CUSABIO developed the CLDN3 recombinant monoclonal antibody using a multi-step process. Initially, B cells were isolated from the spleen of the immunized animal, with the recombinant human CLDN3 protein employed as the immunogen during immunization. Subsequently, RNA was extracted from the B cells and converted into cDNA through reverse transcription. The gene encoding the CLDN3 antibody was extended using a degenerate primer through PCR and inserted into the vector. The recombinant vector was introduced into host cells via transfection, facilitating antibody expression. The CLDN3 recombinant monoclonal antibodies were obtained from the cell culture supernatant and purified using affinity chromatography. To confirm specificity and functionality, this antibody was validated through ELISA, demonstrating its ability to react with human CLDN3 protein.
CUSABIO developed the CLDN3 recombinant monoclonal antibody using a meticulous multi-step process. Initially, B cells were isolated from the spleen of the immunized animal, utilizing recombinant human CLDN3 protein as the immunogen during immunization. Subsequently, RNA was extracted from the B cells and converted into cDNA through reverse transcription. The gene encoding the CLDN3 antibody was amplified using a degenerate primer via PCR and inserted into the vector. The recombinant vector was introduced into host cells through transfection, enabling antibody expression. The CLDN3 recombinant monoclonal antibodies were harvested from the cell culture supernatant and purified using affinity chromatography. To confirm specificity and functionality, this antibody was rigorously validated through ELISA, demonstrating its reactivity with human CLDN3 protein.
CLDN3 plays a critical role in tight junction-specific obliteration of the intercellular space, facilitated by its calcium-independent cell-adhesion activity.
CLDN3 is a tight-junction protein with four-transmembrane domains coded by the intron-less CLDN3 gene on chromosome 7q11.23. In normal tissues, tight junctions form continuous seals around cells, acting as physical barriers that regulate paracellular transport of water and molecules. During tumorigenesis, CLDN3 becomes externally exposed, making it an attractive target for cancer diagnosis and therapy . Despite its widespread expression in normal cells, CLDN3 is considered a viable drug target because tight junction proteins in normal tissues have limited accessibility due to orchestrated cell growth patterns, whereas cancer cells with disrupted tissue architecture may have higher exposure to targeting agents .
Developing antibodies against CLDN3 presents several technical challenges:
Four-transmembrane domain structure makes it difficult to mimic using recombinant proteins
High sequence homology among claudin family members creates specificity issues
Short extracellular loops exhibit low immunogenicity
High homology between human and mouse claudins complicates animal model development
Researchers have overcome these challenges using advanced techniques such as scFv phage display with CLDN3-overexpressing stable cells and CLDN3-embedded lipoparticles as antigens, which better preserve the protein's native conformation .
Thorough validation of CLDN3 antibody specificity should include:
Flow cytometry analysis using cells expressing CLDN3 versus negative controls
Western blotting to confirm molecular weight and expression levels
Cross-reactivity testing against other claudin family members (especially CLDN1, CLDN4, and CLDN5)
Immunofluorescence to confirm membrane localization
Positive and negative tissue controls (e.g., liver bile ducts as positive control, stomach epithelial cells as predominantly negative)
For accurate validation, establish stable CLDN transfectants for each claudin family member to confirm antibody specificity through comparative binding studies .
Based on the research data, several cell models are suitable for CLDN3 antibody investigations:
Cell Type | CLDN3 Status | Recommended Applications |
---|---|---|
Caov-3 | Naturally expressing | Immunofluorescence, functional studies |
OVCAR-3 | Naturally expressing | Immunofluorescence, functional studies |
TOV-112D | Negative control | Background assessment |
hCLDN3/TOV-112D | Stable transfectant | Antibody validation, binding kinetics |
hCLDN3/HEK293 | Stable transfectant | Affinity measurements, binding kinetics |
mCLDN3/HEK293 | Stable transfectant | Cross-species reactivity testing |
Culturing conditions vary by cell type; epithelial cancer cells typically require DMEM supplemented with 10% FBS, 100 units/mL penicillin, and 100 μg/mL streptomycin, while specialized cell lines may need specific media formulations .
