EREG Antibody, Biotin conjugated

Shipped with Ice Packs
In Stock

Description

Antigen and Antibody Specificity

  • Target: EREG protein (epiregulin), a ligand for EGFR, ErbB3, and ErbB4 receptors.

  • Antibody Type: Primarily polyclonal or monoclonal antibodies raised in rabbits or mice.

  • Conjugation: Biotin (vitamin B7) is covalently linked to the antibody, enabling interaction with streptavidin/avidin systems for enhanced detection sensitivity.

PropertyDetailsSource
HostRabbit or mouse
ClonalityPolyclonal or monoclonal (e.g., E235 monoclonal)
Conjugation MethodBiotin-streptavidin binding system
ApplicationsELISA, Western blot, immunohistochemistry (IHC), flow cytometry (FCM)

**2.1 ELISA and Sandwich Assays

Biotin-conjugated EREG antibodies are widely used as detection antibodies in enzyme-linked immunosorbent assays (ELISA). For example:

  • Sandwich ELISA: A capture antibody binds EREG in samples, followed by detection with a biotin-conjugated antibody. Streptavidin-HRP amplifies the signal, enabling quantitative detection down to 3.9 pg/ml .

  • Direct ELISA: Biotin-antibody detects EREG directly on plates, with optical density measured at 405–650 nm .

ELISA Kit ComponentDescriptionDetection RangeSource
Capture AntibodyUnconjugated anti-EREG antibody (e.g., ARG66150)15.6–1000 pg/ml
Detection AntibodyBiotin-conjugated anti-EREG antibody (e.g., CSB-PA007779ND01HU)<3.9 pg/ml (LLD)

**2.2 Western Blot and Immunohistochemistry

  • Western Blot: Biotin-antibodies enhance signal detection under reducing/non-reducing conditions. For example, ARG66151 (anti-EGFR, biotin) shows clear bands for human EGFR at 0.24–250 ng .

  • IHC: Used to localize EREG in cancer tissues. Biotinylated antibodies are paired with streptavidin-alkaline phosphatase or peroxidase for staining .

**2.3 Therapeutic and Targeted Delivery

  • Antibody-Drug Conjugates (ADCs): Biotin-conjugated antibodies enable coupling with cytotoxic agents or nanoparticles (e.g., PCNDs—phase-change nano-droplets) for targeted cancer therapy. In preclinical studies, 9E5 (anti-EREG) conjugated to PCNDs selectively internalized into EREG-expressing cells, inducing intracellular vaporization upon laser activation .

  • Cancer Biomarker: High EREG expression correlates with sensitivity to EGFR inhibitors like erlotinib in head and neck squamous cell carcinoma (HNSCC) .

**3.1 EREG in Oncogenesis

  • EREG-Driven Signaling: EREG activates EGFR, ERK, AKT, and STAT3 pathways, promoting proliferation and survival. In HNSCC, EREG expression mimics EGFR mutations, sustaining signaling even in the presence of EGFR inhibitors .

  • Biomarker Potential: EREG overexpression is observed in colorectal, lung, gastric, and renal cancers. Biotin-conjugated antibodies enable precise quantification of EREG in serum or tissues for diagnostic screening .

**3.2 Key Studies

StudyMethodologyFindingsSource
EREG in HNSCCIHC, Western blot, erlotinib sensitivity assaysHigh EREG expression predicts erlotinib response; EREG sustains EGFR signaling
PCNDs for Cancer TherapyBiotin-9E5 antibody conjugation to phase-change nano-dropletsSelective intracellular vaporization in EREG-positive cells
EREG as a DiagnosticSandwich ELISA with biotin-antibodySensitive detection of EREG in serum (LLD: 3.9 pg/ml)

**4.1 Product Comparison

SupplierProduct CodeApplicationsConjugateReactivityPrice (USD)Source
CusabioCSB-PA007779ND01HUELISABiotinHuman$166–$295
Boster BioA06692ELISA, IHCUnconjugatedHuman, Mouse, Rat$209–$243
DIMA BiotechnologyDMC424Flow CytometryBiotinHuman$670

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Typically, we can ship your order within 1-3 business days of receiving it. The delivery time may vary depending on the purchasing method or location. For specific delivery times, please consult your local distributors.
Synonyms
Proepiregulin [Cleaved into: Epiregulin (EPR)], EREG
Target Names
Uniprot No.

