The CEACAM5 Antibody, FITC conjugated is a fluorescently labeled monoclonal antibody designed to specifically target Carcinoembryonic Antigen-Related Cell Adhesion Molecule 5 (CEACAM5), a glycosylated cell surface protein overexpressed in various cancers, including colorectal, pancreatic, and prostate tumors . FITC (Fluorescein Isothiocyanate) conjugation enables visualization of CEACAM5 expression via fluorescence microscopy, flow cytometry, or immunohistochemistry. These antibodies are critical tools for studying cancer progression, immune evasion mechanisms, and therapeutic targeting of CEACAM5 .
CEACAM5 antibodies are employed across multiple experimental platforms:
Used to detect CEACAM5 expression on tumor cells and assess heterogeneity in cancer populations. For example:
Clone CB30 (Abcam) binds specifically to human CEACAM5, distinguishing adenocarcinomas from mesotheliomas .
Rabbit monoclonal Arcitumomab (Bio-Techne) is CyTOF-ready and compatible with complex immune profiling .
Validated for formalin-fixed, paraffin-embedded (FFPE) tissues to localize CEACAM5 in tumor biopsies. Key antibodies include:
ABIN6939072 (Antibodies-Online), optimized for detecting early gastric carcinoma foci .
Bioss BSM-61419R, a rabbit monoclonal with high specificity for human CEACAM5 .
FITC-conjugated antibodies are precursors for therapeutic ADCs (Antibody-Drug Conjugates). For instance, a single-domain antibody (B9) targeting CEACAM5 showed potent antitumor activity in preclinical models when conjugated with MMAE (monomethyl auristatin E) .
CEACAM5 antibodies bind distinct epitopes, with some recognizing:
A3-B3 domains (e.g., tusamitamab), critical for tumor-specific targeting .
N-terminal IgV-like domain (e.g., B9), involved in homophilic/heterophilic adhesion and immune modulation .
CEACAM5 interacts with CD8α and CD1d, enabling tumor immune evasion. Antibodies like B9 disrupt these interactions, enhancing NK cell cytotoxicity .
B9-MMAE ADC: Achieved IC50 values of 38.14 nM in gastric cancer (MKN-45) and 25.60 nM in pancreatic cancer (BxPC-3) models .
MN-15 Fab: Reduced tumor metastasis in colonic cancer xenografts by inhibiting adhesion and invasion .
PMC3120848: Structural and functional studies of mAb CC4 targeting CEACAM5.
Nature: Epitope mapping of tusamitamab ravtansine binding to CEACAM5.
Antibodies-Online: ABIN6939072 product specifications.
PMC10834580: UdADC B9-MMAE efficacy in CEACAM5+ solid tumors.
Abcam: CB30 antibody validation in flow cytometry.
Bioss: BSM-61419R characterization via SDS-PAGE and IF.
PMC3981948: CEACAM5-CD8α interaction and T cell modulation.
AACR: MN-15 Fab effects on metastasis in colonic cancer.
R&D Systems: FITC-conjugated CEACAM5 antibody (FAB41281R).
Acrobiosystems: Recombinant CEACAM5 protein validation.
AACR: Labetuzumab govitecan in CRPC models.
Bio-Techne: Arcitumomab FITC clone specifications.
CEACAM5 (Carcinoembryonic Antigen-Related Cell Adhesion Molecule 5) is a glycoprotein of approximately 76.8 kDa with 702 amino acid residues that localizes to the cell membrane. It's primarily expressed in columnar epithelial and goblet cells of the colon during normal development and is re-expressed in various carcinomas . CEACAM5 functions in cell adhesion processes and modulates immune responses.
The significance of CEACAM5 in cancer research stems from its differential expression across cancer types. For instance, it's been identified as a promising cell surface antigen in neuroendocrine prostate cancer (NEPC) . Interestingly, research has found that CEACAM5 inhibits lymphatic metastasis in head and neck squamous cell carcinoma (HNSCC) by reducing MDM2 expression and suppressing epithelial-mesenchymal transition . Anti-CEA positivity is detected in adenocarcinomas from numerous organs, including lung, colon, stomach, esophagus, pancreas, gallbladder, salivary gland, ovary, and endocervix .
