demonstrated variation in recommended surveillance test based on HCC risk category and patient factors. US + AFP, risk-stratified, and precision surveillance detected more HCC cases per 100,000 population than no surveillance, with a higher proportion of early-stage cases for precision surveillance (67.6%) than risk-stratified (63.8%), universal ultrasound (63.2%), and no surveillance (38.0%). Compared to no surveillance, precision surveillance was most cost-effective, with an ICER of $104,614/QALY gained, whereas US + AFP and risk-stratified surveillance were both dominated. Compared to US + AFP, risk-stratified surveillance was cost saving and dominated US + AFP, whereas precision surveillance was cost-effective, with an ICER of $98,103/QALY gained. Results were sensitive to survival with early-stage HCC, cost of early-stage HCC treatment, and surveillance utilization. Precision surveillance remained the most cost-effective when WTP thresholds exceeded $110,000/QALY gained. Interpretation A precision surveillance strategy is the most cost-effective method for HCC surveillance. This approach could maximize surveillance benefits in high-risk patients, while minimizing surveillance harms in low-risk individuals. Funding National Cancer Institute (U01 CA230694, R01 CA222900, R01 CA212008, and U24ca086368) and Cancer Prevention Research Institute of Texas (CPRIT) (RP200554).
Collapse
Affiliation(s)
| | | | - Naoto Fujiwara
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yujin Hoshida
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Neehar D. Parikh
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
| | - Amit G. Singal
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
37
|
Sanchez JI, Fontillas AC, Kwan SY, Sanchez CI, Calderone TL, Lee JL, Elsaiey A, Cleere DW, Wei P, Vierling JM, Victor DW, Beretta L. Metabolomics biomarkers of hepatocellular carcinoma in a prospective cohort of patients with cirrhosis. JHEP Rep 2024; 6:101119. [PMID: 39139459 PMCID: PMC11321296 DOI: 10.1016/j.jhepr.2024.101119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 05/01/2024] [Accepted: 05/08/2024] [Indexed: 08/15/2024] Open
Abstract
Background & Aims The effectiveness of surveillance for hepatocellular carcinoma (HCC) in patients with cirrhosis is limited, due to inadequate risk stratification and suboptimal performance of current screening modalities. Methods We developed a multicenter prospective cohort of patients with cirrhosis undergoing surveillance with MRI and applied global untargeted metabolomics to 612 longitudinal serum samples from 203 patients. Among them, 37 developed HCC during follow-up. Results We identified 150 metabolites with significant abundance changes in samples collected prior to HCC (Cases) compared to samples from patients who did not develop HCC (Controls). Tauro-conjugated bile acids and gamma-glutamyl amino acids were increased, while acyl-cholines and deoxycholate derivatives were decreased. Seven amino acids including serine and alanine had strong associations with HCC risk, while strong protective effects were observed for N-acetylglycine and glycerophosphorylcholine. Machine learning using the 150 metabolites, age, gender, and PNPLA3 and TMS6SF2 single nucleotide polymorphisms, identified 15 variables giving optimal performance. Among them, N-acetylglycine had the highest AUC in discriminating Cases and Controls. When restricting Cases to samples collected within 1 year prior to HCC (Cases-12M), additional metabolites including microbiota-derived metabolites were identified. The combination of the top six variables identified by machine learning (alpha-fetoprotein, 6-bromotryptophan, N-acetylglycine, salicyluric glucuronide, testosterone sulfate and age) had good performance in discriminating Cases-12M from Controls (AUC 0.88, 95% CI 0.83-0.93). Finally, 23 metabolites distinguished Cases with LI-RADS-3 lesions from Controls with LI-RADS-3 lesions, with reduced abundance of acyl-cholines and glycerophosphorylcholine-related lysophospholipids in Cases. Conclusions This study identified N-acetylglycine, amino acids, bile acids and choline-derived metabolites as biomarkers of HCC risk, and microbiota-derived metabolites as contributors to HCC development. Impact and implications The effectiveness of surveillance for hepatocellular carcinoma (HCC) in patients with cirrhosis is limited. There is an urgent need for improvement in risk stratification and new screening modalities, particularly blood biomarkers. Longitudinal collection of paired blood samples and MRI images from patients with cirrhosis is particularly valuable in assessing how early blood and imaging markers become positive during the period when lesions are observed to obtain a diagnosis of HCC. We generated a multicenter prospective cohort of patients with cirrhosis under surveillance with contrast MRI, applied untargeted metabolomics on 612 serum samples from 203 patients and identified metabolites associated with risk of HCC development. Such biomarkers may significantly improve early-stage HCC detection for patients with cirrhosis undergoing HCC surveillance, a critical step to increasing curative treatment opportunities and reducing mortality.
