EPCAM Human

Epithelial Cell Adhesion Molecule Human Recombinant
Shipped with Ice Packs
In Stock

Description

Molecular Structure of EPCAM Human

EPCAM is a 30–40 kDa type I transmembrane glycoprotein composed of 314 amino acids. Its structure includes three domains:

DomainCharacteristicsAmino Acid Range
Extracellular (EpEX)Contains epidermal growth factor (EGF)-like and thyroglobulin-like domains; mediates homotypic adhesion24–265
TransmembraneSingle-spanning α-helix anchoring the protein to the cell membrane266–288
Intracellular (EpICD)Short cytoplasmic tail involved in signaling via proteolytic cleavage (e.g., γ-secretase)289–314
  • The extracellular domain forms a heart-shaped dimer stabilized by three N-glycosylation sites (Asn74, Asn111, Asn198) .

  • Recombinant EPCAM proteins (e.g., PRO-1322, 9277-EP) are produced in E. coli or HEK293 cells, with variations in glycosylation and molecular weight (30–42 kDa) .

Biological Functions

EPCAM plays multifaceted roles in epithelial biology and pathology:

  • Cell Adhesion: Mediates Ca²⁺-independent homotypic interactions via EpEX domain oligomerization .

  • Signaling: Cleavage of EpICD by γ-secretase activates oncogenic pathways (e.g., upregulates c-myc, cyclins A/E) .

  • Stem Cell Regulation: Maintains pluripotency in embryonic and cancer stem cells .

  • Morphogenesis: Essential for gastrulation in zebrafish and placental development in mice .

Diagnostic and Therapeutic Applications

ApplicationDetailsReference
Cancer BiomarkerOverexpressed in 80% of epithelial cancers (e.g., colorectal, breast, prostate)
Circulating Tumor Cells (CTCs)Gold-standard marker for CTC detection in liquid biopsies
ImmunotherapyTargeted by monoclonal antibodies (e.g., MOC31, UBS54); limited by pancreatic toxicity in clinical trials

Genetic Disorders

  • Congenital Tufting Enteropathy (CTE): Linked to biallelic EPCAM mutations causing intestinal malabsorption .

  • Lynch Syndrome: EPCAM deletions induce epigenetic silencing of MSH2, increasing colorectal cancer risk .

Recombinant EPCAM Proteins in Research

ProductSourceMolecular WeightKey Features
PRO-1322E. coli30.1 kDaNon-glycosylated; His-tagged; used in adhesion assays
9277-EPHEK29328 kDa (predicted)Glycosylated; supports fibroblast adhesion (ED₅₀: 0.4–2.4 µg/mL)

Research Advancements

  • Structural Insights: Crystal structures reveal EpEX dimerization and glycan shielding of proteolytic sites, informing drug design .

  • Transcriptional Regulation: EPCAM promoter activity is modulated by Wnt/β-catenin and repressed by Zeb1 during epithelial-mesenchymal transition (EMT) .

  • Therapeutic Challenges: Humanized anti-EPCAM antibodies (e.g., MOC31-hFc) show variable biodistribution in transgenic models, highlighting engineering hurdles .

