CLPX3 Antibody

Shipped with Ice Packs
In Stock

Description

Biological Significance of Claudin-3 (CLDN3)

Claudin-3 is a tetraspanin tight junction protein overexpressed in epithelial cancers, including ovarian, prostate, pancreatic, and breast cancers . Its extracellular loops (ECL1 and ECL2) become exposed during tumorigenesis, making it a viable target for antibody-based therapies .

Key Features of CLDN3Description
Protein Structure220 amino acids; four transmembrane domains; two extracellular loops (ECL1/ECL2)
Role in CancerCorrelates with poor prognosis, metastasis, and chemoresistance
ExpressionElevated in 60–80% of epithelial tumors

Human Monoclonal Antibody h4G3

Developed via scFv phage display, h4G3 is a human IgG1 antibody with:

  • Sub-nanomolar affinity for CLDN3 (KD < 1 nM)

  • Cross-species reactivity: Binds human and murine CLDN3 without cross-reactivity to CLDN4, CLDN6, or CLDN9

  • Mechanisms of Action:

    • Induces antibody-dependent cellular cytotoxicity (ADCC) via FcγRIIIa activation

    • Localizes to tumors in xenograft models (20.4% ID/g tumor uptake)

Bispecific Antibody-Drug Conjugates (BsADCs)

A novel EpCAM × CLDN3 bispecific ADC (BsADC 3) demonstrated:

ParameterBsADC 3 Performance
Drug-to-Antibody Ratio3.9–4.12 (via HIC analysis)
In Vitro IC500.72 μg/mL (OVCAR-3 cells) vs. 7.95 μg/mL (A549)
In Vivo Half-Life9.2 days (total antibody); 9.7 days (conjugated)
Safety ProfileNo hepatotoxicity (AST/ALT within normal ranges)

Theranostic Imaging

A dual-labeled CLDN3 antibody (NOTA-¹¹¹In/FNR648) achieved:

  • 83.4% specificity in ovarian cancer (OVCAR-3) cells

  • 24-hour tumor uptake: 20.4 ± 7.4% ID/g in murine models

Research Gaps and Future Directions

  • Toxicity Challenges: Earlier CLDN3-targeting agents (e.g., CPE toxin) caused hepatic injury, necessitating safer formats like h4G3 or BsADCs .

  • Dual Targeting: Co-targeting CLDN3 and EpCAM enhances tumor specificity and internalization efficiency .

  • Clinical Trials: No phase III data available; preclinical models show promise for ADC and CAR-T applications .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CLPX3 antibody; At1g33360 antibody; F10C21.5 antibody; T16O9.5CLP protease regulatory subunit CLPX3 antibody; mitochondrial antibody
Target Names
CLPX3
Uniprot No.

Target Background

Function
The CLPX3 antibody targets the ATP-dependent specificity component of the mitochondrial Clp protease. This component directs the protease to specific substrates. Notably, CLPX3 can also exhibit chaperone functions in the absence of ClpP.
Database Links

KEGG: ath:AT1G33360

STRING: 3702.AT1G33360.1

UniGene: At.39953

Protein Families
ClpX chaperone family
Subcellular Location
Mitochondrion.

Q&A

What is Claudin-3 and why is it an important research target?

Claudin-3 is a 23 kDa multipass membrane protein belonging to the claudin family of epithelial tight junction proteins. It plays a critical role in maintaining cellular barriers and regulating paracellular permeability. Claudin-3 has gained significant research interest because it is upregulated by epidermal growth factor (EGF) and during inflammatory processes. Additionally, its expression is elevated in various epithelial cancers, making it a potential biomarker and therapeutic target. In pathological conditions affecting the blood-brain barrier, Claudin-3 expression is notably lost, further highlighting its importance in normal physiological function. The protein's interaction with Clostridium perfringens exotoxin, which can induce epithelial cell lysis, has also made it relevant for targeted therapy research .

How do I select the appropriate anti-CLDN3 antibody for flow cytometry applications?

When selecting an anti-CLDN3 antibody for flow cytometry, consider antibodies specifically validated for this application, such as PE-conjugated monoclonal antibodies. Verification of specificity is crucial due to the high homology between claudin family members. Look for antibodies tested against cells with known CLDN3 expression levels, such as PC-3 human prostate cancer cell lines. For optimal results, follow manufacturer protocols for staining membrane-associated proteins, which typically include appropriate fixation methods without permeabilization to preserve the native conformation of extracellular epitopes. Always include appropriate isotype controls (such as IC003P when using FAB4620P) to accurately assess specific binding versus background. Optimal dilutions should be empirically determined for each specific cell type being studied .