Collapse
Affiliation(s)
- Jessica I. Sanchez
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Antoine C. Fontillas
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Suet-Ying Kwan
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Caren I. Sanchez
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Tiffany L. Calderone
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Jana L. Lee
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - Ahmed Elsaiey
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Darrel W. Cleere
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Peng Wei
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - John M. Vierling
- Margaret M. and Albert B. Alkek Department of Medicine, Section of Gastroenterology and Hepatology, Baylor College of Medicine, Houston, TX, USA
| | - David W. Victor
- Department of Gastroenterology, Houston Methodist Hospital, Houston, TX, USA
| | - Laura Beretta
- Department of Molecular and Cellular Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| |
Collapse
|
38
|
Cumming J, Scott N, Howell J, Flores JE, Pavlyshyn D, Hellard ME, Winata LSH, Ryan M, Sutherland T, Thompson AJ, Doyle JS, Sacks-Davis R. Improving Hepatocellular Carcinoma Surveillance Outcomes in Patients with Cirrhosis after Hepatitis C Cure: A Modelling Study. Cancers (Basel) 2024; 16:2745. [PMID: 39123472 PMCID: PMC11312194 DOI: 10.3390/cancers16152745] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/29/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND & AIMS Hepatocellular carcinoma (HCC) presents a significant global health challenge, particularly among individuals with liver cirrhosis, with hepatitis C (HCV) a major cause. In people with HCV-related cirrhosis, an increased risk of HCC remains after cure. HCC surveillance with six monthly ultrasounds has been shown to improve survival. However, adherence to biannual screening is currently suboptimal. This study aimed to evaluate the effect of increased HCC surveillance uptake and improved ultrasound sensitivity on mortality among people with HCV-related cirrhosis post HCV cure. METHODS This study utilized mathematical modelling to assess HCC progression, surveillance, diagnosis, and treatment among individuals with cirrhosis who had successfully been treated for HCV. The deterministic compartmental model incorporated Barcelona Clinic Liver Cancer (BCLC) stages to simulate disease progression and diagnosis probabilities in 100 people with cirrhosis who had successfully been treated for hepatitis C over 10 years. Four interventions were modelled to assess their potential for improving life expectancy: realistic improvements to surveillance adherence, optimistic improvements to surveillance adherence, diagnosis sensitivity enhancements, and improved treatment efficacy Results: Realistic adherence improvements resulted in 9.8 (95% CI 7.9, 11.6) life years gained per cohort of 100 over a 10-year intervention period; 17.2 (13.9, 20.3) life years were achieved in optimistic adherence improvements. Diagnosis sensitivity improvements led to a 7.0 (3.6, 13.8) year gain in life years, and treatment improvements improved life years by 9.0 (7.5, 10.3) years. CONCLUSIONS Regular HCC ultrasound surveillance remains crucial to reduce mortality among people with cured hepatitis C and cirrhosis. Our study highlights that even minor enhancements to adherence to ultrasound surveillance can significantly boost life expectancy across populations more effectively than strategies that increase surveillance sensitivity or treatment efficacy.
Collapse
Affiliation(s)
- Jacob Cumming
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Population Health and Immunity, Walter and Eliza Hall Institute, Parkville, VIC 3052, Australia
| | - Nick Scott
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Jessica Howell
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Joan Ericka Flores
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
| | - Damian Pavlyshyn
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
| | - Margaret E. Hellard
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
- Department of Infectious Diseases, The Alfred and Monash University, Melbourne, VIC 3004, Australia
- Doherty Institute and School of Population and Global Health, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Leon Shin-han Winata
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
| | - Marno Ryan
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
| | - Tom Sutherland
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Alexander J. Thompson
- Department of Gastroenterology, St Vincent’s Hospital, Melbourne, VIC 3065, Australia
- Department of Medicine, University of Melbourne, Melbourne, VIC 3052, Australia
| | - Joseph S. Doyle
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Department of Infectious Diseases, The Alfred and Monash University, Melbourne, VIC 3004, Australia
| | - Rachel Sacks-Davis
- Disease Elimination Program, Burnet Institute, Melbourne, VIC 3004, Australia (J.S.D.); (R.S.-D.)
- Department of Epidemiology and Preventive Medicine, Monash University, Melbourne, VIC 3004, Australia
| |
Collapse
|
39
|
Kuzuya T, Kawabe N, Muto H, Wada Y, Komura G, Nakano T, Tanaka H, Nakaoka K, Ohno E, Funasaka K, Nagasaka M, Miyahara R, Hirooka Y. Early Changes in Alpha-Fetoprotein and Des-γ-Carboxy Prothrombin Are Useful Predictors of Antitumor Response to Durvalumab Plus Tremelimumab Therapy for Advanced Hepatocellular Carcinoma. Curr Oncol 2024; 31:4225-4240. [PMID: 39195298 PMCID: PMC11353012 DOI: 10.3390/curroncol31080315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2024] [Revised: 07/24/2024] [Accepted: 07/25/2024] [Indexed: 08/29/2024] Open
Abstract
The relationship between antitumor response and tumor marker changes was evaluated in patients with advanced hepatocellular carcinoma treated with durvalumab plus tremelimumab (Dur/Tre). Forty patients were enrolled in this retrospective evaluation of treatment outcomes. According to the Response Evaluation Criteria for Solid Tumors version 1.1 at 8 weeks, the objective response (OR) rate was 25% and the disease control (DC) rate was 57.5%. The median alpha-fetoprotein (AFP) ratio at 4 weeks was 0.39 in patients who achieved OR at 8 weeks (8W-OR group), significantly lower than the 1.08 in the non-8W-OR group (p = 0.0068); however, it was 1.22 in patients who did not achieve DC at 8 weeks (non-8W-DC group), significantly higher than the 0.53 in the 8W-DC group (p = 0.0006). Similarly, the median des-γ-carboxy-prothrombin (DCP) ratio at 4 weeks was 0.15 in the 8W-OR group, significantly lower than the 1.46 in the non-8W-OR group (p < 0.0001); however, it was 1.23 in the non-8W-DC group, significantly higher than the 0.49 in the 8W-DC group (p = 0.0215). Early changes in tumor markers after Dur/Tre initiation were associated with antitumor response. In particular, changes in AFP and DCP at 4 weeks may offer useful biomarkers for early prediction of both response and progressive disease following Dur/Tre.
