v
Search
Advanced

Publications > Journals > Journal of Clinical and Translational Hepatology> Article Full Text

  • OPEN ACCESS

Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications

  • Zixin Liang1,#,
  • Shanshan Li2,#,
  • Zhiyu Wang1,
  • Junting Zhou1,
  • Ziyue Huang3,
  • Jiehan Li4,
  • Haolin Bao1,
  • Judy Wai Ping Yam5,*  and
  • Yi Xu1,2,5,6,7,8,9,10,* 
 Author information 

Abstract

Hepatocellular carcinoma (HCC) is one of the deadliest malignant tumors in the world, and its incidence and mortality have increased year by year. HCC research has increasingly focused on understanding its pathogenesis and developing treatments.The Wnt signaling pathway, a complex and evolutionarily conserved signal transduction system, has been extensively studied in the genesis and treatment of several malignant tumors. Recent investigations suggest that the pathogenesis of HCC may be significantly influenced by dysregulated Wnt/β-catenin signaling. This article aimed to examine the pathway that controls Wnt signaling in HCC and its mechanisms. In addition, we highlighted the role of this pathway in HCC etiology and targeted treatment.

Graphical Abstract

Keywords

Hepatocellular carcinoma, Wnt signaling, Pathogenesis, Proliferation, Invasion, Metastasis, Therapy

Introduction

Hepatocellular carcinoma (HCC) has steadily become a malignancy with a very high incidence and fatality rate in China, and even globally.1,2 After the pathological diagnosis of patients with liver cancer, it was found that the type with the highest incidence was HCC. Annually, around 700,000 new cases of liver cancer are reported worldwide, of which primary liver cancer accounts for about 300,000–400,000 cases, and ninety percent of these patients have HCC.3 The pathogenesis of HCC has not yet been thoroughly studied, but the main causes of HCC are viral infections, long-term alcohol abuse, exposure to liver-damaging toxins, long-term chronic liver damage, and some immune and metabolic diseases.4 Due to its high degree of malignancy, invisibility, and inconspicuous onset symptoms, many patients are diagnosed at a more advanced stage, losing the chance for radical treatment by surgery, and can only adopt conservative treatments.5 Still, the indications for surgery are limited and restricted to HCC patients with good liver function, appropriate mass size, and no vascular invasion. Conservative treatments for advanced HCC mainly include chemotherapy, immunotherapy, targeted therapy, radiofrequency ablation, and transarterial chemoembolization.6,7 However, conservative treatment is not a radical therapy, so the curative effect is unsatisfactory. These treatments are expensive and have many side effects, which bring inevitable physical and mental trauma to patients.8 As research on HCC progresses, the understanding of its pathogenesis has deepened, and new ideas and methods for clinical treatment have been proposed. However, despite this, the incidence and five-year survival rate of HCC have not been significantly improved.9 The Wnt signaling pathway plays a crucial role in processes such as cell proliferation, differentiation, migration, and embryonic development.10 In recent years, an increasing number of studies have revealed the significance of the Wnt signaling pathway in cancer, particularly in the initiation and progression of malignant tumors such as HCC, colorectal cancer, and breast cancer.11,12 Aberrant activation of the Wnt pathway has been shown to be a key factor driving tumor cell proliferation, invasion, and metastasis. Recent genomic and transcriptomic analyses have classified HCC into three molecular subtypes: C1, C2, and C3.13 In the C1 subtype, Wnt activation is linked to increased hepatocyte proliferation and anti-apoptotic abilities, often through abnormal regulation of β-catenin stability and nuclear translocation.14 In the C2 subtype, Wnt signaling enhances invasiveness and metastasis by activating the epithelial-mesenchymal transition (EMT), with β-catenin upregulated.15 Although less studied in the C3 subtype, Wnt signaling may modulate immune cell function by regulating the immunosuppressive microenvironment in HCC.16 Therefore, research on HCC remains urgent, and integrating progress in Wnt signaling pathway research and revealing its potential mechanisms can help inspire new research directions and clinical diagnosis and treatment approaches.

Wnt signaling pathway

The Wnt signaling pathway plays a vital role in maintaining the body’s homeostasis and physiological balance.17 Wnt signaling was first revealed in the 1970s and described as an embryonic developmental mutation-associated gene-phenotype. In experimental animal studies conducted at that time, Wnt signaling was found to be associated with mammary tumor gene activation in mice.18,19 The first human tumor linked to Wnt signal transduction progresses from familial adenomatous polyposis to colon cancer.20 As a result, an increasing body of research has demonstrated that the Wnt signaling pathway is primarily responsible for initiating intracellular signaling pathways and facilitating cell-to-cell information transfer, contributing to the development of many cancers.21,22 The atypical planar cell polarity (PCP) route, the Wnt/Ca2+ pathway, and the canonical Wnt/β-catenin pathway are the three main branches of the Wnt signaling pathway.23 Upon learning that the Wnt signaling pathway contributes to the growth of malignancies, researchers began to explore the pathways through which Wnt signaling is regulated. In conjunction with previous studies on Wnt signaling in different tumors, researchers found that extracellular vesicles (EVs) in tumors contribute to tumor progression and metastasis by regulating Wnt signaling. Tumors where this mechanism has been clearly identified include cervical, breast, and colon cancers, among others.24,25 According to current research, the primary method by which EVs modulate Wnt signaling is by altering the Wnt/β-catenin pathway, the traditional route.26 EVs carry miRNAs that regulate the activity of the Wnt signaling pathway by targeting key genes.27 For example, Wu et al. examined the methylation status of miR-137 using 1 TCGA data and 3 GEO datasets, confirming that miR-137 inhibits CRC cell proliferation, migration, invasion, and xenograft tumor growth. miR-137 accelerates the degradation of c-Myc and β-catenin through the inhibition of RNF4, which affects protein stability and the inhibition of the Wnt pathway, highlighting its therapeutic potential in CRC.28 Lin et al. enhanced the LNC-POTEM-4/miR-149-5p/Wnt4-regulated signaling axis by inhibiting negative regulators of the Wnt pathway (such as adenomatous polyposis coli (APC) and axis inhibition protein (AXIN) or increasing the stability and nuclear translocation of β-catenin, promoting tumor cell proliferation and metastasis.29 Additionally, EVs can serve as carriers of Wnt signaling molecules, such as Wnt3a and Wnt5a, which bind to Frizzled receptors on recipient cells, activating the Wnt pathway and regulating target gene expression. This mechanism not only facilitates signaling between cancer cells but may also influence the behavior of immune cells and endothelial cells in the tumor microenvironment (TME).30 The majority of patients with hepatocellular carcinoma have verified mutations encoding Wnt components. One such gene is CTNNB1, which encodes the β-catenin protein. Hepatocellular carcinoma development is thought to be closely associated with abnormal activation of this system.31,32 CTNNB1 mutations lead to abnormal β-catenin stability and nuclear translocation, enhancing Wnt/β-catenin signaling activity, promoting tumor cell proliferation, and inhibiting apoptosis, while also affecting the TME.33 Overactivation of β-catenin can induce the secretion of immune-suppressive cytokines such as transforming growth factor β (TGF-β) and IL-10, inhibit the activity of effector T cells, and promote the accumulation of Treg and MDSC cells, leading to immune evasion.34 Furthermore, mutations in AXIN and glycogen synthase kinase 3β (GSK-3β) can sustain Wnt/β-catenin signaling, enhancing β-catenin stability and further driving tumor growth, while also altering the immune microenvironment, promoting immune evasion and inflammation.35

Wnt/β-catenin signaling pathway

The Wnt/β-catenin signaling pathway is composed of three primary components: proteins associated with the Wnt signaling pathway, complexes that regulate the degradation-related fate of β-catenin proteins, and crucial transcriptional regulators found in the nucleus. Each of these components has unique qualities that either prevent or promote cancer.36 The main functions of β-catenin proteins are to act as mediators of intercellular adhesion between neighboring cells and, in the presence of Wnt receptors, to function as key Wnt signaling components, activating downstream gene transduction and responding to nuclear signals.37,38 The most important degradation complexes of the pathway typically consist of a complex containing AXIN, GSK-3β, casein kinase I, and APC, which are capable of phosphorylating and degrading β-catenin proteins to maintain them at a stable level, preventing downstream genes from being transcribed.39,40 The degradation complex is simultaneously prepared for action by the activation of the Wnt signaling pathway.41 Another important aspect is that the control of Wnt signaling activity requires multiple positive and negative receptors, such as Dickkopf-1.42 This mechanism helps maintain the typical Wnt signaling pathway in a hypoactive state. Together, these mechanisms keep β-catenin proteins at consistently low levels and prevent Wnt signaling overexpression. Transmembrane Frizzled (FZD), a glycoprotein present on the surface of cells, is the primary ligand that binds to the Wnt classical pathway. Nineteen isoforms of the human Wnt ligand gene are also known to be involved in Wnt signaling on the cell surface. Furthermore, it has been demonstrated that Wnt can selectively recognize FZD on the cell surface and that the expression of Wnt is significantly higher in tumor tissues than in normal tissues.22 The Wnt signaling pathway becomes pathogenic when there is continuous accumulation of β-catenin. One key event that leads to this occurrence is the inhibition of the axonemal protein complex from phosphorylating β-catenin proteins. Cytoplasmic disheveled (DVL) proteins and the co-receptor low-density lipoprotein receptor-associated proteins 5/6 (LRP5/6) are the main mediators of this activity. Accumulated DVL is phosphorylated in the cytosol by a variety of protein kinases, causing AXIN to lose its degradation function by interacting with DVL and FZD, allowing β-catenin proteins to relocate to the nucleus and carry out a transcriptional function (Fig. 1).43–45

Wnt/β-Catenin signaling pathway.
Fig. 1  Wnt/β-Catenin signaling pathway.

FZD, frizzled; DVL, dishevelled; APC, adenomatous polyposis coli; CK1, casein kinase 1; AXIN, axis inhibition protein; TCF, T-cell Factor; LEF, lymphoid enhancer binding factor; LRP5/6, low-density lipoprotein receptor-related protein 5/6; DKK1, dickkopf-related protein 1.

Non-canonical Wnt signaling pathway

Atypical Wnt signaling pathways include the Wnt/PCP and Wnt/Ca2+ signaling pathways (Fig. 2). Although researchers have not paid as much attention to these atypical pathways as to the canonical pathways, it has been established that many important physiological and pathological processes in the human body are impacted by abnormal Wnt pathways.46 Studies have shown that Wnt3, Wnt5, and Wnt10 are involved in initiating downstream target genes through atypical Wnt signaling pathways.47 This finding suggests that atypical Wnt signaling pathways may share similarities with canonical Wnt pathways, meaning anomalies in these pathways and certain route elements may be connected to the initiation and spread of tumors. Additionally, researchers have found that both pathways begin with the Wnt ligand binding to FZD, by comparing their initiation mechanisms. RHO-associated kinaseand c-Jun N-terminal kinases are two proteins linked to cytoskeletal control and target gene transcription that are activated when the Wnt/PCP pathway is triggered. This is due to the significant amount of DVL transmission information pulled by the Wnt/PCP pathway.48,49 PLC activation increases the breakdown of phosphatidylinositol (4,5)-biphosphates into diacylglycerol and inositol (1,4,5)-triphosphates, which in turn increases cytosolic calcium ion release in response to Wnt/Ca2+ signaling.50,51 Cyclic guanosine monophosphate expression is downregulated in response to increased calcium ions, whereas protein kinase C, calcineurin, and calmodulin-dependent protein kinase II are upregulated. Calmodulin-dependent protein kinase II and calcineurin both activate the nuclear factor of activated T-cells, which translocate to the nucleus and is involved in target gene regulation, and Nemo-like kinase, which suppresses the canonical Wnt signaling pathway.52–54

Non-canonical Wnt signaling pathway.
Fig. 2  Non-canonical Wnt signaling pathway.