When designing experiments with FITC-conjugated CEACAM5 antibodies, the following controls are essential for reliable interpretation:
Isotype control: Include a FITC-conjugated antibody of the identical isotype (e.g., IgG1 as identified in product specifications) but without specificity for CEACAM5 . This controls for non-specific binding due to Fc receptor interactions or other non-target binding.
Negative cell controls: Incorporate cell lines known not to express CEACAM5. According to antibody validation protocols, CEACAM5 is not found in benign glands, stroma, or malignant prostatic cells .
Positive cell controls: Include known CEACAM5-expressing cells such as colorectal, gastric, or lung cancer cell lines with confirmed expression . The antibody validation data shows that 5/5 colorectal, 2/2 gastric, and 2/2 lung cancer cell lines express CEACAM5 .
Unstained sample: Process a portion of your sample without any antibody to establish baseline autofluorescence, particularly important with FITC due to its emission spectrum.
Titration series: For quantitative applications, a titration series of the antibody will determine optimal concentration for maximum signal-to-noise ratio.
Sample preparation significantly impacts the detection of CEACAM5 using FITC-conjugated antibodies across different applications:
For flow cytometry:
Cell dissociation method affects epitope preservation. Gentle dissociation with Versene-EDTA is recommended as demonstrated in protocols detecting CEACAM5 on DU145, 22Rv1, and MSKCC EF1 cells .
Wash buffer composition influences binding efficacy. Optimal results are achieved using monoclonal antibody wash buffer (PBS + 0.1% FBS + 0.1% sodium azide) .
Incubation temperature and duration affect binding kinetics. Room temperature incubation for 30 minutes in the dark has been validated for CEACAM5 detection .
For immunohistochemistry:
Fixation method impacts epitope accessibility. The antibody described in search results is suitable for staining formalin/paraffin tissues while maintaining specificity .
Different fixatives (paraformaldehyde vs. formalin) can yield different staining patterns and intensities.
Antigen retrieval methods may be necessary to expose epitopes masked during fixation.
For immunofluorescence:
Cell permeabilization requirements depend on the epitope location. Since CEACAM5 is a membrane protein, minimal permeabilization is optimal for surface epitope detection.
Blocking protocol optimization reduces background fluorescence, particularly important with FITC due to potential tissue autofluorescence overlap.
Detecting low CEACAM5 expression levels requires methodological optimization across multiple parameters:
Sample preparation optimization:
Maximize cell viability and minimize stress during processing, as stressed cells can alter surface protein expression
Implement rigorous blocking of Fc receptors (1-2% normal serum from host species) to reduce background binding
Process samples consistently at 4°C to prevent receptor internalization
Signal amplification strategies:
Implement a two-step detection system when direct FITC conjugation provides insufficient sensitivity
Consider biotin-streptavidin systems which offer 3-4× signal amplification compared to direct conjugation
For extremely low expression, tyramide signal amplification can provide 10-50× signal enhancement
Instrument optimization:
Calibrate cytometer using standardized FITC beads covering the relevant fluorescence range
Adjust PMT voltage to position negative control population in the first decade of the log scale
Increase event collection (minimum 30,000-50,000 events) to reliably detect rare populations
Analysis refinements:
Apply fluorescence-minus-one (FMO) controls to set precise gates
Consider alternative metrics like stain index or resolution factor rather than simple MFI
Employ probability binning or frequency difference gating for subtle shifts in population distributions
Flow cytometry binding assays have been validated for detecting CEACAM5 expression across concentration ranges from 0.0128 to 200 nM, with distinct protocols for CEACAM5-positive versus negative cell lines .
Cross-reactivity with other CEACAM family members represents a significant challenge for CEACAM5 antibody specificity. Several strategies can mitigate these concerns:
Epitope-focused selection:
Choose antibodies targeting unique regions of CEACAM5. The specific antibody described in search results recognizes proteins of 80-200 kDa identified as CEA family members but doesn't react with nonspecific cross-reacting antigen (NCA) or human polymorphonuclear leucocytes .
Target the A2 domain of CEACAM5 which contains distinguishing sequences from other family members.