Product Specs

Introduction
EPCAM, a carcinoma-associated antigen, belongs to a family of at least two type I membrane proteins. It plays a role in embryonic stem cell proliferation and differentiation. EPCAM serves as a target for immunotherapy in human carcinomas. Expressed on most normal epithelial cells and gastrointestinal carcinomas, EPCAM acts as a homotypic calcium-independent cell adhesion molecule. It facilitates physical homophilic interactions between intestinal epithelial cells (IECs) and intraepithelial lymphocytes (IELs) at the mucosal epithelium, contributing to the immunological barrier against mucosal infection. Mutations in the EPCAM gene can lead to congenital tufting enteropathy.
Description
Recombinant human EPCAM, produced in E. coli, is a single, non-glycosylated polypeptide chain consisting of 267 amino acids (residues 24-265). It has a molecular weight of 30.1 kDa. The protein includes a 25-amino acid His-tag fused at the N-terminus and is purified using proprietary chromatographic techniques.
Physical Appearance
Clear, colorless, and sterile-filtered solution.
Formulation
The EPCAM protein solution is provided at a concentration of 1 mg/ml in a buffer containing 20 mM Tris-HCl (pH 8.0), 0.4 M urea, and 10% glycerol.
Stability
For short-term storage (up to 2-4 weeks), the product can be stored at 4°C. For extended storage, it is recommended to freeze the product at -20°C. Adding a carrier protein (0.1% HSA or BSA) is advised for long-term storage. Repeated freezing and thawing should be avoided.
Purity
The purity of the EPCAM protein is greater than 85%, as determined by SDS-PAGE analysis.
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.
Source
Escherichia Coli.
Amino Acid Sequence
MGSSHHHHHH SSGLVPRGSH MGSHMQEECV CENYKLAVNC FVNNNRQCQC TSVGAQNTVI CSKLAAKCLV MKAEMNGSKL GRRAKPEGALQNNDGLYDPD CDESGLFKAK QCNGTSMCWC VNTAGVRRTD KDTEITCSER VRTYWIIIEL KHKAREKPYD SKSLRTALQK EITTRYQLDP KFITSILYEN NVITIDLVQN SSQKTQNDVD IADVAYYFEK DVKGESLFHS KKMDLTVNGE QLDLDPGQTL IYYVDEKAPE FSMQGLK.

Q&A

What is EpCAM and what is its basic structure?

EpCAM is a 38-kDa transmembrane glycoprotein that forms a heart-shaped dimer at cell surfaces. The extracellular part (EpEX) consists of three domains arranged in a triangular fashion: N-Domain (ND), Thyroglobulin type 1A domain (TY), and C-Domain (CD). The protein contains three N-glycosylation sites (Asn74, 111, 198) that contribute to protein stability and cover lateral protein surfaces . The intracellular cytoplasmic domain anchors EpCAM to the cytoskeleton through interaction with α-actinin and contains a putative PDZ binding site at the C-terminus that facilitates interactions with signaling or structural proteins .

How is EpCAM expressed in normal and cancerous human tissues?

EpCAM expression is predominantly limited to normal and malignant epithelia, making it an effective diagnostic marker for carcinoma cells in mesenchymal organs such as blood, bone marrow, or lymph nodes . A comprehensive tissue microarray analysis of 14,832 samples from 120 different tumor categories revealed that EpCAM staining was detectable in 99 tumor categories . Among 78 epithelial tumor types, 60 categories showed ≥90% EpCAM positivity, including adenocarcinomas, neuroendocrine neoplasms, and germ cell tumors . Lower expression was observed in hepatocellular carcinomas, adrenocortical tumors, renal cell neoplasms, and poorly differentiated carcinomas .

What biological functions does EpCAM perform beyond cellular adhesion?

Although initially classified as a cell adhesion molecule, EpCAM demonstrates diverse biological functions far beyond intercellular adhesion. Recent studies have revealed its role in regulating cell proliferation, migration, stemness, and epithelial-to-mesenchymal transition in both normal and neoplastic epithelial cells . EpCAM and its fragments interact with various proteins including claudins, CD44, and E-cadherin, and regulate growth-relevant proteins such as c-Myc, Cyclin A, E, and D1 .

How is EpCAM gene expression regulated?

The EPCAM gene expression is controlled at the transcriptional level through multiple mechanisms. The proximal promoter region lacks typical TATA and CAAT boxes but contains eukaryotic promoter elements like initiator consensus sequences and GC boxes, as well as binding sequences for transcription factors such as SP-1, AP-1, AP2, Ets, ESE-1, and E-pal-like transcription factors . Epigenetic regulation through DNA methylation also influences EpCAM expression, though correlation with tissue expression varies by cancer type . Histone modifications and enzymes like histone acetyl transferase p300/CBP contribute to EPCAM gene regulation, with TNFα stimulation leading to repression through NF-kB recruitment of p300/CBP .

What are the best methods to detect EpCAM in human samples for research purposes?

