CEA Human, His refers to a recombinant form of human carcinoembryonic antigen (CEA) with a C-terminal polyhistidine (His) tag. This glycoprotein, encoded by the CEACAM5 gene, belongs to the immunoglobulin superfamily and is primarily used as a tumor marker due to its overexpression in gastrointestinal, pulmonary, breast, and thyroid cancers . The His tag facilitates purification via immobilized metal affinity chromatography (IMAC), making it a critical tool for research and therapeutic development .
Cell Adhesion: Mediates intercellular interactions in mucosal tissues .
Immune Modulation: Suppresses T-cell activation and promotes immune evasion .
Metastasis: Enhances anoikis resistance and liver metastasis in colorectal cancer .
Cancer Biomarker: Elevated serum CEA (>2.5 ng/mL) correlates with tumor burden and metastasis .
Therapeutic Target: CAR T-cell therapies and antibody-drug conjugates (e.g., labetuzumab govitecan) exploit CEA overexpression in tumors .
Prognostic Marker: Preoperative CEA >20 ng/mL predicts metastatic disease .
Post-Treatment Monitoring: Levels normalize within 4–6 weeks after successful tumor resection .
Purification: Proprietary chromatographic techniques yield >95% purity (SDS-PAGE) .
Formulation: 0.5 mg/mL in phosphate-buffered saline (pH 7.4) with 10% glycerol .
Parameter | Specification |
---|---|
Catalogue Number | PRO-2407 |
Stability | Long-term storage requires 0.1% HSA/BSA |
Glycosylation Profile | Heterogeneous, tissue-specific |
Non-Specific Elevation: Elevated CEA occurs in smokers, pancreatitis, and cirrhosis .
Therapeutic Challenges: Immune responses to murine-derived anti-CEA antibodies limit clinical use .
Carcinoembryonic Antigen Related Cell Adhesion Molecule 5, Carcinoembryonic Antigen-Related Cell Adhesion Molecule 5, Meconium Antigen 100, CEA, Carcinoembryonic Antigen, CD66e Antigen, CD66e, Carcinoembryonic antigen-related cell adhesion molecule 5, Carcinoembryonic antigen, CEA.
KLTIESTPFN VAEGKEVLLL VHNLPQHLFG YSWYKGERVD GNRQIIGYVI GTQQATPGPA YSGREIIYPN ASLLIQNIIQ NDTGFYTLHV IKSDLVNEEA TGQFRVYPEL PKPSISSNNS KPVEDKDAVA FTCEPETQDA TYLWWVNNQS LPVSPRLQLS NGNRTLTLFN VTRNDTASYK CETQNPVSAR RSDSVILNVL YGPDAPTISP LNTSYRSGEN LNLSCHAASN PPAQYSWFVN GTFQQSTQEL FIPNITVNNS GSYTCQAHNS DTGLNRTTVT TITVYAEPPK PFITSNNSNP VEDEDAVALT CEPEIQNTTY LWWVNNQSLP VSPRLQLSND NRTLTLLSVT RNDVGPYECG IQNKLSVDHS DPVILNVLYG PDDPTISPSY TYYRPGVNLS LSCHAASNPP AQYSWLIDGN IQQHTQELFI SNITEKNSGL YTCQANNSAS GHSRTTVKTI TVSAELPKPS ISSNNSKPVE DKDAVAFTCE PEAQNTTYLW WVNGQSLPVS PRLQLSNGNR TLTLFNVTRN DARAYVCGIQ NSVSANRSDP VTLDVLYGPD TPIISPPDSS YLSGANLNLS CHSASNPSPQ YSWRINGIPQ QHTQVLFIAK ITPNNNGTYA CFVSNLATGR NNSIVKSITV SASGTSPGLS ALEHHHHHH.
CEA (Carcinoembryonic antigen) is an oncofetal glycoprotein that belongs to the immunoglobulin (Ig) superfamily of proteins. It is also known as Carcinoembryonic antigen-related cell adhesion molecule 5 (CEACAM5), CD antigen CD66e, and Meconium antigen 100. Structurally, CEA is a glycophosphatidylinositol (GPI)-linked membrane-anchoring protein that is exposed on the cell surface facing the extracellular matrix. The protein can exist in both membrane-bound and soluble forms, as its anchoring region can be cleaved by phospholipase C and phospholipase D, resulting in soluble CEA that circulates through blood vessels .
The histidine tag (His-tag) in CEA Human, His preparations serves multiple research-critical functions. This poly-histidine sequence, typically consisting of six histidine residues, facilitates protein purification through immobilized metal affinity chromatography. For CEA research, the His-tag enables consistent protein isolation with high purity, crucial for experimental reproducibility. Additionally, the tag provides a uniform anchoring point for immobilization in binding assays, as demonstrated in protocols where CEA His Human immobilized at 1 μg/ml can bind Human CEA Antibody with a measurable EC50 of 2.323 ng/ml . This standardized orientation ensures that functional epitopes remain accessible, making His-tagged CEA especially valuable for antibody development and therapeutic targeting studies.
