Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

Article Types

Countries / Regions

Search Results (363)

Search Parameters:
Keywords = HCC metastasis

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
17 pages, 1827 KB  
Review
The Role of Cadherin 17 (CDH17) in Cancer Progression via Wnt/β-Catenin Signalling Pathway: A Systematic Review and Meta-Analysis
by Bipusha Tha Shrestha, Yahui Feng, Aaron Lad, Anthony Bates, Jing Chen, Karen Brown, Feier Zeng and Ning Wang
Int. J. Mol. Sci. 2025, 26(20), 9838; https://doi.org/10.3390/ijms26209838 - 10 Oct 2025
Viewed by 496
Abstract
Cadherin 17 (CDH17) is a cell adhesion glycoprotein essential for epithelial integrity. It is frequently overexpressed in various cancers, where it is associated with aggressive behaviour. While evidence indicates that CDH17 functions as an upstream regulator of Wnt/β-catenin signalling, findings are inconsistent across [...] Read more.
Cadherin 17 (CDH17) is a cell adhesion glycoprotein essential for epithelial integrity. It is frequently overexpressed in various cancers, where it is associated with aggressive behaviour. While evidence indicates that CDH17 functions as an upstream regulator of Wnt/β-catenin signalling, findings are inconsistent across tumour types, limiting the assessment of CDH17 as a biomarker or therapeutic target for Wnt pathway in cancer. In this study, we systematically review and meta-analyse the relationship between CDH17 and Wnt/β-catenin signalling in human cancers and evaluate whether CDH17 modulation affects tumour behaviour through Wnt-related mechanisms. Our search of Medline, Web of Science and Scopus identified five studies examining CDH17 expression in the Wnt/β-catenin pathway in vitro and in vivo. All five studies identified CDH17 as a key driver of canonical Wnt signalling, directly influencing cancer progression in hepatocellular carcinoma (HCC), gastric cancer (GC), and colorectal cancer (CRC). Meta-analysis (MA) showed that CDH17 inhibition consistently reduced Wnt/β-catenin downstream T-cell factor/lymphoid enhancer-binding factor (TCF/LEF) transcriptional activity (MD = −1.32, 95% CI: −1.64 to −0.99, p < 0.00001). Narrative synthesis found that CDH17 suppression decreased total and nuclear β-catenin, phosphorylated glycogen synthase kinase-3 beta (GSK-3β), and cyclin D1 while increasing tumour suppressors, retinoblastoma (Rb) and p53/p21. These changes were associated with reduced proliferation, colony formation, migration, invasion and cell cycle arrest. In vivo, CDH17 suppression resulted in 80–95% tumour growth suppression (Mean Difference (MD) = −96.67, 95% CI: [−144.35, −48.98], p < 0.0001), with immunohistochemistry confirming cytoplasmic β-catenin sequestration and lower cyclin D1 levels. Collectively, these findings show CDH17 as a critical upstream effector sustaining Wnt/β-catenin signalling, cancer progression, tumour proliferation, stem cell properties, and metastasis, and support CDH17 inhibition as a promising therapeutic target across multiple cancer types. Full article
Show Figures

Figure 1

23 pages, 2667 KB  
Article
Reactivation of the PI3K/mTOR Signaling Pathway Confers Resistance to the FGFR4 Inhibitor FGF401
by Hung Huynh and Wai Har Ng
Int. J. Mol. Sci. 2025, 26(19), 9818; https://doi.org/10.3390/ijms26199818 (registering DOI) - 9 Oct 2025
Viewed by 159
Abstract
Hepatocellular carcinoma (HCC) is a deadly liver cancer characterized by dysregulated signaling and aberrant cell-cycle control. The FGFR4/FGF19 pathway is dysregulated in HCC and other cancers. Inhibitors targeting the FGF19/FGFR4 pathway, including the FGF19/FGFR4 inhibitor FGF401, have been investigated in HCC and other [...] Read more.
Hepatocellular carcinoma (HCC) is a deadly liver cancer characterized by dysregulated signaling and aberrant cell-cycle control. The FGFR4/FGF19 pathway is dysregulated in HCC and other cancers. Inhibitors targeting the FGF19/FGFR4 pathway, including the FGF19/FGFR4 inhibitor FGF401, have been investigated in HCC and other cancers; however, nearly all patients who initially respond eventually develop resistance shortly after starting therapy, highlighting the urgent need for new treatment strategies to overcome drug resistance. In the present study, we report that chronic treatment of the FGF19/FGFR4-expressing HCC25−0705A line with FGF401 led to acquired resistance. FGF401-resistant tumors exhibited upregulation of FGFRs and activation of the PI3K/AKT/mTOR/p70S6K pathway. Combination therapy with FGF401 and the mammalian target of rapamycin (mTOR) inhibitor everolimus (FGF401/everolimus) resulted in more complete tumor growth inhibition, delayed the onset of resistance, and prolonged overall survival (OS) in mice bearing orthotopic HCC tumors. The FGF401/everolimus combination effectively suppressed tumor cell proliferation; promoted apoptosis; reduced tumor hypoxia via blood vessel normalization; and downregulated key proteins involved in proliferation, survival, metastasis, and angiogenesis. These preclinical findings provide a strong rationale for clinical trials combining FGFR4 and mTOR inhibitors in HCC patients with FGF19/FGFR4/mTOR-dependent tumors. Full article
Show Figures