For detecting EpCAM in human samples, immunohistochemistry (IHC) remains the gold standard. Recent methodological advancements involve using rabbit recombinant antibodies like MSVA-326R for optimal specificity . The recommended protocol involves:

  • Tissue preparation: Deparaffinization with xylol, rehydration through graded alcohol series

  • Antigen retrieval: Heat-induced for 5 minutes in an autoclave at 121°C in pH 7.8 Tris-EDTA buffer

  • Blocking: Peroxidase blocking solution for 10 minutes

  • Primary antibody application: Anti-EpCAM at 1:150 dilution, 37°C for 60 minutes

  • Visualization: Through appropriate secondary antibody system

When comparing detection methods, researchers should consider that EpCAM shows higher positivity rates than CKpan in testicular seminomas and neuroendocrine neoplasms, while CKpan performs better in hepatocellular carcinomas, mesotheliomas, and poorly differentiated non-neuroendocrine tumors .

How can researchers effectively study EpCAM signaling pathways?

EpCAM signaling involves regulated intramembrane proteolysis (RIP), which requires specific experimental design to capture the dynamic process. The signaling cascade includes:

  • Initial cleavage by ADAM17/TACE (triggered by cell-to-cell contact), releasing the soluble fragment EpEX

  • Secondary cleavage by γ-secretase complexes at distinct ε- and γ-sites, producing soluble extracellular Aβ-like fragments and intracellular domain EpIC

  • Release of EpIC into the cytosol where it mediates downstream signaling

To effectively study these pathways, researchers should employ cell-based assays that can track the proteolytic fragments using mass spectrometry for precise identification of cleavage sites, as demonstrated by Tsaktanis et al. . Co-immunoprecipitation experiments are valuable for identifying interaction partners of EpIC and other EpCAM fragments.

What transgenic models are available for EpCAM research, and how should they be utilized?

Several transgenic mouse models have been developed to study EpCAM biology in vivo. Constitutive and inducible CTE-associated murine models have been engineered by creating EPCAM knockout mice. These models demonstrate enhanced intestinal permeability and migration as well as decreased ion transport .

For studying anti-EpCAM antibody biodistribution and effects, a transgenic mouse tumor model that expresses human EpCAM similar to carcinoma patients has been developed. This model allows evaluation of treatment-associated effects before clinical trials and has been used to study the in vivo behavior of humanized and mouse-derived anti-EpCAM antibodies .

Key considerations when using these models include:

  • Comparing pharmacokinetics and tissue distribution patterns of different antibody formats

  • Monitoring dose-dependent uptake in EpCAM-expressing normal and tumor tissues

  • Evaluating potential toxicity in EpCAM-expressing normal tissues

How can researchers accurately quantify EpCAM expression levels in clinical samples?

Accurate quantification of EpCAM expression in clinical samples requires standardized methodologies:

  • Tissue microarray (TMA) approach: Using freshly prepared TMA sections immunostained in a single experiment to ensure consistent results across multiple samples

  • Scoring system: Developing a standardized scoring system combining intensity and percentage of positive cells

  • Digital pathology: Implementing automated image analysis for objective quantification

  • Validation: Comparing results with alternative methods like RT-PCR or Western blotting

  • Reference standards: Including known positive and negative controls in each experiment

When interpreting results, researchers should be aware that EpCAM expression can vary by tumor type and differentiation state, with positivity rates ≥90% in well-differentiated adenocarcinomas but lower rates in poorly differentiated carcinomas .

How is EpCAM utilized for circulating tumor cell (CTC) detection, and what are the methodological challenges?

EpCAM has become the most commonly used epithelial marker for capturing circulating tumor cells (CTCs) in the blood circulation of carcinoma patients . The methodological approach typically involves:

  • Enrichment: Using anti-EpCAM antibodies conjugated to magnetic beads to isolate EpCAM-positive cells from blood samples

  • Identification: Further characterizing captured cells with additional markers to confirm their tumoral origin

  • Molecular analysis: Performing genomic or transcriptomic analysis on isolated CTCs

Challenges in this methodology include:

  • False negatives due to EpCAM downregulation during epithelial-to-mesenchymal transition

  • Variable EpCAM expression levels across different carcinoma types

  • Need for highly sensitive detection methods due to the rarity of CTCs

  • Requirement for fresh samples and standardized processing protocols

Researchers should consider implementing complementary markers alongside EpCAM to improve CTC detection sensitivity, particularly in carcinomas known to have lower EpCAM expression like hepatocellular or renal cell carcinomas .

What is the role of EpCAM in distinguishing malignant mesotheliomas from adenocarcinomas?

