EPHA10 Antibody, FITC conjugated

Shipped with Ice Packs
In Stock

Description

2.1. Detection and Quantification

  • Flow Cytometry: Used to identify cell-surface EPHA10 expression in cancer cell lines (e.g., BT-549 TNBC cells) . The FITC conjugate exhibits strong fluorescence intensity, enabling precise quantification .

  • Immunofluorescence: Localizes EPHA10 in tumor microenvironments, including co-localization with immunosuppressive myeloid cells like tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) .

  • Western Blot: Validates EPHA10 expression in lysates from human, mouse, and rat tissues, with a predicted band size of 109 kDa .

2.2. Functional Studies

  • Therapeutic Development: While this antibody is not directly therapeutic, it aids in characterizing EphA10-targeting monoclonal antibodies (mAbs) and chimeric antigen receptor (CAR)-T cells . For example, clone #4 of anti-EPHA10 mAbs suppressed tumor growth in triple-negative breast cancer (TNBC) models .

  • Mechanistic Insights: EPHA10 knockdown reduces tumorigenic behaviors such as invasion and gelatin degradation in pancreatic cancer cells, highlighting its role in metastasis .

4.1. Cancer Biomarker Potential

EPHA10 is overexpressed in aggressive cancers, including TNBC and pancreatic ductal adenocarcinoma . Its co-localization with immunosuppressive cells in tumors suggests a role in immune evasion . The FITC-conjugated antibody facilitates studies exploring EPHA10 as a diagnostic or prognostic marker.

4.2. Therapeutic Linkages

  • Monoclonal Antibodies: Anti-EPHA10 mAbs (e.g., clone #4) inhibit tumor growth in syngeneic mouse models by enhancing T cell–mediated immunity .

  • Bispecific Antibodies: A bispecific antibody targeting EPHA10 and CD3 redirects cytotoxic T cells to tumors, demonstrating preclinical efficacy .

Comparative Analysis

FeatureEPHA10 FITC Antibody Therapeutic mAbs
TargetEPHA10 detectionEPHA10 inhibition
ConjugateFITCNone (naked antibody or CAR-T)
Primary UseResearchPreclinical therapy
Key FindingsLocalizes EPHA10 in TAMs Reduces tumor volume by 40–60%

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. Delivery times may vary depending on the purchasing method and location. Please consult your local distributor for specific delivery times.
Synonyms
Ephrin type-A receptor 10 (EC 2.7.10.1), EPHA10
Target Names
EPHA10
Uniprot No.

Target Background

Function
EPHA10 is a receptor for members of the ephrin-A family. It binds to EFNA3, EFNA4, and EFNA5.
Gene References Into Functions
  1. We identified a novel interaction between EphA10s and EphA10 that promotes malignant transformation in breast cancer. Our findings suggest potential clinical benefits. PMID: 28427223
  2. Our research demonstrated the physical association and cellular co-localization of EPHA7 and EPHA10 in breast carcinoma cells. The nuclear co-localization of these two receptors in invasive MDA-MB-231 cells suggests their involvement in transcriptional activation of genes involved in invasiveness. PMID: 27566654
  3. Ephrin receptor A10 is a promising drug target potentially useful for treating various breast cancers, including triple-negative breast cancers. PMID: 24946238
  4. The overexpression of EphA10 in prostate cancers suggests its potential as a target for prostate cancer therapy, which motivated the studies reported here. PMID: 24924629
  5. The expression of EPHA10 at both the gene and protein level in clinical breast cancer tissues is significantly associated with lymph node metastasis and stage progression. PMID: 24403271
  6. EPHA10 does not interact with EPHB6 in breast neoplasms. PMID: 21737611
  7. CLL B-cells exhibit a more heterogeneous Eph/EFN profile, particularly EFNA4, EphB6, and EphA10. PMID: 18819711

Show More

Hide All

Database Links

HGNC: 19987

OMIM: 611123

KEGG: hsa:284656

STRING: 9606.ENSP00000362139

UniGene: Hs.129435

Protein Families
Protein kinase superfamily, Tyr protein kinase family, Ephrin receptor subfamily
Subcellular Location
[Isoform 1]: Cell membrane; Single-pass type I membrane protein.; [Isoform 3]: Cell membrane; Single-pass type I membrane protein.; [Isoform 2]: Secreted.
Tissue Specificity
Mainly expressed in testis.

