EPCAM Antibody

Epithelial Cell Adhesion Molecule, Mouse Anti Human
Shipped with Ice Packs
In Stock

Description

Introduction to EpCAM Antibodies

EpCAM (Epithelial Cell Adhesion Molecule), originally identified as a tumor antigen in colorectal cancer, is a transmembrane glycoprotein overexpressed in >95% of epithelial cancers, including colorectal, breast, pancreatic, and ovarian carcinomas . EpCAM antibodies are engineered to target this protein, leveraging its role in tumor cell adhesion, proliferation, and metastasis to deliver cytotoxic effects or block oncogenic signaling. These antibodies have evolved from murine-derived monoclonals to fully humanized variants with enhanced specificity and reduced immunogenicity .

Development and Types of EpCAM Antibodies

Key Antibodies in Development/Use:

AntibodyTypeTarget EpitopeMechanismClinical Status
EdrecolomabMurine IgG2aEpCAM surfaceADCC/CDCPhase III (ineffective)
AdecatumumabHumanized IgG1EpCAM surfaceADCC, blocks EpICD signalingPhase II (breast cancer)
CatumaxomabTrifunctionalEpCAM/CD3T-cell recruitment, ADCCApproved (malignant ascites)
MOC31PEImmunotoxinEpCAMPseudomonas exotoxin deliveryPhase I
m801 (E3)Fully human IgG1EpCAM extracellularHigh-affinity binding (1.09 nM) Preclinical
EpAb2-6MonoclonalEGF-II/TY domainInhibits EpICD production Preclinical

Key Advances:

  • Fully human antibodies (e.g., m801) exhibit high affinity (1–10 nM binding) and reduced immunogenicity .

  • Bispecific antibodies (e.g., Catumaxomab) redirect T-cell cytotoxicity to EpCAM+ tumor cells .

  • Immunotoxins (e.g., MOC31PE) conjugate cytotoxic drugs to EpCAM-specific antibodies for targeted delivery .

Mechanisms of Action

EpCAM antibodies employ multiple strategies to inhibit tumor growth:

MechanismExample AntibodyKey Findings
ADCC/CDCEdrecolomabActivates NK cells/macrophages; limited efficacy in Phase III .
EpICD InhibitionEpAb2-6Blocks EpICD nuclear translocation, reducing WNT/Ras signaling .
T-cell RecruitmentCatumaxomabBinds CD3+ T-cells to EpCAM+ tumor cells, enhancing immune synapse formation .
Toxin DeliveryMOC31PEDelivers Pseudomonas exotoxin to EpCAM+ cells, inducing apoptosis .

Clinical Applications and Challenges

Approved Therapies:

  • Catumaxomab: Approved for malignant ascites in ovarian cancer; shows partial response in intrapleural administration .

Ongoing/Failed Trials:

  • Edrecolomab: Failed Phase III trials due to low affinity and toxicity .

  • Adecatumumab: Efficacy observed in high EpCAM-expressing breast cancer patients .

  • MOC31PE: Phase I trials demonstrated dose-dependent toxicity (e.g., hypoalbuminemia) .

Synergistic Combinations:

  • EpAb2-6 + Chemotherapy: Enhances irinotecan/fluorouracil efficacy in colon cancer models .

Future Directions

  1. Bispecific Antibodies: Cotargeting EpCAM with immune checkpoints (e.g., PD-1) to overcome resistance .

  2. Immunotoxins: Optimizing toxin potency while minimizing off-target effects (e.g., Oportuzumab Monatox) .

  3. Fully Human Antibodies: Phage-display libraries enable high-affinity candidates (e.g., m801) with therapeutic potential .

  4. Liquid Biopsy Applications: EpCAM-targeting antibodies may improve circulating tumor cell detection for metastasis monitoring .