EpCAM immunohistochemistry serves as a valuable diagnostic tool for distinguishing malignant mesotheliomas from adenocarcinomas. Comparative analysis has shown that while adenocarcinomas typically express high levels of EpCAM, malignant mesotheliomas consistently show negative or minimal EpCAM staining .

When implementing this diagnostic approach:

  • Use standardized antibodies and protocols for consistent results

  • Include appropriate positive and negative controls

  • Compare EpCAM with other markers like TROP2, which similarly shows positivity in epithelial tumors but not in malignant mesotheliomas

  • Consider that CKpan markers may show higher positivity rates in mesotheliomas compared to EpCAM

This differential expression pattern makes EpCAM a reliable marker in the diagnostic workup of pleural and peritoneal malignancies.

How should researchers interpret conflicting data regarding EpCAM's prognostic significance across different cancer types?

Conflicting reports on EpCAM's prognostic significance across cancer types reflect its complex, context-dependent biological functions. To properly interpret such conflicting data, researchers should:

  • Consider tumor type-specific biology: EpCAM may function differently in various tissue contexts

  • Evaluate methodological differences: Varied antibodies, scoring systems, and cutoff values may explain discrepancies

  • Account for tumor heterogeneity: Expression may vary within different regions of the same tumor

  • Correlate with molecular subtypes: EpCAM's significance may differ across molecular subtypes of the same cancer

  • Analyze in conjunction with other markers: Combinatorial marker patterns may provide clearer prognostic information than EpCAM alone

A meta-analysis approach combining multiple studies with clear documentation of methodological differences is recommended to reconcile conflicting findings .

What are the key considerations in developing EpCAM-targeted therapeutics for cancer?

Developing EpCAM-targeted therapeutics requires careful consideration of several factors:

  • Target specificity and accessibility: Understanding the differential expression between tumor and normal epithelial tissues to minimize on-target/off-tumor effects

  • Antibody format selection: Different antibody formats (mouse-derived, humanized, or fully human) demonstrate varied pharmacokinetics and tissue distribution patterns

  • Potential toxicity: Severe pancreatitis has been observed with some humanized or fully human anti-EpCAM antibodies in clinical settings

  • Epitope selection: Different epitopes may affect antibody efficacy and safety profiles

  • Molecular engineering: The design of antibodies significantly impacts in vivo behavior even when targeting the same epitope

Research has shown that mouse-derived mAbs directed to EpCAM have been used to treat colon carcinoma patients with tolerable toxic side effects but limited antitumor effects, while humanized or fully human anti-EpCAM mAbs produced stronger antitumor activity but more severe toxicity .

How can researchers effectively study EpCAM's role in cancer stem cells and tumor initiation?

Investigating EpCAM's role in cancer stem cells requires specialized methodologies:

  • Isolation techniques: Flow cytometry-based sorting of EpCAM-high cell populations from tumor samples

  • Functional assays: Sphere formation, serial transplantation, and limiting dilution assays to assess stemness properties

  • Lineage tracing: In vivo models to track the fate of EpCAM-positive cells during tumor development

  • Molecular profiling: RNA-seq and proteomics to identify stemness-associated pathways activated in EpCAM-high cells

  • Signaling pathway analysis: Focus on EpCAM's interactions with Wnt/β-catenin signaling and other stemness-related pathways

These approaches help elucidate how EpCAM contributes to cancer stemness, which has significant implications for understanding tumor initiation, progression, and therapeutic resistance .

What experimental methods best assess the functional consequences of EpCAM's regulated intramembrane proteolysis?

To study the functional consequences of EpCAM's regulated intramembrane proteolysis (RIP), researchers should employ:

  • Site-directed mutagenesis: To create cleavage-resistant EpCAM variants by mutating key amino acids at cleavage sites

  • Pharmacological inhibitors: Using ADAM17/TACE inhibitors or γ-secretase inhibitors to block specific steps in the RIP process

  • Fragment-specific antibodies: Developing antibodies that specifically recognize EpEX, EpIC, or Aβ-like fragments

  • Reporter systems: Creating fusion proteins with fluorescent or luminescent tags to monitor real-time proteolysis

  • Downstream signaling assays: Measuring activation of known EpIC targets to assess functional consequences

These methodologies help distinguish between functions mediated by full-length EpCAM versus those dependent on proteolytic processing, providing insights into the mechanistic basis of EpCAM's diverse biological effects .

