GPC3 Antibody

Shipped with Ice Packs
In Stock

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Our standard lead time for dispatching products is 1-3 working days after receiving your order. Delivery times may vary depending on the purchasing method and location. Please consult your local distributors for specific delivery timeframes.
Synonyms
DGSX antibody; Glypican proteoglycan 3 antibody; Glypican-3 [Precursor] antibody; Gpc3 antibody; GPC3_HUMAN antibody; GTR2 2 antibody; GTR2-2 antibody; Heparan sulphate proteoglycan antibody; Intestinal protein OCI 5 antibody; Intestinal protein OCI-5 antibody; MXR7 antibody; OCI 5 antibody; OCI-5 antibody; OCI5 antibody; SDYS antibody; Secreted glypican-3 antibody; SGB antibody; SGBS antibody; SGBS1 antibody
Target Names
Uniprot No.

Target Background

Function
Glypican 3 (GPC3) is a cell surface proteoglycan that carries heparan sulfate. It plays a crucial role in regulating various signaling pathways, including hedgehog, Wnt, and non-canonical Wnt pathways.

GPC3 negatively regulates the hedgehog signaling pathway by competing with the hedgehog receptor PTC1 for binding to hedgehog proteins. This interaction leads to internalization and lysosomal degradation of the complex.

Conversely, GPC3 positively regulates the canonical Wnt signaling pathway by binding to the Wnt receptor Frizzled and facilitating the binding of Frizzled to Wnt ligands. GPC3 also positively regulates the non-canonical Wnt signaling pathway.

Furthermore, GPC3 interacts with CD81, decreasing its availability for binding to the transcriptional repressor HHEX. This results in nuclear translocation of HHEX and subsequent transcriptional repression. GPC3 also exhibits dipeptidyl peptidase activity, inhibiting DPP4.