Collapse
Affiliation(s)
- Teiji Kuzuya
- Department of Gastroenterology and Hepatology, Fujita Health University, 1-98 Dengakugakubo, Kutsukake-Cho, Toyoake 470-1192, Japan; (N.K.); (H.M.); (Y.W.); (G.K.); (T.N.); (H.T.); (K.N.); (E.O.); (K.F.); (M.N.); (R.M.); (Y.H.)
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Liang J, Li PY, Norman J, Lauzon M, Yeo YH, Trivedi H, Ayoub WS, Kuo A, Friedman ML, Sankar K, Gong J, Osipov A, Hendifar A, Todo T, Kim I, Voidonikolas G, Brennan TV, Wisel SA, Steggarda J, Kosari K, Saouaf R, Nissen N, Yao F, Mehta N, Yang JD. Development and validation of a biomarker index for HCC treatment response. Hepatol Commun 2024; 8:e0466. [PMID: 38896084 PMCID: PMC11186807 DOI: 10.1097/hc9.0000000000000466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/29/2024] [Indexed: 06/21/2024] Open
Abstract
BACKGROUND Serum AFP-L3%, AFP, and DCP are useful biomarkers for HCC detection, but their utility in assessing treatment response remains unknown. We aim to evaluate the accuracy of a biomarker model in the detection of posttreatment viable tumors. METHODS For model derivation, recipients with HCC undergoing liver transplant from 2018 to 2022 who had biomarkers collected within 3 months before transplant were included. We developed a generalized linear model for detecting posttreatment viable tumors with the 3 biomarkers as covariates, which we termed the "LAD Score." An independent cohort of 117 patients with HCC was used for external validation. RESULTS Among 205 recipients of transplant, 70.2% had evidence of viable tumor on explant. The median LAD score was higher among patients with viable versus nonviable tumors (1.06 vs. 0.465, p < 0.001). The LAD score had a sensitivity of 55.6% and a specificity of 85.1% at the cutoff of 0.927, which was more accurate than imaging for detecting posttreatment viable tumors (AUROC 0.736 vs. 0.643, respectively; p = 0.045). The superior performance of the LAD score over imaging is primarily driven by its greater accuracy in detecting tumors <2 cm in diameter (AUROC of the LAD score 0.721 vs. imaging 0.595, p = 0.02). In the validation data set, the LAD score had an AUROC of 0.832 (95% CI: 0.753, 0.911) with a sensitivity of 72.5% and a specificity of 89.4% at the cutoff of 0.927. CONCLUSIONS Our findings suggest the utility of LAD score in treatment response assessment after locoregional therapy for HCC, particularly in detecting small tumors. A larger prospective study is in progress to validate its accuracy and evaluate its performance in recurrence monitoring.
Collapse
Affiliation(s)
- Jeff Liang
- Department of Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Po-Yi Li
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, California, USA
| | - Joshua Norman
- Department of Internal Medicine, Stanford University, Palo Alto, California, USA
| | - Marie Lauzon
- Biostatistics and Bioinformatics Research Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yee Hui Yeo
- Department of Internal Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hirsh Trivedi
- Department of Internal Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Walid S. Ayoub
- Department of Internal Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Alexander Kuo
- Department of Internal Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Marc L. Friedman
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kamya Sankar
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jun Gong
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Arsen Osipov
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Andrew Hendifar
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Tsuyoshi Todo
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Irene Kim
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Georgios Voidonikolas
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Todd V. Brennan
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Steven A. Wisel
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Justin Steggarda
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Kambiz Kosari
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Rola Saouaf
- Department of Radiology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Nicholas Nissen
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Francis Yao
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, California, USA
- Department of Surgery, Division of Transplant Surgery, University of California, San Francisco, San Francisco, California, USA
| | - Neil Mehta
- Department of Medicine, Division of Gastroenterology, University of California, San Francisco, San Francisco, California, USA
| | - Ju Dong Yang
- Department of Internal Medicine, Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
41
|
Tang SC, Wu YY, Lin ZW, Chen QJ, Luo C, Li YT, Fu J, Zheng LF, You PH, You S, You WY, Lin KC, Zhou WP, Lin KY, Zeng YY. Prognostic implications of preoperative, postoperative, and dynamic changes of alpha-fetoprotein and des-gamma (γ)-carboxy prothrombin expression pattern for hepatocellular carcinoma after hepatic resection: a multicenter observational study. Front Oncol 2024; 14:1425292. [PMID: 38903723 PMCID: PMC11188428 DOI: 10.3389/fonc.2024.1425292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Accepted: 05/23/2024] [Indexed: 06/22/2024] Open
Abstract
BACKGROUND The utility of pre- and post-operative alpha-fetoprotein (AFP) and des-gamma (γ)-carboxy prothrombin (DCP) expression patterns and their dynamic changes as predictors of the outcome of hepatic resection for hepatocellular carcinoma (HCC) has yet to be well elucidated. METHODS From a multicenter database, AFP and DCP data during the week prior to surgery and the first post-discharge outpatient visit (within 1-2 months after surgery) were collected from patients with HCC who underwent hepatectomy. AFP-DCP expression patterns were categorized according to the number of positive tumor markers (AFP ≥ 20ng/mL, DCP ≥ 40mAU/mL), including double-negative, single-positive, and double-positive. Changes in the AFP-DCP expression patterns were delineated based on variations in the number of positive tumor markers when comparing pre- and post-operative patterns. RESULTS Preoperatively, 53 patients (8.3%), 337 patients (52.8%), and 248 patients (38.9%) exhibited double-negative, single-positive, and double-positive AFP-DCP expression patterns, respectively. Postoperatively, 463 patients (72.6%), 130 patients (20.4%), and 45 patients (7.0%) showed double-negative, single-positive, and double-positive AFP-DCP expression patterns, respectively. Survival analysis showed a progressive decrease in recurrence-free (RFS) and overall survival (OS) as the number of postoperative positive tumor markers increased (both P < 0.001). Multivariate analysis showed that postoperative AFP-DCP expression pattern, but not preoperative AFP-DCP expression pattern, was an independent risk factor for RFS and OS. Further analysis showed that for patients with positive preoperative markers, prognosis gradually improves as positive markers decrease postoperatively. In particular, when all postoperative markers turned negative, the prognosis was consistent with that of preoperative double-negative patients, regardless of the initial number of positive markers. CONCLUSIONS AFP-DCP expression patterns, particularly postoperative patterns, serve as vital sources of information for prognostic evaluation following hepatectomy for HCC. Moreover, changes in AFP-DCP expression patterns from pre- to post-operation enable dynamic prognostic risk stratification postoperatively, aiding the development of individualized follow-up strategies.