FZD, frizzled; DVL, disheveled gene; ROCK, Rho-associated coiled-coil kinase; RhoA, Ras homolog gene family member A; DAAM1/2, dishevelled-associated activator of morphogenesis 1/2; JNK, Jun N-terminal kinase C-Jun-N-terminal kinase; PLC, phospholipase C; cGMP, cyclic guanosine monophosphate; DAG, diacylglycerol; IP3, inositol trisphosphate; PKC, protein kinase C; NLK, nemo-like kinase; NFAT, nuclear factor of activated T-cells; ↑, up-regulated expression; ↓, down-regulated expression.

The role of the Wnt signaling pathway in HCC

Involvement in pathogenesis

The Wnt/β-catenin protein signaling pathway plays a significant role in the development of HCC and is essential for regulating several physiological and pathological liver activities. There is approximately a 40% genetic variation between its key components, and aberrant expression and mutations have been demonstrated to be crucial for the development and dedifferentiation of HCC.55,56 EVs in HCC maintain a certain level of invasiveness of HCC cells by regulating Wnt signaling and delivering contents between cells, which in turn mediates the biological behavior of the cells and promotes tumor progression.57 It has been confirmed that a large amount of abnormally activated Wnt and its contents exist in HCC tissues, further activating downstream target genes through its signaling pathway, which maintains the high invasiveness of HCC.58,59

The most common gene mutation in the Wnt/β-catenin signaling pathway occurs in exon 3 of the β-catenin-encoding CTNNB1 gene.60 According to one study, about 30% of the CTNNB1 gene, which codes for β-catenin proteins, is mutated in HCC cells. This mutation interacts with T cell factor (TCF) 4, a transcription factor specific to Wnt cells, to initiate metabolism and proliferation before Wnt response elements are recruited. This results in blocked phosphorylation of β-catenin proteins, which, in combination with mutant histone modifying factors and chromatin remodeling factors, prevents their degradation by the proteasome, allowing the translocation of β-catenin proteins to the nucleus. The findings indicate that a compound containing APC, AXIN1, GSK3β, and CK1 is responsible for this process.61–63 Two of the most well-studied mechanisms of aberrant activation of the Wnt/β-catenin signaling pathway are cellular mutations and epigenetic modifications. Promoter methylation and the aberrant synthesis of non-coding RNAs are among the other factors. Regional and ethnic factors affect CTNNB1 mutations, with significantly more CTNNB1 mutations found in patients from Asia and Europe compared to HCC patients from Africa.64 In addition, etiological factors of HCC also play a role. Research indicates that CTNNB1 mutations are more common in HCC caused by HCV than in HCC caused by HBV, and alcohol-associated HCC has the highest correlation with the frequency of CTNNB1 mutations, approximately 42%.65,66 Mutations affecting AXIN, GSK-3β, and APC, or their lack of function as AXIN complex constituents, can also trigger the pathway and cause cancer. APC gene inactivation results in cancer through the combined effects of GSK-3β phosphorylation, hepatic β-catenin overexpression, and AXIN mutations.67,68 Zhang et al. found in earlier research that the Wnt3/FZD7-mediated β-catenin signaling pathway was more highly expressed in HCC. Subsequent analysis of the effects of Wnt3 binding to the N-glycosylation site-directed mutation of FZD7 on HCC cell development showed that Wnt3/β-catenin signaling pathway-related protein levels were negatively impacted by downregulating Wnt3’s ability to bind to FZD7, which in turn affected cell invasion, migration, and proliferation.69 Similarly, related studies have demonstrated that a variety of Wnts act in conjunction with various FZD receptors, overexpressing and decreasing the levels of secreted frizzled-associated proteins in tissues and leading to their methylation in HCC and its surrounding tumor microenvironment. Initially, secreted frizzled-associated proteins functioned to oppose the Wnt/β-catenin signaling pathway; however, as their quantity decreased and inactivation occurred, the ability of the pathway to be activated increased, gradually accumulating and promoting HCC tumorigenesis.70–72 This study has validated two important factors that lead to aberrant activation of the Wnt/β-catenin protein signaling pathway in HCC tumor formation: the previously reported CTNNB1 gene mutations and the loss of function or mutations in AXIN, GSK-3β, and APC complexes.73,74 Among these, missense mutations or deletions account for most loss-of-function mutations of GSK-3β and APC axis protein complexes, resulting in a decrease in the negative regulatory function of the pathway. These also facilitate the translocation of β-catenin proteins from the cytoplasm to the nucleus.75,76 Mutations in CTNNB1 are more likely to occur in both ways. Missense mutations or deletions of APC hypermethylate the WNT inhibitory factor-1 promoter, preventing the WNT inhibitory factor-1 from being expressed, causing cells to lose their usual control and encouraging the growth of HCC cells.77 Activation of receptor tyrosine kinases, high levels of R-spondins, and TGF-β-dependent activation in HCC are just a few of the ways that the Wnt/β-catenin signaling pathway is activated. The interaction of multiple HCC-related high-risk factors, either together or separately, leads to HCC tumorigenesis, which is extremely complex. These mechanisms are still under investigation.78–80

Shaping the TIME

The Wnt signaling pathway regulates the immune system in HCC through multiple mechanisms. First, Wnt signaling upregulates immunosuppressive factors such as TGF-β and IL-10, promoting the accumulation of Treg and MDSC cells, which in turn inhibit the function of effector T cells and weaken the anti-tumor immune response.81 Abnormal activation of the Wnt/β-catenin pathway can also alter the interactions between HCC tumor cells and immune cells, further facilitating immune escape.82 Secondly, Wnt signaling may influence the polarization of tumor-associated macrophages, inducing their conversion to an immunosuppressive phenotype, which enhances immune evasion mechanisms in the TME and impedes the immune system’s ability to eliminate the tumor.83 Additionally, the Wnt pathway regulates cytokines and chemokines in the TME, reinforcing the immunosuppressive environment and supporting tumor cell survival and expansion under immune pressure.84

Promote malignant biological behavior

Proliferation

When the Wnt signaling pathway is activated, the amount of β-catenin protein increases. This protein can enter the nucleus and bind to TCF or lymphatic factor (LEF) to regulate the production of downstream target genes such as c-Myc and Cyclin D1.85,86 These genes are upregulated, which promotes cell cycle progression and boosts cell proliferation.87 In HCC cells, the Wnt signaling pathway is often aberrantly activated, releasing β-catenin from the GSK-3β degradation complex and causing it to accumulate in the cytoplasm.88 Subsequently, β-catenin penetrates the nucleus and associates with TCF/LEF family members to form a transcriptional complex.89 A cell’s Frizzled and LRP5/6 receptors are the sites where Wnt ligands bind to activate the Wnt signaling pathway, which in turn inhibits GSK-3β activity.90 GSK-3β is typically responsible for phosphorylating β-catenin, which accelerates its breakdown.91 GSK-3β inhibition prevents β-catenin from being phosphorylated and eliminated, causing it to accumulate in the cytoplasm.92 The accumulating β-catenin is then translocated to the nucleus, where it binds to TCF/LEF transcription factors.93 Once the transcriptional complexes are formed, downstream genes are activated to express themselves.94 These β-catenin-induced genes promote rapid cell division by facilitating the transition from the G1 phase to the S phase of the cell cycle.95 The G1/S phase transition requires CDK4/6 activity, which can be increased by upregulating β-catenin.96 Additionally, β-catenin can prevent the production of genes linked to apoptosis, such as Bax and Bim, thereby inhibiting apoptosis.97 Studies have demonstrated that increased β-catenin accelerates HCC cell proliferation, invasion, and metastasis, and encourages tumor development.98

Activation of the Wnt signaling pathway can sustain liver stem cells’ capacity for self-renewal, allowing them to proliferate without differentiating into fully developed hepatocytes.99 HCC may originate from these undifferentiated stem cells.100 Several genes linked to stem cell properties can also be activated by the accumulation of β-catenin in the nucleus.101 Studies have indicated that HCC cells driven by the Wnt signaling pathway exhibit stem cell-like characteristics, including the ability to differentiate in multiple directions and self-renew.102 By upregulating the expression of genes related to the cell cycle, inhibiting apoptosis, and promoting HCC cell proliferation, the Wnt pathway enables HCC cells to survive and grow in harsh conditions.103 The Wnt signaling pathway also promotes cell division by upregulating the production of cyclins and kinases, thereby maintaining the stem cell pool, activating DNA damage repair mechanisms, and helping HCC cells repair DNA damage, which supports their self-renewal ability.104,105

Invasion and metastasis

The increased malignant biological behavior of HCC patients is a major cause of their high mortality rate. Wnt signaling pathway activation and the development of EMT are closely related.106 EMT enables epithelial cells, such as liver cells, to shed their epithelial characteristics and acquire mesenchymal characteristics, such as the ability to migrate and invade. EMT is considered a key indicator of tumor cells’ propensity to infiltrate.107 The Wnt signaling pathway regulates the expression of N- and E-cadherin, which can further this process.108 E-cadherin is a protein responsible for the adhesion of epithelial cells. The Wnt signaling pathway prevents the expression of E-cadherin by accumulating β-catenin, and its decreased expression leads to reduced intercellular adhesion, promoting EMT.109 The β-catenin-TCF/LEF complex can activate mesenchymal markers such as vimentin, fibronectin, and N-cadherin. The upregulation of these proteins imparts interstitial properties to cells.110,111 The mesenchymal characteristics acquired during EMT facilitate the migration of HCC cells to surrounding tissues. Mesenchymal cells also exhibit enhanced invasion ability, allowing them to enter the blood circulation by passing through the vascular wall and basement membrane, thereby increasing the metastatic potential of HCC.112 Additionally, HCC cells with mesenchymal characteristics enable epithelial cells to survive even in the absence of adhesion, inhibiting tumor cell apoptosis.113 HCC cells’ ability to upregulate angiogenesis-related factors during EMT, such as vascular endothelial growth factor (VEGF), helps supply nutrients and oxygen for tumor development, potentially promoting the formation of tumor blood vessels.114 The Wnt signaling pathway can increase the expression of matrix metalloproteinase (MMP) genes, such as MMP-2 and MMP-9, and facilitate β-catenin’s nuclear translocation and binding to the transcription factor TCF/LEF. These enzymes promote tumor cell motility and invasion by breaking down collagen and elastin, two components of the extracellular matrix (ECM). While MMP activity is negatively regulated by tissue inhibitors of metalloproteinases (TIMPs), MMPs are crucial for matrix disintegration.115 By promoting the breakdown of TIMPs, the Wnt signaling pathway can increase MMP activity and reduce ECM inhibition. In some cases, the Wnt signaling pathway may directly increase TIMP expression, but this typically occurs in conjunction with the degradation of MMPs to maintain proper ECM degradation.116