Determining binding kinetics of anti-CLDN3 antibodies requires specialized approaches:
Use real-time cell-based affinity measurement systems (e.g., LigandTracer)
Label antibodies with fluorescent dyes (e.g., DyLight 488) without compromising binding capacity
Establish proper positive and negative cell lines (e.g., CLDN3-transfected versus parental cells)
Perform stepwise concentration increases (e.g., 3nM and 9nM for human CLDN3, 30nM and 90nM for mouse CLDN3)
Monitor association until equilibrium at each concentration
Measure dissociation by replacing medium and monitoring fluorescence decay
Analyze binding curves using appropriate software (e.g., TraceDrawer)
This methodology allows precise determination of association and dissociation rate constants, providing sub-nanomolar affinity measurements for high-quality antibodies like h4G3 .
Cross-reactivity between claudin family members presents a significant challenge due to high sequence homology. Effective strategies include:
Generate chimeric ECL fusion proteins between different claudins (e.g., CLDN1/CLDN3) to map epitope specificity
Establish a panel of HEK293 cells expressing different claudin family members (CLDN1, 3, 4, 5, 6, 8, 9, and 17)
Employ comparative flow cytometry to quantitatively assess binding to various claudins
Conduct competitive binding assays with known claudin binders (e.g., C-CPE)
Perform detailed epitope mapping through alanine scanning mutagenesis of extracellular loops
These approaches allow researchers to identify antibodies with genuine specificity for CLDN3 without cross-reactivity to other family members, a critical factor for therapeutic and diagnostic applications .
Evaluating ADCC activity of anti-CLDN3 antibodies requires:
Select appropriate target cells with varying CLDN3 expression levels
Use suitable effector cells (e.g., NK-92MI cells)
Establish optimal effector-to-target (E:T) ratios through titration experiments
Quantify cytotoxicity using methods like lactate dehydrogenase (LDH) release assays
Include proper controls:
Isotype control antibodies
CLDN3-negative cell lines
FcγR blocking experiments to confirm mechanism
Correlate ADCC activity with CLDN3 expression levels through flow cytometry quantification
Research indicates that high-quality anti-CLDN3 antibodies can demonstrate ADCC according to CLDN3 expression levels in various cancer cells through activation of FcγRIIIa (CD16a) .
For successful in vivo biodistribution studies of anti-CLDN3 antibodies:
Use fluorescence-conjugated antibodies (optimize dye-to-antibody ratio to prevent function loss)
Establish xenograft models with both CLDN3-positive and CLDN3-negative tumors
Consider tumor size and vascularization variables
Calculate optimal antibody dose based on body weight and pharmacokinetic properties
Determine optimal imaging timepoints (typically 24-72 hours post-injection)
Perform ex vivo validation of specific tumor localization
Quantify biodistribution in major organs to assess off-target accumulation
Compare with non-specific control antibodies of the same isotype
Research with fluorescence-conjugated h4G3 demonstrated that anti-CLDN3 antibodies can successfully localize to tumor sites in xenograft mice bearing CLDN3-expressing tumors, supporting their potential diagnostic and therapeutic applications .
Different antibody formats exhibit distinct advantages and limitations for CLDN3 targeting:
Format | Molecular Weight | Tissue Penetration | Half-life | ADCC Potential | Applications |
---|---|---|---|---|---|
IgG | 150 kDa | Moderate | Long (days) | High | Imaging, therapeutic |
Fab | 50 kDa | Good | Short (hours) | None | Imaging, blocking |
scFv | 25-30 kDa | Excellent | Very short | None | CAR-T, bispecifics |
For targeting claudins in solid tumors, researchers should consider:
Using scFv or Fab fragments for enhanced tumor penetration
Employing full IgG when effector functions (ADCC) are desirable
Adapting formats based on specific application needs (diagnostics vs. therapeutics)
Evaluating format-specific binding kinetics, as different formats may demonstrate altered affinity
Proper controls for immunohistochemistry with anti-CLDN3 antibodies should include:
Positive tissue controls: Liver sections where strong membranous CLDN3 staining should be observed in bile ducts, with weak to moderate membranous staining in hepatocytes (often predominantly at the bile secreting pole)
Negative tissue controls: Stomach sections where most epithelial cells should be CLDN3 negative, while a small subset of epithelial cells in the neck and glandular pits should show strong membranous CLDN3 staining
Technical controls:
Isotype-matched irrelevant antibody
Primary antibody omission
Antigen competition (pre-absorption)
CLDN3 knockout or knockdown tissues/cells
These controls help distinguish specific staining from background and confirm the validity of immunohistochemical findings with anti-CLDN3 antibodies .