Q&A

What is EPHA10 and why is it considered a promising target for cancer immunotherapy?

EPHA10 is a receptor tyrosine kinase belonging to the Eph receptor family that plays critical roles in cell signaling and regulation. It has gained significant attention as a cancer therapeutic target for several reasons:

  • EPHA10 is highly expressed in several cancer types including triple-negative breast cancer (TNBC), pancreatic cancer, and oral squamous cell carcinoma (OSCC), while showing limited expression in normal tissues (primarily restricted to testis) .

  • This restricted expression pattern makes EPHA10 an ideal target for cancer-specific therapies with potentially minimal adverse effects on healthy tissues .

  • EPHA10 expression has been correlated with tumor progression, lymph node metastasis, and higher tumor stage in breast cancer .

  • Studies have detected EPHA10 not only in cancer cells but also in immunosuppressive myeloid cells in the tumor microenvironment, suggesting multiple mechanisms by which targeting EPHA10 could affect tumor progression .

When designing experiments targeting EPHA10, researchers should consider its potential roles in both tumor cells directly and in modulating the immune microenvironment.

How can I validate the specificity of FITC-conjugated EPHA10 antibodies in my experimental system?

Ensuring specificity of EPHA10 antibodies is crucial for reliable research outcomes. Based on published validation methods, consider implementing the following multi-step approach:

  • Cross-reactivity testing: Evaluate binding to other EphA family members (EphA1-EphA8) using ELISA assays with recombinant proteins. Highly specific antibodies should recognize only EPHA10, not other isoforms that share similar architecture .

  • Cell-based validation:

    • Compare staining between EPHA10-expressing cells and mock controls using flow cytometry and immunofluorescence microscopy .

    • Include positive controls (cells with confirmed EPHA10 expression like BT-549 breast cancer cells) and negative controls (cells with no or low EPHA10 expression) .

  • Competitive binding assays: Pre-incubate your FITC-conjugated antibody with recombinant EPHA10 protein before cell staining to demonstrate signal reduction through competitive binding.

  • Knockdown validation: Compare staining intensity between wild-type cells and those with EPHA10 knockdown (using siRNA or CRISPR-Cas9 systems) to confirm signal specificity .

  • Western blot correlation: Correlate fluorescence intensity with protein expression levels determined by Western blot to ensure concordance between detection methods .

This comprehensive validation strategy will help ensure that your FITC-conjugated EPHA10 antibody provides specific and reliable detection for downstream applications.

What are optimal protocols for immunofluorescence staining using FITC-conjugated EPHA10 antibodies?

For successful immunofluorescence staining with FITC-conjugated EPHA10 antibodies, consider the following optimized protocol based on published literature:

  • Sample preparation:

    • For cultured cells: Grow cells on coverslips, fix with 4% paraformaldehyde for 15 minutes at room temperature, and permeabilize with 0.1% Triton X-100 if detecting intracellular domains.

    • For tissue sections: Use freshly frozen or paraffin-embedded sections (4-6 μm thickness). For paraffin sections, perform antigen retrieval using citrate buffer (pH 6.0).

  • Blocking: Block non-specific binding with 5% normal serum in PBS containing 0.1% Tween-20 for 1 hour at room temperature.

  • Primary antibody incubation: Apply FITC-conjugated EPHA10 antibody at an optimized dilution (typically 1:100-1:500 for polyclonal antibodies) . Incubate overnight at 4°C in a humidified chamber protected from light to prevent photobleaching.

  • Washing: Wash 3 times with PBS containing 0.1% Tween-20 for 5 minutes each.

  • Nuclear counterstaining: Counterstain nuclei with DAPI (1 μg/mL) for 5 minutes at room temperature.

  • Mounting: Mount with anti-fade mounting medium to minimize photobleaching.

  • Imaging considerations:

    • Use appropriate filter sets for FITC (excitation ~495 nm, emission ~520 nm).

    • Capture images promptly as FITC is prone to photobleaching.

    • Include appropriate controls (secondary antibody-only, isotype control) to distinguish specific from non-specific staining.

  • Co-localization studies: For analyzing EPHA10 expression in specific cell populations within the tumor microenvironment, consider multi-color immunofluorescence with markers for TAMs (F4/80, CD163) or MDSCs (CD11b, Gr-1) as described in the literature .