The Wnt signaling pathway can also increase integrin expression. Integrins bind to ECM components, enhancing cell migration and invasion, and activating Rho family GTPases, including Rac1 and RhoA. These GTPases affect cell morphology and movement by regulating the polymerization and recombination of cytoskeletal proteins (such as actin and tubulin). These functions are primarily achieved by upregulating cell migration-related genes, such as MMPs, TIMPs, and cytoskeleton-related proteins.117,118 Moreover, Wnt signaling pathway activation may aid in modifying the HCC tumor microenvironment. In addition to promoting VEGF production, it can also support immunosuppression by inhibiting T cell activation and function, and by triggering the release of various cytokines and growth factors from tumor cells, such as platelet-derived growth factor, TGF-β, and fibroblast growth factor, all of which promote tumor invasion and metastasis.119,120 Furthermore, it can regulate the expression of cell adhesion molecules and chemokines, influencing immune cell recruitment and tumor cell infiltration. This suggests that tumor cells have an increased propensity to penetrate blood vessels and the lymphatic system, facilitating their entry into the blood circulation, infiltration, and metastasis.121–123

Therapy

Chemotherapy is the mainstay of treatment for patients with advanced HCC. However, the emergence of targeted therapy has brought new hope for these patients. Compared with traditional chemotherapy methods, targeted therapy is more specific. It has less impact on the normal internal environment of the human body, fewer side effects, better drug compliance, and more obvious efficacy.124 Because of the Wnt signaling pathway’s crucial role in HCC, targeting it has become a feasible therapeutic strategy.125 The related studies include Wnt signaling pathway inhibitors, downstream Wnt signaling pathway inhibitors, combination therapies targeting cell adhesion and migration, etc. (Table 1).126–134

Table 1

Drugs acting on the Wnt/β-catenin signaling pathway in HCC

MedicinesAction mechanismClinical stageReferences
LGK974Inhibition of Porcupine reduces the secretion of Wnt ligandsClinical phase I126
PRI-724Inhibit the interaction between β-catenin and CBP, block the signal transductionClinical phase I127
ICG-001Inhibition of β-catenin binding to CBP blocks Wnt/β-catenin pathwayPreclinical study/clinical phase I128
FH535Inhibition of Wnt/β-catenin and PPAR pathwaysPreclinical study129
XAV939Inhibit Tankyrase, increase AXIN level and reduce the accumulation of β-cateninPreclinical study130
VantictumabAntibody drugs, targeting Frizzled receptors, block Wnt signalingPreclinical study/clinical phase I131
DKK1Blocking Wnt binding to Frizzled receptor and inhibiting Wnt pathwayPreclinical study132
NiclosamideInhibition of Wnt/β-catenin pathway reduces cell invasion ability.Preclinical study133
SalinomycinBy inhibiting the Wnt/β-catenin pathway, it reduces the migration and invasion of cancer cells.Preclinical study134

β-catenin inhibitors, which are further subdivided into GSK-3β inhibitors and small molecule β-catenin inhibitors, are the most significant class of Wnt signaling pathway inhibitors.135 β-catenin inhibitors in small molecules function in two ways. First, they bind directly to β-catenin, blocking its entry into the nucleus and activation of downstream genes. The second mechanism limits the activity of ubiquitinating enzymes (such as MMP7) to reduce β-catenin breakdown, stabilizing it and lowering its activity.136 Importantly, GSK-3β inhibits the Wnt signaling pathway. Inhibiting its activity stabilizes β-catenin and leads to its accumulation in the nucleus. This indirect suppression of the Wnt signaling pathway has led to the development of GSK-3β inhibitors.137 In addition, Wnt competitive inhibitors are substances that competitively bind to Wnt receptors, preventing the activation of the Wnt signaling pathway. Currently, clinical trials for small molecule β-catenin inhibitors are underway, represented by Inducible Cytotoxic GPC3-001, which has entered the clinical trial stage. In patients with HCC, phase I clinical research has shown some anti-tumor activity and good tolerability; the phase II clinical trial is currently in progress to assess its safety and efficacy in more detail.138 Wnt response inhibitors (IWR) are the main type of GSK-3β inhibitor. Research on IWR-1 has attracted widespread attention and has been clinically tested in various cancer types, showing certain anti-tumor activity and potential for the treatment of HCC.139 Representative Wnt competitive inhibitors include κBα (IκBα) analog inhibitors. IκBα analogs inhibit the Nuclear Factor kappa-light-chain-enhancer of activated B cells (NF-κB) signaling pathway by mimicking IκBα, which decreases IκB kinase activity and increases the expression of anti-apoptotic proteins (such as Bcl-2) while decreasing pro-apoptotic proteins (such as Bax), promoting apoptosis of HCC tumor cells.140,141 Clinical studies of related competitive inhibitors and antibodies in HCC are ongoing, and these drugs may provide new treatment options for HCC patients.

The downstream components of the Wnt signaling pathway are primarily responsible for obtaining, transmitting, and amplifying Wnt signals, including related receptors, TCF/LEF transcription factors, and effector molecules. An essential part of the Wnt signaling pathway is Frizzled (Fzd). Fzd receptors initiate signal transduction by attaching to Wnt ligands on the cell membrane. Fzd inhibitors interact with Frizzled receptors to prevent Wnt protein from binding, thus interrupting Wnt signal transduction, reducing downstream target gene transcription, lowering HCC cell survival and proliferation, and inhibiting tumor growth (Table 2).142–144 Inhibition of Fzd3 has been shown to suppress HCC cell proliferation and invasion while promoting tumor growth in animal models, providing a strong basis for further human studies.145,146 TCF/LEF transcription factors are crucial in the downstream Wnt signaling cascade. Inhibitors of TCF/LEF have the potential to restrict the expression of Wnt target genes by blocking the interaction between β-catenin and TCF/LEF, disrupting the Wnt signaling pathway and decreasing tumor cell proliferation and survival.147 Specific inhibitors of TCF/LEF include BAY 11-7082. Recent research has demonstrated that BAY 11-7082, in conjunction with CDK4/6 inhibitors, is beneficial against a range of malignancies, including HCC, in animal trials.148 Furthermore, studies have shown a negative correlation between tumor growth and the production of WISP1, a downstream effector of the Wnt signaling pathway. WISP1 stops HCC progression by negatively regulating β-catenin/TCF/LEF signaling, suggesting a potential therapeutic avenue for targeting Wnt downstream effector molecules to halt HCC progression.149

Table 2

Drugs targeting downstream effector molecules of Wnt signaling in HCC

MedicinesAction mechanismClinical stageReferences
PNU-74654It directly binds to β-catenin, prevents its formation with TCF complex, and reduces gene transcriptionPreclinical study142
MSABInhibition of β-catenin-mediated transcriptional activity reduces Wnt signaling downstream gene expressionPreclinical study143
PBP(Pyrvinium pamoate)By interfering with the Wnt signaling pathway and inhibiting the activity of β-catenin, the transcription of downstream genes is inhibitedPreclinical study144

MMPs are important players in the Wnt signaling pathway, stimulating HCC tumor cells’ adhesion, motility, and invasion. Inhibition of MMPs can reduce these processes and prevent premature metastasis of tumor cells.150 MMP inhibitors directly block the activity of MMPs through chemical structures or biological pathways, reducing ECM degradation and regulating MMP gene expression or protein synthesis to lower MMP production.151 The current research on MMP inhibitors focuses on Batimastat (BB-94) and Marimastat (BB-2516).152 MMP inhibitors have been demonstrated in several clinical trials to reduce HCC tumor invasion and improve prognosis. Studies have shown that BB-94 can prevent HCC cells from migrating and inhibit angiogenesis in the tumor vascular system. Furthermore, the tumor blood vessels that are prevented from growing cannot supply enough oxygen and nutrients for the remaining HCC cells.153 MMP inhibitors target MMPs overexpressed in the tissues and cells of hepatocellular carcinoma. After years of research, many clinical drugs have been produced, such as Bamalabaster, Novartis, BAY12-9566, AG-3340, OPB-3206, KBR07785, and KBR-8301.154–156 Cell adhesion molecules (CAMs) are important for mediating interactions between cells or between cells and the extracellular matrix.157 CAM modulators can affect CAM content and activation pathways, influencing tumor cell adhesion, migration, and invasion (Table 3).158–175 Various types of CAM modulators include E-cadherin agonists, which can enhance intercellular adhesion and reduce tumor cell invasion by promoting E-cadherin expression and function.176 SMADS agonists can initiate SMADS phosphorylation and nuclear translocation through the TGF-β/SMAD signaling pathway. By binding to the E-cadherin promoter region and promoting E-cadherin production, SMADS interacts with β-catenin to improve cell adhesion and Wnt signaling. Studies have indicated that interferon-α and TGF-β1 may effectively block the development of the β-catenin/TCF4/Smads complex, suggesting that these molecules could be employed as potential therapeutics for HCC patients.177,178 Integrin modulators inhibit integrins from binding to ligands, reducing cell adhesion and migration. Examples include antibody-coupled drugs including ramucirumab and atezolizumab, which can bind integrins and target tumor cells.179,180 Cetuximab, a small molecule integrin blocker, may be particularly effective in preventing advanced HCC by obstructing the interaction between integrins and EGFR.181