What are the recommended storage conditions for maintaining CLDN3 antibody stability?

For optimal stability of CLDN3 antibodies, particularly conjugated antibodies like PE-labeled anti-CLDN3, storage at 2-8°C (standard refrigeration) is typically recommended. Most conjugated antibodies remain stable for approximately 12 months from the date of receipt when properly stored. Importantly, PE-conjugated antibodies require protection from light exposure, as fluorophores are susceptible to photobleaching. Avoid freezing conjugated antibodies, as freeze-thaw cycles can compromise the stability of both the protein structure and the fluorophore conjugate. If long-term storage beyond the recommended period is necessary, the addition of preservatives like sodium azide at low concentrations (0.02-0.05%) may help maintain stability, though potential interference with subsequent applications should be considered .

How can I verify the specificity of a CLDN3 antibody?

To verify CLDN3 antibody specificity, implement a multi-faceted validation approach. First, perform comparative binding assays using HEK293T cells overexpressing CLDN3 versus wild-type controls. Second, conduct cross-reactivity tests against related claudin family members, particularly CLDN4, CLDN6, and CLDN9, which share 90% and 80% homology with CLDN3 in the ECD1 region, respectively. Third, utilize knockout validation by confirming lack of binding in CLDN3 knockout cell lines. Fourth, examine binding profiles across multiple cell lines with known differential CLDN3 expression levels (e.g., strong binding to high-expressing OVCAR-3 and NCI-H1781 cells versus weak binding to low-expressing A549 cells). Finally, confirm specificity through immunoblotting, looking for a single band at the expected molecular weight (23 kDa) and absence of non-specific bands .

What methodologies are recommended for generating monoclonal antibodies against CLDN3?

The generation of highly specific anti-CLDN3 monoclonal antibodies requires specialized immunization strategies due to CLDN3's complex membrane topology and high homology with other claudin family members. A successful approach involves immunizing CLDN3 knockout C57BL/6J mice with human CLDN3 mRNA lipid nanoparticles (LNPs), employing adjuvants such as AddaVax to enhance immune response. Following three immunization cycles at two-week intervals, harvest splenocytes from mice demonstrating high antibody titers and proceed with hybridoma generation through fusion with myeloma cell lines. Screen the resulting hybridomas by flow cytometry using HEK293T-CLDN3 overexpressing cells versus controls. For candidates showing specific binding, extract RNA using commercial kits (e.g., RNeasy), generate cDNA, and sequence to identify unique antibody sequences. The monoclonal antibodies can then be humanized into full-size human IgG1 format and produced in CHO-S cells for further characterization and application .

How can I assess the internalization capabilities of anti-CLDN3 antibodies for developing antibody-drug conjugates?

To evaluate the internalization efficiency of anti-CLDN3 antibodies for ADC development, implement a pH-sensitive fluorescence assay using pHAb reactive dyes. These dyes remain non-fluorescent at neutral pH but fluoresce in acidic environments (pH < 6), making them ideal for tracking endosomal-lysosomal trafficking. Conjugate your anti-CLDN3 antibody with pHAb reactive dyes according to manufacturer protocols (e.g., Promega G9841), typically using a molar ratio of 4:1 (dye:antibody). Incubate antibody-dye conjugates (at 5 μg/mL) with target cells expressing varying levels of CLDN3 (e.g., OVCAR-3, NCI-H1781, and A549 as high, medium, and low CLDN3 expressors). Monitor internalization kinetics via flow cytometry over time (typically 0-24 hours), comparing signal intensity between cell lines. Include non-binding antibody controls to establish background levels. This approach provides quantitative assessment of both the rate and extent of receptor-mediated endocytosis, critical parameters for predicting ADC efficacy .

What factors should be considered when designing binding affinity studies for CLDN3 antibodies?