Collapse
Affiliation(s)
- Shi-Chuan Tang
- Department of Hepatopancreatobiliary Surgery, The First Clinical Medical College of Fujian Medical University and First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Ye-Ye Wu
- Department of Hepatic Surgery II, Eastern Hepatobiliary Surgery Hospital, Navy Medical University (Second Military Medical University), Shanghai, China
| | - Zhi-Wen Lin
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Qing-Jing Chen
- Department of Hepatopancreatobiliary Surgery, The First Clinical Medical College of Fujian Medical University and First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Cong Luo
- Department of Hepatopancreatobiliary Surgery, The People’s Hospital of Zizhong County, Zizhong, China
| | - Yun-Tong Li
- Department of Hepatobiliary Surgery, Zhongshan Hospital of Xiamen University, Xiamen, China
| | - Jun Fu
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Li-Fang Zheng
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Peng-Hui You
- Bioinformatics Sample Bank, Biobank in Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Song You
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Wu-Yi You
- Department of Radiation, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Ke-Can Lin
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Wei-Ping Zhou
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Navy Medical University (Second Military Medical University), Shanghai, China
| | - Kong-Ying Lin
- Department of Hepatopancreatobiliary Surgery, The First Clinical Medical College of Fujian Medical University and First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| | - Yong-Yi Zeng
- Department of Hepatopancreatobiliary Surgery, The First Clinical Medical College of Fujian Medical University and First Affiliated Hospital of Fujian Medical University, Fuzhou, China
- Department of Hepatopancreatobiliary Surgery, Mengchao Hepatobiliary Hospital of Fujian Medical University, Fuzhou, China
| |
Collapse
|
42
|
An C, Wei R, Yao W, Han W, Li W, Shi G, Wu P. Association of serum AFP trajectories and hepatocellular carcinoma outcomes after hepatic arterial infusion chemotherapy: A longitudinal, multicenter study. Cancer Med 2024; 13:e7319. [PMID: 38819606 PMCID: PMC11141330 DOI: 10.1002/cam4.7319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/16/2023] [Accepted: 12/30/2023] [Indexed: 06/01/2024] Open
Abstract
AIM This study aims to investigate α-fetoprotein (AFP) trajectories for prediction of survival outcomes after hepatic arterial infusion chemotherapy (HAIC) treatment in large hepatocellular carcinoma (HCC). METHODS From May 2014 to June 2020, 889 eligible patients with large HCC underwent HAIC were retrospectively enrolled from five hospitals. A latent class growth mixed (LCGM) model was applied to distinguish potential AFP level dynamic changing trajectories. Inverse-probability-of-treatment weighted (IPTW) analyses were performed to eliminate unmeasured confounders through marginal structural models. Multivariate Cox proportional hazard regression analyses were used to determine the overall survival (OS) in patients with large HCC. Performance of these serum markers for survival prediction was compared by areas under receiver operating characteristic analysis with the Delong test. RESULTS The median follow-up time was 23.7 (interquartile range, 3.8-115.3). A total of 1009 patients with large HCC, who underwent HAIC with AFP repeatedly measured 3-10 times, were enrolled in the study. Three distinct trajectories of these serum AFP were identified using the LCGM model: high stable (37.0%; n = 373), low stable (15.7%; n = 159), and sharp-falling (47.3%; n = 477). Multivariate Cox proportional hazard regression analyses found that ALBI stage 2-3, BCLC-C stage and high-stable AFP trajectories were associated with OS. AFP trajectories yield the optimal predictive performance in all risk factors. CONCLUSIONS The AFP trajectories based on longitudinal AFP change showed outstanding performance for predicting survival outcomes after HAIC treatment in large HCC, which provide a potential monitoring tool for improving clinical decision-making.