Table 3

Drugs targeting cell adhesion and migration in the Wnt signaling pathway in HCC

MedicinesAction mechanismClinical stageReferences
Batimastat (BB-94)Broad-spectrum MMP inhibitors inhibit MMP-1, MMP-2, MMP-9, etc., and reduce tumor invasion and migrationClinical stage I/II158
Marimastat (BB-2516)Broad-spectrum MMP inhibitors inhibit MMP-1, MMP-2, MMP-9, etc., and reduce tumor invasion and migrationClinical Phase II159
Tanomastat (BAY 12-9566)MMP inhibitors, targeting MMP-2 and MMP-9, inhibit the degradation of extracellular matrix and prevent tumor metastasisClinical Phase II160
TIMP-2Endogenous MMP inhibitors directly inhibit MMP-2 activity and reduce matrix degradation and tumor invasionPreclinical study161
DoxycyclineAntibiotics, non-specific MMP inhibitors, inhibit MMP-2 and MMP-9, reduce tumor invasion and metastasisPreclinical study/clinical phase I162
Ilomastat (GM6001)Broad-spectrum MMP inhibitors, inhibit MMP-1, MMP-2, MMP-9, reduce extracellular matrix degradationPreclinical study163
AG3340 (Prinomastat)Selective MMP inhibitors, mainly inhibit MMP-2 and MMP-9, reduce extracellular matrix degradation and invasionClinical phase I164
ONO-4817Selective inhibition of MMP-2 and MMP-9 reduced tumor cell invasion and matrix degradationPreclinical study165
MinocyclineInhibit the activity of MMP-2 and MMP-9, reduce matrix degradation and tumor cell migrationPreclinical study166
E7820Inhibit the expression of integrin α2, reduce tumor cell adhesion and angiogenesisClinical stage I / II167
VolociximabTargeting integrin α5β1 prevents the binding of tumor cells to the extracellular matrix and inhibits tumor invasionClinical Phase II168
Etaracizumab (Abegrin)Targeting integrin αvβ3 reduces tumor cell adhesion and angiogenesis, and inhibits tumor growth and metastasisClinical Phase II169
CilengitideInhibition of integrin αvβ3 and αvβ5 inhibits tumor angiogenesis and tumor cell invasion.Clinical Phase II170
Simtuzumab (GS-6624)Anti-LOXL2 (lysyl oxidase-like protein 2) antibody prevents extracellular matrix remodeling and reduces tumor invasionClinical Phase II171
DisulfiramIt inhibits the expression of cell adhesion molecules (such as N-cadherin) and prevents cancer cell migration and invasionPreclinical study172
ADH-1Targeting N-cadherin inhibits intercellular adhesion and reduces invasion and metastasis of tumor cellsPreclinical study173
Galunisertib (LY2157299)TGF-β inhibitors inhibit the expression of adhesion molecules and reduce tumor cell invasion and metastasisClinical Phase II174
MetforminIndirectly inhibit the expression of cell adhesion molecules, reduce the invasion ability of tumor cellsPreclinical study175

Several therapeutic modalities related to the Wnt signaling pathway, including immune checkpoint inhibitors and CAR-T cell therapy, are now in the preclinical or animal experimental stages, in addition to the previously mentioned treatment strategies.182,183 The combination of Wnt signaling pathway-related treatments with other therapeutic measures (such as immunotherapy, VEGF inhibitor therapy, etc.) can improve efficacy, eliminate the side effects of monotherapy, and reduce drug resistance.184 Targeting the Wnt signaling pathway holds great potential in clinical practice, but it also faces several challenges. Therapeutic approaches targeting the Wnt pathway may lead to non-specific off-target effects, affecting the function of normal cells and tissues, particularly in the liver, intestine, and bones.185 Although targeting the Wnt pathway has shown promising efficacy in vitro and in animal models, drug resistance is a common issue in clinical applications. Activation of the Wnt signaling pathway involves multiple regulatory mechanisms, including the stability of β-catenin, upstream Frizzled receptors, and downstream transcription factors. The complexity of these regulatory processes allows tumor cells to bypass targeted therapies in various ways.186 Additionally, abnormal stability or nuclear translocation of β-catenin may be maintained through mutations or other pathways, reducing the effectiveness of drugs.187 The regulation of the Wnt pathway is highly complex, involving multiple key molecules, and different types of tumors may depend on different Wnt signaling subtypes, which complicates targeted Wnt therapy and introduces issues of tumor heterogeneity.188 The Wnt signaling pathway’s high degree of intricacy and variety in regulation is a result of its numerous interactions with other signaling pathways. Accurate targeting is extremely challenging, which highlights the significance of combination therapy. Due to the Wnt pathway’s intricate role in the human body, therapeutic techniques for it are currently being developed slowly and require further research.

Conclusions

The initiation, development, and metastasis of HCC are closely dependent on the Wnt signaling pathway. One mechanism driving aberrant activation of this pathway is the production of excess Wnt ligands, which leads to mutations in Wnt receptors or downstream effector molecules, such as β-catenin, and the loss of Wnt pathway inhibitors. However, despite increasing research suggesting the potential for targeted therapies that modulate the Wnt signaling pathway, several challenges remain. The complexity of the Wnt pathway, the diversity of cancer types, and the issue of drug resistance are major obstacles. These factors contribute to the absence of clinically available Wnt signaling inhibitors, making effective treatment difficult.

In conclusion, while our understanding of the Wnt signaling pathway in HCC is still evolving, there is hope for the development of more precise targeted therapies in the future. These therapies would aim to control toxic side effects while providing effective treatment. Additionally, personalized treatment plans based on a patient’s specific tumor genome and clinical features may become feasible. Identifying biomarkers to select patients who are most likely to benefit from Wnt-targeted therapies represents an exciting direction for future research in this area.

Declarations

Funding

This research was funded by the National Natural Science Foundation of China (82472743; 82300921); the Beijing Xisike Clinical Oncology Research Foundation (Y-QL202201-0020); the Beijing Science and Technology Innovation Medical Development Foundation (KC2023-JX-0186-FM046); the Anhui Engineering Technology Research Center of Biochemical Pharmaceutical (Bengbu Medical University)(2024SYKFD01); the Opening Project of the Scientific and Technological Innovation Major Base of Guangxi (2022-36-Z05-GXSWBX202201); the Key Laboratory of Clinical Laboratory Diagnosis and Translational Research of Zhejiang Province (2022E10022); the Opening Project of Fujian Provincial Key Laboratory of Tumor Biotherapy (FJZL2023001); the Opening Project of Fujian Provincial Key Laboratory of Translational Cancer Medicine (TCM2024-3); the Opening Project of Fujian Provincial Key Laboratory of Innovative Drug Target Research (FJ-YW-2024KF02); the Thematic Research Support Scheme of the State Key Laboratory of Liver Research, The University of Hong Kong (SKLLR/TRSS/2022/08); the Chen Xiaoping Foundation for the Development of Science and Technology of Hubei Province (CXPJJH123003-005); the Postgraduate Research & Practice Innovation Program of Harbin Medical University (YJSCX2024-110HYD); the Construction Project of Shanghai Key Laboratory of Molecular Imaging (18DZ2260400).

Conflict of interest

JWPY has been an Editorial Board Member of Journal of Clinical and Translational Hepatology since 2021. The other authors have no conflict of interests related to this publication.

Authors’ contributions

Study concept and design (ZL, SL, ZW, JZ, ZH, JL, HB), writing of the manuscript (ZL, SL), revision (JWPY, YX). All authors have read and agreed to the published version of the manuscript.