When designing binding affinity studies for CLDN3 antibodies, multiple technical considerations must be addressed for accurate measurements. Surface plasmon resonance (SPR) using platforms such as BIAcore 8K represents an optimal approach, requiring careful preparation of both antibody and antigen samples. Immobilize antibodies (diluted to 3 μg/mL) on CM5 sensor chips via amine coupling, then prepare CLDN3 antigens at an initial concentration of 200 nM followed by a 2-fold serial dilution series down to 1.5625 nM to capture a full binding curve. Include reference flow cells for background subtraction and implement multiple regeneration cycles to ensure consistent surface activity. When analyzing data with software such as BIAevaluation 3.2, apply appropriate binding models (typically 1:1 Langmuir binding) to determine association (ka), dissociation (kd) rates, and equilibrium dissociation constants (KD). For membrane proteins like CLDN3, consider using recombinant extracellular domains rather than full-length protein to avoid potential artifacts from detergent solubilization .

How do I optimize cytotoxicity assays for evaluating anti-CLDN3 antibody-drug conjugates?

To optimize cytotoxicity assays for evaluating anti-CLDN3 ADCs, a systematic approach is required. Begin by selecting multiple cell lines with varying CLDN3 expression levels (e.g., OVCAR-3 and NCI-H1781 for high expression, A549 for low expression) to establish a correlation between target expression and cytotoxic effects. Seed cells in 96-well plates and allow 24 hours for adherence before applying ADCs at concentrations ranging from 0.001-100 μg/mL using a minimum of 8 concentration points in triplicate. Include appropriate controls: naked antibody, non-binding ADC (e.g., IgG-ADC with identical linker-payload), and free payload. After a 72-hour incubation period, assess cell viability using CCK-8 assay, measuring absorbance at 450 nm and calculating the percentage of viable cells relative to untreated controls. For accurate IC50 determination, normalize data and apply four-parameter logistic regression analysis. Additionally, conduct parallel flow cytometry experiments to correlate CLDN3 expression levels with cytotoxic potency across cell lines .

How can I design bispecific antibodies incorporating anti-CLDN3 binding domains for enhanced tumor targeting?

Designing effective bispecific antibodies (BsAbs) incorporating anti-CLDN3 requires strategic engineering to optimize dual-target binding. Implement Knob-into-hole and CrossMab technologies to ensure correct heavy chain pairing and proper light chain association, respectively. Begin by selecting anti-CLDN3 antibodies with confirmed specificity against CLDN3 but not CLDN4, CLDN6, or CLDN9, despite high sequence homology. For the second binding arm, consider targets frequently co-expressed with CLDN3 in tumors but minimal in healthy tissues, such as EpCAM. When constructing the BsAb, strategically position the CLDN3 binding domain to maintain optimal binding kinetics, as avidity effects can be significantly impacted by the bispecific format. Express constructs in CHO-S cells using transient transfection with optimized heavy:light chain plasmid ratios (typically 1:2), and harvest supernatant when cell viability drops to approximately 50%. Purify using Protein A affinity chromatography followed by size-exclusion chromatography to remove aggregates and half-antibody species. Extensively characterize the resulting BsAbs for dual antigen binding, simultaneous engagement capability, and thermal stability .

What are the critical considerations for developing anti-CLDN3 antibody-drug conjugates with optimal therapeutic windows?

Developing anti-CLDN3 ADCs with optimal therapeutic windows requires meticulous optimization of multiple parameters. First, antibody selection should focus on clones demonstrating high tumor selectivity, considering that CLDN3 shows expression in some normal tissues. Target antibodies with rapid internalization kinetics in CLDN3-expressing cancer cells, as demonstrated by pHAb dye assays. For conjugation chemistry, cysteine-based approaches after partial reduction of interchain disulfides offer a controlled drug-to-antibody ratio (DAR); aim for a DAR of approximately 4 (3.9-4.2) to balance potency with pharmacokinetic properties. Select linker-payload combinations appropriate for the intracellular trafficking of CLDN3; the GGFG linker coupled with topoisomerase I inhibitors like deruxtecan (DXd) has demonstrated promising results. Establish detailed in vitro cytotoxicity profiles across cell lines with varying CLDN3 expression levels to predict the therapeutic window, and conduct side-by-side comparisons with reference ADCs conjugated to non-binding antibodies (IgG-ADC) to confirm target specificity. Finally, evaluate potential synergistic effects when combining with agents targeting complementary pathways in cancer cells .

How should researchers address potential cross-reactivity concerns with anti-CLDN3 antibodies given the high homology within the claudin family?

