1
|
Singh SP, Kumar K, Kulkarni A, Arora V, Choudhury A, Chaubal A, Rathi S, Shah S, Taneja S, Kumar A, Duseja A, Kumar G, Nagaraja Rao P, Saraswat V, Sarin SK. Predictors of Non-response to Atezolizumab Plus Bevacizumab in Patients With Unresectable Hepatocellular Carcinoma: A Multicentre Real World Study (HCC-AB Study). J Clin Exp Hepatol 2025; 15:102513. [PMID: 40129631 PMCID: PMC11930068 DOI: 10.1016/j.jceh.2025.102513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Accepted: 02/04/2025] [Indexed: 03/26/2025] Open
Abstract
Background The approved immunotherapies for patients with advanced HCC are Atezolizumab and Bevacizumab. However, patients in India present late and healthcare is often available through self-financing. To rationalise the therapy, we conducted a large multicentre study to identify the baseline predictors of non-response to atezolizumab and bevacizumab in advanced unresectable HCC. Methods A dose of atezolizumab 1200 mg and bevacizumab 15 mg/kg was used every 3 weeks from 6 centres across India. A total of 278 patients were screened, and 160 were included in the study. The study included patients with locally advanced metastatic or inoperable hepatocellular carcinoma who were at least 18 years of age and those who received <3 injections were excluded. Fifty-four percent of the included patients were BCLC-B and 46% were BCLC-C. The primary objective was to study overall survival and progression-free survival. While identifying radiological response, objective response rate and adverse effects were secondary objectives. Results The mean age was 61.9 ± 11.7 years, 88% were male, 55% had NASH, 16.3% had hepatitis C, 18.8% had hepatitis B and the rest were alcohol. The mean Model for End-Stage Liver Disease (MELD) is 12.05 ± 4.46, Albumin-Bilirubin Score (ALBI) is -2.04 ± 0.57. Fifty-five percent received first-line and 45% as second/other line therapy. The median overall survival was 10 (95% confidence interval [CI]: 6.1-15.6) months. Progression-free survival was found to be 8 (95%CI: 5.1-14.7) months overall. Eleven (6.9%) achieved complete response, 28 (17.5%) partial response, 33 (20.6%) had stable disease and 88 (55%) had progressive disease. On multivariate analysis, CRP>1 mg/dl (P-0.007), PIVKA-II>400 mAU/mL (P-0.019), AFP>100 ng/ml (P-0.009), presence of diabetes (P-0.042) were associated with non-response to atezolizumab and bevacizumab injection. Fifty-three percent of patients developed any grade of adverse effect, and 20% developed grade 3/4 adverse events amounting to the stoppage of therapy. Conclusion Non-response to atezolizumab and bevacizumab immunotherapy was predicted by CRP>1 mg/dl, PIVKA-II>400mAU/ml, AFP>100 ng/ml and the presence of diabetes.
Collapse
Affiliation(s)
- Satender P. Singh
- Department of Hepatology, Institute of Liver and Biliary Sciences, Vasant Kunj, New Delhi, India
| | - Karan Kumar
- Department of Hepatology and Liver Transplant, Mahatma Gandhi Hospital, Jaipur, India
| | - Anand Kulkarni
- Department of Hepatology and Liver Transplant, Asian Institute of Gastroenterology, Hyderabad, India
| | - Vinod Arora
- Department of Hepatology, Institute of Liver and Biliary Sciences, Vasant Kunj, New Delhi, India
| | - Ashok Choudhury
- Department of Hepatology, Institute of Liver and Biliary Sciences, Vasant Kunj, New Delhi, India
| | - Alisha Chaubal
- Department of Hepatology and Liver Intensive Care, Global Hospitals, Mumbai, India
| | - Sahaj Rathi
- Department of Hepatology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Samir Shah
- Department of Hepatology and Liver Intensive Care, Global Hospitals, Mumbai, India
| | - Sunil Taneja
- Department of Hepatology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Ashish Kumar
- Department of Gastroenterology and Hepatology, Sir Ganga Ram Hospital, New Delhi, India
| | - Ajay Duseja
- Department of Hepatology, Post Graduate Institute of Medical Education and Research, Chandigarh, India
| | - Guresh Kumar
- Department of Hepatology, Institute of Liver and Biliary Sciences, Vasant Kunj, New Delhi, India
| | - P. Nagaraja Rao
- Department of Hepatology and Liver Transplant, Asian Institute of Gastroenterology, Hyderabad, India
| | - Vivek Saraswat
- Department of Hepatology and Liver Transplant, Mahatma Gandhi Hospital, Jaipur, India
| | - Shiv K. Sarin
- Department of Hepatology, Institute of Liver and Biliary Sciences, Vasant Kunj, New Delhi, India
| |
Collapse
|
2
|
Wang A, Mizejewski GJ, Zhang C. Growth inhibitory peptides: a potential novel therapeutic approach to cancer treatment. Eur J Pharmacol 2025; 996:177554. [PMID: 40147579 DOI: 10.1016/j.ejphar.2025.177554] [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: 01/02/2025] [Revised: 03/02/2025] [Accepted: 03/24/2025] [Indexed: 03/29/2025]
Abstract
Cancer remains a major global public health concern, with considerable interest in exploring biological molecules for cancer treatment and prevention. Growth inhibitory peptide (GIP), a promising new class of biological therapeutics, has drawn attention for its distinct anti-tumor properties. Derived from human alpha-fetoprotein (HAFP), this synthetic 34-amino-acid peptide has demonstrated substantial anti-tumor effects across various cancer cell lines, effectively inhibiting tumor cell proliferation, migration, and metastasis. Studies reveal that GIP mediates its effects through a range of mechanisms, including interactions with G protein-coupled receptors (GPCRs), anti-cell adhesion activities, inhibition of cell spreading and metastatic processes, morphological alterations, platelet aggregation inhibition, immune enhancement, cell membrane disruption, ion channel blockade, and cell cycle arrest. While GIP has exhibited promising anti-tumor activity in both in vitro and in vivo models, further investigation is essential to advance its development as a therapeutic drug, particularly regarding pharmacokinetics, safety profiles, storage stability, and clinical efficacy.
Collapse
Affiliation(s)
- Aixin Wang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China
| | - G J Mizejewski
- Division of Translational Medicine, Molecular Diagnostics Laboratory, Wadsworth Center, New York State Department of Health Biggs Laboratory, Empire State Plaza Albany, NY 12237, USA
| | - Chao Zhang
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China; Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, PR China.
| |
Collapse
|
3
|
Wang MD, Wang BD, Diao YK, Li C, Yao LQ, Liu H, Zeng YY, Chen Z, Wu H, Xu XF, Gu LH, Xu JH, Yin DX, Li YC, Chen FJ, Kow AWC, Pawlik TM, Shen F, Yang T. Tumor biology characteristics score based on alpha-fetoprotein and protein induced by vitamin K absence or antagonist II as a predictor for recurrence and survival after curative resection for hepatocellular carcinoma: a multicenter cohort study. J Gastrointest Surg 2025; 29:102038. [PMID: 40157713 DOI: 10.1016/j.gassur.2025.102038] [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: 01/25/2025] [Revised: 03/03/2025] [Accepted: 03/22/2025] [Indexed: 04/01/2025]
Abstract
BACKGROUND Current hepatocellular carcinoma (HCC) staging systems lack comprehensive assessment of tumor biologic characteristics. This study aimed to develop and validate a tumor biology characteristics score (TBCS) based on alpha-fetoprotein (AFP) and protein induced by vitamin K absence or antagonist II (PIVKA-II) to predict long-term oncologic outcomes after HCC resection. METHODS In this multicenter retrospective cohort study, patients who underwent curative resection for HCC between June 2018 and December 2022 were included. TBCS (range, 2-6 points) was calculated by combining preoperative AFP (<20, 20-199, and ≥200 ng/mL) and PIVKA-II levels (<40, 40-399, and ≥400 mAU/mL). Patients were stratified into low (2 points), medium (3-4 points), and high TBCS groups (5-6 points). The primary outcomes were recurrence-free survival (RFS) and overall survival (OS). RESULTS A total of 695 patients were analyzed; the low, medium, and high TBCS groups comprised 132 (19.0%), 233 (33.5%), and 330 patients (47.5%), respectively. Notably, 5-year RFS was 30.4%, 14.7%, and 9.7%, whereas 5-year OS was 42.1%, 35.5%, and 23.5% for low, medium, and high TBCS groups, respectively (both P <.001). Multivariate analysis identified TBCS as an independent predictor of both RFS (medium TBCS: hazard ratio [HR], 1.583; 95% CI, 1.219-2.057; P =.001; high TBCS: HR, 1.895; 95% CI, 1.473-2.438; P <.001) and OS (high TBCS: HR, 1.781; 95% CI, 1.353-2.343; P <.001). CONCLUSION The novel TBCS combining AFP and PIVKA-II effectively stratified patients with HCC into distinct prognostic groups after curative-intent resection, independently predicting both RFS and OS. This score may help identify high-risk patients for more intense postoperative recurrence surveillance and receipt of adjuvant therapies.
Collapse
Affiliation(s)
- Ming-Da Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Bai-Dong Wang
- Second Military Medical University (Naval Medical University), Shanghai, China
| | - Yong-Kang Diao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Lan-Qing Yao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Han Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Centre, First Hospital of Jilin University, Changchun, Jilin, China
| | - Yong-Yi Zeng
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fujian, China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
| | - Han Wu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Xin-Fei Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Li-Hui Gu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Jia-Hao Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China
| | - Dong-Xu Yin
- School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China
| | - Yu-Chen Li
- Department of Graduate, Bengbu Medical University, Bengbu, Anhui, China
| | - Fu-Jie Chen
- Department of Graduate, Bengbu Medical University, Bengbu, Anhui, China
| | - Alfred Wei Chieh Kow
- Division of Hepatopancreaticobiliary Surgery and Liver Transplantation, Department of Surgery, National University Health System, Singapore, Singapore
| | - Timothy M Pawlik
- Department of Surgery, Ohio State University, Wexner Medical Centre, Columbus, OH, United States
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Naval Medical University, Shanghai, China.
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, China; School of Public Health, Hangzhou Medical College, Hangzhou, Zhejiang, China; Department of Graduate, Bengbu Medical University, Bengbu, Anhui, China; Eastern Hepatobiliary Clinical Research Institute, Third Affiliated Hospital of Naval Medical University, Shanghai, China.
| |
Collapse
|
4
|
Niaki NM, Hatefnia F, Heidari MM, Tabean M, Mobed A. Alpha-Fetoprotein (AFP) biosensors. Clin Chim Acta 2025; 573:120293. [PMID: 40216053 DOI: 10.1016/j.cca.2025.120293] [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: 03/15/2025] [Revised: 04/08/2025] [Accepted: 04/08/2025] [Indexed: 04/25/2025]
Abstract
Alpha-fetoprotein (AFP) is a glycoprotein mainly produced during fetal development, and elevated levels in adults are frequently associated with liver diseases, especially hepatocellular carcinoma (HCC), as well as certain germ cell tumors. Measuring AFP in biological samples is crucial for early diagnosis, monitoring disease progression, and evaluating treatment efficacy. While traditional detection methods like enzyme-linked immunosorbent assay (ELISA) and radioimmunoassay are dependable, they often face limitations such as lengthy processes, complexity, and the need for specialized equipment. In recent years, biosensing technologies have emerged as promising alternatives for detecting AFP, offering advantages like increased sensitivity, real-time monitoring, and ease of use. Various biosensing platforms, including electrochemical, optical, and piezoelectric sensors, have been developed to enable quick and specific detection of AFP. These sensors employ molecular recognition elements, such as antibodies, aptamers, or nanoparticles, to selectively bind AFP, producing a measurable signal. This article explores the structure and mechanisms of action of AFP, the diseases linked to it, and describes several biosensing technologies. It also reviews recent advancements in AFP biosensing, discussing their principles, performance, and potential applications in clinical settings. Furthermore, the article highlights the challenges and future prospects for developing cost-effective, portable, and multiplexed AFP biosensors, underscoring their potential to revolutionize early disease detection and personalized healthcare.
Collapse
Affiliation(s)
- Nava Moghadasian Niaki
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Faezeh Hatefnia
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Mohammad Mahdi Heidari
- Department of Pediatrics, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Mahsa Tabean
- Women's Reproductive Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
| | - Ahmad Mobed
- Social Determinants of Health Research Center, Health Management and Safety Promotion, Iran.
| |
Collapse
|
5
|
da Fonseca LG, Piñero F, Anders M, Bermudez C, Demirdjian E, Varón A, Perez D, Rodriguez J, Beltrán O, Ridruejo E, Caballini P, Araujo A, Florez JDT, Marín JI, Villa M, Orozco F, Poniachik J, Marciano S, Bessone F, Mendizabal M. Immune-mediated adverse events following atezolizumab and bevacizumab in a multinational Latin American cohort of unresectable hepatocellular carcinoma. Oncotarget 2025; 16:348-360. [PMID: 40387836 PMCID: PMC12088043 DOI: 10.18632/oncotarget.28721] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Accepted: 04/28/2025] [Indexed: 05/20/2025] Open
Abstract
AIMS Latin America has been underrepresented in trials evaluating immunotherapy for hepatocellular carcinoma (HCC). We aimed to describe the incidence of immune-related adverse events (irAEs) and their impact on outcomes in a Latin American cohort. METHODS A multicenter prospective study was conducted in Argentina, Brazil, Chile, and Colombia, including patients who received atezolizumab plus bevacizumab. A time-covarite proportional hazard analysis evaluated the effect of irAEs. RESULTS 99 patients were included. The median treatment duration was 6 months, with a median survival of 17.0 months (95% CI: 12.6-19.8). The irAE incidence rate was 2.1 cases per 100 persons-months (cumulative incidence 18.1% (95% CI: 11.1-27.2%)). Median time to irAE was 2.3 months (range 1.4-4.8), most frequently hepatitis (n = 6), thyroiditis (n = 5), and 8/18 required steroids. Follow-up, treatment duration, and overall survival were similar regardless of the occurrence of irAEs (HR = 1.71, 95% CI: 0.76-3.86; P = 0.19). Baseline alpha-feto protein ≥400 ng/ml (HR: 2.9 (95% CI: 1.1-7.6)) was independently associated with irAE. CONCLUSION The incidence of irAEs in this cohort is lower than reported in controlled trials, withouut impact on survival outcomes. Education and early recognition are crucial to ensure that these events are identified and addressed.
Collapse
Affiliation(s)
- Leonardo Gomes da Fonseca
- Instituto do Cancer do Estado de São Paulo, Hospital das Clínicas, Universidade São Paulo, Brazil
- Co-first authorship
| | - Federico Piñero
- Hepatology and Liver Transplant Unit, Hospital Universitario Austral, Argentina
- Co-first authorship
| | | | - Carla Bermudez
- Department of Hepatology and Liver Transplantation, Hospital Italiano de Buenos Aires, Argentina
| | | | - Adriana Varón
- Department of Hepatology, Fundación Cardioinfantil, Colombia
| | - Daniela Perez
- Department of Gastroenterology, Hospital Padilla, Tucumán, Argentina
| | - Jorge Rodriguez
- Department of Liver Transplantation, Hospital Central de Mendoza, Argentina
| | - Oscar Beltrán
- Department of Hepatology, Fundación Cardioinfantil, Colombia
| | - Ezequiel Ridruejo
- Department of Hepatology, Centro de Educación Médica e Investigaciones Clínicas (CEMIC), Argentina
| | - Pablo Caballini
- Department of Gastroenterology, Hospital Centenario de Rosario, Santa Fe, Argentina
| | - Alexandre Araujo
- Department of Gastroenterology, Hospital das Clínicas de Porto Alegre, Universidade Federal do Rio Grande do Sul, Brazil
| | | | - Juan Ignacio Marín
- Department of Hepatology and Liver Transplantation, Hospital Pablo Tobón Uribe, Medellín, Colombia
| | - Marina Villa
- Department of Internal Medicine, Hospital Comarcal de Blanes, Córdoba, Argentina
| | | | - Jaime Poniachik
- Department of Gastroenterology, Hospital Clínico de la Universidad de Chile, Chile
| | - Sebastián Marciano
- Department of Hepatology and Liver Transplantation, Hospital Italiano de Buenos Aires, Argentina
| | - Fernando Bessone
- Department of Gastroenterology, Hospital Centenario de Rosario, Santa Fe, Argentina
| | - Manuel Mendizabal
- Hepatology and Liver Transplant Unit, Hospital Universitario Austral, Argentina
| |
Collapse
|
6
|
Kim H, Jang M, Kim E. Exploring the Multifunctional Role of Alpha-Fetoprotein in Cancer Progression: Implications for Targeted Therapy in Hepatocellular Carcinoma and Beyond. Int J Mol Sci 2025; 26:4863. [PMID: 40430002 PMCID: PMC12112184 DOI: 10.3390/ijms26104863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2025] [Revised: 05/16/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Alpha-fetoprotein (AFP) is a well-known biomarker for liver cancer, and its clinical utility is widely recognized. Recent studies have revealed that AFP plays a multifaceted role in various malignant tumors, including liver cancer. This suggests that AFP is not merely a biomarker but also contributes significantly to the complex process of tumor formation, emphasizing the importance of targeting AFP in therapeutic approaches. Consequently, innovative research and development are essential to overcome the current limitations of AFP-targeted therapies, enhance treatment efficacy, and minimize side effects. This review explores the role of AFP in cancer development and progression, highlights the biological functions of AFP and related pathways, and discusses the clinical implications of AFP-targeted therapies. Ongoing research on AFP will significantly contribute to our understanding of the biological mechanisms of cancer and aid in developing effective and safe treatments. Ultimately, advancements in AFP-targeted therapeutic approaches are expected to play a crucial role in the future of cancer management.