Target Background

Function
Epiregulin (EREG) acts as a ligand for the epidermal growth factor receptor (EGFR) and ERBB4. It stimulates tyrosine phosphorylation of EGFR and ERBB4. EREG contributes to various physiological processes including inflammation, wound healing, tissue repair, and oocyte maturation by regulating angiogenesis and vascular remodeling, and by stimulating cell proliferation.
Gene References Into Functions
  1. In Caco-2 CFTR-shRNA cells, EREG, an EGFR ligand, is overexpressed due to an active IL-1beta autocrine loop that indirectly activates EGFR. This constitutes new signaling effectors for the CFTR signaling pathway, downstream of CFTR, Cl(-) , and IL-1beta. PMID: 29091309
  2. Our study demonstrated that EGFRs were constitutively activated in metastatic lung subtypes of salivary adenoid cystic carcinoma cells, and this activation was induced by autocrine expression of epiregulin. PMID: 26958807
  3. This study investigates how EGFR ligands epiregulin (EREG) and epigen (EPGN) stabilize different dimeric conformations of the EGFR extracellular region. The results reveal how responses to different EGFR ligands are determined by receptor dimerization strength and signaling dynamics. These findings have broad implications for understanding receptor tyrosine kinase (RTK) signaling specificity. PMID: 28988771
  4. EREG and MMP-1 were found to be elevated in nasal polyp and uncinate tissues in patients with Chronic rhinosinusitis with nasal polyps. PMID: 28398769
  5. Upregulation of EREG expression through promoter demethylation might be a crucial mechanism in activating the EGFR pathway during the genesis of colorectal adenocarcinoma (CRC) and potentially other cancers. PMID: 27270421
  6. EREG and AREG are strongly regulated by methylation, and their expression is associated with CIMP status and primary tumor site. PMID: 27272216
  7. This research examines the three-dimensional structure of the EPR antibody (the 9E5(Fab) fragment) in the presence and absence of EPR. PMID: 26627827
  8. These studies collectively identify a novel pathway involving EREG and MMP-1 that contributes to the formation of early-stage breast cancer. PMID: 26215578
  9. The findings suggest that EREG is one of the molecules implicated in glioma malignancy. PMID: 24470554
  10. The data indicate that the effects of epiregulin (EREG) and V-ATPase (TCIRG1) single nucleotide polymorphism (SNP) on pulmonary tuberculosis susceptibility, if present, are dependent on gene-gene interactions in West African populations. PMID: 24898387
  11. Patients homozygous for the minor allele A of EREG rs12641042 had a significantly higher 3-year survival rate than patients with allele C (HR 0.48; P=0.034). However, significance was lost in multivariable analysis. PMID: 25203737
  12. Epiregulin is a transcriptional target of TSC2 (tuberin). PMID: 24748662
  13. Epiregulin promotes the proliferation of liver progenitor cells and DNA synthesis by hepatocytes. It is upregulated in the serum of patients with liver injury. PMID: 24812054
  14. Plasma HGF and EREG levels are associated with resistance to treatment with anti-EGFR antibodies in KRAS wild-type patients with metastatic colorectal cancer. PMID: 24800946
  15. The data suggest that EREG (epiregulin), AREG (amphiregulin), and BTC (betacellulin) induced prostaglandin E2 production by induction of COX-2 (prostaglandin-endoperoxide synthase 2) through MAP kinase signaling in granulosa cells. PMID: 24092824
  16. In pre-treated K-ras wild-type status colorectal cancer, patients with high EREG gene expression appear to benefit more from cetuximab therapy compared with those with low expression. PMID: 24335920
  17. EREG may contribute to glioma progression under the control of IRE1a. PMID: 24330607
  18. Keratinocyte hyperproliferation in cholesteatoma is promoted through overexpression of epiregulin by subepithelial fibroblasts via epithelial-mesenchymal interactions, which may play a crucial role in the pathogenesis of middle ear cholesteatoma. PMID: 23826119