Collapse
Affiliation(s)
- Chao An
- Department of Minimal Invasive InterventionState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Ran Wei
- Department of Gastrointestinal SurgeryThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouChina
| | - Wang Yao
- Department of Interventional OncologyThe First Affiliated Hospital of Sun Yat‐Sen UniversityGuangzhouGuangdongChina
| | - Wenwen Han
- Department of International Radiology and Vascular SurgeryThe First Affiliated Hospital of Jinan UniversityGuangzhouChina
| | - Wang Li
- Department of Minimal Invasive InterventionState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐Sen University Cancer CenterGuangzhouChina
| | - Ge Shi
- Medical Cosmetic and Plastic Surgery Center, The Sixth Affiliated Hospital, Sun Yat‐Sen UniversityGuangzhouChina
| | - Peihong Wu
- Department of Minimal Invasive InterventionState Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat‐Sen University Cancer CenterGuangzhouChina
| |
Collapse
|
43
|
Norman JS, Mehta N. Reply to: "Enhancing HCC recurrence prediction after liver transplantation: From DCP plus AFP-L3 model to GALAD score". J Hepatol 2024; 80:e273-e274. [PMID: 38346579 DOI: 10.1016/j.jhep.2024.02.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2024] [Accepted: 02/09/2024] [Indexed: 03/26/2024]
Affiliation(s)
- Joshua S Norman
- Department of Internal Medicine, Stanford University, CA, USA
| | - Neil Mehta
- Division of Gastroenterology and Hepatology, Department of Medicine, University of California, San Francisco, USA.
| |
Collapse
|
44
|
Teufel A, Kudo M, Qian Y, Daza J, Rodriguez I, Reissfelder C, Ridruejo E, Ebert MP. Current Trends and Advancements in the Management of Hepatocellular Carcinoma. Dig Dis 2024; 42:349-360. [PMID: 38599204 DOI: 10.1159/000538815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/08/2024] [Indexed: 04/12/2024]
Abstract
BACKGROUND Hepatocellular carcinoma (HCC) remains a significant global health burden with a high mortality rate. Over the past 40 years, significant progress has been achieved in the prevention and management of HCC. SUMMARY Hepatitis B vaccination programs, the development of direct acting antiviral drugs for Hepatitis C, and effective surveillance strategies provide a profound basis for the prevention of HCC. Advanced surgery and liver transplantation along with local ablation techniques potentially offer cure for the disease. Also, just recently, the introduction of immunotherapy opened a new chapter in systemic treatment. Finally, the introduction of the BCLC classification system for HCC, clearly defining patient groups and assigning reasonable treatment options, has standardized treatment and become the basis of almost all clinical trials for HCC. With this review, we provide a comprehensive overview of the evolving landscape of HCC management and also touch on current challenges. KEY MESSAGE A comprehensive and multidisciplinary approach is crucial for effective HCC management. Continued research and clinical trials are imperative to further enhance treatment options and will ultimately reduce the global burden of this devastating disease.
Collapse
Affiliation(s)
- Andreas Teufel
- Division of Hepatology, Division of Clinical Bioinformatics, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health (CPD), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Masatoshi Kudo
- Department of Gastroenterology and Hepatology, Kindai University Faculty of Medicine, Osaka, Japan
| | - Yuquan Qian
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Jimmy Daza
- Division of Hepatology, Division of Clinical Bioinformatics, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health (CPD), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Isaac Rodriguez
- Division of Hepatology, Division of Clinical Bioinformatics, Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Clinical Cooperation Unit Healthy Metabolism, Center for Preventive Medicine and Digital Health (CPD), Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Christoph Reissfelder
- Department of Surgery, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- DKFZ-Hector Cancer Institute, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
| | - Ezequiel Ridruejo
- Hepatology Section, Department of Medicine, Center for Medical Education and Clinical Research, Buenos Aires, Argentina
| | - Matthias P Ebert
- DKFZ-Hector Cancer Institute, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Department of Medicine II, Medical Faculty Mannheim, Heidelberg University, Mannheim, Germany
- Molecular Medicine Partnership Unit, EMBL, Heidelberg, Germany
| |
Collapse
|
45
|
Qiao W, Li J, Xiong Y, Zheng J, Jin R, Hu C. GALAD score as a prognostic model for recurrence of hepatocellular carcinoma after local ablation. J Cancer Res Clin Oncol 2024; 150:241. [PMID: 38713414 PMCID: PMC11076334 DOI: 10.1007/s00432-024-05760-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2024] [Accepted: 04/22/2024] [Indexed: 05/08/2024]
Abstract
BACKGROUND Currently, the high recurrence rate still forms severe challenges in hepatocellular carcinoma (HCC) treatment. The GALAD score, including age, gender, alpha-fetoprotein (AFP), lens culinaris agglutinin-reactive AFP (AFP-L3), and des-gamma-carboxyprothrombin (DCP) was developed as a diagnostic model. However, evidence is still lacking to confirm the capability of the GALAD score to predict the recurrence of HCC. METHODS This study included 390 HCC patients after local ablation at Beijing You'an Hospital from January 1, 2018, to December 31, 2022. Firstly, the area under the receiver operating characteristic (ROC) curve (AUC) was calculated to assess the predictive capability of the GALAD score. Then, the Kaplan-Meier (KM) curve and log-rank test were used to compare the prognosis between two groups classified by GALAD score. Finally, a nomogram for high-risk patients was established by Lasso-Cox regression. It was assessed by ROC curves, calibration curves, and decision curve analysis (DCA). RESULTS The ROC curve (AUC: 0.749) and KM curve showed the GALAD score had good predictive ability and could clearly stratify patients into two groups through the risk of recurrence. Prognostic factors selected by Lasso-Cox regression contained tumor number, tumor size, and globulin. The nomogram for high-risk patients showed reliable discrimination, calibration, and clinical utility. CONCLUSION This research displayed that the GALAD score is an effective model for predicting the recurrence of HCC. Meanwhile, we found the poor prognosis of the high-risk group and created a nomogram for these patients.