References

  1. Wong CM, Tsang FH, Ng IO. Non-coding RNAs in hepatocellular carcinoma: molecular functions and pathological implications. Nat Rev Gastroenterol Hepatol 2018;15(3):137-151 View Article PubMed/NCBI
  2. Affo S, Yu LX, Schwabe RF. The Role of Cancer-Associated Fibroblasts and Fibrosis in Liver Cancer. Annu Rev Pathol 2017;12:153-186 View Article PubMed/NCBI
  3. Bakrania A, Zheng G, Bhat M. Nanomedicine in Hepatocellular Carcinoma: A New Frontier in Targeted Cancer Treatment. Pharmaceutics 2021;14(1):41 View Article PubMed/NCBI
  4. Llovet JM, Burroughs A, Bruix J. Hepatocellular carcinoma. Lancet 2003;362(9399):1907-1917 View Article PubMed/NCBI
  5. Chakraborty E, Sarkar D. Emerging Therapies for Hepatocellular Carcinoma (HCC). Cancers (Basel) 2022;14(11):2798 View Article PubMed/NCBI
  6. Anstee QM, Reeves HL, Kotsiliti E, Govaere O, Heikenwalder M. From NASH to HCC: current concepts and future challenges. Nat Rev Gastroenterol Hepatol 2019;16(7):411-428 View Article PubMed/NCBI
  7. Thandra KC, Barsouk A, Saginala K, Aluru JS, Rawla P, Barsouk A. Epidemiology of non-alcoholic fatty liver disease and risk of hepatocellular carcinoma progression. Clin Exp Hepatol 2020;6(4):289-294 View Article PubMed/NCBI
  8. Vogel A, Martinelli E, ESMO Guidelines Committee. Updated treatment recommendations for hepatocellular carcinoma (HCC) from the ESMO Clinical Practice Guidelines. Ann Oncol 2021;32(6):801-805 View Article PubMed/NCBI
  9. Jemal A, Ward EM, Johnson CJ, Cronin KA, Ma J, Ryerson B, et al. Annual Report to the Nation on the Status of Cancer, 1975-2014, Featuring Survival. J Natl Cancer Inst 2017;109(9):djx030 View Article PubMed/NCBI
  10. Zhou Y, Xu J, Luo H, Meng X, Chen M, Zhu D. Wnt signaling pathway in cancer immunotherapy. Cancer Lett 2022;525:84-96 View Article PubMed/NCBI
  11. Zhao H, Ming T, Tang S, Ren S, Yang H, Liu M, et al. Wnt signaling in colorectal cancer: pathogenic role and therapeutic target. Mol Cancer 2022;21(1):144 View Article PubMed/NCBI
  12. Xu X, Zhang M, Xu F, Jiang S. Wnt signaling in breast cancer: biological mechanisms, challenges and opportunities. Mol Cancer 2020;19(1):165 View Article PubMed/NCBI
  13. Chaisaingmongkol J, Budhu A, Dang H, Rabibhadana S, Pupacdi B, Kwon SM, et al. Common Molecular Subtypes Among Asian Hepatocellular Carcinoma and Cholangiocarcinoma. Cancer Cell 2017;32(1):57-70.e3 View Article PubMed/NCBI
  14. Wang M, Guo H, Zhang B, Shang Y, Zhang S, Liu X, et al. Transcription factors-related molecular subtypes and risk prognostic model: exploring the immunogenicity landscape and potential drug targets in hepatocellular carcinoma. Cancer Cell Int 2024;24(1):9 View Article PubMed/NCBI
  15. Yang S, Zheng L, Li L, Zhang J, Wang J, Zhao H, et al. Integrative multiomics analysis identifies molecular subtypes and potential targets of hepatocellular carcinoma. Clin Transl Med 2024;14(6):e1727 View Article PubMed/NCBI
  16. Zeng C, Zhang L, Luo C, Yang C, Huang X, Fan L, et al. A stratification model of hepatocellular carcinoma based on expression profiles of cells in the tumor microenvironment. BMC Cancer 2022;22(1):613 View Article PubMed/NCBI
  17. Nusse R, Clevers H. Wnt/β-Catenin Signaling, Disease, and Emerging Therapeutic Modalities. Cell 2017;169(6):985-999 View Article PubMed/NCBI
  18. Bender W, Peifer M. Oncogenes take wing. Cell 1987;50(4):519-520 View Article PubMed/NCBI
  19. Sharma RP, Chopra VL. Effect of the Wingless (wg1) mutation on wing and haltere development in Drosophila melanogaster. Dev Biol 1976;48(2):461-465 View Article PubMed/NCBI
  20. Nishisho I, Nakamura Y, Miyoshi Y, Miki Y, Ando H, Horii A, et al. Mutations of chromosome 5q21 genes in FAP and colorectal cancer patients. Science 1991;253(5020):665-669 View Article PubMed/NCBI
  21. Pez F, Lopez A, Kim M, Wands JR, Caron de Fromentel C, Merle P. Wnt signaling and hepatocarcinogenesis: molecular targets for the development of innovative anticancer drugs. J Hepatol 2013;59(5):1107-1117 View Article PubMed/NCBI
  22. Chan KK, Lo RC. Deregulation of Frizzled Receptors in Hepatocellular Carcinoma. Int J Mol Sci 2018;19(1):313 View Article PubMed/NCBI
  23. Yao H, Ashihara E, Maekawa T. Targeting the Wnt/β-catenin signaling pathway in human cancers. Expert Opin Ther Targets 2011;15(7):873-887 View Article PubMed/NCBI
  24. Luga V, Zhang L, Viloria-Petit AM, Ogunjimi AA, Inanlou MR, Chiu E, et al. Exosomes mediate stromal mobilization of autocrine Wnt-PCP signaling in breast cancer cell migration. Cell 2012;151(7):1542-1556 View Article PubMed/NCBI
  25. Qiu JJ, Sun SG, Tang XY, Lin YY, Hua KQ. Extracellular vesicular Wnt7b mediates HPV E6-induced cervical cancer angiogenesis by activating the β-catenin signaling pathway. J Exp Clin Cancer Res 2020;39(1):260 View Article PubMed/NCBI
  26. Taelman VF, Dobrowolski R, Plouhinec JL, Fuentealba LC, Vorwald PP, Gumper I, et al. Wnt signaling requires sequestration of glycogen synthase kinase 3 inside multivesicular endosomes. Cell 2010;143(7):1136-1148 View Article PubMed/NCBI
  27. Petővári G, Tóth G, Turiák L, Kiss AL, Pálóczi K, Sebestyén A, et al. Dynamic Interplay in Tumor Ecosystems: Communication between Hepatoma Cells and Fibroblasts. Int J Mol Sci 2023;24(18):13996 View Article PubMed/NCBI
  28. Wu Y, Li H, Long Y, Zhang Z, Zhang F, Pan R, et al. Epigenetic Suppression of miR-137 Induces RNF4 Expression, Facilitating Wnt Signaling in Colorectal Cancer. Mol Carcinog 2024 View Article PubMed/NCBI
  29. Lin C, Wu J, Wang Z, Xiang Y. Long non-coding RNA LNC-POTEM-4 promotes HCC progression via the LNC-POTEM-4/miR-149-5p/Wnt4 signaling axis. Cell Signal 2024;124:111412 View Article PubMed/NCBI
  30. Martin-Medina A, Lehmann M, Burgy O, Hermann S, Baarsma HA, Wagner DE, et al. Increased Extracellular Vesicles Mediate WNT5A Signaling in Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2018;198(12):1527-1538 View Article PubMed/NCBI
  31. Han SI, Lim SC. Expression and Prognostic Significance of CDK8 and β-Catenin in Hepatocellular Carcinoma. In Vivo 2020;34(3):1387-1394 View Article PubMed/NCBI
  32. Li YH, Yang SL, Zhang GF, Wu JC, Gong LL, Ming-Zhong, et al. Mefloquine targets β-catenin pathway and thus can play a role in the treatment of liver cancer. Microb Pathog 2018;118:357-360 View Article PubMed/NCBI
  33. Xu C, Xu Z, Zhang Y, Evert M, Calvisi DF, Chen X. β-Catenin signaling in hepatocellular carcinoma. J Clin Invest 2022;132(4):e154515 View Article PubMed/NCBI
  34. de Man SMA, van Amerongen R. Zooming in on the WNT/CTNNB1 Destruction Complex: Functional Mechanistic Details with Implications for Therapeutic Targeting. Handb Exp Pharmacol 2021;269:137-173 View Article PubMed/NCBI
  35. Tran FH, Zheng JJ. Modulating the wnt signaling pathway with small molecules. Protein Sci 2017;26(4):650-661 View Article PubMed/NCBI
  36. Wu S, Liu Z, Chen J, Ma T, Wang L, Huang T, et al. PORCN promotes hepatocellular carcinogenesis via Wnt/β-catenin-dependent manner. Transl Cancer Res 2023;12(7):1703-1714 View Article PubMed/NCBI
  37. Lv D, Chen L, Du L, Zhou L, Tang H. Emerging Regulatory Mechanisms Involved in Liver Cancer Stem Cell Properties in Hepatocellular Carcinoma. Front Cell Dev Biol 2021;9:691410 View Article PubMed/NCBI
  38. Fan J, Wei Q, Liao J, Zou Y, Song D, Xiong D, et al. Noncanonical Wnt signaling plays an important role in modulating canonical Wnt-regulated stemness, proliferation and terminal differentiation of hepatic progenitors. Oncotarget 2017;8(16):27105-27119 View Article PubMed/NCBI
  39. He K, Gan WJ. Wnt/β-Catenin Signaling Pathway in the Development and Progression of Colorectal Cancer. Cancer Manag Res 2023;15:435-448 View Article PubMed/NCBI
  40. Li VS, Ng SS, Boersema PJ, Low TY, Karthaus WR, Gerlach JP, et al. Wnt signaling through inhibition of β-catenin degradation in an intact Axin1 complex. Cell 2012;149(6):1245-1256 View Article PubMed/NCBI
  41. Polakis P. Wnt signaling in cancer. Cold Spring Harb Perspect Biol 2012;4(5):a008052 View Article PubMed/NCBI
  42. Niehrs C. The complex world of WNT receptor signalling. Nat Rev Mol Cell Biol 2012;13(12):767-779 View Article PubMed/NCBI
  43. Cadigan KM, Waterman ML. TCF/LEFs and Wnt signaling in the nucleus. Cold Spring Harb Perspect Biol 2012;4(11):a007906 View Article PubMed/NCBI
  44. Steinhart Z, Angers S. Wnt signaling in development and tissue homeostasis. Development 2018;145(11):dev146589 View Article PubMed/NCBI
  45. Kretzschmar K, Clevers H. Wnt/β-catenin signaling in adult mammalian epithelial stem cells. Dev Biol 2017;428(2):273-282 View Article PubMed/NCBI
  46. Thompson MD, Monga SP. WNT/beta-catenin signaling in liver health and disease. Hepatology 2007;45(5):1298-1305 View Article PubMed/NCBI
  47. Zhan T, Rindtorff N, Boutros M. Wnt signaling in cancer. Oncogene 2017;36(11):1461-1473 View Article PubMed/NCBI
  48. VanderVorst K, Dreyer CA, Konopelski SE, Lee H, Ho HH, Carraway KL. Wnt/PCP Signaling Contribution to Carcinoma Collective Cell Migration and Metastasis. Cancer Res 2019;79(8):1719-1729 View Article PubMed/NCBI
  49. Pataki CA, Couchman JR, Brábek J. Wnt Signaling Cascades and the Roles of Syndecan Proteoglycans. J Histochem Cytochem 2015;63(7):465-480 View Article PubMed/NCBI
  50. Farb MG, Karki S, Park SY, Saggese SM, Carmine B, Hess DT, et al. WNT5A-JNK regulation of vascular insulin resistance in human obesity. Vasc Med 2016;21(6):489-496 View Article PubMed/NCBI
  51. Zhu Y, Li X. Advances of Wnt Signalling Pathway in Colorectal Cancer. Cells 2023;12(3):447 View Article PubMed/NCBI
  52. Molkentin JD. Calcineurin-NFAT signaling regulates the cardiac hypertrophic response in coordination with the MAPKs. Cardiovasc Res 2004;63(3):467-475 View Article PubMed/NCBI