  19. Depletion of Epiregulin with shRNA inhibited SCAP proliferation. PMID: 23829318
  20. The data indicate that epiregulin (EREG) expression significantly correlated with KRAS expression or KRAS copy number in KRAS-mutant non-small-cell lung cancer (NSCLC) cell lines. PMID: 22964644
  21. EREG-AREG and NRG1, which are members of the epidermal growth factor (EGF) family, seem to modulate Bhecet's Disease susceptibility through main effects and gene-gene interactions. PMID: 23625463
  22. We did not find a correlation between the presence of a K-ras mutation and the presence of Epiregulin and Amphiregulin in colon cancer tissue. PMID: 23885463
  23. Apical mistrafficking of EREG crystallizes an apical EGFR signaling complex that may be uncoupled from basolateral regulatory restraints leading to cell transformation. PMID: 23671122
  24. FBXL11 inhibited osteo/dentinogenic differentiation potential in MSC cells by associating with BCOR, then increasing histone K4/36 methylation in Epiregulin promoter to repress Epiregulin transcription. PMID: 23074094
  25. EREG gene expression was low in 7 out of 11 gastric cancer cells, and this downregulation was mediated by aberrant CpG methylation of the EREG promoter. PMID: 22508389
  26. Epiregulin (EREG) variation is associated with susceptibility to tuberculosis. PMID: 22170233
  27. The data suggest that expression status of AR and EPI mRNAs might be evaluated as dynamic predictors of response in KRAS WT patients receiving any cetuximab-based therapy. PMID: 21161326
  28. It is suggested that follow-up of the expression of Ep can serve as a reliable early indication of the development of ovarian cancer. PMID: 21769422
  29. Blockade of epiregulin reduced the growth of hTERT-BJ cells and colony formation of hTERT-transformed fibroblasts. Moreover, inhibition of epiregulin function in immortal hTERT-BJ cells triggered a senescence program. PMID: 12702554
  30. Epiregulin might be a more important tumor growth regulator of malignant fibrous histiocytoma through autocrine or paracrine pathways, when compared with betacellulin. PMID: 15274392
  31. Upregulation of the epiregulin and amphiregulin expression is part of the signal transduction pathway that leads to ovulation and luteinization in the human ovary. PMID: 15474502
  32. The findings demonstrated that PGE2 may mimic LH action at least in part by the activation of amphiregulin and epiregulin biosynthesis in human granulosa cells. PMID: 16888076
  33. Epiregulin, COX2, and MMP1 and 2 collectively facilitate the assembly of new tumor blood vessels, the release of tumor cells into the circulation, and the breaching of lung capillaries by circulating tumor cells to seed pulmonary metastasis. PMID: 17429393
  34. This is the first report of EREG expression in breast cancer (45.5% of breast cancers studied). It is preferentially expressed in breast tumors co-expressing HER2/HER4. PMID: 17962208
  35. Epiregulin played an autocrine role in the proliferation of corneal epithelial cells presumably through cross-induction with other EGF family members. PMID: 18079685
  36. Hamartomatous TSC skin tumors are induced by paracrine factors released by two-hit cells in the dermis, and proliferation with mTOR activation of the overlying epidermis is an effect of epiregulin. PMID: 18292222
  37. Increased epiregulin is associated with oral squamous cell carcinomas. PMID: 18497965
  38. Epiregulin has a protective effect against apoptosis in the human corpus luteum. PMID: 18835871
  39. The regulatory mechanism of epiregulin expression in Ki-ras-transformed 267B1 prostate epithelial cells was studied. PMID: 18948081
  40. Epiregulin expression correlates with advanced disease, is EGFR dependent, and confers invasive properties on non-small cell lung cancer cells. PMID: 19138957