GPC3 is essential for limb patterning and skeletal development by regulating cellular responses to BMP4. It modulates the effects of growth factors BMP2, BMP7, and FGF7 on renal branching morphogenesis. GPC3 is also crucial for coronary vascular development and regulates cell movements during gastrulation.
Gene References Into Functions
  1. The areas under the receiver operating curve (AUROC) value, sensitivity, and specificity of GPC3 for hepatoblastoma (HB) pretreatment group versus all controls were all significantly lower than those of alpha-fetoprotein (AFP). PMID: 28378832
  2. GPC3 operates through a complex molecular signaling network. From the balance of these interactions, the inhibition of breast metastatic spread induced by GPC3 emerges. PMID: 30267212
  3. Its surface is modified with anti-GPC3 antibody. PMID: 29916268
  4. Data suggest that transcriptionally targeted delivery of transgene in HCC cells can be achieved using the GPC3 promoter, with limited toxicity to normal liver cells. PMID: 29563582
  5. High GPC3 expression is associated with Hepatocellular Carcinoma. PMID: 28429175
  6. Study demonstrated that GPC3 expression is inversely associated with glucose metabolism, suggesting that GPC3 may play a role in regulating glucose metabolism in hepatocellular carcinoma. PMID: 29398870
  7. The intravenous injection of SF-PL/siGPC3 into nude mice bearing subcutaneous human HepG2 xenografts effectively inhibited tumor growth and also increased the survival rates of animals. These results highlight the potential of the PEI-modified liposomal nanomedicine carrying SF and siGPC3 to improve Hepatocellular carcinoma treatment. PMID: 29106433
  8. Invasive hepatocellular carcinoma (HCC) samples and HCC cell lines with high metastatic potential exhibited higher MXR7 expression. Overexpression of MXR7 promoted epithelial-mesenchymal transition (EMT) progress, and MXR7 depletion repressed the EMT phenotype. Human MXR7 protein is a mediator of EMT and metastasis in HCC. PMID: 28812296
  9. Overexpression of GPC3 was significantly associated with poor prognosis in patients with hepatocellular carcinoma. PMID: 29901640
  10. These data show that glycanation and convertase maturation are not required for soluble mutant GPC3 to inhibit hepatocellular carcinoma cell proliferation. PMID: 29345911
  11. Data indicate that several microRNAs target the oncogenic functions of glypican-3 (GPC3). PMID: 28476031
  12. The presence of GPC3 distinguishes aggressive from non-aggressive odontogenic tumors. PMID: 27647326
  13. GPC3 is a potential metastasis suppressor gene and holds value as a prognostic marker in gastric cancer. PMID: 27259271
  14. In this study, we systematically evaluated a series of CAR constructs targeting glypican-3 (GPC3), which is selectively expressed on several solid tumors. We compared GPC3-specific CARs that encoded CD3zeta (Gz) alone or with costimulatory domains derived from CD28 (G28z), 4-1BB (GBBz), or CD28 and 4-1BB (G28BBz). PMID: 27530312
  15. Data indicate that glypican-3 (GPC3) is an important regulator of epithelial-mesenchymal transition (EMT) in breast cancer and a potential target for procedures against breast cancer metastasis. PMID: 27507057
  16. Glypican-3 overexpression in Wilms tumor correlates with poor overall survival. PMID: 28432433
  17. Glypican-3 plays a role in HBV-related hepatocellular carcinoma. PMID: 27286460
  18. MOSPD1 is a possible candidate gene for DORV, probably in combination with GPC3. Further studies of the combined functions of MOSPD1 and GPC3 are needed, and identification of additional patients with MOSPD1 and GPC3 duplication should be pursued. PMID: 28636109
  19. Glypican-3 is correlated with the clinical malignant behavior of hepatocellular carcinoma, and its phenotype changes from positive to negative during tumor cell differentiation. PMID: 28087980
  20. The diagnostic sensitivity for hepatocellular carcinoma increased to 72.8% (206 of the 283) when glypican 3 was combined with alpha-fetoprotein. PMID: 26370140
  21. The lncRNA glypican 3 antisense transcript 1 (GPC3-AS1) has been reported to be a potential biomarker for hepatocellular carcinoma (HCC) screening. We observed a significant upregulation of GPC3-AS1 in HCC. Increased expression of GPC3-AS1 was associated with alpha-fetoprotein, tumor size, microvascular invasion, encapsulation, Barcelona Clinic Liver Cancer stage, and worse prognosis of HCC patients. PMID: 27573079
  22. The study provides the first evidence that GPC3 can modulate the PCSK9 extracellular activity as a competitive binding partner to the LDLR in HepG2 cells. PMID: 27758865
  23. By subsequent Sanger sequencing of genomic DNA, we could map the chromosomal break points to define a deletion size of 43,617 bp including exons 5 and 6 of the GPC3 gene. PMID: 28371070