Collapse
Affiliation(s)
| | | | - Eunmi Kim
- Cancer Molecular Biology Branch, Division of Cancer Biology, National Cancer Center, Goyang-si 10408, Republic of Korea; (H.K.); (M.J.)
| |
Collapse
|
7
|
Løppke C, Ugleholdt R, Secher CF, Sand NT, Mujollari J, Gustavsson T, Dagil R, Theander TG, Salanti A, Rohrberg KS, Agerbæk MØ. Multi-cancer detection of circulating tumor cells by targeting oncofetal chondroitin sulfate. NPJ Precis Oncol 2025; 9:144. [PMID: 40379789 PMCID: PMC12084527 DOI: 10.1038/s41698-025-00936-3] [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] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 05/05/2025] [Indexed: 05/19/2025] Open
Abstract
Liquid biopsies for the detection of circulating tumor cells (CTCs) are a promising strategy for personalized cancer management. However, traditional CTC detection platforms are often constrained to epithelial cancers, vulnerable to phenotypic changes, and rely on specialized devices for standardized detection, restricting the clinical utility across diverse cancer types and healthcare settings. In this study, we present a tumor-agnostic, platform-independent CTC detection strategy based on recognition of the cancer-specific glycosylation, oncofetal chondroitin sulfate (ofCS). Through coupling of the ofCS-binding protein, VAR2CSA, to a fluorophore-carrying dextran polymer, we successfully detected ofCS-positive CTCs from blood samples in two diverse and independent cohorts comprising early- and late-stage cancer patients of both epithelial and non-epithelial tumor origin. In addition, no ofCS-positive cells were detected in non-malignant controls. Thus, targeting of ofCS has the potential to expand the range of patients who could benefit from CTC analysis, enhancing the clinical utility in various cancer settings.
Collapse
Affiliation(s)
- Caroline Løppke
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Randi Ugleholdt
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Medicine, Copenhagen University Hospital - Herlev and Gentofte, Herlev, Denmark
| | - Christine F Secher
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | | | - Joana Mujollari
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Tobias Gustavsson
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Robert Dagil
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Thor G Theander
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Ali Salanti
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark
| | - Kristoffer S Rohrberg
- Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
- Department of Oncology, Copenhagen University Hospital, Rigshospitalet, Denmark
| | - Mette Ø Agerbæk
- Centre for translational Medicine and Parasitology at Department of Immunology and Microbiology, Faculty of Health and Medical Sciences, University of Copenhagen and Department of Infectious Disease, Copenhagen University Hospital, 2200, Copenhagen, Denmark.
- VarCT Diagnostics, 2000, Frederiksberg, Denmark.
| |
Collapse
|
8
|
Rafaqat S, Noshair I, Shahid M, Bibi S, Hafeez R, Hamid H. Correlation between prognostic markers and clinical parameters in hepatocellular carcinoma: Pathophysiological aspects to therapeutic targets. World J Gastrointest Oncol 2025; 17:106278. [DOI: 10.4251/wjgo.v17.i5.106278] [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: 02/20/2025] [Revised: 03/08/2025] [Accepted: 03/17/2025] [Indexed: 05/15/2025] Open
Abstract
One of the main causes of cancer-related morbidity and mortality globally is hepatocellular carcinoma (HCC). At every stage of the disease, HCC may now be treated using a variety of therapy techniques. Nevertheless, despite the abundance of effective therapeutic choices, the prognosis for patients with HCC is still typically dismal. Prognostic indicators are crucial when assessing prognosis and tracking tumor metastases or recurrence. There are many prognostic markers in HCC. We mainly focused on newly reported prognostic markers such as MEX3A, apolipoprotein B, alpha-fetoprotein, circulating tumor cells, SAMD13, Agrin, and Glypican-3 in the pathogenesis of HCC. Further, we highlighted how these prognostic markers correlated to clinical parameters such as tumor node metastasis, tumor diameter, differentiation, hepatocirrhosis, vascular invasion, and others in HCC. Therefore, identifying specific prognostic biomarkers of HCC helps to provide a great opportunity to improve the prognosis in patients with HCC and provide therapeutic targets.
Collapse
Affiliation(s)
- Saira Rafaqat
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Iqra Noshair
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Momina Shahid
- Department of Zoology, University of Narowal, Narowal 54000, Pakistan
| | - Sadaf Bibi
- Department of Zoology, Government College University, Lahore 54000, Pakistan
| | - Ramsha Hafeez
- Department of Zoology, Lahore College for Women University, Lahore 54000, Pakistan
| | - Hafsa Hamid
- Department of Biotechnology, Lahore College for Women University, Lahore 54000, Pakistan
| |
Collapse
|
9
|
Terzapulo X, Dyussupova A, Ilyas A, Boranova A, Shevchenko Y, Mergenbayeva S, Filchakova O, Gaipov A, Bukasov R. Detection of Cancer Biomarkers: Review of Methods and Applications Reported from Analytical Perspective. Crit Rev Anal Chem 2025:1-46. [PMID: 40367278 DOI: 10.1080/10408347.2025.2497868] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2025]
Abstract
One in five deaths in developed countries is related to cancer. The cancer prevalence is likely to grow with aging population. The affordable and accurate early diagnostics of cancer based on detection of cancer biomarkers at low concentration during its early stages is one of the most efficient way to decrease mortality and human suffering from cancer. The data from 201 analytical papers are tabulated in 9 tables, illustrated in 8 figures and used for comparative analysis of methods applied for cancer biomarker detection, including polymerase chain reaction, Loop-mediated isothermal amplification (LAMP), mass spectrometry, enzyme-linked immunosorbent assay, electroanalytical methods, immunoassays, surface enhanced Raman scattering, Fourier Transform Infrared and others in terms of above-mentioned performance parameters. Median and/or average limit of detection (LOD) are calculated and compared between different analytical methods. We also described and compared LOD of the methods used for detection of three frequently detected cancer biomarkers: carcinoembryonic antigen, prostate-specific antigen and alpha-fetoprotein. Among those methods of detection, the reported electrochemical sensors often demonstrate relatively high sensitivity/low LOD while they often have a moderate instrumental cost and fast time to results. The review tabulates, compares and discusses analytical papers, which report LOD of cancer biomarkers and comprehensive quantitative comparison of various analytical methods is made. The discussion of those techniques applied for cancer biomarker detection included brief summary of pro and cons for each of those methods.
Collapse
Affiliation(s)
- Xeniya Terzapulo
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aigerim Dyussupova
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aisha Ilyas
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Aigerim Boranova
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Yegor Shevchenko
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Saule Mergenbayeva
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Olena Filchakova
- Biology Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| | - Abduzhappar Gaipov
- Department of Medicine, Nazarbayev University School of Medicine, Astana, Republic of Kazakhstan
| | - Rostislav Bukasov
- Chemistry Department, School of Sciences and Humanities, Nazarbayev University, Astana, Republic of Kazakhstan
| |
Collapse
|
10
|
Liu M, Qiu Y, Xie E, Qian P, Yang S, Zhao S, Yan W, Huang X, Han S. Development of a chitosanase 3-like protein 1 assay kit and study of its application in patients with hepatocellular carcinoma. BMC Biotechnol 2025; 25:35. [PMID: 40355912 PMCID: PMC12070687 DOI: 10.1186/s12896-025-00970-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
OBJECTIVE The detection kit for plasma Chitinase-3-like Protein 1 was developed using the magnetic bead chemiluminescence method, in order to investigate the diagnostic value of DD, FDP, CHI3L1, AFP-L3 and PIVKA-II in hepatocellular carcinoma. METHOD The CHI3L1 detection kit was developed using the chemiluminescence method. The luminescence value obtained from the chemiluminescence analyzer was utilized for sensitive detection of CHI3L1, and the performance of the kit was evaluated accordingly. Moreover, this study enrolled 200 patients with hepatocellular carcinoma who were treated at the Oncology Department of the Affiliated Hospital of Jiangnan University between August 2022 and November 2023 as study subjects, while 100 healthy individuals undergoing physical examinations during the same period served as a control group. The plasma CHI3L1 levels in these subjects were measured using our institute's developed kit. Simultaneously, DD, FDP, AFP-L3, and PIVKA-II levels were assessed in all subjects to investigate their relationship with general pathology in patients with hepatocellular carcinoma. Additionally, ROC curves were generated to evaluate both single and combined detections' diagnostic efficacy for hepatocellular carcinoma. RESULT The serological index changes of DD, FDP, AFP-L3, PIVKA-II, and CHI3L1 were not associated with patient gender. The concentrations of AFP-L3 and PIVKA-II in the 45-59 age group were significantly higher than in other groups (P < 0.05). Additionally, DD, CHI3L1, and PIVKA-II levels were markedly elevated in patients with tumors > 5 cm, medium-to-high differentiation, nerve invasion, lymph node metastasis, or distant metastasis. In advanced liver cancer (stages III-IV), DD, FDP, and CHI3L1 concentrations were significantly higher than in early-stage patients (stages I-II). For single diagnostic analysis, the AUC for CHI3L1 was 0.923, while the combined AUC for all five indices was 0.961, indicating greater diagnostic value when used together. The CHI3L1 chemiluminescence detection kit had a minimum detection limit of 1.50 ng/mL, with precision and accuracy within 10%, and R > 0.99. Compared to a clinical reference kit, the correlation coefficient (R) was 0.994, meeting clinical performance evaluation criteria. CONCLUSION The CHI3L1 chemiluminescence kit developed meets clinical requirements. CHI3L1 can be used as an indicator for early screening of liver cancer, and the detection value of combined five indicators DD, FDP, AFP-L3, PIVKA-II and CHI3L1 is higher than that of single detection.
Collapse
Affiliation(s)
- Min Liu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Yanru Qiu
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Erfu Xie
- Department of Laboratory Medicine, The First Afiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu, China
| | - Pu Qian
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Shuxian Yang
- Department of Laboratory Medicine, The First Afiliated Hospital with Nanjing Medical University, Nanjing City, Jiangsu, China
| | - Simin Zhao
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Wenjun Yan
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China
| | - Xuan Huang
- Department of Laboratory Medicine, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China.
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China.
| | - Shuang Han
- Department of Pathology, Affiliated Hospital of Jiangnan University, No. 1000 Hefeng Road, Wuxi City, Jiangsu Province, 214122, China.
- Wuxi Medical College, Jiangnan University, Wuxi, Jiangsu, China.
| |
Collapse
|
11
|
Gan D, Wang Y, Yang X, Huang J, Zhang L, Guo B, Li P, Gou D. Diagnostic value of TAP, PIVKA-II, and AFP in hepatocellular carcinoma and their prognostic value for patients treated with transarterial chemoembolization. Lab Med 2025; 56:297-304. [PMID: 39749461 DOI: 10.1093/labmed/lmae104] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025] Open
Abstract
OBJECTIVE The diagnosis and prognosis of hepatocellular carcinoma (HCC) present significant challenges in clinical practice. This study aimed to evaluate the clinical utility of tumor abnormal protein (TAP), Prothrombin induced by vitamin K absence-II (PIVKA-II), and alpha-fetoprotein (AFP) in diagnosing HCC as well as to investigate their prognostic significance in patients with HCC undergoing transarterial chemoembolization. METHODS A total of 93 HCC patients were enrolled and 101 healthy individuals served as controls. Fresh venous blood samples were collected, and TAP, PIVKA-II, and AFP levels were measured by chemiluminescence immunoassay. RESULTS Significant differences in TAP, PIVKA-II, and AFP levels were found between HCC patients and healthy individuals. The combined assay of TAP, AFP, and PIVKA-II showed better diagnostic performance for HCC. Patients who underwent transarterial chemoembolization and achieved complete response (CR) had lower levels of prechemotherapy serum TAP, AFP, and PIVKA-II. There are significant differences in levels of TAP, AFP, and PIVKA-II between CR and partial response (PR), CR and stable disease (SD), and CR and progressive disease (PD). CONCLUSION Combined detection of TAP, PIVKA-II, and AFP has better diagnostic performance for HCC. Higher levels of prechemotherapy serum TAP, AFP, and PIVKA-II are significantly associated with poor clinical chemoresponse.
Collapse
Affiliation(s)
- Delu Gan
- Department of Clinical Laboratory, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yali Wang
- Department of Clinical Laboratory, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Xin Yang
- Department of Head and Neck Oncology, Chongqing University Cancer Hospital, Chongqing, China
| | - Juan Huang
- Department of Information Center, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Lijun Zhang
- Department of Clinical Laboratory, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Bianqin Guo
- Department of Clinical Laboratory, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| | - Pu Li
- Department of Clinical Laboratory, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Dan Gou
- Department of Clinical Laboratory, Chongqing Key Laboratory of Translational Research for Cancer Metastasis and Individualized Treatment, Chongqing University Cancer Hospital, Chongqing, China
| |
Collapse
|
12
|
An Y, Liu W, Deng Y, Huang W, Huang J. SLC7A11-HSPB1 Axis: A Novel Mechanism for Hepatocellular Carcinoma Progression and Ferroptosis Regulation. Biomed J 2025:100869. [PMID: 40339903 DOI: 10.1016/j.bj.2025.100869] [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: 11/19/2024] [Revised: 03/27/2025] [Accepted: 05/02/2025] [Indexed: 05/10/2025] Open
Abstract
BACKGROUND SLC7A11, a plasma membrane protein, has been implicated as an oncogene in various cancers, including hepatocellular carcinoma (HCC). Its role in HCC pathogenesis, particularly in relation to ferroptosis, is not well understood. This study aims to investigate the function of SLC7A11 with ferroptosis and its interaction in development of HCC. METHODS AND MATERIAL Clinical HCC tissue samples were used to analyze the expression of SLC7A11 by RT-PCR. The impact of SLC7A11 on HCC cell viability, proliferation, and migration was assessed by CCK-8, AlamarBlue and Transwell. Protein-protein interactions were explored using co-immunoprecipitation and immunofluorescence. The effect of SLC7A11 on ferroptosis was evaluated by iron levels, ROS, and GSH. The impact of sorafenib and doxorubicin (DOX) on HCC cells was analyzed using cell viability assay. RESULTS SLC7A11 was found to be highly expressed in HCC tissues and was correlated with tumor size and poor prognosis. Overexpression of SLC7A11 in HCC cells promoted cell viability, proliferation, and migration. Additionally, SLC7A11 overexpression mitigated erastin-induced ferroptosis, as evidenced by decreased ROS levels and increased GSH levels. We also discovered that SLC7A11 interacted with HSPB1. HSPB1 inhibited erastin-induced ferroptosis. Furthermore, a portion of the cell death induced by sorafenib and DOX is attributed to ferroptosis, with HSPB1 and SLC7A11 inhibiting the death induced by the two drugs, respectively. CONCLUSIONS SLC7A11 plays a significant role in HCC progression by inhibiting ferroptosis, and its interaction with HSPB1 is a critical pathway in this process. Targeting the SLC7A11-HSPB1 axis may provide a novel therapeutic strategy for HCC treatment, highlighting the importance of understanding the mechanisms of ferroptosis in cancer cells.
Collapse
Affiliation(s)
- Yan An
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200050, China
| | - Weilong Liu
- Institute of Hepatology, National clinical research center for infectious diseases, Guangdong Key Lab for Diagnosis &Treatment of Emerging Infectious Diseases, Shenzhen Third People's Hospital, Southern University of Science and Technology, Shenzhen, 518112, China
| | - Yuliang Deng
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wanqiu Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| | - Jian Huang
- Key Laboratory of Systems Biomedicine (Ministry of Education), Shanghai Center for Systems Biomedicine, Shanghai Jiao Tong University, Shanghai, 200240, China.
| |
Collapse
|
13
|
Ozer G, Ozcan HN, Ardicli B, Kutluk T, Oguz B, Haliloglu M. Radiological and clinical signatures to differentiate hepatocellular carcinoma from hepatoblastoma in children older than 5 years of age: a feasibility study. Pediatr Radiol 2025; 55:946-954. [PMID: 39961817 PMCID: PMC12065736 DOI: 10.1007/s00247-025-06190-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 01/30/2025] [Accepted: 02/03/2025] [Indexed: 04/12/2025]
Abstract
BACKGROUND Hepatoblastoma and hepatocellular carcinoma (HCC) are the most common primary malignant liver tumors in children. Although some characteristic imaging findings have been described in both hepatoblastoma and HCC, it is difficult to distinguish between these two tumors over the 5 years of age. OBJECTIVE To investigate clinical and radiological findings that may help differentiate hepatoblastoma and HCC over 5 years of age. MATERIALS AND METHODS From 2007 to 2022, 19 consecutive patients older than 5 years old diagnosed with primary liver malignancy were yielded from our radiology archive retrospectively. Imaging features, age, sex, treatment, and follow-up data were recorded. RESULTS A total of 19 patients (16 boys; median age 7.5, min-max 5-17), ten HCCs and nine hepatoblastomas, were included. Serum alpha-fetoprotein (sAFP) values were significantly higher in hepatoblastoma patients (n=9), compared to the HCC (n=10) (P=0.002). Tumor size and PRETEXT stages were higher in hepatoblastoma patients; however, there was no statistical difference (P=0.06). Initial MRI was available for six patients with hepatoblastoma and seven patients with HCC, and there was no difference regarding ADCmin values. CONCLUSION In the differential diagnosis of primary malignant liver tumor in a child older than 5 years of age, higher sAFP level may support the diagnosis of hepatoblastoma rather than HCC.
