EPHA10 Antibody, HRP conjugated is a polyclonal antibody derived from rabbit hosts that specifically targets the Ephrin type-A receptor 10 protein. The antibody is chemically linked to Horseradish Peroxidase (HRP), an enzyme that facilitates detection through colorimetric or chemiluminescent reactions in various immunoassays. This conjugation eliminates the need for secondary antibody steps, streamlining experimental protocols and potentially reducing background noise in research applications .
The antibody is generated using a KLH-conjugated synthetic peptide derived from human EPHA10, specifically targeting the amino acid region 151-250 of the full 1008 amino acid sequence. This strategic targeting ensures specificity for the EPHA10 receptor while minimizing cross-reactivity with other related proteins . The polyclonal nature of this antibody means it recognizes multiple epitopes on the EPHA10 antigen, potentially increasing sensitivity compared to monoclonal alternatives.
The EPHA10 antibody exhibits specific reactivity patterns that define its utility in different experimental models and systems. Understanding these patterns is essential for researchers selecting appropriate antibodies for their specific research questions.
The antibody has been confirmed to react with EPHA10 from multiple species, demonstrating versatility across different experimental models:
This multi-species reactivity makes the antibody valuable for comparative studies and translational research where findings in animal models might be extrapolated to human systems.
Based on sequence homology and structural similarities, the antibody is predicted to recognize EPHA10 from several additional species, though these interactions have not been experimentally validated:
The wide range of predicted reactivity suggests conservation of the epitope region across multiple species, highlighting the evolutionary significance of this receptor domain.
The EPHA10 antibody, HRP conjugated, demonstrates utility across multiple immunological techniques, making it a versatile tool in the researcher's arsenal. Each application requires specific dilution parameters for optimal results.
The antibody has been validated for several common laboratory techniques with specific dilution recommendations for each application:
Application | Recommended Dilution | Description |
---|---|---|
Western Blotting (WB) | 1:300-5000 | Detection of denatured EPHA10 protein separated by electrophoresis |
Enzyme-Linked Immunosorbent Assay (ELISA) | 1:500-1000 | Quantitative detection of EPHA10 in solution |
Immunohistochemistry - Paraffin (IHC-P) | 1:200-400 | Visualization of EPHA10 in paraffin-embedded tissue sections |
Immunohistochemistry - Frozen (IHC-F) | 1:100-500 | Detection of EPHA10 in frozen tissue sections |
These dilution ranges provide flexibility for researchers to optimize conditions based on their specific sample types and detection systems . The HRP conjugation provides direct detection capability, eliminating the need for secondary antibody incubation steps in these applications.
In Western blot applications, the EPHA10 antibody has demonstrated specific binding to its target protein. When used at concentrations of 1-2 μg/mL with 293 cell lysates, the antibody detects a band corresponding to the predicted molecular weight of 109 kDa for the EPHA10 protein . This specificity in protein detection makes the antibody valuable for expression studies and protein characterization experiments.
Understanding the biological context of the target protein provides essential background for interpreting experimental results obtained using the EPHA10 antibody.
EPHA10 has emerged as a protein of interest in cancer research, particularly in studies of Triple-Negative Breast Cancer (TNBC). Recent research has focused on generating monoclonal antibodies with high specificity for EphA10 for potential therapeutic applications . The expression patterns and functions of EPHA10 in normal versus pathological states continue to be areas of active investigation, with the HRP-conjugated polyclonal antibody serving as an important tool in these studies.
Achieving reliable and reproducible results with the EPHA10 antibody requires attention to several methodological details that can influence experimental outcomes.
Proper sample preparation is critical for successful detection of EPHA10 using this antibody:
For Western blotting: Complete lysis of cells or tissues in appropriate buffers containing protease inhibitors helps preserve the integrity of the target protein.
For immunohistochemistry: Proper fixation and antigen retrieval methods are essential, as over-fixation may mask epitopes and prevent antibody binding.
For ELISA applications: Samples should be appropriately diluted in recommended buffers to ensure they fall within the assay's dynamic range .