Product Specs

Introduction
EPCAM is a carcinoma-associated antigen and belongs to a family which includes at least 2 type I membrane proteins. The EPCAM protein has a role in embryonic stem cells proliferation and differentiation. EPCAM is used as a target for immunotherapy treatment of human carcinomas. EPCAM is expressed on most normal epithelial cells and gastrointestinal carcinomas and acts as a homotypic calcium-independent cell adhesion molecule. Epithelial cell adhesion molecules (EPCAM) can act as a physical homophilic interaction molecule between intestinal epithelial cells (IECs) and intraepithelial lymphocytes (IELs) at the mucosal epithelium for supplying immunological barrier as a first line of defense against mucosal infection. EPCAM gene mutations result in congenital tufting enteropathy.
Physical Appearance
Sterile filtered colorless solution.
Formulation
1mg/ml containing PBS, pH-7.4, 10% Glycerol and 0.02% Sodium Azide.
Storage Procedures
For periods up to 1 month store at 4°C, for longer periods of time, store at -20°C. Prevent freeze thaw cycles.
Stability / Shelf Life
12 months at -20°C. 1 month at 4°C.
Applications
The antibody has been tested by ELISA, Western blot analysis to assure specificity and reactivity. Since application varies, however, each investigation should be titrated by the reagent to obtain optimal results. Recommended starting dilution is 1:500.
Synonyms
Epithelial cell adhesion molecule, Ep-CAM, Adenocarcinoma-associated antigen, Cell surface glycoprotein Trop-1, Epithelial cell surface antigen, Epithelial glycoprotein, EGP, Epithelial glycoprotein 314, EGP314, hEGP314, KS 1/4 antigen, KSA, Major gastrointestinal tumor-associated protein GA733-2, Tumor-associated calcium signal transducer 1, CD326, EPCAM, GA733-2, M1S2, M4S1, MIC18, TACSTD1, TROP1, ESA, MK-1, DIAR5, EGP-2, EGP40, KS1/4, HNPCC8.
Purification Method
EPCAM antibody was purified from mouse ascitic fluids by protein-A affinity chromatography.
Type
Mouse Anti Human Monoclonal.
Clone

PAT36F10AT.

Immunogen
Anti-human EPCAM mAb, clone PAT37F9AT, is derived from hybridization of mouse F0 myeloma cells with spleen cells from BALB/c mice immunized with a recombinant human EPCAM protein 24-265 amino acids purified from E. coli.
Ig Subclass
Mouse IgG1 heavy chain and k light chain.

Q&A

What is EPCAM and why is it significant for antibody development?

EPCAM (Epithelial Cell Adhesion Molecule) is a transmembrane glycoprotein with a molecular weight of 34.9 kDa, consisting of 314 amino acid residues in humans. It primarily localizes to the cell membrane and demonstrates notable expression across multiple tissues, particularly in the appendix and colon . EPCAM functions as a homophilic interaction molecule between intestinal epithelial cells (IECs) and intraepithelial lymphocytes (IELs), providing an immunological barrier against mucosal infections .

The significance of EPCAM as an antibody target stems from its role as a tumor-associated antigen that shows elevated expression in various cancer cell lines and tumor tissues . This overexpression pattern has established EPCAM as a valuable biomarker for detection and potential therapeutic targeting, making anti-EPCAM antibodies crucial tools in cancer research.

Researchers should note that EPCAM undergoes post-translational modifications, particularly glycosylation, which can affect antibody recognition and binding characteristics . When designing experiments with anti-EPCAM antibodies, these modifications should be considered, especially when comparing results across different tissue types or cell lines.

What are the primary applications of EPCAM antibodies in research settings?

EPCAM antibodies serve multiple critical functions in research environments:

  • Detection Methods: These antibodies are widely employed in Western blotting (WB), flow cytometry (FCM), immunohistochemistry (IHC), immunoprecipitation (IP), ELISA, and immunofluorescence (IF) .

  • Circulating Tumor Cell (CTC) Research: Anti-EPCAM antibodies provide a means for immunomagnetic enrichment and detection of EpCAM-expressing circulating tumor cells from blood samples, serving as liquid biopsy tools .