Collapse
Affiliation(s)
- Gozde Ozer
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey.
| | - H Nursun Ozcan
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey
| | - Burak Ardicli
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey
| | - Tezer Kutluk
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey
| | - Berna Oguz
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey
| | - Mithat Haliloglu
- Hacettepe University School of Medicine, Department of Radiology, Sıhhiye, 06100, Ankara, Turkey
| |
Collapse
|
14
|
Mollaei Z, Asle-Rousta M, Asaadi Tehrani G. Protective effect of menthol against diethylnitrosamine-induced hepatocellular carcinoma in mice by downregulating CTNNB1 and HIF-1α. AVICENNA JOURNAL OF PHYTOMEDICINE 2025; 15:1167-1176. [PMID: 40365190 PMCID: PMC12068496 DOI: 10.22038/ajp.2024.25230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
Objective This study examined the impact of menthol, a natural monoterpene, on diethylnitrosamine (DEN)-induced molecular and histopathological changes in the livers of male mice. Materials and Methods Forty male mice were divided into four groups: Control, Menthol (M), DEN, and DEN-M. The DEN and DEN-M groups received an intraperitoneal injection of DEN (25 mg/kg) at the age of 14 days. The M and DEN-M groups were also given menthol (50 mg/kg, three times a week for six months) via gavage. The expression of genes related to liver carcinoma was analyzed using real-time PCR. Subsequently, the liver tissue was microscopically examined following staining with hematoxylin-eosin. Results After one month, menthol reduced the infiltration of inflammatory cells in the liver tissue of mice injected with DEN. It also prevented the increase in the expression of alpha-fetoprotein (AFP) (p<0.001), programmed cell death 6 (p<0.05), hypoxia-inducible factor-1 alpha (HIF-1α) (p<0.001), and vascular endothelial growth factor (VEGF) (p<0.001) in DEN-M animals compared with DEN group. After six months of session, the expression of AFP (p<0.05), HIF-1α (p<0.05), secreted frizzled-related protein 1 (p<0.001), and catenin beta 1 (p<0.01) was lower in group DEN-M compared with group DEN. Menthol also partially prevented DEN-induced various histopathological changes in the liver after six months of treatment. Conclusion We concluded that menthol inhibits Wnt signaling and suppresses the expression of HIF-1α and VEGF in the liver of DEN-injected mice. It is probably a suitable option for the prevention and treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Zahra Mollaei
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
| | | | - Golnaz Asaadi Tehrani
- Department of Genetics, Zanjan Branch, Islamic Azad University, Zanjan, Iran
- Aerospace and Mechanical Engineering Department, Notre Dame University, Indiana, USA
| |
Collapse
|
15
|
Zhong W, Zhao Z, Fang X, Sun J, Wei Y, Li F, Han B, Jin C. Constructing a neural network model based on tumor-infiltrating lymphocytes (TILs) to predict the survival of hepatocellular carcinoma patients. PeerJ 2025; 13:e19351. [PMID: 40292102 PMCID: PMC12032962 DOI: 10.7717/peerj.19351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2024] [Accepted: 03/31/2025] [Indexed: 04/30/2025] Open
Abstract
Background Hepatocellular carcinoma (HCC) is the most common primary liver cancer worldwide, and early pathological diagnosis is crucial for formulating treatment plans. Despite the widespread attention to pathology in the treatment of HCC patients, a large amount of information contained in pathological images is often overlooked. Methods We retrospectively collected clinical data and pathological slide images from (a) 331 HCC patients at Qingdao University Affiliated Hospital between January 2013 and December 2016 and (b) 180 HCC patients from The Cancer Genome Atlas (TCGA). After data screening, precise quantification of various cell types was achieved using QuPath software. Key factors related to the survival prognosis of pathologically confirmed HCC patients were identified through Cox regression and neural network models, and potential therapeutic targets were screened. Results Our study showed that tumour-infiltrating lymphocytes (TILs) had a protective effect. We quantified the TILs index by machine learning and built a neural network model to predict the prognostic risk of patients (ROC = 0.836 for training set ROC validation set). 95% CI [0.7688-0.896], and there was a significant difference in prognosis in the high-low risk group predicted by the model (p = 2.6e-18, HR = 0.18, 95% CI [0.12-0.27], and TNFSF4 was identified as a possible immunotherapy target. Conclusion This study included a total of 511 patients, divided into a training cohort of 331 cases (from Qingdao University Hospital between January 2013 and December 2016) and a validation cohort of 180 cases (TCGA). The results revealed that tumor-infiltrating lymphocytes (TILs) have a protective effect and successfully predicted the survival risk of liver cancer patients using machine learning and neural network technology. The discovery of TNFSF4 provides a new potential target for immunotherapy.
Collapse
Affiliation(s)
- Wenqing Zhong
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Ziyin Zhao
- Organ Transplantation Center, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Xin Fang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| | - Jingyi Sun
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Yanbing Wei
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Fengda Li
- Department of Hepatobiliary Surgery, Gao mi People’s Hospital, Weifang, Shandong, China
| | - Bing Han
- Department of Hepatobiliary and Pancreatic Surgery, The Affiliated Hospital of Qingdao University, Qingdao, Shandong, China
| | - Cheng Jin
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai, Shanghai, China
| |
Collapse
|
16
|
Lin Y, Ma Y, Chen Y, Huang Y, Lin J, Xiao Z, Cui Z. Diagnostic and prognostic performance of serum GPC3 and PIVKA-II in AFP-negative hepatocellular carcinoma and establishment of nomogram prediction models. BMC Cancer 2025; 25:721. [PMID: 40247208 PMCID: PMC12007284 DOI: 10.1186/s12885-025-14025-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2023] [Accepted: 03/26/2025] [Indexed: 04/19/2025] Open
Abstract
OBJECTIVE A significant proportion, ranging from 20 to 40%, of individuals with hepatocellular carcinoma (HCC) do not exhibit elevated Alpha-fetoprotein (AFP) levels. This study aimed to evaluate the utility of serum glypican-3 (GPC3) and protein induced by vitamin K absence or antagonist II (PIVKA-II) in an AFP-negative HCC (N-HCC) population, and to develop nomogram diagnostic and prognostic prediction models utilizing GPC3 and PIVKA-II. METHODS Serum GPC3 and PIVKA-II levels were measured in this case-control study, followed by the establishment of a receiver operating characteristic (ROC) curve, restricted cubic spline (RCS), and Kaplan-Meier survival curve. Additionally, a diagnostic prediction nomogram was constructed using univariate and multivariate logistic regression. Furthermore, we utilized least absolute shrinkage and selection operator (LASSO) regression and multivariate Cox regression to develop a prognostic prediction nomogram. The performance of these models was evaluated using ROC curve analysis and decision curve analysis (DCA). RESULTS Serum GPC3 and PIVKA-II expression levels were significantly elevated in untreated patients with N-HCC (especially stageI and tumor size < 3 cm) compared to those with AFP-negative benign liver disease (N-BLD). Derived from ROC analysis, the diagnostic cutoff points for GPC3 and PIVKA-II were set at 0.100 ng/mL and 40.00 mAU/mL, respectively. PIVKA-II demonstrated sensitivity and specificity of 84.62% and 90.38%, surpassing GPC3's 76.92% and 73.08%. The area under the ROC curve (AUC) for a diagnostic prediction nomogram incorporating GPC3, PIVKA-II, and gamma-glutamyltransferase (GGT) was 0.943 (95% CI: 0.912-0.974), superior to models using GPC3 or PIVKA-II alone. This model showed 95.20% sensitivity and 81.70% specificity in differentiating N-HCC from N-BLD. Stratifying patients into high-risk and low-risk groups using cutoff values established by RCS for GPC3 (0.124 ng/mL) and PIVKA-II (274 mAU/mL) revealed significant associations between these risk stratifications and patient survival. Finally, the use of GPC3-highrisk, cirrhosis, albumin (ALB), portal venous thrombosis (PVT), and surgical treatment as five parameters in the nomogram prognostic prediction model effectively differentiated between high- and low-risk prognostic patients with N-HCC with relatively high accuracy. CONCLUSIONS Serum GPC3 and PIVKA-II demonstrate clinical significance in the timely detection and prognosis assessment of N-HCC. The application of nomogram prediction models based on GPC3 and PIVKA-II stands as an important adjunctive tool for diagnosing and prognosticating N-HCC.
Collapse
Affiliation(s)
- Yingying Lin
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Yuefei Ma
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Yan Chen
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Yepei Huang
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China
| | - Jinchuan Lin
- Department of Laboratory Medicine, the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350005, China
- Department of Laboratory Medicine, National Regional Medical Center, Binhai Campus of the First Affiliated Hospital, Fujian Medical University, Fuzhou, 350212, China
| | - Zhenzhou Xiao
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China.
| | - Zhaolei Cui
- Laboratory of Biochemistry and Molecular Biology Research, Department of Clinical Laboratory, Clinical Oncology School of Fujian Medical University, Fujian Cancer Hospital, Fuzhou, 350014, Fujian, China.
| |
Collapse
|
17
|
Wen Y, Zhou S, Xu Y, Zhang C, Feng Z, Song Y, Ding B, Peng C, Tan H, Wang C, Feng J, Pei J, He G, Fu S, Wang L, Cai L, Liu S, Pan M. Donafenib versus sorafenib in triple therapy for unresectable hepatocellular carcinoma: a propensity score-matched multicenter analysis. World J Surg Oncol 2025; 23:143. [PMID: 40221746 PMCID: PMC11993997 DOI: 10.1186/s12957-025-03767-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND In recent years, triple therapy (molecular targeted agent + PD-1 inhibitor + transarterial therapy) has emerged as a promising strategy for unresectable hepatocellular carcinoma (uHCC). However, the optimal molecular targeted agent choice within triple therapy remains unclear. Donafenib is currently the only targeted drug with superior survival benefits compared with sorafenib monotherapy. This study aimed to compare donafenib-based versus sorafenib-based triple therapy in patients with uHCC, providing preliminary evidence to guide molecular targeted agent selection in this emerging treatment paradigm. METHODS This retrospective study enrolled 106 patients with initially uHCC who received triple therapy combining either donafenib or sorafenib with PD-1 inhibitors and transarterial therapies. A 1:2 nearest neighbour propensity score matching was used to minimize selection bias. The primary endpoints were overall survival (OS) and progression-free survival (PFS) based on Kaplan-Meier analysis. The secondary endpoints included objective response rate (ORR), surgical conversion rate and adverse events (AEs). Statistical comparisons used Cox regression for survival data and chi-squared/ t-tests for other metrics, with p < 0.05 indicating significance. RESULTS After matching, 30 patients received sorafenib-based triple therapy (Sor-P-T/H group) and 50 patients received donafenib-based triple therapy (Don-P-T/H group). Although the median OS was not attained, the Don-P-T/H regimen demonstrated a statistically significant survival advantage (HR = 0.317, P = 0.004). Moreover, the Don-P-T/H group demonstrated significantly higher median PFS (9.00 vs. 4.62 months, P = 0.005), ORR (64% vs. 40%, P = 0.037) and surgical conversion rate (26.0% vs. 3.3%, P = 0.01) compared to the Sor-P-T/H group. The two groups showed no notable difference in the overall severity of adverse events but the Don-P-T/H group demonstrated less liver impairment. CONCLUSION Donafenib may be more advantageous than sorafenib in triple therapy for patients with uHCC.
Collapse
Affiliation(s)
- Yaohong Wen
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Shuyi Zhou
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, People's Republic of China
| | - Yuyan Xu
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Cheng Zhang
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Zhoubin Feng
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Yinghui Song
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, People's Republic of China
| | - Bai Ding
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, People's Republic of China
| | - Chuang Peng
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, People's Republic of China
- Hunan Provincial Key Laboratory of Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
- Clinical Medical Technology Research Center of Hunan Provincial for Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China
| | - Hongkun Tan
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Chunming Wang
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Jianan Feng
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Jingyuan Pei
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Guolin He
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Shunjun Fu
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China
| | - Lvhuan Wang
- Suzhou Zelgen Biopharmaceuticals Co, Ltd, Suzhou, China
| | - Lei Cai
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China.
| | - Sulai Liu
- Department of Hepatobiliary Surgery/Central Laboratory, Hunan Provincial People's Hospital (The First Affiliated Hospital of Hunan Normal University), Changsha, Hunan Province, People's Republic of China.
- Hunan Engineering Research Center of Digital Hepatobiliary Medicine, Changsha, Hunan Province, China.
- Hunan Provincial Key Laboratory of Biliary Disease Prevention and Treatment, Changsha, Hunan Province, China.
| | - Mingxin Pan
- General Surgery Center, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Southern Medical University, No.253 Gongye Dadao Zhong, Haizhu District, Guangzhou, 510280, Guangdong Province, China.
| |
Collapse
|
18
|
Ivaskiene T, Kaspute G, Ramanavicius A, Prentice U. Molecularly Imprinted Polymer Advanced Hydrogels as Tools for Gastrointestinal Diagnostics. Gels 2025; 11:269. [PMID: 40277704 PMCID: PMC12026608 DOI: 10.3390/gels11040269] [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: 03/10/2025] [Revised: 03/27/2025] [Accepted: 04/01/2025] [Indexed: 04/26/2025] Open
Abstract
Gastroenterology faces significant challenges due to the global burden of gastrointestinal (GI) diseases, driven by socio-economic disparities and their wide-ranging impact on health and healthcare systems. Advances in molecularly imprinted polymers (MIPs) offer promising opportunities for developing non-invasive, cost-effective diagnostic tools that enhance the accuracy and accessibility of GI disease detection. This research explores the potential of MIP-based sensors in revolutionizing gastrointestinal diagnostics and improving early detection and disease management. Biomarkers are vital in diagnosing, monitoring, and personalizing disease treatment, particularly in gastroenterology, where advancements like MIPs offer highly selective and non-invasive diagnostic solutions. MIPs mimic natural recognition mechanisms, providing stability and sensitivity even in complex biological environments, making them ideal for early disease detection and real-time monitoring. Their integration with advanced technologies, including conducting polymers, enhances their functionality, enabling rapid, point-of-care diagnostics for gastrointestinal disorders. Despite regulatory approval and scalability challenges, ongoing innovations promise to revolutionize diagnostics and improve patient outcomes through precise approaches.
Collapse
Affiliation(s)
- Tatjana Ivaskiene
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
| | - Greta Kaspute
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
| | - Arunas Ramanavicius
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| | - Urte Prentice
- State Research Institute Centre for Innovative Medicine, LT-08410 Vilnius, Lithuania; (T.I.); (G.K.)
- Department of Nanotechnology, State Research Institute Center for Physical Sciences and Technology (FTMC), LT-10257 Vilnius, Lithuania
- Department of Physical Chemistry, Faculty of Chemistry and Geosciences, Institute of Chemistry, Vilnius University, LT-03225 Vilnius, Lithuania
| |
Collapse
|
19
|
Wang Y, Chi S, Tian Y, Li X, Zhang H, Xu Y, Huang C, Gao Y, Jin G, Fu Q, Cao W, Chen C, Ding H, Zhang Y, Hong Y, Li J, Sun X, Li E, Zhang Y, Yao W, Liu R, Hua Y, Huang H, Xu M, Zhang B, Tao W, Yang T, Gao Y, Wang X, Lin C, Li J, Zhang Q, Liang T. Construction of an artificially intelligent model for accurate detection of HCC by integrating clinical, radiological, and peripheral immunological features. Int J Surg 2025; 111:2942-2952. [PMID: 39878177 DOI: 10.1097/js9.0000000000002281] [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: 10/11/2024] [Accepted: 12/30/2024] [Indexed: 01/31/2025]
Abstract
BACKGROUND Integrating comprehensive information on hepatocellular carcinoma (HCC) is essential to improve its early detection. We aimed to develop a model with multimodal features (MMF) using artificial intelligence (AI) approaches to enhance the performance of HCC detection. MATERIALS AND METHODS A total of 1092 participants were enrolled from 16 centers. These participants were allocated into the training, internal validation, and external validation cohorts. Peripheral blood specimens were collected prospectively and subjected to mass cytometry analysis. Clinical and radiological data were obtained from electrical medical records. Various AI methods were employed to identify pertinent features and construct single-modal models with optimal performance. The XGBoost algorithm was utilized to amalgamate these models, integrating multimodal information and facilitating the development of a fusion model. Model evaluation and interpretability were demonstrated using the SHapley Additive exPlanations method. RESULTS We constructed the electronic health record, BioScore, RadiomicScore, and DLScore models based on clinical, radiological, and peripheral immunological features, respectively. Subsequently, these single-modal models were amalgamated to develop an all-in-one MMF model. The MMF model exhibited enhanced performance compared to models comprising only single-modal features in detecting HCC. This superiority in performance was confirmed through the internal and external validation cohorts, yielding area under the curve (AUC) values of 0.985 and 0.915, respectively. Additionally, the MMF model improved the detection ability in subpopulations of HCCs that were negative for alpha-fetoprotein and those with small size, with AUC values of 0.974 and 0.996, respectively. CONCLUSIONS Integrating MMF improved the performance of the model for HCC detection.
