The antibody targets the phosphorylated form of EGFR at Y1068, a key residue in the kinase domain that mediates receptor dimerization and signal transduction . Phosphorylation at Y1068 activates pathways such as RAS-RAF-MEK-ERK and PI3K-AKT, which are implicated in oncogenesis and targeted therapy resistance . The recombinant monoclonal format ensures consistent production via genetic engineering, avoiding reliance on animal-derived sources .
Peptide Inhibition Assay: Pre-treatment with phospho-specific peptide abolishes WB signal, confirming specificity .
EGF Stimulation: A431 cells treated with 50–100 ng/mL EGF show time-dependent phosphorylation (5–30 minutes) .
Cross-Reactivity Testing: Reactivity with mouse/rat EGFR confirmed in select antibodies .
EGF-Induced Phosphorylation: EGF stimulation (100 ng/mL, 5–30 minutes) robustly induces Y1068 phosphorylation in A431 cells, detectable via WB and FC .
Pathway Activation: Meprinα protease enhances EGFR-Y1068 phosphorylation, promoting ERK1/2 activation and cell migration .
Glycosylation Impact: Observed band sizes (175–190 kDa) exceed predicted MW due to glycosylation and other post-translational modifications .
Cancer Research: EGFR-Y1068 phosphorylation is linked to oncogenesis and resistance to EGFR inhibitors (e.g., gefitinib) .
Diagnostic Utility: IHC detection of phospho-EGFR-Y1068 in cervical cancer tissues highlights its potential in biomarker studies .
The phospho-EGFR (Y1068) recombinant monoclonal antibody is produced through the utilization of protein technology and DNA recombinant techniques. Initially, animals are immunized with a synthesized peptide derived from human phospho-EGFR (Y1068). Subsequently, B cells are extracted from the immunized animals. Through a rigorous screening process, positive B cells are selected and undergo single clone identification. The light and heavy chains of the phospho-EGFR (Y1068) antibody are then amplified using PCR and inserted into a plasmid vector, resulting in a recombinant vector. This recombinant vector is then transfected into host cells for the expression of the antibody. Finally, the phospho-EGFR (Y1068) recombinant monoclonal antibody is purified from the cell culture supernatant using affinity chromatography. This antibody is recommended for use in ELISA and WB for the detection of human EGFR phosphorylated at the Y1068 residue.
Epidermal growth factor receptor (EGFR) serves as a receptor for ligands belonging to the EGF family, triggering the activation of multiple signaling cascades. These cascades effectively translate extracellular cues into appropriate cellular responses. Notable ligands include EGF, TGFA/TGF-alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG, and HBEGF/heparin-binding EGF. Ligand binding induces receptor homo- and/or heterodimerization, leading to autophosphorylation on critical cytoplasmic residues. This phosphorylation event facilitates the recruitment of adapter proteins like GRB2, which in turn activates intricate downstream signaling cascades.
EGFR activates at least four major downstream signaling cascades, encompassing the RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC, and STATs modules. It may also activate the NF-kappa-B signaling cascade. Notably, EGFR directly phosphorylates other proteins like RGS16, stimulating its GTPase activity and potentially coupling EGF receptor signaling to G protein-coupled receptor signaling. EGFR also phosphorylates MUC1, enhancing its interaction with SRC and CTNNB1/beta-catenin.
EGFR positively regulates cell migration through its interaction with CCDC88A/GIV. This interaction retains EGFR at the cell membrane following ligand stimulation, thereby promoting EGFR signaling and triggering cell migration. EGFR plays a crucial role in enhancing learning and memory performance. Isoform 2 of EGFR may act as an antagonist of EGF action. In the context of microbial infection, EGFR acts as a receptor for hepatitis C virus (HCV) in hepatocytes, facilitating its entry into cells. EGFR mediates HCV entry by promoting the formation of the CD81-CLDN1 receptor complexes essential for HCV entry and enhancing membrane fusion of cells expressing HCV envelope glycoproteins.
EGFR phosphorylation at Y1068 serves as a critical binding site for signaling adaptors, particularly the Grb2:Cbl complex. This phosphorylation site is not only necessary but also sufficient for EGFR ubiquitination, along with Y1045 and Y1086. These phosphorylation events trigger downstream signaling cascades that regulate cell proliferation, survival, and differentiation. The phosphorylation status at Y1068 is therefore a key indicator of EGFR activation and function in both normal and pathological contexts .
