EPHA2 Antibody

EPH receptor A2, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction

EPHA2 antibodies represent a targeted therapeutic approach for cancers overexpressing the EPHA2 receptor tyrosine kinase. These antibodies exploit EPHA2's role in oncogenesis, particularly its ligand-independent kinase activity and association with aggressive tumor phenotypes. By inducing receptor degradation, blocking oncogenic signaling, or activating immune-mediated cytotoxicity, EPHA2 antibodies have shown promise in preclinical and clinical studies .

Receptor Degradation via Autophosphorylation

Agonistic EPHA2 antibodies (e.g., EA1.2, IC1) bind to the extracellular domain of EPHA2, triggering autophosphorylation and subsequent internalization/degradation. This mechanism inhibits metastatic behaviors like soft agar colonization and tubular network formation in breast cancer models (e.g., MDA-MB-231) . Biochemical studies confirm that active antibodies (e.g., EA1.2) induce dose-dependent tyrosine phosphorylation, while neutral antibodies (e.g., EN2) do not .

Antibody-Dependent Cellular Cytotoxicity (ADCC)

Afucosylated antibodies (e.g., DS-8895a) enhance ADCC by recruiting natural killer (NK) cells or monocytes to lyse EPHA2-expressing tumor cells. DS-8895a demonstrated >80% tumor cell killing in vitro and reduced tumor volume in xenograft models (e.g., MDA-MB-231, SNU-16 gastric cancer) .

AntibodyMechanismKey Preclinical Efficacy
EA1.2Autophosphorylation → DegradationInhibits soft agar growth (MDA-MB-231)
DS-8895aADCC, Ephrin-A1 antagonismReduces tumor volume (SNU-16, MDA-MB-231)
1C1 (MEDI-547)ADC (mcMMAF conjugate)IC₅₀: 3 ng/mL in breast cancer models

In Vitro and In Vivo Efficacy

  • EA1.2: Blocked tubular network formation in Matrigel assays (metastatic melanoma cells) .

  • DS-8895a: Synergized with cisplatin in gastric cancer models, enhancing tumor response .

  • 1C1-mcMMAF: Induced caspase-3/7 activation and tumor regression at 1 mg/kg in xenografts .

  • BT5528: Peptide-cytotoxin conjugate with low off-target effects, showing tumor volume reduction in EPHA2-positive models .

Completed Trials

Trial (NCT)PhaseCompoundDiseaseOutcome
NCT007960551MEDI-547Solid tumorsTerminated (severe coagulopathy)
NCT020047171DS-8895aGastric/EsophagealStable disease/partial responses
NCT015913561/2DasatinibBreast cancer (bone)Completed, limited efficacy

Ongoing Trials

Trial (NCT)PhaseCompoundDiseaseStatus
NCT034239921BT5528Solid tumorsRecruiting
NCT038785241bDasatinibAdvanced cancersRecruiting

Off-Target Effects

MEDI-547 (1C1-mcMMAF) caused severe coagulopathy in clinical trials, attributed to endothelial cell toxicity . This highlights the need for antibodies with high specificity to avoid vascular damage.

Contextual Oncogenic Role

EPHA2 exhibits dual roles: promoting metastasis in some contexts and suppressing proliferation in others. Antisense strategies must balance receptor downregulation with oncogenic compensation .

Precision Medicine

Trials now prioritize patient stratification based on EPHA2 expression levels (e.g., NCT03878524) . Biomarkers for EPHA2 activation status could optimize therapeutic outcomes.

Next-Generation Antibodies

  • Afucosylated variants: DS-8895a enhances ADCC without agonistic effects, reducing off-target risks .

  • Peptide-cytotoxin conjugates: BT5528 avoids hematologic toxicity, showing promise in phase 1/2 trials .

  • Single-chain antibodies: D2 scFv’s structural mimicry of ephrin-A1 offers potential for tumor-specific delivery .