  • Tumor Characterization: Researchers use these antibodies to identify and characterize epithelial tumor cells based on EPCAM expression levels, which can correlate with prognostic outcomes .

  • Therapeutic Development: Anti-EPCAM antibodies serve as foundations for developing antibody-drug conjugates (ADCs) and immunotherapeutic approaches against EPCAM-expressing tumors .

For research requiring dual targeting approaches, studies have demonstrated that combining antibodies against EPCAM with those targeting other markers (such as CD-49f for EPCAM-low populations) can enhance detection sensitivity in heterogeneous samples .

How should researchers select appropriate anti-EPCAM antibodies for specific applications?

Selection criteria for anti-EPCAM antibodies should be guided by several experimental considerations:

  • Epitope Specificity: Consider whether antibodies target the EpCL region (amino acids 24-80), which studies have shown to be more antigenic than the EpRE region (81-265) . Epitope mapping analyses can identify antibodies with distinct binding properties relevant to your research question.

  • Application Compatibility: Verify antibody validation for specific applications. Not all anti-EPCAM antibodies perform equally across different techniques; some may excel in flow cytometry but perform poorly in fixed tissue IHC applications .

  • Species Reactivity: Many anti-EPCAM antibodies show cross-reactivity between human, mouse, and rat samples . Researchers should carefully verify species reactivity when working with model organisms.

  • Clonality Considerations: For detection of native conformational epitopes, select monoclonal antibodies validated for recognizing the protein in its native state. For studies requiring detection of denatured protein, consider antibodies validated for Western blotting .

  • Clone Selection: When available, review citation records and published validation data for specific clones. For instance, search results mention clone E144 with 61 citations and 29 figures, providing evidence of successful application in peer-reviewed research .

What methods exist for developing novel anti-EPCAM monoclonal antibodies?

Researchers seeking to develop custom anti-EPCAM antibodies can employ several methodological approaches:

  • Cell-Based Immunization and Screening (CBIS): This approach involves immunizing mice with cells expressing EPCAM (e.g., CHO/EPCAM cells) followed by hybridoma generation through fusion with P3U1 cells. Initial screening through flow cytometry identifies hybridoma supernatants positive for EPCAM-expressing cells while negative for control cells. Secondary validation through immunohistochemistry and Western blotting confirms specificity, as demonstrated in the development of EpMab-16 (IgG2a, κ) .

  • Transchromosomic Mouse Technology: For generating fully human anti-EPCAM antibodies, transchromosomic mice carrying mini-chromosomes with human immunoglobulin loci have proven effective. These TC-mAb mice maintain engineered mouse artificial chromosomes (MAC) containing human Ig heavy and kappa light chain loci (IGH and IGK) in a mouse Ig knockout background, facilitating production of diverse human antibodies against EPCAM .

  • Native Conformational Recognition Analysis: To identify antibodies recognizing native EPCAM, researchers can employ conformational epitope mapping using differential binding studies between native and denatured protein. In one study, analysis of 72 mAbs reacting with native EPCAM revealed the EpCL region (amino acids 24-80) exhibited higher antigenicity than the EpRE region (81-265) .

When developing novel antibodies, standardized evaluation of functional properties such as antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) can identify candidates with therapeutic potential .

How can researchers evaluate EPCAM expression in tissue samples?

Robust evaluation of EPCAM expression in tissue samples requires standardized methodologies:

  • Immunohistochemistry Protocol: Treatment of formalin-fixed paraffin-embedded tissue sections with anti-EPCAM antibodies (e.g., OCAb9-1 at 1 μg/ml) for 1 hour at room temperature, followed by polymer-based detection systems such as the Super Sensitive IHC detection system provides reliable results. Visualization using DAB (0.02%) containing 0.03% H₂O₂ as a chromogen, with hematoxylin counterstaining, allows for assessment of EPCAM localization and expression patterns .