Addressing cross-reactivity concerns with anti-CLDN3 antibodies requires a comprehensive epitope-focused strategy. Begin by analyzing sequence alignments between CLDN3 and related family members, particularly CLDN4 (90% homology), CLDN6 and CLDN9 (both 80% homology) in the ECD1 region. Target immunization strategies toward the more divergent ECD2 region or unique conformational epitopes. Implement a hierarchical screening approach starting with binding assays against a panel of HEK293T cells overexpressing individual claudin family members (CLDN3, CLDN4, CLDN6, CLDN9). For promising candidates, perform competitive binding experiments to identify antibodies recognizing non-overlapping epitopes. Confirm specificity through epitope mapping techniques such as hydrogen-deuterium exchange mass spectrometry or X-ray crystallography of the antibody-antigen complex. For therapeutically intended antibodies, conduct cross-reactivity tissue microarray studies across human tissues expressing various claudin family members. Finally, validate species cross-reactivity with mouse and cynomolgus monkey CLDN3 to enable appropriate preclinical toxicology studies while maintaining specificity against other claudin family members in these species .

What methodologies should be employed to investigate the mechanism of action of anti-CLDN3 antibodies in disrupting tight junction functionality?

Investigating how anti-CLDN3 antibodies affect tight junction functionality requires multi-parameter analysis at molecular, cellular, and functional levels. At the molecular level, implement proximity ligation assays to quantify interactions between CLDN3 and other tight junction components (occludin, ZO-1) before and after antibody treatment. At the cellular level, utilize confocal microscopy with dual immunofluorescence labeling to track real-time redistribution of CLDN3 and associated proteins. Measure tight junction strand remodeling through freeze-fracture electron microscopy, which allows visualization of tight junction architecture disruption. Functionally assess epithelial barrier integrity through transepithelial electrical resistance (TEER) measurements in polarized cell models (e.g., Caco-2 or MDCK cells transfected with human CLDN3) treated with antibodies at various concentrations (1-50 μg/ml) and time points (1-72 hours). Complement TEER with paracellular flux assays using fluorescent tracers of different molecular weights (e.g., FITC-dextran 4kDa, 10kDa, 40kDa) to characterize size-selective permeability changes. Additionally, investigate potential intracellular signaling cascades activated by antibody binding through phosphoproteomic analysis, focusing on pathways known to regulate tight junction assembly and disassembly .

How can researchers troubleshoot inconsistent binding of anti-CLDN3 antibodies in flow cytometry experiments?

Inconsistent binding of anti-CLDN3 antibodies in flow cytometry experiments may stem from multiple factors requiring systematic troubleshooting. First, verify CLDN3 expression levels in your cell population, as expression can vary with culture conditions and cell density; consider using positive control cell lines with established CLDN3 expression (e.g., OVCAR-3). Second, optimize cell preparation protocols, as excessive enzymatic dissociation (particularly trypsin) can cleave extracellular epitopes of membrane proteins like CLDN3; use gentler dissociation methods such as EDTA-based solutions or commercially available non-enzymatic dissociation buffers. Third, adjust fixation procedures, as overfixation may mask epitopes; test paraformaldehyde concentrations between 1-4% or consider using live cell staining protocols. Fourth, examine antibody concentration and incubation conditions; perform titration experiments (0.1-10 μg/mL) at different temperatures (4°C, room temperature, 37°C) and durations (15-60 minutes). Fifth, assess buffer composition, particularly ensuring physiological pH and the presence of protein blockers (1-5% BSA or serum) to reduce non-specific binding. Finally, for conjugated antibodies like PE-anti-CLDN3, verify fluorophore integrity by testing with known positive controls, as fluorophores can degrade with improper storage or repeated freeze-thaw cycles .

What strategies can enhance the production and purification yields of anti-CLDN3 bispecific antibodies?