Product Specs

Introduction
EPHA2, a member of the ephrin receptor subfamily within the protein-tyrosine kinase family, plays a crucial role in developmental processes, particularly in the nervous system. EPH and EPH-related receptors are known for their involvement in these events. EPH subfamily receptors typically possess a single kinase domain and an extracellular region characterized by a cysteine-rich domain and two fibronectin type III repeats. The ephrin receptors are classified into two groups based on sequence similarities in their extracellular domains and their binding affinities for ephrin-A and ephrin-B ligands. Specifically, the EPHA2 gene encodes a protein that exhibits binding affinity for ephrin-A ligands. Mutations in the EPHA2 gene have been identified as the underlying cause of certain genetically inherited cataract disorders.
Formulation
The antibody is supplied as a 1 mg/ml solution in a buffer consisting of PBS at pH 7.4 with 0.1% sodium azide.
Storage Procedures
For storage durations of up to one month, maintain the product at 4°C. For extended storage periods, store at -20°C. To preserve product integrity, it is crucial to avoid repeated freeze-thaw cycles.
Stability / Shelf Life
The product has a shelf life of 12 months when stored at -20°C and remains stable for 1 month at 4°C.
Applications
The EPHA2 antibody has undergone rigorous testing using ELISA, Western blot, and immunohistochemistry analyses to ensure both specificity and reactivity. However, optimal working dilutions should be determined empirically for each application. For Western blotting, a recommended dilution range of 1:500 to 1:2,000 is suggested, with a starting dilution of 1:500. For immunohistochemistry analysis, a dilution range of 1:100 to 1:200 is recommended, with a starting dilution of 1:100.
Synonyms
EPH receptor A2, Ephrin type-A receptor 2, Tyrosine-protein kinase receptor ECK, Epithelial cell kinase, EPHA2, ECK, ARCC2.
Purification Method
EPHA2 antibody was purified from mouse ascitic fluids by protein-G affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone
P3D7AT.
Immunogen
Anti-human EPHA2 mAb, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with recombinant human EPHA2 amino acids 1-154 purified from E. coli.
Ig Subclass
Mouse IgG2b heavy chain and κ light chain.

Q&A

What is the significance of EphA2 receptor in cancer biology?

EphA2 represents a paradoxical target in cancer research due to its unique signaling mechanism. Unlike typical receptor tyrosine kinases, EphA2 demonstrates ligand-independent activity in cancer cells while maintaining kinase activity. The receptor is significantly overexpressed in multiple aggressive carcinomas compared to normal tissues, with the highest levels consistently found in the most aggressive tumor cells . This differential expression pattern provides an opportunity for selective targeting of malignant cells while minimizing effects on normal tissues. Importantly, when EphA2 binds its ligand (ephrinA1) or ligand-mimicking antibodies, it undergoes phosphorylation and subsequent degradation, leading to negative regulation of tumor cell growth and migration . This dual nature makes EphA2 an attractive therapeutic target, as strategies that restore normal receptor function can potentially reverse malignant phenotypes.

How do EphA2 antibodies differ mechanistically from other cancer-targeting antibodies?

Unlike many therapeutic antibodies that function by blocking receptor-ligand interactions, EphA2 antibodies can work through a distinctive mechanism that mimics the natural ligand ephrinA1. Specifically, biologically active antibodies (such as EA1.2 mentioned in research) induce EphA2 phosphorylation and subsequent degradation . This mechanism results in selective inhibition of malignant behaviors, particularly anchorage-independent growth, without affecting normal cell function . This selectivity is demonstrated by experiments showing that EphA2 antibodies inhibit soft agar colonization by breast tumor cells without affecting monolayer growth of nontransformed mammary epithelial cells . The ability to distinguish between normal and malignant cell behavior provides a therapeutic window not available with many other targeted antibodies, which often affect normal tissues expressing the target protein.

What experimental methods are most reliable for detecting EphA2 expression in tissue samples?

For robust detection of EphA2 in tissue samples, multiple complementary approaches should be employed:

  • Fluorescence-based ELISA (FluorELISA): This method selects for antibody reactivity against live cells, preserving conformational epitopes that might be lost in fixed or denatured samples. Research shows this technique effectively distinguished between EphA2-overexpressing tumor cells (MDA-MB-231) and EphA2-deficient cells (BT474) .

  • Immunoprecipitation followed by Western blot: This two-step process confirms specificity by immunoprecipitating with the test antibody and blotting with a validated anti-EphA2 antibody. Studies demonstrate this approach identifies EphA2-specific antibodies while eliminating cross-reactive candidates .

  • Immunofluorescence microscopy: This provides spatial information about EphA2 distribution, revealing the characteristic pattern of diffuse membrane staining in EphA2-positive cells .

