BPG is committed to discovery and dissemination of knowledge
Minireviews Open Access
Copyright ©The Author(s) 2025. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Hepatol. Sep 27, 2025; 17(9): 107671
Published online Sep 27, 2025. doi: 10.4254/wjh.v17.i9.107671
Targeting glypican-3 as a new frontier in liver cancer therapy
Chen-Shiou Wu, Department of Medical Research, Taichung Veterans General Hospital, Taichung 407219, Taiwan
Teng-Yu Lee, Division of Gastroenterology and Hepatology, Department of Internal Medicine, Taichung Veterans General Hospital, Taichung 407219, Taiwan
Teng-Yu Lee, School of Medicine, Chung Shan Medical University, Taichung 40201, Taiwan
Hsu-Wen Chao, Department of Physiology, School of Medicine, Taipei Medical University, Taipei 110301, Taiwan
Hsu-Wen Chao, Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei 110301, Taiwan
Hsu-Wen Chao, Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung 807378, Taiwan
ORCID number: Hsu-Wen Chao (0000-0002-8122-924X).
Author contributions: Wu CS wrote the original draft; Lee TY and Chao HW provided guidance and critically reviewed the manuscript. All authors read and approved the final manuscript.
Conflict-of-interest statement: The authors declare no conflict of interest related to this manuscript.
Open Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Hsu-Wen Chao, PhD, Associate Professor, Department of Physiology, School of Medicine, Taipei Medical University, Wuxing St, Xinyi District, Taipei 110301, Taiwan. chaohw3619@tmu.edu.tw
Received: March 28, 2025
Revised: May 23, 2025
Accepted: August 1, 2025
Published online: September 27, 2025
Processing time: 181 Days and 21.9 Hours

Abstract

Glypican-3 (GPC3) is a tumor-associated antigen that is specifically expressed in hepatocellular carcinoma (HCC) and having relatively low levels in normal tissues. This unique expression pattern positions GPC3 as a potential target for precision therapy and drug development in HCC. Recent studies have shown significant advancements in GPC3-targeted therapies and immunotherapies, particularly for patients with advanced or treatment-resistant HCC. Although certain clinical trials have yielded suboptimal results, numerous ongoing studies continue to explore its therapeutic efficacy. This mini-review focuses on the latest research developments regarding GPC3 as a therapeutic target across various HCC treatment strategies, including monoclonal antibodies, bispecific antibodies, chimeric antigen receptor-T-cell therapies, and other innovative approaches. In addition, the limitations of GPC3-targeted therapies and their future application prospects in HCC treatment are discussed. The review particularly emphasizes the unmet need for future research directions, such as combination immunotherapy strategies and novel drug designs. Through the integration of innovative technologies and clinical validation, GPC3 holds strong potential as a promising breakthrough in the treatment of HCC, offering new opportunities for enhancing patient outcomes and improving therapeutic efficacy.

Key Words: Glypican-3; Hepatocellular carcinoma; Chimeric antigen receptor T-cell; Wnt; Immunotherapy

Core Tip: This review provides a comprehensive overview of glypican-3 (GPC3)-targeted strategies in hepatocellular carcinoma (HCC), including monoclonal and bispecific antibodies, chimeric antigen receptor-T cell therapies, vaccines, and photodynamic approaches. It further explores GPC3’s role in molecular imaging, radiomics, and liquid biopsy. Despite challenges in clinical translation, ongoing trials and novel combination therapies highlight the potential of GPC3-based approaches to improve treatment specificity, overcome resistance, and guide personalized therapy in HCC.



INTRODUCTION

The glypican (GPC) family belongs to the heparan sulfate (HS) proteoglycan family and is characterized by its unique structural components, including a core protein, HS glycosaminoglycan chains, and a glycosylphosphatidylinositol anchor that tethers the protein to the cell surface[1]. Among the six members of this family (GPC1 to GPC6), glypican-3 (GPC3) stands out as a key molecular marker and therapeutic target in hepatocellular carcinoma (HCC), due to its tumor-specific overexpression and oncogenic functions. In normal adult liver, the expression level of GPC3 is extremely low[2]. Notably, GPC3 expression is markedly upregulated in HCC and melanoma[3], while it is significantly downregulated or completely silenced in several malignancies, including ovarian cancer[4], breast cancer[5], and clear cell renal cell carcinoma[6]. These contrasting expression patterns highlight the critical role of GPC3 in tumorigenesis, emphasizing its relevance as a potential target in cancer therapy.

GPC3 is not merely a biomarker but also an active driver of HCC progression, contributing to tumor proliferation, immune evasion, and poor prognosis. Its oncogenic effects, primarily through activation of the c-Myc signaling pathway, make it a crucial target for both diagnostic and therapeutic interventions. Given the high incidence and mortality rates seen in HCC patients, developing effective GPC3-targeted strategies could very well address the significantly unmet clinical need currently seen. In HCC, GPC3 is overexpressed in more than 70% of cases when compared to adjacent normal tissue, with this upregulation being strongly associated with adverse clinical outcomes[7]. Moreover, GPC3 promotes tumor progression primarily through activation of the c-Myc signaling pathway[8], establishing it as a key molecular marker for both diagnostic and therapeutic applications in HCC. Despite advancements in HCC treatment, current options such as surgical resection, liver transplantation, tyrosine kinase inhibitors (TKIs) and immune checkpoint inhibitors (ICIs) remain limited due to high recurrence rates and insufficient long-term efficacy[9,10]. Therefore, there remains an urgent need for novel molecularly targeted therapies to help improve patient survival. Unlike other biomarkers with overlapping expression in normal liver or non-malignant diseases, GPC3 demonstrates exceptional specificity for HCC, making it not only an ideal diagnostic marker but also an attractive therapeutic target. This unique expression profile has propelled the development of multiple GPC3-targeted therapies, all of which hold great promise for transforming HCC treatment.

GPC3 is primarily expressed on the surface of HCC cells but undergoes proteolytic cleavage by the enzyme Furin at Arg358 and Ser359, resulting in the release of soluble GPC3 (sGPC3) into the bloodstream[11]. Additionally, GPC3 has been detected in small extracellular vesicles (EVs) isolated from the serum of HCC patients, highlighting its potential as a blood-based biomarker for HCC diagnosis and prognosis[12]. Although sGPC3 alone has a sensitivity of only 40%, its high specificity for HCC makes it an indispensable component of multi-marker diagnostic panels, enhancing early detection and improving diagnostic accuracy when combined with other biomarkers[13]. While sGPC3 demonstrates high specificity for HCC, its presence has also been detected in other malignancies, including lung cancer (13.5%) and thyroid cancer (13.2%), further highlighting the importance of using it as part of a multi-marker diagnostic panel rather than as a single diagnostic indicator[14]. To overcome the limitations of single biomarkers, combining multiple serum markers has emerged as a promising strategy to enhance HCC detection and monitoring. Our studies highlighted the added value of combining Protein induced by vitamin K absence-II (PIVKA-II) with alpha-fetoprotein (AFP), particularly in AFP-negative patients, as well as its potential to predict recurrence[15]. Additionally, a highly sensitive glycan microarray has enabled more reliable detection of AFP-L3 in hepatitis B virus (HBV)-related HCC, even at low AFP levels[16]. These findings suggest the value of integrating novel detection platforms and complementary biomarkers, such as GPC3, lens culinaris agglutinin-reactive fraction of AFP (AFP-L3) and PIVKA-II, in order to develop more accurate and clinically applicable multi-marker diagnostic panels.

Importantly, GPC3-targeted therapies offer a new frontier in HCC treatment by leveraging its tumor-specific expression. Given the limitations of current HCC treatments, GPC3-targeted therapies represent a promising development in the field. Various strategies, including monoclonal antibodies, antibody-drug conjugates (ADCs) and chimeric antigen receptor (CAR)-T-cell therapies, are all in active development, aiming to harness GPC3’s tumor-specific expression for precise treatment. However, challenges such as optimizing delivery, overcoming tumor heterogeneity, and minimizing immune evasion must be addressed to fully realize GPC3-targeted therapy as a transformative clinical solution. These strategies aim to enhance antitumor efficacy while also minimizing off-target toxicity, addressing a major limitation to the current treatments. However, challenges remain in translating these therapies into clinical practice, including optimizing drug delivery, overcoming tumor heterogeneity, and mitigating resistance mechanisms. Further research is still needed in order to refine these approaches and better develop combinatorial strategies that will enhance treatment effectiveness.

This review highlights the pivotal role of GPC3 in HCC, from its fundamental oncogenic mechanisms to its emerging role as a critical biomarker and therapeutic target. As novel GPC3-based strategies advance, they hold the potential to reshape the current landscape of HCC diagnosis and treatment, addressing the urgent need for more effective therapeutic solutions. We have critically examined the molecular mechanisms underlying GPC3's biological functions and subsequently highlighted its significance as a therapeutic target in HCC diagnosis and treatment. Additionally, we analyzed the current challenges in translating GPC3-based therapies from bench to bedside so as to optimize their clinical implementation.

MOLECULAR AND BIOLOGICAL FUNCTIONS OF GPC3

GPC3 is essential for embryonic development and organogenesis. During embryogenesis, GPC3 regulates multiple regulatory pathways by modulating growth factor signaling cascades, including Wnt, fibroblast growth factor (FGF), bone morphogenetic protein (BMP), and insulin-like growth factor (IGF)[17-21] (Figure 1). These interactions influence downstream pathways involved in cellular proliferation, differentiation and tissue morphogenesis.

Figure 1
Figure 1 Diverse growth factor signaling pathways coordinated by glypican-3 in hepatocellular carcinoma. Glypican-3 (GPC3) plays a crucial role in hepatocellular carcinoma (HCC) by interacting with multiple ligands through modulation by GPC3-conjugated heparan sulfate. These ligands include Wnt, fibroblast growth factor 2 (FGF 2), hepatocyte growth factor (HGF), bone morphogenetic protein (BMP), and insulin-like growth factor (IGF). Through these interactions, GPC3 regulates key oncogenic signaling pathways, thereby contributing to tumor proliferation, migration, invasion, and angiogenesis. Specifically, FGF2, HGF, BMP, and IGF activate their respective receptors (FGFR, C-MET, BMPR, and IGF1R), triggering downstream signaling cascades such as AKT/mTOR, c-Myc, and SMAD pathways, which ultimately drive tumor progression. Moreover, GPC3 enhances Wnt signaling by facilitating Wnt ligand binding to Frizzled receptors, leading to the activation of the β-catenin and Hippo/YAP pathways, which further promote oncogenic signaling in HCC. GPC3: Glypican-3; HGF: Hepatocyte growth factor; IGF: Insulin-like growth factor.