Figure 1

20 pages, 1591 KB  
Case Report
Highlighting the Importance of Signaling Pathways and Immunohistochemistry Features in HCC: A Case Report and Literature Review
by Madalin Alexandru Hasan, Ioana Larisa Paul, Simona Cavalu, Ovidiu Laurean Pop, Lorena Paduraru, Ioan Magyar and Mihaela Doina Chirila
Reports 2025, 8(4), 197; https://doi.org/10.3390/reports8040197 - 3 Oct 2025
Viewed by 186
Abstract
Background and Clinical Significance: In hepatocellular carcinoma (HCC), numerous signaling pathways become aberrantly regulated, resulting in sustained cellular proliferation and enhanced metastatic potential. Tumors that lack PYGO2 may not show the same types of tissue remodeling or regenerative features driven by the Wnt/β-catenin [...] Read more.
Background and Clinical Significance: In hepatocellular carcinoma (HCC), numerous signaling pathways become aberrantly regulated, resulting in sustained cellular proliferation and enhanced metastatic potential. Tumors that lack PYGO2 may not show the same types of tissue remodeling or regenerative features driven by the Wnt/β-catenin pathway, which could make the tumor behave differently from others that are Wnt-positive. PIK3CA-positive tumors are often associated with worse prognosis due to the aggressive nature of the PI3K/AKT pathway activation. This is linked to higher chances of metastasis, recurrence, and resistance to therapies that do not target this pathway. Case presentation: In this paper we present a rare case of hepatocellular carcinoma with PIK3CA-positive and PYGO2-negative signaling pathways, several key aspects of the tumor’s behavior, prognosis, and treatment options. Although alpha-fetoprotein (AFP) levels were significantly elevated, the CT and MRI examination showed characteristics of malignancy, HCC with secondary hepatic lesions and associated perfusion disturbances. The case particularities and immunohistochemistry features are highlighted in the context of literature review, the PIK3CA mutation suggesting the activation of the PI3K/AKT/mTOR pathway, a critical signaling pathway involved in cell survival, proliferation, and metabolism. Conclusions: Due to the aggressive nature of PIK3CA mutations, close monitoring and consideration of immunotherapy and targeted treatments are of crucial importance. Full article
(This article belongs to the Section Oncology)
20 pages, 3824 KB  
Article
Spatial Transcriptomics Reveals Distinct Architectures but Shared Vulnerabilities in Primary and Metastatic Liver Tumors
by Swamy R. Adapa, Sahanama Porshe, Divya Priyanka Talada, Timothy M. Nywening, Mattew L. Anderson, Timothy I. Shaw and Rays H. Y. Jiang
Cancers 2025, 17(19), 3210; https://doi.org/10.3390/cancers17193210 - 1 Oct 2025
Viewed by 602
Abstract
Background: Primary hepatocellular carcinoma (HCC) and liver metastases differ in origin, progression, and therapeutic response, yet a direct high-resolution spatial comparison of their tumor microenvironments (TMEs) within the liver has not previously been performed. Methods: We applied high-definition spatial transcriptomics to [...] Read more.
Background: Primary hepatocellular carcinoma (HCC) and liver metastases differ in origin, progression, and therapeutic response, yet a direct high-resolution spatial comparison of their tumor microenvironments (TMEs) within the liver has not previously been performed. Methods: We applied high-definition spatial transcriptomics to fresh-frozen specimens of one HCC and one liver metastasis (>16,000 genes per sample, >97% mapping rates) as a proof-of-principle two-specimen study, cross-validated in human proteomics and patients’ survival datasets. Transcriptional clustering revealed spatially distinct compartments, rare cell states, and pathway alterations, which were further compared against an independent systemic dataset. Results: HCC displayed an ordered lineage architecture, with transformed hepatocyte-like tumor cells broadly dispersed across the tissue and more differentiated hepatocyte-derived cells restricted to localized zones. By contrast, liver metastases showed two sharply compartmentalized domains: an invasion zone, where proliferative stem-like tumor cells occupied TAM-rich boundaries adjacent to hypoxia-adapted tumor-core cells, and a plasticity zone, which formed a heterogeneous niche of cancer–testis antigen–positive germline-like cells. Across both tumor types, we detected a conserved metabolic program of “porphyrin overdrive,” defined by reduced cytochrome P450 expression, enhanced oxidative phosphorylation gene expression, and upregulation of FLVCR1 and ALOX5, reflecting coordinated rewiring of heme and lipid metabolism. Conclusions: In this pilot study, HCC and liver metastases demonstrated fundamentally different spatial architectures, with metastases uniquely harboring a germline/neural-like plasticity hub. Despite these organizational contrasts, both tumor types converged on a shared program of metabolic rewiring, highlighting potential therapeutic targets that link local tumor niches to systemic host–tumor interactions. Full article
Show Figures