Following recommended protocols for each application type ensures optimal antibody performance and reliable experimental outcomes.
When working with EPHA10 antibody, HRP conjugated, several quality control measures should be implemented:
Include appropriate positive and negative controls to validate antibody specificity.
Verify the predicted molecular weight of detected bands in Western blot applications (approximately 109 kDa).
Store the antibody in aliquots to minimize freeze-thaw cycles that could degrade the HRP enzyme and reduce activity.
Be aware of potential cross-reactivity with other EphA family members, though the antibody has been shown to be specific for EPHA10 in validation studies .
These quality control measures help ensure the reliability and reproducibility of experimental results obtained with this antibody.
The EPHA10 antibody, HRP conjugated, has potential applications beyond standard laboratory techniques, opening possibilities for innovative research directions.
Recent research has explored the development of monoclonal antibodies against EPHA10 for potential therapeutic applications, particularly in TNBC models . The polyclonal HRP-conjugated antibody can serve as a valuable tool in these development pipelines:
For screening EPHA10 expression in patient-derived samples to identify potential responders to targeted therapies.
In pharmacodynamic studies to monitor changes in EPHA10 expression following experimental treatments.
As a comparator for evaluating the binding characteristics of novel therapeutic antibodies being developed against the same target.
These applications highlight the translational potential of this research tool in bridging basic science and clinical investigations.
The HRP conjugation of this antibody makes it compatible with various modern analytical platforms:
High-throughput automated Western blotting systems for large-scale protein expression profiling.
Multiplex immunoassays where different antibodies targeting various proteins can be used simultaneously.
Quantitative ELISA systems for precise measurement of EPHA10 levels in research samples, with commercially available kits demonstrating sensitivity down to 0.109 ng/ml .
The versatility of this antibody in different analytical contexts expands its utility across diverse research applications and technological platforms.
EPHA10 (ephrin receptor A10) is a receptor tyrosine kinase that has gained significant attention as a cancer biomarker due to its unique expression pattern. It is virtually undetectable in most normal tissues except the male testis, yet shows high expression in several malignancies . This selective expression profile makes it an ideal target for cancer therapeutics with potentially minimal adverse effects on normal tissues.
Research has demonstrated that EPHA10 expression correlates with tumor progression and poor prognosis in several cancer types, including triple-negative breast cancer (TNBC) . It has been significantly linked to lymph node metastasis and higher tumor staging in human breast cancer specimens . Additionally, EPHA10 is expressed not only in cancer cells but also in immunosuppressive myeloid cells within the tumor microenvironment, including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) .
EPHA10 antibodies serve multiple critical research applications:
Detection and Localization: For identifying EPHA10 expression in tissue samples and cell lines using techniques such as immunohistochemistry (IHC), immunofluorescence (IF), and flow cytometry .
Protein Analysis: For quantifying and characterizing EPHA10 protein via Western blotting and ELISA .
Therapeutic Development: As potential direct therapeutic agents or as components of targeted delivery systems for cancer treatment .
CAR-T Cell Therapy: For developing chimeric antigen receptor T cells that specifically target EPHA10-expressing tumors .
HRP-conjugated EPHA10 antibodies particularly enhance detection sensitivity in applications such as Western blotting, ELISA, and IHC through enzymatic signal amplification.
EPHA10 exhibits a highly restricted expression pattern that makes it particularly valuable as a cancer target. In normal tissues, EPHA10 expression is essentially undetectable except in male testis tissue . This limited normal tissue distribution suggests that EPHA10-targeted therapies may have minimal off-target effects.
In contrast, EPHA10 shows significantly elevated expression in the tumor regions of multiple cancer types. Research using human tissue microarrays has demonstrated that EPHA10 expression levels are significantly higher in breast, lung, and ovarian cancer tumor regions compared to adjacent normal tissues . Within the tumor microenvironment, EPHA10 is expressed both by cancer cells and by immunosuppressive myeloid cells including TAMs and MDSCs, but not by tumor-infiltrating T cells as confirmed by immunofluorescence staining .