Validation approaches:
Perform Western blot analysis against recombinant proteins of various CEACAM family members
Test antibodies on cell panels expressing individual CEACAM family members
Validate using genetic approaches such as siRNA knockdown experiments, which have been successfully demonstrated in CEACAM5 research
Experimental controls:
Include competing soluble CEACAM5 to demonstrate binding specificity
Perform pre-absorption studies with purified antigens
Implement parallel staining with different anti-CEACAM5 antibodies targeting distinct epitopes
Cross-reactivity quantification:
Establish binding ratios against different CEACAM family members
Document cross-reactivity profiles in detailed methods sections when publishing
While some cross-reactivity may be unavoidable, documentation of the specificity profile allows for appropriate experimental design and accurate data interpretation.
Multiplex immunofluorescence incorporating FITC-conjugated CEACAM5 antibodies enables comprehensive analysis of CEACAM5 in complex tissue microenvironments:
Panel design considerations:
Account for spectral overlap between FITC (emission ~520nm) and other fluorophores
Place FITC-conjugated CEACAM5 antibody in a channel distinct from fluorophores with similar emission spectra
Include markers for relevant cell types, proliferation status, and EMT markers based on research showing CEACAM5's role in inhibiting EMT
Technical implementation:
Optimize antibody concentration and incubation conditions for each marker individually before multiplexing
Determine appropriate sequence of antibody application to prevent steric hindrance
Validate multiplex panel against single-stain controls to confirm no reduction in signal or specificity
Analysis approach:
Implement advanced image analysis tools capable of cell segmentation and phenotyping
Quantify spatial relationships between CEACAM5+ cells and other cell populations
Correlate CEACAM5 expression with markers of tumor progression or response
Validation strategy:
Confirm key findings using alternative methods (flow cytometry, Western blotting)
Compare results across multiple patient samples or experimental models
Consider tissue-specific autofluorescence quenching methods to improve signal-to-noise ratio
Multiplex immunofluorescence has been successfully applied to characterize CEACAM5 expression in metastatic tumors from lethal metastatic castration-resistant prostate cancer cases, demonstrating its utility in complex tissue analysis .
The relationship between CEACAM5 expression and cancer progression is complex and appears to be cancer-type specific, requiring sophisticated methodological approaches:
Expression patterns across cancer types:
In neuroendocrine prostate cancer (NEPC), CEACAM5 has been identified as a promising cell surface antigen, suggesting potential for targeted therapies .
In head and neck squamous cell carcinoma (HNSCC), CEACAM5 levels were significantly higher in tissue without lymph node metastasis than in tissue with lymph node metastasis, indicating a potential protective role .
According to antibody validation data, CEACAM5 is expressed in adenocarcinomas from multiple organs including lung, colon, stomach, esophagus, pancreas, and others .
Mechanistic insights:
In HNSCC, CEACAM5 inhibits epithelial-mesenchymal transition (EMT) by reducing MDM2 expression, thereby suppressing lymph node metastasis .
Cell culture experiments demonstrate that CEACAM5 inhibits proliferation, migration, and invasion while promoting apoptosis of HNSCC cells .
Mouse xenograft models confirm that CEACAM5 inhibits lymph node metastasis in vivo .
Methodological approaches:
Multiparameter analysis combining CEACAM5 with EMT markers (E-cadherin, vimentin, etc.)
Longitudinal sampling to track expression changes during disease progression
Spatial analysis techniques to correlate CEACAM5 expression with invasive fronts or metastatic foci
Functional assays including EdU proliferation, colony formation, migration, invasion, and apoptosis assays
These findings highlight the need for cancer-type specific investigation of CEACAM5 function rather than generalizing across all malignancies.
Comprehensive validation of FITC-conjugated CEACAM5 antibodies requires a multi-faceted approach:
Specificity validation:
Sensitivity validation:
Antibody titration to determine:
Minimum concentration for reliable detection
Optimal concentration for maximum signal-to-noise ratio
Dynamic range of detection
Limit of detection assessment:
Serial dilution of target-expressing cells
Spike-in experiments with known quantities of recombinant protein
Comparison with gold-standard detection methods
Performance across applications:
All validation data should be systematically documented and included in publications to ensure reproducibility.