  24. This is the first study in which the optimal HLA-A*0201 GPC3 epitopes were screened from a large number of candidates predicted by three software. The optimized HLA-A*0201 GPC3 peptides will provide new epitope candidates for hepatocellular carcinoma (HCC) immunotherapy. PMID: 27102087
  25. GPC3 and KRT19 overexpression are associated with carcinogenesis, progression, and poor prognosis in patients with PDAC and a valuable biomarker for diagnosis of PDAC. PMID: 27689616
  26. The clinical implication of GPC3 detection and targeting in the management of patients with hepatocellular carcinoma. Review. PMID: 26755876
  27. Glypican 3 expression showed a significant difference between endometrioid endometrial carcinoma and serous endometrial carcinoma, and it was significantly correlated with tumor grade, stage, and myometrial invasion. PMID: 26722522
  28. Data show that notum and glypican-1 and glypican-3 gene expression during colorectal cancer (CRC) development and present evidence to suggest them as potential new biomarkers of CRC pathogenesis. PMID: 26517809
  29. GPC3 expression was measured in hepatocellular carcinoma at different stages and correlated with prognosis. CK19+/GPC3+ HCC has the highest risk of intrahepatic metastasis, microvascular invasion, regional lymph node involvement, and distant metastasis. PMID: 26977595
  30. Review: Glypican-3 is a highly specific biomarker for the diagnosis of hepatocellular carcinoma and a promising therapeutic target. PMID: 26256079
  31. In South Korean hepatocellular carcinoma patients, GPC3 expression was more frequent in hepatocellular carcinoma with aggressive features, but it was not an independent prognostic biomarker. PMID: 26764243
  32. In this meta-analysis, GPC3 was found to be acceptable as a serum marker for the diagnosis of hepatocellular carcinoma. PMID: 26502856
  33. GPC3 may be a candidate marker for detecting lung squamous cell carcinoma. PMID: 26345955
  34. The study suggests that GPC3 is involved in HCC cell migration and motility through HS chain-mediated cooperation with the HGF/Met pathway, showing how HS targeting has potential therapeutic implications for liver cancer. PMID: 26332121
  35. The potential role of GPC3 in urothelial carcinogenesis warrants further investigation, especially the potential use of Glypican-3 for therapeutic and diagnostic purposes. PMID: 25896897
  36. Downregulation of glypican-3 expression increases migration, invasion, and tumorigenicity of ovarian cancer. PMID: 25967456
  37. GPC3 expression is an independent prognostic factor for postoperative hepatocellular carcinoma. PMID: 25432695
  38. Identify a GPC3-specific T-cell receptor. Expression of this receptor by T cells allows them to recognize and kill GPC3-positive hepatoma cells. PMID: 26052074
  39. High expression of glypican-3 is associated with hepatoblastoma. PMID: 25735325
  40. GPC3 and E-cadherin expressions are not independent prognostic factors in CRC. PMID: 25619476
  41. In HCC patients, sGPC3N levels were significantly increased (mean/median, 405.16/236.19 pg mL(-1) ) compared to healthy controls (p < 0.0001), and 60% of HCC cases (69/115) showed sGPC3N levels that were higher than the upper normal limit. PMID: 25784484
  42. GPC3 contributes to hepatocellular carcinoma progression and metastasis through impacting epithelial-mesenchymal transition of cancer cells, and the effects of GPC3 are associated with ERK activation. PMID: 25572615
  43. Most cases of hepatoblastoma and yolk sac tumor, and some cases of other tumors were found to express GPC3. On the other hand, GPC3 was physiologically expressed during the fetal and neoinfantile period under 1 year of age. PMID: 25344940
  44. OPN, SPINK1, GPC3, and KNPA2 were significantly over-expressed in HCC tissues. These genes may be useful in developing future biomarkers and therapeutic strategies for HCC. PMID: 25862856
  45. Data indicate that zinc-fingers and homeoboxes 2 (ZHX2) suppresses glypican 3 (GPC3) transcription by binding with its core promoter. PMID: 25195714
  46. We propose that the structural changes generated by the lack of cleavage determine a change in the sulfation of the HS chains, and that these hypersulfated chains mediate the interaction of the mutant GPC3 with Ptc. PMID: 25653284
  47. GPC3 is associated with the HCC cell biological behavior. PMID: 25270552
  48. Data indicate that the triple stain of reticulin, glypican-3, and glutamine synthetase is useful in the differentiation of hepatocellular carcinoma, hepatic adenoma, and focal nodular hyperplasia. PMID: 25822763
  49. Data shows that GPC3 gene expression is downregulated in primary clear cell renal cell carcinoma; its overexpression arrest cells in G1 phase suggesting its role as a tumor suppressor in clear cell renal cell carcinoma. PMID: 25168166
  50. This study demonstrated that highly expression of GPC3 could enrich hepatocellular carcinoma -related genes' mRNA expression and positive associate with dysplasia in cirrhotic livers. PMID: 25542894

Show More

Hide All

Database Links

HGNC: 4451

OMIM: 300037

KEGG: hsa:2719

STRING: 9606.ENSP00000377836

UniGene: Hs.644108

Involvement In Disease
Simpson-Golabi-Behmel syndrome 1 (SGBS1)
Protein Families
Glypican family
Subcellular Location
Cell membrane; Lipid-anchor, GPI-anchor; Extracellular side.
Tissue Specificity
Highly expressed in lung, liver and kidney.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : Western Blot (WB)

Sample type: Jurkat whole cell lysate、Hep2B whole cell lysate/Human

Sample dilution: 1:1000

Review: I used CSB-RA009705A0HU antibody to conduct WB detection and detect the cell sample. The sample processing was as follows: After cracking with RIPA lysate, Protein loading Buffer was added to the sample, and the sample was cooked for 20min. The result was as follows: The product met the expectation.

Q&A

What is GPC3 and why is it an important target for antibody development?

GPC3 (Glypican-3) is a cell-surface heparan sulfate proteoglycan that plays a significant role in cancer biology. In humans, the canonical GPC3 protein consists of 580 amino acid residues with a molecular mass of approximately 65.6 kDa and is localized in the cell membrane . GPC3 is highly expressed in most hepatocellular carcinomas and some types of squamous cell carcinomas, while showing limited expression in normal adult tissues . This differential expression pattern makes GPC3 an attractive target for cancer-specific therapies.

GPC3 belongs to the Glypican protein family and is involved in the morphogenesis of anatomical structures and the Wnt signaling pathway, which is frequently dysregulated in cancer . The protein undergoes post-translational modifications including O-glycosylation and protein cleavage. Importantly, overexpression of GPC3 has been significantly associated with poor prognosis in patients with HCC, making it both a prognostic biomarker and therapeutic target .

How do researchers classify different types of GPC3 antibodies?

GPC3 antibodies can be classified based on several key characteristics:

  • Origin and humanization status:

    • Mouse monoclonal antibodies (e.g., YP7, YP8, YP9)

    • Humanized mouse antibodies (e.g., GC33, hYP7)

    • Fully human antibodies (e.g., MDX-1414, HN3)

  • Antibody format:

    • Conventional IgG antibodies

    • Single-chain variable fragments (scFv)

    • Immunotoxin conjugates (e.g., YP7IT, YP9.1IT)

    • Single-domain antibodies (e.g., VH domain antibody HN3)

  • Epitope recognition:

    • N-terminal domain-specific

    • C-terminal domain-specific

    • Conformational epitopes requiring both domains

    • Heparan sulfate (HS) chain-targeting

  • Mechanism of action:

    • Direct inhibition of cell proliferation

    • Antibody-dependent cell-mediated cytotoxicity (ADCC)

    • Complement-dependent cytotoxicity (CDC)

    • Delivery of toxic payloads

This classification framework helps researchers select appropriate antibodies based on their specific experimental or therapeutic objectives.

What are the standard experimental methods for validating GPC3 antibody specificity?

Validating the specificity of GPC3 antibodies requires a multi-faceted approach:

  • Cell-based binding assays:

    • Flow cytometry using GPC3-positive cells (e.g., G1) versus GPC3-negative controls (e.g., A431)

    • Determination of binding efficiency and EC₅₀ values

    • Competitive binding assays to confirm epitope specificity

  • Protein interaction studies:

    • ELISA against recombinant GPC3 proteins

    • Western blotting under reducing and non-reducing conditions

    • Immunoprecipitation followed by mass spectrometry

  • Tissue cross-reactivity:

    • Immunohistochemistry on multiple tumor and normal tissue samples

    • Comparison of staining patterns across different cancer types

    • Correlation with GPC3 mRNA expression data

  • Functional validation:

    • GPC3 knockdown studies as negative controls

    • Comparison of antibody effects with genetic silencing

    • Epitope mapping using truncated GPC3 variants

Importantly, researchers should include both conformational and denatured GPC3 in validation studies, as some antibodies like HN3 specifically recognize conformational epitopes present only in the native form of the GPC3 core protein .

How should researchers design experiments to evaluate the therapeutic potential of GPC3 antibodies?

Designing robust experiments to evaluate GPC3 antibodies requires a comprehensive approach encompassing in vitro and in vivo studies:

In vitro assessment:

  • Binding characterization:

    • Determine affinity (Kd) using surface plasmon resonance

    • Assess binding to cell surface GPC3 via flow cytometry

    • Compare EC₅₀ values across different antibody candidates

  • Functional assays:

    • Cell proliferation assays (e.g., MTT, BrdU incorporation)

    • Cell cycle analysis to identify mechanisms (e.g., G1 arrest)

    • Apoptosis assessment using Annexin V/PI staining

  • Immune effector function evaluation:

    • ADCC assays using human PBMCs from multiple donors

    • CDC assays with varying antibody concentrations

    • Target-dependent cytotoxicity confirmation using GPC3+ and GPC3- cell lines

In vivo assessment:

  • Xenograft tumor models:

    • Establish HCC xenografts in immunodeficient mice

    • Compare antibody effects on tumor growth inhibition

    • Evaluate dose-response relationships and scheduling effects

  • Pharmacokinetic/pharmacodynamic studies:

    • Determine antibody half-life and tissue distribution

    • Assess target engagement in tumors

    • Monitor downstream signaling markers (e.g., Yes-associated protein)

  • Toxicity evaluation:

    • Cross-reactivity assessment in normal tissues

    • Monitoring of liver function and hematological parameters

    • Histopathological examination of major organs

Including appropriate controls is critical - researchers should use isotype-matched control antibodies and compare multiple anti-GPC3 antibodies with different epitope specificities to identify optimal therapeutic candidates.