  • Quantitative Scoring Systems: Implementation of standardized scoring methods enhances reproducibility. One validated approach calculates a total staining score as the product of a proportion score (0-3) and an intensity score (0-3):

    • Proportion score: 0 (<10%), 1 (10-30%), 2 (30-50%), 3 (>50%) of positively stained tumor cells

    • Intensity score: 0 (no staining), 1 (weak), 2 (moderate), 3 (strong)

    • Total score range: 0-9, with scores ≥3 typically classified as high expression

  • Digital Pathology Analysis: Software tools like HistoQuest can provide quantitative assessment of EPCAM expression in tissue microarrays, reducing subjective interpretation variability and enabling more precise correlation with clinical outcomes .

For comparative studies, researchers should maintain consistent antibody concentrations, incubation periods, and detection methods across all samples to minimize technical variations that could confound biological differences.

What are the challenges in detecting EPCAM-low expressing populations?

Detection of EPCAM-low expressing cell populations presents several methodological challenges:

  • Variable Expression Levels: RNA-level analysis demonstrates that only 55.6% and 36.4% of circulating tumor cells show EPCAM positivity in some studies, indicating substantial heterogeneity that standard EPCAM-dependent enrichment methods might miss .

  • Epithelial-Mesenchymal Transition: During cancer progression, cells undergoing epithelial-mesenchymal transition (EMT) may downregulate EPCAM, making these potentially important populations difficult to detect using traditional EPCAM-based approaches.

  • Alternative Capture Strategies: Studies have established alternative capture approaches targeting proteins like CD-49f (encoded by ITGA6) that show significantly higher expression in EPCAM-negative or low-expressing cells. This provides a complementary approach for capturing these biologically relevant populations .

Researchers can address these challenges through:

  • Sequential Enrichment Protocols: Implementing sequential analysis that first depletes EPCAM-positive cells followed by capture with alternative markers can reveal previously undetected populations. This two-step approach has demonstrated the capacity to identify clinically relevant cells from the EPCAM-depleted fraction .

  • Synergistic Multi-marker Approaches: Combining antibodies targeting different surface proteins simultaneously has demonstrated synergistic effects in CTC capture. For instance, using antibodies against both Trop-2 (frequently co-expressed with EPCAM) and CD-49f (enriched in EPCAM-low cells) increased CTC detection significantly compared to either antibody alone .

What is the prognostic significance of EPCAM expression heterogeneity?

The heterogeneity of EPCAM expression has important prognostic implications:

Understanding these relationships requires sophisticated approaches that can capture both populations while preserving their distinct identities for comparative analysis.

How are anti-EPCAM antibodies utilized in antibody-drug conjugate development?

Development of antibody-drug conjugates (ADCs) targeting EPCAM involves several specialized approaches:

  • Direct Labeling Techniques: Anti-EPCAM antibodies can be directly labeled with cytotoxic agents such as DM1 (a maytansine derivative) using methods like the affinity peptide-based chemical conjugation (CCAP) method . This approach has demonstrated efficacy even with lower-affinity antibodies, suggesting advantages in therapeutic development.

  • Efficacy Assessment: Cytotoxicity evaluation against human cancer cell lines (e.g., colon cancer) provides critical data on the therapeutic potential of anti-EPCAM ADCs. Both high-affinity and low-affinity mAbs have shown clear cytotoxicity when conjugated using appropriate methods .

  • Antibody Selection Criteria: For ADC development, antibodies must demonstrate specific binding to EPCAM-expressing cancer cells while minimizing off-target effects. The search results indicate that fully human mAbs generated from TC-mAb mice provide a diverse repertoire of candidates for ADC development .

The effectiveness of anti-EPCAM ADCs is influenced by both antibody properties (affinity, specificity, internalization rate) and conjugation methodology, highlighting the importance of comprehensive characterization during development.

What emerging applications exist for dual-targeting strategies with EPCAM antibodies?

Innovative dual-targeting approaches involving EPCAM antibodies are advancing several research areas:

  • Complementary Marker Combinations: The combination of antibodies targeting Trop-2 (predominantly expressed on EPCAM high-expressing CTCs) and CD-49f (particularly present on EPCAM low-expressing CTCs) has demonstrated synergistic effects in capturing heterogeneous CTC populations, providing more comprehensive tumor representation than single-marker approaches .