Enhancing production and purification yields of anti-CLDN3 bispecific antibodies requires optimization at multiple stages of the manufacturing process. During vector design, implement balanced expression of all chains by using optimized promoters and signal peptides, while incorporating the Knob-into-hole and CrossMab technologies to ensure proper chain pairing. For transfection, optimize the ratio of heavy and light chain plasmids (typically 1:2) and implement high-density transient transfection systems using CHO-S cells adapted to serum-free suspension culture. During cell culture, implement fed-batch processes with optimized nutrient supplements and maintain culture at lower temperatures (30-32°C) during the production phase to reduce proteolytic activity while enhancing proper protein folding. For harvesting, collect supernatant when cell viability is approximately 50%, then implement clarification through depth filtration before affinity purification. During purification, use Protein A Plus Agarose chromatography followed by additional polishing steps such as ion exchange and size exclusion chromatography to remove aggregates, half-antibodies, and host cell proteins. Finally, formulate in stabilizing buffers containing appropriate excipients (e.g., trehalose, polysorbate 80) to prevent aggregation during storage .

How should researchers address potential experimental artifacts when evaluating anti-CLDN3 antibody internalization?

When evaluating anti-CLDN3 antibody internalization, several experimental artifacts can confound results and require specific mitigation strategies. First, distinguish between active receptor-mediated endocytosis and passive uptake by implementing temperature controls; compare internalization at 37°C (active transport) versus 4°C (inhibited endocytosis). Second, account for potential pH-dependent fluorescence artifacts in pHAb dye assays by establishing appropriate calibration curves using ionophores (e.g., nigericin) to equilibrate intracellular and extracellular pH. Third, differentiate surface-bound versus truly internalized antibodies by implementing acid wash steps (pH 2.5-3.0 glycine buffer) to strip surface-bound antibodies without affecting internalized fractions. Fourth, consider the impact of antibody valency, as bivalent binding may artificially enhance apparent internalization through receptor crosslinking; compare monovalent Fab fragments with complete IgG to assess this effect. Fifth, validate internalization through complementary techniques such as confocal microscopy with Z-stack analysis to visualize subcellular localization. Finally, account for recycling of internalized antibodies by performing pulse-chase experiments with differently labeled antibodies to distinguish between newly internalized and recycled molecules .

What are the best practices for validating the cytotoxic potency of anti-CLDN3 antibody-drug conjugates across different experimental systems?

Validating cytotoxic potency of anti-CLDN3 ADCs across experimental systems requires rigorous standardization and multiple complementary approaches. Implement a tiered testing strategy beginning with 2D monolayer cytotoxicity assays using CCK-8 or similar viability reagents across cell lines with quantified CLDN3 expression levels. Progress to 3D spheroid models, which better recapitulate in vivo architecture and often demonstrate different sensitivity profiles; monitor spheroid growth through diameter measurements and viability using reagents capable of penetrating spheroids (e.g., CellTiter-Glo 3D). For mechanistic validation, conduct cell cycle analysis and apoptosis assays (Annexin V/PI staining) to confirm the expected mechanism of action of the payload. Evaluate bystander killing effects using co-culture systems of CLDN3-positive and CLDN3-negative cells labeled with different tracking dyes. For in vivo validation, establish xenograft models with varying CLDN3 expression levels and implement rigorous dosing schedules, including regular body weight monitoring and tumor volume measurements. Include appropriate controls: naked antibody, non-binding ADC with identical linker-payload (IgG-ADC), and vehicle. Finally, quantify intratumoral accumulation through ex vivo analysis of ADC concentration and perform immunohistochemistry to correlate CLDN3 expression with response .

How might anti-CLDN3 antibodies be utilized in combination with emerging immunotherapy approaches?

Anti-CLDN3 antibodies present significant potential for integration with emerging immunotherapy approaches through multiple mechanistic strategies. First, consider developing CLDN3-targeted bispecific T-cell engagers (BiTEs) that simultaneously bind CLDN3 on tumor cells and CD3 on T cells, leveraging the high specificity of anti-CLDN3 antibodies for tumor recognition while recruiting cytotoxic T cells to the tumor microenvironment. Second, explore anti-CLDN3 antibody-cytokine fusion proteins (immunocytokines) that can deliver immunostimulatory cytokines (IL-2, IL-12, IFN-γ) specifically to the tumor microenvironment, potentially converting "cold" tumors to "hot" immunologically active tumors. Third, investigate combination approaches of anti-CLDN3 ADCs with immune checkpoint inhibitors (anti-PD-1/PD-L1, anti-CTLA-4), as ADC-induced immunogenic cell death may enhance tumor antigen release and presentation, potentially synergizing with checkpoint blockade. Fourth, develop CLDN3-targeted delivery of Toll-like receptor (TLR) agonists to stimulate innate immunity within tumors. Finally, examine potential roles of anti-CLDN3 antibodies in modulating tight junction integrity within the tumor microenvironment, which might enhance penetration of immunotherapeutic agents or increase exposure of tumor antigens to immune surveillance .