For optimal IHC results with HRP-conjugated EPHA10 antibodies, follow this methodological approach:
Tissue Preparation:
Fix tissues in 10% neutral buffered formalin for 24-48 hours
Process and embed in paraffin
Section at 4-5 μm thickness onto positively charged slides
Antigen Retrieval:
Perform heat-induced epitope retrieval using citrate buffer (pH 6.0) for 20 minutes
Allow slides to cool to room temperature for 20 minutes
Blocking and Primary Antibody Incubation:
Block endogenous peroxidase activity with 3% hydrogen peroxide for 10 minutes
Block non-specific binding with 5% normal serum for 30 minutes
Apply HRP-conjugated EPHA10 antibody at optimized dilution (typically 1:100 to 1:500) and incubate at 4°C overnight
Detection and Visualization:
Since the antibody is HRP-conjugated, no secondary antibody is required
Develop signal with DAB substrate for 5-10 minutes (optimize timing based on signal intensity)
Counterstain with hematoxylin, dehydrate, and mount
Controls:
Include EPHA10-positive tissues (breast cancer or testis) as positive controls
Include normal tissues (except testis) as negative controls
Consider including absorption controls to confirm specificity
For multiplexed staining, carefully consider that EPHA10 co-localizes with TAMs and MDSCs but not with T cells in the tumor microenvironment .
Validating antibody specificity is crucial for generating reliable research data. For HRP-conjugated EPHA10 antibodies, implement these complementary validation approaches:
Positive and Negative Tissue Controls:
Test on tissues known to express EPHA10 (breast, lung, or ovarian cancer samples; testis)
Confirm absence of staining in tissues known to lack EPHA10 expression (most normal tissues)
Competitive Binding Assay:
Pre-incubate the antibody with recombinant EPHA10 protein before application
Specific antibodies will show reduced or eliminated staining
Cellular Models:
Cross-Reactivity Assessment:
Multiple Detection Methods:
Correlate IHC results with other detection methods such as Western blot, RT-PCR, or flow cytometry
For rigorous validation, a flow cytometry-based assay comparing fluorescence intensity between cells expressing human EPHA10 and mock controls can effectively demonstrate antibody specificity, as shown with other EPHA10 antibodies .
For optimal Western blotting results with HRP-conjugated EPHA10 antibodies:
Sample Preparation:
Extract proteins using RIPA buffer supplemented with protease inhibitors
Determine protein concentration using BCA or Bradford assay
Load 20-50 μg of protein per lane
Gel Electrophoresis:
Use 8-10% SDS-PAGE (EPHA10 is ~120 kDa)
Include positive control lysates from EPHA10-expressing cancer cells
Transfer:
Transfer to PVDF membrane (recommended over nitrocellulose for EPHA10)
Use wet transfer at 100V for 90 minutes or 30V overnight at 4°C
Blocking and Antibody Incubation:
Block with 5% non-fat milk in TBST for 1 hour at room temperature
Dilute HRP-conjugated EPHA10 antibody in blocking buffer (typically 1:1000 to 1:5000)
Incubate overnight at 4°C with gentle agitation
Detection:
Since the antibody is HRP-conjugated, proceed directly to detection
Develop using enhanced chemiluminescence (ECL) substrate
Expose to X-ray film or use digital imaging system
Optimization Notes:
EPHA10 may require extended transfer times due to its high molecular weight
If background is high, increase washing time or add 0.1% Tween-20 to blocking buffer
For validation, compare results across multiple EPHA10-expressing and non-expressing samples
HRP-conjugated EPHA10 antibodies offer powerful tools for investigating the tumor microenvironment (TME) when used in these methodological contexts:
Multiplex Immunohistochemistry:
Combine with antibodies against immune cell markers to identify EPHA10 expression patterns
Sequential staining protocols using HRP-conjugated EPHA10 antibody followed by tyramide signal amplification and antibody stripping
This approach can reveal co-localization of EPHA10 with immunosuppressive myeloid cells such as TAMs (F4/80+/CD163+) and MDSCs (CD11b+/Gr-1+)
Tissue Microarray Analysis:
Apply to cancer tissue microarrays to assess correlation between EPHA10 expression and:
Infiltration of different immune cell populations
Expression of immune checkpoint molecules (e.g., PD-L1)
Patient outcomes and treatment responses
Dual Chromogenic IHC:
Use HRP-conjugated EPHA10 antibodies with other enzyme-conjugated antibodies (e.g., alkaline phosphatase)
Visualize with contrasting chromogens (DAB for EPHA10, Fast Red for other markers)
Quantify co-expression using digital image analysis
Research has shown that EPHA10 expression correlates with increased PD-L1 expression and immunosuppression , suggesting its role in immune evasion mechanisms. When analyzing the TME, focus particularly on the co-localization of EPHA10 with TAMs and MDSCs, which are known to suppress antitumor immunity through various mechanisms .
Developing effective EPHA10 antibody-based targeted delivery systems requires careful optimization of several parameters:
Antibody Conjugation Chemistry:
For liposomal delivery systems, conjugate anti-EPHA10 antibodies to liposomes using EDCI and sulfo-NHS chemistry in pH 7.4 PBS with 2-hour incubation at room temperature
For nanoparticle systems, consider maleimide-thiol coupling after antibody reduction
Purify conjugated systems using gel filtration on Sepharose 4B columns equilibrated in PBS
Target Binding and Specificity:
Payload Considerations:
For siRNA delivery, develop pH-sensitive liposomes modified with cholesterol-Schiff base-polyethylene glycol (Chol-SIB-PEG)
For toxic payloads, calculate optimal drug-to-antibody ratios to maintain binding while maximizing efficacy
Consider stimuli-responsive release mechanisms (pH, redox, enzymatic) based on tumor microenvironment
In Vivo Validation:
Perform antibody chasing assays with fluorescently labeled antibodies in xenograft models to confirm tumor-specific accumulation
Monitor pharmacokinetics and biodistribution to ensure minimal accumulation in non-target organs
Assess therapeutic efficacy in appropriate syngeneic models that maintain intact immune systems
Research has demonstrated that anti-EPHA10 antibodies can specifically accumulate in tumor regions according to EPHA10 levels without significant accumulation in other organs, making them excellent candidates for targeted delivery systems .
EPHA10 antibody targeting significantly impacts T cell-mediated antitumor immunity through multiple mechanisms:
Direct Effects on Immunosuppressive Myeloid Cells:
EPHA10 is expressed on immunosuppressive myeloid cells including TAMs and MDSCs, but not on T cells
Anti-EPHA10 antibody treatment likely modulates these immunosuppressive cells, potentially reducing their suppressive functions
This indirect mechanism helps restore T cell function in the tumor microenvironment
Enhancement of CTL-Mediated Responses:
Studies with anti-EPHA10 mAb clone #4 have demonstrated enhanced tumor regression in syngeneic TNBC mouse models
This was accompanied by improved CTL-mediated antitumor immunity without significant toxicity
The therapeutic effect appears to be dose-dependent, with higher doses (300 μg/mouse vs. 150 μg/mouse) showing greater efficacy
Relationship with Immune Checkpoint Pathways:
CAR-T Cell Approaches:
The efficacy of anti-EPHA10 therapy in improving antitumor immunity has been demonstrated across multiple syngeneic mouse models of TNBC, including 4T1 and EMT6, suggesting broad applicability across different TNBC subtypes .
When working with HRP-conjugated EPHA10 antibodies, researchers may encounter several technical challenges. Here are methodological solutions to common problems:
For particularly challenging samples, consider using amplification systems such as biotin-streptavidin or tyramide signal amplification to enhance detection sensitivity without increasing background.
Determining optimal antibody concentration requires systematic titration across different applications. Follow these methodological approaches:
For Western Blotting:
Prepare a titration series ranging from 1:500 to 1:5000 dilution
Use positive control lysates (EPHA10-expressing cancer cells) alongside negative controls
Select the dilution that provides clear specific bands with minimal background
For novel samples, consider starting with a broader range (1:100 to 1:10,000)
For Immunohistochemistry/Immunofluorescence:
Prepare a dilution series from 1:50 to 1:500
Apply to serial sections of known positive tissue (breast cancer or testis)
Evaluate for:
Signal-to-noise ratio
Specificity of staining pattern (membrane localization)
Correlation with expected expression patterns
The optimal dilution shows specific staining with minimal background
For ELISA:
Perform a checkerboard titration using:
Coating antigen at 0.1-10 μg/mL
Antibody dilutions from 1:100 to 1:10,000
Generate standard curves at each antibody concentration
Select the dilution providing the widest dynamic range with lowest EC50
For Flow Cytometry:
Each new lot of antibody should undergo re-titration, as conjugation efficiency may vary between manufacturing batches. Document optimal conditions for reproducibility across experiments.
Proper storage and handling of HRP-conjugated antibodies is critical for maintaining enzymatic activity and binding specificity. Follow these evidence-based practices:
Storage Conditions:
Store antibody at -20°C for long-term storage (up to 1 year)
For working stocks, store at 4°C for up to 1 month
Add 50% glycerol to stock solutions to prevent freeze-thaw damage
Include preservatives (0.02% sodium azide or 0.1% thimerosal) for stocks stored at 4°C
Note: Sodium azide inhibits HRP activity, so dilute sufficiently before use
Aliquoting Strategy:
Prepare single-use aliquots (10-50 μL) upon receipt
Use siliconized or low-protein binding tubes
Label with date, concentration, and freeze-thaw count
Never refreeze thawed aliquots
Thawing Protocol:
Thaw rapidly at room temperature with gentle agitation
Do not use heat sources as they may denature the antibody
Centrifuge briefly after thawing to collect solution
Use immediately after thawing for optimal results
Dilution Guidelines:
Use freshly prepared buffers for dilutions
Include carrier proteins (0.1-1% BSA) in dilution buffers
For most applications, prepare dilutions immediately before use
For multi-day experiments, prepare fresh dilutions daily
Stability Monitoring:
Include consistent positive controls in each experiment
Monitor signal intensity over time as indicator of stability
Consider including HRP activity controls (commercial standards)
If signal decreases >30% from baseline, use a new aliquot
Environmental Factors:
Protect from extended exposure to light
Avoid repeated temperature fluctuations
Maintain pH between 6.0-8.0 for optimal HRP stability
Avoid oxidizing agents and metal ions
Following these practices will help maintain antibody performance, ensure experimental reproducibility, and maximize the usable lifetime of valuable HRP-conjugated EPHA10 antibodies.
EPHA10 antibodies hold significant potential for integration into combination immunotherapy strategies through several mechanistic pathways:
Combination with Immune Checkpoint Inhibitors:
Enhancement of CAR-T Cell Approaches:
Develop dual-targeting strategies combining:
This approach could potentially overcome resistance mechanisms by addressing both tumor cells and their protective microenvironment simultaneously
Antibody-Drug Conjugates (ADCs) with Immunomodulatory Payloads:
Engineer anti-EPHA10 ADCs carrying payloads that can:
Directly kill EPHA10+ tumor cells
Release immunostimulatory molecules into the tumor microenvironment
Deplete immunosuppressive cell populations
These "immunomodulatory ADCs" could serve as bridges between targeted therapy and immunotherapy
Combination with Cancer Vaccines:
Anti-EPHA10 antibody treatment enhances CTL-mediated antitumor immunity
This effect could amplify responses to cancer vaccines by:
Reducing immunosuppression in the tumor microenvironment
Potentially releasing tumor antigens through antibody-dependent cellular cytotoxicity
Creating a more favorable environment for vaccine-induced T cells
Research in syngeneic TNBC mouse models has already demonstrated that anti-EPHA10 antibody treatment enhances tumor regression and improves therapeutic response rates , providing a strong rationale for combination approaches.
