Phospho-EGFR (S1071) Antibody

Shipped with Ice Packs
In Stock

Description

Western Blot (WB)

  • Used to detect phosphorylated EGFR (S1071) in lysates from human, mouse, or rat tissues/cell lines .

  • Optimized dilution: 1:500–1:2000 .

ELISA

  • Quantifies phospho-EGFR (S1071) levels in biological samples at a dilution of 1:5000 .

Functional Insights

  • EGFR phosphorylation at S1071 is linked to receptor activation and downstream signaling cascades (e.g., RAS-RAF-MEK-ERK, PI3K-AKT) .

  • Post-translational modifications at S1071 may regulate EGFR internalization or interaction with adaptor proteins like GRB2 .

Technical Validation and Specificity

  • Specificity: The antibody shows no cross-reactivity with non-phosphorylated EGFR or other phosphorylated tyrosine residues (e.g., Tyr1068, Tyr1173) .

  • Validation: Tested in WB using HEK293T cells treated with EGF to induce EGFR phosphorylation .

Biological Context of EGFR S1071 Phosphorylation

  • EGFR Function: A receptor tyrosine kinase critical for cell growth, survival, and migration. Dysregulation is implicated in cancers (e.g., lung, colorectal) .

  • Phosphorylation Role: S1071 phosphorylation may modulate receptor dimerization, kinase activity, or interaction with downstream effectors .

Comparison with Other EGFR Phospho-Specific Antibodies

Antibody TargetApplicationsSpecies ReactivityKey Differences
Phospho-EGFR (S1071) WB, ELISAHuman, Mouse, RatTargets serine phosphorylation (vs. tyrosine)
Phospho-EGFR (Y1068) WB, IHCHuman, Mouse, RatDetects tyrosine phosphorylation
Phospho-EGFR (Y1173) WB, IHC, IF/ICCHuman, Mouse, RatBinds dual phosphorylation sites (Y1173/Y1197)

Research Implications

  • Cancer Studies: Enables tracking of EGFR activation status in tumor samples, aiding in therapeutic response studies (e.g., tyrosine kinase inhibitors) .

  • Mechanistic Insights: Useful for dissecting phosphorylation-dependent EGFR interactions in signal transduction pathways .

Limitations and Cautions

  • Research Use Only (RUO): Not validated for diagnostic or therapeutic applications .

  • Species Limitations: Reactivity confirmed only in human, mouse, and rat models .

Product Specs

Buffer
The antibody is provided as a liquid solution in phosphate buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA) and 0.02% sodium azide.
Form
Liquid
Lead Time
Typically, we can ship the products within 1-3 business days after receiving your order. Delivery time may vary depending on the shipping method and destination. Please consult your local distributors for specific delivery times.
Synonyms
Avian erythroblastic leukemia viral (v erb b) oncogene homolog antibody; Cell growth inhibiting protein 40 antibody; Cell proliferation inducing protein 61 antibody; EGF R antibody; EGFR antibody; EGFR_HUMAN antibody; Epidermal growth factor receptor (avian erythroblastic leukemia viral (v erb b) oncogene homolog) antibody; Epidermal growth factor receptor (erythroblastic leukemia viral (v erb b) oncogene homolog avian) antibody; Epidermal growth factor receptor antibody; erb-b2 receptor tyrosine kinase 1 antibody; ERBB antibody; ERBB1 antibody; Errp antibody; HER1 antibody; mENA antibody; NISBD2 antibody; Oncogen ERBB antibody; PIG61 antibody; Proto-oncogene c-ErbB-1 antibody; Receptor tyrosine protein kinase ErbB 1 antibody; Receptor tyrosine-protein kinase ErbB-1 antibody; SA7 antibody; Species antigen 7 antibody; Urogastrone antibody; v-erb-b Avian erythroblastic leukemia viral oncogen homolog antibody; wa2 antibody; Wa5 antibody
Target Names
Uniprot No.