What cell lines and models are most appropriate for studying GPC3 antibodies?

Selecting appropriate experimental models is crucial for meaningful GPC3 antibody research:

Cell line models:

Cell Line CategoryExamplesCharacteristicsApplications
GPC3-positive HCCHep3B, HepG2, Huh-7Endogenous GPC3 expressionTarget engagement, functional studies
GPC3-engineeredG1 (A431 overexpressing GPC3)Controlled GPC3 expressionBinding specificity, mechanism studies
GPC3-negative controlsA431, normal hepatocytesAbsence of GPC3Specificity assessment
GPC3-knockdownMHCC97-H with GPC3 siRNAReduced GPC3 expressionValidation of antibody specificity

Animal models:

  • Xenograft models:

    • Subcutaneous implantation of HCC cells in nude mice

    • Orthotopic liver implantation for more relevant microenvironment

    • Patient-derived xenografts for heterogeneity studies

  • Genetically engineered models:

    • GPC3-inducible expression systems

    • Models with humanized immune components for ADCC studies

    • Liver-specific GPC3 overexpression models

  • Ex vivo systems:

    • Patient-derived tumor slices for antibody penetration studies

    • 3D spheroid cultures to mimic tumor architecture

    • Co-culture systems with immune effector cells

The selection should be guided by the specific research question, with consideration of GPC3 expression levels, species-specific differences in GPC3 structure, and the intended mechanism of antibody action being evaluated.

How can researchers accurately quantify GPC3 expression levels when evaluating antibody efficacy?

Accurate quantification of GPC3 expression is essential for interpreting antibody efficacy data:

  • Protein-level quantification:

    • Flow cytometry for cell surface GPC3 (antibody binding capacity)

    • Western blotting with calibrated standards for total protein

    • ELISA for secreted/shed GPC3 fragments

    • Mass spectrometry for absolute quantification

  • Transcript-level assessment:

    • RT-qPCR with validated reference genes

    • RNA-seq for comprehensive expression profiling

    • In situ hybridization for spatial expression in tissues

    • Single-cell RNA analysis for heterogeneity evaluation

  • Spatial expression analysis:

    • Immunohistochemistry with digital image analysis

    • Multiplexed immunofluorescence for co-expression studies

    • Tissue microarrays for high-throughput screening

    • Correlation between staining intensity and therapeutic response

  • Dynamic expression monitoring:

    • Serial sampling during treatment courses

    • Inducible expression systems for controlled experiments

    • Correlation with treatment response metrics

    • Assessment of isoform switching under treatment pressure

How do different GPC3 antibody formats influence their therapeutic efficacy?

Different antibody formats exhibit distinct properties that affect their therapeutic potential:

Conventional IgG antibodies (e.g., hYP7, hYP9.1b):

  • Engage immune effector functions (ADCC, CDC)

  • Longer half-life in circulation

  • Limited tumor penetration due to size

  • The humanized YP7 antibody (hYP7) demonstrated superior ADCC and CDC activity compared to hYP9.1b in GPC3-positive cancer cells

Immunotoxin conjugates (e.g., YP7IT, YP9.1IT):

  • Direct cytotoxicity without immune system requirement

  • YP9.1IT showed the highest affinity (EC₅₀ = 3 nM) and cytotoxicity (EC₅₀ = 1.9 ng/ml)

  • YP7IT demonstrated strong cytotoxicity (EC₅₀ = 5 ng/ml) despite similar binding as YP8IT

  • Potential for off-target toxicity requires careful epitope selection

Single-domain antibodies (e.g., HN3):

  • Superior tissue penetration

  • Recognition of unique conformational epitopes

  • Direct inhibition of cell proliferation without immune effector functions

  • HN3 binding requires both N- and C-terminal domains of GPC3

Comparative efficacy factors:

  • The binding affinity of therapeutic antibodies ranges from 0.4-10 nM depending on format and epitope

  • Direct growth inhibition vs. immune-mediated killing affects efficacy in different immune contexts

  • Conformational epitope recognition (as with HN3) may provide superior selectivity and functional inhibition

  • Tissue penetration and pharmacokinetics differ substantially between formats

These format-dependent characteristics should guide selection based on the specific therapeutic context, including tumor location, immune status, and target expression pattern.