  • Flow cytometry: This enables quantification of cell surface EphA2 on viable cells within heterogeneous samples, allowing researchers to analyze expression in specific cell populations.

When designing experimental protocols, it's critical to recognize that some EphA2 epitopes are conformation-dependent and may be lost under reducing conditions during SDS-PAGE, necessitating nonreducing conditions for detection with certain antibodies .

How should researchers design functional assays to evaluate EphA2 antibody efficacy?

Designing functional assays for EphA2 antibodies requires careful consideration of the biology underlying EphA2's role in cancer. Based on research findings, the following approaches are recommended:

  • Three-dimensional growth assays: Soft agar colonization assays are particularly valuable for evaluating EphA2 antibody efficacy, as they specifically assess anchorage-independent growth—a hallmark of malignant transformation. Research demonstrates that the oncogenic potential of EphA2 is more apparent in three-dimensional assays than in monolayer cultures . These assays should include appropriate controls such as isotype-matched non-targeting antibodies.

  • Tubular network formation assays: EphA2 antibodies can prevent tumor cells from forming tubular networks on reconstituted basement membranes (Matrigel), providing a sensitive indicator of metastatic character . This assay evaluates the ability of antibodies to inhibit behaviors specific to metastatic cells.

  • Comparative growth studies: Compare effects on malignant versus normal cells using identical conditions. Research shows that effective EphA2 antibodies inhibit tumor cell growth without affecting nontransformed epithelial cells, confirming selective targeting .

  • Time-course experiments: Assess antibody effects at multiple time points, as research indicates that inhibition of monolayer growth may only become apparent after cells reach confluence .

  • Phosphorylation and degradation analysis: Measure EphA2 phosphorylation and subsequent degradation following antibody treatment to confirm the mechanism of action. In published studies, biologically active antibodies significantly increased the phosphotyrosine content of EphA2 in a dose-dependent manner .

These assays collectively provide a comprehensive evaluation of antibody efficacy while distinguishing between effects on normal versus malignant cell behaviors.

What controls are essential when evaluating EphA2 antibody specificity?

Rigorous control strategies are critical for validating EphA2 antibody specificity:

  • Cell line controls: Utilize both EphA2-overexpressing cell lines (e.g., MDA-MB-231) and EphA2-deficient cells (e.g., BT474) as positive and negative controls respectively . This comparison establishes baseline specificity for EphA2 recognition.

  • Isotype-matched controls: Include appropriate isotype-matched non-targeting antibodies to distinguish EphA2-specific effects from non-specific binding or Fc receptor-mediated effects.

  • Reciprocal immunoprecipitation validation: Confirm antibody specificity through reciprocal experiments—immunoprecipitate with the test antibody and blot with a validated EphA2 antibody, then perform the reverse process . This double-confirmation approach strengthens confidence in antibody specificity.

  • Reduced versus non-reduced conditions: Test antibody recognition under both reducing and non-reduced conditions, as research shows some antibodies (like EA1.2) may only recognize epitopes in their native conformation .

  • Cross-reactivity assessment: Evaluate potential cross-reactivity with other Eph family members, particularly closely related ones like EphA1, using cells that express these related receptors but not EphA2.

  • Epitope mapping: When possible, characterize the specific epitope recognized by the antibody, which provides insight into its functional properties and potential cross-reactivity.

  • Functional validation: Confirm that antibodies predicted to be agonistic can indeed induce EphA2 phosphorylation, internalization, and degradation as demonstrated in published research .

Implementing these controls ensures that observed effects are truly attributable to EphA2-specific interactions rather than off-target or non-specific mechanisms.

How can researchers optimize antibody-induced EphA2 phosphorylation and degradation?

Optimizing antibody-induced EphA2 phosphorylation and degradation requires strategic experimental design based on known molecular mechanisms:

  • Dose optimization: Titrate antibody concentrations to identify the optimal dose for inducing EphA2 phosphorylation. Research demonstrates that effective antibodies like EA1.2 increase EphA2 phosphotyrosine content in a dose-dependent manner .

  • Time course analysis: Conduct time course experiments to determine the kinetics of EphA2 phosphorylation and subsequent degradation. Initial phosphorylation can be detected within minutes (8-10 minutes in published studies), while degradation occurs over a longer timeframe .