Target Background

Function
The epidermal growth factor receptor (EGFR) is a transmembrane receptor tyrosine kinase that plays a crucial role in regulating cell growth, proliferation, differentiation, and survival. EGFR is activated by a variety of ligands, including EGF, TGFA/TGF-alpha, AREG, epigen/EPGN, BTC/betacellulin, epiregulin/EREG, and HBEGF/heparin-binding EGF. Ligand binding induces receptor dimerization and autophosphorylation, leading to the activation of downstream signaling pathways such as the RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC, and STATs modules. EGFR signaling can also activate the NF-kappa-B pathway. Additionally, EGFR directly phosphorylates other proteins such as RGS16, enhancing its GTPase activity and potentially coupling EGFR signaling to G protein-coupled receptor signaling. EGFR also phosphorylates MUC1, increasing its interaction with SRC and CTNNB1/beta-catenin. EGFR positively regulates cell migration through interaction with CCDC88A/GIV, which maintains EGFR at the cell membrane following ligand stimulation, promoting EGFR signaling and triggering cell migration. EGFR plays a 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 and facilitates its cell entry. EGFR mediates HCV entry by promoting the formation of the CD81-CLDN1 receptor complexes that are essential for HCV entry and by enhancing membrane fusion of cells expressing HCV envelope glycoproteins.
Gene References Into Functions
  1. Amphiregulin, present in non-small-cell lung carcinoma-derived exosomes, induces osteoclast differentiation through the activation of the EGFR pathway. PMID: 28600504
  2. Combining vorinostat with an EGFR tyrosine kinase inhibitor (TKI) may reverse EGFRTKI resistance in non-small cell lung cancer (NSCLC). PMID: 30365122
  3. The feasibility of utilizing the radiocobalt labeled antiEGFR affibody conjugate ZEGFR:2377 as an imaging agent is being explored. PMID: 30320363
  4. Among all transfection complexes studied, 454 lipopolyplexes modified with the bidentate PEG-GE11 agent demonstrated the best EGFR-dependent uptake, as well as luciferase and NIS gene expression. PMID: 28877405
  5. EGFR amplification was higher in the oral squamous cell carcinoma (OSCC) group than in the control group (P=0.018) and was associated with advanced clinical stage (P=0.013), irrespective of age. Patients with EGFR overexpression exhibited worse survival rates, as did patients with T3-T4 tumors and positive margins. EGFR overexpression negatively impacts disease progression. PMID: 29395668
  6. Clonal analysis revealed that the dominant JAK2 V617F-positive clone in Polycythemia Vera harbors EGFR C329R substitution, suggesting that this mutation may contribute to clonal expansion. PMID: 28550306
  7. Baseline circulating tumor cell count could serve as a predictive biomarker for EGFR-mutated and ALK-rearranged non-small cell lung cancer, providing better guidance and monitoring of patients during molecular targeted therapies. PMID: 29582563
  8. High EGFR expression is associated with cystic fibrosis. PMID: 29351448
  9. Findings suggest a mechanism for EGFR inhibition to suppress respiratory syncytial virus by activating endogenous epithelial antiviral defenses. PMID: 29411775
  10. This study detected the emergence of the T790M mutation within the EGFR cDNA in a subset of erlotinib-resistant PC9 cell models through Sanger sequencing and droplet digital PCR-based methods, demonstrating that the T790M mutation can arise de novo following treatment with erlotinib. PMID: 29909007
  11. The present study revealed that miR145 regulates the EGFR/PI3K/AKT signaling pathway in patients with non-small cell lung cancer. PMID: 30226581
  12. Among NSCLC patients treated with EGFR-TKI, those with T790M mutations were found to frequently also exhibit 19 deletions, compared to T790M-negative patients. Moreover, T790M-positive patients had a longer progression-free survival (PFS). Therefore, screening these patients for T790M mutations may contribute to improved survival. PMID: 30150444
  13. High EGFR expression is associated with Breast Carcinoma. PMID: 30139236
  14. Results indicated that CAV-1 could promote anchorage-independent growth and anoikis resistance in detached SGC-7901 cells, which was associated with the activation of Src-dependent epidermal growth factor receptor-integrin beta signaling as well as the phosphorylation of PI3K/Akt and MEK/ERK signaling pathways. PMID: 30088837
  15. Findings suggest that FOXK2 inhibits the malignant phenotype of clear-cell renal cell carcinoma and acts as a tumor suppressor potentially through the inhibition of EGFR. PMID: 29368368
  16. EGFR mutation status in advanced non-small cell lung cancer (NSCLC) patients altered significantly. PMID: 30454543
  17. Different Signaling Pathways in Regulating PD-L1 Expression in EGFR Mutated Lung Adenocarcinoma. PMID: 30454551
  18. Internal tandem duplication of the kinase domain delineates a genetic subgroup of congenital mesoblastic nephroma transcending histological subtypes. PMID: 29915264
  19. The expression level of EGFR increased along with higher stages and pathological grades of breast tumor-like carcinoma (BTCC), and the significantly increased expression of HER-2 was statistically associated with clinical stages and tumor recurrence. Additionally, the expression level of HER-2 increased with the higher clinical stage of BTCC. EGFR expression and HER-2 levels were positively associated in BTCC samples. PMID: 30296252
  20. Results demonstrate that GGA2 interacts with the EGFR cytoplasmic domain to stabilize its expression and reduce its lysosomal degradation. PMID: 29358589
  21. Combination therapy of apatinib with icotinib for primary acquired resistance to icotinib might be an option for patients with advanced pulmonary adenocarcinoma with EGFR mutations, but clinicians must also be mindful of the side effects caused by such therapy. PMID: 29575765