Show More

Hide All

Database Links

HGNC: 3443

OMIM: 602061

KEGG: hsa:2069

STRING: 9606.ENSP00000244869

UniGene: Hs.115263

Subcellular Location
[Epiregulin]: Secreted, extracellular space.; [Proepiregulin]: Cell membrane; Single-pass type I membrane protein.
Tissue Specificity
In normal adults, expressed predominantly in the placenta and peripheral blood leukocytes. High levels were detected in carcinomas of the bladder, lung, kidney and colon.

Q&A

What is the significance of EREG antibody in oncology research?

Epiregulin (EREG) has emerged as a critical biomarker in cancer research, particularly in non-small cell lung cancer (NSCLC). Immunohistochemical studies have demonstrated that EREG expression correlates significantly with nodal metastasis and shorter survival duration in NSCLC patients . When detected in tumor biopsy samples, EREG staining was observed in 64.7% (237 of 366) of NSCLC cases, predominantly in the cytoplasmic compartment . Furthermore, EREG has been shown to confer invasive properties on cancer cells, as demonstrated through Matrigel invasion assays where anti-epiregulin neutralizing antibodies inhibited the invasive capacity of EGFR-mutant NSCLC cell lines . This makes EREG antibodies valuable tools for both diagnostic applications and for understanding the biological mechanisms of cancer progression.

What are the typical protocols for using EREG antibody, biotin conjugated in immunohistochemistry?

When using biotin-conjugated EREG antibodies in immunohistochemistry, researchers typically follow a protocol similar to that used for other biotinylated antibodies in cancer tissue analysis. First, tissue sections are deparaffinized, rehydrated, and subjected to antigen retrieval using appropriate buffer systems (typically citrate or EDTA-based) . Endogenous peroxidase activity is quenched using hydrogen peroxide (3-5% solution), and non-specific binding is blocked with appropriate sera or protein solutions.

The biotin-conjugated EREG antibody is then applied at optimized concentrations (typically 1-10 μg/mL) and incubated for 60-90 minutes at room temperature or overnight at 4°C . After washing with PBS or TBS buffer, streptavidin-HRP is applied for 30-60 minutes. Following additional washing steps, the reaction is visualized using 3,3'-diaminobenzidine (DAB) or other chromogens . Counterstaining with hematoxylin provides nuclear detail before mounting.

Importantly, researchers should always include appropriate positive controls (tissues known to express EREG, such as specific NSCLC samples) and negative controls (primary antibody omission or isotype-matched control antibodies) .

How can site-specific biotin conjugation improve the performance of EREG antibodies compared to conventional conjugation methods?

Site-specific biotin conjugation represents a significant advancement over conventional random conjugation methods, particularly for critical reagents like EREG antibodies. Conventional conjugation typically targets primary amine groups (lysine residues) throughout the antibody structure, which can lead to heterogeneous conjugates with variable performance . In contrast, site-specific conjugation directs the biotin molecule to precise locations, preserving antibody binding properties.

Research has demonstrated that site-specific conjugation using reductive alkylation approaches enables the majority of antibody molecules to be labeled at the desirable N-terminus, which minimizes modification of the protein after labeling . This precision is especially crucial for therapeutic antibodies and their detection in anti-drug antibody (ADA) assays, where maintaining epitope recognition is paramount.

Comparative biophysical characterization has revealed that site-specifically biotin-conjugated antibodies demonstrate superior quality compared to those prepared using conventional amine coupling methods . For EREG antibodies, which may target specific epitopes critical for neutralizing function, site-specific conjugation helps ensure consistent performance in both research and clinical applications.

What strategies can minimize biotin interference in immunoassays using biotin-conjugated EREG antibodies?

Biotin interference is a significant concern in immunoassays utilizing the biotin-streptavidin detection system. When analyzing samples with high endogenous biotin, such as those from individuals taking biotin supplements or certain therapeutic preparations, several strategies can mitigate interference:

  • Sample pre-treatment: Implementing a streptavidin pre-treatment step to sequester free biotin in samples before performing the assay .

  • Alternative detection systems: For samples suspected of high biotin content, consider using non-biotin detection methods such as direct HRP-conjugated antibodies or alternative tags .

  • Serial dilution assessment: Performing serial dilutions of samples can help identify potential biotin interference, as demonstrated in the table below from experimental data:

Sample DilutionBiotin Concentration (ng/mL)Signal OutcomeInterference Assessment
Undiluted5250ReducedHigh interference
1:22620ReducedModerate interference
1:41310Slightly reducedLow interference
1:8650NormalMinimal interference
1:16320NormalNo significant interference
1:32160NormalNo interference

This data demonstrates how serial dilution can reveal and potentially overcome biotin interference effects .

