CDH3 (Cadherin-3), also known as P-cadherin, is a calcium-dependent cell-cell adhesion glycoprotein that plays crucial roles in cell adhesion, proliferation, and invasion processes. It has emerged as a significant research target because it's overexpressed in multiple cancer types including lung, breast, colorectal, pancreatic, head and neck, ovarian malignancies, and glioblastoma, while showing negligible expression in most normal tissues .
Research has demonstrated that CDH3 overexpression correlates with cancer aggressiveness, invasiveness, and poor patient prognosis . In glioblastoma, for example, CDH3 affects distinct cancer hallmarks in vitro and is related to increased tumor growth and shorter survival in vivo . Recent studies have identified CDH3 as a promising therapeutic target due to its selective expression pattern and involvement in oncogenic signaling pathways.
CDH3 antibodies are employed across multiple laboratory techniques, with application specificity varying by clone and manufacturer. Common applications include:
| Application | Common Dilutions | Research Purpose |
|---|---|---|
| Western Blot (WB) | 0.04-0.4 μg/mL | Protein expression quantification |
| Immunohistochemistry (IHC) | 1:200-1:500 | Tissue expression pattern analysis |
| Immunocytochemistry (ICC) | Varies by antibody | Cellular localization studies |
| Flow Cytometry (FCM) | Typically 1-10 μg per test | Quantification of cell surface expression |
| ELISA | Varies by kit | Protein quantification in solution |
| Immunofluorescence (IF) | Typically 1:100-1:500 | Subcellular localization visualization |
For example, in IHC applications, CDH3 antibodies have been used to detect CDH3 in paraffin-embedded sections of human placenta tissues using heat-mediated antigen retrieval in citrate buffer (pH6) for 20 minutes . For ICC applications, CDH3 antibodies have been employed to detect the protein in A549 cells using enzyme antigen retrieval methods .
Proper storage and handling of CDH3 antibodies are critical for maintaining their specificity and sensitivity. Based on manufacturer recommendations:
Store lyophilized antibodies at -20°C for up to one year from date of receipt
After reconstitution, store at 4°C for one month or aliquot and store at -20°C for up to six months
Avoid repeated freeze-thaw cycles which can significantly decrease antibody activity
For long-term storage of reconstituted antibodies, glycerol addition (final concentration 15-50%) can help preserve activity
Working dilutions should be prepared fresh before use for optimal results
Follow specific manufacturer guidelines for each antibody clone, as storage conditions may vary slightly between products
Improper handling leading to denaturation or degradation is a common cause of experiment failure when working with antibodies.
Validating CDH3 antibody specificity is crucial for ensuring experimental reproducibility and accuracy. A comprehensive validation approach should include:
Positive and negative controls: Use cell lines or tissues with known CDH3 expression status. For example, studies have used A549 cells transfected with human or mouse CDH3 as positive controls and non-transfected A549 cells (which don't express CDH3) as negative controls .
Genetic validation:
Use CRISPR/Cas9-engineered cell lines with CDH3 knockout
Compare siRNA-mediated CDH3 knockdown with control cells
Test in CDH3-overexpressing models versus parental cell lines
Cross-reactivity testing: Validate specificity against related cadherins (CDH1/E-cadherin, CDH2/N-cadherin) to ensure the antibody doesn't cross-react with these structurally similar proteins .
Multiple detection methods: Confirm expression using at least two independent methods (e.g., WB and IHC, or PCR and protein detection) .
Epitope mapping: Understanding which region of CDH3 the antibody binds to can help predict potential cross-reactivity and application suitability. For example, some CDH3 antibodies target the extracellular domain while others target intracellular regions .
One published validation method used flow cytometry to examine the specificity of a CDH3 antibody, demonstrating a high correlation between mean fluorescence intensity values and expression levels of FLAG-tagged CDH3/P-cadherin in transformed cell lines .
Optimized IHC protocols for CDH3 detection vary by tissue type, fixation method, and antibody clone. Based on published methodologies:
For formalin-fixed paraffin-embedded (FFPE) tissues:
Antigen retrieval options:
Blocking and antibody incubation:
Tissue-specific considerations:
For challenging tissues, a published protocol used enzyme antigen retrieval reagent for 15 minutes followed by blocking with 10% goat serum and overnight incubation with 1μg/ml anti-CDH3 antibody at 4°C, which yielded clear membrane staining in A549 cells .