  22. This report presents a rare case presenting as multiple lung adenocarcinomas with four different EGFR gene mutations detected in three lung tumors. PMID: 29577613
  23. The study supports the involvement of EGFR, HER2, and HER3 in basal cell carcinoma (BCC) aggressiveness and in tumor differentiation towards different histological subtypes. PMID: 30173251
  24. The ratio of sFlt-1/sEGFR could be employed as a novel candidate biochemical marker for monitoring the severity of preterm preeclampsia. sEndoglin and sEGFR may be involved in the pathogenesis of small for gestational age in preterm preeclampsia. PMID: 30177039
  25. The study confirmed the prognostic effect of EGFR and VEGFR2 on recurrent disease and survival rates in patients with epithelial ovarian cancer. PMID: 30066848
  26. Data suggest that diagnostic or therapeutic chest radiation may predispose patients with decreased stromal PTEN expression to secondary breast cancer, and that prophylactic EGFR inhibition may reduce this risk. PMID: 30018330
  27. Findings propose a unique regulatory feature of PHLDA1 to inhibit the ErbB receptor oligomerization process and thereby control the activity of the receptor signaling network. PMID: 29233889
  28. The study observed the occurrence of not only EGFR C797S mutation but also L792F/Y/H in three NSCLC clinical subjects with acquired resistance to osimertinib treatment. PMID: 28093244
  29. Data show that the expression level of epidermal growth factor-like domain 7 (EGFL7) and epidermal growth factor receptor (EGFR) in invasive growth hormone-producing pituitary adenomas (GHPA) was much higher than that of non-invasive GHPA. PMID: 29951953
  30. Concurrent mutations, in genes such as CDKN2B or RB1, were associated with worse clinical outcome in lung adenocarcinoma patients with EGFR active mutations. PMID: 29343775
  31. The ER-alpha36/EGFR signaling loop promotes growth of hepatocellular carcinoma cells. PMID: 29481815
  32. High EGFR expression is associated with colorectal cancer. PMID: 30106444
  33. High EGFR expression is associated with gefitinib resistance in lung cancer. PMID: 30106446
  34. High EGFR expression is associated with tumor-node-metastasis in non-small cell lung cancer. PMID: 30106450
  35. Data suggest that Thr264 in TRPV3 is a key ERK1 phosphorylation site mediating EGFR-induced sensitization of TRPV3 to stimulate signaling pathways involved in regulating skin homeostasis. (TRPV3 = transient receptor potential cation channel subfamily V member-3; ERK1 = extracellular signal-regulated kinase-1; EGFR = epidermal growth factor receptor) PMID: 29084846
  36. The EGFR mutation frequency in Middle East and African patients is higher than that observed in white populations but still lower than the frequency reported in Asian populations. PMID: 30217176
  37. EGFR-containing exosomes derived from cancer cells could favor the development of a liver-like microenvironment, promoting liver-specific metastasis. PMID: 28393839
  38. The results indicate that the EGF-STAT3 signaling pathway promotes and maintains colorectal cancer (CRC) stemness. Additionally, a crosstalk between STAT3 and Wnt activates the Wnt/beta-catenin signaling pathway, which is also responsible for cancer stemness. Thus, STAT3 is a potential therapeutic target for CRC treatment. PMID: 30068339
  39. This result suggested that the T790M mutation is not only associated with EGFR-TKI resistance but also may play a functional role in the malignant progression of lung adenocarcinoma. PMID: 29887244
  40. LOX regulates EGFR cell surface retention to drive tumor progression. PMID: 28416796
  41. In a Han Chinese population, EGFR gene polymorphisms, rs730437 and rs1468727, and haplotype A-C-C were shown to be potential protective factors against the development of Alzheimer's Disease. PMID: 30026459
  42. EGFR proteins at different cellular locations in lung adenocarcinoma might influence the biology of cancer cells and are an independent indicator of a more favorable prognosis and treatment response. PMID: 29950164
  43. This report presents the crystal structure of EGFR T790M/C797S/V948R in complex with EAI045, a novel type of EGFR TKI that binds to EGFR reversibly and does not rely on Cys 797. PMID: 29802850
  44. Overexpression of miR-452-3p promoted cell proliferation and mobility while suppressing apoptosis. MiR-452-3p enhanced EGFR and phosphorylated AKT (pAKT) expression but inhibited p21 expression level. MiR-452-3p promoted hepatocellular carcinoma (HCC) cell proliferation and mobility by directly targeting the CPEB3/EGFR axis. PMID: 29332449
  45. This study shows that the D2A sequence of the UPAR induces cell growth through alphaVbeta3 integrin and EGFR. PMID: 29184982
  46. BRAF and EGFR inhibitors are capable of synergizing to increase cytotoxic effects and decrease stem cell capacities in BRAF(V600E)-mutant colorectal cancer cells. PMID: 29534162
  47. This study confirms a direct correlation between MSI1 and EGFR and may support the significant role of MSI1 in the activation of EGFR through NOTCH/WNT pathways in esophageal squamous cell carcinoma. PMID: 30202417
  48. Three lines of tyrosine kinase inhibitors (TKIs) therapy can prolong survival in non-small cell lung cancer (NSCLC) patients. Elderly patients can benefit from TKI therapy. EGFR mutation-positive patients can benefit from second-line or third-line TKI therapy. PMID: 29266865
  49. EGFR 19Del and L858R mutations are good biomarkers for predicting the clinical response of EGFR-TKIs. 19Del mutations may have a better clinical outcome. PMID: 29222872
  50. HMGA2-EGFR constitutively induced a higher level of phosphorylated STAT5B than EGFRvIII. PMID: 29193056