  • Phosphotyrosine detection methodology: Use phosphotyrosine-specific antibodies (such as 4G10) in combination with EphA2 immunoprecipitation to specifically detect phosphorylated receptor . This approach allows for quantitative assessment of phosphorylation levels.

  • Inhibitor studies: Utilize lysosomal and proteasomal inhibitors to characterize the degradation pathway and confirm the mechanism through which antibody binding leads to EphA2 downregulation.

  • Receptor trafficking analysis: Employ fluorescently labeled antibodies and co-localization studies with endosomal and lysosomal markers to track EphA2 internalization and degradation in real-time.

  • Comparison with natural ligand: Include ephrinA1-Fc as a positive control, as it represents the natural ligand that induces EphA2 phosphorylation and degradation. This comparison helps determine whether antibodies fully recapitulate the natural ligand's effects.

  • Validation in multiple cell lines: Test optimization protocols across different cell lines with varying levels of EphA2 expression to ensure the robustness of the approach.

These methodological approaches enable researchers to characterize and enhance the two key mechanisms—phosphorylation and degradation—through which EphA2 antibodies exert their anti-tumor effects.

How should researchers address the apparent paradox between EphA2 overexpression and tumor inhibition?

The EphA2 paradox—where a seemingly oncogenic receptor can inhibit tumor growth when properly stimulated—requires careful experimental design and interpretation:

  • Distinguish between ligand-independent and ligand-dependent signaling: EphA2 in tumor cells often exhibits ligand-independent signaling that promotes tumor progression, while ligand (or antibody) binding activates pathways that inhibit malignant behavior . Researchers should explicitly characterize these distinct signaling states through phosphoproteomic analysis.

  • Analyze receptor localization and clustering: Examine whether EphA2 is properly localized to cell-cell contacts in normal versus tumor cells. Research indicates that in malignant cells, EphA2 fails to bind ephrinA1 on adjacent cells despite overexpression . This mislocalization contributes to the paradoxical function.

  • Characterize downstream signaling pathways: Identify the distinct signaling cascades activated in unliganded versus ligand-bound states. This can be accomplished through phosphoproteomic approaches combined with pathway inhibitor studies.

  • Context-dependent effects: Evaluate the impact of cellular context on EphA2 function by comparing effects in 2D monolayer versus 3D cultures. Research demonstrates that the oncogenic potential of EphA2 is most apparent in three-dimensional assays, while differences may not be detectable in monolayer cultures .

  • Temporal dynamics: Consider the timing of observations, as EphA2 signaling exhibits complex temporal dynamics. Short-term versus long-term effects may differ significantly, as evidenced by research showing inhibition of growth only after cells reached confluence .

Understanding this paradox is essential for developing effective therapeutic approaches, as it explains why antibodies that activate rather than block EphA2 can effectively inhibit tumor growth.

What considerations are important when analyzing EphA2 antibody effects across different tumor types?

  • Baseline EphA2 expression levels: Quantify and normalize for varying baseline EphA2 expression levels across tumor types. Research indicates that multiple epithelial tumors overexpress EphA2, but levels may vary significantly .

  • Receptor activation state: Assess the baseline phosphorylation status of EphA2 in different tumor types, as this may influence antibody efficacy. Some tumors may have partially activated receptors while others have completely ligand-independent signaling.

  • Cell context-specific signaling partners: Identify and account for differences in EphA2 signaling partners across tumor types. The presence or absence of certain adapter proteins or downstream effectors may dramatically alter antibody effects.

  • Growth pattern differences: Recognize that different tumor types may exhibit distinct growth patterns in response to EphA2 targeting. For example, research shows that inhibition of monolayer growth in MDA-MB-231 cells only became apparent after reaching confluence, a pattern also observed in PC-3 prostate cancer cells .

  • Stratified analysis: Stratify analysis based on molecular subtypes within each cancer type to identify patterns of response that correlate with particular molecular features beyond simply EphA2 expression levels.

  • Differential endpoint selection: Select endpoints relevant to each tumor type's biology. While soft agar colonization may be appropriate for some cancers, others might require invasion assays or angiogenesis measurements to capture EphA2's role in that particular context.

  • Time-dependent effects: Include multiple time points in analyses, as research shows that the timing of effects may vary across tumor types, with some showing early responses and others demonstrating efficacy only at later time points .