How should researchers approach studying EpCAM in the context of epithelial-to-mesenchymal transition (EMT)?

EMT represents a critical process in cancer progression where EpCAM expression is dynamically regulated. An effective research approach includes:

  • Dynamic monitoring: Time-course experiments tracking EpCAM expression during EMT induction (e.g., TGF-β treatment)

  • Single-cell analysis: Accounting for heterogeneity in EMT status within tumor populations

  • Correlation studies: Analyzing the relationship between EpCAM expression and EMT markers (E-cadherin, vimentin, etc.)

  • Functional manipulation: Utilizing gain- and loss-of-function approaches to determine how EpCAM affects EMT progression

  • In vivo models: Studying EpCAM expression at invasion fronts and in circulating tumor cells from the same tumor

This comprehensive approach helps elucidate EpCAM's context-dependent roles during the complex process of EMT, which has significant implications for understanding metastasis and therapeutic resistance .

How does EpCAM compare with related markers like TROP2 and CKpan in research applications?

Comparative analysis of EpCAM, TROP2, and CKpan reveals important differences that researchers should consider:

MarkerStrengthsLimitationsBest Applications
EpCAM- High positivity in adenocarcinomas and neuroendocrine tumors
- Excellent for testicular seminomas
- Strong membranous staining pattern
- Lower sensitivity in hepatocellular carcinomas and renal cell tumors
- Variable in poorly differentiated carcinomas
- Mesothelioma vs. adenocarcinoma distinction
- CTC detection
- Neuroendocrine tumor identification
TROP2- Higher positivity in squamous cell carcinomas
- Stronger staining intensity in many carcinomas
- Lower positivity in GI adenocarcinomas
- Limited utility in neuroendocrine and testicular germ cell tumors
- Squamous cell carcinoma identification
- Complementary to EpCAM for broader epithelial detection
CKpan- Higher sensitivity in hepatocellular carcinomas
- Better for mesotheliomas and poorly differentiated tumors
- Lower specificity (some mesenchymal positivity)
- Less effective for neuroendocrine tumors
- Gold standard for epithelial differentiation
- Poorly differentiated carcinoma identification

This comparison indicates that EpCAM shows particularly high positivity rates in seminomas and neuroendocrine neoplasms compared to CKpan, while TROP2 demonstrates higher positivity rates in squamous cell carcinomas but lower rates in many gastrointestinal adenocarcinomas, testicular germ cell tumors, neuroendocrine neoplasms, and renal cell tumors .

What are the optimal preservation and processing methods for EpCAM detection in tissue samples?

To ensure optimal EpCAM detection, researchers should follow these tissue preservation and processing guidelines:

  • Fixation: 10% neutral buffered formalin for 24-48 hours is recommended for reliable EpCAM preservation

  • Processing: Standard paraffin embedding protocols with careful temperature control to prevent antigen degradation

  • Section preparation: 3-5 μm thick sections are optimal for immunohistochemical staining

  • Antigen retrieval: Heat-induced epitope retrieval in Tris-EDTA buffer (pH 7.8) at 121°C for 5 minutes provides optimal results

  • Storage considerations: Freshly cut sections yield better results than stored slides

These methodological details are critical for obtaining consistent and reliable EpCAM detection across different laboratories and studies.

How can researchers effectively validate novel anti-EpCAM antibodies for research applications?

Thorough validation of novel anti-EpCAM antibodies requires a systematic approach:

  • Epitope characterization: Identify the specific region of EpCAM recognized by the antibody

  • Specificity testing: Confirm binding to recombinant EpCAM and absence of cross-reactivity with related proteins like TROP2

  • Western blot validation: Verify detection of correct molecular weight bands in EpCAM-positive cell lines

  • Immunohistochemical testing: Compare staining patterns with established EpCAM antibodies across a tissue panel

  • Knockout/knockdown controls: Test antibody on EpCAM-knockout or knockdown samples to confirm specificity

  • Application-specific validation: Evaluate performance in the specific application (flow cytometry, immunoprecipitation, etc.)

Researchers should also consider the importance of antibody format (monoclonal vs. polyclonal, species of origin) and any post-translational modifications that might affect epitope recognition .

What are the most promising emerging areas of EpCAM research?