Collapse
Affiliation(s)
- Yangyang Wang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Shengqiang Chi
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Yu Tian
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Xueyao Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Hang Zhang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiting Xu
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Chao Huang
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Yiwei Gao
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
| | - Gaowei Jin
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Qihan Fu
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Department of Oncology, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Wanyue Cao
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Cao Chen
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Haonan Ding
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yuquan Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
| | - Yupeng Hong
- Department of Oncology, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, China
| | - Junjian Li
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, China
| | - Xu Sun
- Department of General Surgery, Huzhou Central Hospital, Zhejiang University School of Medicine, Huzhou, China
| | - Enliang Li
- Department of Hepatobiliary and Pancreatic Surgery, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Yuhua Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The Cancer Hospital of the University of Chinese Academy of Sciences (Zhejiang Cancer Hospital), Institute of Basic Medicine and Cancer, Chinese Academy of Sciences, Hangzhou, China
| | - Weiyun Yao
- Department of Surgery, Changxing People's Hospital, Huzhou, China
| | - Runtian Liu
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - Yongfei Hua
- Department of General Surgery, Ningbo Medical Center Lihuili Eastern Hospital, Ningbo, China
| | - Haifeng Huang
- Department of General Surgery, Shengzhou People's Hospital, Shengzhou, China
| | - Minghui Xu
- Department of General Surgery, Haining People's Hospital, Haining, China
| | - Bo Zhang
- Department of General Surgery, Shenzhen University Luohu People's Hospital, Shenzhen, China
| | - Weifeng Tao
- Department of General Surgery, Shangyu People's Hospital of Shaoxing, Shangyu, China
| | - Tianxing Yang
- Department of Medical Oncology, Sanmen People's Hospital, Taizhou, China
| | - Yuming Gao
- Department of General Surgery, Jixi County People's Hospital, Jixi, China
| | - Xiaoguang Wang
- Department of General Surgery, Jiaxing Second People's Hospital, Jiaxing, China
| | - Cheng Lin
- Zhejiang Puluoting Health Technology Co Ltd, Hangzhou, China
| | - Jingsong Li
- Research Center for Data Hub and Security, Zhejiang Lab, Hangzhou, China
- The Engineering Research Center of EMR and Intelligent Expert System, Ministry of Education, College of Biomedical Engineering and Instrument Science, Zhejiang University, Hangzhou, China
| | - Qi Zhang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| | - Tingbo Liang
- Department of Hepatobiliary and Pancreatic Surgery, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- MOE Joint International Research Laboratory of Pancreatic Diseases, Hangzhou, China
- Zhejiang Provincial Key Laboratory of Pancreatic Disease, the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, China
- Zhejiang Clinical Research Center of Hepatobiliary and Pancreatic Diseases, Hangzhou, China
- Cancer Center of Zhejiang University, Hangzhou, China
| |
Collapse
|
20
|
Crawford CEW, Burslem GM. Acetylation: a new target for protein degradation in cancer. Trends Cancer 2025; 11:403-420. [PMID: 40055119 PMCID: PMC11981854 DOI: 10.1016/j.trecan.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 01/15/2025] [Accepted: 01/24/2025] [Indexed: 04/11/2025]
Abstract
Acetylation is an increasing area of focus for cancer research as it is closely related to a variety of cellular processes through modulation of histone and non-histone proteins. However, broadly targeting acetylation threatens to yield nonselective toxic effects owing to the vital role of acetylation in cellular function. There is thus a pressing need to elucidate and characterize the specific cancer-relevant roles of acetylation for future therapeutic design. Acetylation-mediated protein homeostasis is an example of selective acetylation that affects a myriad of proteins as well as their correlated functions. We review recent examples of acetylation-mediated protein homeostasis that have emerged as key contributors to tumorigenesis, tumor proliferation, metastasis, and/or drug resistance, and we discuss their implications for future exploration of this intriguing phenomenon.
Collapse
Affiliation(s)
- Callie E W Crawford
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA
| | - George M Burslem
- Department of Biochemistry and Biophysics, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA; Department of Cancer Biology and Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, PA 19104, USA.
| |
Collapse
|
21
|
Adugna A, Azanaw Amare G, Jemal M. Current Advancements in Serum Protein Biomarkers for Hepatitis B Virus-Associated Hepatocyte Remodeling and Hepatocellular Carcinoma. Immun Inflamm Dis 2025; 13:e70171. [PMID: 40192058 PMCID: PMC11973733 DOI: 10.1002/iid3.70171] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Revised: 02/08/2025] [Accepted: 02/28/2025] [Indexed: 04/10/2025] Open
Abstract
BACKGROUND Hepatitis B virus (HBV)-related liver cancer is the third most common cause of cancer-related death globally. Hepatocyte remodeling, also known as hepatocyte transformation and immortalization, and hepatocellular carcinoma (HCC), are brought on by persistent inflammation caused by HBV in the host hepatocytes. One of the main concerns in the perspective of HBV-induced hepatocyte remodeling and liver cancer is accurately identifying cancer stages to maximize early screening and detection. Biological signatures have a significant impact on solving this problem. OBJECTIVE This review article aimed to discuss the novel serum protein biomarkers for HBV-induced hepatocyte remodeling and HCC. METHODS The information was collected from various peer-reviewed journals through electronic searches utilizing various search engines, including PubMed, Google Scholar, HINARI, and Cochrane Library from 2017 to 2024. Keywords for searches included "serum protein biomarkers in HBV-HCC," "blood-based biomarkers in HBV-HCC," and "viral biomarkers for HBV-HCC." RESULTS Recently, novel protein signatures have been discovered for the early diagnosis, treatment, and prognosis of HBV-induced hepatic cell remodeling and HCC from proteomic data sets. We have discussed the recent literature on the clinical utility of the protein signatures for the diagnosis and forecasting of HBV-associated hepatocyte remodeling and HCC, including golgi protein 73 (GP73), glypican-3 (GPC3), midkine (MDK), des-γ-carboxy-prothrombin (DCP), von Willebrand factor (vWF), pentraxin 3 (PTX3), pseudouridine synthases 7 (PUSs 7), squamous cell carcinoma antigen (SCCA), and osteopontin (OPN). CONCLUSION All these protein markers also exhibit the survival of HBV-related HCC patients, the proliferation, migration, antiapoptosis, mitogenesis, transformation, and angiogenesis of HBV-infected hepatocytes.
Collapse
Affiliation(s)
- Adane Adugna
- Medical Laboratory SciencesCollege of Health SciencesDebre Markos UniversityDebre MarkosEthiopia
| | - Gashaw Azanaw Amare
- Medical Laboratory SciencesCollege of Health SciencesDebre Markos UniversityDebre MarkosEthiopia
| | - Mohammed Jemal
- Department of Biomedical Sciences, School of MedicineDebre Markos UniversityDebre MarkosEthiopia
| |
Collapse
|
22
|
Akaoka M, Yanagaki M, Kubota H, Haruki K, Furukawa K, Taniai T, Onda S, Hamura R, Tsunematsu M, Shirai Y, Matsumoto M, Shimoda M, Ikegami T. ARID4B Promotes the Progression of Hepatocellular Carcinoma Through the PI3K/AKT Pathway. Ann Surg Oncol 2025; 32:3009-3018. [PMID: 39751985 DOI: 10.1245/s10434-024-16790-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 12/12/2024] [Indexed: 01/04/2025]
Abstract
BACKGROUND AT-rich interaction domain 4B (ARID4B) is a transcriptional activator that regulates the phosphatidylinositol 3-kinase (PI3K)/AKT pathway in prostate cancer. However, the role of ARID4B in hepatocellular carcinoma (HCC) has remained unclear. METHODS This study included 162 patients who had undergone primary hepatic resection for HCC between 2008 and 2019. Their HCC samples were immunohistochemically stained for ARID4B, and ARID4B score was calculated from the intensity and percentage of staining. We retrospectively investigated the association of ARID4B score with disease-free and overall survival, and primary recurrence patterns of HCC. Furthermore, human HCC cell lines (HuH-1 and HuH-7) were knocked down for ARID4B using small-interfering RNA (siRNA), and the expression of PI3K/AKT proteins, cell proliferation, migration, and invasion ability were assessed. RESULTS In multivariate analyses, negative HBs-antigen (p = 0.02), multiple tumors (p < 0.01), microvascular invasion (p = 0.03), and high ARID4B score (p = 0.01) were independent predictors of disease-free survival, while tumor size >5 cm (p = 0.03), microvascular invasion (p < 0.01), and high ARID4B score (p = 0.04) were independent predictors of overall survival. A high ARID4B score was associated with high serum α-fetoprotein (AFP) level (p = 0.04), poor tumor differentiation (p < 0.01), and microvascular invasion (p < 0.01). ARID4B scores were significantly lower in the no recurrence, intrahepatic recurrence, and extrahepatic recurrence groups, in that order. Knockdown of ARID4B using siRNA in human HCC cell lines significantly suppressed the PI3K/AKT pathway, cell proliferation, migration, and invasion. CONCLUSIONS ARID4B may activate the PI3K/AKT signaling pathway in HCC and may be a prognostic factor after hepatic resection for HCC.
Collapse
Affiliation(s)
- Munetoshi Akaoka
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Mitsuru Yanagaki
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan.
| | - Hoshiho Kubota
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Koichiro Haruki
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Kenei Furukawa
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Tomohiko Taniai
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Shinji Onda
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Ryoga Hamura
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masashi Tsunematsu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Yoshihiro Shirai
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Michinori Matsumoto
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| | - Masayuki Shimoda
- Department of Pathology, The Jikei University School of Medicine, Tokyo, Japan
| | - Toru Ikegami
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The Jikei University School of Medicine, Tokyo, Japan
| |
Collapse
|
23
|
Zhang F, Wang YS, Li SP, Zhao B, Huang N, Song RP, Meng FZ, Feng ZW, Zhang SY, Song HC, Chen XP, Liu LX, Wang JZ. Alpha-fetoprotein combined with initial tumor shape irregularity in predicting the survival of patients with advanced hepatocellular carcinoma treated with immune-checkpoint inhibitors: a retrospective multi-center cohort study. J Gastroenterol 2025; 60:442-455. [PMID: 39714631 PMCID: PMC11922967 DOI: 10.1007/s00535-024-02202-y] [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: 08/12/2024] [Accepted: 12/07/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND Immune checkpoint inhibitors (ICIs) are playing a significant role in the treatment of hepatocellular carcinoma (HCC). This study aims to explore the prognostic value of alpha-fetoprotein (AFP) and initial tumor shape irregularity in patients treated with ICIs. METHODS In this retrospective, multi-center study, 296 HCC patients were randomly divided into the training set and the validation set in a 3:2 ratio. The training set was used to evaluate prognostic factors and to develop an easily applicable ATSI (AFP and Tumor Shape Irregularity) score, which was verified in the validation set. RESULTS The ATSI score was developed from two independent prognostic risk factors: baseline AFP ≥ 400 ng/ml (HR 1.73, 95% CI 1.01-2.96, P = 0.046) and initial tumor shape irregularity (HR 1.94, 95% CI 1.03-3.65, P = 0.041). The median overall survival (OS) was not reached (95% CI 28.20-NA) in patients who met no criteria (0 points), 25.8 months (95% CI 14.17-NA) in patients who met one criterion (1 point), and 17.03 months (95% CI 11.73-23.83) in patients who met two criteria (2 points) (P = 0.001). The median progression-free survival (PFS) was 10.83 months (95% CI 9.27-14.33) for 0 points, 8.03 months (95% CI 6.77-10.57) for 1 point, and 5.03 months (95% CI 3.83-9.67) for 2 points (P < 0.001). The validation set effectively verified these results (median OS, 37.43/24.27/14.03 months for 0/1/2 points, P = 0.028; median PFS, 13.93/8.30/4.90 months for 0/1/2 points, P < 0.001). CONCLUSIONS The ATSI score can effectively predict prognosis in HCC patients receiving ICIs.
Collapse
Affiliation(s)
- Feng Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Yong-Shuai Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Shao-Peng Li
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
| | - Bin Zhao
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Nan Huang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Rui-Peng Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Fan-Zheng Meng
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Zhi-Wen Feng
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China
| | - Shen-Yu Zhang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Hua-Chuan Song
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China
| | - Xiao-Peng Chen
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, Anhui, 241000, China.
| | - Lian-Xin Liu
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| | - Ji-Zhou Wang
- Department of Hepatobiliary Surgery, Centre for Leading Medicine and Advanced Technologies of IHM, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui, 230001, China.
- Anhui Provincial Key Laboratory of Hepatopancreatobiliary Surgery, Hefei, Anhui, 230001, China.
- Anhui Provincial Clinical Research Center for Hepatobiliary Diseases, Hefei, Anhui, 230001, China.
| |
Collapse
|
24
|
Banjan B, Vishwakarma R, Ramakrishnan K, Dev RR, Kalath H, Kumar P, Soman S, Raju R, Revikumar A, Rehman N, Abhinand CS. Targeting AFP-RARβ complex formation: a potential strategy for treating AFP-positive hepatocellular carcinoma. Mol Divers 2025; 29:1337-1352. [PMID: 38955977 DOI: 10.1007/s11030-024-10915-8] [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: 03/21/2024] [Accepted: 06/09/2024] [Indexed: 07/04/2024]
Abstract
Alpha-fetoprotein (AFP) is a glycoprotein primarily expressed during embryogenesis, with declining levels postnatally. Elevated AFP levels correlate with pathological conditions such as liver fibrosis, cirrhosis, and hepatocellular carcinoma (HCC). Recent investigations underscore AFP's intracellular role in HCC progression, wherein it forms complexes with proteins like Phosphatase and tensin homolog (PTEN), Caspase 3 (CASP3), and Retinoic acid receptors and Retinoid X receptors (RAR/RXR). RAR and RXR regulate gene expression linked to cell death and tumorigenesis in normal physiology. AFP impedes RAR/RXR dimerization, nuclear translocation, and function, promoting gene expression favoring cancer progression in HCC that provoked us to target AFP as a drug candidate. Despite extensive studies, inhibitors targeting AFP to disrupt complex formation and activities remain scarce. In this study, employing protein-protein docking, amino acid residues involved in AFP-RARβ interaction were identified, guiding the definition of AFP's active site for potential inhibitor screening. Currently, kinase inhibitors play a significant role in cancer treatment and, the present study explores the potential of repurposing FDA-approved protein kinase inhibitors to target AFP. Molecular docking with kinase inhibitors revealed Lapatinib as a candidate drug of the AFP-RARβ complex. Molecular dynamics simulations and binding energy calculations, employing Mechanic/Poisson-Boltzmann Surface Area (MM-PBSA), confirmed Lapatinib's stability with AFP. The study suggests Lapatinib's potential in disrupting the AFP-RARβ complex, providing a promising avenue for treating molecularly stratified AFP-positive HCC or its early stages.
Collapse
Affiliation(s)
- Bhavya Banjan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Riya Vishwakarma
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Krishnapriya Ramakrishnan
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Radul R Dev
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Haritha Kalath
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Pankaj Kumar
- Nitte (Deemed to Be University), Department of Pharmaceutical Chemistry, NGSMPS, NGSM Institute of Pharmaceutical Sciences, Mangalore, 575018, Karnataka, India
| | - Sowmya Soman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India
| | - Amjesh Revikumar
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India
- Kerala Genome Data Centre, Kerala Development and Innovation Strategic Council, Vazhuthacaud, Thiruvananthapuram, Kerala, 695014, India
| | - Niyas Rehman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, 575018, India.
| | - Chandran S Abhinand
- Centre for Systems Biology and Molecular Medicine (CSBMM), Yenepoya Research Centre, Yenepoya (Deemed to Be University), Deralakatte, Mangalore, 575018, Karnataka, India.
| |
Collapse
|
25
|
Pinto E, Lazzarini E, Pelizzaro F, Gambato M, Santarelli L, Potente S, Zanaga P, Zappitelli T, Cardin R, Burra P, Farinati F, Romualdi C, Boscarino D, Tosello V, Indraccolo S, Russo FP. Somatic Copy Number Alterations in Circulating Cell-Free DNA as a Prognostic Biomarker for Hepatocellular Carcinoma: Insights from a Proof-of-Concept Study. Cancers (Basel) 2025; 17:1115. [PMID: 40227625 PMCID: PMC11988118 DOI: 10.3390/cancers17071115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2025] [Revised: 03/13/2025] [Accepted: 03/21/2025] [Indexed: 04/15/2025] Open
Abstract
BACKGROUND/OBJECTIVES Despite advances in hepatocellular carcinoma (HCC) management, prognosis remains poor. Advanced-stage diagnosis often excludes curative treatments, and current biomarkers (e.g., alpha-fetoprotein [AFP]) have limited utility in early detection. Liquid biopsy has emerged as a promising cancer detection tool, with circulating cell-free DNA (ccfDNA) showing significant diagnostic potential. This proof-of-concept study aimed to investigate the potential role of tumor fraction (TF) within ccfDNA as a biomarker in HCC patients. METHODS A total of sixty patients were recruited, including thirteen with chronic liver disease (CLD), twenty-four with cirrhosis, and twenty-three with HCC. Plasma samples were collected, and ccfDNA was extracted for shallow whole genome sequencing (sWGS) analysis. The TF was calculated by focusing on somatic copy number alterations (SCNAs) within the ccfDNA. RESULTS Among patients with CLD and cirrhosis (n = 37), ctDNA was undetectable in all but one cirrhotic patient who exhibited a significant tumor fraction (TF) of 17% and subsequently developed HCC. Conversely, five out of twenty-three HCC patients (21.7%) displayed detectable ctDNA with TF levels ranging from 3.0% to 32.6%. Patients with detectable ctDNA were characterized by more aggressive oncological features, including a higher number of nodules (p = 0.005), advanced-stage disease (60% BCLC C, p = 0.010), and poorer response to therapy (80% PD, p = 0.001). Moreover, the overall survival (OS) was significantly reduced in patients with detectable ctDNA (median OS: 17 months; CI 95% 4.5-26.5) compared to those without (median OS: 24.0 months; CI 95% 7.0-66.0; log-rank p = 0.002). CONCLUSIONS Our results suggest that the analysis of TF by sWGS is a promising non-invasive tool for the identification of HCC with aggressive clinical behavior, whereas it is not sensitive enough for early HCC detection. This molecular assay can improve prognostic stratification in HCC patients.