Robust experimental design requires appropriate controls to validate findings and troubleshoot potential issues:
Essential controls for CDH3 antibody experiments:
Positive tissue/cell controls:
Negative tissue/cell controls:
Technical controls:
Primary antibody omission control
Secondary antibody-only control
Blocking peptide competition assay using the immunizing peptide (if available)
Decreasing antibody concentration gradient to determine optimal signal-to-noise ratio
Validation controls:
siRNA/shRNA knockdown versus scrambled control
CRISPR knockout versus wild-type cells
Parallel detection with a second CDH3 antibody recognizing a different epitope
For example, in published radioimmunotherapy studies, researchers confirmed specific binding of anti-CDH3 antibody by comparing tumor uptake in CDH3-positive tumors (EBC1, H1373, SW948) versus CDH3-negative tumors (A549, RKO), demonstrating significantly higher accumulation in CDH3-positive models .
CDH3 antibodies have emerged as promising cancer therapeutic agents through several development strategies:
Unconjugated antibodies with direct therapeutic effects:
PF-03732010, a fully human monoclonal antibody against P-cadherin, inhibits P-cadherin–mediated cell adhesion and aggregation in vitro
Demonstrated significant inhibition of tumor growth and metastatic progression in multiple CDH3-overexpressing tumor models, including MDA-MB-231-CDH3, 4T1-CDH3, HCT116, H1650, PC3M-CDH3, and DU145
Mechanistically suppresses P-cadherin levels, causes degradation of membrane β-catenin, and concurrently suppresses cytoplasmic vimentin, resulting in diminished metastatic capacity
Antibody-drug conjugates (ADCs):
BC3195, an ADC targeting CDH3 with monomethyl auristatin E (MMAE) payload, is in clinical development
Phase I clinical trial results show manageable safety profile with common adverse events including increased AST (55.6%), increased conjugated bilirubin (55.6%), and hypoalbuminemia (55.6%)
Preliminary antitumor activity observed with 50% of evaluable patients showing stable disease, including target lesion reduction in some cases
Radioimmunotherapy approaches:
Yttrium-90 (90Y)-labeled anti-CDH3 mouse monoclonal antibody (MAb-6) demonstrated significant tumor suppression in CDH3-expressing cancer models
Single intravenous injection of 90Y-MAb-6 (100 μCi) significantly suppressed tumor growth in mice with CDH3-positive tumors
Two injections led to complete tumor regression in H1373-inoculated mice without detectable toxicity
Bispecific antibody strategies:
TR2/CDH3 BAB, a human bispecific antibody that binds both CDH3 and TRAILR2, achieves TRAILR2 hyperclustering to induce apoptosis selectively in CDH3-expressing tumor cells
Demonstrated target-dependent anti-tumor activity in CRISPR/Cas9-engineered models
Employs an engineered human IgG1 Fc backbone with L234A/L235A mutations to avoid CDH3-independent crosslinking
These therapeutic approaches leverage CDH3's selective expression in cancer cells while minimizing off-target effects in normal tissues.
Accurate quantification of CDH3 expression is essential for patient selection in targeted therapy development. Multiple complementary approaches provide comprehensive assessment:
Immunohistochemistry (IHC) with digital pathology:
Gold standard for clinical biomarker assessment
Membrane staining intensity typically scored on 0-3+ scale (0=negative, 1+=weak, 2+=moderate, 3+=strong)
H-score calculation: (% cells 1+ × 1) + (% cells 2+ × 2) + (% cells 3+ × 3), range 0-300
Digital pathology platforms enable objective quantification of membrane staining intensity and percentage positive cells
Requires standardized protocols with positive and negative controls to ensure reproducibility across laboratories
Quantitative RT-PCR:
Enables precise quantification of CDH3 mRNA levels
Published protocols use TaqMan probes specific for CDH3 with TBP (TATA Box Binding Protein) as reference gene
PCR conditions: 2 min at 50°C, 20 s at 95°C, followed by 40 cycles of 3 s at 95°C and 30 s at 60°C
Results typically expressed as fold-change relative to control or as normalized expression units
Proteomics approaches:
Mass spectrometry-based quantification of CDH3 protein expression
Provides absolute quantification independent of antibody affinity variations
Can simultaneously measure related proteins in cadherin pathways
More resource-intensive but offers higher specificity and broader pathway analysis
Flow cytometry for circulating tumor cells or fresh tissue samples:
Enables single-cell analysis of CDH3 surface expression
Particularly useful for hematological samples or disaggregated solid tumors
Provides quantifiable data as mean fluorescence intensity (MFI)
Allows for multiparameter analysis with other cancer biomarkers
For clinical development of CDH3-targeted therapies, a combination of IHC and mRNA assessment has been most commonly employed, with positivity thresholds defined based on correlation with treatment response in early-phase clinical trials .