These considerations enable more nuanced interpretation of antibody efficacy data and may reveal tumor type-specific mechanisms that could inform clinical translation strategies.

How can antibody-induced EphA2 degradation be distinguished from other mechanisms of protein downregulation?

Distinguishing antibody-induced EphA2 degradation from other downregulation mechanisms requires specific experimental approaches:

  • Direct comparison with ligand-induced degradation: Compare antibody effects with ephrinA1-Fc (the natural ligand) treatment to determine whether the degradation pattern mimics physiological ligand-induced downregulation .

  • Phosphorylation requirement: Determine whether phosphorylation precedes degradation by using phosphorylation-specific antibodies in time course experiments. Research demonstrates that biologically active antibodies like EA1.2 first increase EphA2 phosphotyrosine content before degradation occurs .

  • Inhibitor studies: Employ specific inhibitors of different degradation pathways:

    • Lysosomal inhibitors (e.g., chloroquine, bafilomycin A)

    • Proteasomal inhibitors (e.g., MG132, bortezomib)

    • Endocytosis inhibitors (e.g., dynasore, pitstop)
      These studies can identify which cellular machinery is involved in antibody-induced EphA2 degradation.

  • Mutation analysis: Utilize EphA2 mutants with altered phosphorylation sites or internalization motifs to determine the structural requirements for antibody-induced degradation.

  • Subcellular fractionation: Track the movement of EphA2 between membrane and intracellular compartments following antibody treatment using biochemical fractionation techniques.

  • Pulse-chase experiments: Employ metabolic labeling to distinguish between effects on EphA2 synthesis versus degradation rates.

  • Co-localization studies: Use immunofluorescence microscopy to visualize EphA2 trafficking to early endosomes, late endosomes, and lysosomes following antibody treatment.

These approaches collectively provide strong evidence for antibody-specific degradation mechanisms, distinguishing them from transcriptional downregulation, microRNA-mediated suppression, or other protein stability regulators.

How can EphA2 antibodies be utilized to study bidirectional signaling in the tumor microenvironment?

EphA2 antibodies provide powerful tools for investigating the complex bidirectional signaling between tumor cells and their microenvironment:

  • Forward versus reverse signaling discrimination: Utilize domain-specific antibodies that selectively activate forward signaling (through EphA2) without triggering reverse signaling (through ephrinA ligands). This approach can dissect the relative contributions of each signaling direction to tumor progression.

  • Cell type-specific antibody treatments: Apply EphA2 antibodies to co-culture systems containing tumor cells and stromal components to observe effects on each cell population. Research indicates that EphA2 on tumor cells interacts with ephrinA1 anchored to the membrane of adjacent cells, suggesting important intercellular communication .

  • Spatial mapping of receptor-ligand interactions: Employ fluorescently labeled antibodies in tissue sections to visualize the spatial distribution of EphA2-ephrinA interactions at tumor-stroma boundaries. This mapping can identify regions of active bidirectional signaling.

  • Phosphoproteomic profiling: Combine cell sorting with phosphoproteomic analysis following antibody treatment to characterize signaling cascades activated in both EphA2-expressing tumor cells and ephrinA-expressing stromal cells.

  • Genetic manipulation strategies: Use EphA2 antibodies in conjunction with genetic approaches that selectively modify forward or reverse signaling capabilities to create experimental systems with controlled bidirectional signaling properties.

  • Extracellular vesicle analysis: Investigate whether EphA2 antibody treatment alters the release or content of tumor-derived extracellular vesicles, which may carry EphA2 receptors and influence stromal cell behavior.

  • In vivo imaging: Develop antibody-based imaging approaches to visualize EphA2-ephrinA interactions in living organisms, providing insights into the dynamic nature of bidirectional signaling during tumor progression.

Understanding this bidirectional communication is critical since research demonstrates that the defect in ligand binding in cancer cells contributes to their malignant phenotype, and restoring proper EphA2-ephrinA interactions can reverse metastatic behavior .

What are the latest developments in antibody-drug conjugates targeting EphA2?

Antibody-drug conjugates (ADCs) targeting EphA2 represent an evolving therapeutic strategy with several important developments:

  • Optimization of antibody selection: Research has focused on identifying antibodies that not only bind EphA2 but also induce efficient internalization—a critical property for ADC efficacy. Studies like those with MEDI-547, which combines a fully human monoclonal antibody against both human and murine EphA2 (1C1) with the tubulin polymerization inhibitor monomethylauristatin F (MMAF), demonstrate this approach .

  • Payload diversification: Beyond conventional cytotoxic agents like MMAF, researchers are exploring alternative payloads including DNA-damaging agents, RNA polymerase inhibitors, and immunomodulatory molecules to overcome resistance mechanisms.

  • Linker technology advancement: Development of novel linker chemistries that optimize stability in circulation while ensuring efficient payload release in the tumor environment. This includes pH-sensitive linkers that respond to the acidic endosomal environment following EphA2 internalization.

  • Dual-targeting strategies: Creation of bispecific antibodies that simultaneously target EphA2 and another tumor-associated antigen to improve specificity and overcome potential resistance through antigen loss.

  • Patient selection biomarkers: Development of companion diagnostics that predict response to EphA2-targeted ADCs based not just on expression levels but also on internalization efficiency and recycling rates.

  • Combination approaches: Investigation of synergistic combinations between EphA2-ADCs and other therapeutic modalities, including checkpoint inhibitors, based on the understanding that EphA2 signaling may influence the tumor immune microenvironment.

  • Target validation in diverse tumor types: Expansion of therapeutic applications beyond the initial focus on endometrial cancer to other malignancies with EphA2 overexpression, supported by evidence that EphA2 levels predict poor outcomes across multiple cancer types .

These developments leverage the finding that EphA2 is frequently overexpressed in cancer cells compared to normal tissues, providing a therapeutic window for selective targeting with potent cytotoxic payloads.

How can antisense and RNA interference approaches complement antibody strategies against EphA2?

Antisense and RNA interference (RNAi) technologies offer complementary approaches to antibody strategies for targeting EphA2, each with unique advantages:

  • Validation of antibody mechanisms: Antisense-based targeting of EphA2 provides crucial validation of antibody effects by demonstrating that direct reduction of EphA2 protein levels inhibits malignant phenotypes. Research shows that antisense-based EphA2 targeting inhibits soft agar colonization, similar to the effects observed with EphA2 antibodies, suggesting that antibodies exert their effects by downregulating EphA2 .

  • Access to intracellular domains: While antibodies primarily engage the extracellular domain of EphA2, antisense and RNAi approaches can effectively target the intracellular tyrosine kinase domain and other regions inaccessible to antibodies.

  • Combination therapy potential: Combining antibodies with antisense or RNAi technologies can simultaneously target different aspects of EphA2 biology—antibodies inducing receptor degradation while nucleic acid-based approaches prevent new synthesis.

  • Kinetics of action: Antisense and RNAi typically demonstrate slower onset but potentially more complete target suppression compared to antibodies. This difference in kinetics can be exploited in experimental designs requiring different temporal patterns of EphA2 inhibition.

  • Overcoming resistance mechanisms: Tumor cells that develop resistance to antibody-induced EphA2 degradation (through altered trafficking or processing) may remain sensitive to direct suppression of EphA2 expression via antisense or RNAi.

  • Delivery challenges and solutions: While delivery of nucleic acid therapeutics presents challenges, advances in nanoparticle formulations, conjugated delivery systems, and even antibody-directed delivery of siRNA offer promising solutions.

  • Conditional and inducible systems: Genetic approaches enable the development of conditional and inducible EphA2 knockdown systems that can provide temporal control over EphA2 expression, facilitating studies of EphA2's role at different stages of tumor progression.

These complementary approaches provide researchers with a diverse toolkit for investigating EphA2 biology and developing therapeutic strategies, each offering distinct advantages depending on the specific research or clinical question.

What factors can affect EphA2 antibody binding in experimental settings?

Multiple factors can significantly influence EphA2 antibody binding, requiring careful experimental design and interpretation:

  • Epitope accessibility: The conformation of EphA2 can dramatically affect epitope accessibility. Research demonstrates that some antibodies (including EA1.2) only recognize EphA2 epitopes under non-reducing conditions, indicating conformation-dependent recognition . Researchers should test both reducing and non-reducing conditions when evaluating antibody binding.

  • Post-translational modifications: Glycosylation and other modifications on the extracellular domain of EphA2 may mask epitopes or create new ones. Different tumor types or cell lines may exhibit varied modification patterns that affect antibody binding.