  • Control implementation: Including specific assay controls to monitor direct cross-reactions between the antigen, blocking reagents, target antibodies, biotin, and HRP-streptavidin interactions .

How does EREG expression correlate with EGFR mutation status in cancer samples, and what implications does this have for biotin-conjugated EREG antibody applications?

EREG expression demonstrates a strong correlation with EGFR mutation status in cancer samples, particularly in non-small cell lung cancer. Research has shown that NSCLC cell lines with activating EGFR mutations express significantly higher levels of EREG compared to those with wild-type EGFR . This relationship has important implications for biotin-conjugated EREG antibody applications in both diagnostic and research contexts.

In EGFR-mutant NSCLC cell lines (such as HCC827, H3255, and HCC2279), EREG expression is not merely correlative but functionally dependent on EGFR signaling. Treatment with the EGFR tyrosine kinase inhibitor gefitinib sharply reduces EREG mRNA levels, demonstrating that EREG is part of a feed-forward loop by which EGFR maintains the activity of ErbB dimeric complexes in EGFR-mutant NSCLC cells .

For biotin-conjugated EREG antibody applications, these findings suggest:

  • Diagnostic stratification: EREG expression detection using biotin-conjugated antibodies could help stratify NSCLC patients based on likely EGFR mutation status, potentially identifying candidates for EGFR-targeted therapies.

  • Therapeutic monitoring: Monitoring changes in EREG expression using these antibodies may provide insights into treatment efficacy for EGFR-targeted therapies.

  • Mechanism studies: Biotin-conjugated EREG antibodies can be valuable tools for investigating the interplay between EGFR signaling and EREG expression in different cellular contexts .

What are the optimal analytical validation parameters for assays employing biotin-conjugated EREG antibodies?

Rigorous analytical validation is essential for assays employing biotin-conjugated EREG antibodies. Key validation parameters should include:

  • Specificity assessment: Evaluate cross-reactivity with related ErbB ligands such as EGF, TGF-α, and amphiregulin through competitive binding studies and western blot analysis . This is particularly important given the structural similarities between these growth factors.

  • Sensitivity determination: Establish the lower limit of detection (LLOD) and lower limit of quantification (LLOQ) using recombinant EREG protein standards. For immunohistochemical applications, sensitivity should be determined using tissues with known EREG expression levels .

  • Linearity evaluation: Assess signal linearity across a range of EREG concentrations to ensure accurate quantification. This should include evaluation of potential hook effects at high concentrations.

  • Reproducibility testing: Determine intra- and inter-assay coefficients of variation (CVs) through repeated measurements of quality control samples containing low, medium, and high EREG concentrations. Acceptable CVs should typically be <15% for quantitative assays and <20% for semi-quantitative applications .

  • Biotin interference assessment: Evaluate the impact of varying biotin concentrations (20-5250 ng/mL) on assay performance to establish tolerance limits and appropriate sample dilution strategies .

  • Antibody binding stability: Monitor the stability of biotin-conjugated EREG antibodies under various storage conditions (4°C, -20°C, -80°C) and after multiple freeze-thaw cycles to establish optimal handling procedures.

How can researchers optimize site-specific biotin conjugation ratios for EREG antibodies?

Optimizing site-specific biotin conjugation ratios for EREG antibodies requires a systematic approach to ensure maximum functionality while maintaining antibody binding properties. The key considerations include:

  • Biotin-to-antibody ratio determination: Start with a range of molar ratios (typically 2:1 to 20:1 biotin:antibody) during the conjugation reaction . Each ratio should be evaluated for conjugation efficiency using MALDI-TOF mass spectrometry or other appropriate analytical techniques.

  • Reductive alkylation approach: For N-terminal site-specific conjugation, researchers should optimize the reductive alkylation conditions. This involves careful control of pH (typically 5.0-6.0), temperature (usually 25-37°C), and reaction time (4-24 hours) . These parameters significantly influence the selectivity for N-terminal versus lysine modification.

  • Functional assessment: Test each conjugate ratio for retention of EREG binding using ELISA or surface plasmon resonance (SPR). The optimal ratio will provide sufficient detection sensitivity without compromising antibody affinity or specificity .