Under physiological conditions, GPC3 is essential for the development of various organ systems. In liver development, it regulates hepatocyte proliferation and differentiation, ensuring proper liver formation[17,22]. In kidney development, GPC3 influences the migration and differentiation of nephron progenitor cells[19,23]. It also plays a critical role in skeletal and limb formation through BMP signaling[20] In the gastrointestinal tract, GPC3 contributes to both intestinal cell differentiation and tissue organization, while in the neural system, it is thought to affect brain and spinal cord development by regulating neural stem cell differentiation and migration[24,25]. Through these diverse functions, GPC3 serves as an essential regulator of embryonic tissue patterning and organogenesis.

In pathological conditions such as HCC, GPC3 is aberrantly re-expressed or overexpressed, serving as a critical regulator of multiple oncogenic signaling pathways. Through its core protein and HS side chains, GPC3 interacts with both ligands and receptors, functioning as a co-receptor and scaffold to facilitate signal complex formation, thereby activating downstream effectors such as c-Myc, AKT, and ERK that drive proliferation, survival, and metastasis in HCC cells.

Among the multiple oncogenic pathways regulated by GPC3, the Wnt/β-catenin pathway is one of the most frequently activated (Figure 2). GPC3 enhances Wnt signaling by directly binding to Wnt ligands and increasing their local concentration near receptors[26,27]. This process facilitates the assembly of Wnt-Frizzled (FZD)-LRP5/6 receptor complexes[28], involving both the GPC3 core protein and HS side chains for stabilizing Wnt-FZD interactions[29]. Upon activation, this pathway leads to β-catenin nuclear translocation and transcription of cell cycle-related genes like cyclin D1. Notably, mutations within the N-terminal Wnt-binding domain of GPC3 impair its Wnt-binding capacity and suppress tumor growth in murine HCC models[26], while sGPC3 acts as a decoy receptor that sequesters Wnt ligands to inhibit signal transmission[30].

Figure 2
Figure 2 Glypican-3-mediated Wnt/β-catenin and YAP/TAZ signaling axes in hepatocellular carcinoma. GPC3 acts as a central signaling molecule that regulates two key pathways in hepatocellular carcinoma. One is the canonical Wnt/β-catenin pathway, where activated β-catenin translocates to the nucleus and induces the expression of oncogenic genes. The other is the alternative Wnt–YAP/TAZ pathway, in which activated YAP/TAZ translocate to the nucleus and promote tumor progression through transcriptional regulation. GPC3: Glypican-3.

GPC3 also modulates the Hippo/YAP signaling cascade. Certain Wnt ligands, upon binding to FZD receptors, can bypass β-catenin and suppress LATS1/2 kinase activity, promoting YAP nuclear translocation and transcriptional activity[31,32]. The Hippo pathway, generally tumor-suppressive in liver tissue, is frequently inactivated during hepatocarcinogenesis[33]. In HCC, GPC3 knockdown reduces YAP activity, while monoclonal antibodies targeting GPC3 (e.g., HN3) further suppress YAP-mediated transcription and tumor cell proliferation[34]. Activated YAP can coactivate TEAD and TCF transcription factors with nuclear β-catenin, inducing oncogenesis-associated genes and mediating recruitment of M2-polarized tumor-associated macrophages, enhancing angiogenesis and immune evasion[35].

GPC3 regulates multiple growth factor pathways through its core protein and HS chains[36]. For example, GPC3 binds FGF2 through its HS chains to promote FGFR activation and downstream MAPK, PKC, and PI3K/AKT pathways[36], with SULF2 enhancing GPC3 expression and FGF signaling in HCC[37]. The HS chains of GPC3 regulate BMP signaling, which typically activates the intracellular SMAD pathway upon receptor binding, leading to nuclear translocation and transcriptional regulation[38]. GPC3 is frequently overexpressed in HCC and regulates both FGF2 and BMP-7 signaling pathways[39]. In the hepatocyte growth factor (HGF)/c-Met axis, GPC3 promotes cell motility and invasion through HS chain-mediated interactions, with antibodies targeting the HS domain of GPC3 (e.g., HS20) significantly inhibiting HGF-induced migration and 3D spheroid formation in HCC cells[40]. GPC3 also supports IGF pathway signaling by binding IGF-II and IGF-1R via a proline-rich N-terminal domain, promoting downstream MAPK/ERK and PI3K/AKT signaling that facilitates proliferation, metabolic reprogramming, and apoptosis resistance[21]. Additionally, GPC3 inhibits ubiquitination and degradation of IGF-1R by sequestering the adapter protein Grb10, sustaining prolonged receptor activation[41].

GPC3 in hypoxia and oncogenic signaling

Recent studies have shown that under hypoxic conditions, hypoxia-inducible factor 1-alpha upregulates GPC3 mRNA expression and promotes its secretion via exosomes in HCC cells[42] (Figure 3). These GPC3-enriched exosomes have been shown to enhance HCC cell proliferation, migration, and epithelial-mesenchymal transition (EMT), primarily through activation of the β-catenin signaling cascade[42]. This activation is accompanied by upregulation of β-catenin and c-Myc, and downregulation of GSK-3β. Moreover, exosomal GPC3 facilitates angiogenic activity in HUVECs, indicating its role in remodeling the tumor microenvironment[42]. Notably, GPC3 knockdown attenuates these tumor-promoting effects, suggesting that targeting exosomal GPC3 may offer a promising therapeutic strategy to limit HCC progression both in vitro and in vivo[42]. These findings establish GPC3 as a master regulator of multiple oncogenic pathways in HCC, suggesting that targeting GPC3, particularly its exosomal form, may offer a multifaceted therapeutic approach to simultaneously inhibit tumor growth, metastasis, and angiogenesis in HCC patients.

Figure 3
Figure 3 Hypoxia-induced exosomal release of glypican-3 and its effects on hepatocellular carcinoma progression and angiogenesis. Under hypoxic conditions, hepatocellular carcinoma cells upregulate glypican-3 expression and release it via exosomes, contributing to tumor progression and angiogenesis in the tumor microenvironment. GPC3: Glypican-3; HCC: Hepatocellular carcinoma; EMT: Epithelial-mesenchymal transition.
GPC3 in metabolic adaptation and tumor stemness

In addition to its role in oncogenic signaling, GPC3 plays a pivotal role in metabolic adaptation under hypoxic conditions. It promotes lactylation and cellular glycolysis, thus enhancing tumor stemness, metabolic plasticity and immune evasion[43] (Figure 4). By increasing lactate accumulation and c-Myc signaling, GPC3 facilitates metabolic reprogramming that supports tumor adaptation to the microenvironment[43]. The relationship between GPC3 and c-Myc appears bidirectional, forming a positive feedback regulatory loop that drives HCC progression[8]. Luciferase reporter assays and chromatin immunoprecipitation analyses have demonstrated that c-Myc directly binds to the GPC3 promoter region, enhancing its transcriptional activity and protein expression[8]. Conversely, GPC3 enhances c-Myc expression and signaling. This relationship is further evidenced by functional studies where c-Myc overexpression rescues the diminished proliferative capacity, stemness, and glycolytic activity caused by GPC3 silencing, while GPC3 knockdown reduces c-Myc expression and lactate-driven post-translational modifications, leading to diminished tumor cell viability[43]. These findings suggest that GPC3 is not only a key player in signal transduction but also a critical regulator of tumor metabolism. Its ability to enhance metabolic adaptation further strengthens its role in promoting tumor aggressiveness and resistance to therapy. Understanding how GPC3 influences the metabolic landscape of HCC may help open new avenues for targeted therapeutic strategies that aim to disrupt its role in tumor cell survival.

Figure 4
Figure 4 Glypican-3-mediated regulation of c-Myc stability and metabolic adaptation under hypoxia. Glypican-3 (GPC3) regulates tumor metabolic adaptation and stemness by modulating c-Myc protein stability under normoxic and hypoxic conditions. Under normoxic conditions, c-Myc is more prone to ubiquitination and degradation when GPC3 expression is low. Under hypoxia, GPC3 is upregulated and promotes global protein lactylation, which specifically prevents c-Myc degradation, thereby stabilizing c-Myc protein and enhancing glycolysis and stemness. GPC3: Glypican-3.
Differential functions of GPC3 across cancer types

In cervical cancer, elevated GPC3 expression promotes tumor progression. Studies have shown that GPC3 antisense long non-coding RNA (GPC3-AS1) is upregulated and cooperates with ELK1 to regulate GPC3 expression, enhancing cell proliferation and migration, thereby revealing its regulatory mechanism in cervical cancer[44]. However, the functionality of GPC3 exhibits marked heterogeneity across different tumor types. In breast cancer, GPC3 acts as a tumor suppressor by inhibiting EMT through p38 MAPK signaling pathway activation, subsequently preventing metastasis and inducing tumor dormancy[45,46]. Notably, certain studies have demonstrated the capacity of GPC3 to trigger apoptotic responses in cancer cells, suggesting its potential role as a tumor suppressor gene[47]. This tumor-suppressive function is further evidenced by the significant downregulation of GPC3 expression in papillary thyroid cancer and non-small cell lung cancer[48,49]. These findings indicate the tumor-specific nature of GPC3 expression, which is likely modulated by tissue-specific signaling networks and microenvironmental factors.

The role of GPC3 in the cancer immune microenvironment

In the tumor immune microenvironment, the correlation between elevated GPC3 expression and therapeutic response has emerged as a significant area of investigation. Studies have revealed that high GPC3 expression correlates with diminished clinical efficacy and may contribute to resistance against combination therapy involving anti-PD-1 (atezolizumab) and anti-VEGF (bevacizumab) in HCC patients, potentially through interference with immune and angiogenic co-regulatory mechanisms[50]. Furthermore, research has demonstrated that combining GPC3-targeted therapy with TGF-β signaling pathway inhibition significantly enhances the anti-tumor efficacy of natural killer (NK) cells against HCC, suggesting the potential benefits of multi-targeted therapeutic approaches in immunotherapy[51]. Mechanistically, this synergistic effect occurs because TGF-β signaling normally suppresses NK cell cytotoxicity, limiting the effectiveness of GPC3-CAR NK cells alone[51]. By genetically disrupting TGFBR2 or employing dominant-negative inhibition strategies to block TGF-β signaling, NK cells can retain their cytolytic function even within highly immunosuppressive tumor microenvironments[51]. This dual-targeting approach creates a sustained and potent anti-tumor response that neither strategy could achieve independently, demonstrating significant therapeutic synergy against HCC. Parallel investigations have identified significant GPC3 upregulation in the cancer-associated fibroblasts (CAFs) of advanced gastric cancer patients, correlating strongly with poor prognosis and reduced responsiveness to PD-1 treatment[52]. These findings suggest that targeting GPC3-high CAFs could potentially enhance the therapeutic efficacy of PD-1 blockade in gastric cancer patients[52]. GPC3 secreted by CAFs induces the expression of multiple immunosuppressive and oncogenic markers, such as PD-L1, TIM-3, CD24, Cyclin D1, and c-Myc, in gastric cancer cells, thereby promoting tumor immune evasion[52]. These findings suggest that immune checkpoint blockade alone, such as anti-PD-1 therapy, may be insufficient in GPC3-enriched tumor microenvironments. In vivo models have further confirmed that neutralizing GPC3 in CAFs significantly restores CD8+ T cell activity and enhances the therapeutic efficacy of PD-1 blockade[52]. Thus, a multi-targeted strategy combining anti-GPC3 therapy with immune checkpoint inhibition may help overcome CAF-mediated immunosuppression and improve treatment outcomes in GPC3-high gastric cancer. These collective findings underscore the crucial role of GPC3 in modulating the tumor microenvironment and immune escape mechanisms, highlighting its significance in cancer immunotherapy development.

INNOVATIVE APPLICATIONS OF GPC3 IN HCC MOLECULAR IMAGING AND DIAGNOSTICS
Molecular imaging

Recent advances have enhanced GPC3-based molecular imaging for HCC diagnosis. A single-domain antibody probe (ssHN3) conjugated for selective GPC3 binding and labeled with [89Zr] demonstrated improved tumor uptake and reduced liver accumulation, optimizing PET imaging for HCC detection[53]. Another novel PET probe, [18F]AlF-NOTA-IPB-GPC3P, outperformed [18F]AlF-GPC263, showing higher cellular uptake, better internalization, and prolonged tumor retention[54]. Additionally, [68Ga]Ga-T2P, a dual-specific probe targeting GPC3 and PSMA, significantly enhanced both diagnostic sensitivity and imaging precision, underscoring the potential of multi-target probes in liver cancer detection[55].

Radiomics

Advanced radiomic models have improved non-invasive prediction of GPC3 expression in HCC. A multi-sequence magnetic resonance imaging-based radiomic scoring model integrating tumor morphology and microvascular invasion achieved high predictive accuracy [area under the curve (AUC) = 0.979][56]. Similarly, a contrast-enhanced computed tomography radiomics model demonstrated superior performance over serum AFP testing (AUC = 0.867), while integrating clinical risk factors further improved diagnostic accuracy to AUC = 0.895[57]. These methods offer effective, non-invasive approaches to assess GPC3 expression.

Serum biomarker detection

Technological advancements have significantly improved serum-based GPC3 detection. A sandwich-type ultrasensitive electrochemical aptamer sensor using both gold-polyaniline nanomaterials and graphene oxide-platinum palladium nanoparticles (PrGO-Hemin-PdNP) showed exceptional sensitivity for GPC3 detection, thus enhancing early HCC diagnosis[58]. A single-tube immuno-RPA and CRISPR-Cas13a system enables rapid, extraction-free detection of GPC3 in EVs, offering a promising diagnostic tool[59]. Additionally, time-resolved fluorescence immunochromatography (TRFIS) allows 15-minute plasma exosome-based GPC3 detection, demonstrating the potential for rapid HCC screening[60].

Circulating tumor cell detection

Circulating tumor cell (CTC)-based technologies have advanced GPC3 detection in HCC. A dual fluorescence aptamer-based sensing system achieved 92% sensitivity and 100% specificity, with a detection limit of just 2 cells/mL[61]. The d-SCOUT technology, capable of quantifying multiple HCC CTC biomarkers including GPC3, offers high sensitivity and reproducibility, further enhancing diagnostic precision[61].

Together, these advancements demonstrate that GPC3 has been innovatively applied across molecular imaging, radiomics, serum detection and CTC analysis, significantly improving the sensitivity and accuracy of HCC diagnosis. These technologies provide novel, non-invasive tools for early detection and disease monitoring, reinforcing GPC3’s clinical potential as both a diagnostic and prognostic marker in liver cancer.

TRANSLATIONAL PROGRESS OF GPC3-DIRECTED THERAPIES IN HCC

Given its selective expression in HCC, various GPC3-targeting strategies have been developed, including monoclonal antibodies, ADCs, photodynamic therapy (PDT), DNA vaccines, and CAR-T-cell therapies (Figure 5). Although still under development, these approaches have demonstrated promising antitumor activity in preclinical models. In particular, targeting the C-terminal domain of GPC3 helps prevent loss of tumor specificity due to proteolytic cleavage. However, while several Phase I clinical trials have been completed, multiple trials are still ongoing (Table 1).

Figure 5
Figure 5 Applications of glypican-3-targeted therapies. Glypican-3 (GPC3) serves as a promising target for hepatocellular carcinoma (HCC) therapy, with various therapeutic strategies under development. I-124 codrituzumab, a radiolabeled monoclonal antibody targeting GPC3, enables tumor imaging and potential radioimmunotherapy applications. Antibody-drug conjugates utilize GPC3-targeting antibodies conjugated with cytotoxic agents to selectively eliminate cancer cells. Photodynamic therapy, an emerging approach, employs light-activated GPC3-targeting molecules to induce localized cytotoxicity. GPC3-specific chimeric antigen receptor (CAR)-T cells have been engineered to target tumor cells, with their efficacy further enhanced by the secretion of cytokines such as IL-15, IL-21, and IL-7, which improve CAR-T cell persistence and strengthen antitumor activity. Bispecific CAR-T cells, designed to simultaneously recognize GPC3 and B7-H3, offer a dual-targeting strategy that enhances tumor specificity, boosts immune activation, and mitigates antigen escape, ultimately improving the effectiveness of CAR-T-cell therapy. In DNA vaccine development, PLGA/PEI nanoparticles are utilized to encapsulate and deliver GPC3 and HMGB1, aiming to stimulate immune responses and activate cytotoxic T lymphocyte. ERY974, a bispecific T cell-redirecting antibody, is designed to target both GPC3 on tumor cells and CD3 on T cells, facilitating potent T cell-mediated tumor killing. Collectively, these GPC3-targeted strategies offer significant potential for advancing HCC treatment by leveraging multiple mechanisms to enhance tumor specificity, immune activation, and therapeutic efficacy. CTL: Cytotoxic T lymphocyte; CAR-T: Chimeric antigen receptor-T.
Table 1 Current clinical trials targeting glypican-3.
No.
NCT number
Study title
Study status
Stage
Enrollment (estimated)
Primary endpoint
1NCT05352542GPC3-targeting CART Cell in Treatment of Advanced Hepatocellular CarcinomaTerminatedPhase 110Incidence, severity, and type of TEAEs; DLT rate; RP2D finding; CAR positive T cells; CAR transgene levels in peripheral blood
2NCT02395250Anti-GPC3 CAR T for Treating Patients with Advanced HCCCompletedPhase 113AEs attributed to the administration of the anti-GPC3 CAR T cells
3NCT03146234CAR-GPC3 T Cells in Patients with Refractory Hepatocellular CarcinomaCompletedNA7Safety and tolerance
4NCT039802884th Generation Chimeric Antigen Receptor T Cells Targeting Glypican-3CompletedPhase 16DLT and MTD
5NCT02905188Glypican 3-specific Chimeric Antigen Receptor Expressing T Cells for Hepatocellular Carcinoma (GLYCAR)CompletedPhase 19Number of patients with DLT; RP2D
6NCT03884751Chimeric Antigen Receptor T Cells Targeting Glypican-3CompletedPhase 19DLT, MTD
7NCT02748837A Study of ERY974 in Patient with Advanced Solid TumorsCompletedPhase 129Dose escalation: MTD determination; cohort expansion: Preliminary assessment of change in tumor size
8NCT05070156B010-A Injection for Treating Patients with GPC3 Positive Advanced Hepatocellular CarcinomaActive not recruitingPhase 13Evaluate incidence of TEAEs [safety and tolerability] after B010-A injection
9NCT02932956Glypican 3-specific Chimeric Antigen Receptor Expressed in T Cells for Patients with Pediatric Solid Tumors (GAP)Active not recruitingPhase 110Number of patients with DLT
10NCT04405778A Study of TAK-102 in Adult with Previously-Treated Solid TumorsActive not recruitingPhase 111Number of participants with first cycle DLTs; number of participants with one or more TEAE; number of participants with AEs of clinical interest
11NCT03198546GPC3-CAR-T Cells for Immunotherapy of Cancer with GPC3 ExpressionRecruitingPhase 130Number of patients with DLT
12NCT05003895GPC3 Targeted CAR-T Cell Therapy in Advanced GPC3 Expressing Hepatocellular Carcinoma (HCC)RecruitingPhase 138To determine the safety and feasibility of T-cells, expressing a novel humanized anti-GPC3 chimeric antigen receptor, in patients with advanced HCC, expressing GPC3
13NCT04928677A Study of Codrituzumab in Children and Young Adults with Solid Tumors and Have Not Responded to Treatment or Have Come Back After TreatmentRecruitingPhase 150Estimate the MTD
14NCT06196294GPC3/Mesothelin-CAR-γδT Cells Against CancersRecruitingPhase 130Number of patients with DLT
15NCT06461624Clinical Trial of Autologous GPC3 CAR-T Cells (CBG166) Therapy for Advanced Hepatocellular CarcinomaRecruitingPhase 115DLT; AEs; MTD
16NCT05620706Study of GPC-3 CAR-T Cells in Treating with Hepatocellular CarcinomaRecruitingNA20AE/SAE
17NCT05410717CLDN6/GPC3/Mesothelin/AXL-CAR-NK Cell Therapy for Advanced Solid TumorsRecruitingPhase 1200Safety by CTCAE V5.0
18NCT05047510GPC3 Targeted Fluorescence Image Guided Surgery of Hepatocellular CarcinomaRecruitingNA60HCC lesions (numbers of intraoperatively detected hepatocellular carcinoma lesions)
19NCT05779917Mesothelin/GPC3/GUCY2C-CAR-T Cells Against CancersRecruitingPhase 130Number of patients with DLT
20NCT06383520Study on the Clinical Application Value of PET Imaging Targeting GPC3 in Hepatocellular CarcinomaRecruitingEarly Phase 1100Visual and standardized uptake values assessment of lesions and biodistribution
21NCT05926726GPC3-directed CAR-T in the Treatment Amongst Subjects With Advanced Hepatocellular CarcinomaRecruitingNA12TRAEs; DLT; RP2D of JWATM214 in HCC patients
22NCT06687941A Study to Evaluate the Tolerability, Safety, and PK of AST-201 in Patients With GPC3-positive Advanced Solid TumorsRecruitingPhase 170DLT
23NCT06726161Study of the Theranostic Pair RYZ811 (diagnostic) and RYZ801 (therapeutic) to Identify and Treat Subjects with GPC3+ Unresectable HCCRecruitingPhase 170Evaluate RYZ801 dose and safety; Evaluate RYZ811 safety and distribution
24NCT03198052GPC3/Mesothelin/Claudin18.2/GUCY2C/B7-H3/PSCA/PSMA/MUC1/TGFβ/HER2/Lewis-Y/AXL/EGFR-CAR-T Cells Against CancersRecruitingPhase 130Number of patients with DLT
25NCT06652243Clinical Study of SN301A Injection in the Treatment of Hepatocellular CarcinomaRecruitingPhase 112DLT; Incidence and severity of AEs and SAEs (safety and tolerability)
26NCT06478693A Study of MT-303 in Adults With Advanced or Metastatic GPC3-Expressing Cancers, Including HCCRecruitingPhase 148Type, incidence and severity of AEs; RP2D; change from baseline in vital signs; change in laboratory parameters; change from baseline in ECG parameters
27NCT06084884A Phase I/II Study to Evaluate AZD5851 in GPC3+ Advanced/Recurrent Hepatocellular CarcinomaRecruitingPhase 1
Phase 2
94Incidence of participants with DLTs, AEs, including AESIs and SAEs; determination of the recommended dose of AZD5851 for expansion phase
28NCT05120271BOXR1030 T Cells in Subjects With Advanced GPC3-Positive Solid TumorsRecruitingPhase 1; Phase 2110Dose limiting toxicity; MTD; RP2D; TEAEs
29NCT05783570To Evaluate the Safety, Tolerability and Preliminary Efficacy of EU307RecruitingPhase 112AEs (including DLT); production of replication competent lentiviruses; development of anti-drug antibodies
30NCT04377932Interleukin-15 Armored Glypican 3-specific Chimeric Antigen Receptor Expressed in T Cells for Pediatric Solid TumorsRecruitingPhase 124Number of patients with DLT
31NCT04715191Interleukin-15 and -21 Armored Glypican-3-specific Chimeric Antigen Receptor Expressed in T Cells for Pediatric Solid TumorsRecruitingPhase 124Number of patients with DLT
32NCT06427941A Phase 1 Study of BGB-B2033, Alone or in Combination With Tislelizumab, in Participants With Advanced or Metastatic Solid TumorsRecruitingPhase 170DLT
33NCT05103631Interleukin-15 Armored Glypican 3-specific Chimeric Antigen Receptor Expressed in Autologous T Cells for Solid TumorsRecruitingPhase 127Number of patients with DLT
34NCT06088459NWRD06 DNA Plasmid for HCC After Radical ResectionRecruitingPhase 19DLT; all AEs
35NCT04756648Phase I Clinical Trial of CT0180 Cells in the Treatment of Hepatocellular CarcinomaRecruitingPhase 121DLT; MTD; AE; AESI
36NCT06560827CT011 Autologous CAR-T Cells in Patients With Hepatocellular Carcinoma at Risk of Recurrence After Surgical ResectionRecruitingPhase 130Incidence and severity of TEAE; incidence and severity of TRAE; incidence and severity of AESI
37NCT04842812Engineered TILs/CAR-TILs to Treat Advanced Solid TumorsRecruitingPhase 140Safety of TILs/CAR-TILs treatment in advanced solid cancers
38NCT06715839Target-specific immunoPET Imaging of Digestive System CarcinomaRecruitingNA400Biodistribution; standardized uptake value; radiation dosimetry; diagnostic value in patients with digestive system malignancy; the correlation between the expression of specific target and tracer uptake value
39NCT05263830Glypican-3 as a Prognostic Factor in Patients With Hepatocellular Carcinoma Treated by ImmunotherapyRecruitingNA120Evaluate concentration of circulating GPC-3
40NCT04506983GPC3-CAR-T Cells for the Hepatocellular CarcinomaSuspendedPhase 112Percentage of AEs
41NCT06815432GPC-3 CAR T CELLS for Recurrent GPC-3 Positive GlioblastomaNot yet recruitingPhase 127Number of patients with DLT
42NCT06641453CAR-T Cell Therapy Targeting GPC3 in Patients with Advanced GPC3-Positive Hepatocellular CarcinomaNot yet recruitingPhase 1; Phase 230Incidence of TRAEs; incidence of DLTs
43NCT06198296Immunotherapy for Adults with GPC3-Positive Solid Tumors Using IL-15 and IL-21 Armored GPC3-CAR T CellsNot yet recruitingPhase 121Number of patients with DLT
44NCT06590246A Study to Evaluate C-CAR031 in Glypican-3 (GPC3)+ Advanced/recurrent Hepatocellular Carcinoma (HCC)Not yet recruitingPhase 1; Phase 2121Safety and tolerability; anti-tumor activity
45NCT06795022First in Human Study to Evaluate AZD9793 in Participants With Advanced or Metastatic Solid TumoursNot yet recruitingPhase 1; Phase 2304Number of patients with AEs; number of patients with SAEs; number of patients with AESIs; number of patients with DLT; objective response rate
46NCT02723942CAR-T Cell Immunotherapy for HCC Targeting GPC3WithdrawnPhase 1; Phase 20Radiological assessment
47NCT04093648T Cells co- Expressing a Second Generation Glypican 3-specific Chimeric Antigen Receptor With Cytokines Interleukin-21 and 15 as Immunotherapy for Patients With Liver Cancer (TEGAR)WithdrawnPhase 10DLT rate
48NCT04121273GPC3-targeted CAR-T Cell for Treating GPC3 Positive Advanced HCCUnknownPhase 120Number of patient with dose limiting toxicity
49NCT03084380Anti-GPC3 CAR-T for Treating GPC3-positive Advanced Hepatocellular Carcinoma (HCC)UnknownPhase 1; Phase 220Safety: Measured by occurrence of study related adverse effects defined by NCI CTCAE 4.0
50NCT03130712A Study of GPC3-targeted T Cells by Intratumor Injection for Advanced HCC (GPC3-CART)UnknownPhase 1; Phase 210Safety of CAR-T cell infusion mediated by intratumoral injection as measured by number of participants with AEs
51NCT02715362A Study of GPC3 Redirected Autologous T Cells for Advanced HCCUnknownPhase 1; Phase 230Safety of CAR-T cell infusion mediated by TAI as measured by number of participants with AEs
52NCT05344664Novel GPC3 CAR-T Cell Therapy for Hepatocellular CarcinomaUnknownPhase 112Percentage of AEs
53NCT02876978Anti-GPC3 CAR T for Recurrent or Refractory Lung Squamous Cell CarcinomaUnknownPhase 120Safety and tolerance: Occurrence of study related AEs
54NCT04076137Targeted T-cell Therapy in Solid TumorsUnknownEarly Phase 110Overrall survival
55NCT03146637Study of Activated CIK Armed With Bispecific Antibody for Advanced Liver CancerUnknownPhase 280Overrall survival
56NCT04728139Assessment of Glypican 3 as Apredictive Marker in Colorectal Cancer PatientsUnknownNA90Evaluation of Glypican 3 role as a predictive and diagnostic marker of colorectal marker
57NCT05059821Personalized Cancer Vaccine in Egyptian Cancer PatientsUnknownPhase 110Assessment of the safety of the personalized cancer vaccine; assessment of immunological response

Among the earliest GPC3-targeting therapeutic approaches, codrituzumab, a humanized monoclonal antibody against GPC3, was developed to leverage antibody-dependent cytotoxicity through its interaction with CD16/FcγRIIIa[62]. However, despite its promising preclinical efficacy, codrituzumab did not demonstrate any significant clinical benefit in a randomized Phase II placebo-controlled trial involving previously treated advanced HCC patients[62] (NCT01507168). Several factors may have contributed to this outcome. First, the clinical dosing only achieved approximately 50%-60% GPC3 target saturation, which may have been insufficient to trigger effective antitumor responses. Second, therapeutic benefit was observed primarily in patients with both high GPC3 and high CD16 expression. The absence of prospective biomarker-based patient selection likely resulted in a diluted overall treatment effect. Furthermore, interindividual variation in CD16 expression may have influenced the efficiency of NK cell-mediated antibody-dependent cellular cytotoxicity, further impacting response. These findings underscore the importance of tumor and immune microenvironment heterogeneity in determining therapeutic efficacy. Building on these insights, I-124 codrituzumab (GC33) was designed as a radiolabeled diagnostic tool for detecting GPC3-expressing HCC. In clinical evaluations, I-124 codrituzumab successfully localized tumors in most HCC patients without exhibiting cross-reactivity with normal tissues, demonstrating its potential as a non-invasive diagnostic tool[63]. Beyond monoclonal antibodies, GPC3 has emerged as a promising target for ADCs. Two GPC3-specific ADCs, hYP7-DC and hYP7-PC, were developed using duocarmycin SA and pyrrolobenzodiazepine dimer as cytotoxic payloads[7]. These ADCs demonstrated potent anti-tumor activity at picomolar concentrations against GPC3-positive cancer cell lines while sparing GPC3-negative cells[7]. Recently, GPC3-targeting peptide-drug conjugates have been explored for PDT, offering a selective tumor-killing strategy. Among them, conjugate 8b, designed by linking a GPC3-targeting peptide to chlorin e6, demonstrated strong tumor-targeting ability, potent anti-tumor efficacy, and minimal toxicity to normal cells[64]. In a HepG2 xenograft model, light-activated conjugate 8b achieved complete tumor elimination without harming surrounding tissues, highlighting its clinical potential[64].

ERY974 is a humanized IgG4 bispecific T cell-redirecting antibody that simultaneously targets GPC3 on tumor cells and CD3 on T cells to promote T cell-mediated tumor killing[65]. In preclinical studies, ERY974 demonstrated strong antitumor activity against various GPC3-positive tumors and was capable of converting non-inflamed tumor microenvironments into inflamed ones, thereby enhancing T cell infiltration[65]. In addition, combination therapy with ERY974 and chemotherapeutic agents such as paclitaxel, cisplatin, and capecitabine showed synergistic efficacy by promoting T cell infiltration into the tumor core and enhancing drug activation, significantly improving responses in poorly immunogenic tumors[66]. The phase I clinical trial has been completed, where a starting dose of 3.0 ng/kg was selected, which is considered to be within a safe and potentially effective range based on both safety and efficacy perspectives[67]. As GPC3-targeted therapies continue to evolve, the focus has increasingly shifted toward immune cell-based treatments. In particular, CAR-T-cell therapy has shown impressive potential against GPC3-positive tumors in preclinical studies.

Preclinical evaluation of GPC3-CAR-T cells

In preclinical mouse models, GPC3-CAR-T cells demonstrated significant anti-tumor activity, eliminating approximately 66% of the tumor burden. Beyond direct tumor cell killing, these cells also influence Wnt signaling, which is critical in HCC progression. However, challenges such as limited persistence and the immunosuppressive tumor microenvironment hinder their therapeutic efficacy. To address these limitations, researchers are actively refining GPC3-CAR-T cell design in order to enhance both their functionality and clinical potential.

Patients in clinical trials NCT02905188 and NCT02932956 received GPC3 CAR-T cell therapy, which was found to be safe but did not generate a significant antitumor response[68]. However, challenges such as limited persistence and the immunosuppressive tumor microenvironment hindered its therapeutic efficacy. To overcome these limitations, researchers developed a modified version co-expressing IL-15 (15.CAR), as IL-15 has been shown to enhance expansion, intratumoral persistence, and antitumor activity in GPC3-CAR-T cells. In clinical trials NCT05103631 and NCT04377932, 15.CAR-T cells exhibited significantly improved expansion, resulting in a disease control rate of 66% and an objective response rate of 33%[68].

Cytokine co-expression to boost CAR-T cell persistence

A major limitation of CAR-T therapy in HCC is the absence of essential survival signals, such as IL-15 and IL-21, within the tumor microenvironment[69,70]. GPC3-CAR-T cells co-expressing IL-15 and IL-21 exhibited enhanced proliferation and anti-tumor activity, significantly improving therapeutic outcomes in HCC mouse models[71]. Additionally, a novel GPC3-7-19-CAR-T construct, engineered to secrete IL-7 and CCL19, demonstrated increased tumor-killing efficiency and promoted immune cell infiltration within the tumor microenvironment, further strengthening therapeutic efficacy[72].

Metabolic regulation for CAR-T enhancement

Metabolic adaptation is critical for CAR-T cell persistence and function. Overexpression of GLUT1 or AGK enhanced glycolytic metabolism and anti-apoptotic capacity, improving GPC3-CAR-T cell persistence and cytotoxicity via PI3K/Akt pathway activation in HCC xenograft models[73]. In addition to metabolic adaptation, T-cell exhaustion markers such as CD39 also play a crucial role in CAR-T cell persistence and function[74,75]. Optimal anti-tumor efficacy was observed in moderate CD39-expressing GPC3-CAR-T cells, as excessive or insufficient CD39 expression impaired function[76]. Targeting CD39 with either Mitochondrial Division Inhibitor 1 (Mdivi-1) or shRNA led to the highest proportion of CD39int CAR-T cells, resulting in improved tumor infiltration and robust anti-tumor activity in vivo[76].

Multi-target CAR-T therapies for enhanced efficacy

One of the major obstacles in CAR-T therapy is antigen escape, which occurs due to antigen loss, shedding, or low expression, as well as limited T-cell homing and immunosuppressive tumor microenvironments[77,78].

A recent preclinical study developed GPC3-BiTE CAR-T cells, which not only express a GPC3-directed CAR but also secrete BiTE molecules targeting B7H3[79]. These BiTEs engage CD3 on non-transduced T cells, expanding their recruitment and cytotoxic activity against HCC. This dual-targeting approach enabled non-transduced T cells to attack GPC3-/B7H3+ tumor cells, compensating for the limitations of single-antigen targeting and addressing antigen heterogeneity. Compared to conventional GPC3-CAR-T cells, BiTE-secreting CAR-T cells exhibited superior cytotoxicity against GPC3+/B7H3+ tumor cells and effectively targeted GPC3-/B7H3+ HCC cells, enhancing CD69 expression and IFN-γ production[79]. These findings highlight the potential of GPC3-BiTE CAR-T therapy in mitigating antigen escape and improving therapeutic outcomes.

To further enhance T-cell homing and infiltration, researchers engineered GPC3-CAR-T cells co-expressing IL-21 and CXCL9 in combination with PD-1 blockade, significantly improving tumor infiltration and tumor suppression in HCC xenograft models[80]. Mechanistically, this study demonstrates that GPC3-CAR-T cells engineered to co-express IL-21 and CXCL9 exhibit enhanced proliferation, cytokine secretion (including IL-2, IFN-γ, and TNF-α), and improved tumor infiltration[80]. When combined with PD-1 blockade, these modified CAR-T cells further increase their effector differentiation, such as cytotoxic T lymphocytes, natural killer T cells, and effector memory T (TEM) cells, and their cytotoxic activity by alleviating T cell exhaustion[80]. This dual approach not only promotes T cell recruitment via CXCL9-mediated chemotaxis but also overcomes immune suppression through PD-1 pathway inhibition. Together, the combination therapy markedly enhances antitumor efficacy in both in vitro and in vivo HCC models, offering a promising strategy to overcome the immunosuppressive tumor microenvironment and improve immunotherapeutic outcomes for GPC3-positive liver cancer patients.

Allogeneic and novel T-cell therapies

Conventional autologous CAR-T cell therapy faces challenges in manufacturing, T-cell homing, antigen heterogeneity, and tumor immune evasion. To overcome these limitations, researchers developed GPC3-CAR/sIL-15 Vδ1 T cells, leveraging the unique properties of Vδ1 γδ T cells, which exhibit MHC-independent cytotoxicity and a reduced risk of graft-versus-host disease (GvHD)[81]. By engineering these CAR-T cells to secrete soluble IL-15 (sIL-15), researchers improved both T-cell survival and persistence, counteracting the immunosuppressive tumor microenvironment. In vitro studies demonstrated that GPC3-CAR/sIL-15 Vδ1 T cells exhibited potent cytotoxicity against both high (HepG2) and low (PLC/PRF/5) GPC3-expressing HCC cells, suggesting their ability to overcome antigen heterogeneity[81]. In a HepG2 xenograft mouse model, a single intravenous injection of GPC3-CAR/sIL-15 Vδ1 T cells significantly inhibited tumor growth while predominantly accumulating at tumor sites, highlighting strong tumor infiltration capabilities. Compared to non-sIL-15-expressing CAR-T cells, those expressing sIL-15 demonstrated a prolonged expansion and sustained tumor suppression without requiring IL-2 supplementation. Moreover, this strategy reduced T-cell exhaustion markers (PD-1, TIM-3, LAG-3) and promoted memory T-cell formation, enhancing long-term immune responses. Importantly, no GvHD-related toxicity was observed, indicating a high safety profile[81]. These findings suggest that GPC3-CAR/sIL-15 Vδ1 T cells hold promise as an off-the-shelf therapy for HCC and other GPC3-expressing tumors.

GPC3-targeted DNA vaccine for enhanced T-Cell response

Beyond CAR-T cell therapy, a novel PLGA/PEI-HMGB1/GPC3 dual-target DNA vaccine was developed to enhance CD8+ T-cell responses and counteract immune suppression in HCC[82].

HMGB1, an inflammatory mediator overexpressed in solid tumors, contributes to cancer progression and immune modulation[83]. While blocking extracellular HMGB1 has been shown to inhibit tumor growth and enhance immune checkpoint blockade efficacy, HMGB1 also plays a role in adaptive immunity by promoting dendritic cell maturation, Th1 differentiation, and T-cell proliferation[84,85]. By leveraging HMGB1 as an immune adjuvant, researchers designed a GPC3-targeted vaccine that enhances dendritic cell activation and antigen presentation, leading to a stronger T-cell response against HCC. The PLGA/PEI nanoparticle delivery system further improved vaccine expression, antigen release, and T-cell efficacy. Preclinical studies in Hepa1-6-hGPC3 mouse models demonstrated significant tumor growth inhibition, along with increased CD8+ T-cell and dendritic cell infiltration[82]. Compared to a GPC3-only vaccine, the dual-target vaccine enhanced IFN-γ secretion and cytotoxic activity, resulting in better tumor control[82]. However, the precise role of HMGB1 in vaccine-induced immunity remains to be elucidated, necessitating further investigation in additional preclinical models.

COMPARISON TO OTHER HCC THERAPIES AND FUTURE PERSPECTIVES

The treatment of advanced HCC has evolved from TKIs, such as sorafenib and lenvatinib, to ICIs, which have improved clinical outcomes to some extent[86,87]. However, tumor heterogeneity, an immunosuppressive microenvironment, and adaptive resistance mechanisms continue to hinder treatment efficacy, leading to limited tumor responses and patient survival[9,10]. These challenges highlight the urgent need for novel, tumor-specific therapeutic strategies.

As a tumor-specific antigen highly expressed in HCC, GPC3 represents a promising therapeutic target with the potential to overcome current treatment limitations. Multiple clinical trials are evaluating GPC3-directed approaches, including CAR-T cell therapy, DNA vaccines, T-cell redirecting antibodies, and allogeneic strategies. Among these, CAR-T cell therapy has seen major advancements through cytokine modulation, metabolic reprogramming, dual-targeting strategies, and combination therapies with ICIs, significantly improving persistence, tumor infiltration and antitumor efficacy. Additionally, emerging modalities such as vaccines and multifunctional antibody therapies continue to expand the immunotherapy landscape, offering new opportunities for enhancing immune responses against HCC.

Although there has been significant progress, further improvements are needed to boost effectiveness, overcome resistance, and better identify the right patients for treatment. Future research should focus on integrating GPC3-targeted therapies into personalized treatment plans, exploring combination approaches, and enhancing drug delivery to maximize effectiveness. HBV-related HCC progression involves the epigenetic silencing of tumor-suppressive miRNAs, such as let-7c, through HBx-induced EZH2 upregulation, leading to HMGA2-mediated metastasis[88]. This underscores the complexity of HCC oncogenesis and highlights the potential of combinatorial strategies that target both surface antigens like GPC3 and intracellular oncogenic pathways.

CONCLUSION

With ongoing advancements in both CAR-T technology and clinical trials, GPC3-targeted therapies are poised to revolutionize HCC treatment, addressing key limitations within the existing therapies. By integrating innovative dual-targeting designs, metabolic enhancements, and immune checkpoint blockade, these therapies have the potential to overcome antigen escape, enhance T-cell function, and improve treatment durability. As research continues, GPC3-based immunotherapies may offer breakthrough solutions for HCC, bringing new hope for improved patient outcomes and long-term disease control.

ACKNOWLEDGEMENTS

The authors thank Mr. Tsung-Hung Hung from the Medical Art Department, IBMS, Academia Sinica, for figure preparation.

Footnotes

Provenance and peer review: Invited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country of origin: Taiwan

Peer-review report’s classification

Scientific Quality: Grade A, Grade B, Grade C

Novelty: Grade A, Grade B, Grade C

Creativity or Innovation: Grade A, Grade B, Grade C

Scientific Significance: Grade B, Grade B, Grade C

P-Reviewer: Wang W, Lecturer, Postdoc, China; Yang Z, PhD, Professor, Editorial Director, China S-Editor: Qu XL L-Editor: A P-Editor: Zhang YL

References
1.  Li N, Gao W, Zhang YF, Ho M. Glypicans as Cancer Therapeutic Targets. Trends Cancer. 2018;4:741-754.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 97]  [Cited by in RCA: 105]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
2.  Baumhoer D, Tornillo L, Stadlmann S, Roncalli M, Diamantis EK, Terracciano LM. Glypican 3 expression in human nonneoplastic, preneoplastic, and neoplastic tissues: a tissue microarray analysis of 4,387 tissue samples. Am J Clin Pathol. 2008;129:899-906.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 171]  [Cited by in RCA: 192]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
3.  Ho M, Kim H. Glypican-3: a new target for cancer immunotherapy. Eur J Cancer. 2011;47:333-338.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 181]  [Cited by in RCA: 181]  [Article Influence: 12.9]  [Reference Citation Analysis (0)]
4.  Lin H, Huber R, Schlessinger D, Morin PJ. Frequent silencing of the GPC3 gene in ovarian cancer cell lines. Cancer Res. 1999;59:807-810.  [PubMed]  [DOI]
5.  Xiang YY, Ladeda V, Filmus J. Glypican-3 expression is silenced in human breast cancer. Oncogene. 2001;20:7408-7412.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 151]  [Cited by in RCA: 158]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
6.  Valsechi MC, Oliveira AB, Conceição AL, Stuqui B, Candido NM, Provazzi PJ, de Araújo LF, Silva WA Jr, Calmon Mde F, Rahal P. GPC3 reduces cell proliferation in renal carcinoma cell lines. BMC Cancer. 2014;14:631.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 30]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
7.  Fu Y, Urban DJ, Nani RR, Zhang YF, Li N, Fu H, Shah H, Gorka AP, Guha R, Chen L, Hall MD, Schnermann MJ, Ho M. Glypican-3-Specific Antibody Drug Conjugates Targeting Hepatocellular Carcinoma. Hepatology. 2019;70:563-576.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 43]  [Cited by in RCA: 86]  [Article Influence: 14.3]  [Reference Citation Analysis (0)]
8.  Li L, Jin R, Zhang X, Lv F, Liu L, Liu D, Liu K, Li N, Chen D. Oncogenic activation of glypican-3 by c-Myc in human hepatocellular carcinoma. Hepatology. 2012;56:1380-1390.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 56]  [Cited by in RCA: 68]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
9.  Yang X, Yang C, Zhang S, Geng H, Zhu AX, Bernards R, Qin W, Fan J, Wang C, Gao Q. Precision treatment in advanced hepatocellular carcinoma. Cancer Cell. 2024;42:180-197.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 155]  [Reference Citation Analysis (0)]
10.  Ladd AD, Duarte S, Sahin I, Zarrinpar A. Mechanisms of drug resistance in HCC. Hepatology. 2024;79:926-940.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 52]  [Cited by in RCA: 107]  [Article Influence: 107.0]  [Reference Citation Analysis (0)]
11.  Capurro M, Shi W, Izumikawa T, Kitagawa H, Filmus J. Processing by convertases is required for glypican-3-induced inhibition of Hedgehog signaling. J Biol Chem. 2015;290:7576-7585.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 19]  [Cited by in RCA: 25]  [Article Influence: 2.5]  [Reference Citation Analysis (0)]
12.  Mauro M, Ugo P, Walton Z, Ali S, Rastellini C, Cicalese L. Glypican-3 (GPC-3) Structural Analysis and Cargo in Serum Small Extracellular Vesicles of Hepatocellular Carcinoma Patients. Int J Mol Sci. 2023;24:10922.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]
13.  Chen M, Li G, Yan J, Lu X, Cui J, Ni Z, Cheng W, Qian G, Zhang J, Tu H. Reevaluation of glypican-3 as a serological marker for hepatocellular carcinoma. Clin Chim Acta. 2013;423:105-111.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 37]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
14.  Batbaatar B, Gurbadam U, Tuvshinsaikhan O, Narmandakh NE, Khatanbaatar G, Radnaabazar M, Erdene-Ochir D, Boldbaatar M, Byambaragchaa M, Amankyeldi Y, Chogsom M, Ganbileg N, Batdelger A, Demchig T, Nyam-Osor L, Bayartugs B, Batmunkh E, Munkhjargal B, Lonjid T, Khasbagana B, Batmunkh M, Jav S, Semchin M. Evaluation of glypican‑3 in patients with hepatocellular carcinoma. Mol Clin Oncol. 2025;22:1.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 2]  [Reference Citation Analysis (0)]
15.  Kim DY, Toan BN, Tan CK, Hasan I, Setiawan L, Yu ML, Izumi N, Huyen NN, Chow PK, Mohamed R, Chan SL, Tanwandee T, Lee TY, Hai TTN, Yang T, Lee WC, Chan HLY. Utility of combining PIVKA-II and AFP in the surveillance and monitoring of hepatocellular carcinoma in the Asia-Pacific region. Clin Mol Hepatol. 2023;29:277-292.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 68]  [Cited by in RCA: 76]  [Article Influence: 38.0]  [Reference Citation Analysis (0)]
16.  Wu CS, Lee TY, Chou RH, Yen CJ, Huang WC, Wu CY, Yu YL. Development of a highly sensitive glycan microarray for quantifying AFP-L3 for early prediction of hepatitis B virus-related hepatocellular carcinoma. PLoS One. 2014;9:e99959.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 19]  [Cited by in RCA: 22]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
17.  Kolluri A, Ho M. The Role of Glypican-3 in Regulating Wnt, YAP, and Hedgehog in Liver Cancer. Front Oncol. 2019;9:708.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 47]  [Cited by in RCA: 80]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
18.  Gao W, Kim H, Feng M, Phung Y, Xavier CP, Rubin JS, Ho M. Inactivation of Wnt signaling by a human antibody that recognizes the heparan sulfate chains of glypican-3 for liver cancer therapy. Hepatology. 2014;60:576-587.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 111]  [Cited by in RCA: 140]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
19.  Grisaru S, Cano-Gauci D, Tee J, Filmus J, Rosenblum ND. Glypican-3 modulates BMP- and FGF-mediated effects during renal branching morphogenesis. Dev Biol. 2001;231:31-46.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 139]  [Cited by in RCA: 139]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
20.  Dwivedi PP, Grose RH, Filmus J, Hii CS, Xian CJ, Anderson PJ, Powell BC. Regulation of bone morphogenetic protein signalling and cranial osteogenesis by Gpc1 and Gpc3. Bone. 2013;55:367-376.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 38]  [Cited by in RCA: 42]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
21.  Cheng W, Tseng CJ, Lin TT, Cheng I, Pan HW, Hsu HC, Lee YM. Glypican-3-mediated oncogenesis involves the Insulin-like growth factor-signaling pathway. Carcinogenesis. 2008;29:1319-1326.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 65]  [Cited by in RCA: 71]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
22.  Grozdanov PN, Yovchev MI, Dabeva MD. The oncofetal protein glypican-3 is a novel marker of hepatic progenitor/oval cells. Lab Invest. 2006;86:1272-1284.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 72]  [Cited by in RCA: 83]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
23.  Iglesias BV, Centeno G, Pascuccelli H, Ward F, Peters MG, Filmus J, Puricelli L, de Kier Joffé EB. Expression pattern of glypican-3 (GPC3) during human embryonic and fetal development. Histol Histopathol. 2008;23:1333-1340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 44]  [Reference Citation Analysis (0)]
24.  Capurro MI, Xu P, Shi W, Li F, Jia A, Filmus J. Glypican-3 inhibits Hedgehog signaling during development by competing with patched for Hedgehog binding. Dev Cell. 2008;14:700-711.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 256]  [Cited by in RCA: 278]  [Article Influence: 16.4]  [Reference Citation Analysis (0)]
25.  Akkermans O, Delloye-Bourgeois C, Peregrina C, Carrasquero-Ordaz M, Kokolaki M, Berbeira-Santana M, Chavent M, Reynaud F, Raj R, Agirre J, Aksu M, White ES, Lowe E, Ben Amar D, Zaballa S, Huo J, Pakos I, McCubbin PTN, Comoletti D, Owens RJ, Robinson CV, Castellani V, Del Toro D, Seiradake E. GPC3-Unc5 receptor complex structure and role in cell migration. Cell. 2022;185:3931-3949.e26.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 15]  [Cited by in RCA: 31]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
26.  Li N, Wei L, Liu X, Bai H, Ye Y, Li D, Li N, Baxa U, Wang Q, Lv L, Chen Y, Feng M, Lee B, Gao W, Ho M. A Frizzled-Like Cysteine-Rich Domain in Glypican-3 Mediates Wnt Binding and Regulates Hepatocellular Carcinoma Tumor Growth in Mice. Hepatology. 2019;70:1231-1245.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 58]  [Cited by in RCA: 88]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
27.  Capurro M, Martin T, Shi W, Filmus J. Glypican-3 binds to Frizzled and plays a direct role in the stimulation of canonical Wnt signaling. J Cell Sci. 2014;127:1565-1575.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 96]  [Cited by in RCA: 113]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
28.  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;59:1107-1117.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 194]  [Cited by in RCA: 214]  [Article Influence: 17.8]  [Reference Citation Analysis (0)]
29.  Zheng X, Liu X, Lei Y, Wang G, Liu M. Glypican-3: A Novel and Promising Target for the Treatment of Hepatocellular Carcinoma. Front Oncol. 2022;12:824208.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 64]  [Article Influence: 21.3]  [Reference Citation Analysis (0)]
30.  Zittermann SI, Capurro MI, Shi W, Filmus J. Soluble glypican 3 inhibits the growth of hepatocellular carcinoma in vitro and in vivo. Int J Cancer. 2010;126:1291-1301.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 16]  [Cited by in RCA: 59]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
31.  Park HW, Kim YC, Yu B, Moroishi T, Mo JS, Plouffe SW, Meng Z, Lin KC, Yu FX, Alexander CM, Wang CY, Guan KL. Alternative Wnt Signaling Activates YAP/TAZ. Cell. 2015;162:780-794.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 566]  [Cited by in RCA: 556]  [Article Influence: 55.6]  [Reference Citation Analysis (0)]
32.  Feng M, Gao W, Wang R, Chen W, Man YG, Figg WD, Wang XW, Dimitrov DS, Ho M. Therapeutically targeting glypican-3 via a conformation-specific single-domain antibody in hepatocellular carcinoma. Proc Natl Acad Sci U S A. 2013;110:E1083-E1091.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 128]  [Cited by in RCA: 152]  [Article Influence: 12.7]  [Reference Citation Analysis (0)]
33.  Sohn BH, Shim JJ, Kim SB, Jang KY, Kim SM, Kim JH, Hwang JE, Jang HJ, Lee HS, Kim SC, Jeong W, Kim SS, Park ES, Heo J, Kim YJ, Kim DG, Leem SH, Kaseb A, Hassan MM, Cha M, Chu IS, Johnson RL, Park YY, Lee JS. Inactivation of Hippo Pathway Is Significantly Associated with Poor Prognosis in Hepatocellular Carcinoma. Clin Cancer Res. 2016;22:1256-1264.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 74]  [Cited by in RCA: 97]  [Article Influence: 9.7]  [Reference Citation Analysis (0)]
34.  Fleming BD, Ho M. Glypican-3 Targeting Immunotoxins for the Treatment of Liver Cancer. Toxins (Basel). 2016;8:274.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 19]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
35.  Guo X, Zhao Y, Yan H, Yang Y, Shen S, Dai X, Ji X, Ji F, Gong XG, Li L, Bai X, Feng XH, Liang T, Ji J, Chen L, Wang H, Zhao B. Single tumor-initiating cells evade immune clearance by recruiting type II macrophages. Genes Dev. 2017;31:247-259.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 139]  [Cited by in RCA: 226]  [Article Influence: 28.3]  [Reference Citation Analysis (0)]
36.  Acevedo VD, Ittmann M, Spencer DM. Paths of FGFR-driven tumorigenesis. Cell Cycle. 2009;8:580-588.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 97]  [Cited by in RCA: 112]  [Article Influence: 7.0]  [Reference Citation Analysis (0)]
37.  Lai JP, Sandhu DS, Yu C, Han T, Moser CD, Jackson KK, Guerrero RB, Aderca I, Isomoto H, Garrity-Park MM, Zou H, Shire AM, Nagorney DM, Sanderson SO, Adjei AA, Lee JS, Thorgeirsson SS, Roberts LR. Sulfatase 2 up-regulates glypican 3, promotes fibroblast growth factor signaling, and decreases survival in hepatocellular carcinoma. Hepatology. 2008;47:1211-1222.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 165]  [Cited by in RCA: 173]  [Article Influence: 10.2]  [Reference Citation Analysis (0)]
38.  Dwivedi PP, Lam N, Powell BC. Boning up on glypicans--opportunities for new insights into bone biology. Cell Biochem Funct. 2013;31:91-114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 23]  [Cited by in RCA: 25]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
39.  Midorikawa Y, Ishikawa S, Iwanari H, Imamura T, Sakamoto H, Miyazono K, Kodama T, Makuuchi M, Aburatani H. Glypican-3, overexpressed in hepatocellular carcinoma, modulates FGF2 and BMP-7 signaling. Int J Cancer. 2003;103:455-465.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 178]  [Cited by in RCA: 188]  [Article Influence: 8.5]  [Reference Citation Analysis (0)]
40.  Gao W, Kim H, Ho M. Human Monoclonal Antibody Targeting the Heparan Sulfate Chains of Glypican-3 Inhibits HGF-Mediated Migration and Motility of Hepatocellular Carcinoma Cells. PLoS One. 2015;10:e0137664.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 49]  [Cited by in RCA: 60]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
41.  Cheng W, Huang PC, Chao HM, Jeng YM, Hsu HC, Pan HW, Hwu WL, Lee YM. Glypican-3 induces oncogenicity by preventing IGF-1R degradation, a process that can be blocked by Grb10. Oncotarget. 2017;8:80429-80442.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 5]  [Cited by in RCA: 6]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
42.  Wang P, Tong K, Li Y, Li X, Zhang Y, Gu J, Lei P, Yan S, Hu P. The role and mechanism of HIF-1α-mediated glypican-3 secretion in hypoxia-induced tumor progression in hepatocellular carcinoma. Cell Signal. 2024;114:111007.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 4]  [Reference Citation Analysis (0)]
43.  Yao G, Yang Z. Glypican-3 knockdown inhibits the cell growth, stemness, and glycolysis development of hepatocellular carcinoma cells under hypoxic microenvironment through lactylation. Arch Physiol Biochem. 2024;130:546-554.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 5]  [Cited by in RCA: 11]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
44.  Hu R, Zhu Z. ELK1-activated GPC3-AS1/GPC3 axis promotes the proliferation and migration of cervical cancer cells. J Gene Med. 2019;21:e3099.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 12]  [Cited by in RCA: 22]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
45.  Guereño M, Delgado Pastore M, Lugones AC, Cercato M, Todaro L, Urtreger A, Peters MG. Glypican-3 (GPC3) inhibits metastasis development promoting dormancy in breast cancer cells by p38 MAPK pathway activation. Eur J Cell Biol. 2020;99:151096.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 10]  [Cited by in RCA: 25]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
46.  Fernández D, Guereño M, Lago Huvelle MA, Cercato M, Peters MG. Signaling network involved in the GPC3-induced inhibition of breast cancer progression: role of canonical Wnt pathway. J Cancer Res Clin Oncol. 2018;144:2399-2418.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 18]  [Cited by in RCA: 16]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
47.  Chen C, Huang X, Ying Z, Wu D, Yu Y, Wang X, Chen C. Can glypican-3 be a disease-specific biomarker? Clin Transl Med. 2017;6:18.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 13]  [Cited by in RCA: 14]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
48.  Hu H, Quan G, Yang F, Du S, Ding S, Lun Y, Chen Q. MicroRNA-96-5p is negatively regulating GPC3 in the metastasis of papillary thyroid cancer. SAGE Open Med. 2023;11:20503121231205710.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Reference Citation Analysis (0)]
49.  Fei X, Zhang J, Zhao Y, Sun M, Zhao H, Li S. miR-96 promotes invasion and metastasis by targeting GPC3 in non-small cell lung cancer cells. Oncol Lett. 2018;15:9081-9086.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 7]  [Cited by in RCA: 15]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
50.  Zhu AX, Abbas AR, de Galarreta MR, Guan Y, Lu S, Koeppen H, Zhang W, Hsu CH, He AR, Ryoo BY, Yau T, Kaseb AO, Burgoyne AM, Dayyani F, Spahn J, Verret W, Finn RS, Toh HC, Lujambio A, Wang Y. Molecular correlates of clinical response and resistance to atezolizumab in combination with bevacizumab in advanced hepatocellular carcinoma. Nat Med. 2022;28:1599-1611.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 3]  [Cited by in RCA: 352]  [Article Influence: 117.3]  [Reference Citation Analysis (0)]
51.  Thangaraj JL, Coffey M, Lopez E, Kaufman DS. Disruption of TGF-β signaling pathway is required to mediate effective killing of hepatocellular carcinoma by human iPSC-derived NK cells. Cell Stem Cell. 2024;31:1327-1343.e5.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 22]  [Reference Citation Analysis (0)]
52.  Li D, Wang Y, Shi C, Fu S, Sun YF, Li C. Targeting GPC3(high) cancer-associated fibroblasts sensitizing the PD-1 blockage therapy in gastric cancer. Ann Med. 2023;55:2189295.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 19]  [Reference Citation Analysis (0)]
53.  Fayn S, King AP, Gutsche NT, Duan Z, Buffington J, Olkowski CP, Fu Y, Hong J, Sail D, Baidoo KE, Swenson RE, Cheloha RW, Ho M, Choyke PL, Escorcia FE. Site-Specifically Conjugated Single-Domain Antibody Successfully Identifies Glypican-3-Expressing Liver Cancer by Immuno-PET. J Nucl Med. 2023;64:1017-1023.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 30]  [Reference Citation Analysis (0)]
54.  Mo C, Sun P, Liang H, Chen Z, Wang M, Fu L, Huang S, Tang G. Synthesis and preclinical evaluation of a novel probe [(18)F]AlF-NOTA-IPB-GPC3P for PET imaging of GPC3 positive tumor. Bioorg Chem. 2024;147:107352.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
55.  Chen L, Cheng S, Zhu D, Bao G, Wang Z, Deng X, Liu X, Ma X, Zhao J, Zhu L, Zhu X. Synthesis and Preclinical Evaluation of Dual-Specific Probe Targeting Glypican-3 and Prostate-Specific Membrane Antigen for Hepatocellular Carcinoma PET Imaging. Mol Pharm. 2025;22:209-220.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
56.  Li SQ, Yang CX, Wu CM, Cui JJ, Wang JN, Yin XP. Prediction of glypican-3 expression in hepatocellular carcinoma using multisequence magnetic resonance imaging-based histology nomograms. Quant Imaging Med Surg. 2024;14:4436-4449.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 1]  [Reference Citation Analysis (0)]
57.  Sun S, Xiao S, Jiang Z, Xiao J, He Q, Wang M, Fan Y. Radiomic Analysis of Contrast-Enhanced CT Predicts Glypican 3-Positive Hepatocellular Carcinoma. Curr Med Imaging. 2024;20:e15734056277475.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
58.  Li G, Guo F, Liang J, Wan B, Liang J, Zhou Z. Sandwich-type supersensitive electrochemical aptasensor of glypican-3 based on PrGO-Hemin-PdNP and AuNP@PoPD. Mikrochim Acta. 2024;191:340.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
59.  Xiao X, Lin X, Ting CL, Huang X, Zeng B, Liu T, Zeng T. Extraction-free, immuno-RPA-CRISPR/Cas13a-based one-pot detection of glypican-3 directly from extracellular vesicles. Anal Chim Acta. 2024;1297:342385.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Reference Citation Analysis (0)]
60.  Li J, Su J, Li M, Wu Y, Chen H, Fu X, Yao H, Chen J, Liu Y, Zan J. Rapid evaluation of hepatocellular carcinoma by detecting plasma exosomes with time-resolved fluorescence immunochromatographic test strips. Mikrochim Acta. 2024;192:39.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
61.  Jiang P, Zhan Z, Peng Y, Wu C, Wang Y, Wu L, Shi S, Ying B, Wei Y, Chen P, Chen J. Steric Hindrance-Mediated Enzymatic Reaction Enable Homogeneous Dual Fluorescence Indicators Aptasensing of Hepatocellular Carcinoma CTCs. Anal Chem. 2024;96:10705-10713.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