Figure 1

24 pages, 5386 KB  
Article
Kuwanon A Targeted YWHAB in Hepatocellular Carcinoma Cells to Inhibit the Raf/MEK/ERK Signaling Pathway
by Jingyang Xu, Hongbo Chang, Yongzhao Wang, Yi Du, Liping Zhong and Hongjuan Cui
Cells 2025, 14(19), 1487; https://doi.org/10.3390/cells14191487 - 23 Sep 2025
Viewed by 315
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, and the lung is one of the most frequent metastatic sites for HCC. In this study, we aimed to identify a mild active substance in Morus alba L. that can inhibit [...] Read more.
Hepatocellular carcinoma (HCC) is one of the most common malignant tumors worldwide, and the lung is one of the most frequent metastatic sites for HCC. In this study, we aimed to identify a mild active substance in Morus alba L. that can inhibit the pulmonary metastasis of HCC and reduce the drug resistance of clinical therapies. Further deepen the understanding of the anti-cancer functions of the mulberry active substances. In this study, we have screened and identified a flavonoid compound extracted from the root bark of the Morus alba L. named Kuwanon A (KA). Our research demonstrated that KA directly targeted the YWHAB (tyrosine 3-monooxygenase/tryptophan 5-monooxygenase activation protein beta) and mediated its dimer dissociation. Thereby inhibiting the MAPK pathway and affecting downstream biological functions, including cell cycle arrest and migration/invasion inhibition. The experiment results proved that KA could inhibit the proliferation and metastasis of highly metastatic HCC cells both in vitro and in vivo. Additionally, when KA was combined with the clinical drug sorafenib, it exhibited a synergistic effect in inhibiting HCC cell proliferation, migration, and invasion. In conclusion, KA demonstrated a favorable anti-tumor effect in HCC cells. Full article
Show Figures

Graphical abstract

19 pages, 1781 KB  
Article
Physiopathological Features in a Three-Dimensional In Vitro Model of Hepatocellular Carcinoma: Hypoxia-Driven Oxidative Stress and ECM Remodeling
by Maria Giovanna Rizzo, Enza Fazio, Claudia De Pasquale, Emanuele Luigi Sciuto, Giorgia Cannatà, Cristiana Roberta Multisanti, Federica Impellitteri, Federica Gilda D’Agostino, Salvatore Pietro Paolo Guglielmino, Caterina Faggio and Sabrina Conoci
Cancers 2025, 17(18), 3082; https://doi.org/10.3390/cancers17183082 - 21 Sep 2025
Viewed by 495
Abstract
Background: Hypoxia is a hallmark of solid tumors, including hepatocellular carcinoma (HCC), where it drives oxidative stress and extracellular matrix (ECM) remodeling, promoting tumor invasion and metastasis. Investigating these mechanisms in patients remains challenging due to the complexity of the tumor microenvironment. [...] Read more.
Background: Hypoxia is a hallmark of solid tumors, including hepatocellular carcinoma (HCC), where it drives oxidative stress and extracellular matrix (ECM) remodeling, promoting tumor invasion and metastasis. Investigating these mechanisms in patients remains challenging due to the complexity of the tumor microenvironment. Methods: We developed a scaffold-free three-dimensional (3D) spheroid model of HCC using human hepatocellular carcinoma HepG2 cells (ATCC HB-8065). To characterize hypoxia-driven processes, a multiparametric approach combining MTT assays for metabolic activity, confocal microscopy for viability and ECM organization, flow cytometry for apoptosis and ROS detection, qRT-PCR for gene expression, and FTIR spectroscopy for biochemical profiling were performed. Results: The 3D model exhibited progressive ROS accumulation, stabilization of HIF-1α, and metabolic reprogramming toward aerobic glycolysis. In parallel, ECM remodeling was evident, with increased expression of SPARC and FN1 and collagen fiber alignment, reflecting an invasive tumor phenotype. Conclusions: This scaffold-free 3D HCC model recapitulates key physiopathological features of tumor progression, providing a robust and physiologically relevant platform to investigate the hypoxia–ROS–ECM relationship and to support preclinical evaluation of targeted therapeutic strategies. Full article
(This article belongs to the Special Issue Extracellular Matrix Proteins in Cancer)
Show Figures