  • Signal-to-noise optimization: Evaluate background signal in relevant assay formats. Excessive biotin conjugation can increase non-specific binding, while insufficient conjugation may result in inadequate detection sensitivity .

  • Stability evaluation: Assess the stability of different conjugate ratios during storage and under assay conditions. Higher conjugation ratios sometimes lead to reduced stability due to conformational changes in the antibody structure .

The optimal conjugation ratio typically allows the majority of antibody molecules to be labeled with biotin at the desired position while minimizing off-target modifications that could impact binding properties.

What controls should be included when using biotin-conjugated EREG antibodies in experimental designs?

A comprehensive set of controls is essential when using biotin-conjugated EREG antibodies to ensure reliable and interpretable results:

  • Antigen omission control: Wells without the target antigen should be included to assess non-specific binding of the biotin-conjugated EREG antibody to the solid phase .

  • Primary antibody omission control: Include samples treated with all reagents except the biotin-conjugated EREG antibody to evaluate background from secondary detection systems .

  • Isotype-matched control: Use an irrelevant biotin-conjugated antibody of the same isotype to determine non-specific binding due to antibody class characteristics .

  • Biotin interference control: Include samples with known high biotin concentrations (5250 ng/mL) alongside serial dilutions (2620, 1310, 650, 320, 160, 80, 40, and 20 ng/mL) to assess potential interference effects .

  • Competitive inhibition control: Pre-incubate the biotin-conjugated EREG antibody with soluble recombinant EREG protein before application to verify binding specificity .

  • Cross-reactivity assessment: Include related ErbB ligands (EGF, TGF-α, amphiregulin) to evaluate potential cross-reactivity of the EREG antibody .

  • System suitability controls: Incorporate positive and negative control samples with known EREG expression levels to confirm assay performance across experiments .

  • Streptavidin-only control: Apply streptavidin-HRP without prior biotin-conjugated antibody to assess direct binding of streptavidin to sample components .

These controls collectively enable researchers to distinguish true positive signals from various sources of background or non-specific interactions.

How can researchers evaluate whether biotin conjugation affects the binding characteristics of EREG antibodies?

Evaluating the impact of biotin conjugation on EREG antibody binding characteristics requires multiple complementary approaches:

  • Comparative affinity measurement: Determine the binding kinetics (kon, koff, and KD) of unconjugated and biotin-conjugated EREG antibodies using surface plasmon resonance (SPR) or bio-layer interferometry (BLI) . A significant increase in KD (decreased affinity) after conjugation may indicate compromised binding properties.

  • Epitope mapping: Perform epitope binning or mapping studies to ensure that biotin conjugation hasn't altered the antibody's ability to recognize specific EREG epitopes . This is particularly important when the antibody's functionality depends on binding to specific regions of the EREG protein.

  • Competitive binding assays: Compare the ability of conjugated and unconjugated antibodies to compete for EREG binding sites. Equal competition suggests preservation of binding characteristics .

  • Functional neutralization assessment: If the EREG antibody has neutralizing properties, evaluate whether biotin conjugation affects its ability to inhibit EREG-mediated cellular responses. This can be assessed using invasion assays or EGFR phosphorylation studies, as EREG signaling through EGFR has been shown to promote invasion in NSCLC cell lines .

  • Thermal stability analysis: Measure melting temperatures (Tm) of conjugated and unconjugated antibodies using differential scanning calorimetry (DSC) or differential scanning fluorimetry (DSF). Significant decreases in Tm after conjugation may indicate structural destabilization .

  • Size-exclusion chromatography: Analyze both antibody preparations to detect potential aggregation or conformational changes resulting from biotin conjugation .

How should researchers interpret discrepancies between EREG antibody staining patterns and clinical outcomes?

When interpreting discrepancies between EREG antibody staining patterns and clinical outcomes, researchers should consider several factors that may explain these variations:

  • Heterogeneity of EREG expression: EREG expression in tumors may exhibit spatial heterogeneity, with varying expression levels across different regions of the tumor . This heterogeneity can lead to sampling bias if tissue microarrays or limited biopsy samples are analyzed.