What are the key challenges in developing anti-GPC3 antibodies with optimal tumor selectivity?

Developing highly selective anti-GPC3 antibodies presents several challenges:

  • GPC3 isoform complexity:

    • Up to 3 different isoforms have been reported

    • Antibody epitopes may be present in some but not all isoforms

    • Developmental and pathological expression patterns differ across isoforms

  • Post-translational modifications:

    • O-glycosylation and protein cleavage affect epitope accessibility

    • Heparan sulfate chains may mask or create epitopes

    • Tissue-specific glycosylation patterns alter antibody recognition

  • Cross-reactivity concerns:

    • Homology with other glypican family members

    • GPC3 orthologs in mouse, rat, bovine, frog, chimpanzee and chicken show species differences

    • Expression in placenta may pose safety concerns for certain applications

  • Conformational epitopes:

    • Native protein structure is required for some antibodies (e.g., HN3)

    • Conformational changes in the tumor microenvironment may alter binding

    • In vitro screening may not predict in vivo epitope accessibility

  • Heterogeneity in target expression:

    • Variable GPC3 expression between and within tumors

    • Potential expression changes during treatment

    • Regional differences in tumor microenvironment affecting antibody distribution

Strategies to overcome these challenges include comprehensive epitope mapping, advanced humanization techniques (maintaining critical non-CDR residues like proline at position 41 in VH regions), and selection of antibodies recognizing conserved structural features rather than variable regions .

How do GPC3 antibodies modulate signaling pathways in hepatocellular carcinoma?

GPC3 antibodies can modulate various signaling pathways in HCC through different mechanisms:

  • Wnt/β-catenin pathway inhibition:

    • GPC3 normally enhances Wnt signaling in HCC

    • Antibodies targeting specific GPC3 domains can disrupt Wnt ligand binding

    • Downstream effects include reduced β-catenin nuclear translocation and decreased expression of target genes

  • Yes-associated protein (YAP) signaling modulation:

    • The HN3 antibody induces cell-cycle arrest at G1 phase through YAP signaling

    • This represents a previously unrecognized mechanism for GPC3-targeted therapy

    • YAP is a critical oncogenic transcription co-activator in the Hippo pathway

  • Growth factor signaling interference:

    • GPC3 can act as a co-receptor for various growth factors

    • Antibodies may prevent growth factor presentation to their cognate receptors

    • Reduced activation of downstream MAPK and PI3K/AKT pathways

  • Cell cycle regulation:

    • Anti-proliferative effects through G1 phase arrest

    • Modulation of cyclin-dependent kinase inhibitors

    • Altered expression of cell cycle regulatory proteins

  • Immune pathway engagement:

    • Fc-dependent recruitment of immune effectors (for IgG formats)

    • Enhanced antigen presentation through antibody-mediated uptake

    • Potential synergy with checkpoint inhibitors

The pathway modulation varies substantially between antibodies, with some like HN3 directly inhibiting proliferation signaling while others primarily function through immune effector recruitment. Understanding these mechanistic differences is essential for rational combination therapy design and biomarker development .

What are the critical factors in humanizing mouse anti-GPC3 antibodies?

Humanization of mouse anti-GPC3 antibodies involves several critical considerations:

  • CDR grafting strategy:

    • Combined KABAT/IMGT complementarity determining regions (CDR) approach provides optimal results

    • Grafting into human IgG germline framework preserves binding specificity

    • Careful selection of human framework with highest homology to mouse sequence

  • Framework residue preservation:

    • Proline at position 41 in heavy chain variable regions (VH) is particularly important

    • This non-CDR residue significantly impacts humanization success

    • Other vernier zone residues may require preservation to maintain CDR conformation

  • N-glycosylation consideration:

    • N-glycosylation motifs within VH CDR2 (e.g., residue 52a) should be evaluated

    • Potential impacts on antibody function assessed through expression systems with differing glycosylation capacity