Discrepancies in CEACAM5 detection across different methodologies are common and can be reconciled through systematic analysis:
Sources of methodological discrepancies:
Epitope accessibility variations:
Different fixation methods may mask or expose distinct epitopes
Native conformation in flow cytometry versus denatured state in Western blotting
Post-translational modifications (glycosylation of CEACAM5) affecting antibody binding
Detection sensitivity differences:
Direct FITC conjugation provides one-step detection but may offer lower sensitivity than amplified systems
Limit of detection varies significantly between methods (Western blot, flow cytometry, IHC)
Signal-to-noise ratio differences across platforms
Sample preparation effects:
Reconciliation strategies:
Method correlation studies:
Analyze the same samples using multiple techniques in parallel
Establish conversion factors between methodologies
Identify systematic biases through Bland-Altman analysis
Reference standards:
Include calibrated reference materials across methods
Use cell lines with defined CEACAM5 expression levels as internal controls
Implement standardized protocols for multi-site consistency
Biological validation:
Focus on relative changes rather than absolute values when comparing across methods
Verify the functional relevance of expression differences using appropriate assays
Consider the biological question when selecting the most appropriate methodology
Data integration:
Develop computational approaches to integrate multi-modal data
Apply normalization algorithms specific to each methodology
Implement machine learning to identify patterns across diverse data types
FITC-conjugated CEACAM5 antibodies serve as critical tools in the development pipeline for CEACAM5-targeted therapeutics:
Target validation and patient selection:
Flow cytometric quantification of CEACAM5 expression across patient samples identifies appropriate candidates for therapy
Immunofluorescence analysis of tissue specimens maps CEACAM5 distribution and accessibility
Correlation of expression levels with therapeutic response helps establish predictive biomarkers
Therapeutic mechanism assessment:
Monitoring CEACAM5 receptor occupancy during treatment
Tracking receptor internalization dynamics following therapeutic antibody binding
Measuring changes in target expression in response to treatment
Specific therapeutic applications:
Antibody-drug conjugates: The search results describe labetuzumab govitecan, a CEACAM5 antibody-drug conjugate being investigated for prostate cancer .
T-cell engagers: NILK-2301, a novel CEACAM5xCD3 T-cell engager, has been developed for CEACAM5-expressing solid tumors .
Potential CAR-T approaches targeting CEACAM5 expression
Resistance monitoring:
Detecting changes in CEACAM5 expression following treatment
Identifying alterations in epitope structure affecting therapeutic binding
Evaluating changes in internalization kinetics that might impact ADC efficacy
Flow cytometry-based binding assays have been specifically developed to assess CEACAM5-targeting therapeutics, with protocols testing concentration ranges from 0.0128 to 200 nM for binding to CEACAM5-expressing cell lines .
The discovery of CEACAM5's context-dependent roles in cancer progression has significant implications for therapeutic development:
Contrasting roles by cancer type:
In most epithelial cancers, CEACAM5 is traditionally associated with tumor progression
In head and neck squamous cell carcinoma (HNSCC), CEACAM5 inhibits lymphatic metastasis by reducing MDM2 expression and suppressing epithelial-mesenchymal transition
This dichotomy necessitates cancer-specific therapeutic strategies
Mechanistic insights informing therapy:
CEACAM5 inhibits cell proliferation and migration while promoting apoptosis in HNSCC cells
These effects were demonstrated both in vitro using HNSCC cell lines and in vivo using mouse xenograft models
Understanding the molecular mechanisms (e.g., MDM2 regulation) opens opportunities for combination therapies
Therapeutic strategy implications:
Cancer type-specific approach is crucial - targeting CEACAM5 may be beneficial in some cancers but potentially detrimental in others
Expression level assessment is essential for patient selection
Combination with EMT inhibitors might enhance efficacy in appropriate contexts
Biomarker potential:
CEACAM5 expression correlates with prognosis in HNSCC, with higher levels associated with better clinical outcomes
This prognostic value supports its use as a stratification marker in clinical trials
Longitudinal monitoring may provide insights into treatment response and resistance development
These findings highlight the importance of comprehensive biological understanding before therapeutic targeting, as CEACAM5's role appears more nuanced than previously recognized.