Collapse
Affiliation(s)
- Wenying Qiao
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai Street, Fengtai District, Beijing, People's Republic of China
- Beijing Di'tan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, People's Republic of China
- Changping Laboratory, Beijing, People's Republic of China
| | - Jiashuo Li
- Beijing Di'tan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, People's Republic of China
| | - Yiqi Xiong
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai Street, Fengtai District, Beijing, People's Republic of China
| | - Jiasheng Zheng
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai Street, Fengtai District, Beijing, People's Republic of China.
| | - Ronghua Jin
- Beijing Di'tan Hospital, Capital Medical University, 8 Jingshun East Street, Chaoyang District, Beijing, People's Republic of China.
- Changping Laboratory, Beijing, People's Republic of China.
| | - Caixia Hu
- Interventional Therapy Center for Oncology, Beijing You'an Hospital, Capital Medical University, 8 Xitoutiao, Youanmenwai Street, Fengtai District, Beijing, People's Republic of China.
| |
Collapse
|
46
|
Lai Q, Ito T, Iesari S, Ikegami T, Nicolini D, Larghi Laureiro Z, Rossi M, Vivarelli M, Yoshizumi T, Hatano E, Lerut J. Role of protein induced by vitamin-K absence-II in transplanted patients with HCC not producing alpha-fetoprotein. Liver Transpl 2024; 30:472-483. [PMID: 37729520 DOI: 10.1097/lvt.0000000000000259] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2023] [Accepted: 08/30/2023] [Indexed: 09/22/2023]
Abstract
Elevated Protein Induced by Vitamin-K Absence-II (PIVKA-II) has been shown to be an adverse prognostic factor in HCC patients undergoing liver transplantation (LT). No definitive data are available about the impact of PIVKA-II concerning post-LT recurrence in patients not secreting (≤ 20 ng/mL) alpha-fetoprotein (AFP). An observational retrospective study of the East-West HCC-LT consortium is reported. Between 2000 and 2019, 639 HCC patients were enrolled in 5 collaborative European and Japanese centers. To minimize the initial selection bias, an inverse probability therapy weighting method was adopted to analyze the data. In the post-inverse probability therapy weighting population, PIVKA-II (HR = 2.00; 95% CI: 1.52-2.64; p < 0.001) and AFP (HR=1.82; 95% CI: 1.48-2.24; p < 0.001) were the most relevant independent risk factors for post-LT recurrence. A sub-analysis focusing only on patients who are AFP non-secreting confirmed the negative role of PIVKA-II (HR=2.06, 95% CI: 1.26-3.35; p =0.004). When categorizing the entire population into 4 groups according to the AFP levels (≤ or > 20 ng/mL) and PIVKA (≤ or > 300 mUA/mL) at the time of LT, the lowest recurrence rates were observed in the low AFP-PIVKA-II group (5-year recurrence rate = 8.0%). Conversely, the high AFP-PIVKA-II group had the worst outcome (5-year recurrence rate = 35.1%). PIVKA-II secretion is a relevant risk factor for post-LT HCC recurrence. The role of this marker is independent of the AFP status. Combining both tumor markers, especially in the setting of LT, should be of great relevance for adding information about predicting the post-LT risk of tumor recurrence and selecting these patients for transplantation.
Collapse
Affiliation(s)
- Quirino Lai
- Department of General and Specialistic Surgery, General Surgery and Organ Transplantation Unit, Sapienza University of Rome, Rome, Italy
| | - Takashi Ito
- Department of Surgery, Kyoto University, Kyoto, Japan
| | - Samuele Iesari
- Department of Surgery, Universitè catholique de Louvain, Brussels, Belgium
| | - Toru Ikegami
- Department of Surgery and Science, Kyushu University, Fukuoka, Japan
| | - Daniele Nicolini
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, Ancona, Italy
| | - Zoe Larghi Laureiro
- Department of General and Specialistic Surgery, General Surgery and Organ Transplantation Unit, Sapienza University of Rome, Rome, Italy
| | - Massimo Rossi
- Department of General and Specialistic Surgery, General Surgery and Organ Transplantation Unit, Sapienza University of Rome, Rome, Italy
| | - Marco Vivarelli
- Department of Experimental and Clinical Medicine, Polytechnic University of Marche, Ancona, Italy
| | | | | | - Jan Lerut
- Institute for Experimental and Clinical Research IREC-Université catholique de Louvain, Brussels, Belgium
| |
Collapse
|
47
|
Yeo YH, Lee YT, Tseng HR, Zhu Y, You S, Agopian VG, Yang JD. Alpha-fetoprotein: Past, present, and future. Hepatol Commun 2024; 8:e0422. [PMID: 38619448 PMCID: PMC11019827 DOI: 10.1097/hc9.0000000000000422] [Citation(s) in RCA: 24] [Impact Index Per Article: 24.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 01/29/2024] [Indexed: 04/16/2024] Open
Abstract
Alpha-fetoprotein (AFP) is a glycoprotein that plays an important role in immune regulation with critical involvement in early human development and maintaining the immune balance during pregnancy. Postfetal development, the regulatory mechanisms controlling AFP undergo a shift and AFP gene transcription is suppressed. Instead, these enhancers refocus their activity to maintain albumin gene transcription throughout adulthood. During the postnatal period, AFP expression can increase in the setting of hepatocyte injury, regeneration, and malignant transformation. It is the first oncoprotein discovered and is routinely used as part of a screening strategy for HCC. AFP has been shown to be a powerful prognostic biomarker, and multiple HCC prognosis models confirmed the independent prognostic utility of AFP. AFP is also a useful predictive biomarker for monitoring the treatment response of HCC. In addition to its role as a biomarker, AFP plays important roles in immune modulation to promote tumorigenesis and thus has been investigated as a therapeutic target in HCC. In this review article, we aim to provide an overview of AFP, encompassing the discovery, biological role, and utility as an HCC biomarker in combination with other biomarkers and how it impacts clinical practice and future direction.