What are the emerging applications of anti-CLDN3 antibodies in detecting circulating tumor cells and liquid biopsy approaches?

Anti-CLDN3 antibodies offer promising applications in liquid biopsy approaches, particularly for detecting circulating tumor cells (CTCs) from epithelial cancers where CLDN3 is overexpressed. Developing highly specific anti-CLDN3 antibodies conjugated to capture surfaces (magnetic beads, microfluidic chips) can enable positive selection of CLDN3-expressing CTCs from peripheral blood samples. This approach may complement existing EpCAM-based CTC isolation methods, potentially capturing EpCAM-low/CLDN3-high CTCs that would be missed by conventional approaches. For enhanced sensitivity, implement a dual-capture system using both anti-CLDN3 and anti-EpCAM antibodies, followed by downstream confirmation through immunofluorescence staining or molecular characterization. Additionally, anti-CLDN3 antibodies can be employed to detect CLDN3-expressing extracellular vesicles (EVs), which may serve as biomarkers for cancer diagnosis and monitoring. As demonstrated in research on high-grade serous ovarian carcinoma, specialized nanowire-based approaches incorporating antibodies against tumor markers can isolate cancer-specific EVs from bodily fluids, offering potential for early detection and disease monitoring through minimally invasive sampling .

How might structural biology approaches enhance the development of next-generation anti-CLDN3 antibodies with improved specificity and functionality?

Structural biology approaches can revolutionize anti-CLDN3 antibody development through multiple avenues. First, implement high-resolution cryo-electron microscopy to determine the structure of CLDN3 in its native membrane environment, revealing conformational epitopes that may be absent in recombinant proteins. Second, utilize X-ray crystallography of CLDN3-antibody complexes to precisely map epitope-paratope interactions, enabling structure-guided antibody engineering to enhance specificity against homologous claudin family members. Third, employ hydrogen-deuterium exchange mass spectrometry to identify dynamic regions and conformational changes in CLDN3 upon antibody binding, potentially revealing allosteric effects that could be therapeutically exploited. Fourth, implement molecular dynamics simulations to predict the effects of antibody binding on tight junction assembly and stability, guiding the development of antibodies that either stabilize or disrupt these structures based on therapeutic goals. Fifth, employ directed evolution approaches coupled with structural insights to develop antibodies targeting CLDN3-specific regions with enhanced binding properties. Finally, utilize structural data to design bispecific constructs with optimal spatial orientation of binding domains, ensuring efficient simultaneous engagement of CLDN3 and secondary targets like EpCAM for improved tumor targeting .

What considerations should guide the development of anti-CLDN3 antibodies for targeting cancers with acquired resistance to conventional therapies?

Developing anti-CLDN3 antibodies for targeting therapy-resistant cancers requires strategic considerations addressing multiple resistance mechanisms. First, conduct comprehensive profiling of CLDN3 expression across therapy-resistant versus therapy-sensitive tumor samples to identify potential correlations between CLDN3 expression and resistance phenotypes. Second, investigate whether CLDN3 localization changes in resistant cells, as redistribution from tight junctions to other cellular compartments may affect antibody accessibility and internalization. Third, design ADCs with payloads specifically selected to overcome known resistance mechanisms, such as MMAE for taxane-resistant tumors or topoisomerase inhibitors like DXd for tumors resistant to DNA-damaging agents. Fourth, develop bispecific antibodies targeting both CLDN3 and secondary targets involved in resistance pathways (e.g., growth factor receptors, anti-apoptotic proteins) to simultaneously address multiple therapeutic vulnerabilities. Fifth, evaluate the impact of the tumor microenvironment on anti-CLDN3 antibody efficacy in resistant models, as stromal barriers and hypoxia can affect antibody penetration and activity. Finally, investigate combination approaches of anti-CLDN3 antibodies with agents that modulate resistance pathways, such as inhibitors of drug efflux pumps, anti-apoptotic proteins, or DNA damage repair mechanisms, to develop rational combination strategies specifically tailored for therapy-resistant disease contexts .

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 2025 TheBiotek. All Rights Reserved.