Verification of CEA Human, His activity requires multifaceted approaches that assess both binding capability and functional activity. A standard binding verification involves immobilization assays where CEA is coated at defined concentrations (typically 1 μg/ml) and its ability to bind specific antibodies is measured through detection systems such as anti-mouse Fc antibodies. Functional verification should include cell adhesion assays that assess CEA's ability to mediate homotypic and heterotypic cellular interactions, immune modulation assays that measure its impact on immune cell function, and anoikis resistance assays that evaluate its role in cell survival after detachment. Additionally, metastasis models using xenografts can assess CEA's contribution to metastatic potential .
Post-translational modifications of CEA, particularly glycosylation patterns, critically influence both detection sensitivity and functional properties. CEA contains multiple N-glycosylation sites that can vary between normal and cancerous tissues, affecting epitope accessibility and antibody recognition. Research indicates that altered glycosylation can modify CEA's role in cellular adhesion and immune evasion mechanisms. When designing experiments, researchers should consider employing multiple detection antibodies that recognize different epitopes to account for glycosylation heterogeneity. Additionally, glycan analysis techniques such as lectin arrays or mass spectrometry should be incorporated to characterize CEA glycoforms that may correlate with specific cancer progression stages or therapeutic responses .
Genetic modification of T-cell receptors (TCRs) for specific CEA recognition requires precise methodological approaches. Current research demonstrates success with isolating CEA-reactive TCRs from HLA-A2.1 transgenic mice immunized with CEA:691-699 peptide. These isolated TCRs can then be genetically introduced into human peripheral blood lymphocytes through RNA electroporation or retroviral transduction. Critical to this process is the introduction of amino acid substitutions throughout the complementarity determining regions (CDR1, CDR2, and CDR3) of both TCR α and β chains to enhance recognition of CEA . Researchers should employ functional verification through assays that measure T-cell activation, cytokine production, and specific lysis of CEA-expressing target cells to confirm the efficacy of the modified TCRs.
Addressing data variability in CEA quantification requires rigorous standardization and quality control protocols. ELISA-based approaches for CEA quantification demonstrate intra-assay variation of 3.2%-4.8% and inter-assay variation of 4.0%-6.5% . To minimize this variability, researchers should:
Establish consistent standard curves using recombinant CEA Human, His
Perform duplicate or triplicate measurements for each sample
Include control samples of known concentration in each assay
Account for interfering substances such as hemolytic, icteric, and lipemic components
Consider the dynamic range limitations (e.g., 0.596-100 ng/ml for standard ELISA)
Implement statistical methods like Bland-Altman plots to analyze method agreement
Additionally, researchers should be aware that factors such as smoking can elevate CEA levels, potentially confounding research results when using clinical samples .
Differentiating between membrane-bound and soluble CEA forms requires specialized experimental approaches. Membrane-bound CEA can be specifically detected through cell surface biotinylation followed by immunoprecipitation, flow cytometry with non-permeabilized cells, or confocal microscopy with membrane-specific counterstains. Soluble CEA analysis requires techniques like ultracentrifugation to separate membrane vesicles from truly soluble protein, followed by Western blotting or ELISA. Additionally, researchers can utilize phospholipase C treatment of cells to experimentally release GPI-anchored CEA, providing a controlled system to study the transition between the two forms. Functional studies comparing these forms should assess their differential effects on cell signaling, immune modulation, and metastatic potential .
Optimized binding assay protocols for CEA Human, His characterization typically employ a sandwich-based approach. The established protocol involves immobilizing CEA Human, His at 1 μg/ml (100 μl/well) which can then bind Human CEA Antibody (such as 1A5C3, Mouse) with EC50=2.323 ng/ml when detected by secondary antibodies like M6 Goat Anti-Mouse Fc . For research validity, background subtraction from data points before curve fitting is essential. Key optimization factors include:
Parameter | Recommended Condition | Notes |
---|---|---|
Coating buffer | PBS pH 7.4 | Maintains protein conformation |
Blocking solution | 1-3% BSA or casein | Reduces non-specific binding |
Incubation time | 1-2 hours at RT or overnight at 4°C | Temperature-dependent |
Washing steps | 3-5× with PBS-T (0.05% Tween-20) | Critical for reducing background |
Detection system | HRP-conjugated secondary antibody | Provides sensitive quantification |
Development of reliable CEA ELISA systems requires strict adherence to quality control parameters. Critical factors include antibody selection, where monoclonal antibodies directed towards unique antigenic sites of the CEA molecule provide highest specificity. The established dynamic range for CEA ELISA is typically 0.596-100 ng/ml, with analytical sensitivity calculated as the mean plus two standard deviations of twenty replicate analyses of Standard 0, typically <0.596 ng/ml .