Collapse
Affiliation(s)
- Yee Hui Yeo
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Yi-Te Lee
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Hsian-Rong Tseng
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
| | - Yazhen Zhu
- Department of Molecular and Medical Pharmacology, California NanoSystems Institute, Crump Institute for Molecular Imaging, University of California, Los Angeles, California, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, California, USA
- Department of Pathology and Laboratory Medicine, Ronald Reagan Medical Center, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Sungyong You
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Department of Surgery, Division of Cancer Biology and Therapeutics, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Vatche G. Agopian
- Department of Surgery, David Geffen School of Medicine, University of California, Los Angeles, California, USA
| | - Ju Dong Yang
- Karsh Division of Gastroenterology and Hepatology, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Samuel Oschin Comprehensive Cancer Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Comprehensive Transplant Center, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
48
|
Mendiratta-Lala M, Fetzer D, Kamaya A, Parikh ND, Singal AG. The Future Role of Abdominal US in Hepatocellular Carcinoma Surveillance. Radiology 2024; 311:e232624. [PMID: 38742973 PMCID: PMC11140528 DOI: 10.1148/radiol.232624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Revised: 12/16/2023] [Accepted: 12/26/2023] [Indexed: 05/16/2024]
Abstract
Abdominal US is currently the best-validated surveillance strategy for hepatocellular carcinoma (HCC) in at-risk patients. It is the only modality shown to have completed all five phases of validation and can achieve high sensitivity and specificity for HCC detection, especially when conducted by expert sonographers in high-volume centers. However, US also has limitations, including operator dependency and varying sensitivity in clinical practice. Further, the sensitivity of US for early-stage HCC detection is lower in patients with obesity or nonviral liver disease, increasingly common populations undergoing surveillance. Imaging-based and blood-based surveillance strategies, including abbreviated MRI and biomarker panels, may overcome some limitations of US-based surveillance. Both strategies have promising test performance in phase II and phase III biomarker studies and are undergoing prospective validation. Considering the variation in HCC risk and test performance between patients, there will likely be a shift away from a one-size-fits-all approach and toward precision screening, in which the "best" test is selected based on individual patient characteristics. In this upcoming era of precision HCC screening among patients with cirrhosis, US will likely continue to have an important, albeit reduced, surveillance role.
Collapse
Affiliation(s)
| | | | - Aya Kamaya
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| | - Neehar D. Parikh
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| | - Amit G. Singal
- From the Departments of Radiology (M.M.L.) and Internal Medicine
(N.D.P.), University of Michigan, Ann Arbor, Mich; Department of Radiology
(D.F.) and Department of Internal Medicine, Division of Digestive and Liver
Diseases (A.G.S.), University of Texas Southwestern Medical Center, 5959 Harry
Hines Blvd, Ste 420, POB 1, Dallas, TX 75390-8887; and Department of Radiology,
Stanford University School of Medicine, Stanford, Calif (A.K.)
| |
Collapse
|
49
|
Huang DQ, Tran S, Barnett S, Zou B, Yeo YH, Cheung R, Nguyen MH. Incidence and predictors of hepatocellular carcinoma in NAFLD without diagnosed cirrhosis: a nationwide real-world U.S. study. Hepatol Int 2024; 18:540-549. [PMID: 38079023 DOI: 10.1007/s12072-023-10616-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2023] [Accepted: 11/05/2023] [Indexed: 04/14/2024]
Abstract
BACKGROUND A substantial proportion of patients with nonalcoholic fatty liver disease (NAFLD)-related hepatocellular carcinoma (HCC) do not have cirrhosis. Data regarding the incidence and predictors of HCC development in NAFLD without cirrhosis are limited. We conducted a large, national study of NAFLD patients without documented cirrhosis to examine the incidence and predictors for HCC development. METHODS This retrospective study included 751,603 NAFLD patients (54% female) without documented cirrhosis derived from the deidentified Optum Clinformatics® Data Mart Database. Patients with cirrhosis, platelets < 120,000/µL or FIB-4 values > 2.67 were excluded. RESULTS The mean age was 53.7 ± 15.0 years, 45.9% were male, 39.5% had diabetes, 57.6% were White, 18.4% Hispanic, 8.2% Black and 4.9% were Asian. The mean platelet count was 264,000 ± 72,000/µL, and 96.3% of patients had a FIB-4 < 1.30. Over 1,686,607 person-years of follow-up, there were 76 incident cases of HCC, resulting in an HCC incidence rate of 0.05 per 1000 person-years. There was a higher HCC incidence rate among patients with platelets ≤ 150,000/µL, versus those with platelets > 150,000/µL (0.23 per 1000 person-years, vs. 0.04 per 1000 person-years, p = 0.02) but not in subgroup analyses for age, sex, race/ethnicity or diabetes. Using multivariable Cox proportional hazards model adjusted multiple confounders, platelet count ≤ 150,000/µL remained an independent predictor of HCC development (adjusted HR 5.80, 95% CI 1.67-20.1, p = 0.006). CONCLUSION HCC incidence in NAFLD without documented cirrhosis was below the threshold for cost-effective HCC surveillance in overall and multiple subgroup analyses. Platelet count < 150,000/µL may be a useful predictor of HCC development in this population.