Show More

Hide All

Database Links

HGNC: 3236

OMIM: 131550

KEGG: hsa:1956

STRING: 9606.ENSP00000275493

UniGene: Hs.488293

Involvement In Disease
Lung cancer (LNCR); Inflammatory skin and bowel disease, neonatal, 2 (NISBD2)
Protein Families
Protein kinase superfamily, Tyr protein kinase family, EGF receptor subfamily
Subcellular Location
Cell membrane; Single-pass type I membrane protein. Endoplasmic reticulum membrane; Single-pass type I membrane protein. Golgi apparatus membrane; Single-pass type I membrane protein. Nucleus membrane; Single-pass type I membrane protein. Endosome. Endosome membrane. Nucleus.; [Isoform 2]: Secreted.
Tissue Specificity
Ubiquitously expressed. Isoform 2 is also expressed in ovarian cancers.

Q&A

What is the Phospho-EGFR (S1071) Antibody and what epitope does it recognize?

Phospho-EGFR (S1071) Antibody is a polyclonal antibody specifically designed to recognize the Epidermal Growth Factor Receptor (EGFR) protein only when phosphorylated at serine residue 1071. The antibody is typically generated using synthesized peptides derived from human EGFR encompassing the phosphorylation site of S1071 as immunogens . The specificity for the phosphorylated form allows researchers to distinguish the activated state of EGFR at this particular residue from the non-phosphorylated form, enabling detailed signaling studies.

How does Phospho-EGFR (S1071) differ from other phospho-specific EGFR antibodies?

Phospho-EGFR (S1071) antibody specifically targets serine phosphorylation, while many commonly used phospho-EGFR antibodies target tyrosine residues such as Y1068, Y1086/Y1110, or Y1172 . This distinction is important because:

  • Serine/threonine phosphorylation often regulates receptor desensitization and internalization, whereas tyrosine phosphorylation typically activates downstream signaling cascades .

  • Different kinases are responsible for these modifications - serine/threonine kinases versus tyrosine kinases - representing distinct regulatory mechanisms.

  • The functional outcomes of S1071 phosphorylation may differ significantly from those of tyrosine phosphorylation sites.

This table summarizes key differences between common phospho-EGFR antibodies:

Antibody TargetResidue TypePrimary FunctionCommon Applications
Phospho-EGFR (S1071)SerineReceptor regulationWB, ELISA
Phospho-EGFR (Y1068)TyrosineGRB2 binding, MAPK activationWB, IHC, Flow Cytometry
Phospho-EGFR (Y1086/Y1110)TyrosinePI3K signalingWB, ELISA
Phospho-EGFR (S1046/S1047)SerineReceptor downregulationWB, ICC/IF, IP

What are the recommended experimental applications for Phospho-EGFR (S1071) Antibody?

Based on validation data, Phospho-EGFR (S1071) Antibody is primarily recommended for:

  • Western Blotting (WB): Optimal dilution range 1:500-1:2000

    • Can detect bands at approximately 134-180 kDa depending on post-translational modifications

  • ELISA: Recommended dilution of 1:5000

    • Useful for quantitative assessment of phosphorylation levels

While some vendors may suggest other applications, these two have the most consistent validation data across manufacturers. For specialized applications like immunohistochemistry (IHC) or immunofluorescence (IF), additional validation may be necessary in your specific experimental system.

What are the optimal sample preparation methods for detecting Phospho-EGFR (S1071)?

For effective detection of Phospho-EGFR (S1071), consider these critical sample preparation guidelines:

  • Phosphatase inhibitors: Always include phosphatase inhibitors (e.g., sodium orthovanadate, sodium fluoride, β-glycerophosphate) in lysis buffers to preserve phosphorylation status .