Quality control parameters should include:
Intra-assay precision with coefficient of variation <5% (reported range: 3.2%-4.8%)
Inter-assay precision with coefficient of variation <7% (reported range: 4.0%-6.5%)
Assessment of interfering substances, particularly HAMA (Human Anti-Mouse Antibodies)
Evaluation of high-dose hook effect (absent up to 10,000 ng/ml for validated assays)
Establishment of clinical reference ranges with consideration for smoking status
Experimental controls for T-cell receptor studies targeting CEA require multifaceted design considerations. When isolating and genetically modifying T-cell receptors (TCRs) that specifically bind CEA peptide on human cancer cells, the following control structure is recommended:
Negative controls:
Non-transduced T-cells to assess baseline activity
T-cells with irrelevant TCRs to evaluate specificity
HLA-mismatched target cells to confirm HLA restriction
Positive controls:
T-cells with validated anti-tumor TCRs
Target cells pulsed with excessive peptide concentrations
Experimental validation:
Characterizing CEA's role in metastasis requires integrated in vitro and in vivo methodological approaches. CEA has been functionally associated with anoikis resistance and promotion of liver metastasis . Research protocols should include:
In vitro models:
Anoikis resistance assays using ultra-low attachment plates
3D spheroid formation assays to assess cell survival in suspension
Migration and invasion assays to quantify metastatic potential
Cell adhesion assays to characterize homotypic and heterotypic interactions
In vivo models:
Orthotopic xenograft models with CEA-expressing cells
Experimental metastasis models through tail vein injection
Genetic manipulation studies (knockdown/overexpression) to assess CEA's direct contribution
Molecular characterization:
Analysis of downstream signaling pathways activated by CEA
Assessment of integrin activation and extracellular matrix interactions
Evaluation of immune surveillance evasion mechanisms
CEA Human, His has substantial applications in developing cancer immunotherapies. Research demonstrates successful approaches involving genetic modification of T-cell receptors to specifically target CEA peptides. The methodology involves isolating CEA-reactive TCRs from HLA-A2.1 transgenic mice immunized with CEA:691-699 peptide, followed by genetic introduction of these TCRs into human peripheral blood lymphocytes through RNA electroporation or retroviral transduction .
Advanced approaches include amino acid substitutions throughout complementarity determining regions (CDR1, CDR2, and CDR3) of both TCR α and β chains to enhance recognition specificity and affinity. This strategy transforms previously non-reactive T cells into cells capable of recognizing both peptide-pulsed target cells and HLA-A2.1-matched tumor cells expressing CEA . Such CEA-targeted immunotherapies hold particular promise for gastrointestinal, respiratory, genitourinary, and breast cancers where CEA overexpression is well-documented .
Evaluating CEA as a diagnostic biomarker requires comprehensive analytical validation approaches. The ELISA methodology provides a foundational platform, utilizing a sandwich principle with microtiter wells coated with monoclonal antibodies directed towards unique antigenic sites of the CEA molecule. Sample CEA binds to this antibody and is then detected using enzyme conjugate (anti-CEA antibody conjugated with horseradish peroxidase) .
Critical evaluation parameters include:
Parameter | Typical Values | Significance |
---|---|---|
Dynamic Range | 0.596-100 ng/ml | Defines detection limits |
Analytical Sensitivity | <0.596 ng/ml | Establishes lower detection threshold |
Intra-Assay Precision | 3.2%-4.8% CV | Indicates reproducibility |
Inter-Assay Precision | 4.0%-6.5% CV | Reflects long-term reliability |
Interference Assessment | HAMA, heterophilic antibodies | Identifies false positive sources |
High-Dose-Hook Effect | Absent up to 10,000 ng/ml | Ensures accuracy at high concentrations |
Researchers must also consider population variables, such as elevated CEA levels in smokers, when establishing reference ranges for diagnostic applications .