To validate specificity, researchers should use appropriate positive and negative controls. A commonly employed method is comparing A431 human epithelial carcinoma cells with and without EGF treatment. Western blot analysis should show detection of a specific band at approximately 190 kDa in EGF-treated samples, with minimal to no detection in untreated samples. Additionally, using EGFR kinase inhibitors like C56 can confirm specificity, as they should abrogate the signal in stimulated cells .
Phospho-EGFR (Y1068) antibodies have been validated for multiple applications including Western blotting, flow cytometry, and CyTOF (mass cytometry). For Western blotting, these antibodies typically work well under reducing conditions, with PVDF membranes and appropriate buffer systems. For flow cytometry, cells must be fixed with paraformaldehyde and permeabilized (often with saponin) to facilitate intracellular staining .
While EGFR mutations are strong predictors of response to EGFR tyrosine kinase inhibitors (TKIs), approximately 10% of patients with wild-type EGFR also respond to these therapies. Research has shown that pTyr1068 expression may serve as an independent predictive biomarker for TKI response. In a study of 205 non-small cell lung cancer patients, those with pTyr1068 expression demonstrated superior progression-free survival after EGFR-TKI therapy compared to those without expression (median PFS 7.0 months vs. 1.2 months, P < 0.001). Notably, even in the wild-type EGFR subgroup, pTyr1068-positive patients showed significantly prolonged PFS (4.2 months vs. 1.2 months, P < 0.001) .
Quantitative analysis reveals that EGFR phosphorylation and subsequent signaling follow a dose-response pattern to ligand stimulation. The relationship between EGF concentration and Y1068 phosphorylation is not simply linear but involves complex regulatory mechanisms. Mathematical modeling suggests that cooperativity between multiple phosphorylation sites, including Y1068, contributes to establishing thresholds for cellular responses. This cooperativity is particularly evident in the relationship between phosphorylation and ubiquitination, where Y1068 (along with Y1086) serves as a binding site for Grb2:Cbl complexes that mediate receptor ubiquitination and downregulation .
For optimal detection by Western blotting:
Parameter | Recommended Condition |
---|---|
Membrane | PVDF |
Antibody concentration | 1 μg/mL |
Secondary antibody | HRP-conjugated Anti-Mouse IgG |
Running conditions | Reducing conditions |
Buffer system | Immunoblot Buffer Group 1 |
Expected band size | Approximately 190 kDa |
Positive control | A431 cells treated with 100 ng/mL EGF for 5 minutes |
Negative control | Untreated A431 cells |
It is critical to include phosphatase inhibitors in lysis buffers and to maintain samples at cold temperatures during processing to preserve phosphorylation status .
Successful intracellular flow cytometry for phospho-EGFR (Y1068) requires:
Proper fixation: Use paraformaldehyde (typically 4%) to preserve cellular architecture and phosphorylation status.
Effective permeabilization: Saponin is commonly used to allow antibody access to intracellular antigens.
Blocking: Include appropriate blocking reagents to reduce non-specific binding.
Controls: Always include an isotype control (e.g., Mouse IgG2A) and both stimulated and unstimulated samples.
Secondary detection: For unconjugated primary antibodies, use fluorochrome-conjugated secondary antibodies like Phycoerythrin-conjugated Anti-Mouse IgG F(ab')2.
This approach has been successfully demonstrated in A431 cells treated with EGF, showing clear shifts in phospho-EGFR signal compared to untreated controls .
LANCE Ultra assays offer several methodological advantages for phospho-EGFR (Y1068) detection:
Homogeneous format: No wash steps required, simplifying workflow and reducing variability.
Compatibility: Works with both adherent and suspension cells.
Low sample requirement: Only 15 μL of sample needed.
Time-resolved detection: Utilizes TR-FRET (time-resolved fluorescence resonance energy transfer) technology, reducing background interference.
Quantitative results: Provides ratiometric data (665/615 nm X 10,000) for more reliable quantification.
This technology combines a donor fluorophore (LANCE Europium chelate) on one antibody with an acceptor fluorophore (ULight dye) on another, enabling energy transfer when the antibodies are in close proximity (~10 nm) following excitation at 320 or 340 nm .