  • Lysis conditions: Use RIPA or similar buffers supplemented with protease inhibitors. For membrane proteins like EGFR, addition of 0.1% SDS can improve extraction efficiency .

  • Sample handling: Process samples rapidly at 4°C to minimize dephosphorylation. Flash freezing in liquid nitrogen is recommended if immediate processing is not possible.

  • Positive controls: EGF-stimulated A431 cells (human epithelial carcinoma) serve as excellent positive controls, showing robust phosphorylation at S1071 after treatment with 100 ng/mL recombinant human EGF for 5-10 minutes .

  • Reduction and denaturation: Complete denaturation is critical; use reducing conditions (DTT or β-mercaptoethanol) and heat samples at 95°C for 5 minutes.

How should I design experiments to study the kinetics of EGFR S1071 phosphorylation?

To effectively study the kinetics of EGFR S1071 phosphorylation:

  • Time course experiment design:

    • Establish baseline (0 min) phosphorylation levels

    • Stimulate cells with appropriate ligand (typically EGF at 100 ng/mL)

    • Collect samples at multiple timepoints (30 sec, 2 min, 5 min, 10 min, 30 min, 1 hr, 2 hr)

    • Process all samples simultaneously to minimize technical variation

  • Quantification approach:

    • Always normalize phospho-signal to total EGFR expression levels using a non-phospho-specific EGFR antibody on stripped membranes

    • Use densitometry software for accurate quantification

    • Present data as fold-change in phosphorylation relative to baseline

  • Controls to include:

    • Unstimulated controls at each timepoint to account for basal changes

    • Phosphatase treatment controls to confirm signal specificity

    • EGFR inhibitor controls (e.g., erlotinib) to demonstrate signal regulation

Research has shown that different phosphorylation sites on EGFR demonstrate distinct temporal patterns, with S1071 potentially showing different kinetics compared to tyrosine sites like Y1068 or Y1086 .

How should I interpret discrepancies between phospho-EGFR (S1071) levels and other EGFR phosphorylation sites?

Discrepancies between phosphorylation levels at different EGFR sites are common and biologically significant. When analyzing such differences:

  • Different regulatory mechanisms: Serine phosphorylation (including S1071) is often mediated by downstream kinases rather than receptor autophosphorylation. For example, studies have shown that while EGF stimulation induces rapid Y1068 phosphorylation, S1071 phosphorylation may follow different kinetics and might be regulated by distinct pathways .

  • Functional implications: Tyrosine phosphorylation sites (e.g., Y1068, Y1086) primarily mediate downstream signaling through recruitment of adaptor proteins like GRB2, while serine phosphorylation often regulates receptor trafficking, desensitization, or cross-talk with other pathways .

  • Cell type-specific patterns: Different cell types may exhibit distinct phosphorylation patterns. For example, research on EGFR phosphorylation in lung cancer cells (A549) versus breast cancer cells (MDA-MB-231) showed distinct patterns even with the same stimulus .

  • Interpretative approach: When observing discrepancies, consider:

    • Is the difference stimulus-specific?

    • Does inhibition of specific pathways differentially affect these sites?

    • Are these differences consistent across cell types or unique to your model?

What are common sources of false positives/negatives when using Phospho-EGFR (S1071) antibodies?

Several factors can contribute to false results when using phospho-specific antibodies:

Potential sources of false positives:

  • Cross-reactivity with similar phosphorylation motifs on other proteins

  • Insufficient blocking during Western blot procedures

  • Sample overloading causing non-specific binding

  • Inadequate washing steps in immunoassays

Potential sources of false negatives:

  • Rapid dephosphorylation during sample preparation (insufficient phosphatase inhibitors)

  • Epitope masking due to protein-protein interactions

  • Suboptimal antibody concentration

  • Incomplete protein transfer during Western blotting for large proteins like EGFR

Verification strategies:

  • Phosphatase treatment controls - sample treatment with lambda phosphatase should abolish signal

  • Peptide competition assays - pre-incubation with phosphopeptide should block specific binding

  • siRNA knockdown of EGFR - should reduce or eliminate specific signal

  • Comparison with other well-characterized phospho-specific antibodies using the same samples

How can Phospho-EGFR (S1071) Antibody be used to study receptor trafficking and internalization?