Figure 1

18 pages, 2522 KB  
Article
Therapeutic Effect of Selenium Nanoparticles, Sorafenib, and Selenium–Sorafenib Nanocomplex in the Lungs and Kidneys of Mice with TAA-Induced HCC
by Egor A. Turovsky, Sergey V. Gudkov and Elena G. Varlamova
Biomolecules 2025, 15(9), 1336; https://doi.org/10.3390/biom15091336 - 18 Sep 2025
Viewed by 514
Abstract
Hepatocellular carcinoma is a primary malignant tumor of the liver, which is a serious health problem due to its aggressive nature, late diagnosis, and metastasis to other organs. We present, for the first time, the mRNA expression patterns of a wide range of [...] Read more.
Hepatocellular carcinoma is a primary malignant tumor of the liver, which is a serious health problem due to its aggressive nature, late diagnosis, and metastasis to other organs. We present, for the first time, the mRNA expression patterns of a wide range of genes involved in inflammation, fibrosis, endoplasmic reticulum stress, various forms of cell death, and signaling cascades in the lungs and kidneys of mice with thioacetamide-induced HCC. It is known that HCC often metastasizes to the lungs, and it is also important to understand which pathological processes occur in the kidneys, since the liver and kidneys are key target organs of toxicity. The main goal of this work was to study the pathological processes in the lungs and kidneys in HCC and the effectiveness of selenium nanocomplexes, as well as the well-known drug sorafenib, in mitigating these pathological consequences. These results present a significant contribution to the study of HCC metastasis to the lungs and kidneys and to the development of drugs that are most effective in the late stages of HCC. In addition, a hierarchy of the distribution of the selenium in the liver, kidneys, and lungs was established after the treatment of mice with HCC with selenium nanoparticles and a selenium–sorafenib nanocomplex. These data are important for developing a treatment protocol and determining optimal dosages of the drugs under study, which allows for achieving the desired therapeutic effect and neutralizing the toxic effect of selenium on healthy tissues and organs. Full article
(This article belongs to the Special Issue Advances in Nano-Based Drug Delivery Systems)
Show Figures

Figure 1

15 pages, 1193 KB  
Article
Somatostatin Receptor 2 Overexpression in Hepatocellular Carcinoma: Implications for Cancer Biology and Therapeutic Applications
by Servando Hernandez Vargas, Solmaz Aghaamiri, Jack T. Adams, Tyler M. Bateman, Belkacem Acidi, Sukhen C. Ghosh, Vahid Khalaj, Ahmed O. Kaseb, Hop S. Tran Cao, Majid Momeny and Ali Azhdarinia
Curr. Oncol. 2025, 32(9), 512; https://doi.org/10.3390/curroncol32090512 - 13 Sep 2025
Viewed by 780
Abstract
(1) Background: Somatostatin receptor 2 (SSTR2), a G protein-coupled receptor, is overexpressed in multiple malignancies, including hepatocellular carcinoma (HCC). While SSTR2 has traditionally been viewed as an inhibitory receptor involved in suppressing hormone secretion and cell proliferation, emerging evidence suggests a more complex [...] Read more.
(1) Background: Somatostatin receptor 2 (SSTR2), a G protein-coupled receptor, is overexpressed in multiple malignancies, including hepatocellular carcinoma (HCC). While SSTR2 has traditionally been viewed as an inhibitory receptor involved in suppressing hormone secretion and cell proliferation, emerging evidence suggests a more complex role in cancer biology. However, the functional implications of SSTR2 expression in HCC remain poorly understood. This study aimed to systematically investigate the molecular landscape associated with SSTR2 expression in HCC and evaluate its potential as a therapeutic target. (2) Methods: SSTR2 expression patterns across 22 tumor types were assessed using TNMplot, and its expression in HCC was further validated through The Human Protein Atlas. Integrative analysis of transcriptomic profiles, protein expression data, and somatic copy number alterations was performed using data from The Cancer Genome Atlas (TCGA) to stratify HCC patients by SSTR2 expression levels. Gene Ontology (GO) enrichment analysis was conducted via SRplot to uncover biological processes and signaling pathways associated with SSTR2. Kaplan–Meier survival analyses were performed using GEO datasets to determine the prognostic significance of SSTR2 expression. (3) Results: SSTR2 is moderately expressed in the majority of HCC tumors. Elevated SSTR2 expression correlates with significantly poorer overall and disease-specific survival. High SSTR2 levels are associated with activation of oncogenic signaling cascades related to cell proliferation, epithelial-to-mesenchymal transition (EMT), angiogenesis, and metastasis. Additionally, SSTR2 expression is positively correlated with several receptor tyrosine kinases and oncogenes implicated in HCC progression. (4) Conclusions: Our findings suggest that SSTR2 is not merely a passive biomarker but may contribute to HCC pathogenesis through modulation of oncogenic pathways. These data support the rationale for further development of SSTR2-directed therapeutic strategies to inhibit tumor growth and invasion in HCC patients. Full article
Show Figures