Evaluating CEACAM5-targeted therapies in preclinical models requires careful experimental design and methodological considerations:
Model selection criteria:
Expression profile matching: Select models with CEACAM5 expression patterns resembling the target human cancer
CEACAM5 heterogeneity representation: Incorporate models with varying expression levels to assess dose-response relationships
Species considerations: Account for potential differences between human and mouse CEACAM5 epitopes
Detection and monitoring approaches:
Longitudinal imaging: Implement non-invasive imaging to track therapy response over time
Flow cytometry: Quantify changes in CEACAM5 expression on tumor cells during treatment
Multiplex analysis: Assess CEACAM5 in relation to immune infiltration and tumor microenvironment
Efficacy parameters:
Standard endpoints: Tumor growth inhibition, survival improvement
Mechanism-specific endpoints: For HNSCC models, assess lymph node metastasis inhibition as demonstrated in previous research
Functional correlates: EMT marker modulation, cell proliferation changes, apoptosis induction
Therapeutic resistance evaluation:
Selection pressure effects: Monitor for emergence of CEACAM5-negative subpopulations
Compensatory pathway activation: Assess parallel signaling pathway upregulation
Epitope modulation: Test for changes in antibody binding patterns following treatment
Research demonstrates that CEACAM5 inhibits epithelial-mesenchymal transition in HNSCC by reducing MDM2 expression , suggesting that monitoring EMT markers and MDM2 levels could provide mechanistic insights into therapeutic efficacy.
Single-cell technologies offer unprecedented resolution for investigating CEACAM5 biology and optimizing antibody-based detection:
Single-cell expression profiling:
Reveals heterogeneity in CEACAM5 expression that might be masked in bulk analysis
Identifies cell state-specific expression patterns
Correlates CEACAM5 with other molecular markers at single-cell resolution
Spatial transcriptomics integration:
Maps CEACAM5 expression in spatial context within tissues
Correlates protein expression (by immunofluorescence) with mRNA levels
Identifies microenvironmental factors influencing CEACAM5 expression
Antibody validation enhancements:
Single-cell proteo-genomics confirms specificity at individual cell level
Flow sorting of CEACAM5+ vs CEACAM5- populations for downstream validation
Precise quantification of antibody binding kinetics at cellular level
Therapeutic implications:
Identification of CEACAM5 expression in rare cell populations
Tracking clonal evolution of CEACAM5 expression during therapy
Characterizing resistant populations with altered CEACAM5 biology
These approaches could particularly enhance understanding of the dual role of CEACAM5 in cancer progression, potentially reconciling the contrast between its role in most epithelial cancers and its metastasis-inhibiting function in HNSCC .
Several innovative approaches can enhance FITC-conjugated antibody performance for challenging CEACAM5 detection scenarios:
Conjugation chemistry optimization:
Site-specific conjugation rather than random labeling improves binding consistency
Optimized fluorophore-to-protein ratio balances signal intensity with potential steric hindrance
Linker chemistry selection affecting stability and performance in different microenvironments
Signal enhancement technologies:
Proximity-based amplification systems (e.g., RNAscope) for detecting low-abundance targets
Photostable FITC derivatives reducing photobleaching during extended imaging
Nanobody-based detection systems offering improved tissue penetration and reduced background
Sample treatment innovations:
Adaptive epitope retrieval protocols customized to preserve CEACAM5 conformation
Background reduction agents specifically addressing tissue autofluorescence in the FITC spectrum
Clearing techniques for enhanced imaging depth in tissue specimens
Data acquisition and analysis:
Advanced spectral unmixing algorithms to resolve FITC signal from autofluorescence
Artificial intelligence-based image analysis improving detection sensitivity
Standardized acquisition parameters for cross-laboratory reproducibility
These approaches would be particularly valuable for challenging applications such as detecting the subtle variations in CEACAM5 expression that correlate with lymph node metastasis status in HNSCC, where higher CEACAM5 levels are associated with reduced metastasis .
Structural biology and epitope mapping technologies are poised to revolutionize CEACAM5 antibody development:
High-resolution structural insights:
Cryo-EM structures of CEACAM5 in different conformational states guide rational antibody design
Mapping of functional domains enables targeting of specific CEACAM5 activities
Identification of conserved versus variable regions informs cross-reactivity management
Epitope-specific optimization:
Epitope mapping of the A2 domain of CEACAM5 (an important antibody target ) reveals optimal binding sites
Function-blocking antibodies targeting specific interaction interfaces
Internalization-promoting antibodies for enhanced ADC delivery
Computational antibody design:
In silico modeling to predict antibody-antigen interactions
Structure-based optimization of binding affinity and specificity
Paratope engineering to minimize cross-reactivity with other CEACAM family members
Translation to novel formats: