GPC3 consists of a 70-kDa core protein anchored to the cell membrane via a glycosylphosphatidylinositol (GPI) linkage. It contains 14 conserved cysteine residues and heparan sulfate (HS) chains, which mediate interactions with growth factors and signaling proteins . The GPC3 gene produces four isoforms through alternative splicing:
The protein’s N-terminal subunit (~40 kDa) and C-terminal subunit (~30 kDa) interact with Wnt, Yap, and other signaling pathways .
GPC3 modulates Wnt/β-catenin and Yap signaling, influencing cell proliferation and survival. Its HS chains bind growth factors like FGF2 and HGF, while its core protein interacts with receptors such as Frizzled (FZD) .
Hepatocellular Carcinoma (HCC): Overexpressed in >50% of HCC cases, GPC3 promotes tumor growth via Wnt activation and Yap signaling .
Diagnostic Utility: GPC3 immunostaining (e.g., YP7/hYP7 antibodies) differentiates HCC from non-neoplastic liver tissue .
Other Cancers: Expressed in melanoma, ovarian clear-cell carcinomas, and Wilms’ tumor .
GPC3’s restricted expression in normal tissues makes it an attractive target for precision therapies.
Immunotoxins and ADCs: Fusion proteins (e.g., HN3-Pseudomonas toxin) and antibody-drug conjugates (e.g., hYP7-linker-drug) are under development to enhance tumor-specific cytotoxicity .
CAR-T Cells: Third-generation CAR-T cells targeting GPC3 (e.g., GC33, hYP7) show potent in vivo cytotoxicity in HCC models .
Peptide Vaccines: GPC3-derived peptides induce cytotoxic T-lymphocyte (CTL) responses, with clinical trials demonstrating prolonged recurrence-free survival in HCC patients .
Challenges: Tumor heterogeneity and antigen loss (e.g., GPC3-negative recurrence) limit monotherapy efficacy .
Glypican 3, Intestinal Protein OCI-5, Glypican Proteoglycan 3, GTR2-2, MXR7, Heparan Sulphate Proteoglycan, Secreted Glypican-3, Glypican-3, OCI-5, SGBS1, DGSX, SGBS, SDYS, OCI5, SGB, GPC3.
Sf9, Baculovirus cells.
ADPQPPPPPP DATCHQVRSF FQRLQPGLKW VPETPVPGSD LQVCLPKGPT CCSRKMEEKY QLTARLNMEQ LLQSASMELK FLIIQNAAVF QEAFEIVVRH AKNYTNAMFK NNYPSLTPQA FEFVGEFFTD VSLYILGSDI NVDDMVNELF DSLFPVIYTQ LMNPGLPDSA LDINECLRGA RRDLKVFGNF PKLIMTQVSK SLQVTRIFLQ ALNLGIEVIN TTDHLKFSKD CGRMLTRMWY CSYCQGLMMV KPCGGYCNVV MQGCMAGVVE IDKYWREYIL SLEELVNGMY RIYDMENVLL GLFSTIHDSI QYVQKNAGKL TTTIGKLCAH SQQRQYRSAY YPEDLFIDKK VLKVAHVEHE ETLSSRRREL IQKLKSFISF YSALPGYICS HSPVAENDTL CWNGQELVER YSQKAARNGM KNQFNLHELK MKGPEPVVSQ IIDKLKHINQ LLRTMSMPKG RVLDKNLDEE GFESGDCGDD EDECIGGSGD GMIKVKNQLR FLAELAYDLD VDDAPGNSQQ ATPKDNEIST FHNLGNVHHH HHHH.
GPC3 is a glycophosphatidylinositol (GPI)-anchored cell surface heparan sulfate proteoglycan. The protein core consists of two subunits: an N-terminal subunit (~40 kDa) and a C-terminal subunit (~30 kDa) . The full protein has a predicted molecular weight of approximately 21.5 kDa for specific fragments like the Arg438-Asn554 region . GPC3 undergoes post-translational modifications including the addition of heparan sulfate glycan chains, which are important for some of its biological functions, particularly in growth factor binding .
GPC3 is encoded by the GPC3 gene in humans, with several synonyms including DGSX, GTR2-2, MXR7, OCI-5, SGBS, and SGBS1 . It is classified as a membrane-bound heparan sulfate proteoglycan that plays crucial roles in cell signaling and growth regulation.
GPC3 shows a distinct temporospatial expression pattern:
Expressed during early development in human embryo, fetus, and placental tissues
Also expressed to varying degrees in other cancers including melanoma, ovarian clear-cell carcinomas, yolk sac tumors, neuroblastoma, hepatoblastoma, and Wilms' tumor cells
This oncofetal expression pattern makes GPC3 an interesting candidate as both a biomarker and therapeutic target for cancer, particularly HCC.
GPC3 is highly expressed in HCC but not in normal liver tissue, making it a specific marker for this cancer type . Evidence suggests that GPC3 is actively involved in HCC tumorigenesis through several mechanisms:
Stimulation of canonical Wnt signaling by forming complexes with Wnt molecules
Possible roles in other signaling pathways including TGF-β2, HGF, and Yap
The specificity of GPC3 expression in HCC (versus cholangiocarcinoma or normal liver) has established it as a promising diagnostic marker and therapeutic target for HCC .
Simpson-Golabi-Behmel Syndrome (SGBS) is a rare X-linked overgrowth disorder characterized by pre- and postnatal overgrowth of multiple tissues and organs, and an increased risk for developing embryonic tumors . This syndrome is caused by loss-of-function mutations in the GPC3 gene .
The phenotype of SGBS patients aligns with observations from GPC3 knockout mice, which also show overgrowth phenotypes . These findings indicate that GPC3 normally functions to regulate cell survival and inhibit cell proliferation during development . The association between GPC3 dysfunction and overgrowth syndromes highlights its critical role in growth regulation, providing insight into how its dysregulation might contribute to cancer development.
While GPC3 is most prominently associated with HCC, several other cancer types show altered GPC3 expression:
In LUAD, approximately 1.4% to 2.2% of patients exhibit copy number amplifications in GPC3 . The significance of GPC3 in these various cancer types may differ, with evidence suggesting its involvement in specific signaling pathways relevant to each cancer type.
Several recombinant GPC3 proteins are available for research purposes, including:
When selecting a recombinant protein for research, important considerations include:
Expression system (e.g., HEK293 cells for proper post-translational modifications)
Protein fragment vs. full-length protein
Activity validation (e.g., binding assays with known antibodies)
Endotoxin levels (important for cell-based and in vivo experiments)
For binding studies, researchers should note that different regions of GPC3 interact with different partners, with the C-terminal region being particularly important for some antibody recognition .
Several experimental approaches can be employed to investigate GPC3's interactions with signaling pathways:
Protein-protein interaction studies:
Co-immunoprecipitation to detect direct binding between GPC3 and pathway components
Surface plasmon resonance (SPR) for quantitative binding kinetics
Proximity ligation assays for visualizing interactions in situ
Signaling pathway activation assays:
Luciferase reporter assays for Wnt/β-catenin pathway activation
Western blotting for phosphorylation of downstream effectors
Transcriptional profiling of pathway target genes
Functional studies with GPC3 variants:
When designing these experiments, researchers should consider the cellular context, as GPC3's effects may vary between different cell types and developmental stages.
GPC3 undergoes endoproteolytic processing by proprotein convertases, which is required for its modulation of Wnt signaling . This processing results in two subunits (N-terminal ~40 kDa and C-terminal ~30 kDa) that remain associated through disulfide bonds .
Methodological approaches to study this processing include:
Western blotting with antibodies specific to different domains to monitor processing
Site-directed mutagenesis of cleavage sites to generate non-cleavable variants
Inhibition of proprotein convertases to assess processing-dependent functions
Comparison of signaling activities between processed and unprocessed forms
Understanding this processing is critical for designing therapeutic strategies and interpreting experimental results, as antibodies targeting different epitopes may recognize processed or unprocessed forms differently.
While GPC3 is primarily studied for its direct roles in signaling, emerging evidence suggests potential roles in cancer immune interactions. Researchers investigating this aspect should consider:
GPC3's impact on tumor microenvironment:
Analysis of immune cell infiltration in GPC3-expressing versus non-expressing tumors
Correlation between GPC3 expression and immune checkpoint molecule expression
Assessment of cytokine profiles in the presence of GPC3
Immunotherapeutic targeting approaches:
The humanized anti-GPC3 antibodies hYP7 and hYP9.1b have been shown to induce both ADCC and CDC in GPC3-positive cancer cells but not in GPC3-negative cells, demonstrating the potential of GPC3 as an immunotherapeutic target .
The development of therapeutic anti-GPC3 antibodies involves several critical steps:
Generation of high-affinity mouse monoclonal antibodies:
Humanization strategies:
Functional validation:
Testing binding affinity to cell surface GPC3
Evaluation of ADCC and CDC activities
Assessment of in vivo antitumor efficacy in xenograft models
For example, humanized antibodies hYP7 and hYP9.1b have shown specific binding to GPC3-positive cells with EC50 values of 0.7 nM and 0.4 nM respectively, while demonstrating no binding to GPC3-negative cells . These antibodies have also demonstrated effective ADCC and CDC activities against GPC3-expressing cancer cells .
Methodological approaches for evaluating GPC3-targeted therapies include:
In vitro assessments:
Cell viability and proliferation assays in GPC3-positive versus GPC3-negative cell lines
Pathway inhibition assays (e.g., Wnt reporter assays)
ADCC and CDC assays using various effector cell sources
3D organoid models for improved physiological relevance
In vivo evaluations:
Biomarker strategies:
Monitoring target engagement through biopsies
Assessing pathway inhibition through surrogate markers
Evaluation of immune infiltration changes in immunotherapeutic approaches
For example, the hYP7 antibody has demonstrated inhibition of HCC xenograft tumor growth in nude mice, providing validation for its potential therapeutic efficacy .
GPC3 is composed of a core protein to which heparan sulfate chains are attached. The protein undergoes endoproteolytic processing by proprotein convertases, resulting in multiple peptides that remain associated via disulfide bonds . The predicted molecular mass of GPC3 is approximately 61.6 kDa, but it appears as a smear with an apparent molecular mass of 60-100 kDa under non-reducing conditions in SDS-PAGE .
GPC3 is involved in modulating the activity of growth factors, such as fibroblast growth factors (FGFs), by binding to them and influencing their interaction with their receptors . This interaction is crucial for various cellular processes, including proliferation, differentiation, and migration.
GPC3 is highly expressed in hepatocellular carcinoma (HCC) and is rarely found in normal liver tissues, making it a valuable diagnostic and therapeutic target for HCC . Its overexpression in HCC and other cancers, such as melanoma, highlights its potential as a biomarker for cancer diagnosis and treatment .
Recombinant human GPC3 is produced using various expression systems, including mouse myeloma cell lines (NS0-derived). The recombinant protein is often tagged with a His-tag for purification purposes and is available in both carrier-free and carrier-containing formulations . The carrier protein, typically bovine serum albumin (BSA), enhances protein stability and shelf-life .
The recombinant GPC3 protein is used in research to study its binding interactions, particularly with growth factors like FGF. It is also utilized in the development of diagnostic and therapeutic agents targeting GPC3-expressing tumors .
Recent studies have focused on developing GPC3-specific binding peptides for imaging and therapeutic purposes. For instance, a two-step phage display screening approach identified a GPC3-specific binding peptide, TJ12P2, which shows promise as a PET imaging probe for accurate HCC diagnosis . Such advancements underscore the potential of GPC3 in cancer research and treatment.