Collapse
Affiliation(s)
- Elisa Pinto
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Elisabetta Lazzarini
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Filippo Pelizzaro
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Martina Gambato
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Laura Santarelli
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Sara Potente
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | - Paola Zanaga
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Teresa Zappitelli
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Romilda Cardin
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Patrizia Burra
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Fabio Farinati
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| | - Chiara Romualdi
- Department of Biology, University of Padova, 35121 Padua, Italy; (S.P.); (C.R.)
| | | | - Valeria Tosello
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Stefano Indraccolo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Basic and Translational Oncology Unit, Veneto Institute of Oncology IOV-IRCCS, 35121 Padua, Italy; (E.L.); (L.S.); (V.T.)
| | - Francesco Paolo Russo
- Department of Surgery, Oncology and Gastroenterology, University of Padova, 35121 Padua, Italy; (E.P.); (F.P.); (M.G.); (P.Z.); (T.Z.); (P.B.); (F.F.); (S.I.)
- Gastroenterology Unit, Azienda Ospedale-Università di Padova, 35121 Padua, Italy;
| |
Collapse
|
26
|
Tang P, Zhou F. Efficacy and safety of PD-1/PD-L1 inhibitors combined with tyrosine kinase inhibitors as first-line treatment for hepatocellular carcinoma: a meta-analysis and trial sequential analysis of randomized controlled trials. Front Pharmacol 2025; 16:1535444. [PMID: 40196369 PMCID: PMC11973308 DOI: 10.3389/fphar.2025.1535444] [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: 11/27/2024] [Accepted: 03/04/2025] [Indexed: 04/09/2025] Open
Abstract
Background The use of immune checkpoint inhibitors (ICIs) in treating hepatocellular carcinoma (HCC) has grown significantly. However, the therapeutic benefits of ICIs alone are notably modest. This meta-analysis assesses the efficacy and safety of using PD-1/PD-L1 inhibitors in conjunction with tyrosine kinase inhibitors (TKIs) for patients with advanced or unresectable HCC. Methods An extensive search of the literature was performed using databases such as PubMed, Web of Science, Embase, and the Cochrane Library, capturing randomized controlled trials (RCTs) until 16 October 2024. Efficacy was measured by progression-free survival (PFS), overall survival (OS), objective response rate (ORR), and disease control rate (DCR). Safety was gauged through the occurrence of treatment-related adverse events (TRAEs). Hazard ratios (HRs) for PFS and OS, along with risk ratios (RRs) for ORR, DCR, and TRAEs, were calculated, each with 95% confidence intervals (CIs). Heterogeneity among studies was quantified using Cochran's Q test, I2 statistics, and 95% prediction intervals (PIs). Results This analysis incorporated 4 studies with a total of 2,174 patients. Treatment regimens combining PD-1/PD-L1 inhibitors with TKIs significantly improved PFS (HR = 0.694, 95% CI: 0.527-0.914; 95% PI: 0.228-2.114) and ORR (RR = 2.303, 95% CI: 1.360-3.902; 95% PI: 0.408-12.991) compared with first-line monotherapy or TKI monotherapy in the overall population. Subgroup analysis indicated that the improvements in PFS and OS were particularly significant among patients of Asian descent or those with hepatitis B virus (HBV) infection (all p < 0.05). While the occurrence of any grade TRAEs did not differ significantly between the two groups (RR = 1.016, 95% CI: 0.996-1.036; 95% PI: 0.941-1.097), the incidence of serious (RR = 2.068, 95% CI: 1.328-3.222; 95% PI: 0.487-8.776) and grade ≥3 TRAEs (RR = 1.287, 95% CI: 1.020-1.624; 95% PI: 0.574-2.883) increased in patients treated with the combination of PD-1/PD-L1 inhibitors and TKIs. Conclusion This study revealed that combining PD-1/PD-L1 inhibitors with TKIs in the treatment of advanced or unresectable HCC leads to superior clinical outcomes compared to first-line monotherapy or TKIs alone, particularly in patients with HBV infection and those of Asian descent. Clinicians are advised to be vigilant regarding the potential for TRAEs in clinical settings.
Collapse
Affiliation(s)
- Peng Tang
- Department of Gastroenterology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Fei Zhou
- Department of Obstetrics and Gynaecology, Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
27
|
Giron-Michel J, Padelli M, Oberlin E, Guenou H, Duclos-Vallée JC. State-of-the-Art Liver Cancer Organoids: Modeling Cancer Stem Cell Heterogeneity for Personalized Treatment. BioDrugs 2025; 39:237-260. [PMID: 39826071 PMCID: PMC11906529 DOI: 10.1007/s40259-024-00702-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/21/2024] [Indexed: 01/20/2025]
Abstract
Liver cancer poses a global health challenge with limited therapeutic options. Notably, the limited success of current therapies in patients with primary liver cancers (PLCs) may be attributed to the high heterogeneity of both hepatocellular carcinoma (HCCs) and intrahepatic cholangiocarcinoma (iCCAs). This heterogeneity evolves over time as tumor-initiating stem cells, or cancer stem cells (CSCs), undergo (epi)genetic alterations or encounter microenvironmental changes within the tumor microenvironment. These modifications enable CSCs to exhibit plasticity, differentiating into various resistant tumor cell types. Addressing this challenge requires urgent efforts to develop personalized treatments guided by biomarkers, with a specific focus on targeting CSCs. The lack of effective precision treatments for PLCs is partly due to the scarcity of ex vivo preclinical models that accurately capture the complexity of CSC-related tumors and can predict therapeutic responses. Fortunately, recent advancements in the establishment of patient-derived liver cancer cell lines and organoids have opened new avenues for precision medicine research. Notably, patient-derived organoid (PDO) cultures have demonstrated self-assembly and self-renewal capabilities, retaining essential characteristics of their respective in vivo tissues, including both inter- and intratumoral heterogeneities. The emergence of PDOs derived from PLCs serves as patient avatars, enabling preclinical investigations for patient stratification, screening of anticancer drugs, efficacy testing, and thereby advancing the field of precision medicine. This review offers a comprehensive summary of the advancements in constructing PLC-derived PDO models. Emphasis is placed on the role of CSCs, which not only contribute significantly to the establishment of PDO cultures but also faithfully capture tumor heterogeneity and the ensuing development of therapy resistance. The exploration of PDOs' benefits in personalized medicine research is undertaken, including a discussion of their limitations, particularly in terms of culture conditions, reproducibility, and scalability.
Collapse
Affiliation(s)
- Julien Giron-Michel
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France.
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France.
| | - Maël Padelli
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- Department of Biochemistry and Oncogenetics, Paul Brousse Hospital, AP-HP, Villejuif, France
| | - Estelle Oberlin
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Hind Guenou
- INSERM UMR-S-MD 1197, Paul-Brousse Hospital, Villejuif, France
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
| | - Jean-Charles Duclos-Vallée
- Orsay-Vallée Campus, Paris-Saclay University, Gif-sur-Yvette, France
- INSERM UMR-S 1193, Paul Brousse Hospital, Villejuif, France
- Hepato-Biliary Department, Paul Brousse Hospital, APHP, Villejuif, France
- Fédération Hospitalo-Universitaire (FHU) Hepatinov, Villejuif, France
| |
Collapse
|
28
|
Yao Y, Zhang M, Liu D, Liu X, Li Q, Wang X. Changes in systemic immune-inflammation index (SII) predict the prognosis of patients with hepatitis B-related hepatocellular carcinoma treated with lenvatinib plus PD-1 inhibitors. Clin Transl Oncol 2025; 27:1155-1165. [PMID: 39153177 DOI: 10.1007/s12094-024-03596-0] [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: 04/30/2024] [Accepted: 07/03/2024] [Indexed: 08/19/2024]
Abstract
PURPOSE This study aimed to evaluate the prognostic significance of changes in inflammatory markers in patients with Hepatitis B virus-related hepatocellular carcinoma (HBV-HCC) treated with first-line lenvatinib plus a programmed cell death protein 1 (PD-1) inhibitor. METHODS This study retrospectively included 117 HBV-HCC patients treated with first-line lenvatinib in combination with a PD-1 inhibitor. Independent factors affecting progression-free survival (PFS) and overall survival (OS) were explored based on baseline indicators and inflammatory markers changes after one treatment cycle. RESULTS Multivariate analysis revealed that an alpha-fetoprotein (AFP) level ⩾ 400 ng/mL [hazard ratio (HR), 1.69; 95% confidence interval (CI), 1.11-2.58; P = 0.01] was identified as an independent risk factor, platelet-to-neutrophil ratio (PNR) ⩽ 65.43 (HR 0.50; 95% CI 0.30-0.84; P < 0.01 ) and SII ⩽ 539.47 (HR 0.54; 95% CI 0.30-0.96; P = 0.03) were identified as independent protective factors for PFS. Additionally, multivariate analysis demonstrated that AFP ⩾ 400 ng/mL, HBV-HCC patients with diabetes mellitus (DM), and SII > 303.66 were independent risk factors of OS. The patients whose SII had increased after one cycle of treatment showed a poorer PFS (HR 1.61; 95 %CI 1.10-2.37; P = 0.015) and OS (HR 1.76; 95 % CI 1.15-2.70; P = 0.009) than patients whose SII had decreased. The objective response rate (ORR) was higher in the SII-decreased patients (47.5% vs 32.5%, P = 0.11). Mann-Whitney test found a significant difference in therapeutic response between the SII-increased patients and the SII-decreased patients (P = 0.04). CONCLUSION SII can be associated with outcomes in patients with HBV-HCC treated with first-line lenvatinib plus PD-1 inhibitors.
Collapse
Affiliation(s)
- Yang Yao
- Department of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China
| | - Minyue Zhang
- Department of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China
| | - Di Liu
- Department of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China
| | - Xiaoni Liu
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China
| | - Quanwei Li
- Beijing Institute of Hepatology, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China
| | - Xiaojun Wang
- Department of Integrated Traditional Chinese and Western Medicine, Beijing Youan Hospital, Capital Medical University, No. 8 Xi Tou Tiao, You An Men Wai, Feng Tai District, Beijing, 100069, China.
| |
Collapse
|
29
|
Peris Alvà H, Cano Rodríguez C, Bosch Barragan F, Framis Utset A, Novell Teixidó F, Prieto Del Rey MJ. New-onset hepatic lesions in oncological patients: A challenging diagnosis. RADIOLOGIA 2025; 67:191-201. [PMID: 40187811 DOI: 10.1016/j.rxeng.2023.07.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Accepted: 07/02/2023] [Indexed: 04/07/2025]
Abstract
The appearance of new-onset liver lesions is frequent during imaging follow-up of oncological patients. Most of these lesions will be metastases. But in the presence of atypical radiological findings, there are other diagnoses to consider. Hepatic abscesses, focal nodular hyperplasia-like in patients treated with platinum salts, or hepatocarcinoma in cirrhotic patients are examples of lesions that may appear in the imaging follow-up and should not be confused with metastases. It is essential to establish the nature of the lesion as this will determine the therapeutic management and might avoid unnecessary invasive procedures. The evaluation of previous radiological studies and the global vision of the patient will be primordial. While liver MRI is mainly the indicated imaging technique for these cases, sometimes a biopsy will be unavoidable. In this article, we will discuss through clinical cases some new-onset liver lesions in oncological patients that generated diagnostic doubts and will explain how to orient the diagnosis.
Collapse
Affiliation(s)
- H Peris Alvà
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain.
| | - C Cano Rodríguez
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain
| | - F Bosch Barragan
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain
| | - A Framis Utset
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain
| | - F Novell Teixidó
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain
| | - M J Prieto Del Rey
- Servicio de Radiodiagnóstico (UDIAT), Consorci Sanitari Parc Taulí, Sabadell, Barcelona, Spain
| |
Collapse
|
30
|
Park C, Hwang G, Choi WM, Han JE, Kim C, Lee DY, Heo S, Park RW. Baseline Alpha-Fetoprotein Elevation and the Risk of Hepatocellular Carcinoma in Chronic Hepatitis B: A Multicentre Cohort Study. J Viral Hepat 2025; 32:e70006. [PMID: 39878696 DOI: 10.1111/jvh.70006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/16/2025] [Indexed: 01/31/2025]
Abstract
Alpha-fetoprotein (AFP) level and its changes in chronic hepatitis B (CHB) may influence the risk of future hepatocellular carcinoma (HCC). This study aims to evaluate the HCC risk in CHB patients with no overt HCC but with elevated AFP level and to explore the prognostic role of longitudinal changes in AFP and liver-related laboratory values. This multicentre cohort study included 10,639 CHB patients without a history of HCC from seven medical facilities in South Korea. Patients with a baseline serum AFP test and no HCC diagnosis on imaging within 3 months were included. Patients were categorised into high-AFP (≥ 10 ng/mL) and normal-AFP (< 10 ng/mL) groups. The primary outcome was the incidence of HCC within 2 years, with secondary outcomes focused on longitudinal changes in AFP and liver-related laboratory values. Propensity score matching (PSM) and Cox proportional hazard models were used to assess HCC risk. After 1:4 PSM, 1278 high-AFP and 3731 normal-AFP patients were analysed. The high-AFP group had a significantly higher 2-year incidence of HCC (HR: 4.29; 95% CI: 3.31-5.57). AFP levels increased in patients who developed HCC in both groups (p < 0.01). Among the high-AFP group, patients who did not develop HCC had elevated baseline alanine aminotransferase levels (p < 0.01), which decreased during follow-up (p < 0.01) unlike those who developed HCC. In conclusion, baseline AFP elevation in CHB patients is associated with an increased risk of developing HCC within 2 years. Longitudinal monitoring of AFP and liver-related laboratory values can help in risk stratification.
Collapse
Affiliation(s)
- ChulHyoung Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Gyubeom Hwang
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Won-Mook Choi
- Department of Gastroenterology, Liver Center, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Ji Eun Han
- Department of Gastroenterology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Chungsoo Kim
- Department of Internal Medicine, Section of Cardiovascular Medicine, Yale University School of Medicine, New Haven, Connecticut, USA
- Center for Outcomes Research and Evaluation, Yale-New Haven Hospital, New Haven, Connecticut, USA
| | - Dong Yun Lee
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Subin Heo
- Department of Radiology and Research Institute of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Rae Woong Park
- Department of Biomedical Informatics, Ajou University School of Medicine, Suwon, Republic of Korea
- Department of Biomedical Sciences, Ajou University Graduate School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
31
|
Lee IC, Lei HJ, Wang LC, Yeh YC, Chau GY, Hsia CY, Chou SC, Luo JC, Hou MC, Huang YH. M2BPGi Correlated with Immunological Biomarkers and Further Stratified Recurrence Risk in Patients with Hepatocellular Carcinoma. Liver Cancer 2025; 14:68-79. [PMID: 40144467 PMCID: PMC11936441 DOI: 10.1159/000540802] [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: 02/27/2024] [Accepted: 08/06/2024] [Indexed: 03/28/2025] Open
Abstract
Introduction Novel biomarkers reflecting liver fibrosis and the immune microenvironment may correlate with the risk of hepatocellular carcinoma (HCC) recurrence. This study aimed to evaluate the prognostic value of serum biomarkers in predicting HCC recurrence. Methods Serum biomarkers, including M2BPGi, IL-6, IL-10, CCL5, VEGF-A, soluble PD-1, PD-L1, TIM-3, and LAG-3, were measured in 247 patients with HCC undergoing surgical resection. Factors associated with recurrence-free survival (RFS) and overall survival (OS) were evaluated. The ERASL-post model and IMbrave050 criteria were used to define HCC recurrence risk groups. Results Serum M2BPGi levels significantly correlated with FIB-4 score, aspartate transaminase-to-platelet ratio index, ALBI score, alpha-fetoprotein (AFP), alanine transaminase, aspartate transaminase, IL-10, CCL5, VEGF-A, soluble PD-1, PD-L1, TIM-3, and LAG-3 levels. M2BPGi, VEGF-A, soluble PD-1, and TIM-3 levels significantly correlated with RFS. In multivariate analysis, M2BPGi >1.5 COI (hazard ratio [HR] = 2.100, p < 0.001), tumor size >5 cm (HR = 1.859, p = 0.002), multiple tumors (HR = 2.562, p < 0.001), AFP >20 ng/mL (HR = 2.141, p < 0.001), and microvascular invasion (HR = 1.954, p = 0.004) were independent predictors of RFS. M2BPGi levels significantly stratified the recurrence risk in ERASL-post and IMbrave050 risk groups. An M2BPGi-based model could significantly discriminate RFS in the overall cohort as well as in the IMbrave050 low- and high-risk groups. M2BPGi >1.5 COI was also an independent predictor of OS after resection (HR = 2.707, p < 0.001). Conclusion Serum M2BPGi levels significantly correlated with surrogate markers of liver fibrosis, liver function, and immunology. M2BPGi is a significant predictor of HCC recurrence and survival after resection and could be incorporated into recurrence-prediction models.