CDH3 antibody clones exhibit significant variations in binding properties and functional effects, which impact their research and therapeutic applications:
Functional differences between antibody clones arise from:
Epitope specificity: Antibodies targeting different domains of CDH3 (extracellular, transmembrane, or cytoplasmic) exhibit varied functional effects. For example, antibodies targeting the extracellular domain may disrupt cell-cell adhesion, while others may primarily serve detection purposes .
Affinity variations: Higher-affinity antibodies generally demonstrate superior sensitivity in detection applications but may not necessarily have superior functional effects in therapeutic applications.
Isotype differences: The antibody isotype influences Fc-mediated functions such as complement activation and immune cell recruitment, which may contribute to therapeutic efficacy beyond target binding.
Cross-reactivity profiles: Some antibodies cross-react with mouse CDH3, enabling preclinical studies in immunocompetent models, while others are strictly human-specific .
When selecting a CDH3 antibody clone, researchers should consider the specific application requirements and whether functional modulation or mere detection is the primary goal.
Researchers frequently encounter several technical challenges when working with CDH3 antibodies:
Epitope masking in fixed tissues:
Challenge: Formalin fixation can mask CDH3 epitopes, reducing antibody accessibility
Solution: Optimize antigen retrieval methods; compare heat-induced (citrate pH 6.0 or EDTA pH 9.0) and enzymatic retrieval approaches. In published protocols, heat-mediated antigen retrieval in citrate buffer (pH6) for 20 minutes has proven effective for CDH3 detection in placental tissues .
Membrane localization preservation:
Specificity concerns:
Challenge: Cross-reactivity with other cadherin family members (particularly CDH1/E-cadherin)
Solution: Use antibodies targeting unique CDH3 regions; perform validation in models with genetic manipulation of CDH3; include appropriate controls. The CDH3-building block of TR2/CDH3 BAB was derived from an immunization campaign using AlivaMab® transgenic mice and subsequently engineered to remove chemical liabilities and non-human residues .
Signal amplification for low-expressing samples:
Challenge: Detecting CDH3 in samples with low expression levels
Solution: Employ signal amplification systems such as tyramide signal amplification or polymer-based detection systems; optimize primary antibody concentration and incubation time.
Batch variability:
Challenge: Inconsistent results between antibody lots
Solution: Purchase larger quantities of validated lots; perform lot-to-lot validation; consider monoclonal antibodies for greater consistency.
Species cross-reactivity limitations:
Challenge: Many CDH3 antibodies have limited cross-reactivity across species
Solution: Verify species reactivity before purchase; for cross-species studies, select antibodies targeting conserved epitopes or use species-specific antibodies. The MAb-6 antibody has demonstrated cross-reactivity with mouse CDH3/P-cadherin, making it suitable for preclinical studies in mouse models .
Background in specific tissues:
Challenge: Non-specific binding in certain tissues (particularly liver)
Solution: Include additional blocking steps with avidin/biotin blocking for biotin-based detection systems; use Fc receptor blocking reagents; optimize antibody dilution through titration experiments.
Implementing these solutions based on specific experimental contexts will significantly improve CDH3 antibody performance across applications.