  • Sequential Enrichment Protocols: Targeting EPCAM-depleted fractions with secondary markers has revealed CTC populations not captured by EPCAM-dependent technologies but still harboring valuable predictive information . This sequential approach maximizes detection of diverse tumor cell populations.

  • Morphological and Genetic Characterization: Dual-targeting strategies enable comparative analysis of different CTC subpopulations, revealing that despite morphological and phenotypic differences, EPCAM high-expressing and low-expressing CTCs often share similar chromosomal aberrations and mutations, suggesting a close evolutionary relationship .

These approaches highlight the value of multiparametric strategies that can capture the biological diversity of tumor cells in circulation, providing more comprehensive biomarker information than single-marker methods.

What controls should be implemented when developing or using anti-EPCAM antibodies?

Robust experimental design for anti-EPCAM antibody research requires comprehensive controls:

  • Cell Line Controls: When developing new antibodies, include both positive controls (cells known to express EPCAM, such as Caco-2 colorectal adenocarcinoma cells) and negative controls (EPCAM-negative cell lines like CHO-K1) . This dual-control approach enables verification of antibody specificity.

  • Isotype Controls: Include appropriate isotype-matched control antibodies to distinguish specific binding from non-specific Fc receptor interactions or other background binding phenomena.

  • Blocking Studies: Conduct competitive binding experiments with known anti-EPCAM antibodies or recombinant EPCAM protein to confirm epitope specificity and validate binding characteristics.

  • Cross-Reactivity Assessment: Test antibodies against related family members or potential cross-reactive proteins to ensure specificity for the intended EPCAM target.

  • Application-Specific Controls: For immunohistochemistry, include positive and negative tissue controls with known EPCAM expression patterns. For flow cytometry, employ fluorescence-minus-one (FMO) controls to set accurate gating boundaries .

Implementing these controls systematically enhances data reliability and facilitates troubleshooting when unexpected results occur.

How can researchers optimize immunohistochemical detection of EPCAM?

Optimization of immunohistochemical detection requires attention to several technical parameters:

  • Antibody Concentration Titration: Perform systematic titration experiments to determine optimal antibody concentration. Starting points based on published protocols suggest 1 μg/ml for antibodies like OCAb9-1, but optimal concentration may vary by antibody clone and tissue type .

  • Antigen Retrieval Methods: Compare heat-induced epitope retrieval using citrate buffer (pH 6.0) versus EDTA buffer (pH 9.0) to determine optimal conditions for exposing EPCAM epitopes in formalin-fixed tissues.

  • Detection System Selection: Polymer-based detection systems like the Super Sensitive IHC detection system have demonstrated efficacy for EPCAM visualization. The protocol involves sequential application of Super Enhancer reagent (20 minutes at room temperature) followed by Poly-HRP reagent (30 minutes at room temperature) .

  • Chromogen Development: Use DAB (0.02%) containing 0.03% H₂O₂ as a chromogen with careful timing to achieve optimal signal-to-noise ratio without overdevelopment .

  • Quantification Standards: Implement standardized scoring systems that account for both proportion of positive cells and staining intensity. The total score approach (range 0-9) with defined thresholds for high expression (score ≥3) enables consistent classification across samples .

Optimization should be performed systematically, changing one variable at a time while maintaining others constant to identify the specific contribution of each parameter to staining quality.

What are emerging applications of anti-EPCAM antibodies in cancer immunotherapy?

Anti-EPCAM antibodies are finding innovative applications in cancer immunotherapy development:

  • ADCC and CDC Mechanisms: Specifically engineered anti-EPCAM monoclonal antibodies like EpMab-16 (IgG2a, κ) have demonstrated the capacity to induce antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC), suggesting potential applications in therapeutic antibody development .

  • Bispecific Antibody Constructs: Development of bispecific antibodies incorporating anti-EPCAM binding domains alongside T-cell engaging components represents an emerging approach to redirect immune responses against EPCAM-expressing tumors.