Collapse
Affiliation(s)
- I-Cheng Lee
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Hao-Jan Lei
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Lei-Chi Wang
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Yi-Chen Yeh
- Department of Pathology and Laboratory Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Gar-Yang Chau
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Cheng-Yuan Hsia
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Shu-Cheng Chou
- Department of Surgery, Taipei Veterans General Hospital, Taipei, Taiwan
| | - Jiing-Chyuan Luo
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Ming-Chih Hou
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
| | - Yi-Hsiang Huang
- Division of Gastroenterology and Hepatology, Department of Medicine, Taipei Veterans General Hospital, Taipei, Taiwan
- College of Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei, Taiwan
- Healthcare and Service Center, Taipei Veterans General Hospital, Taipei, Taiwan
| |
Collapse
|
32
|
Yin X, Deng N, Ding XY, Chen JL, Sun W. CRAFITY score and nomogram predict the clinical efficacy of lenvatinib combined with immune checkpoint inhibitors in hepatocellular carcinoma. World J Gastroenterol 2025; 31:101672. [PMID: 39991685 PMCID: PMC11755258 DOI: 10.3748/wjg.v31.i7.101672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/03/2024] [Accepted: 12/30/2024] [Indexed: 01/20/2025] Open
Abstract
BACKGROUND The CRAFITY score is mainly utilized for hepatocellular carcinoma (HCC) patients receiving atezolizumab and bevacizumab, with little investigation in its predictive capacity for alternative regimens, such as lenvatinib and programmed cell death protein 1 (PD-1) inhibitors, which are widely utilized in Chinese clinical practice. AIM To look at the predictive significance of the CRAFITY score in HCC patients taking lenvatinib and PD-1 inhibitors. METHODS The retrospective investigation consisted of 192 patients with incurable HCC who received lenvatinib and PD-1 inhibitors between January 2018 and January 2022. Patients were stratified according to CRAFITY score (based on baseline alpha-fetoprotein and C-reactive protein levels) into CRAFITY-low, CRAFITY-intermediate, and CRAFITY-high groups. Overall survival (OS) and progression-free survival (PFS) were assessed using Kaplan-Meier analysis, and independent prognostic factors were identified through Cox regression analysis. Nomograms were created to forecast survival for a year. RESULTS The median PFS and OS were the longest for patients in the CRAFITY-low group, followed by those in the CRAFITY-intermediate and CRAFITY-high groups (median PFS: 8.4 months, 6.0 months, and 3.1 months, P < 0.0001; median OS: 33.4 months, 19.2 months, and 6.6 months, P < 0.0001). Both the objective response rate (5%, 19.6%, and 22%, P = 0.0669) and the disease control rate (50%, 76.5%, and 80%, P = 0.0023) were considerably lower in the CRAFITY-high group. The findings from the multivariate analysis showed that a nomogram which included the tumor number, prior transarterial chemoembolization history, and CRAFITY score predicted 12-month survival with an area under the curve of 0.788 (95% confidence interval: 0.718-0.859), which was in good agreement with actual data. CONCLUSION The CRAFITY score is a valuable predictor of survival and treatment outcomes in patients receiving lenvatinib and PD-1 inhibitors.
Collapse
Affiliation(s)
- Xue Yin
- Department of Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Na Deng
- Department of Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Xiao-Yan Ding
- Department of Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Jing-Long Chen
- Department of Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| | - Wei Sun
- Department of Cancer Center, Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China
| |
Collapse
|
33
|
Mullick Chowdhury S, Hong F, Rolfo C, Li Z, He K, Wesolowski R, Mortazavi A, Meng L. CNPY2 in Solid Tumors: Mechanisms, Biomarker Potential, and Therapeutic Implications. BIOLOGY 2025; 14:214. [PMID: 40001982 PMCID: PMC11851889 DOI: 10.3390/biology14020214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Revised: 02/10/2025] [Accepted: 02/16/2025] [Indexed: 02/27/2025]
Abstract
Canopy FGF signaling regulator 2 (CNPY2) has emerged as a crucial player in cancer development by promoting cell proliferation, tissue repair, and angiogenesis. This review synthesizes the current understanding of CNPY2's role in solid tumors, particularly renal cell carcinoma, prostate cancer, hepatocellular carcinoma, and non-small-cell lung cancer. CNPY2 modulates key pathways such as p53, MYLIP, NF-κB, and AKT/GSK3β, thereby driving tumor growth and progression. In renal cell carcinoma, CNPY2 paradoxically promotes tumor growth through p53 upregulation, while in hepatocellular carcinoma, CNPY2 drives cell cycle progression via p53 destabilization. In prostate cancer, it enhances tumor progression by stabilizing androgen receptors through MYLIP interaction, and in non-small-cell lung cancer, it contributes to chemoresistance and metastasis through NF-κB and AKT/GSK3β signaling. Additionally, CNPY2 influences the tumor microenvironment, impacting immune function and metastatic potential. As a potential biomarker, CNPY2 shows promise for cancer detection and prognosis, particularly when used in combination with other markers. Early therapeutic strategies, including siRNA and miRNA approaches, are under exploration, though challenges remain due to CNPY2's expression in normal tissues and potential off-target effects. This review underscores the need for further research to fully elucidate CNPY2's oncogenic mechanisms and develop targeted therapies. Improved understanding of CNPY2's diverse roles may lead to novel diagnostic and therapeutic approaches in solid tumors.
Collapse
Affiliation(s)
- Sayan Mullick Chowdhury
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Feng Hong
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Christian Rolfo
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Zihai Li
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Kai He
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
- Pelotonia Institute for Immuno-Oncology, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA
| | - Robert Wesolowski
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Amir Mortazavi
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| | - Lingbin Meng
- Division of Medical Oncology, Department of Internal Medicine, The Ohio State University Comprehensive Cancer Center, Columbus, OH 43210, USA; (S.M.C.); (F.H.); (C.R.); (Z.L.); (K.H.); (R.W.); (A.M.)
| |
Collapse
|
34
|
Gu Y, Jin K, Gao S, Sun W, Yin M, Han J, Zhang Y, Wang X, Zeng M, Sheng R. A preoperative nomogram with MR elastography in identifying cytokeratin 19 status of hepatocellular carcinoma. Br J Radiol 2025; 98:210-219. [PMID: 39657213 DOI: 10.1093/bjr/tqae193] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 08/30/2024] [Accepted: 09/15/2024] [Indexed: 12/17/2024] Open
Abstract
OBJECTIVES Developing a nomogram integrating MR elastography (MRE)-based tumour stiffness and contrast-enhanced MRI in identifying cytokeratin 19 (CK19) status of hepatocellular carcinoma (HCC) preoperatively. METHODS One hundred twenty CK19-negative HCC and 39 CK19-positive HCC patients undergoing curative resection were prospectively evaluated. All received MRE and contrast-enhanced MRI. Clinical and MRI tumour features were compared. Univariate and multivariate logistic regression analyses identified independent predictors for CK19 status. Receiver operating characteristic curve analysis evaluated diagnostic performance. A nomogram was established with calibration and decision curve analysis. RESULTS Multivariate analysis revealed serum alpha fetoprotein (AFP) level (P < 0.001), targetoid appearance (P = 0.007), and tumour stiffness (P = 0.011) as independent significant variables for CK19-positive HCC. The area under the curve for tumour stiffness was 0.729 (95% confidence interval [CI] 0.653, 0.796). Combining these features, a nomogram-based model achieved an area under the curve value of 0.844 (95% CI 0.778, 0.897), with sensitivity, specificity, and accuracy of 76.92%, 85.00%, and 83.02%, respectively. Calibration and decision curve analyses demonstrated good agreement and optimal net benefit. CONCLUSIONS MRE-measured tumour stiffness aids in predicting CK19 status in HCC. The combined nomogram incorporating tumour stiffness, targetoid appearance, and AFP provides a reliable biomarker for CK19-positive HCC. ADVANCES IN KNOWLEDGE MRE-measured tumour stiffness can be used to predict CK19 status in HCC. The nomogram, which integrates tumour stiffness, targetoid appearance, and AFP levels, has shown improved diagnostic performance. It offers a comprehensive preoperative tool for clinical decision-making, further advancing personalized treatment strategies in HCC management.
Collapse
Affiliation(s)
- Yanan Gu
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Kaipu Jin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Geriatric Medical Center, Zhongshan Hospital, Fudan University, Shanghai 201104, China
| | - Shanshan Gao
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Wei Sun
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Minyan Yin
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Jing Han
- Department of Pathology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Yunfei Zhang
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| | - Xiaolin Wang
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Mengsu Zeng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
- Department of Cancer Center, Zhongshan Hospital, Fudan University, Shanghai 200032, China
| | - Ruofan Sheng
- Department of Radiology, Zhongshan Hospital, Fudan University, Shanghai 200032, China
- Shanghai Institute of Medical Imaging, Shanghai 200032, China
| |
Collapse
|
35
|
Cai C, Wang L, Tao L, Zhu H, Ren Y, Li D, Li D. Imaging-Based Prediction of Ki-67 Expression in Hepatocellular Carcinoma: A Retrospective Study. Cancer Med 2025; 14:e70562. [PMID: 39964132 PMCID: PMC11834164 DOI: 10.1002/cam4.70562] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/04/2024] [Accepted: 12/20/2024] [Indexed: 02/21/2025] Open
Abstract
AIM This study aims to develop a non-invasive, preoperative predictive model for Ki-67 expression in HCC patients using enhanced computed tomography (CT) and clinical indicators to improve patient outcomes. METHODS This retrospective study analyzed 595 post-curative hepatectomy HCC patients. Patients were categorized into high (> 20%) and low (≤ 20%) Ki-67 expression groups based on cellular proliferation levels. Radiomic features were extracted from enhanced CT scans and combined with clinical parameters to develop a predictive model for Ki-67 expression. RESULTS Key clinical factors impacting Ki-67 expression in HCC included alpha-fetoprotein (AFP), non-smooth tumor margin, ill-defined pseudo-capsule, and peritumoral star node. From 1441 initially extracted radiomic features, 16 key features were selected using Lasso regression. These features were used to develop a radiomics model, which, when combined with clinical data, yielded an integrated predictive model with high accuracy. The combined model achieved an area under the curve (AUC) of 0.854 in the training group and 0.839 in the validation group. A nomogram based on this model was constructed, and its predictive accuracy was validated through calibration curves and decision curve analysis. A risk scorecard model was also constructed as a practical tool for clinicians to assess the risk level of high Ki-67 expression, facilitating personalized treatment planning. Survival analysis demonstrated significant differences in 3-year overall survival (OS) and progression-free survival (PFS) rates between patients with high and low Ki-67 expression, indicating the model's strong prognostic capability. CONCLUSIONS This study successfully developed a comprehensive model that integrates radiomic and clinical data for the preoperative prediction of Ki-67 expression in HCC patients.
Collapse
Affiliation(s)
- Chiyu Cai
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Liancai Wang
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Lianyuan Tao
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Hengli Zhu
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Yongnian Ren
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Deyu Li
- Department of Hepatobiliary and Pancreatic SurgeryZhengzhou University People's HospitalZhengzhouChina
| | - Dongxiao Li
- Department of Digestive DiseasesZhengzhou University People's HospitalZhengzhouChina
| |
Collapse
|
36
|
Shi J, Zhu X, Yang JB. Advances and challenges in molecular understanding, early detection, and targeted treatment of liver cancer. World J Hepatol 2025; 17:102273. [PMID: 39871899 PMCID: PMC11736488 DOI: 10.4254/wjh.v17.i1.102273] [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: 10/14/2024] [Revised: 11/12/2024] [Accepted: 11/27/2024] [Indexed: 01/06/2025] Open
Abstract
In this review, we explore the application of next-generation sequencing in liver cancer research, highlighting its potential in modern oncology. Liver cancer, particularly hepatocellular carcinoma, is driven by a complex interplay of genetic, epigenetic, and environmental factors. Key genetic alterations, such as mutations in TERT, TP53, and CTNNB1, alongside epigenetic modifications such as DNA methylation and histone remodeling, disrupt regulatory pathways and promote tumorigenesis. Environmental factors, including viral infections, alcohol consumption, and metabolic disorders such as nonalcoholic fatty liver disease, enhance hepatocarcinogenesis. The tumor microenvironment plays a pivotal role in liver cancer progression and therapy resistance, with immune cell infiltration, fibrosis, and angiogenesis supporting cancer cell survival. Advances in immune checkpoint inhibitors and chimeric antigen receptor T-cell therapies have shown potential, but the unique immunosuppressive milieu in liver cancer presents challenges. Dysregulation in pathways such as Wnt/β-catenin underscores the need for targeted therapeutic strategies. Next-generation sequencing is accelerating the identification of genetic and epigenetic alterations, enabling more precise diagnosis and personalized treatment plans. A deeper understanding of these molecular mechanisms is essential for advancing early detection and developing effective therapies against liver cancer.
Collapse
Affiliation(s)
- Ji Shi
- Department of Research and Development, Ruibiotech Company Limited, Beijing 100101, China
| | - Xu Zhu
- Department of Research and Development, Ruibiotech Company Limited, Beijing 100101, China
| | - Jun-Bo Yang
- Shenzhen Branch, Guangdong Laboratory of Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518000, Guangdong Province, China.
| |
Collapse
|
37
|
Zhang W, Zhong Y, Wang J, Zou G, Chen Q, Liu C. Direct repeat region 3' end modifications regulate Cas12a activity and expand its applications. Nucleic Acids Res 2025; 53:gkaf040. [PMID: 39883010 PMCID: PMC11780881 DOI: 10.1093/nar/gkaf040] [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: 12/02/2024] [Revised: 01/13/2025] [Accepted: 01/16/2025] [Indexed: 01/31/2025] Open
Abstract
CRISPR-Cas12a technology has transformative potential, but as its applications grow, enhancing its inherent functionalities is essential to meet diverse demands. Here, we reveal a regulatory mechanism for LbCas12a through direct repeat (DR) region 3' end modifications and de-modifications, which can regulate LbCas12a's cis- and trans-cleavage activities. We extensively explored the effects of introducing phosphorylation, DNA, photo-cleavable linker, DNA modifications at the DR 3' end on LbCas12a's functionality. We find that the temporary inhibitory function of Cas12a can be reactivated by DR 3' end modification corresponding substances, such as alkaline phosphatase (ALP), immunoglobulin G (IgG), alpha-fetoprotein (AFP), DNA exonucleases, ultraviolet radiation, and DNA glycosylases, which greatly expand the scope of application of Cas12a. Clinical applications demonstrated promising results in ALP, AFP, and trace Epstein-Barr virus detection compared to gold standard methods. Our research provides valuable insights into regulating LbCas12a activity through direct modification of DR and significantly expands its potential clinical detection targets, paving the way for future universal clustered regularly interspaced short palindromic repeats (CRISPR) diagnostic strategies.
Collapse
Affiliation(s)
- Wei Zhang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Yinyin Zhong
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Jiaqi Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Guangrong Zou
- Department of Clinical Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| | - Qiaozhen Chen
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, P.R. China
| | - Chaoxing Liu
- Guangdong Provincial Key Laboratory of Digestive Cancer Research, Digestive Diseases Center, Scientific Research Center, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen, Guangdong 518107, P.R. China
| |
Collapse
|
38
|
Hu M, Zhang Y, Zhang P, Liu K, Zhang M, Li L, Yu Z, Zhang X, Zhang W, Xu Y. Targeting APE1: Advancements in the Diagnosis and Treatment of Tumors. Protein Pept Lett 2025; 32:18-33. [PMID: 39648425 DOI: 10.2174/0109298665338519241114103223] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 12/10/2024]
Abstract
With the emergence of the precision medicine era, targeting specific proteins has emerged as a pivotal breakthrough in tumor diagnosis and treatment. Apurinic/apyrimidinic Endonuclease 1 (APE1) is a multifunctional protein that plays a crucial role in DNA repair and cellular redox regulation. This article comprehensively explores the fundamental mechanisms of APE1 as a multifunctional enzyme in biology, with particular emphasis on its potential significance in disease diagnosis and strategies for tumor treatment. Firstly, this article meticulously analyzes the intricate biological functions of APE1 at a molecular level, establishing a solid theoretical foundation for subsequent research endeavors. In terms of diagnostic applications, the presence of APE1 can be detected in patient serum samples, biopsy tissues, and through cellular in situ testing. The precise detection methods enable changes in APE1 levels to serve as reliable biomarkers for predicting tumor occurrence, progression, and patient prognosis. Moreover, this article focuses on elucidating the potential role of APE1 in tumor treatment by exploring various inhibitors, including nucleic acid-based inhibitors and small molecule drug inhibitors categories, and revealing their unique advantages in disrupting DNA repair function and modulating oxidative-reduction activity. Finally, the article provides an outlook on future research directions for APE1 while acknowledging major technical difficulties and clinical challenges that need to be overcome despite its immense potential as a target for tumor therapy.