Phosphorylation at serine residues like S1071 has been implicated in EGFR internalization and trafficking. To study these processes:

  • Subcellular fractionation approach:

    • Separate cellular compartments (membrane, cytosolic, endosomal fractions)

    • Analyze the distribution of phospho-EGFR (S1071) versus total EGFR

    • Compare with markers for different cellular compartments (e.g., Na+/K+ ATPase for plasma membrane, EEA1 for early endosomes)

  • Imaging techniques:

    • Combined immunofluorescence using phospho-EGFR (S1071) antibody with compartment markers

    • Live-cell imaging using fluorescently tagged EGFR combined with phospho-specific antibody staining after fixation

    • Super-resolution microscopy for detailed localization studies

  • Receptor trafficking inhibitors:

    • Use specific inhibitors of different trafficking pathways (e.g., dynamin inhibitors, clathrin inhibitors)

    • Measure effects on S1071 phosphorylation compared to other EGFR phosphorylation sites

    • Compare with receptor downregulation measurement using surface biotinylation assays

Research has shown that antibody combinations targeting different EGFR domains can reduce surface receptor levels by up to 80%, suggesting complex regulation of receptor internalization . The relationship between S1071 phosphorylation and this process remains an active area of investigation.

What is the role of Phospho-EGFR (S1071) in cancer resistance mechanisms to EGFR-targeted therapies?

EGFR-targeted therapies, including tyrosine kinase inhibitors like erlotinib, are important cancer treatments, but resistance frequently develops. Investigating the role of S1071 phosphorylation in this context:

  • Resistance model systems:

    • Compare S1071 phosphorylation levels between sensitive and resistant cell lines

    • Develop acquired resistance models through long-term drug exposure

    • Use patient-derived xenografts from responders versus non-responders

  • Signaling bypass mechanisms:

    • Investigate whether S1071 phosphorylation persists despite tyrosine kinase inhibition

    • Determine which upstream kinases maintain S1071 phosphorylation during drug treatment

    • Assess whether combined inhibition of these kinases restores drug sensitivity

  • Methodological approaches:

    • Phosphoproteomics to comprehensively map changes in EGFR phosphorylation patterns during resistance development

    • Site-directed mutagenesis (S1071A) to determine functional significance

    • In vivo models to validate findings from cell culture systems

Research has shown that while EGFR tyrosine phosphorylation sites like Y1092, Y1110, and Y1172 correlate with sensitivity to erlotinib , the role of serine phosphorylation sites including S1071 in modulating drug response represents an understudied area that could reveal novel resistance mechanisms.

How do different EGFR mutations affect phosphorylation at S1071 compared to tyrosine phosphorylation sites?

EGFR mutations, particularly in lung cancer, significantly impact receptor function and drug sensitivity. Understanding their effect on S1071 phosphorylation:

  • Comparative analysis across mutation types:

    • Common activating mutations (exon 19 deletions, L858R)

    • Resistance mutations (T790M, C797S)

    • Uncommon mutations with variable clinical responses

  • Experimental approach:

    • Isogenic cell lines expressing wild-type versus mutant EGFR

    • Analysis of basal and ligand-induced phosphorylation patterns

    • Temporal dynamics of phosphorylation/dephosphorylation at different sites

  • Structural considerations:

    • How mutations affect the conformation of the kinase domain

    • Impact on accessibility of S1071 to kinases and phosphatases

    • Potential for altered protein-protein interactions affecting serine phosphorylation

How can mass spectrometry complement antibody-based detection of Phospho-EGFR (S1071)?

Mass spectrometry (MS) provides powerful complementary approaches to antibody-based detection:

  • Unbiased phosphorylation site mapping:

    • MS can identify novel and known phosphorylation sites simultaneously

    • Enables discovery of previously uncharacterized sites that may function together with S1071

    • Can detect multiple modifications on the same peptide to reveal combinatorial regulation

  • Quantitative assessment advantages:

    • Label-free quantitation allows precise measurement of phosphorylation stoichiometry

    • Multiple reaction monitoring (MRM) provides highly sensitive targeted quantification

    • SILAC or TMT labeling enables multiplexed comparison across conditions

  • Methodological workflow:

    • EGFR immunoprecipitation followed by tryptic digestion

    • Phosphopeptide enrichment (TiO2, IMAC, or phospho-specific antibodies)

    • High-resolution LC-MS/MS analysis

    • Database searching with phosphorylation as variable modification

Studies utilizing mass spectrometry have identified 30 EGFR phosphorylation sites, including 12 serine phosphorylation sites, providing a comprehensive map of receptor modification that extends beyond what can be studied with available antibodies . These approaches have revealed that five sites (pT693, pY1092, pY1110, pY1172 and pY1197) are inhibited by erlotinib in a concentration-dependent manner .

What is the interplay between S1071 phosphorylation and other post-translational modifications on EGFR?