    • Bacteria-expressed immunotoxins can validate function without N-glycosylation

  • Activity validation hierarchy:

    • Initial binding affinity screening

    • Functional assays for cytotoxicity or mechanism-specific activity

    • Comprehensive immunogenicity assessment

    • In vivo pharmacokinetics and efficacy validation

The humanization process for YP7 and YP9.1 antibodies demonstrated that successful humanization retains or even enhances the original antibody's properties. The humanized YP7 antibody (hYP7) showed strong binding to GPC3-positive cells with an EC₅₀ of 0.7 nM while maintaining specificity and demonstrated superior ADCC and CDC activity compared to hYP9.1b .

How can researchers optimize production systems for different anti-GPC3 antibody formats?

Optimizing production systems for different anti-GPC3 antibody formats requires format-specific approaches:

For full IgG antibodies:

  • Expression system selection:

    • HEK293T cells for humanized antibodies (hYP7, hYP9.1b)

    • CHO cells for stable production and clinical-grade material

    • Yields and glycosylation patterns differ between systems

  • Process optimization:

    • Fed-batch culture conditions for high-density growth

    • Optimized temperature and pH for maximum expression

    • Serum-free adaptation for clinical applications

For immunotoxin conjugates:

  • Expression in E. coli:

    • Periplasmic expression for proper folding

    • Optimization of induction conditions (temperature, IPTG concentration)

    • Refolding protocols for inclusion body recovery

  • Purification strategy:

    • Multiple chromatography steps to ensure purity

    • Endotoxin removal critical for in vivo applications

    • Stability assessment under various storage conditions

For single-domain antibodies:

  • Expression options:

    • Bacterial systems for cost-effective production

    • Yeast systems for enhanced folding and moderate glycosylation

    • Mammalian expression for complex modifications

  • Format-specific considerations:

    • Addition of stabilizing domains or multimerization

    • Half-life extension strategies (PEGylation, albumin fusion)

    • Affinity maturation through directed evolution

Quality control metrics:

  • SDS-PAGE for purity assessment

  • Size exclusion chromatography for aggregation analysis

  • Functional binding assays (EC₅₀ determination)

  • Cell-based cytotoxicity assays comparing different production batches

The choice of production system should align with the antibody's intended application, with more stringent quality requirements for clinical development compared to research use.

What approaches can improve the affinity and specificity of anti-GPC3 antibodies?

Multiple approaches can enhance the performance characteristics of anti-GPC3 antibodies:

  • Antibody discovery platforms:

    • Humanized transgenic mouse platforms (e.g., CAMouse) containing large V(D)J-regions

    • Phage display technology for isolation of high-affinity binders

    • Single-cell DNA sequencing for rare antibody identification

  • Affinity maturation strategies:

    • Targeted mutagenesis of CDR residues

    • Stringent selection with decreasing antigen concentrations

    • Competitive selection against soluble target

    • High-throughput screening with mammalian display

  • Specificity enhancement:

    • Negative selection against related glypican family members

    • Counter-screening against potential cross-reactive epitopes

    • Tissue cross-reactivity profiling to identify off-target binding

    • Computational prediction of potential cross-reactive epitopes

  • Structural optimization:

    • Crystal structure-guided design for epitope targeting

    • Framework stabilization to improve thermostability

    • Charge distribution optimization to reduce non-specific binding

    • Removal of potential deamidation or oxidation sites

Through these approaches, researchers have achieved remarkable improvements in antibody performance. For example, the HN3 antibody developed through phage display technology achieved subnanomolar affinity (Kd = 0.6 nM) for cell-surface GPC3 while maintaining high specificity for a unique conformational epitope .

How does GPC3 expression heterogeneity impact antibody efficacy in clinical settings?