Collapse
Affiliation(s)
- Minghui Hu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Yingyu Zhang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital, and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China
| | - Pin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Kangbo Liu
- Henan Institute for Drug and Medical Device Inspection (Henan Vaccine Issuance Center), Zhengzhou, 450018, China
| | - Mengxin Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Lifeng Li
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Zhidan Yu
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Xianwei Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Wancun Zhang
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan International Joint Laboratory for Prevention and Treatment of Pediatric Disease, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
- Henan Key Laboratory of Children's Genetics and Metabolic Diseases, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| | - Ying Xu
- Health Commission of Henan Province Key Laboratory for Precision Diagnosis and Treatment of Pediatric Tumor, Children's Hospital Affiliated to Zhengzhou University, Zhengzhou, 450018, China
| |
Collapse
|
39
|
Li YT, Zeng XZ. Establishment and Validation of the Novel Necroptosis-related Genes for Predicting Stemness and Immunity of Hepatocellular Carcinoma via Machine-learning Algorithm. Comb Chem High Throughput Screen 2025; 28:146-165. [PMID: 39641162 DOI: 10.2174/0113862073271292231108113547] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 08/21/2023] [Accepted: 09/21/2023] [Indexed: 12/07/2024]
Abstract
BACKGROUND Necroptosis, a recently identified mechanism of programmed cell death, exerts significant influence on various aspects of cancer biology, including tumor cell proliferation, stemness, metastasis, and immunosuppression. However, the role of necroptosis-related genes (NRGs) in Hepatocellular Carcinoma (HCC) remains elusive. METHODS In this study, we assessed the mutation signature, copy number variation, and expression of 37 NRGs in HCC using the TCGA-LIHC dataset. We further validated our results using the ICGC-LIRI-JP dataset. To construct our prognostic model, we utilized the least absolute shrinkage and selection operator (LASSO), and evaluated the predictive efficacy of the NRGs-score using various machine learning algorithms, including K-M curves, time-ROC curves, univariate and multivariate Cox regression, and nomogram. In addition, we analyzed immune infiltration using the CIBERSOFT and ssGSEA algorithms, calculated the stemness index through the one-class logistic regression (OCLR) algorithm, and performed anti-cancer stem cells (CSCs) drug sensitivity analysis using oncoPredict. Finally, we validated the expression of the prognostic NRGs through qPCR both in vitro and in vivo. RESULTS About 18 out of 37 NRGs were found to be differentially expressed in HCC and correlated with clinical outcomes. To construct a prognostic model, six signature genes (ALDH2, EZH2, PGAM5, PLK1, SQSTM1, and TARDBP) were selected using LASSO analysis. These genes were then employed to categorize HCC patients into two subgroups based on NRGs-score (low vs. high). A high NRGs score was associated with a worse prognosis. Furthermore, univariate and multivariate Cox regression analyses were performed to confirm the NRGs-score as an independent risk factor. These analyses revealed strong associations between NRGs-score and critical factors, such as AFP, disease stage, and tumor grade in the HCC cohort. NRGs-score effectively predicted the 1-, 3-, and 5-year survival of HCC patients. Immune infiltration analysis further revealed that the expression of immune checkpoint molecules was significantly enhanced in the high NRGs-score group. Stemness analysis in the HCC cohort showed that NRGs-score was positively correlated with mRNA stemness index, and patients with high NRGs-score were sensitive to CSCs inhibitors. The findings from the external validation cohort provided confirmation that the NRGs-score presented a trait with universal applicability in accurately predicting the survival of HCC. Additionally, the six prognostic genes were consistently differentially expressed in both the HCC cell line and the mouse HCC model. CONCLUSION Our study demonstrated the pivotal role of NRGs in promoting stemness and immune suppression in HCC and established a robust model which could successfully predict HCC prognosis.
Collapse
Affiliation(s)
- Yao-Ting Li
- Department of Forensic Science, Guangdong Police College, 500 Binjiang East Road, Guangzhou 510230, Guangdong, China
| | - Xue-Zhen Zeng
- Department of Pharmacy, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou 510080, Guangdong, China
| |
Collapse
|
40
|
Long Z, Zhang L. Detection of Hepatocellular Carcinoma Using Optimized miRNA Combinations and Interpretable Machine Learning Models. IEEE ACCESS 2025; 13:66078-66093. [DOI: 10.1109/access.2025.3559105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/03/2025]
Affiliation(s)
- Zhengwu Long
- College of Life Science and Technology, Huazhong Agricultural University, Wuhan, Hubei, China
| | - Lisheng Zhang
- Bio-Medical Center, College of Veterinary Medicine, Huazhong Agricultural University, Wuhan, Hubei, China
| |
Collapse
|
41
|
Benderski K, Schneider P, Kordeves P, Fichter M, Schunke J, De Lorenzi F, Durak F, Schrörs B, Akilli Ö, Kiessling F, Bros M, Diken M, Grabbe S, Schattenberg JM, Lammers T, Sofias AM, Kaps L. A hepatocellular carcinoma model with and without parenchymal liver damage that integrates technical and pathophysiological advantages for therapy testing. Pharmacol Res 2025; 211:107560. [PMID: 39730106 DOI: 10.1016/j.phrs.2024.107560] [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: 10/01/2024] [Revised: 12/20/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Hepatocellular Carcinoma (HCC) is the most common form of primary liver cancer, with cirrhosis being its strongest risk factor. Interestingly, an increasing number of HCC cases is also observed without cirrhosis. We developed an HCC model via intrasplenic injection of highly tumorigenic HCC cells, which, due to cellular tropism, invade the liver and allow for a controllable disease progression. Specifically, C57BL/6JRj mice were intrasplenically inoculated with Dt81Hepa1-6 HCC cells, with a subgroup pre-treated with CCl4 to induce cirrhosis (C-HCC). At four weeks post-inoculation, mice were sacrificed, and diseased livers were analyzed via histology, flow cytometry, and RT-qPCR to profile the extracellular matrix (ECM), angiogenesis, and immune cells. In addition, tumor-bearing mice were treated with the first-line therapy, AtezoBev, to assess therapeutic responsiveness of the model. Dt81Hepa1-6 cells displayed similar gene expression as human HCC. After intrasplenic injection, all mice developed multifocal disease. C-HCC mice had a significantly higher tumor load than non-cirrhotic HCC mice. Both HCC and C-HCC models displayed extensive ECM formation, increased levels of vascularization, and immune cell infiltration compared to healthy and non-cancerous cirrhotic livers. AtezoBev treatment produced robust antitumor efficacy, validating the model's suitability for therapy testing. In conclusion, we established a rapidly developing and high-yield HCC model through a simple intrasplenic injection, with or without cirrhotic damage. The model overexpressed key human HCC genes and showed high responsiveness to first-line treatment. Our model uniquely combines all the above-mentioned features, promoting its use towards HCC therapy testing.
Collapse
Affiliation(s)
- Karina Benderski
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Paul Schneider
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Panayiotis Kordeves
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Michael Fichter
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Max Planck Institute for Polymer Research, Ackermannweg 10, Mainz 55128, Germany
| | - Jenny Schunke
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Federica De Lorenzi
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany
| | - Feyza Durak
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Barbara Schrörs
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Özlem Akilli
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany
| | - Matthias Bros
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Mustafa Diken
- TRON - Translational Oncology at the University Medical Center of the Johannes Gutenberg-University Mainz GmbH, Freiligrathstrasse 12, Mainz 55131, Germany
| | - Stephan Grabbe
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany
| | - Jörn M Schattenberg
- Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany
| | - Twan Lammers
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Alexandros Marios Sofias
- Department of Nanomedicine and Theranostics, Institute for Experimental Molecular Imaging (ExMI), RWTH Aachen University Hospital, Forckenbeckstrasse 55, Aachen 52074, Germany; Mildred Scheel School of Oncology (MSSO), Center for Integrated Oncology Aachen (CIOA), RWTH Aachen University Hospital, Pauwelsstrasse 30, Aachen 52074, Germany.
| | - Leonard Kaps
- Department of Dermatology, University Medical Center of the Johannes Gutenberg-University, Langenbeckstrasse 1, Mainz 55131, Germany; Department of Medicine II, Saarland University Medical Center, Saarland University, Kirrberger Strasse 100, Saarbrücken 66123, Germany.
| |
Collapse
|
42
|
Ramakrishnan K, Sanjeev D, Rehman N, Raju R. A Network Map of Intracellular Alpha-Fetoprotein Signalling in Hepatocellular Carcinoma. J Viral Hepat 2025; 32:e14035. [PMID: 39668590 DOI: 10.1111/jvh.14035] [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: 08/06/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 12/14/2024]
Abstract
Alpha fetoprotein (AFP) is a glycoprotein of foetal origin belonging to the albumin protein family. Serum AFP is a long-conceived early-diagnostic biomarker for HCC with its elevated expression in different liver pathologies ranging from hepatitis viral infections to fibrosis, cirrhosis, and HCC. Beyond their utility as biomarkers, in support of its contribution to these clinical outcomes, the function of AFP as an immune suppressor and inducer of malignant transformation in HCC patients is well reported. Multiple reports show that AFP is secreted by hepatocytes, binds to its cognate receptor, AFP-receptor (AFPR), and exerts its actions. However, there is only limited information available in this context. There is an urgent need to gather more insight into the AFP signalling pathway and consider it a classical intracellular signalling pathway, among others. AFP is a highly potent intracellular molecule that has the potential to bind to many interactors like PTEN, Caspase, RAR, and so on. It has been shown that cellular AFP and secreted AFP have different roles in HCC pathophysiology, and a comprehensive map of the AFP signalling pathway is warranted for further theranostic applications.
Collapse
Affiliation(s)
| | - Diya Sanjeev
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, India
| | - Niyas Rehman
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, India
| | - Rajesh Raju
- Centre for Integrative Omics Data Science, Yenepoya (Deemed to Be University), Mangalore, India
| |
Collapse
|
43
|
Boon-yasidhi P, Karnsakul W. Non-Invasive Biomarkers and Breath Tests for Diagnosis and Monitoring of Chronic Liver Diseases. Diagnostics (Basel) 2024; 15:68. [PMID: 39795596 PMCID: PMC11720471 DOI: 10.3390/diagnostics15010068] [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: 11/27/2024] [Revised: 12/23/2024] [Accepted: 12/27/2024] [Indexed: 01/13/2025] Open
Abstract
Background: Chronic liver disease (CLD) presents a significant global health burden, demanding effective tools for diagnosis and monitoring. Traditionally, liver biopsy has been the gold standard for evaluating liver fibrosis and other chronic liver conditions. However, biopsy's invasiveness, associated risks, and sampling variability indicate the need for reliable, noninvasive alternatives. This review examines the utility of noninvasive tests (NITs) in assessing liver disease severity, progression, and therapeutic response in patients with CLD. Result: Key modalities discussed include serum biomarker panels (e.g., FIB-4, APRI, ELF), imaging techniques like transient elastography, and magnetic resonance elastography, each offering unique benefits in fibrosis staging. Emerging biomarkers such as extracellular vesicles and circulating microRNAs show promise in early detection and personalized monitoring. Comparative studies indicate that while no single NIT matches biopsy precision, combinations of these modalities improve diagnostic accuracy and patient outcomes by reducing unnecessary biopsies. Moreover, NITs are instrumental in monitoring dynamic changes in liver health, allowing for more responsive and patient-centered care. Conclusions: Challenges remain, including standardization across tests, cost considerations, and the need for larger, diverse population studies to validate findings. Despite these limitations, NITs are increasingly integrated into clinical practice, fostering a paradigm shift toward noninvasive, accessible liver disease management. Continued advancements in NITs are essential for improved patient outcomes and will likely shape the future standard of care for CLD.
Collapse
Affiliation(s)
- Pasawat Boon-yasidhi
- Department of Pharmacology, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wikrom Karnsakul
- Pediatric Liver Center, Department of Pediatric Gastroenterology, Hepatology and Nutrition, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
44
|
Büdeyri I, Guckelberger O, Oppermann E, Roy D, Sliwinski S, Becker F, Struecker B, Vogl TJ, Pascher A, Bechstein WO, Lorentzen A, Heikenwalder M, Juratli MA. Ezrin Polarization as a Diagnostic Marker for Circulating Tumor Cells in Hepatocellular Carcinoma. Cells 2024; 14:6. [PMID: 39791707 PMCID: PMC11720075 DOI: 10.3390/cells14010006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/08/2024] [Accepted: 12/24/2024] [Indexed: 01/12/2025] Open
Abstract
Hepatocellular carcinoma (HCC) is the sixth most common cancer and the third leading cause of cancer-related death worldwide, with no precise method for early detection. Circulating tumor cells (CTCs) expressing the dynamic polarity of the cytoskeletal membrane protein, ezrin, have been proposed to play a crucial role in tumor progression and metastasis. This study investigated the diagnostic and prognostic potential of polarized circulating tumor cells (p-CTCs) in HCC patients. CTCs were isolated from the peripheral blood of 20 HCC patients and 18 patients with nonmalignant liver disease (NMLD) via an OncoQuick® kit and immunostained with Ezrin-Alexa Fluor 488®, CD146-PE, and CD45-APC. A fluorescence microscopy was then performed for analysis. The HCC group exhibited significantly higher levels of p-CTCs, with median values of 0.56 p-CTCs/mL, compared to 0.02 p-CTCs/mL (p = 0.03) in the NMLD group. CTCs were detected in 95% of the HCC patients, with a sensitivity of 95% and specificity of 89%. p-CTCs were present in 75% of the HCC patients, with a sensitivity of 75% and a specificity of 94%. Higher p-CTC counts were associated with the significantly longer overall survival in HCC patients (p = 0.05). These findings suggest that p-CTCs could serve as valuable diagnostic and prognostic markers for HCC. The incorporation of p-CTCs into diagnostic strategies could enhance therapeutic decision-making and improve patient outcomes.
Collapse
Affiliation(s)
- Ibrahim Büdeyri
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Olaf Guckelberger
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Elsie Oppermann
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Dhruvajyoti Roy
- Department of Breast Surgical Oncology, The University of Texas MD Anderson Cancer Center, Houston, TX 77054, USA
| | - Svenja Sliwinski
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Felix Becker
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Benjamin Struecker
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Thomas J. Vogl
- Department of Diagnostic and Interventional Radiology, Frankfurt University Hospital, Goethe University, 60596 Frankfurt, Germany
| | - Andreas Pascher
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| | - Wolf O. Bechstein
- Department of General, Visceral and Transplant Surgery, Frankfurt University Hospital, 60596 Frankfurt, Germany
| | - Anna Lorentzen
- Department of Biomedicine, Aarhus University, 8200 Aarhus, Denmark
| | - Mathias Heikenwalder
- Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), 69120 Heidelberg, Germany
| | - Mazen A. Juratli
- Department of General, Visceral and Transplant Surgery, University Hospital Muenster, University of Muenster, Albert-Schweitzer-Campus 1, 48149 Muenster, Germany; (I.B.)
| |
Collapse
|
45
|
Chacko N, Ankri R. Non-invasive early-stage cancer detection: current methods and future perspectives. Clin Exp Med 2024; 25:17. [PMID: 39708168 DOI: 10.1007/s10238-024-01513-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 10/21/2024] [Indexed: 12/23/2024]
Abstract
This review paper explores the realm of non-invasive methods for early cancer detection. Early identification is crucial for effective therapeutic intervention, and non-invasive techniques have emerged as promising tools to enhance diagnostic accuracy and improve patient outcomes. The paper thoroughly examines the advantages, limitations, and prospects of various non-invasive approaches, including blood tests, non-blood-based tests, and diverse imaging modalities. It discusses the biomarkers found in blood for early-stage cancer detection, specifying the types of cancer associated with each biomarker. The non-blood-based tests focus on components in saliva, urine, and breath for cancer detection, alongside current studies and future perspectives on various cancers. Optical imaging methods covered in this review include fluorescence imaging in the near-infrared (NIR) region, bioluminescence imaging, and Raman spectroscopy for early-stage cancer detection. The review also highlights the pros and cons of ultrasound imaging in early-stage cancer detection. Additionally, the clinical implications of using AI for cancer detection, both present and future, are explored. This paper provides valuable insights for researchers and clinicians working in the field of non-invasive early-stage cancer detection.