  53. Lang CMR, Chan CK, Veltri A, Lien WH. Wnt Signaling Pathways in Keratinocyte Carcinomas. Cancers (Basel) 2019;11(9):E1216 View Article PubMed/NCBI
  54. De A. Wnt/Ca2+ signaling pathway: a brief overview. Acta Biochim Biophys Sin (Shanghai) 2011;43(10):745-756 View Article PubMed/NCBI
  55. Chen Y, Chen Z, Tang Y, Xiao Q. The involvement of noncanonical Wnt signaling in cancers. Biomed Pharmacother 2021;133:110946 View Article PubMed/NCBI
  56. Wang W, Smits R, Hao H, He C. Wnt/β-Catenin Signaling in Liver Cancers. Cancers (Basel) 2019;11(7):926 View Article PubMed/NCBI
  57. Khalaf AM, Fuentes D, Morshid AI, Burke MR, Kaseb AO, Hassan M, et al. Role of Wnt/β-catenin signaling in hepatocellular carcinoma, pathogenesis, and clinical significance. J Hepatocell Carcinoma 2018;5:61-73 View Article PubMed/NCBI
  58. Lin J, Lu W, Huang B, Yang W, Wang X. The role of tissue-derived extracellular vesicles in tumor microenvironment. Tissue Cell 2024;89:102470 View Article PubMed/NCBI
  59. Carmeliet P, Dor Y, Herbert JM, Fukumura D, Brusselmans K, Dewerchin M, et al. Role of HIF-1alpha in hypoxia-mediated apoptosis, cell proliferation and tumour angiogenesis. Nature 1998;394(6692):485-490 View Article PubMed/NCBI
  60. Sun X, Hu F, Hou Z, Chen Q, Lan J, Luo X, et al. SIX4 activates Akt and promotes tumor angiogenesis. Exp Cell Res 2019;383(1):111495 View Article PubMed/NCBI
  61. Monga SP. β-Catenin Signaling and Roles in Liver Homeostasis, Injury, and Tumorigenesis. Gastroenterology 2015;148(7):1294-1310 View Article PubMed/NCBI
  62. Gougelet A, Torre C, Veber P, Sartor C, Bachelot L, Denechaud PD, et al. T-cell factor 4 and β-catenin chromatin occupancies pattern zonal liver metabolism in mice. Hepatology 2014;59(6):2344-2357 View Article PubMed/NCBI
  63. Gougelet A, Colnot S. A Complex Interplay between Wnt/β-Catenin Signalling and the Cell Cycle in the Adult Liver. Int J Hepatol 2012;2012:816125 View Article PubMed/NCBI
  64. Sanceau J, Poupel L, Joubel C, Lagoutte I, Caruso S, Pinto S, et al. DLK1/DIO3 locus upregulation by a β-catenin-dependent enhancer drives cell proliferation and liver tumorigenesis. Mol Ther 2024;32(4):1125-1143 View Article PubMed/NCBI
  65. Ning X, Shi Z, Liu X, Zhang A, Han L, Jiang K, et al. DNMT1 and EZH2 mediated methylation silences the microRNA-200b/a/429 gene and promotes tumor progression. Cancer Lett 2015;359(2):198-205 View Article PubMed/NCBI
  66. Elmileik H, Paterson AC, Kew MC. Beta-catenin mutations and expression, 249serine p53 tumor suppressor gene mutation, and hepatitis B virus infection in southern African Blacks with hepatocellular carcinoma. J Surg Oncol 2005;91(4):258-263 View Article PubMed/NCBI
  67. Schulze K, Imbeaud S, Letouzé E, Alexandrov LB, Calderaro J, Rebouissou S, et al. Exome sequencing of hepatocellular carcinomas identifies new mutational signatures and potential therapeutic targets. Nat Genet 2015;47(5):505-511 View Article PubMed/NCBI
  68. Colnot S, Decaens T, Niwa-Kawakita M, Godard C, Hamard G, Kahn A, et al. Liver-targeted disruption of Apc in mice activates beta-catenin signaling and leads to hepatocellular carcinomas. Proc Natl Acad Sci U S A 2004;101(49):17216-17221 View Article PubMed/NCBI
  69. Taniguchi K, Roberts LR, Aderca IN, Dong X, Qian C, Murphy LM, et al. Mutational spectrum of beta-catenin, AXIN1, and AXIN2 in hepatocellular carcinomas and hepatoblastomas. Oncogene 2002;21(31):4863-4871 View Article PubMed/NCBI
  70. Zhang XZ, Mo XC, Wang ZT, Sun R, Sun DQ. N-glycosylation of Wnt3 regulates the progression of hepatocellular carcinoma by affecting Wnt/β-catenin signal pathway. World J Gastrointest Oncol 2024;16(6):2769-2780 View Article PubMed/NCBI
  71. Jones SE, Jomary C. Secreted Frizzled-related proteins: searching for relationships and patterns. Bioessays 2002;24(9):811-820 View Article PubMed/NCBI
  72. Liu HY, Sun XJ, Xiu SY, Zhang XY, Wang ZQ, Gu YL, et al. Frizzled receptors (FZDs) in Wnt signaling: potential therapeutic targets for human cancers. Acta Pharmacol Sin 2024;45(8):1556-1570 View Article PubMed/NCBI
  73. Takagi H, Sasaki S, Suzuki H, Toyota M, Maruyama R, Nojima M, et al. Frequent epigenetic inactivation of SFRP genes in hepatocellular carcinoma. J Gastroenterol 2008;43(5):378-389 View Article PubMed/NCBI
  74. He S, Tang S. WNT/β-catenin signaling in the development of liver cancers. Biomed Pharmacother 2020;132:110851 View Article PubMed/NCBI
  75. Li C, Furth EE, Rustgi AK, Klein PS. When You Come to a Fork in the Road, Take It: Wnt Signaling Activates Multiple Pathways through the APC/Axin/GSK-3 Complex. Cells 2023;12(18):2256 View Article PubMed/NCBI
  76. Lu G, Lin J, Song G, Chen M. Prognostic significance of CTNNB1 mutation in hepatocellular carcinoma: a systematic review and meta-analysis. Aging (Albany NY) 2023;15(18):9759-9778 View Article PubMed/NCBI
  77. Jung YS, Stratton SA, Lee SH, Kim MJ, Jun S, Zhang J, et al. TMEM9-v-ATPase Activates Wnt/β-Catenin Signaling Via APC Lysosomal Degradation for Liver Regeneration and Tumorigenesis. Hepatology 2021;73(2):776-794 View Article PubMed/NCBI
  78. Zhang C, Li J, Huang T, Duan S, Dai D, Jiang D, et al. Meta-analysis of DNA methylation biomarkers in hepatocellular carcinoma. Oncotarget 2016;7(49):81255-81267 View Article PubMed/NCBI
  79. Chen J, Gingold JA, Su X. Immunomodulatory TGF-β Signaling in Hepatocellular Carcinoma. Trends Mol Med 2019;25(11):1010-1023 View Article PubMed/NCBI
  80. Zhu W, Qin L. GOLM1-regulated EGFR/RTK recycling is a novel target for combating HCC metastasis. Sci China Life Sci 2017;60(1):98-101 View Article PubMed/NCBI
  81. Timperi E, Barnaba V. Viral Hepatitides, Inflammation and Tumour Microenvironment. Adv Exp Med Biol 2020;1263:25-43 View Article PubMed/NCBI
  82. da Motta Girardi D, Correa TS, Crosara Teixeira M, Dos Santos Fernandes G. Hepatocellular Carcinoma: Review of Targeted and Immune Therapies. J Gastrointest Cancer 2018;49(3):227-236 View Article PubMed/NCBI
  83. Tigue ML, Loberg MA, Goettel JA, Weiss WA, Lee E, Weiss VL. Wnt Signaling in the Phenotype and Function of Tumor-Associated Macrophages. Cancer Res 2023;83(1):3-11 View Article PubMed/NCBI
  84. Aoki T, Nishida N, Kurebayashi Y, Sakai K, Morita M, Chishina H, et al. Two Distinct Characteristics of Immune Microenvironment in Human Hepatocellular Carcinoma with Wnt/β-Catenin Mutations. Liver Cancer 2024;13(3):285-305 View Article PubMed/NCBI
  85. Anna CH, Iida M, Sills RC, Devereux TR. Expression of potential beta-catenin targets, cyclin D1, c-Jun, c-Myc, E-cadherin, and EGFR in chemically induced hepatocellular neoplasms from B6C3F1 mice. Toxicol Appl Pharmacol 2003;190(2):135-145 View Article PubMed/NCBI
  86. Doumpas N, Lampart F, Robinson MD, Lentini A, Nestor CE, Cantù C, et al. TCF/LEF dependent and independent transcriptional regulation of Wnt/β-catenin target genes. EMBO J 2019;38(2):e98873 View Article PubMed/NCBI
  87. Davidson G. The cell cycle and Wnt. Cell Cycle 2010;9(9):1667-1668 View Article PubMed/NCBI
  88. Zhang JH, Jiao LY, Li TJ, Zhu YY, Zhou JW, Tian J. GSK-3β suppresses HCC cell dissociation in vitro by upregulating epithelial junction proteins and inhibiting Wnt/β-catenin signaling pathway. J Cancer 2017;8(9):1598-1608 View Article PubMed/NCBI
  89. Lu FI, Sun YH, Wei CY, Thisse C, Thisse B. Tissue-specific derepression of TCF/LEF controls the activity of the Wnt/β-catenin pathway. Nat Commun 2014;5:5368 View Article PubMed/NCBI
  90. MacDonald BT, He X. Frizzled and LRP5/6 receptors for Wnt/β-catenin signaling. Cold Spring Harb Perspect Biol 2012;4(12):a007880 View Article PubMed/NCBI
  91. Biswas P, Canosa S, Schoenfeld D, Schoenfeld J, Li P, Cheas LC, et al. PECAM-1 affects GSK-3beta-mediated beta-catenin phosphorylation and degradation. Am J Pathol 2006;169(1):314-324 View Article PubMed/NCBI
  92. Lin J, Song T, Li C, Mao W. GSK-3β in DNA repair, apoptosis, and resistance of chemotherapy, radiotherapy of cancer. Biochim Biophys Acta Mol Cell Res 2020;1867(5):118659 View Article PubMed/NCBI
  93. Kim W, Kim M, Jho EH. Wnt/β-catenin signalling: from plasma membrane to nucleus. Biochem J 2013;450(1):9-21 View Article PubMed/NCBI
  94. Novak A, Dedhar S. Signaling through beta-catenin and Lef/Tcf. Cell Mol Life Sci 1999;56(5-6):523-537 View Article PubMed/NCBI
  95. Hume S, Dianov GL, Ramadan K. A unified model for the G1/S cell cycle transition. Nucleic Acids Res 2020;48(22):12483-12501 View Article PubMed/NCBI
  96. Bertoli C, Skotheim JM, de Bruin RA. Control of cell cycle transcription during G1 and S phases. Nat Rev Mol Cell Biol 2013;14(8):518-528 View Article PubMed/NCBI
  97. Huang M, Wang Y, Sun D, Zhu H, Yin Y, Zhang W, et al. Identification of genes regulated by Wnt/beta-catenin pathway and involved in apoptosis via microarray analysis. BMC Cancer 2006;6:221 View Article PubMed/NCBI
  98. Matsumoto S, Kikuchi A. Wnt/β-catenin signaling pathway in liver biology and tumorigenesis. In Vitro Cell Dev Biol Anim 2024;60(5):466-481 View Article PubMed/NCBI
  99. So J, Martin BL, Kimelman D, Shin D. Wnt/β-catenin signaling cell-autonomously converts non-hepatic endodermal cells to a liver fate. Biol Open 2013;2(1):30-36 View Article PubMed/NCBI
  100. Castelli G, Pelosi E, Testa U. Liver Cancer: Molecular Characterization, Clonal Evolution and Cancer Stem Cells. Cancers (Basel) 2017;9(9):E127 View Article PubMed/NCBI
  101. Kahn M. Wnt Signaling in Stem Cells and Cancer Stem Cells: A Tale of Two Coactivators. Prog Mol Biol Transl Sci 2018;153:209-244 View Article PubMed/NCBI
  102. Ji J, Wang XW. Clinical implications of cancer stem cell biology in hepatocellular carcinoma. Semin Oncol 2012;39(4):461-472 View Article PubMed/NCBI
  103. Yan H, Li Z, Shen Q, Wang Q, Tian J, Jiang Q, et al. Aberrant expression of cell cycle and material metabolism related genes contributes to hepatocellular carcinoma occurrence. Pathol Res Pract 2017;213(4):316-321 View Article PubMed/NCBI