For optimal results, validate dilution factors for each new antibody lot and cell/tissue type being studied.

How does EPHA10 expression vary across cancer types and what are the implications for antibody-based targeting strategies?

EPHA10 shows distinct expression patterns across various cancer types, with important implications for targeting strategies:

  • Breast cancer:

    • Particularly high expression in triple-negative breast cancer (TNBC) .

    • Expression correlates with lymph node metastasis and higher tumor stage .

    • Detected in both HER2-positive and HER2-negative breast cancer tissues .

  • Pancreatic cancer:

    • Significantly expressed in pancreatic cancer cell lines including MIA PaCa-2 and AsPC-1 .

    • Overexpression increases proliferation, migration, adhesion, and invasion capabilities .

    • Enhances MMP-9 expression and secretion, contributing to increased invasiveness .

  • Oral squamous cell carcinoma (OSCC):

    • Supports tumor growth and lymphatic metastasis .

    • Regulates epithelial-mesenchymal transition (EMT) and sphere formation .

    • Drives expression of EMT- and stemness-associated transcription factors .

  • Expression in the tumor microenvironment:

    • Present in immunosuppressive myeloid cells including tumor-associated macrophages (TAMs) and myeloid-derived suppressor cells (MDSCs) .

    • Potentially contributes to immunosuppression, as higher EPHA10 expression has been associated with increased PD-L1 expression .

These expression patterns suggest several targeting strategies:

  • For breast cancer, particularly TNBC, direct antibody targeting has shown promise in preclinical models .

  • For pancreatic cancer, targeting EPHA10 might inhibit multiple tumorigenic processes including proliferation and invasion .

  • The presence of EPHA10 in immune cells suggests potential for combination therapies targeting both tumor cells and immunosuppressive components of the microenvironment .

When designing experiments with FITC-conjugated EPHA10 antibodies, consider the specific cancer context and whether you aim to target cancer cells directly, immune cells, or both populations.

What are the best methods for quantifying EPHA10 expression levels using flow cytometry with FITC-conjugated antibodies?

For accurate quantification of EPHA10 expression using flow cytometry with FITC-conjugated antibodies, implement these methodological approaches:

  • Sample preparation optimization:

    • For cell lines: Use single-cell suspensions in flow buffer (PBS with 2% FBS and 2mM EDTA) at 1×10^6 cells/100μL.

    • For tumor samples: Generate single-cell suspensions using gentle enzymatic digestion (collagenase IV, DNase I) to preserve surface antigens.

    • Use viability dyes (e.g., 7-AAD or propidium iodide) to exclude dead cells that can give false-positive signals.

  • Antibody titration:

    • Perform titration experiments (typically 0.1-10 μg/mL) to determine the optimal antibody concentration that provides the highest signal-to-noise ratio.

    • Include isotype controls conjugated to FITC at the same concentration to assess non-specific binding.

  • Compensation setup:

    • For multicolor panels, perform proper compensation using single-color controls to account for spectral overlap, particularly important when FITC is used alongside PE or other fluorophores with overlapping emission spectra.

  • Quantitative analysis approaches:

    • Calculate Mean/Median Fluorescence Intensity (MFI) ratios between sample and isotype control.

    • Use Molecules of Equivalent Soluble Fluorochrome (MESF) beads to standardize measurements across experiments.

    • Consider Quantitative Flow Cytometry (QFCM) using beads with known antibody binding capacity (ABC) to determine the actual number of EPHA10 molecules per cell.

  • Gating strategies:

    • For tumor microenvironment analysis, use a hierarchical gating strategy as described in published studies: first identify viable cells, then CD45+ leukocytes, followed by specific immune cell populations (e.g., CD3-, F4/80+, CD206+ for TAMs or CD11b+, Gr1+ for MDSCs) .

    • For co-expression studies, consider bivariate plots showing EPHA10-FITC versus other markers of interest.

  • Controls and validation:

    • Use cell lines with known EPHA10 expression levels (e.g., BT-549) as positive controls .

    • Include EPHA10 knockdown cells as biological negative controls .

    • Consider parallel analysis with other detection methods (Western blot, qPCR) to validate findings.

This comprehensive approach will ensure reliable quantification of EPHA10 expression across different experimental contexts and sample types.