Collapse
Affiliation(s)
- Daniel Q Huang
- Department of Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Division of Gastroenterology and Hepatology, National University Health System, Singapore, Singapore
| | - Sally Tran
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA, 94304, USA
| | - Scott Barnett
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA, 94304, USA
| | - Biyao Zou
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA, 94304, USA
| | - Yee Hui Yeo
- Division of General Internal Medicine, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Ramsey Cheung
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA, 94304, USA
- Division of Gastroenterology and Hepatology, Veterans Affairs Palo Alto Healthcare System, Palo Alto, CA, USA
| | - Mindie H Nguyen
- Division of Gastroenterology and Hepatology, Stanford University Medical Center, 780 Welch Road, Palo Alto, CA, 94304, USA.
- Department of Epidemiology and Population Health, Stanford University, Palo Alto, CA, USA.
| |
Collapse
|
50
|
deLemos AS, Zhao J, Patel M, Kooken B, Mathur K, Nguyen HM, Mazhar A, McCarter M, Burney H, Kettler C, Chalasani N, Gawrieh S. Lean body mass index is a marker of advanced tumor features in patients with hepatocellular carcinoma. World J Hepatol 2024; 16:393-404. [PMID: 38577534 PMCID: PMC10989303 DOI: 10.4254/wjh.v16.i3.393] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Revised: 12/31/2023] [Accepted: 02/23/2024] [Indexed: 03/27/2024] Open
Abstract
BACKGROUND Obesity is an independent risk factor for the development of hepatocellular carcinoma (HCC) and may influence its outcomes. However, after diagnosis of HCC, like other malignancies, the obesity paradox may exist where higher body mass index (BMI) may in fact confer a survival benefit. This is frequently observed in patients with advanced HCC and cirrhosis, who often present late with advanced tumor features and cancer related weight loss. AIM To explore the relationship between BMI and survival in patients with cirrhosis and HCC. METHODS This is a retrospective cohort study of over 2500 patients diagnosed with HCC between 2009-2019 at two United States academic medical centers. Patient and tumor characteristics were extracted manually from medical records of each institutions' cancer registries. Patients were stratified according to BMI classes: < 25 kg/m2 (lean), 25-29.9 kg/m2 (overweight), and > 30 kg/m2 (obese). Patient and tumor characteristics were compared according to BMI classification. We performed an overall survival analysis using Kaplan Meier by the three BMI classes and after adjusting for Milan criteria. A multivariable Cox regression model was then used to assess known risk factors for survival in patients with cirrhosis and HCC. RESULTS A total of 2548 patients with HCC were included in the analysis of which 11.2% (n = 286) were classified as non-cirrhotic. The three main BMI categories: Lean (n = 754), overweight (n = 861), and obese (n = 933) represented 29.6%, 33.8%, and 36.6% of the total population overall. Within each BMI class, the non-cirrhotic patients accounted for 15% (n = 100), 12% (n = 94), and 11% (n = 92), respectively. Underweight patients with a BMI < 18.5 kg/m2 (n = 52) were included in the lean cohort. Of the obese cohort, 42% (n = 396) had a BMI ≥ 35 kg/m2. Out of 2262 patients with cirrhosis and HCC, 654 (29%) were lean, 767 (34%) were overweight, and 841 (37%) were obese. The three BMI classes did not differ by age, MELD, or Child-Pugh class. Chronic hepatitis C was the dominant etiology in lean compared to the overweight and obese patients (71%, 62%, 49%, P < 0.001). Lean patients had significantly larger tumors compared to the other two BMI classes (5.1 vs 4.2 vs 4.2 cm, P < 0.001), were more likely outside Milan (56% vs 48% vs 47%, P < 0.001), and less likely to undergo transplantation (9% vs 18% vs 18%, P < 0.001). While both tumor size (P < 0.0001) and elevated alpha fetoprotein (P < 0.0001) were associated with worse survival by regression analysis, lean BMI was not (P = 0.36). CONCLUSION Lean patients with cirrhosis and HCC present with larger tumors and are more often outside Milan criteria, reflecting cancer related cachexia from delayed diagnosis. Access to care for hepatitis C virus therapy and liver transplantation confer a survival benefit, but not overweight or obese BMI classifications.
Collapse
Affiliation(s)
| | - Jing Zhao
- Center for Outcomes Research and Evaluation, Atrium Health, Charlotte, NC 28204, United States
| | - Milin Patel
- Department of Medicine, Atrium Health, Charlotte, NC 28204, United States
| | - Banks Kooken
- Department of Medicine, Atrium Health, Charlotte, NC 28204, United States
| | - Karan Mathur
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Hieu Minh Nguyen
- Center for Health System Sciences (CHASSIS), Atrium Health, Charlotte, NC 28204, United States
| | - Areej Mazhar
- Department of Medicine, Northwestern University Feinberg School of Medicine, Chicago, IL 60611, United States
| | - Maggie McCarter
- Center for Outcomes Research and Evaluation, Atrium Health, Charlotte, NC 28204, United States
| | - Heather Burney
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Carla Kettler
- Department of Biostatistics and Health Data Science, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Naga Chalasani
- Department of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| | - Samer Gawrieh
- Department of Gastroenterology and Hepatology, Indiana University School of Medicine, Indianapolis, IN 46202, United States
| |
Collapse
|