  104. Karimaian A, Majidinia M, Bannazadeh Baghi H, Yousefi B. The crosstalk between Wnt/β-catenin signaling pathway with DNA damage response and oxidative stress: Implications in cancer therapy. DNA Repair (Amst) 2017;51:14-19 View Article PubMed/NCBI
  105. Jin Y, Anbarchian T, Wu P, Sarkar A, Fish M, Peng WC, et al. Wnt signaling regulates hepatocyte cell division by a transcriptional repressor cascade. Proc Natl Acad Sci U S A 2022;119(30):e2203849119 View Article PubMed/NCBI
  106. Xue W, Yang L, Chen C, Ashrafizadeh M, Tian Y, Sun R. Wnt/β-catenin-driven EMT regulation in human cancers. Cell Mol Life Sci 2024;81(1):79 View Article PubMed/NCBI
  107. Mittal V. Epithelial Mesenchymal Transition in Tumor Metastasis. Annu Rev Pathol 2018;13:395-412 View Article PubMed/NCBI
  108. Paolillo M, Schinelli S. Extracellular Matrix Alterations in Metastatic Processes. Int J Mol Sci 2019;20(19):4947 View Article PubMed/NCBI
  109. Wei Y, Van Nhieu JT, Prigent S, Srivatanakul P, Tiollais P, Buendia MA. Altered expression of E-cadherin in hepatocellular carcinoma: correlations with genetic alterations, beta-catenin expression, and clinical features. Hepatology 2002;36(3):692-701 View Article PubMed/NCBI
  110. Loh CY, Chai JY, Tang TF, Wong WF, Sethi G, Shanmugam MK, et al. The E-Cadherin and N-Cadherin Switch in Epithelial-to-Mesenchymal Transition: Signaling, Therapeutic Implications, and Challenges. Cells 2019;8(10):1118 View Article PubMed/NCBI
  111. Zhai X, Zhu H, Wang W, Zhang S, Zhang Y, Mao G. Abnormal expression of EMT-related proteins, S100A4, vimentin and E-cadherin, is correlated with clinicopathological features and prognosis in HCC. Med Oncol 2014;31(6):970 View Article PubMed/NCBI
  112. Giannelli G, Koudelkova P, Dituri F, Mikulits W. Role of epithelial to mesenchymal transition in hepatocellular carcinoma. J Hepatol 2016;65(4):798-808 View Article PubMed/NCBI
  113. Li LY, Yang JF, Rong F, Luo ZP, Hu S, Fang H, et al. ZEB1 serves an oncogenic role in the tumourigenesis of HCC by promoting cell proliferation, migration, and inhibiting apoptosis via Wnt/β-catenin signaling pathway. Acta Pharmacol Sin 2021;42(10):1676-1689 View Article PubMed/NCBI
  114. Li W, Wu R, Zhang S, Zhao Y, Li L, Hu Z, et al. Analysis of angiogenesis-related subtypes of hepatocellular carcinoma and tumor microenvironment infiltration feature in hepatocellular carcinoma. Clin Transl Oncol 2023;25(7):2099-2115 View Article PubMed/NCBI
  115. Neth P, Ries C, Karow M, Egea V, Ilmer M, Jochum M. The Wnt signal transduction pathway in stem cells and cancer cells: influence on cellular invasion. Stem Cell Rev 2007;3(1):18-29 View Article PubMed/NCBI
  116. Astudillo P, Larraín J. Wnt signaling and cell-matrix adhesion. Curr Mol Med 2014;14(2):209-220 View Article PubMed/NCBI
  117. Tejeda-Muñoz N, Morselli M, Moriyama Y, Sheladiya P, Pellegrini M, De Robertis EM. Canonical Wnt signaling induces focal adhesion and Integrin beta-1 endocytosis. iScience 2022;25(4):104123 View Article PubMed/NCBI
  118. Schambony A, Kunz M, Gradl D. Cross-regulation of Wnt signaling and cell adhesion. Differentiation 2004;72(7):307-318 View Article PubMed/NCBI
  119. Wang K, Qiu X, Zhao Y, Wang H, Chen L. The Wnt/β-catenin signaling pathway in the tumor microenvironment of hepatocellular carcinoma. Cancer Biol Med 2021;19(3):305-318 View Article PubMed/NCBI
  120. Moeini A, Cornellà H, Villanueva A. Emerging signaling pathways in hepatocellular carcinoma. Liver Cancer 2012;1(2):83-93 View Article PubMed/NCBI
  121. Liu DX, Hao SL, Yang WX. Crosstalk Between β-CATENIN-Mediated Cell Adhesion and the WNT Signaling Pathway. DNA Cell Biol 2023;42(1):1-13 View Article PubMed/NCBI
  122. Zelikson N, Ben S, Caspi M, Tarabe R, Shaleve Y, Pri-Paz Basson Y, et al. Wnt signaling regulates chemokine production and cell migration of circulating human monocytes. Cell Commun Signal 2024;22(1):229 View Article PubMed/NCBI
  123. Qu B, Liu BR, DU YJ, Chen J, Cheng YQ, Xu W, et al. Wnt/β-catenin signaling pathway may regulate the expression of angiogenic growth factors in hepatocellular carcinoma. Oncol Lett 2014;7(4):1175-1178 View Article PubMed/NCBI
  124. Yang C, Zhang H, Zhang L, Zhu AX, Bernards R, Qin W, et al. Evolving therapeutic landscape of advanced hepatocellular carcinoma. Nat Rev Gastroenterol Hepatol 2023;20(4):203-222 View Article PubMed/NCBI
  125. Dahmani R, Just PA, Perret C. The Wnt/β-catenin pathway as a therapeutic target in human hepatocellular carcinoma. Clin Res Hepatol Gastroenterol 2011;35(11):709-713 View Article PubMed/NCBI
  126. Liu J, Pan S, Hsieh MH, Ng N, Sun F, Wang T, et al. Targeting Wnt-driven cancer through the inhibition of Porcupine by LGK974. Proc Natl Acad Sci U S A 2013;110(50):20224-20229 View Article PubMed/NCBI
  127. Gabata R, Harada K, Mizutani Y, Ouchi H, Yoshimura K, Sato Y, et al. Anti-tumor Activity of the Small Molecule Inhibitor PRI-724 Against β-Catenin-activated Hepatocellular Carcinoma. Anticancer Res 2020;40(9):5211-5219 View Article PubMed/NCBI
  128. Huang Y, Sheng H, Xiao Y, Hu W, Zhang Z, Chen Y, et al. Wnt/β-catenin inhibitor ICG-001 enhances the antitumor efficacy of radiotherapy by increasing radiation-induced DNA damage and improving tumor immune microenvironment in hepatocellular carcinoma. Radiother Oncol 2021;162:34-44 View Article PubMed/NCBI
  129. Gedaly R, Galuppo R, Daily MF, Shah M, Maynard E, Chen C, et al. Targeting the Wnt/β-catenin signaling pathway in liver cancer stem cells and hepatocellular carcinoma cell lines with FH535. PLoS One 2014;9(6):e99272 View Article PubMed/NCBI
  130. Ma L, Wang X, Jia T, Wei W, Chua MS, So S. Tankyrase inhibitors attenuate WNT/β-catenin signaling and inhibit growth of hepatocellular carcinoma cells. Oncotarget 2015;6(28):25390-25401 View Article PubMed/NCBI
  131. Fischer MM, Cancilla B, Yeung VP, Cattaruzza F, Chartier C, Murriel CL, et al. WNT antagonists exhibit unique combinatorial antitumor activity with taxanes by potentiating mitotic cell death. Sci Adv 2017;3(6):e1700090 View Article PubMed/NCBI
  132. Chen L, Li M, Li Q, Wang CJ, Xie SQ. DKK1 promotes hepatocellular carcinoma cell migration and invasion through β-catenin/MMP7 signaling pathway. Mol Cancer 2013;12:157 View Article PubMed/NCBI
  133. Zeyada MS, Abdel-Rahman N, El-Karef A, Yahia S, El-Sherbiny IM, Eissa LA. Niclosamide-loaded polymeric micelles ameliorate hepatocellular carcinoma in vivo through targeting Wnt and Notch pathways. Life Sci 2020;261:118458 View Article PubMed/NCBI
  134. Wang F, He L, Dai WQ, Xu YP, Wu D, Lin CL, et al. Salinomycin inhibits proliferation and induces apoptosis of human hepatocellular carcinoma cells in vitro and in vivo. PLoS One 2012;7(12):e50638 View Article PubMed/NCBI
  135. Harb J, Lin PJ, Hao J. Recent Development of Wnt Signaling Pathway Inhibitors for Cancer Therapeutics. Curr Oncol Rep 2019;21(2):12 View Article PubMed/NCBI
  136. Zhang X, Dong N, Hu X. Wnt/β-catenin Signaling Inhibitors. Curr Top Med Chem 2023;23(10):880-896 View Article PubMed/NCBI
  137. Emma MR, Augello G, Cusimano A, Azzolina A, Montalto G, McCubrey JA, et al. GSK-3 in liver diseases: Friend or foe?. Biochim Biophys Acta Mol Cell Res 2020;1867(9):118743 View Article PubMed/NCBI
  138. Katoh M. Multi-layered prevention and treatment of chronic inflammation, organ fibrosis and cancer associated with canonical WNT/β-catenin signaling activation (Review). Int J Mol Med 2018;42(2):713-725 View Article PubMed/NCBI
  139. Lee SC, Kim OH, Lee SK, Kim SJ. IWR-1 inhibits epithelial-mesenchymal transition of colorectal cancer cells through suppressing Wnt/β-catenin signaling as well as survivin expression. Oncotarget 2015;6(29):27146-27159 View Article PubMed/NCBI
  140. Tan Y, Lin XT, Luo YD, Zhang J, Fang L, Zhu YY, et al. Reduced IκBα promotes hepatocellular carcinoma cell proliferation and migration via regulation of NF-κB/Erbin axis. Oncol Lett 2020;20(5):216 View Article PubMed/NCBI
  141. Rothwarf DM, Karin M. The NF-kappa B activation pathway: a paradigm in information transfer from membrane to nucleus. Sci STKE 1999;1999(5):RE1 View Article PubMed/NCBI
  142. Wu MY, Wang CC, Chang YC, Yu CY, Sung WW, Chen CJ, et al. The Therapeutic Role of PNU-74654 in Hepatocellular Carcinoma May Involve Suppression of NF-κB Signaling. Medicina (Kaunas) 2022;58(6):798 View Article PubMed/NCBI
  143. Hwang SY, Deng X, Byun S, Lee C, Lee SJ, Suh H, et al. Direct Targeting of β-Catenin by a Small Molecule Stimulates Proteasomal Degradation and Suppresses Oncogenic Wnt/β-Catenin Signaling. Cell Rep 2016;16(1):28-36 View Article PubMed/NCBI
  144. Pećina-Šlaus N, Aničić S, Bukovac A, Kafka A. Wnt Signaling Inhibitors and Their Promising Role in Tumor Treatment. Int J Mol Sci 2023;24(7):6733 View Article PubMed/NCBI
  145. Lee HC, Kim M, Wands JR. Wnt/Frizzled signaling in hepatocellular carcinoma. Front Biosci 2006;11:1901-1915 View Article PubMed/NCBI
  146. Meng Z, Liu Q, Liu Y, Yang Y, Shao C, Zhang S. Frizzled-3 suppression overcomes multidrug chemoresistance by Wnt/β-catenin signaling pathway inhibition in hepatocellular carcinoma cells. J Chemother 2023;35(7):653-661 View Article PubMed/NCBI
  147. Koelman EMR, Yeste-Vázquez A, Grossmann TN. Targeting the interaction of β-catenin and TCF/LEF transcription factors to inhibit oncogenic Wnt signaling. Bioorg Med Chem 2022;70:116920 View Article PubMed/NCBI
  148. Sheng J, Kohno S, Okada N, Okahashi N, Teranishi K, Matsuda F, et al. Treatment of Retinoblastoma 1-Intact Hepatocellular Carcinoma With Cyclin-Dependent Kinase 4/6 Inhibitor Combination Therapy. Hepatology 2021;74(4):1971-1993 View Article PubMed/NCBI