62.  Abou-Alfa GK, Puig O, Daniele B, Kudo M, Merle P, Park JW, Ross P, Peron JM, Ebert O, Chan S, Poon TP, Colombo M, Okusaka T, Ryoo BY, Minguez B, Tanaka T, Ohtomo T, Ukrainskyj S, Boisserie F, Rutman O, Chen YC, Xu C, Shochat E, Jukofsky L, Reis B, Chen G, Di Laurenzio L, Lee R, Yen CJ. Randomized phase II placebo controlled study of codrituzumab in previously treated patients with advanced hepatocellular carcinoma. J Hepatol. 2016;65:289-295.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 73]  [Cited by in RCA: 90]  [Article Influence: 10.0]  [Reference Citation Analysis (0)]
63.  Carrasquillo JA, O'Donoghue JA, Beylergil V, Ruan S, Pandit-Taskar N, Larson SM, Smith-Jones PM, Lyashchenko SK, Ohishi N, Ohtomo T, Abou-Alfa GK. I-124 codrituzumab imaging and biodistribution in patients with hepatocellular carcinoma. EJNMMI Res. 2018;8:20.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 26]  [Article Influence: 3.7]  [Reference Citation Analysis (0)]
64.  Fang Y, Ma H, Zhang X, Zhang P, Li Y, He S, Sheng C, Dong G. Smart glypican-3-targeting peptide-chlorin e6 conjugates for targeted photodynamic therapy of hepatocellular carcinoma. Eur J Med Chem. 2024;264:116047.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
65.  Ishiguro T, Sano Y, Komatsu SI, Kamata-Sakurai M, Kaneko A, Kinoshita Y, Shiraiwa H, Azuma Y, Tsunenari T, Kayukawa Y, Sonobe Y, Ono N, Sakata K, Fujii T, Miyazaki Y, Noguchi M, Endo M, Harada A, Frings W, Fujii E, Nanba E, Narita A, Sakamoto A, Wakabayashi T, Konishi H, Segawa H, Igawa T, Tsushima T, Mutoh H, Nishito Y, Takahashi M, Stewart L, ElGabry E, Kawabe Y, Ishigai M, Chiba S, Aoki M, Hattori K, Nezu J. An anti-glypican 3/CD3 bispecific T cell-redirecting antibody for treatment of solid tumors. Sci Transl Med. 2017;9:eaal4291.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 117]  [Cited by in RCA: 120]  [Article Influence: 15.0]  [Reference Citation Analysis (0)]
66.  Sano Y, Azuma Y, Tsunenari T, Kayukawa Y, Shinozuka J, Fujii E, Amano J, Nishito Y, Maruyama T, Kinoshita Y, Sakamoto Y, Yoshida A, Miyazaki Y, Sato Y, Teramoto-Seida C, Ishiguro T, Tanaka T, Kitazawa T, Endo M. Combination of T cell-redirecting bispecific antibody ERY974 and chemotherapy reciprocally enhances efficacy against non-inflamed tumours. Nat Commun. 2022;13:5265.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 12]  [Reference Citation Analysis (0)]
67.  Komatsu SI, Kayukawa Y, Miyazaki Y, Kaneko A, Ikegami H, Ishiguro T, Nakamura M, Frings W, Ono N, Sakata K, Fujii T, Kishishita S, Kitazawa T, Endo M, Sano Y. Determination of starting dose of the T cell-redirecting bispecific antibody ERY974 targeting glypican-3 in first-in-human clinical trial. Sci Rep. 2022;12:12312.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 2]  [Cited by in RCA: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
68.  Steffin D, Ghatwai N, Montalbano A, Rathi P, Courtney AN, Arnett AB, Fleurence J, Sweidan R, Wang T, Zhang H, Masand P, Maris JM, Martinez D, Pogoriler J, Varadarajan N, Thakkar SG, Lyon D, Lapteva N, Zhuyong M, Patel K, Lopez-Terrada D, Ramos CA, Lulla P, Armaghany T, Grilley BJ, Gottschalk S, Dotti G, Metelitsa LS, Heslop HE, Brenner MK, Sumazin P, Heczey A. Interleukin-15-armoured GPC3 CAR T cells for patients with solid cancers. Nature. 2025;637:940-946.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 22]  [Cited by in RCA: 37]  [Article Influence: 37.0]  [Reference Citation Analysis (0)]
69.  Pilipow K, Roberto A, Roederer M, Waldmann TA, Mavilio D, Lugli E. IL15 and T-cell Stemness in T-cell-Based Cancer Immunotherapy. Cancer Res. 2015;75:5187-5193.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 64]  [Cited by in RCA: 101]  [Article Influence: 10.1]  [Reference Citation Analysis (0)]
70.  Davis MR, Zhu Z, Hansen DM, Bai Q, Fang Y. The role of IL-21 in immunity and cancer. Cancer Lett. 2015;358:107-114.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 78]  [Cited by in RCA: 118]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
71.  Batra SA, Rathi P, Guo L, Courtney AN, Fleurence J, Balzeau J, Shaik RS, Nguyen TP, Wu MF, Bulsara S, Mamonkin M, Metelitsa LS, Heczey A. Glypican-3-Specific CAR T Cells Coexpressing IL15 and IL21 Have Superior Expansion and Antitumor Activity against Hepatocellular Carcinoma. Cancer Immunol Res. 2020;8:309-320.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 121]  [Cited by in RCA: 178]  [Article Influence: 35.6]  [Reference Citation Analysis (0)]
72.  Lu LL, Xiao SX, Lin ZY, Bai JJ, Li W, Song ZQ, Zhou YH, Lu B, Wu WZ. GPC3-IL7-CCL19-CAR-T primes immune microenvironment reconstitution for hepatocellular carcinoma therapy. Cell Biol Toxicol. 2023;39:3101-3119.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 8]  [Cited by in RCA: 25]  [Article Influence: 12.5]  [Reference Citation Analysis (0)]
73.  Sun RX, Liu YF, Sun YS, Zhou M, Wang Y, Shi BZ, Jiang H, Li ZH. GPC3-targeted CAR-T cells expressing GLUT1 or AGK exhibit enhanced antitumor activity against hepatocellular carcinoma. Acta Pharmacol Sin. 2024;45:1937-1950.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 16]  [Cited by in RCA: 1]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
74.  Gerhardt L, Hong MMY, Yousefi Y, Figueredo R, Maleki Vareki S. IL-12 and IL-27 Promote CD39 Expression on CD8+ T Cells and Differentially Regulate the CD39+CD8+ T Cell Phenotype. J Immunol. 2023;210:1598-1606.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 2]  [Cited by in RCA: 10]  [Article Influence: 5.0]  [Reference Citation Analysis (0)]
75.  Zou F, Tan J, Liu T, Liu B, Tang Y, Zhang H, Li J. The CD39(+) HBV surface protein-targeted CAR-T and personalized tumor-reactive CD8(+) T cells exhibit potent anti-HCC activity. Mol Ther. 2021;29:1794-1807.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 11]  [Cited by in RCA: 59]  [Article Influence: 14.8]  [Reference Citation Analysis (0)]
76.  Zou F, Wei J, Zhuang J, Liu Y, Tan J, Huang X, Liu T. Moderate expression of CD39 in GPC3-CAR-T cells shows high efficacy against hepatocellular carcinoma. Front Med. 2024;18:708-720.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
77.  Sun L, Gao F, Gao Z, Ao L, Li N, Ma S, Jia M, Li N, Lu P, Sun B, Ho M, Jia S, Ding T, Gao W. Shed antigen-induced blocking effect on CAR-T cells targeting Glypican-3 in Hepatocellular Carcinoma. J Immunother Cancer. 2021;9:e001875.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 37]  [Cited by in RCA: 74]  [Article Influence: 18.5]  [Reference Citation Analysis (0)]
78.  Majzner RG, Mackall CL. Tumor Antigen Escape from CAR T-cell Therapy. Cancer Discov. 2018;8:1219-1226.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 412]  [Cited by in RCA: 756]  [Article Influence: 108.0]  [Reference Citation Analysis (0)]
79.  Cao G, Zhang G, Liu M, Liu J, Wang Q, Zhu L, Wan X. GPC3-targeted CAR-T cells secreting B7H3-targeted BiTE exhibit potent cytotoxicity activity against hepatocellular carcinoma cell in the in vitro assay. Biochem Biophys Rep. 2022;31:101324.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 10]  [Reference Citation Analysis (0)]
80.  Chen S, Gong F, Liu S, Xie Y, Ye X, Lin X, Wang X, Zheng Q, Liu Q, Sun Y. IL-21- and CXCL9-engineered GPC3-specific CAR-T cells combined with PD-1 blockade enhance cytotoxic activities against hepatocellular carcinoma. Clin Exp Med. 2024;24:204.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Reference Citation Analysis (0)]
81.  Makkouk A, Yang XC, Barca T, Lucas A, Turkoz M, Wong JTS, Nishimoto KP, Brodey MM, Tabrizizad M, Gundurao SRY, Bai L, Bhat A, An Z, Abbot S, Satpayev D, Aftab BT, Herrman M. Off-the-shelf Vδ1 gamma delta T cells engineered with glypican-3 (GPC-3)-specific chimeric antigen receptor (CAR) and soluble IL-15 display robust antitumor efficacy against hepatocellular carcinoma. J Immunother Cancer. 2021;9:e003441.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 24]  [Cited by in RCA: 126]  [Article Influence: 31.5]  [Reference Citation Analysis (0)]
82.  Shi X, Ding J, Zheng Y, Wang J, Sobhani N, Neeli P, Wang G, Zheng J, Chai D. HMGB1/GPC3 dual targeting vaccine induces dendritic cells-mediated CD8(+)T cell immune response and elicits potential therapeutic effect in hepatocellular carcinoma. iScience. 2023;26:106143.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in RCA: 9]  [Reference Citation Analysis (0)]
83.  Xue J, Suarez JS, Minaai M, Li S, Gaudino G, Pass HI, Carbone M, Yang H. HMGB1 as a therapeutic target in disease. J Cell Physiol. 2021;236:3406-3419.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 48]  [Cited by in RCA: 200]  [Article Influence: 40.0]  [Reference Citation Analysis (0)]
84.  Liu Y, Yan W, Tohme S, Chen M, Fu Y, Tian D, Lotze M, Tang D, Tsung A. Hypoxia induced HMGB1 and mitochondrial DNA interactions mediate tumor growth in hepatocellular carcinoma through Toll-like receptor 9. J Hepatol. 2015;63:114-121.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 136]  [Cited by in RCA: 197]  [Article Influence: 19.7]  [Reference Citation Analysis (0)]
85.  Bianchi ME, Manfredi AA. High-mobility group box 1 (HMGB1) protein at the crossroads between innate and adaptive immunity. Immunol Rev. 2007;220:35-46.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Cited by in Crossref: 442]  [Cited by in RCA: 478]  [Article Influence: 28.1]  [Reference Citation Analysis (0)]
86.  Llovet JM, Pinyol R, Yarchoan M, Singal AG, Marron TU, Schwartz M, Pikarsky E, Kudo M, Finn RS. Adjuvant and neoadjuvant immunotherapies in hepatocellular carcinoma. Nat Rev Clin Oncol. 2024;21:294-311.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 152]  [Cited by in RCA: 142]  [Article Influence: 142.0]  [Reference Citation Analysis (0)]
87.  Cappuyns S, Corbett V, Yarchoan M, Finn RS, Llovet JM. Critical Appraisal of Guideline Recommendations on Systemic Therapies for Advanced Hepatocellular Carcinoma: A Review. JAMA Oncol. 2024;10:395-404.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in Crossref: 82]  [Cited by in RCA: 75]  [Article Influence: 75.0]  [Reference Citation Analysis (0)]
88.  Wu CS, Chien YC, Yen CJ, Wu JY, Bai LY, Yu YL. EZH2-mediated epigenetic silencing of tumor-suppressive let-7c/miR-99a cluster by hepatitis B virus X antigen enhances hepatocellular carcinoma progression and metastasis. Cancer Cell Int. 2023;23:199.  [RCA]  [PubMed]  [DOI]  [Full Text]  [Full Text (PDF)]  [Cited by in RCA: 8]  [Reference Citation Analysis (0)]