  • Context-dependent EREG functions: EREG may have different functional effects depending on the molecular context of the tumor. Research has shown that while EREG expression correlates with advanced nodal (N2) disease in NSCLC (p = 0.04), its impact on survival may be influenced by other molecular alterations . After correcting for covariates (age, pathologic nodal and tumor stage, and histologic subtype), the correlation with shorter survival duration becomes more evident (p = 0.014) .

  • Signaling pathway integration: EREG is part of a complex signaling network involving other ErbB ligands and receptors. The expression and activity of these other components may modulate EREG's effects on clinical outcomes . For instance, in EGFR-mutant NSCLC cells, EREG expression is regulated by specific downstream mediators of EGFR, including MAPK/ERK and p38 .

  • Technical considerations: Differences in biotin-conjugated antibody performance, detection systems, or scoring methods across studies can contribute to discrepancies. Standardization of immunohistochemical protocols and scoring systems is essential for reliable comparisons .

  • Multivariate analysis approach: When discrepancies arise, multivariate analysis incorporating additional clinicopathological and molecular variables should be performed to identify potential confounding factors or synergistic effects .

What are the potential sources of false positive and false negative results when using biotin-conjugated EREG antibodies?

Understanding potential sources of false results is crucial for accurate data interpretation when using biotin-conjugated EREG antibodies:

Sources of false positive results:

  • Endogenous biotin interference: High levels of endogenous biotin in biological samples can bind to streptavidin-HRP and generate signals even in the absence of the target antigen . This is particularly problematic in samples from patients taking biotin supplements or certain therapeutics.

  • Cross-reactivity with related proteins: EREG belongs to the EGF family of growth factors, which share structural similarities. Insufficient antibody specificity may lead to detection of other family members like EGF, TGF-α, or amphiregulin .

  • Non-specific binding: Inadequate blocking or the presence of Fc receptors in tissue samples can cause non-specific binding of the biotin-conjugated antibody, resulting in background staining .

  • Endogenous peroxidase activity: Insufficient quenching of endogenous peroxidase activity in tissue sections can lead to false positive signals when using HRP-based detection systems .

Sources of false negative results:

  • Epitope masking: Biotin conjugation near the antibody's antigen-binding region can sterically hinder epitope recognition, particularly if random conjugation methods are used rather than site-specific approaches .

  • Fixation artifacts: Formalin fixation can cross-link proteins and mask epitopes, potentially preventing antibody binding. This underscores the importance of appropriate antigen retrieval methods .

  • Pre-analytical variables: Prolonged ischemia time before fixation, improper fixation duration, or suboptimal processing can degrade EREG protein in tissue samples .

  • Biotin-streptavidin system oversaturation: Excess free biotin in samples can saturate available streptavidin binding sites, preventing detection of biotinylated antibodies bound to the target antigen .

  • Threshold setting: Inappropriate threshold settings for positive staining can lead to false classification of low-level positive samples as negative .

How can multiplex assays incorporating biotin-conjugated EREG antibodies be optimized to reduce cross-reactivity?

Optimizing multiplex assays that include biotin-conjugated EREG antibodies requires strategic approaches to minimize cross-reactivity:

  • Antibody selection and validation: Choose EREG antibodies with demonstrated specificity against related ErbB ligands. Validate each antibody individually before incorporation into multiplex formats, testing against a panel of related proteins to assess cross-reactivity profiles .

  • Sequential detection strategies: Implement sequential rather than simultaneous detection for biotin-conjugated antibodies. This approach allows complete washing and blocking between detection steps to minimize cross-detection .

  • Alternative conjugation chemistry: Consider using orthogonal labeling strategies where only one antibody in the multiplex panel uses biotin conjugation. Other antibodies can employ different detection tags such as fluorophores, enzymes, or metal isotopes for mass cytometry .

  • Spatial separation techniques: For tissue-based multiplex assays, employ spectral unmixing algorithms or sequential staining and imaging with antibody stripping between cycles to reduce signal overlap .

  • Cross-blocking experiments: Perform cross-blocking experiments with all antibodies in the multiplex panel to ensure they target distinct, non-overlapping epitopes on their respective targets .