EGFR undergoes multiple types of post-translational modifications that may interact with S1071 phosphorylation:

  • Cross-talk with other phosphorylation sites:

    • Hierarchical phosphorylation events where one site influences modification of another

    • Competitive phosphorylation where different kinases target overlapping regions

    • Combinatorial effects where specific patterns of multi-site phosphorylation create unique signaling outputs

  • Interaction with non-phosphorylation modifications:

    • Ubiquitination - potentially influenced by serine phosphorylation status

    • Glycosylation - may affect accessibility of kinases to specific residues

    • Acetylation - emerging modification with potential regulatory functions

  • Experimental approaches:

    • Site-directed mutagenesis to create phosphomimetic (S1071D/E) or phospho-dead (S1071A) mutants

    • Kinase inhibitor profiling to identify relevant upstream enzymes

    • Proteomics analysis of interactomes specific to phosphorylation status

  • Methodological considerations:

    • Use of appropriate phosphatase inhibitors to preserve physiological modification patterns

    • Analysis of intact proteins to maintain modification relationships

    • Consideration of spatio-temporal regulation of different modifications

Research has shown that AIB1 knockdown affects both tyrosine and serine/threonine phosphorylation of EGFR, suggesting complex regulatory networks governing receptor modification . Understanding these relationships is critical for developing more effective therapeutic strategies.

How does phosphorylation at S1071 compare between different isoforms and family members of EGFR?

EGFR (ErbB1) belongs to a family of receptors including ErbB2, ErbB3, and ErbB4, with significant implications for comparing phosphorylation patterns:

  • Comparative analysis across ErbB family members:

    • Sequence alignment to identify equivalent or distinct phosphorylation sites

    • Conservation of S1071 or analogous sites across the family

    • Distinct regulatory mechanisms potentially affecting these sites

  • EGFR isoform considerations:

    • Alternative splicing produces EGFR variants with potentially different regulation

    • Isoform 2 may act as an antagonist of EGF action , potentially with altered phosphorylation patterns

    • Expression pattern differences (e.g., isoform 2 expressed in ovarian cancers )

  • Experimental approaches:

    • Isoform-specific detection using selective antibodies

    • Recombinant expression of different family members for comparative analysis

    • Receptor co-expression studies to determine heterodimer effects on phosphorylation

  • Functional implications:

    • How phosphorylation patterns influence homo- versus heterodimerization

    • Differential sensitivity to therapeutic antibodies or small molecule inhibitors

    • Potential for isoform-specific targeting strategies

Research indicates that EGFR isoforms may have distinct functions, with isoform 2 potentially acting as an antagonist of EGF action . Understanding the phosphorylation patterns across these variants could reveal important regulatory mechanisms and therapeutic opportunities.

What are emerging technologies for sensitive detection of low-abundance Phospho-EGFR (S1071)?

Recent technological advances have significantly improved detection of low-abundance phosphorylation events:

  • Enhanced sensitivity methods:

    • Proximity ligation assay (PLA) - allows visualization of specific phosphorylation events in situ with single-molecule sensitivity

    • Single-molecule pull-down (SiMPull) - combines immunoprecipitation with single-molecule fluorescence detection

    • Nanoscale antibody arrays - miniaturized immunoassays with femtomolar sensitivity

  • Signal amplification approaches:

    • Tyramide signal amplification (TSA) - enzymatic deposition of fluorescent tyramide for significantly enhanced signal

    • Rolling circle amplification (RCA) - DNA amplification linked to antibody binding events

    • Quantum dot-conjugated secondary antibodies - brighter and more stable than conventional fluorophores

  • Novel sample preparation strategies:

    • Laser capture microdissection to isolate specific cell populations from heterogeneous tissues

    • Digital protein analysis through single-cell western blotting

    • Selective phosphopeptide enrichment using titanium dioxide or immobilized metal affinity chromatography

Research on combination antibody treatments has demonstrated the ability to downregulate EGFR surface expression by up to 80% , highlighting the need for increasingly sensitive detection methods to capture the remaining receptor population and its phosphorylation status.

How can phospho-flow cytometry be optimized for Phospho-EGFR (S1071) detection in heterogeneous samples?

Phospho-flow cytometry offers unique advantages for analyzing phosphorylation events in mixed cell populations:

  • Sample preparation optimization:

    • Critical fixation timing to capture transient phosphorylation events

    • Permeabilization protocol selection based on epitope accessibility

    • Buffer compositions that preserve phosphorylation while enabling antibody penetration

  • Antibody validation for flow cytometry:

    • Confirmation of specificity in positive control systems (e.g., EGF-stimulated A431 cells)

    • Titration to determine optimal concentration for signal-to-noise ratio

    • Testing with phosphatase-treated controls to confirm phospho-specificity

  • Multi-parameter analysis strategies:

    • Combined surface marker and phospho-epitope detection

    • Inclusion of total EGFR measurement for normalization

    • Integration with cell cycle analysis or viability markers

  • Data analysis approaches:

    • Phosphorylation-specific gating strategies

    • Population-level versus single-cell analysis

    • Visualization tools for complex phosphorylation relationships

While Phospho-EGFR (S1071) antibodies have not been extensively validated for flow cytometry, approaches similar to those used for phospho-EGFR (Y1068) detection in flow cytometry could be adapted, with appropriate optimization and validation steps.