When incorporating CEA into multi-biomarker cancer panels, several methodological considerations become critical for research validity. CEA's functional association with cellular interaction, cell adhesion, immune response, and anoikis resistance suggests complementary biomarkers should include factors involved in these pathways . Key considerations include:
Biomarker selection strategy:
Include markers with independent prognostic value
Select biomarkers representing different cancer hallmarks
Consider tissue-specific markers alongside CEA
Analytical harmonization:
Standardize sample processing procedures
Utilize multiplexed assay platforms when possible
Develop normalization strategies for cross-platform comparison
Data integration approaches:
Apply machine learning algorithms for pattern recognition
Develop weighted scoring systems based on biomarker performance
Implement longitudinal analysis methods for temporal changes
Validation requirements:
Perform cross-validation in independent cohorts
Establish clear incrementing value beyond CEA alone
Determine clinical decision thresholds through ROC analysis
Optimizing CEA-based theranostic approaches requires integrating diagnostic capabilities with therapeutic targeting. The structural characteristics of CEA as a GPI-linked membrane protein with well-defined epitopes make it suitable for dual-purpose applications . Optimization strategies include:
Antibody engineering:
Development of bispecific antibodies targeting CEA and effector cells
Creation of antibody-drug conjugates (ADCs) with CEA-specific binding domains
Engineering of imaging-capable antibodies with dual radiotracer/therapeutic payloads
T-cell-based approaches:
Targeted nanoparticle delivery:
Conjugation of CEA-specific antibodies to nanoparticles carrying therapeutic payloads
Development of theranostic nanoparticles with imaging and therapeutic capabilities
Optimization of particle size and surface chemistry for improved tumor penetration
Companion diagnostic integration:
Development of standardized ELISA or other detection methods to identify optimal patient populations
Establishment of CEA threshold levels that predict therapeutic response
Creation of real-time monitoring systems for treatment efficacy
Interference in CEA detection assays presents significant challenges to research validity. Common interfering substances include hemolytic, icteric, and lipemic components in serum samples, as well as Human Anti-Mouse Antibodies (HAMA) and heterophilic antibodies that can produce false positive results . Effective strategies to overcome these issues include:
Sample preprocessing:
Implement centrifugation protocols to remove lipemic components
Dilute samples with high bilirubin or hemoglobin content
Use sample additives that block heterophilic antibody interference
Assay modification:
Include blocking agents specific for HAMA interference
Utilize specialized buffer systems to minimize matrix effects
Implement wash procedures optimized for reducing non-specific binding
Alternative detection methods:
Consider mass spectrometry-based approaches for complex samples
Employ epitope-specific capture/detection antibody pairs
Utilize species-matched antibody systems to reduce HAMA effects
Validation protocols:
CEA belongs to a family of related proteins (CEACAM family), presenting specificity challenges in research contexts. Key strategies to ensure specificity include:
Epitope-focused approaches:
Select antibodies targeting CEA-specific epitopes not shared with related molecules
Implement competitive binding assays with recombinant CEACAM proteins
Utilize epitope mapping to confirm antibody specificity
Molecular techniques:
Employ RT-qPCR to distinguish CEA (CEACAM5) gene expression from related genes
Utilize siRNA knockdown to confirm specificity of functional observations
Implement CRISPR-Cas9 gene editing for precise functional studies
Analytical validation:
CEA Human, His experimental models present several critical limitations that researchers must address through careful experimental design:
Structural considerations:
The His-tag may influence protein folding or function in some experimental contexts
The tag could potentially mask or create epitopes not present in native CEA
Potential differences between recombinant and naturally expressed CEA glycosylation patterns
Functional limitations:
In vitro studies may not fully recapitulate the complex tumor microenvironment interactions
Murine models may not accurately reflect human immune interactions with CEA
GPI-anchored vs. soluble CEA may demonstrate different functional properties
Analytical constraints:
When faced with conflicting data in CEA research, systematic interpretation approaches are essential:
Methodological reconciliation:
Compare experimental protocols for differences in protein preparation, immobilization methods, and detection systems
Evaluate buffer compositions, incubation conditions, and washing procedures
Consider differences in CEA sources (recombinant vs. native, different expression systems)
Sample-related factors:
Assess variation in glycosylation patterns between sample sources
Consider potential interfering substances in clinical samples
Evaluate differences in CEA isoforms or cleavage products
Experimental design differences:
Compare antibody specificity profiles and epitope targets
Evaluate differences in control systems and normalization approaches
Consider cell line differences in expression studies
Reconciliation approaches:
The Human Recombinant CEA with His Tag is produced in Sf9 Baculovirus cells. It is a single, glycosylated polypeptide chain containing 659 amino acids and has a molecular mass of approximately 72.3 kDa . The recombinant protein is expressed with an 8 amino acid His tag at the C-terminus, which facilitates its purification through chromatographic techniques .
CEA is an important biomarker in oncology. Its levels are used to screen for cancer, evaluate the recurrence or dissemination of the disease, and determine the success of surgical removal of malignant tumors . Normal CEA levels range from 0.0 to 2.5 ng/ml in non-smokers, and levels above 20 ng/ml before treatment are often associated with metastatic cancer . Monitoring CEA levels post-treatment can help in detecting tumor recurrence .
The recombinant CEA protein with His tag is used in various research applications, including:
The recombinant CEA protein is supplied as a sterile, filtered colorless solution. It is formulated in phosphate-buffered saline (pH 7.4) with 10% glycerol . For short-term storage, it can be kept at 4°C for 2-4 weeks. For long-term storage, it is recommended to store the protein at -20°C with a carrier protein to prevent multiple freeze-thaw cycles .