Graphical abstract

17 pages, 2154 KB  
Article
Impact of Serum Albumin Levels on Prognosis and Recurrence in Patients with Hepatocellular Carcinoma
by Naoko Hayata, Atsushi Hosui, Tomohide Kurahashi, Shigeki Suemura, Akane Namiki, Akino Okamoto, Takafumi Tanimoto, Hiroki Murai, Kohsaku Ohnishi, Motohiro Hirao, Takuya Yamada and Naoki Hiramatsu
Cancers 2025, 17(18), 2971; https://doi.org/10.3390/cancers17182971 - 11 Sep 2025
Viewed by 675
Abstract
Background: Liver function is a critical factor, both in the selection of treatment and in the prediction of prognosis in patients with hepatocellular carcinoma (HCC). The ALBI grade, introduced as a more objective method of assessing liver function, utilizes serum albumin (Alb) [...] Read more.
Background: Liver function is a critical factor, both in the selection of treatment and in the prediction of prognosis in patients with hepatocellular carcinoma (HCC). The ALBI grade, introduced as a more objective method of assessing liver function, utilizes serum albumin (Alb) and total bilirubin (Bil) levels. Although albumin is widely recognized for its role in maintaining colloid osmotic pressure and regulating plasma volume, recent studies have implicated it in tumor progression, invasion, and metastasis. The purpose of this study is to determine the impact of serum albumin levels on overall survival (OS) and tumor invasion/metastasis in HCC patients with the same liver function (ALBI grade) at the time of diagnosis. Methods: In this study, 285 patients diagnosed with primary HCC at our institution from 2015 to 2019 were classified by ALBI grade and analyzed. Among them, 78 patients with ALBI grade 2 status were selected to evaluate the impact of albumin level. To further isolate the effect of albumin rather than bilirubin, patients in the ALBI grade 2 cohort were divided into two groups based on mean values of Alb (3.5 g/dL) and Bil (1.0 mg/dL). Alb normal group (Group A): Alb ≥ 3.5 g/dL, Bil ≥ 1.0 mg/dL (n = 42). Bil normal group (Group T): Alb < 3.5 g/dL, Bil < 1.0 mg/dL (n = 36). Liver function was almost the same in these two groups based on the ALBI grade. OS, progression-free survival (PFS), types of recurrence, and pathological findings were compared between the two groups. OS was analyzed by the log-rank test, and comparisons between the two groups were performed by the t-test and chi-square test, with p < 0.05 indicating statistical significance. Results: OS was significantly worse in Group T than in Group A before and after propensity score matching based on age, performance status, and HCC stage (p < 0.001 and p = 0.011). Among the 44 patients who received curative treatment (surgery or radiofrequency ablation), OS was also significantly worse in Group T (p < 0.001). An analysis of the recurrence patterns of 44 curatively treated patients revealed that Group T had significantly shorter PFS (p < 0.001), and all recurrence patterns were multiple (p = 0.002). Pathological analysis in 28 surgical patients showed that serosal invasion was present in significantly more patients in Group T (p = 0.003). Conclusions: Low serum albumin levels in patients with HCC indicate both liver dysfunction and increased tumor invasion and metastasis. Nutritional support and albumin supplementation may help reduce intrahepatic metastases and improve prognosis. Further studies are needed to explore the underlying mechanisms and therapeutic potential. Full article
Show Figures