  149. Dropmann A, Alex S, Schorn K, Tong C, Caccamo T, Godoy P, et al. The TGF-β1 target WISP1 is highly expressed in liver cirrhosis and cirrhotic HCC microenvironment and involved in pro- and anti-tumorigenic effects. Biochem Biophys Res Commun 2024;732:150409 View Article PubMed/NCBI
  150. Naim A, Pan Q, Baig MS. Matrix Metalloproteinases (MMPs) in Liver Diseases. J Clin Exp Hepatol 2017;7(4):367-372 View Article PubMed/NCBI
  151. Scheau C, Badarau IA, Costache R, Caruntu C, Mihai GL, Didilescu AC, et al. The Role of Matrix Metalloproteinases in the Epithelial-Mesenchymal Transition of Hepatocellular Carcinoma. Anal Cell Pathol (Amst) 2019;2019:9423907 View Article PubMed/NCBI
  152. Wojtowicz-Praga SM, Dickson RB, Hawkins MJ. Matrix metalloproteinase inhibitors. Invest New Drugs 1997;15(1):61-75 View Article PubMed/NCBI
  153. Xiao L, Wang M. Batimastat nanoparticles associated with transcatheter arterial chemoembolization decrease hepatocellular carcinoma recurrence. Cell Biochem Biophys 2014;70(1):269-272 View Article PubMed/NCBI
  154. Heath EI, O’Reilly S, Humphrey R, Sundaresan P, Donehower RC, Sartorius S, et al. Phase I trial of the matrix metalloproteinase inhibitor BAY12-9566 in patients with advanced solid tumors. Cancer Chemother Pharmacol 2001;48(4):269-274 View Article PubMed/NCBI
  155. Lv Y, Zhao X, Zhu L, Li S, Xiao Q, He W, et al. Targeting intracellular MMPs efficiently inhibits tumor metastasis and angiogenesis. Theranostics 2018;8(10):2830-2845 View Article PubMed/NCBI
  156. Belotti D, Paganoni P, Giavazzi R. MMP inhibitors: experimental and clinical studies. Int J Biol Markers 1999;14(4):232-238 View Article PubMed/NCBI
  157. Stevens AJ, Harris AR, Gerdts J, Kim KH, Trentesaux C, Ramirez JT, et al. Programming multicellular assembly with synthetic cell adhesion molecules. Nature 2023;614(7946):144-152 View Article PubMed/NCBI
  158. Giannelli G, Bergamini C, Fransvea E, Marinosci F, Quaranta V, Antonaci S. Human hepatocellular carcinoma (HCC) cells require both alpha3beta1 integrin and matrix metalloproteinases activity for migration and invasion. Lab Invest 2001;81(4):613-627 View Article PubMed/NCBI
  159. Monvoisin A, Bisson C, Si-Tayeb K, Balabaud C, Desmoulière A, Rosenbaum J. Involvement of matrix metalloproteinase type-3 in hepatocyte growth factor-induced invasion of human hepatocellular carcinoma cells. Int J Cancer 2002;97(2):157-162 View Article PubMed/NCBI
  160. Erlichman C, Adjei AA, Alberts SR, Sloan JA, Goldberg RM, Pitot HC, et al. Phase I study of the matrix metalloproteinase inhibitor, BAY 12-9566. Ann Oncol 2001;12(3):389-395 View Article PubMed/NCBI
  161. Giannelli G, Bergamini C, Marinosci F, Fransvea E, Quaranta M, Lupo L, et al. Clinical role of MMP-2/TIMP-2 imbalance in hepatocellular carcinoma. Int J Cancer 2002;97(4):425-431 View Article PubMed/NCBI
  162. Meng J, Sun B, Zhao X, Zhang D, Zhao X, Gu Q, et al. Doxycycline as an inhibitor of the epithelial-to-mesenchymal transition and vasculogenic mimicry in hepatocellular carcinoma. Mol Cancer Ther 2014;13(12):3107-3122 View Article PubMed/NCBI
  163. Lee KW, Kim MS, Kang NJ, Kim DH, Surh YJ, Lee HJ, et al. H-Ras selectively up-regulates MMP-9 and COX-2 through activation of ERK1/2 and NF-kappaB: an implication for invasive phenotype in rat liver epithelial cells. Int J Cancer 2006;119(8):1767-1775 View Article PubMed/NCBI
  164. Shalinsky DR, Brekken J, Zou H, McDermott CD, Forsyth P, Edwards D, et al. Broad antitumor and antiangiogenic activities of AG3340, a potent and selective MMP inhibitor undergoing advanced oncology clinical trials. Ann N Y Acad Sci 1999;878:236-270 View Article PubMed/NCBI
  165. Sawada S, Murakami K, Yamaura T, Mitani N, Tsukada K, Saiki I. Therapeutic and analysis model of intrahepatic metastasis reflects clinical behavior of hepatocellular carcinoma. Jpn J Cancer Res 2002;93(2):190-197 View Article PubMed/NCBI
  166. Liu FY, Wu YH, Zhou SJ, Deng YL, Zhang ZY, Zhang EL, et al. Minocycline and cisplatin exert synergistic growth suppression on hepatocellular carcinoma by inducing S phase arrest and apoptosis. Oncol Rep 2014;32(2):835-844 View Article PubMed/NCBI
  167. Mita M, Kelly KR, Mita A, Ricart AD, Romero O, Tolcher A, et al. Phase I study of E7820, an oral inhibitor of integrin alpha-2 expression with antiangiogenic properties, in patients with advanced malignancies. Clin Cancer Res 2011;17(1):193-200 View Article PubMed/NCBI
  168. Ramakrishnan V, Bhaskar V, Law DA, Wong MH, DuBridge RB, Breinberg D, et al. Preclinical evaluation of an anti-alpha5beta1 integrin antibody as a novel anti-angiogenic agent. J Exp Ther Oncol 2006;5(4):273-286 PubMed/NCBI
  169. Delbaldo C, Raymond E, Vera K, Hammershaimb L, Kaucic K, Lozahic S, et al. Phase I and pharmacokinetic study of etaracizumab (Abegrin), a humanized monoclonal antibody against alphavbeta3 integrin receptor, in patients with advanced solid tumors. Invest New Drugs 2008;26(1):35-43 View Article PubMed/NCBI
  170. Cai J, Wang J, Jiang C, Ye L, He X, Huang J, et al. Combined inhibition of surface CD51 and γ-secretase-mediated CD51 cleavage improves therapeutic efficacy in experimental metastatic hepatocellular carcinoma. J Hepatol 2023;79(6):1418-1434 View Article PubMed/NCBI
  171. Chen W, Yang A, Jia J, Popov YV, Schuppan D, You H. Lysyl Oxidase (LOX) Family Members: Rationale and Their Potential as Therapeutic Targets for Liver Fibrosis. Hepatology 2020;72(2):729-741 View Article PubMed/NCBI
  172. Goto K, Arai J, Stephanou A, Kato N. Novel therapeutic features of disulfiram against hepatocellular carcinoma cells with inhibitory effects on a disintegrin and metalloproteinase 10. Oncotarget 2018;9(27):18821-18831 View Article PubMed/NCBI
  173. Blaschuk OW. Potential Therapeutic Applications of N-Cadherin Antagonists and Agonists. Front Cell Dev Biol 2022;10:866200 View Article PubMed/NCBI
  174. Kelley RK, Gane E, Assenat E, Siebler J, Galle PR, Merle P, et al. A Phase 2 Study of Galunisertib (TGF-β1 Receptor Type I Inhibitor) and Sorafenib in Patients With Advanced Hepatocellular Carcinoma. Clin Transl Gastroenterol 2019;10(7):e00056 View Article PubMed/NCBI
  175. Wang K, Zhang K, Zhang X, Chen D, Jiang S. Recent Insights of Metformin on Hepatocellular Carcinoma (HCC). Mini Rev Med Chem 2023;23(11):1154-1166 View Article PubMed/NCBI
  176. Fan X, Jin S, Li Y, Khadaroo PA, Dai Y, He L, et al. Genetic And Epigenetic Regulation Of E-Cadherin Signaling In Human Hepatocellular Carcinoma. Cancer Manag Res 2019;11:8947-8963 View Article PubMed/NCBI
  177. Xu F, Liu C, Zhou D, Zhang L. TGF-β/SMAD Pathway and Its Regulation in Hepatic Fibrosis. J Histochem Cytochem 2016;64(3):157-167 View Article PubMed/NCBI
  178. Ceballos MP, Parody JP, Alvarez Mde L, Ingaramo PI, Carnovale CE, Carrillo MC. Interferon-α2b and transforming growth factor-β1 treatments on HCC cell lines: Are Wnt/β-catenin pathway and Smads signaling connected in hepatocellular carcinoma?. Biochem Pharmacol 2011;82(11):1682-1691 View Article PubMed/NCBI
  179. Choucair K, Kamran S, Saeed A. Clinical Evaluation of Ramucirumab for the Treatment of Hepatocellular Carcinoma (HCC): Place in Therapy. Onco Targets Ther 2021;14:5521-5532 View Article PubMed/NCBI
  180. Kuwano A, Yada M, Narutomi F, Nagasawa S, Tanaka K, Kurosaka K, et al. Therapeutic efficacy of atezolizumab plus bevacizumab for hepatocellular carcinoma with WNT/β-catenin signal activation. Oncol Lett 2022;24(1):216 View Article PubMed/NCBI
  181. Huether A, Höpfner M, Baradari V, Schuppan D, Scherübl H. EGFR blockade by cetuximab alone or as combination therapy for growth control of hepatocellular cancer. Biochem Pharmacol 2005;70(11):1568-1578 View Article PubMed/NCBI
  182. Harkus U, Wankell M, Palamuthusingam P, McFarlane C, Hebbard L. Immune checkpoint inhibitors in HCC: Cellular, molecular and systemic data. Semin Cancer Biol 2022;86(Pt 3):799-815 View Article PubMed/NCBI
  183. Donne R, Lujambio A. The liver cancer immune microenvironment: Therapeutic implications for hepatocellular carcinoma. Hepatology 2023;77(5):1773-1796 View Article PubMed/NCBI
  184. Bupathi M, Kaseb A, Meric-Bernstam F, Naing A. Hepatocellular carcinoma: Where there is unmet need. Mol Oncol 2015;9(8):1501-1509 View Article PubMed/NCBI
  185. Thuru X, Magnez R, Vergoten G, Bailly C. A Potential Off-Target Effect of the Wnt/β-Catenin Inhibitor KYA1797K: PD-L1 Binding and Checkpoint Inhibition. Biomed Hub 2023;8(1):1-9 View Article PubMed/NCBI
  186. Zhong Z, Virshup DM. Wnt Signaling and Drug Resistance in Cancer. Mol Pharmacol 2020;97(2):72-89 View Article PubMed/NCBI
  187. Tian M, Wang X, Sun J, Lin W, Chen L, Liu S, et al. IRF3 prevents colorectal tumorigenesis via inhibiting the nuclear translocation of β-catenin. Nat Commun 2020;11(1):5762 View Article PubMed/NCBI
  188. Katoh M. Canonical and non-canonical WNT signaling in cancer stem cells and their niches: Cellular heterogeneity, omics reprogramming, targeted therapy and tumor plasticity (Review). Int J Oncol 2017;51(5):1357-1369 View Article PubMed/NCBI

About this Article

Cite this article
Liang Z, Li S, Wang Z, Zhou J, Huang Z, Li J, et al. Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications. J Clin Transl Hepatol. Published online: Jan 14, 2025. doi: 10.14218/JCTH.2024.00401.
Copy        Export to RIS        Export to EndNote
Article History
Received Revised Accepted Published
October 29, 2024 December 13, 2024 December 23, 2024 January 14, 2025
DOI http://dx.doi.org/10.14218/JCTH.2024.00401
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Unraveling the Role of the Wnt Pathway in Hepatocellular Carcinoma: From Molecular Mechanisms to Therapeutic Implications

Zixin Liang, Shanshan Li, Zhiyu Wang, Junting Zhou, Ziyue Huang, Jiehan Li, Haolin Bao, Judy Wai Ping Yam, Yi Xu
  • Reset Zoom
  • Download TIFF