Several cutting-edge research directions are advancing our understanding of EpCAM biology:

  • Single-cell analysis of EpCAM expression heterogeneity within tumors and its correlation with functional cellular states

  • Structural biology approaches to elucidate the complete 3D structure of EpCAM in different conformational states

  • Investigation of EpCAM's role in the tumor microenvironment and immune cell interactions

  • Development of novel bispecific antibodies targeting EpCAM and immune checkpoints

  • Exploration of EpCAM as a biomarker for response to immunotherapy and targeted treatments

  • Understanding the role of soluble EpCAM fragments in the circulation as liquid biopsy biomarkers

These emerging areas represent promising opportunities for researchers to make significant contributions to the field .

How can researchers better understand the dual roles of EpCAM in promoting both adhesion and proliferation?

To resolve the apparent paradox of EpCAM's dual functions in adhesion and proliferation, researchers should:

  • Develop domain-specific functional assays: Separate experiments targeting different EpCAM domains to identify region-specific functions

  • Investigate conformational states: Study how EpCAM's conformation might switch between adhesion-promoting and signaling-promoting states

  • Analyze protein interaction networks: Use proteomics to identify context-specific binding partners in different cellular states

  • Employ real-time imaging: Monitor EpCAM dynamics and localization during cell adhesion versus proliferation events

  • Create domain-swap chimeras: Generate fusion proteins with domains from related molecules to map functional regions

This multifaceted approach can help elucidate how a single protein coordinates seemingly contradictory cellular processes in a context-dependent manner .

What novel methodologies are being developed to target EpCAM for precision oncology applications?

Innovative approaches for EpCAM-targeted precision oncology include:

  • Antibody-drug conjugates (ADCs): Engineering anti-EpCAM antibodies linked to potent cytotoxic payloads with optimized linker chemistry

  • CAR-T cell therapy: Developing chimeric antigen receptor T cells targeting EpCAM with built-in safety switches

  • RNA therapeutics: Using siRNA or antisense oligonucleotides to modulate EpCAM expression in tumors

  • Combination approaches: Targeting EpCAM in conjunction with immune checkpoint inhibitors or conventional chemotherapy

  • Theranostic applications: Developing dual-purpose agents that simultaneously image and treat EpCAM-positive tumors

Researchers exploring these approaches should carefully consider potential on-target/off-tumor toxicity given EpCAM's expression in normal epithelial tissues and implement appropriate safety strategies .

Product Science Overview

Introduction

The Epithelial Cell Adhesion Molecule (EpCAM), also known as CD326, is a transmembrane glycoprotein that plays a crucial role in cell-cell adhesion, signaling, migration, proliferation, and differentiation . It is encoded by the EPCAM gene in humans and is widely expressed in various epithelial tissues .

Discovery and Structure

EpCAM was first identified in 1979 as a tumor-associated antigen on colorectal carcinomas . It consists of an extracellular domain (EpEX) with 265 amino acid residues and an intracellular domain (EpICD) with 26 amino acid residues . The molecule mediates calcium-independent homotypic cell-cell adhesion in epithelial tissues .

Functions

EpCAM is involved in several critical cellular processes:

  • Cell Adhesion: It mediates homotypic cell-cell adhesion, which is essential for maintaining the integrity of epithelial tissues .
  • Cell Signaling: EpCAM participates in intracellular signaling pathways, including the WNT and Ras/Raf pathways .
  • Cell Proliferation and Differentiation: It plays a role in regulating cell proliferation and differentiation, contributing to tissue homeostasis .
  • Migration and Invasion: EpCAM is implicated in cell migration and invasion, particularly in the context of cancer metastasis .
Clinical Significance

EpCAM is overexpressed in many carcinomas, including colorectal, breast, and prostate cancers . Its high expression levels in tumors make it a valuable prognostic marker and a potential therapeutic target . EpCAM-specific recombinant immunotoxins have shown potent antitumor activity in preclinical studies .

Applications in Cancer Therapy

EpCAM has been explored as a target for cancer immunotherapy. Recombinant immunotoxins, which combine EpCAM-specific antibodies with cytotoxic agents, have demonstrated selective antitumor activity . These immunotoxins can inhibit protein synthesis and reduce the viability of EpCAM-positive carcinoma 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.