Graphical abstract

14 pages, 586 KB  
Article
Surgical Outcomes of Lenvatinib Treatment Followed by Liver Resection for Advanced Hepatocellular Carcinoma Larger than 10 cm
by Hideki Yokoo, Shoichiro Mizukami, Hiroyuki Takahashi, Tomoki Takizawa, Katsuro Enomoto, Kai Makino, Hiroki Takahata, Yuki Adachi and Koji Imai
Cancers 2025, 17(17), 2818; https://doi.org/10.3390/cancers17172818 - 28 Aug 2025
Viewed by 709
Abstract
Background: Hepatocellular carcinoma (HCC) larger than 10 cm has a poor prognosis, with high recurrence rates, particularly distant metastases. This study examined whether lenvatinib treatment followed by liver resection improves the outcomes for large HCCs compared with upfront surgery. Methods: We retrospectively analyzed [...] Read more.
Background: Hepatocellular carcinoma (HCC) larger than 10 cm has a poor prognosis, with high recurrence rates, particularly distant metastases. This study examined whether lenvatinib treatment followed by liver resection improves the outcomes for large HCCs compared with upfront surgery. Methods: We retrospectively analyzed 30 patients with HCC larger than 10 cm who underwent hepatectomy at our institution between January 2008 and December 2023. The study cohort included 30 patients: 9 received preoperative lenvatinib treatment followed by hepatectomy (LEN group), while 21 patients underwent upfront surgery (UFS group). We compared the clinicopathological characteristics, surgical outcomes, recurrence patterns, and survival between the two groups. Results: The median duration of lenvatinib administration was 1.8 months, with partial response in two patients (22.2%) and stable disease in seven patients (77.7%). While lenvatinib treatment significantly decreased serum albumin levels (p < 0.05) and increased ALBI scores (p = 0.03), the surgical outcomes including blood loss, operation time, and complication rates were comparable between the two groups. The 3-year recurrence-free survival rate was significantly higher in the LEN group compared with the UFS group (66.7% vs. 16.1%, p = 0.027). The 3-year overall survival rate was also higher in the LEN group, though not statistically significant (85.7% vs. 56.1%, p = 0.059). Notably, distant metastasis rates were lower in the LEN group compared with the UFS group (11.1% vs. 47.6%, p = 0.10). Conclusions: Preoperative lenvatinib treatment followed by hepatectomy for large HCC (> 10 cm) may reduce recurrence, particularly distant metastases, and potentially improve long-term survival. This approach may be a promising strategy for large HCCs, which traditionally have a poor prognosis with upfront surgery alone. Full article
Show Figures

Graphical abstract

23 pages, 4819 KB  
Review
Dysregulation of MicroRNAs in Hepatocellular Carcinoma: Targeting Oncogenic Signaling Pathways for Innovative Therapies
by Yusra Zarlashat, Judit Halász and Edit Dósa
Int. J. Mol. Sci. 2025, 26(17), 8365; https://doi.org/10.3390/ijms26178365 - 28 Aug 2025
Viewed by 701
Abstract
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and the third leading cause of cancer-related death. Hyperactivation of oncogenes and suppression of tumor suppressor genes/proteins drive HCC initiation and progression. MicroRNAs (miRNAs) critically modulate HCC biology by regulating proliferation, apoptosis, [...] Read more.
Hepatocellular carcinoma (HCC) is one of the most common malignancies worldwide and the third leading cause of cancer-related death. Hyperactivation of oncogenes and suppression of tumor suppressor genes/proteins drive HCC initiation and progression. MicroRNAs (miRNAs) critically modulate HCC biology by regulating proliferation, apoptosis, and metastasis. Acting either as tumor suppressors or oncomiRs, they shape core signaling pathways, including PI3K/Akt/mTOR, Hippo–YAP/TAZ, Wnt/β-catenin, RAS/MAPK, and p53. Their dysregulation in tissues and body fluids renders them promising diagnostic biomarkers and therapeutic targets. Preclinical studies demonstrate that miRNA-based strategies—either restoring tumor-suppressive miRNAs (e.g., miR-34a, miR-125a-5p) or inhibiting oncogenic miRNAs (e.g., miR-660-5p)—can suppress HCC progression and reduce treatment resistance. Combination approaches, such as pairing miR-122 mimics with miR-221 inhibitors or delivering miR-326 via nanoparticles, further enhance efficacy by simultaneously targeting multiple oncogenic pathways. This review summarizes recent advances in miRNA-mediated regulation of HCC signaling and highlights their clinical potential, including ongoing trials of miRNA-based diagnostics and therapeutics for early detection, prognostication, and personalized treatment. Full article
(This article belongs to the Special Issue Pathogenesis and Molecular Treatment of Primary Liver Cancer)
Show Figures