Detecting CLDN3 in tight junctions requires specialized techniques:
Sample preparation:
Use fresh tissues or cells grown to confluence to maintain intact tight junctions
Consider different fixation methods (4% paraformaldehyde is often optimal)
Test permeabilization conditions carefully (excessive permeabilization may disrupt tight junction structure)
Staining optimization:
Use antibodies recognizing native conformations of CLDN3
Employ 1:100–1:200 dilutions for immunohistochemistry applications
Include membrane markers to demonstrate co-localization
Use confocal microscopy for detailed junction visualization
Validation approaches:
These optimizations help ensure specific detection of CLDN3 in its native tight junction context rather than in other cellular compartments where it might be mislocalized.
CLDN3 antibodies offer several valuable approaches for cancer diagnostics:
Tissue-based diagnostics:
Immunohistochemical assessment of CLDN3 overexpression
Evaluation of subcellular localization (membrane vs. cytoplasmic)
Cancer subtyping based on CLDN3 expression patterns
Liquid biopsy applications:
Detection of CLDN3-expressing circulating tumor cells
Identification of CLDN3-positive extracellular vesicles
Monitoring treatment response through sequential sampling
Molecular imaging:
Fluorescence-labeled antibodies for intraoperative guidance
PET/SPECT imaging with radiolabeled antibodies
Assessment of target expression before therapeutic intervention
Research indicates that high-quality CLDN3 antibodies can specifically localize to tumor sites in vivo, supporting their potential as diagnostic tools .
CLDN3 antibodies show promising characteristics for CAR-T development:
Antibody selection considerations:
High-affinity scFv fragments are ideal CAR components
Specificity for CLDN3 without cross-reactivity to other claudins is essential
Epitope accessibility in tumor contexts must be confirmed
CAR design factors:
Incorporate anti-CLDN3 scFvs with optimal spacer length
Select appropriate costimulatory domains (CD28, 4-1BB)
Consider dual-targeting approaches to enhance specificity
Preclinical testing requirements:
Validate CAR-T activity against cell lines with varying CLDN3 expression
Assess potential on-target/off-tumor toxicity against normal CLDN3-expressing tissues
Evaluate efficacy in xenograft models of CLDN3-expressing tumors
The specificity of antibodies like h4G3 makes them suitable candidates for CAR development targeting CLDN3-expressing tumors across multiple cancer types .
Generating stable CLDN3-expressing cell lines presents unique challenges that researchers can address through:
Vector selection:
Use pcDNA3.1(+) or similar expression vectors with strong promoters
Consider lentiviral systems for hard-to-transfect cell lines
Include selection markers (e.g., G418 resistance)
Transfection optimization:
Compare multiple transfection reagents (e.g., FuGENE HD)
Optimize cell density and DNA:reagent ratios
Allow sufficient recovery time before selection
Clonal selection strategy:
Isolate individual colonies using clonal cylinders
Screen multiple clones for consistent expression levels
Validate membrane localization of CLDN3
Expression maintenance:
These approaches help establish reliable cell models for antibody characterization and functional studies.
Distinguishing between claudin-3 and claudin-4 binding is particularly challenging due to their structural similarities but can be achieved through:
Comparative binding studies:
Test antibody binding to cells expressing only CLDN3 or only CLDN4
Perform dose-response studies to identify differential affinities
Use competition assays with known claudin-specific binders
Domain swapping experiments:
Create chimeric constructs with swapped extracellular loops between CLDN3 and CLDN4
Express chimeras in claudin-negative cell lines
Map binding epitopes through differential recognition patterns
Specialized binding assays:
These approaches can confirm whether an antibody like h4G3 truly recognizes CLDN3 specifically without cross-reactivity to CLDN4, an important distinction given that both are potential cancer targets .