How can I assess the functionality of anti-EPHA10 antibodies in activating immune responses against tumor cells?

Evaluating the immune-activating potential of anti-EPHA10 antibodies requires multi-dimensional functional assays. Based on published methodologies, consider these approaches:

  • T cell-mediated cytotoxicity assays:

    • Co-culture system: Establish co-cultures of EPHA10-expressing tumor cells with isolated T cells at various effector-to-target (E:T) ratios (2.5:1 to 20:1) .

    • Cytotoxicity measurement: Assess tumor cell killing using lactate dehydrogenase (LDH) release, calcein-AM release, or real-time impedance-based assays.

    • Controls: Include isotype control antibodies and EPHA10-negative cell lines to confirm specificity.

  • Immune cell activation analysis:

    • Measure T cell activation markers (CD25, CD69) and effector molecules (granzyme B, perforin, IFN-γ) by flow cytometry or ELISA after co-culture with EPHA10-expressing cells and antibodies .

    • Assess proliferation of T cells using CFSE dilution or Ki-67 staining when exposed to antibody-coated tumor cells.

  • Bispecific antibody testing:

    • If working with bispecific antibodies targeting both EPHA10 and CD3, compare monomeric versus dimeric formats at varying concentrations (10^-3 to 10^1 μg/mL) .

    • Determine the minimum E:T ratio required for effective tumor cell lysis (published data shows dimeric bispecific antibodies can be effective at E:T ratios as low as 2.5:1) .

  • In vivo evaluation approaches:

    • Syngeneic models: Use immunocompetent mice with EPHA10-expressing tumors to assess antibody efficacy in the context of an intact immune system .

    • Mechanistic studies: Compare antibody efficacy between immunocompetent and immunodeficient (e.g., BALB/c-SCID) mice to confirm immune-mediated effects .

    • Tumor infiltration analysis: Evaluate changes in tumor-infiltrating lymphocyte populations, particularly activated CD8+/GrB+ CTLs, using flow cytometry or multiplex immunofluorescence .

  • Combination therapy assessment:

    • Test anti-EPHA10 antibodies in combination with immune checkpoint inhibitors (anti-PD-1, anti-CTLA-4) to evaluate potential synergistic effects.

    • Consider combination with standard-of-care therapies for specific cancer types to assess complementary mechanisms of action.

These methodological approaches provide a comprehensive framework for evaluating the immune-activating potential of anti-EPHA10 antibodies across in vitro and in vivo systems.

What are the key considerations for in vivo imaging studies using fluorescently labeled anti-EPHA10 antibodies?

When conducting in vivo imaging with fluorescently labeled anti-EPHA10 antibodies, several methodological considerations can optimize experimental outcomes:

  • Fluorophore selection and conjugation:

    • While the query specifically mentions FITC conjugation, consider that FITC has limitations for in vivo imaging due to its shorter wavelength (520 nm emission) and susceptibility to photobleaching.

    • Published studies have successfully used longer-wavelength fluorophores like Alexa-647 for in vivo imaging of anti-EPHA10 antibodies .

    • If using FITC, ensure conjugation chemistry preserves antibody binding capacity and consider higher antibody doses to compensate for lower tissue penetration.

  • Animal model selection:

    • Xenograft models using human EPHA10-expressing cell lines (e.g., MDA-MB-231, MDA-MB-435) in immunodeficient mice have been validated for antibody distribution studies .

    • For functional studies combining imaging with therapeutic assessment, consider syngeneic models in immunocompetent mice to preserve immune interactions .

  • Antibody administration and imaging protocol:

    • Administer labeled antibodies intravenously (typically 100-150 μg per mouse) .

    • For FITC-conjugated antibodies, use imaging systems with appropriate excitation (488 nm) and emission filters.

    • Perform longitudinal imaging to track antibody distribution and persistence (published studies show detectable signal for up to 6 days with Alexa-647 conjugated antibodies) .

  • Biodistribution assessment:

    • Quantify fluorescence intensity as the ratio between tumor and contralateral normal tissue .

    • Include multiple timepoints (24h, 48h, 72h, 6d) to determine optimal imaging window .

    • Harvest and image major organs (liver, kidney, spleen, lung, heart, testis) ex vivo to assess potential off-target accumulation .

  • Validation and controls:

    • Include isotype-matched control antibodies conjugated with the same fluorophore to distinguish specific from non-specific accumulation .