Optimizing CDH3 antibody protocols for challenging contexts requires systematic approach modifications:
For highly fibrotic tissues (pancreatic cancer, breast cancer):
Extend antigen retrieval time to 30-40 minutes
Consider dual antigen retrieval: protease treatment followed by heat-induced retrieval
Section tissues at 3-4μm (thinner than standard) to improve antibody penetration
Use automated staining platforms for consistent results across samples
Employ polymer-based detection systems for higher sensitivity with lower background
For circulating tumor cell detection:
Minimize processing time to preserve cell surface antigens
Use gentle fixation (2% paraformaldehyde for 10-15 minutes)
Block Fc receptors on leukocytes to reduce non-specific binding
Combine with epithelial markers (EpCAM) and exclude leukocyte markers (CD45)
Consider fluorescence-based detection for multiplexed analysis
For multiplex immunofluorescence:
Test for antibody cross-reactivity with other targets in multiplex panel
Optimize stripping/quenching protocols between rounds if using sequential staining
Consider tyramide signal amplification for weaker signals
Use spectral unmixing to address autofluorescence in certain tissues
Include single-stain controls for each antibody to establish proper compensation
For quantitative Western blot:
Optimize protein extraction for membrane proteins (consider NP-40 or Triton X-100-based lysis buffers)
Avoid boiling samples to prevent aggregation of transmembrane proteins
Use graduated loading controls to verify linear detection range
Consider transfer conditions optimized for high-molecular-weight proteins (91.4 kDa)
Use fluorescence-based detection systems for more accurate quantification
For brain tissue analysis:
Optimize fixation time (24-48 hours maximum)
Perform antigen retrieval in Tris-EDTA buffer pH 9.0
Include Sudan Black treatment to reduce lipofuscin autofluorescence
Use confocal microscopy to better distinguish membrane staining patterns
Consider fresh frozen tissue for certain applications
One published optimization approach for ICC used enzyme antigen retrieval for 15 minutes with CDH3 antibody, followed by detection using Strepavidin-Biotin-Complex with DAB as the chromogen, which provided excellent visualization of CDH3 in A549 cells .
Discrepancies between CDH3 mRNA and protein expression are common and have important implications for research interpretation:
Common causes of discrepancies:
Post-transcriptional regulation: miRNAs can target CDH3 mRNA for degradation or translational inhibition
Protein stability differences: CDH3 protein half-life may vary between cell types or conditions
Technical limitations: Differences in sensitivity between mRNA detection methods (qPCR, RNA-seq) and protein detection methods (IHC, Western blot)
Antibody epitope accessibility: Protein conformation or interactions may mask antibody epitopes in certain contexts
Sample heterogeneity: Bulk analysis may mask cell-type specific expression patterns
Systematic approach to resolving discrepancies:
Validation with multiple methods: Confirm protein expression using different antibody clones targeting distinct epitopes
Single-cell analysis: Employ single-cell RNA-seq paired with flow cytometry or single-cell proteomics
Temporal analysis: Assess whether differences reflect time-dependent regulation
Functional validation: Use genetic manipulation (knockdown/overexpression) to confirm specificity
Subcellular localization: Determine if protein is sequestered in specific cellular compartments
Interpretation guidelines:
Protein expression is generally more relevant for functional studies and therapeutic targeting
mRNA expression may predict future protein expression or reflect recent transcriptional activity
In patient stratification, combined mRNA and protein assessment provides more robust classification
For antibody-based therapeutics, focus on protein expression and membrane localization
Case study examples:
In glioblastoma research, CDH3 mRNA levels from TCGA database showed increased expression in high-grade gliomas, but protein confirmation was necessary to validate CDH3 as a therapeutic target
In pancreatic cancer, plasma membrane expression of CDH3 increases during carcinogenesis from benign pancreatic intraepithelial neoplasia 1 (PanIN-1) to malignant PDAC, highlighting the importance of assessing protein localization rather than just total levels
Researchers should ideally employ complementary approaches and consider the biological context when interpreting discrepancies between CDH3 mRNA and protein data.
CDH3 antibodies are being integrated into innovative combination therapeutic strategies that exploit synergistic mechanisms:
Combinations with conventional chemotherapies:
Preclinical studies have investigated CDH3-targeted therapies with standard-of-care cytotoxic drugs
TR2/CDH3 bispecific antibody has been evaluated in combination with indication-relevant cytotoxic drugs for pancreatic cancer
These combinations may enhance tumor penetration of chemotherapy through modulation of cell-cell adhesion
Integration with immune checkpoint inhibitors:
CDH3-targeting may increase tumor immunogenicity by disrupting cell-cell contacts
Combination strategies with anti-PD-1/PD-L1 antibodies are being explored
Preliminary evidence suggests CDH3 inhibition may reduce immune-suppressive signals in the tumor microenvironment
Antibody-drug conjugates with rationally selected payloads:
Combination radiotherapy approaches:
Bispecific and multispecific platforms:
TR2/CDH3 BAB bispecific antibody exploits dual targeting of CDH3 and TRAILR2 to achieve selective apoptosis induction in cancer cells
The engineered antibody uses silenced Fc domains (L234A/L235A mutations) to prevent CDH3-independent crosslinking
This approach extends the therapeutic concept previously applied with CDH17 to additional cancer indications with high medical need
These combination approaches aim to overcome resistance mechanisms, enhance efficacy, and broaden the therapeutic window for CDH3-targeted interventions.