  • Chimeric Antigen Receptor (CAR) T-cell Therapy: EPCAM-targeting single-chain variable fragments derived from characterized anti-EPCAM antibodies are being incorporated into CAR constructs for adoptive cell therapy approaches against EPCAM-expressing solid tumors.

  • Combination Therapeutic Strategies: The differential prognostic significance of EPCAM high-expressing versus low-expressing CTCs suggests potential value in combinatorial approaches targeting both populations to prevent escape of treatment-resistant subclones .

These emerging approaches leverage the specificity of anti-EPCAM antibodies while enhancing their therapeutic potential through innovative engineering and combination strategies.

What future developments might enhance EPCAM antibody utility in liquid biopsy applications?

Several promising directions could enhance the utility of EPCAM antibodies in liquid biopsy applications:

  • Multiparametric CTC Capture Systems: Further development of technologies combining antibodies targeting complementary markers (e.g., EPCAM, Trop-2, CD-49f) could enhance sensitivity for detecting heterogeneous CTC populations, providing more comprehensive tumor representation .

  • Integration with Molecular Profiling: Combining CTC capture using optimized anti-EPCAM antibodies with downstream single-cell sequencing could enable more detailed characterization of tumor heterogeneity and evolution during treatment.

  • Automation and Standardization: Development of automated systems for sequential enrichment protocols could enhance reproducibility and clinical utility of approaches that target both EPCAM-positive and EPCAM-negative/low CTCs .

  • Predictive Biomarker Development: Research correlating the presence and characteristics of EPCAM-expressing CTCs with treatment response could establish more precise predictive biomarkers to guide therapeutic decision-making.

These developments would address current limitations in capturing the full spectrum of circulating tumor cells while enhancing the clinical utility of liquid biopsy approaches based on EPCAM detection.

Product Science Overview

Introduction

Epithelial Cell Adhesion Molecule (EpCAM), also known as CD326, is a transmembrane glycoprotein that plays a crucial role in cell-cell adhesion within epithelial tissues. It is involved in various cellular processes, including signaling, migration, proliferation, and differentiation . EpCAM has gained significant attention due to its role in cancer biology, where it is often overexpressed in epithelial-derived tumors and serves as a biomarker for cancer diagnosis and prognosis .

Discovery and Structure

EpCAM was first identified in the late 1970s as a tumor-associated antigen recognized by monoclonal antibody 17-1A after immunization of mice with human colorectal cancer cells . It belongs to the GA733 protein family and is characterized by its homophilic binding properties, which facilitate Ca²⁺-independent cell-cell adhesion . The molecule consists of an extracellular domain (EpEX), a single transmembrane domain, and an intracellular domain (EpICD) .

Functions and Mechanisms

EpCAM is involved in a variety of cellular functions beyond cell adhesion. It participates in signal transduction, acting as a ligand for receptors and as a transcriptional cofactor . The extracellular domain (EpEX) and intracellular domain (EpICD) can be cleaved and released from the membrane, further influencing cellular signaling pathways . EpCAM’s role in cancer progression is complex and context-dependent, involving interactions with signaling pathways such as Wnt/β-catenin, TGF-β/SMAD, and PI3K/AKT/mTOR .

Clinical Applications

EpCAM’s overexpression in various carcinomas makes it a valuable target for cancer diagnosis and therapy. It is used as a diagnostic marker for detecting circulating tumor cells (CTCs) and cancer stem cells (CSCs) . Additionally, EpCAM-targeted therapies, including monoclonal antibodies and immunotherapies, are being developed to treat epithelial-derived tumors .

Mouse Anti Human EpCAM Antibodies

Mouse anti-human EpCAM antibodies are monoclonal antibodies generated by immunizing mice with human EpCAM. These antibodies are widely used in research and clinical settings to study EpCAM expression, function, and its role in cancer. They are also employed in diagnostic assays to detect EpCAM-positive cells in patient samples .

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.