Graphical abstract

10 pages, 826 KB  
Article
Differential Associations of PIVKA-II with Epithelial and Mesenchymal Features in HCC and PDAC
by Antonella Farina, Gaia Cicolani, Valentina Viggiani, Matteo Maini, Antonio Angeloni and Emanuela Anastasi
Int. J. Mol. Sci. 2025, 26(15), 7581; https://doi.org/10.3390/ijms26157581 - 5 Aug 2025
Viewed by 658
Abstract
Hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC) are aggressive malignancies characterized by a poor prognosis and resistance to conventional therapies. Mounting evidence suggests the pivotal role of epithelial–mesenchymal transition (EMT) in tumor progression, metastasis, and therapeutic resistance in these cancers. Protein induced [...] Read more.
Hepatocellular carcinoma (HCC) and pancreatic ductal adenocarcinoma (PDAC) are aggressive malignancies characterized by a poor prognosis and resistance to conventional therapies. Mounting evidence suggests the pivotal role of epithelial–mesenchymal transition (EMT) in tumor progression, metastasis, and therapeutic resistance in these cancers. Protein induced by vitamin K absence II (PIVKA-II)—a valuable HCC detector—has ultimately emerged as a potentially relevant biomarker in PDAC, serving as both a serum biomarker and a prognostic indicator. This study investigates the putative link between PIVKA-II expression and the EMT process in HCC and PDAC. Using a Western blot analysis and electrochemiluminescence immunoassay (ECLIA), we quantified PIVKA-II serum levels alongside two canonical EMT markers—Vimentin and E-cadherin—in selected cohorts. Emerging data suggest a dual, context-dependent role for PIVKA-II. Beyond its diagnostic value in both malignancies, its co-expression with EMT markers points to a potential mechanistic involvement in tumor invasiveness and phenotypic plasticity. Notably, the selective detection of E-cadherin in HCC implies limited EMT activation and a preservation of the epithelial phenotype, whereas the higher expression of Vimentin in PDAC reflects a more substantial shift toward EMT. We provide a comprehensive analysis of key molecular markers, their involvement in EMT-driven pathophysiological mechanisms, and their potential as novel diagnostic tools. Full article
(This article belongs to the Section Macromolecules)
Show Figures

Figure 1

14 pages, 4954 KB  
Article
The Niemann–Pick C1 Protein of Patients with Hepatocellular Carcinoma Is Associated with Survival Time in Males and Tumor Size in Females
by Florian Weber, Katja Evert, Alexander Scheiter, Sophie von Sachsen-Coburg, Kirsten Utpatel and Christa Buechler
Biomedicines 2025, 13(7), 1707; https://doi.org/10.3390/biomedicines13071707 - 13 Jul 2025
Viewed by 610
Abstract
Background/Objectives: The Niemann–Pick C1 (NPC1) protein regulates cellular cholesterol homeostasis, which is disrupted in hepatocellular carcinoma (HCC). Sex differences in cholesterol metabolism may also be related to NPC1 expression in HCC. A sex-specific analysis was, therefore, performed to investigate this further. Methods: The [...] Read more.
Background/Objectives: The Niemann–Pick C1 (NPC1) protein regulates cellular cholesterol homeostasis, which is disrupted in hepatocellular carcinoma (HCC). Sex differences in cholesterol metabolism may also be related to NPC1 expression in HCC. A sex-specific analysis was, therefore, performed to investigate this further. Methods: The expression of NPC1 protein in hepatocytes was assessed using immunohistochemistry in HCC tissues from 264 male and 59 female patients, as well as in non-tumor tissues from 41 males and 7 females. Results: The disease etiology was documented for 40% of these patients, and NPC1 protein levels in the tumors of patients with alcoholic, metabolic, and viral liver disease were comparable. The severity of underlying liver fibrosis was similar in both females and males. No difference in hepatocyte NPC1 protein expression was observed between males and females in non-tumor and tumor tissues. However, NPC1 expression was strongly increased in tumor tissues in both sexes. NPC1 protein levels were positively associated with T stage and Union for International Cancer Control (UICC) stage in both sexes. NPC1 protein levels were negatively correlated with overall survival, recurrence-free survival, and metastasis-free survival time in males only. Univariate Cox regression analysis showed a significant association of NPC1 protein levels with metastasis-free survival in males. Positive correlations of NPC1 protein with tumor size and negative associations with tumor inflammation were observed only in women. Conclusions: This study showed that hepatocyte NPC1 protein levels are highly elevated in HCC tissue in both sexes but are more closely associated with survival in male patients than in female patients. Full article
Show Figures