What is the potential for Phospho-EGFR (S1071) as a biomarker in cancer diagnosis or treatment selection?

Phosphorylation status of EGFR has significant potential as a biomarker in cancer:

  • Diagnostic applications:

    • Differential phosphorylation patterns between normal and malignant tissues

    • Correlation with specific cancer subtypes or stages

    • Potential for non-invasive detection in liquid biopsies

  • Predictive biomarker potential:

    • Association with response to EGFR-targeted therapies

    • Indication of specific resistance mechanisms

    • Guidance for combination therapy approaches

  • Methodological considerations for clinical implementation:

    • Sample collection and preservation protocols to maintain phosphorylation status

    • Standardization of detection methods across clinical laboratories

    • Development of quantitative cutoff values for clinical decision-making

  • Evidence from existing research:

    • Studies have identified three sites (pY1092, pY1110, pY1172) correlating with activating mutations while three sites (pY1110, pY1172, pY1197) correlated with erlotinib sensitivity

    • S1071 phosphorylation status could potentially provide complementary information to these established markers

The relationship between S1071 phosphorylation and clinical outcomes remains an important area for investigation, with potential significance for patient stratification and treatment selection strategies.

How does the phosphorylation status of EGFR at S1071 compare between different cancer types and stages?

Understanding the pattern of S1071 phosphorylation across cancer types could provide valuable insights:

  • Comparative analysis across cancer types:

    • Epithelial cancers with known EGFR involvement (lung, colorectal, head and neck)

    • Comparison with cancers where EGFR plays a variable role (breast, pancreatic)

    • Correlation with EGFR expression levels and mutation status

  • Progression analysis:

    • Changes in phosphorylation patterns during tumor development

    • Association with invasive or metastatic phenotypes

    • Comparison between primary tumors and metastatic lesions

  • Methodological approaches:

    • Tissue microarray analysis with phospho-specific immunohistochemistry

    • Quantitative image analysis for standardized scoring

    • Complementary proteomic profiling of selected samples

Research has demonstrated that EGFR phosphorylation patterns differ between cancer types, with studies showing distinct patterns between lung cancer cells (A549), breast cancer cells (MDA-MB-231), and pancreatic cancer cells (PANC-1) . These differences may reflect tissue-specific signaling networks and could influence therapeutic vulnerabilities.

How can computational modeling integrate Phospho-EGFR (S1071) data to understand system-level regulation?

Systems biology approaches can provide valuable insights into complex EGFR signaling networks:

  • Kinetic modeling approaches:

    • Ordinary differential equation (ODE) models of EGFR phosphorylation/dephosphorylation

    • Parameter estimation using time-course experimental data

    • Sensitivity analysis to identify critical regulatory nodes

  • Network-based analysis:

    • Integration of phospho-EGFR (S1071) data with larger signaling networks

    • Identification of feedback and feedforward loops involving this site

    • Prediction of system-level responses to perturbations

  • Multi-scale modeling considerations:

    • Linking molecular events (phosphorylation) to cellular phenotypes

    • Integration of spatial aspects of receptor trafficking

    • Incorporation of tissue-level heterogeneity

  • Data requirements and experimental design:

    • Time-resolved measurements across multiple conditions

    • Quantitative data for model training and validation

    • Targeted perturbation experiments to test model predictions

Research has shown that EGFR activates at least four major downstream signaling cascades: RAS-RAF-MEK-ERK, PI3 kinase-AKT, PLCgamma-PKC and STATs modules . Integrating S1071 phosphorylation data into these signaling networks could reveal novel regulatory mechanisms and therapeutic opportunities.

What are the best experimental designs for studying the dynamics of S1071 phosphorylation in the context of feedback regulation?

EGFR signaling involves complex feedback mechanisms that influence phosphorylation dynamics:

  • Temporal sampling strategies:

    • High-density early timepoints (seconds to minutes) to capture rapid phosphorylation events

    • Extended timepoints (hours to days) to observe adaptation and feedback effects

    • Synchronized cell populations to reduce heterogeneity

  • Perturbation approaches:

    • Dose-response studies with varying EGF concentrations

    • Pulses versus sustained stimulation paradigms

    • Inhibitor time-course studies with pathway-specific inhibitors

  • Readout technologies:

    • Multiplexed detection of multiple phosphorylation sites simultaneously

    • Live-cell reporters for real-time monitoring where feasible

    • Single-cell approaches to capture population heterogeneity

  • Experimental considerations for feedback analysis:

    • Combined inhibition of receptor and downstream pathways

    • Chemical-genetic approaches for rapid and specific kinase inhibition

    • Protein synthesis inhibitors to distinguish between immediate and delayed feedback mechanisms

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.