  • Titration optimization: Carefully titrate each biotin-conjugated antibody to determine the minimum concentration that provides adequate signal-to-noise ratios. This minimizes potential cross-reactivity while maintaining sensitivity .

  • Multiplexed controls: Implement comprehensive controls, including single-stain controls, fluorescence-minus-one (FMO) controls, and isotype controls for each antibody in the panel to accurately assess and correct for spectral overlap and non-specific binding .

  • Data analysis algorithms: Utilize advanced computational approaches such as machine learning algorithms to distinguish true signals from background or cross-reactive signals in complex multiplex data sets.

How might site-specific biotin conjugation of EREG antibodies advance personalized medicine approaches for cancer treatment?

Site-specific biotin conjugation of EREG antibodies has significant potential to advance personalized medicine for cancer treatment through several innovative approaches:

  • Companion diagnostics development: Site-specifically biotin-conjugated EREG antibodies could serve as superior reagents for companion diagnostic assays that identify patients likely to benefit from EGFR-targeted therapies . The enhanced consistency and performance of these conjugates compared to conventional methods would improve diagnostic accuracy.

  • Circulating biomarker detection: These optimized antibodies could enable more sensitive detection of soluble EREG in liquid biopsies, potentially allowing for real-time monitoring of treatment response and disease progression without invasive tissue sampling .

  • Theranostic applications: Leveraging the biotin-streptavidin system's versatility, EREG antibodies could be developed into theranostic tools that both detect EREG-expressing tumors and deliver therapeutic payloads. The site-specific conjugation ensures consistent antibody performance in both diagnostic and therapeutic functions .

  • Predictive modeling integration: Quantitative data from assays using precisely conjugated EREG antibodies could improve predictive models that guide treatment decisions. Research has shown that EREG expression correlates with advanced disease and poorer outcomes in NSCLC, making it a valuable input parameter for such models .

  • Resistance mechanism identification: As EREG expression is EGFR-dependent and regulated by specific downstream mediators like MAPK/ERK and p38, site-specifically conjugated antibodies could help characterize resistance mechanisms to EGFR inhibitors by accurately monitoring changes in EREG expression patterns .

  • Combination therapy rationales: Precise quantification of EREG using optimized biotin-conjugated antibodies could identify patients who might benefit from combining EGFR inhibitors with other targeted therapies, such as those targeting invasion pathways that EREG has been shown to promote in cancer cells .

What emerging technologies might enhance the specificity and sensitivity of biotin-conjugated EREG antibody applications?

Several emerging technologies hold promise for enhancing the specificity and sensitivity of biotin-conjugated EREG antibody applications:

  • Click chemistry approaches: Bioorthogonal click chemistry reactions offer greater specificity for site-directed biotin conjugation, potentially improving the consistency and performance of EREG antibodies while maintaining their native binding properties .

  • Nanobody and single-domain antibody platforms: These smaller antibody formats provide advantages for site-specific biotin conjugation due to their reduced structural complexity. Their application to EREG detection could improve tissue penetration and reduce background in imaging applications .

  • Proximity ligation assays (PLA): Combining biotin-conjugated EREG antibodies with PLA technology could significantly enhance detection sensitivity by generating amplifiable DNA signals only when antibodies bind in close proximity, allowing visualization of protein-protein interactions involving EREG .

  • Digital ELISA platforms: Technologies like Simoa (single molecule array) could be adapted for use with biotin-conjugated EREG antibodies, potentially enabling detection at femtomolar concentrations in biological fluids—orders of magnitude more sensitive than conventional ELISA .

  • CRISPR-based antibody engineering: CRISPR/Cas technologies enable precise genetic modification to introduce specific amino acids at defined positions in antibodies, facilitating highly controlled biotin conjugation sites that preserve binding characteristics .

  • Artificial intelligence for image analysis: Machine learning algorithms can enhance the interpretation of immunohistochemical data from biotin-conjugated EREG antibodies, improving quantification accuracy and identifying subtle staining patterns that correlate with clinical outcomes .

  • Microfluidic immunoassay platforms: Integration of biotin-conjugated EREG antibodies into microfluidic devices could reduce sample volume requirements, increase throughput, and improve sensitivity through optimized reaction kinetics and reduced diffusion distances .

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.