Collapse
Affiliation(s)
- Neelima Chacko
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel
| | - Rinat Ankri
- Department of Physics, Faculty of Natural Science, Ariel University, 40700, Ariel, Israel.
| |
Collapse
|
46
|
Xu N, Wu Z, Pan J, Xu X, Wei Q. CAR-T cell therapy: Advances in digestive system malignant tumors. MOLECULAR THERAPY. ONCOLOGY 2024; 32:200872. [PMID: 39377038 PMCID: PMC11456800 DOI: 10.1016/j.omton.2024.200872] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Malignant tumors of the digestive system have had a notoriously dismal prognosis throughout history. Immunotherapy, radiotherapy, surgery, and chemotherapy are the primary therapeutic approaches for digestive system cancers. The rate of recurrence and metastasis, nevertheless, remains elevated. As one of the immunotherapies, chimeric antigen receptor T cell (CAR-T) therapy has demonstrated a promising antitumor effect in hematologic cancer. Despite undergoing numerous clinical trials, the ineffective antitumor effect and adverse effects of CAR-T cell therapy in the treatment of digestive system cancers continue to impede its clinical translation. It is necessary to surmount the restricted options for targeting proteins, the obstacles that impede CAR-T cell infiltration into solid tumors, and the limited survival time in vivo. We examined and summarized the developments, obstacles, and countermeasures associated with CAR-T therapy in digestive system cancers. Emphasis was placed on the regulatory functions of potential antigen targets, the tumor microenvironment, and immune evasion in CAR-T therapy. Thus, our analysis has furnished an all-encompassing comprehension of CAR-T cell therapy in digestive system cancers, which will generate tremendous enthusiasm for subsequent in-depth research into CAR-T-based therapies in digestive system cancers.
Collapse
Affiliation(s)
- Nan Xu
- Zhejiang University School of Medicine, Hangzhou 310058, China
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Zhonglin Wu
- Key Laboratory of Integrated Oncology and Intelligent Medicine of Zhejiang Province, Affiliated Hangzhou First People’s Hospital, School of Medicine, Westlake University, Hangzhou 310006, China
| | - Jun Pan
- Zhejiang University School of Medicine, Hangzhou 310058, China
| | - Xiao Xu
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
- Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou 310029, China
| | - Qiang Wei
- School of Clinical Medicine, Hangzhou Medical College, Hangzhou 310053, China
| |
Collapse
|
47
|
Wang M, Qian G, Xiao H, Liu X, Sun L, Chen Z, Lin K, Yao L, Li C, Gu L, Xu J, Sun X, Qiu W, Pawlik TM, Yee Lau W, Lv G, Shen F, Yang T. Prognostic significance of postoperative serological incomplete conversion of AFP and PIVKA-II after hepatic resection for hepatocellular carcinoma: a multicenter analysis of 1755 patients. Oncologist 2024; 29:e1723-e1733. [PMID: 38907676 PMCID: PMC11630741 DOI: 10.1093/oncolo/oyae139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 05/14/2024] [Indexed: 06/24/2024] Open
Abstract
BACKGROUND The value of serum biomarkers, particularly alpha-fetoprotein (AFP) and protein induced by vitamin K absence or antagonist-II (PIVKA-II), gains increasing attention in prognostic evaluation and recurrence monitoring for patients with hepatocellular carcinoma (HCC). This study investigated the implications of serological incomplete conversion (SIC) of these 2 biomarkers as prognostic indicators for long-term outcomes after HCC resection. METHODS A multicenter observational study was conducted on a cohort of HCC patients presenting with AFP (>20 ng/mL) or PIVKA-II (>40 mAU/mL) positivity who underwent curative-intent resection. Based on their postoperative AFP and PIVKA-II levels at first postoperative follow-up (4~8 weeks after surgery), these patients were stratified into the serological incomplete conversion (SIC) and serological complete conversion (SCC) groups. The study endpoints were recurrence and overall survival (OS). RESULTS Among 1755 patients, 379 and 1376 were categorized as having SIC and SCC, respectively. The SIC group exhibited 1- and 5-year OS rates of 67.5% and 26.3%, with the corresponding recurrence rates of 53.2% and 79.0%, respectively; while the SCC group displayed 1- and 5-year OS rates of 95.8% and 62.5%, with the corresponding recurrence rates of 16.8% and 48.8%, respectively (both P < .001). Multivariate Cox regression analysis demonstrated that postoperative SIC was an independent risk factor for both increased recurrence (HR: 2.40, 95% CI, 2.04-2.81, P < .001) and decreased OS (HR: 2.69, 95% CI, 2.24-3.24, P < .001). CONCLUSION The results emphasize that postoperative incomplete conversion of either AFP or PIVKA-II is a significant prognostic marker, indicating a higher risk for adverse oncologic outcomes following HCC resection. This revelation has crucial implications for refining postoperative adjuvant therapy and surveillance strategies for HCC patients.
Collapse
Affiliation(s)
- Mingda Wang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Guojun Qian
- Department of Ultrasonic Intervention, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Hongmei Xiao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Xingkai Liu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Liyang Sun
- Department of Hepatobiliary and Pancreatic Surgery, Zhejiang Provincial People’s Hospital, Affiliated People’s Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, People’s Republic of China
| | - Zhong Chen
- Department of Hepatobiliary Surgery, Affiliated Hospital of Nantong University, Nantong, People’s Republic of China
| | - Kongying Lin
- Department of Hepatobiliary Surgery, Mengchao Hepatobiliary Hospital, Fujian Medical University, Fuzhou, People’s Republic of China
| | - Lanqing Yao
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Chao Li
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Lihui Gu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Jiahao Xu
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Xiaodong Sun
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Wei Qiu
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Timothy M Pawlik
- Department of Surgery, Ohio State University, Wexner Medical Center, Columbus, OH, United States
| | - Wan Yee Lau
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
- Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, People’s Republic of China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, First Hospital of Jilin University, Changchun, Jilin, People’s Republic of China
| | - Feng Shen
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| | - Tian Yang
- Department of Hepatobiliary Surgery, Eastern Hepatobiliary Surgery Hospital, Second Military Medical University (Naval Medical University), Shanghai, People’s Republic of China
| |
Collapse
|
48
|
Tian J, Pan S, Wang Y, Yu Y, Wang S, Shen Y, Yang L, Liu X, Qiu Q, Luan J, Wang F, Meng F. Early Alpha-Fetoprotein Response Predicts Sustained Tumor Response Following Immune Checkpoint Inhibitors Combined with Targeted Therapy in Liver Cancer. Biomedicines 2024; 12:2769. [PMID: 39767676 PMCID: PMC11672884 DOI: 10.3390/biomedicines12122769] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 11/27/2024] [Accepted: 12/03/2024] [Indexed: 01/11/2025] Open
Abstract
Background: Although immune checkpoint inhibitors (ICI) have revolutionized liver cancer treatment, some patients experience early tumor progression after therapy, missing the window for other potential treatments, such as neoadjuvant therapy. Therefore, identifying the predictive factors for early progression is critical for timely therapeutic adjustment and the optimization of patient outcomes. Methods: This retrospective study enrolled patients with liver cancer who received their first ICI combined with targeted therapy at the Fifth Medical Center of the PLA General Hospital between June 2022 and December 2023. Early tumor progression was defined as tumor progression within 6 months of therapy initiation. Multivariate logistic regression analysis was used to identify independent risk factors for early tumor progression, and overall survival (OS) curves were generated using the Kaplan-Meier method. Results: A total of 159 patients were enrolled. Multivariate logistic regression analysis indicated that patients with an early alpha-fetoprotein (AFP) response had a significantly reduced risk of early tumor progression (OR = 0.34, 95% CI: 0.13-0.84, p = 0.019), suggesting that an early AFP response is a protective factor against early progression. The area under curve (AUC) for the predictive model was 0.73 (95% CI: 0.63-0.83, p < 0.001). Stratified survival analysis showed that the median overall survival (mOS) in the early AFP response group was significantly longer than that in the poor response group (17.3 months vs. 6.1 months, HR = 2.11, 95% CI: 1.19-2.74, p = 0.009). Conclusions: Early AFP response is not only an effective biomarker for identifying high-risk patients prone to early tumor progression but is also significantly associated with long-term survival in liver cancer patients treated with ICI combined with targeted therapy. This finding will enable clinicians to make timely therapeutic adjustments and optimize treatment outcomes, thereby improving both progression-free survival and overall survival.
Collapse
Affiliation(s)
- Jiahe Tian
- Peking University 302 Clinical Medical School, Beijing 100191, China;
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Shida Pan
- Beijing Ditan Hospital, Capital Medical University, Beijing 100015, China;
| | - Yilin Wang
- Medical School of Chinese PLA, Beijing 100853, China;
| | - Yingying Yu
- The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China;
| | - Siyu Wang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Yingjuan Shen
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Luo Yang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Xiaomeng Liu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Qin Qiu
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Junqing Luan
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Fusheng Wang
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| | - Fanping Meng
- Peking University 302 Clinical Medical School, Beijing 100191, China;
- Department of Infectious Diseases, The Fifth Medical Centre of Chinese PLA General Hospital, National Clinical Research Centre for Infectious Diseases, Beijing 100039, China; (S.W.); (Y.S.); (L.Y.); (X.L.); (Q.Q.); (J.L.)
| |
Collapse
|
49
|
Zhou J, Zhang Y, Zhang J, Chen J, Jiang H, Zhang L, Zhong X, Zhang T, Chen L, Wang Y, Xu Y, Wang J. New strategy of LI-RADS v2018 to improve the sensitivity for small hepatocellular carcinoma ≤ 3.0 cm on extracellular-contrast enhanced MRI. Eur J Radiol 2024; 181:111830. [PMID: 39547000 DOI: 10.1016/j.ejrad.2024.111830] [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: 08/22/2024] [Revised: 10/15/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024]
Abstract
INTRODUCTION We aimed to modify LI-RADS version 2018 to improve sensitivity and determine the value of the combination of high alpha-fetoprotein (AFP) levels for small HCC (sHCC, ≤ 30 mm) diagnosis. METHODS A total of 984 patients at high risk for HCC, with 1204 observations (including 997 small observations ≤ 30 mm), who underwent extracellular contrast-enhanced MRI were enrolled from five independent centers. Blinded readers evaluated the LI-RADS features and categorized each observation according to the LI-RADS v2018, modified LI-RADS and EASL. Odds ratios of LI-RADS major features (MFs) and several high AFP levels for sHCC diagnosis were analyzed using multivariable logistic regression. The modified LR-5 criteria was developed by including no APHE at any size with two MFs, and non-rim APHE with one MF (≥ 10 mm) or with two MFs (< 10 mm). The diagnostic performance of each version of the LR-5 was compared using generalized estimating equations. RESULTS APHE, washout, enhancing capsule and five high AFP levels were independently associated with sHCC. In three datasets, the modified LI-RADS had higher sensitivities for sHCC (76.8 ∼ 85.5 % vs. 73.7 ∼ 75.9 %, P < 0.05) to the LR-5 v2018. The modified LI-RADS with AFP ≥ 200 ng/mL as an additional feature or as an alternative to threshold growth provided higher sensitivities for sHCC than LI-RADS v2018 (82.1 ∼ 90.1 % vs. 73.7 ∼ 75.9 %, all P < 0.05), modified LI-RADS (82.1 ∼ 90.1 % vs. 76.8 ∼ 85.5 %, all P < 0.05) and EASL version 2018 (82.1 ∼ 90.1 % vs. 73.3 ∼ 74.7 %, all P < 0.05), with comparable specificities (all P > 0.05). CONCLUSION The new strategy of LI-RADS v2018 provides significantly higher sensitivity and comparable specificity than those of LI-RADS v2018 for sHCC diagnosis on ECA-MRI.
Collapse
Affiliation(s)
- Jinhui Zhou
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, No 600, Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Yao Zhang
- Department of Radiology, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Lujiang Road 17, Hefei 230001, China
| | - Jing Zhang
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jingbiao Chen
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, No 600, Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Hang Jiang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, No 600, Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Linqi Zhang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, No 600, Tianhe Road, Guangzhou, Guangdong 510630, China
| | - Xi Zhong
- Department of Radiology, Affiliated Cancer Hospital & Institute of Guangzhou Medical University, 78 Hengzhigang Rd, Guangzhou, Guangdong 510095, China
| | - Tianhui Zhang
- Department of Radiology, Meizhou People's Hospital, Meizhou, Guangdong 514031, China
| | - Lichun Chen
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University Yuedong Hospital, Meizhou, Guangdong 514733, China
| | - Yufeng Wang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University Yuedong Hospital, Meizhou, Guangdong 514733, China
| | - Yikai Xu
- Department of Medical Imaging Center, Nanfang Hospital, Southern Medical University, Guangzhou 510515, China
| | - Jin Wang
- Department of Radiology, The Third Affiliated Hospital of Sun Yat-sen University, No 600, Tianhe Road, Guangzhou, Guangdong 510630, China; Organ Transplantation Institute, Sun Yat-sen University; Organ Transplantation Research Center of Guangdong Province, Guangdong Province Engineering Laboratory for Transplantation Medicine, Guangzhou 510630, China; Guangdong Key Laboratory of Liver Disease Research, the Third Affiliated Hospital of Sun Yat-sen University, Guangzhou 510630, China.
| |
Collapse
|
50
|
Ueno M, Takeda H, Takai A, Morimura H, Nishijima N, Iwamoto S, Okuyama S, Umeda M, Seta T, Ikeda A, Goto T, Miyamoto S, Kayahara T, Uenoyama Y, Matsumura K, Nakano S, Mishima M, Inuzuka T, Eso Y, Takahashi K, Marusawa H, Osaki Y, Hatano E, Seno H. CRAFITY score as a predictive marker for refractoriness to atezolizumab plus bevacizumab therapy in hepatocellular carcinoma: a multicenter retrospective study. J Gastroenterol 2024; 59:1107-1118. [PMID: 39289234 PMCID: PMC11541291 DOI: 10.1007/s00535-024-02150-7] [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: 06/05/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
BACKGROUND Although atezolizumab plus bevacizumab (Atezo/Bev) therapy has been used as the preferred first-line treatment for advanced hepatocellular carcinoma (HCC), up to 26% of patients do not achieve disease control, suggesting alternative treatments might be more beneficial for such patients. We investigated key predictors for refractoriness to Atezo/Bev therapy, particularly in the first-line setting. METHODS We retrospectively analyzed 302 patients with HCC who received Atezo/Bev therapy between October 2020 and September 2022 across nine hospitals in Japan. Refractoriness was defined as best overall response (BOR) of progressive disease or stable disease and a progression-free survival (PFS) of < 180 days (RECIST v1.1). Clinical benefit was defined as BOR of partial/complete response or stable disease with PFS of ≥ 180 days. Baseline characteristics and potential predictors, identified through literature review, were compared between these groups. Stratifications of overall survival (OS), and PFS were also assessed. RESULTS Refractoriness was observed in 126 (41.7%) patients, while 154 (51.0%) achieved clinical benefit. Due to a significant association between the treatment line and refractory rate, the subsequent analysis focused on the first-line cohort (n = 214; 72 [33.6%] patients showed refractoriness). Among 13 potential predictors, the CRP and AFP in immunotherapy (CRAFITY) score had the best predictive performance, with refractory rates of 24.6%, 44.6%, and 57.9% in CRAFITY-0, 1, and 2 patients, respectively (p < 0.001). OS and PFS were also well-stratified by this scoring system. CONCLUSIONS Approximately one-third of patients were refractory to first-line Atezo/Bev therapy. The CRAFITY score demonstrated superior performance in predicting refractoriness.
Collapse
Affiliation(s)
- Masayuki Ueno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Haruhiko Takeda
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Atsushi Takai
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan.
| | - Hiroki Morimura
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Norihiro Nishijima
- Department of Gastroenterology and Hepatology, Meiwa Hospital, Nishinomiya, Japan
| | - Satoru Iwamoto
- Department of Gastroenterology, Kyoto Medical Center, Kyoto, Japan
| | - Shunsuke Okuyama
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Makoto Umeda
- Department of Gastroenterology and Hepatology, Hyogo Prefectural Amagasaki General Medical Center, Amagasaki, Japan
| | - Takeshi Seta
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
- Department of Health Informatics, Graduate School of Medicine and School of Public Health, Kyoto University, Kyoto, Japan
| | - Atsuyuki Ikeda
- Department of Gastroenterology and Hepatology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Tomoyuki Goto
- Department of Medical Oncology, Shiga General Hospital, Moriyama, Japan
| | | | - Takahisa Kayahara
- Department of Gastroenterology and Hepatology, Kurashiki Central Hospital, Kurashiki, Japan
| | - Yoshito Uenoyama
- Department of Gastroenterology and Hepatology, Japanese Red Cross Wakayama Medical Center, Wakayama, Japan
| | - Kazuyoshi Matsumura
- Department of Gastroenterology and Hepatology, Shiga General Hospital, Moriyama, Japan
| | - Shigeharu Nakano
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Masako Mishima
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Tadashi Inuzuka
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Yuji Eso
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| | - Ken Takahashi
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
- Division of Cancer Immunotherapy, Center for Cancer Immunotherapy and Immunobiology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroyuki Marusawa
- Department of Gastroenterology and Hepatology, Osaka Red Cross Hospital, Osaka, Japan
| | - Yukio Osaki
- Department of Gastroenterology and Hepatology, Meiwa Hospital, Nishinomiya, Japan
| | - Etsuro Hatano
- Division of Hepato-Biliary-Pancreatic Surgery and Transplantation, Department of Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroshi Seno
- Department of Gastroenterology and Hepatology, Graduate School of Medicine, Kyoto University, 54 Kawahara-cho, Shogoin, Sakyo-ku, Kyoto, 606-8507, Japan
| |
Collapse
|