  • Receptor clustering: The clustering state of EphA2 on the cell surface can influence antibody avidity and epitope accessibility. Pre-treatment conditions that alter membrane fluidity or receptor organization may affect binding efficiency.

  • Ligand occupancy: Pre-existing bound ligand (ephrinA1) may compete with antibodies for binding sites or induce conformational changes that affect antibody recognition. Assessing the native ligand occupancy state is important for interpreting binding data.

  • Fixation artifacts: For immunohistochemistry or immunofluorescence applications, fixation methods can significantly impact epitope preservation. Research utilized fluorescence-based ELISA protocols specifically to select antibodies that recognize conformational epitopes on viable tumor cells rather than denatured proteins .

  • Cold competition: Unintended competition between different antibodies can occur when performing sequential or simultaneous staining. This is particularly relevant when using multiple antibodies to different EphA2 epitopes.

  • Receptor internalization dynamics: The rate of constitutive EphA2 internalization and recycling varies across cell types. Antibody binding studies should account for these dynamics, particularly during longer incubation periods.

  • Expression level variability: Extreme differences in EphA2 expression levels between samples can affect the apparent binding affinity due to avidity effects or receptor saturation. Normalization approaches should be considered when comparing binding across samples with widely varying expression.

Understanding and controlling for these factors is essential for generating reproducible and interpretable EphA2 antibody binding data.

How can researchers differentiate between cytostatic and cytotoxic effects of EphA2 antibodies?

Differentiating between cytostatic (growth-inhibitory) and cytotoxic (cell-killing) effects of EphA2 antibodies requires multi-parameter assessment:

  • Growth curve analysis: Perform detailed growth curve analysis with multiple time points rather than single endpoint measurements. Research demonstrates that EphA2 antibodies like EA1.2 inhibit growth of MDA-MB-231 cells only after reaching confluence, suggesting cytostatic rather than immediate cytotoxic effects .

  • Comparing growth in different contexts: Assess effects in both anchorage-dependent (monolayer) and anchorage-independent (soft agar) conditions. Studies show that EphA2 antibody effects were more pronounced in soft agar assays than in monolayer cultures, indicating context-dependent action .

  • Cell cycle analysis: Utilize flow cytometry to determine whether antibody treatment induces cell cycle arrest (cytostatic) or sub-G1 accumulation (indicative of cell death).

  • Apoptosis markers: Employ multiple complementary approaches to detect apoptosis:

    • Annexin V/PI staining for early/late apoptosis discrimination

    • Caspase activation assays for apoptotic pathway engagement

    • TUNEL assays for DNA fragmentation

    • Assessment of mitochondrial membrane potential

  • Live-cell imaging: Conduct time-lapse microscopy to directly observe cellular responses to antibody treatment, distinguishing between growth arrest and cell death morphology.

  • Reversibility testing: Remove antibody after treatment period and monitor recovery. Cytostatic effects are typically reversible upon drug removal, while cytotoxic effects are not.

  • Cellular metabolism assessment: Measure effects on cellular metabolism using assays like MTT or ATP production, which can distinguish between viable but non-proliferating cells (cytostatic effect) versus dead cells (cytotoxic effect).

  • Combinatorial approach: Compare EphA2 antibody effects with known cytostatic (e.g., CDK inhibitors) and cytotoxic (e.g., doxorubicin) agents to establish reference response patterns.

This multi-parameter approach provides a comprehensive understanding of how EphA2 antibodies affect tumor cell viability and growth, informing both mechanistic insights and therapeutic applications.

What are the key considerations for transitioning from in vitro to in vivo studies with EphA2 antibodies?

Transitioning from in vitro to in vivo studies with EphA2 antibodies requires careful planning and consideration of multiple factors:

  • Antibody species cross-reactivity: Ensure that the antibody recognizes both human and murine EphA2 if using mouse models with human xenografts, as interactions with host tissue EphA2 may be biologically relevant. Research specifically notes that MEDI-547 was composed of an antibody against both human and murine EphA2, highlighting this important consideration .

  • Pharmacokinetic optimization: Determine antibody half-life in circulation and tissue distribution patterns to establish appropriate dosing schedules. Consider antibody format (full IgG vs. F(ab')2 vs. Fab) and species to optimize circulating half-life.