GPC3 expression heterogeneity presents significant challenges for antibody-based therapeutics:

  • Inter-tumor heterogeneity:

    • Variability in GPC3 expression levels between different patients

    • Correlation between GPC3 expression and clinical outcomes differs by geographic region

    • Studies from Japan showed significant correlation between high GPC3 expression and poor disease-free survival (DFS) (HR = 1.64, 95% CI: 1.02–2.64)

    • Studies from China showed less consistent correlation (HR = 1.59, 95% CI: 0.86–2.95)

  • Intra-tumor heterogeneity:

    • Variable expression within different regions of the same tumor

    • Potential for treatment-resistant subpopulations with altered expression

    • Dynamic changes in expression during disease progression

  • Clinical implications:

    • Need for patient stratification based on GPC3 expression level and pattern

    • Differential response to antibody therapy based on expression intensity

    • Association between high GPC3 expression and treatment response

    • Treatment method impacts prognostic significance (significant association with poor outcomes in patients treated by hepatectomy)

  • Biomarker strategies:

    • Quantitative IHC scoring systems for patient selection

    • Circulating GPC3 as complementary biomarker

    • Multiparametric approaches combining GPC3 with other markers

    • Serial monitoring for expression changes during treatment

These factors highlight the importance of comprehensive GPC3 expression analysis before and during treatment to optimize therapeutic strategies and potentially identify combination approaches for heterogeneously expressing tumors.

What is the current evidence for combining GPC3 antibodies with other cancer therapies?

Emerging evidence supports several promising combination approaches:

  • Combination with immune checkpoint inhibitors:

    • Anti-GPC3 antibodies may enhance tumor antigen presentation

    • ADCC/CDC functions can recruit immune cells to the tumor microenvironment

    • Potential for converting "cold" tumors to "hot" immunogenic tumors

    • Synergistic effects with PD-1/PD-L1 blockade

  • Combination with conventional chemotherapy:

    • GPC3 antibodies may sensitize cancer cells to cytotoxic agents

    • Potential for targeting different cancer cell subpopulations

    • Sequential administration strategies to optimize immune effector recruitment

    • Enhanced efficacy in preclinical models

  • Targeted therapy combinations:

    • Synergy with tyrosine kinase inhibitors targeting complementary pathways

    • Combined blockade of Wnt signaling through different mechanisms

    • Potential for overcoming resistance to single-agent therapies

    • Rational combinations based on signaling pathway analysis

  • Locoregional therapy enhancement:

    • GPC3 antibodies may complement transarterial chemoembolization (TACE)

    • Potential for improved outcomes following surgical resection

    • Combination with radiofrequency ablation in early-stage disease

    • Antibody-directed radiation therapy approaches

The scientific rationale for these combinations is based on the unique mechanisms of GPC3 antibodies. For example, the HN3 antibody's ability to induce cell-cycle arrest via YAP signaling provides a complementary mechanism to immune checkpoint inhibition, potentially addressing multiple aspects of tumor biology simultaneously .

How can researchers translate laboratory findings on GPC3 antibodies to clinical applications?

Successful translation of GPC3 antibodies from laboratory to clinic involves several critical steps:

  • Preclinical development optimization:

    • Humanization strategies preserving critical binding residues

    • Production of clinical-grade material under GMP conditions

    • Comprehensive toxicology assessment in relevant species

    • PK/PD modeling to inform clinical dosing strategies

  • Patient selection strategy development:

    • Standardized GPC3 expression assays for enrollment criteria

    • Identification of predictive biomarkers beyond GPC3 expression

    • Stratification based on tumor characteristics (hepatitis association, multifocality)

    • Consideration of geographical differences in GPC3 association with outcomes

  • Clinical trial design considerations:

    • Selection of appropriate endpoints based on mechanism of action

    • Early incorporation of pharmacodynamic biomarkers

    • Adaptive designs allowing for rapid identification of responding populations

    • Rational combination strategies based on preclinical evidence

  • Translational research integration:

    • Paired biopsies to assess on-target effects

    • Immune monitoring for ADCC/CDC-mediating antibodies

    • Correlation of signaling pathway modulation with clinical response

    • Resistance mechanism investigation through longitudinal sampling

Current clinical development status exemplifies this approach. The humanized GC33 antibody targeting a C-terminal GPC3 epitope has progressed to clinical trials for liver cancer therapy. Meanwhile, newer antibodies like HN3 offer additional mechanisms (direct proliferation inhibition) and formats (single-domain) that may address limitations of first-generation approaches .

Quick Inquiry

Personal Email Detected
Please use an institutional or corporate email address for inquiries. Personal email accounts ( such as Gmail, Yahoo, and Outlook) are not accepted. *
© Copyright 2025 TheBiotek. All Rights Reserved.