Research has demonstrated that phospho-EGFR (Y1068) expression may serve as a valuable predictive biomarker for EGFR-TKI therapy response, independent of EGFR mutation status. In non-small cell lung cancer, patients with pTyr1068 expression showed significantly better outcomes with EGFR-TKI treatment. Particularly notable was the identification of a subgroup of patients with wild-type EGFR but positive pTyr1068 expression who responded well to EGFR-TKIs, with a median progression-free survival of 15.6 months (95% CI: 7.28-23.9). This suggests that immunohistochemical assessment of pTyr1068 could help identify additional patients who might benefit from EGFR-targeted therapies beyond those selected by mutation testing alone .
The relationship between different EGFR phosphorylation sites provides complex regulatory information. While pTyr1068 expression correlates with improved outcomes on EGFR-TKI therapy, pTyr1173 shows an inverse relationship, with patients expressing pTyr1173 having shorter progression-free survival (4.8 months vs. 7.7 months, P = 0.016). This differential impact of various phosphorylation sites highlights the complexity of EGFR signaling networks and suggests that comprehensive phosphorylation profiling might provide more nuanced predictive information than single-site analysis. The molecular basis for these differences may lie in the recruitment of distinct adaptor proteins and activation of different downstream signaling pathways .
Understanding phospho-EGFR (Y1068) status in the context of treatment resistance provides important mechanistic insights. Patients who initially respond to EGFR-TKIs based on pTyr1068 expression may develop resistance through various mechanisms, including:
Bypass pathway activation: Alternative signaling pathways may become activated, reducing dependence on EGFR.
Phosphatase upregulation: Increased expression or activity of phosphatases that dephosphorylate Y1068.
Altered receptor trafficking: Changes in receptor internalization and degradation pathways.
Secondary mutations: Acquisition of mutations that prevent inhibitor binding while maintaining Y1068 phosphorylation.
Monitoring changes in pTyr1068 status during treatment could potentially help identify emerging resistance mechanisms and inform subsequent treatment decisions .
False negative results when detecting phospho-EGFR (Y1068) may stem from several methodological issues:
Rapid dephosphorylation: Phosphorylation is highly labile and can be lost during sample preparation. Always include phosphatase inhibitors in lysis buffers.
Insufficient stimulation: For positive controls, ensure adequate stimulation (e.g., 100 ng/mL EGF for 5 minutes for A431 cells).
Antibody concentration: Using too dilute antibody preparation (optimal concentration is typically 1 μg/mL for Western blot).
Ineffective permeabilization: For flow cytometry, inadequate permeabilization with saponin can prevent antibody access to intracellular antigens.
Sample degradation: Repeated freeze-thaw cycles or prolonged storage at inappropriate temperatures can degrade phosphorylated proteins .
To distinguish between kinase-dependent and kinase-independent phosphorylation mechanisms:
Use specific EGFR kinase inhibitors (e.g., C56 at 1 μM) as pretreatment before stimulation. If Y1068 phosphorylation is abrogated by the inhibitor, it suggests kinase-dependent mechanisms.
Compare phosphorylation patterns across multiple sites - different mechanisms may preferentially affect certain sites.
Investigate time-course dynamics, as kinase-dependent and independent mechanisms often show different temporal profiles.
Examine phosphorylation in the presence of Src inhibitors, as Src-mediated phosphorylation of EGFR can occur independently of EGFR's intrinsic kinase activity in some contexts.
Research has shown that ozone-induced EGFR (Y1068) phosphorylation in human bronchial epithelial cells requires EGFR kinase activity, as it was abrogated by kinase inhibition .
When working with complex biological samples such as tissue lysates or clinical specimens:
Sample preservation: Rapid freezing and proper storage are essential to maintain phosphorylation status.
Normalization strategies: Use total EGFR levels for normalization rather than housekeeping proteins alone.
Heterogeneity considerations: Cell-type specific expression patterns may be masked in whole-tissue lysates.
Validation across methods: Confirm key findings using orthogonal techniques (e.g., both Western blot and immunohistochemistry).
Contextual interpretation: Consider the broader signaling environment, including the status of phosphatases, RTK crosstalk, and downstream effectors.
Appropriate controls: Include both positive controls (e.g., EGF-stimulated cell lines) and negative controls (untreated samples or those treated with kinase inhibitors).
These considerations are particularly important in clinical research settings where sample quality and processing methods can significantly impact results and their interpretation .