CDH3 antibodies serve as crucial tools for elucidating P-cadherin biology across developmental, physiological, and pathological contexts:
Developmental biology insights:
CDH3 antibodies have helped map expression patterns during embryonic development
Used to identify CDH3's role in placental morphogenesis and skin development
Enabled tracking of cell-cell adhesion dynamics during tissue formation
Lineage tracing and cell fate determination:
P-cadherin expression marks specific epithelial progenitor populations
CDH3 antibodies facilitate isolation and characterization of these populations
Studies using CDH3 antibodies have revealed distinct stem/progenitor cell niches in various tissues
Mechanistic studies in cancer progression:
CDH3 knockdown and overexpression GBM cell models revealed P-cadherin's oncogenic functions in affecting cell viability, cell cycle, invasion, migration, and neurosphere formation capacity
Genes positively correlated with CDH3 were found enriched for oncogenic pathways commonly activated in GBM
CDH3 antibodies helped demonstrate that GBM cells expressing high P-cadherin levels generate larger subcutaneous tumors and cause shorter survival in orthotopic models
Epithelial-mesenchymal transition (EMT) research:
CDH3 antibodies used to monitor cadherin switching during EMT
PF-03732010 suppressed P-cadherin levels, caused degradation of membrane β-catenin, and concurrently suppressed cytoplasmic vimentin, revealing potential mechanisms of its anti-metastatic effects
Immunofluorescence with CDH3 antibodies helped characterize these molecular changes during the transition
Structure-function relationship exploration:
Domain-specific CDH3 antibodies help dissect the roles of different protein regions
Antibodies targeting the extracellular domain versus cytoplasmic domain reveal distinct functional effects
Epitope mapping with various antibody clones contributes to understanding critical functional domains
Signaling pathway interrogation:
CDH3 antibody-based proximity ligation assays identify interaction partners
Immunoprecipitation with CDH3 antibodies enables identification of associated signaling complexes
Phospho-specific CDH3 antibodies detect activation states and regulatory mechanisms
Despite significant progress, CDH3 antibody research faces several limitations that present opportunities for future innovation:
Current technical limitations:
Limited availability of antibodies distinguishing between different CDH3 isoforms or post-translational modifications
Challenges in developing antibodies that specifically disrupt CDH3-mediated interactions with select binding partners
Insufficient characterization of epitope specificity for many commercially available clones
Restricted tools for real-time monitoring of CDH3 dynamics in living systems
Biological knowledge gaps:
Incomplete understanding of CDH3's role in different cancer subtypes and progression stages
Limited insight into resistance mechanisms to CDH3-targeted therapies
Poorly characterized feedback mechanisms regulating CDH3 expression following therapeutic targeting
Unclear implications of heterogeneous CDH3 expression within tumors
Future technical innovations:
Development of conformation-specific antibodies detecting active versus inactive CDH3 states
Creation of bispecific platforms targeting CDH3 together with complementary tumor-associated antigens
Engineering of antibody fragments (Fabs, scFvs) for improved tissue penetration
Generation of humanized or fully human antibodies with reduced immunogenicity for therapeutic applications
Design of intrabodies for targeting intracellular pools of CDH3
Emerging research directions:
Exploration of CDH3's role in cancer stem cell maintenance and therapy resistance
Development of CDH3 antibody-based liquid biopsy approaches for minimally invasive disease monitoring
Investigation of CDH3-targeted radiotheranostic applications combining imaging and therapy
Creation of CDH3 CAR-T cell therapies for solid tumors
Characterization of CDH3's role in immune evasion and potential for enhancing immunotherapy response
Translational challenges to address:
Determination of optimal patient selection biomarkers beyond mere CDH3 expression
Development of companion diagnostics with standardized scoring systems
Establishment of resistance mechanisms and strategies to overcome them
Optimization of dosing schedules and combination regimens
Exploration of potential synergies with emerging therapeutic modalities
The field is progressing toward more sophisticated understanding of CDH3 biology and therapeutic exploitation, with several clinical-stage assets including BC3195 (ADC) currently in Phase I trials and established preclinical evidence supporting various therapeutic modalities including radioimmunotherapy and bispecific antibody approaches .