    • Perform blocking studies with unlabeled anti-EPHA10 antibodies to confirm binding specificity.

    • Correlate imaging findings with ex vivo immunohistochemistry to validate target engagement.

  • Data analysis considerations:

    • Apply appropriate background subtraction and normalization techniques to account for tissue autofluorescence.

    • Consider photon attenuation at different tissue depths when quantifying signals.

    • Report tumor-to-background ratios rather than absolute fluorescence intensity to enable cross-experimental comparisons.

These methodological considerations will help researchers design robust in vivo imaging studies to assess the tumor-targeting capabilities of fluorescently labeled anti-EPHA10 antibodies.

How does EPHA10 signaling differ from other EphA family receptors, and what implications does this have for antibody development?

EPHA10 possesses unique characteristics among the EphA family that directly impact antibody development strategies:

  • Catalytic deficiency and signaling mechanism:

    • EPHA10 is classified as a catalytically defective receptor protein tyrosine kinase, unlike most other EphA family members that possess active kinase domains .

    • Despite this catalytic deficiency, EPHA10 still influences signaling cascades, potentially through:

      • Acting as a scaffold protein for signaling complexes

      • Modulating the activity of binding partners

      • Activating downstream pathways through unconventional mechanisms

  • Downstream signaling pathways:

    • EPHA10 overexpression increases phosphorylation of ERK, JNK, AKT, FAK, and NF-κB pathways, which are critical for cell proliferation, survival, adhesion, migration, and invasion .

    • In oral squamous cell carcinoma, EPHA10 interacts with ephrin A4 (EFNA4) to promote cell migration and sphere formation through regulation of SNAIL, NANOG, and OCT4 expression .

  • Expression pattern differences:

    • Unlike other EphA receptors that show broader tissue distribution, EPHA10 expression in normal tissues is largely restricted to testis .

    • This restricted expression pattern in normal tissues coupled with upregulation in multiple cancer types makes EPHA10 uniquely suitable as an antibody target .

  • Implications for antibody development:

    • Epitope selection: Since EPHA10 is catalytically defective, therapeutic antibodies should target:

      • The ligand-binding domain to interfere with interactions with ephrin ligands

      • Regions involved in protein-protein interactions that may mediate scaffold functions

      • Epitopes that trigger receptor internalization rather than focusing solely on kinase inhibition

    • Cross-reactivity considerations: The EphA family shares significant structural homology, making specificity a critical concern. Published studies demonstrate the importance of validating antibodies against all EphA family members (EphA1-EphA8) to ensure EPHA10 specificity .

    • Functional screening approaches: When developing anti-EPHA10 antibodies, screening should assess:

      • Ability to interfere with downstream signaling pathways (ERK, JNK, AKT)

      • Effects on cellular phenotypes (migration, invasion, sphere formation)

      • Capacity to induce immune-mediated killing rather than focusing only on direct inhibition of tumor cell proliferation

    • Format considerations: The catalytically defective nature of EPHA10 suggests therapeutic antibodies may be particularly effective when developed as:

      • Antibody-drug conjugates that utilize EPHA10's expression pattern rather than relying on inhibiting its kinase activity

      • Bispecific antibodies that redirect immune cells to EPHA10-expressing cells

      • CAR-T cell therapies targeting EPHA10 epitopes

Understanding these unique aspects of EPHA10 enables rational design of antibodies that exploit its distinctive biology for more effective therapeutic approaches.

What are the advantages and limitations of FITC-conjugated antibodies compared to other fluorophores for EPHA10 detection in experimental systems?

When selecting fluorophores for EPHA10 detection, researchers should consider these comparative advantages and limitations of FITC conjugation:

Advantages of FITC conjugation:

  • Established compatibility: FITC has well-established conjugation chemistry with antibodies, making it widely available and standardized.

  • Instrumentation accessibility: Almost all flow cytometers, fluorescence microscopes, and imaging systems have appropriate filter sets for FITC detection (excitation ~495 nm, emission ~520 nm).

  • Multicolor panel integration: FITC works well in multicolor panels with minimal compensation requirements when paired with fluorophores like PE-Cy5, APC, and Pacific Blue.

  • Cost-effectiveness: FITC-conjugated antibodies are generally less expensive than antibodies conjugated with newer-generation fluorophores.

  • Signal amplification compatibility: FITC-conjugated antibodies work well with tyramide signal amplification (TSA) systems for detecting low-abundance targets like EPHA10 in certain tissues.

Limitations of FITC conjugation:

  • Photobleaching: FITC is particularly susceptible to photobleaching compared to newer fluorophores, limiting its utility in extended imaging sessions or when multiple scans are required .

  • pH sensitivity: FITC fluorescence decreases significantly at acidic pH, which can be problematic when studying endocytosis of EPHA10 antibodies or trafficking to acidic compartments.

  • Tissue autofluorescence: Background autofluorescence from tissues is often highest in the green spectrum where FITC emits, potentially obscuring specific signals, particularly in fixed tissues.

  • In vivo imaging limitations: FITC's relatively short wavelength limits tissue penetration for in vivo imaging, whereas longer-wavelength fluorophores like Alexa-647 have been successfully used for in vivo tracking of anti-EPHA10 antibodies .

  • Quantum yield changes upon conjugation: FITC can lose quantum yield when conjugated to proteins, sometimes resulting in lower brightness compared to theoretical expectations.

Alternative fluorophores for EPHA10 detection:

  • Alexa Fluor dyes: Alexa-488 offers similar spectral properties to FITC but with improved photostability and brightness. Alexa-647 has been successfully used for in vivo imaging of anti-EPHA10 antibodies with excellent results .

  • Phycoerythrin (PE): Offers much higher brightness than FITC, beneficial for detecting low-level EPHA10 expression, though its large size may affect antibody trafficking.

  • Quantum dots: Provide exceptional brightness and photostability for long-term imaging of EPHA10 localization, though their size may impact binding kinetics.

  • Near-infrared fluorophores: For in vivo applications, fluorophores like IRDye800 provide superior tissue penetration and lower autofluorescence compared to FITC.

The optimal choice depends on your specific experimental requirements, with FITC being well-suited for routine flow cytometry and standard microscopy applications, while other fluorophores may offer advantages for specialized applications like in vivo imaging or detection of low EPHA10 expression levels.

How can I design experiments to evaluate the therapeutic potential of anti-EPHA10 antibodies in combination with immune checkpoint inhibitors?

Designing robust experiments to assess combination therapy with anti-EPHA10 antibodies and immune checkpoint inhibitors requires careful methodological planning:

  • Preclinical model selection:

    • Syngeneic models: Use immunocompetent mouse models (BALB/c mice with 4T1 or EMT6 TNBC cells) that express EPHA10 and allow for intact immune system interactions .

    • Humanized models: Consider NSG mice engrafted with human immune cells and human EPHA10-expressing tumors for evaluating fully human antibodies.

    • Rationale: Published studies demonstrate that anti-EPHA10 antibody efficacy requires an intact immune system, as evidenced by lack of effect in BALB/c-SCID mice .

  • Treatment arms design:

    • Monotherapy groups: Anti-EPHA10 antibody alone (150-300 μg/mouse, twice weekly), checkpoint inhibitor alone (anti-PD-1, anti-PD-L1, or anti-CTLA-4 at standard doses) .

    • Combination groups: Anti-EPHA10 plus checkpoint inhibitor at the same doses used in monotherapy.

    • Controls: Isotype-matched control antibodies for both treatments.

    • Dosing schedule: Test sequential versus concurrent administration to determine optimal timing.

  • Primary endpoints assessment:

    • Tumor growth inhibition: Measure tumor volume over time using calipers or bioluminescence imaging.

    • Survival analysis: Compare median survival and generate Kaplan-Meier curves.

    • Response rate categorization: Classify responses as progressive disease, stable disease, partial response, or complete response based on established criteria .

    • Metastasis evaluation: Assess lymph node and distant metastasis when relevant.

  • Mechanistic assessments:

    • Immunophenotyping analysis:

      • Quantify tumor-infiltrating lymphocytes (TILs), particularly focusing on CD8+/Granzyme B+ CTLs, which have been shown to increase with anti-EPHA10 therapy .

      • Analyze immunosuppressive cell populations (TAMs, MDSCs) that express EPHA10 for changes in frequency and phenotype .

      • Measure PD-1/PD-L1 expression levels before and after anti-EPHA10 treatment.

    • Functional immune assays:

      • Ex vivo cytotoxicity assays using TILs isolated from treated tumors.

      • Cytokine profiling (IFN-γ, TNF-α, IL-2) to assess T cell functionality.

      • Multiplexed immunohistochemistry to evaluate spatial relationships between immune cells and tumor cells.

    • Signaling pathway analysis:

      • Assess changes in phosphorylation status of ERK, JNK, AKT, FAK, and NF-κB pathways, which are modulated by EPHA10 .

      • Evaluate expression of EMT and stemness markers that might be affected by EPHA10 targeting .

  • Optimizing combination strategies:

    • Dose-finding studies: Test multiple dose levels of anti-EPHA10 antibodies (e.g., 50, 150, 300 μg/mouse) in combination with fixed doses of checkpoint inhibitors to identify optimal combinations .

    • Schedule optimization: Compare concurrent administration versus sequential treatment (anti-EPHA10 followed by checkpoint inhibitor or vice versa).

    • Mechanism-based combinations: Consider additional combination partners based on EPHA10 biology:

      • Anti-angiogenic agents (EPHA10 affects MMP-9 production) .

      • Targeted therapies against pathways activated by EPHA10 (ERK/JNK inhibitors) .

  • Translational biomarker assessment:

    • Patient selection markers: Identify threshold levels of EPHA10 expression that predict response.

    • Pharmacodynamic markers: Develop assays to confirm target engagement and downstream pathway modulation.

    • Resistance mechanisms: Monitor for adaptive resistance through evaluation of alternative immune checkpoints or EPHA10 downregulation.

This comprehensive experimental approach will generate robust data on the potential synergy between anti-EPHA10 antibodies and immune checkpoint inhibitors, providing a foundation for clinical translation.

How can I develop and validate EphA10-specific CAR-T cells using anti-EphA10 antibodies?

Developing effective EphA10-specific CAR-T cells requires careful consideration of antibody selection, CAR design, and validation methods:

  • Anti-EphA10 antibody selection for CAR construction:

    • Specificity assessment: Begin with antibodies validated for exclusive binding to EphA10 without cross-reactivity to other EphA family members .

    • Epitope mapping: Select antibodies targeting extracellular domains that maintain accessibility when EphA10 is expressed on cell surfaces .

    • Affinity optimization: Test antibodies with different binding affinities (nanomolar to picomolar range) as very high-affinity antibodies may impair CAR-T cell detachment and serial killing.

    • Published example: Anti-EphA10 mAb clone #4 has been successfully used to derive effective CAR-T cells with significant inhibitory effects on TNBC cells in vitro and in vivo .

  • CAR construct design:

    • Single-chain variable fragment (scFv) generation: Clone and sequence the variable regions of the heavy and light chains from the selected anti-EphA10 antibody.

    • Extracellular domain engineering: Connect VH and VL with a flexible linker (e.g., (G4S)3) to maintain proper folding and antigen recognition.

    • Hinge and transmembrane selection: Test different options (CD8α, IgG4, CD28) as these components affect CAR expression and function.

    • Costimulatory domains: Compare second-generation CARs containing CD28 or 4-1BB costimulatory domains to optimize persistence and cytotoxicity.

    • Vector selection: Use lentiviral or retroviral vectors for stable CAR expression and clinical translatability.

  • In vitro validation methodology:

    • CAR expression verification: Assess surface expression using protein L or anti-Fab antibodies by flow cytometry.

    • Target-specific activation: Measure upregulation of CD25, CD69, and cytokine production (IFN-γ, TNF-α, IL-2) upon co-culture with EphA10+ tumor cells.

    • Cytotoxicity assays: Perform dose-response killing assays with multiple E:T ratios against:

      • EphA10-expressing cancer cell lines (TNBC: MDA-MB-231, 4T1; Pancreatic: MIA PaCa-2, AsPC-1)

      • EphA10-negative control cells to confirm specificity

      • Cell lines with CRISPR-mediated EphA10 knockout as additional controls

    • Repeated challenge assays: Assess CAR-T cell persistence and serial killing capacity through sequential tumor cell challenges.

  • In vivo validation approaches:

    • Animal models:

      • For human CAR-T cells: Use immunodeficient mouse models (NSG) with established human EphA10+ tumors.

      • For murine CAR-T cells: Use syngeneic models with murine EphA10+ tumors in immunocompetent mice .

    • Efficacy endpoints: Measure tumor growth inhibition, survival benefit, and complete response rates.

    • Persistence and trafficking: Track CAR-T cell expansion, persistence, and tumor infiltration using flow cytometry and immunohistochemistry.

    • Safety evaluation: Monitor for on-target/off-tumor toxicity, particularly in testis where low-level EphA10 expression occurs in normal tissues .

  • Potential enhancement strategies:

    • Armored CARs: Engineer CAR-T cells to secrete cytokines (IL-12, IL-18) to enhance function in immunosuppressive tumor microenvironments.

    • Combination approaches: Test EphA10 CAR-T cells in combination with checkpoint inhibitors or after antibody pre-treatment to enhance efficacy.

    • Logic-gated CARs: Develop dual-receptor systems requiring both EphA10 and a second tumor-associated antigen for activation to enhance specificity.

By following this comprehensive development and validation pathway, researchers can create effective EphA10-directed CAR-T cell therapies with potential applications across multiple cancer types expressing this target.

What methodological approaches can effectively distinguish between EphA10 expression on tumor cells versus immune cells in the tumor microenvironment?

Accurate differentiation of EphA10 expression between tumor cells and immune cells requires integrated multi-platform approaches:

  • Multiplex immunofluorescence methodology:

    • Panel design: Combine FITC-conjugated anti-EphA10 antibodies with lineage-specific markers:

      • Tumor cells: Cytokeratin, EpCAM

      • T cells: CD3

      • Macrophages: F4/80, CD163

      • MDSCs: CD11b, Gr-1

    • Sequential staining protocol: Implement tyramide signal amplification (TSA) for sequential antibody staining and stripping to minimize cross-reactivity.

    • Multispectral imaging: Use platforms like Vectra Polaris or Mantra to spectrally unmix fluorophores and accurately quantify co-localization.

    • Spatial analysis: Apply computational spatial analysis to quantify cell-type specific expression and proximity relationships.

  • Flow cytometry-based approaches:

    • Dissociation optimization: Use gentle enzymatic dissociation protocols (collagenase IV + DNase I) that preserve surface epitopes while achieving single-cell suspensions.

    • Viability discrimination: Include fixable viability dyes to exclude dead cells that may bind antibodies non-specifically.

    • Gating strategy: Implement hierarchical gating (as published in literature):

      • First gate viable cells → identify cell populations (tumor vs. immune) → within immune cells, separate subpopulations (TAMs, MDSCs) → assess EphA10 expression within each population .

    • Controls: Include fluorescence-minus-one (FMO) controls for accurate boundary setting with FITC-conjugated antibodies.

  • Single-cell RNA sequencing integration:

    • Perform scRNA-seq on tumor samples to identify cell-type specific EphA10 mRNA expression patterns.

    • Validate at protein level using CITE-seq approaches with oligo-conjugated anti-EphA10 antibodies.

    • Correlate EphA10 expression with cell-type specific transcriptional signatures.

  • Laser capture microdissection approaches:

    • Identify regions enriched for specific cell types using rapid immunohistochemistry.

    • Microdissect distinct areas (tumor nests vs. immune-rich stroma).

    • Analyze EphA10 expression by qPCR or proteomics in the isolated populations.

  • In situ hybridization combined with immunofluorescence:

    • Perform RNAscope for EphA10 mRNA detection with high sensitivity.

    • Combine with immunofluorescence for cell-type markers.

    • Quantify co-localization of EphA10 mRNA with specific cell populations.

  • Validation and quantification strategies:

    • Quantitative image analysis: Use digital pathology software (HALO, QuPath) with machine learning algorithms to classify cells and quantify EphA10 expression intensity in different populations.

    • Expression level comparison: Develop standardized methods to compare EphA10 expression levels between cell types using mean fluorescence intensity ratios or molecules of equivalent soluble fluorochrome (MESF).

    • Spatial context preservation: Map EphA10+ immune cells relative to tumor boundaries to assess potential functional implications.

Published studies have demonstrated successful implementation of these approaches, revealing that EphA10 is expressed not only in tumor regions but also co-localizes with TAMs and MDSCs (appearing as merged signals in multiplex imaging), while not co-localizing with T cells . These methodological approaches provide a comprehensive framework for distinguishing cell-type specific EphA10 expression patterns within the complex tumor microenvironment.

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.