Elucidating the mechanisms of EPHA10-mediated immunosuppression requires sophisticated experimental approaches:
Single-Cell RNA Sequencing of the Tumor Microenvironment:
Isolate cells from EPHA10-high versus EPHA10-low tumors
Perform scRNA-seq to identify:
Transcriptional differences in immune cell populations
Altered signaling pathways in EPHA10-expressing cells
Changes in immunosuppressive molecule expression
Correlate findings with spatial information using spatial transcriptomics
Functional Assays of Myeloid-T Cell Interactions:
Co-culture with T cells to assess:
T cell proliferation (CFSE dilution assay)
Cytokine production (intracellular cytokine staining)
Cytotoxic activity against tumor targets
Compare effects of anti-EPHA10 antibody treatment on these interactions
Phospho-Proteomic Analysis:
Perform phospho-proteomics on EPHA10+ cells with and without antibody treatment
Map EPHA10 signaling networks and their connection to immunosuppressive pathways
Identify key nodes that could be targeted simultaneously with EPHA10
Genetic Manipulation Approaches:
Generate EPHA10 knockout or knockdown in specific cell populations using:
Conditional knockout mouse models
Cell type-specific CRISPR-Cas9 delivery
Compare with effects of antibody blockade to identify:
On-target versus off-target effects
Signaling-dependent versus independent mechanisms
In Vivo Imaging of Immune Cell Dynamics:
Utilize intravital microscopy to visualize:
T cell trafficking in EPHA10-high versus EPHA10-low tumors
Interactions between EPHA10+ myeloid cells and T cells
Effects of anti-EPHA10 antibody treatment on these dynamics
The finding that EPHA10 co-localizes with immunosuppressive myeloid cells (TAMs and MDSCs) but not with T cells in the tumor microenvironment provides a critical starting point for these mechanistic investigations.
Several innovative approaches could enhance the specificity and efficacy of EPHA10-targeted therapeutics:
Bispecific Antibody Platforms:
Develop bispecific antibodies targeting:
EPHA10 on tumor cells and CD3 on T cells to redirect T cell cytotoxicity
EPHA10 and immunosuppressive receptors (e.g., PD-L1) for dual checkpoint blockade
EPHA10 and myeloid cell markers to specifically target immunosuppressive populations
Optimize affinity and geometry for maximal efficacy with minimal adverse effects
Advanced Antibody Engineering:
Generate site-specific conjugation methods for ADCs to maintain binding properties
Develop antibody fragments (Fabs, scFvs) for improved tumor penetration
Engineer Fc modifications to enhance:
Antibody-dependent cellular cytotoxicity (ADCC)
Complement-dependent cytotoxicity (CDC)
Extended half-life through FcRn interactions
Responsive Nanoparticle Delivery Systems:
Create pH-sensitive liposomes conjugated with anti-EPHA10 antibodies that:
Remain stable in circulation
Release payloads specifically in the acidic tumor microenvironment
Can deliver combination therapeutics (e.g., siRNA plus small molecules)
Incorporate stimuli-responsive elements that respond to tumor-specific triggers
Improved CAR-T Cell Designs:
Develop next-generation EPHA10-specific CAR-T cells with:
Inducible expression systems for safety
Additional costimulatory domains for enhanced persistence
Resistance to exhaustion in the immunosuppressive TME
Logic-gated activation requiring multiple tumor antigens
Combination with Epigenetic Modifiers:
Investigate whether epigenetic drugs can increase EPHA10 expression in:
Cancer stem cells that may have lower expression
Tumor cells that have lost expression as an escape mechanism
This approach could expand the population of targetable cells
Research has already demonstrated that anti-EPHA10 mAbs with high specificity for EPHA10 (without cross-reactivity to other EphA family members) can effectively target tumor regions in vivo . Building on this foundation with these innovative approaches could significantly enhance therapeutic outcomes.