  • Model selection relevance: Choose appropriate models that recapitulate the EphA2 biology observed in human tumors. Research demonstrates the use of orthotopic models for endometrial cancer to evaluate EphA2-targeted therapies, providing physiologically relevant tumor microenvironments .

  • Dosing optimization: Establish dose-response relationships, as antibody effects may be concentration-dependent. Research shows that antibodies like EA1.2 increase EphA2 phosphotyrosine content in a dose-dependent manner in vitro, suggesting that dosing will be critical in vivo as well .

  • Time course considerations: Design experiments with appropriate time points for assessment, as effects may vary temporally. In vitro studies demonstrate that growth inhibition by EphA2 antibodies may only become apparent at later time points (e.g., after cells reach confluence) .

  • Relevant endpoints: Select endpoints that capture EphA2-specific biology beyond simple tumor volume measurements. Consider assessing metastasis, angiogenesis, and tumor microenvironment changes given EphA2's roles in these processes.

  • Biomarker development: Develop and validate pharmacodynamic biomarkers that confirm antibody engagement with EphA2 in vivo, such as measurements of receptor phosphorylation, degradation, or downstream signaling changes in tumor biopsies.

  • Combination strategies: Consider potential combination approaches based on in vitro findings. If EphA2 antibodies primarily demonstrate cytostatic effects in vitro, combinations with cytotoxic agents may be warranted for in vivo studies.

  • Imaging approaches: Incorporate non-invasive imaging techniques to monitor treatment effects longitudinally, potentially including labeled antibodies to track tumor targeting and receptor dynamics.

Addressing these considerations helps ensure that in vivo studies are optimally designed to evaluate the therapeutic potential of EphA2 antibodies and generate translatable findings.

Product Science Overview

Introduction

The EPH receptor A2 (EphA2) is a member of the Eph receptor family, which is the largest subgroup of receptor tyrosine kinases (RTKs). Eph receptors and their ligands, ephrins, play crucial roles in various biological processes, including cell growth, migration, and tissue development. EphA2, in particular, has been extensively studied for its involvement in cancer progression and metastasis.

Structure and Function

EphA2 is a transmembrane protein that consists of an extracellular domain, a single transmembrane segment, and an intracellular tyrosine kinase domain. The extracellular domain is responsible for binding to ephrin ligands, which are membrane-bound proteins. Upon ligand binding, EphA2 undergoes dimerization and autophosphorylation, leading to the activation of downstream signaling pathways.

EphA2 primarily interacts with ephrin-A family members, including ephrin-A1 to ephrin-A5. These interactions regulate various cellular processes such as cell adhesion, shape, and mobility. EphA2 is also involved in angiogenesis, the formation of new blood vessels, which is a critical process in tumor growth and metastasis.

Role in Cancer

EphA2 is overexpressed in various types of cancers, including breast, lung, prostate, and colorectal cancers. Its overexpression is often associated with poor prognosis and increased tumor aggressiveness. EphA2 promotes cancer cell proliferation, migration, and invasion by activating several signaling pathways, including the PI3K/AKT and MAPK pathways.

In addition to its role in cancer cell behavior, EphA2 also contributes to the tumor microenvironment by promoting angiogenesis and vasculogenic mimicry. Vasculogenic mimicry refers to the ability of aggressive cancer cells to form vessel-like structures, providing an alternative blood supply to the tumor.

Therapeutic Target

Given its significant role in cancer progression, EphA2 has emerged as a potential therapeutic target. Several strategies have been developed to inhibit EphA2 signaling, including monoclonal antibodies, small molecule inhibitors, and ligand-based approaches. These therapies aim to block EphA2 activation, thereby inhibiting tumor growth and metastasis.

Mouse Anti-Human EphA2 Antibody

Mouse anti-human EphA2 antibodies are monoclonal antibodies developed to specifically recognize and bind to human EphA2. These antibodies are commonly used in research to study EphA2 expression and function in various experimental settings. They are also utilized in diagnostic applications to detect EphA2 levels in tissue samples.

One such antibody is the Mouse Anti-Human EphA2 Monoclonal Antibody (Clone 371805), which has been shown to detect EphA2 in direct ELISAs and Western blots without cross-reactivity with other Eph receptors . This antibody is used in various applications, including flow cytometry and immunocytochemistry, to study EphA2 expression in cancer cells .

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 2024 Thebiotek. All Rights Reserved.