Figure 1

20 pages, 7746 KB  
Article
PBX3-HMGCR Axis Promotes Hepatocellular Carcinoma Progression Through Enhancing De Novo Cholesterol Biosynthesis
by Xia Zhang, Li Qiu, Lei Zhang, Wenfang Li, Debing Xiang, Jian Wang, Shourong Wu and Vivi Kasim
Int. J. Mol. Sci. 2025, 26(11), 5210; https://doi.org/10.3390/ijms26115210 - 29 May 2025
Viewed by 842
Abstract
Tumor cells alter lipid metabolic pathways to meet their demands for energy and membrane biosynthesis. Despite its crucial role in tumor cell growth, survival, and metastasis, the mechanisms underlying tumor cell lipid metabolic reprogramming remain poorly understood. Pre-B-cell leukemia transcription factor 3 (PBX3), [...] Read more.
Tumor cells alter lipid metabolic pathways to meet their demands for energy and membrane biosynthesis. Despite its crucial role in tumor cell growth, survival, and metastasis, the mechanisms underlying tumor cell lipid metabolic reprogramming remain poorly understood. Pre-B-cell leukemia transcription factor 3 (PBX3), a member of the PBX family, could promote tumorigenesis; however, whether it is involved in tumor lipid metabolic reprogramming remains unknown. Herein, we found that PBX3 significantly promotes tumor growth by enhancing lipid accumulation in HCC cells. By assessing the effect of PBX3 on the expression levels of lipid metabolism-related genes, we found that PBX3 could positively regulate the expression of 3-hydroxy-3-methylglutaryl CoA reductase (HMGCR), a rate-limiting enzyme in the cholesterol biosynthesis pathway. Mechanistically, we revealed that PBX3 could directly bind to the −167/−151 region of HMGCR promoter, thereby increasing its transcriptional activity and, subsequently, its expression level. This leads to the increase of HCC cell cholesterol biosynthesis and, eventually, to the increase of the in vivo tumorigenic potential. Collectively, our research revealed an unprecedented regulatory mechanism of cholesterol metabolism in HCC cells through PBX3 positive regulation on HMGCR expression levels. These findings provide novel insights into tumor metabolic reprogramming and uncover a previously unknown physiological function for PBX3. Moreover, these results suggest the potential of targeting PBX3 as an anti-tumor therapeutic strategy. Full article
(This article belongs to the Special Issue Molecular Advances in Cancer and Cell Metabolism—2nd Edition)
Show Figures

Figure 1

15 pages, 10924 KB  
Article
FBXO10 Drives Hepatocellular Carcinoma Proliferation via K63-Linked Ubiquitination and Stabilization of FRMPD1
by Wuguang Liu, Bin Xu, Kashif Kifayat, Yuhong Xie, Xiaolong Liu, Chengyong Dong and Liming Wang
Curr. Issues Mol. Biol. 2025, 47(6), 391; https://doi.org/10.3390/cimb47060391 - 24 May 2025
Viewed by 783
Abstract
Aberrant ubiquitination drives hepatocellular carcinoma (HCC) progression, yet the role of FBXO10—a key F-box E3 ubiquitin ligase component—remains uncharacterized. Through bioinformatics analyses and functional validation, we establish FBXO10 as a critical oncogenic driver in HCC. Transcriptomic data from public databases (TIMER, UALCAN, GEO) [...] Read more.
Aberrant ubiquitination drives hepatocellular carcinoma (HCC) progression, yet the role of FBXO10—a key F-box E3 ubiquitin ligase component—remains uncharacterized. Through bioinformatics analyses and functional validation, we establish FBXO10 as a critical oncogenic driver in HCC. Transcriptomic data from public databases (TIMER, UALCAN, GEO) revealed significant FBXO10 upregulation in HCC tissues, with elevated expression predicting advanced tumor stage, metastasis, and reduced survival. Functionally, FBXO10 silencing suppressed HCC cell proliferation while its overexpression promoted tumor growth. Mechanistic studies revealed that FBXO10 directly interacts with FRMPD1 to mediate its K63-linked polyubiquitination and stabilization, independent of transcriptional regulation. FRMPD1 restoration rescued FBXO10-mediated proliferation, confirming its role as the key downstream effector. Clinically, FBXO10 expression correlated with TP53 mutations and adverse clinicopathological features. Our findings reveal a novel FBXO10–FRMPD1 axis promoting hepatocarcinogenesis through post-translational stabilization, positioning FBXO10 as both a prognostic biomarker and therapeutic target in HCC. Full article
(This article belongs to the Section Biochemistry, Molecular and Cellular Biology)
Show Figures

Graphical abstract

Back to TopTop