CLDN6/9 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Therapeutic CLDN6-Specific Antibodies

Recent efforts focus on developing antibodies with strict CLDN6 specificity for cancer therapies, including antibody-drug conjugates (ADCs) and bispecific T-cell engagers. Key examples include:

CLDN6-Specific Monoclonal Antibodies (MAbs)

AntibodyVH CDR3 LengthCLDN6 EC50 (nM)CLDN9 BindingKey Epitope Residues
IM301182.8 ± 0.8InsufficientE48, E154, R158
IM302181.0 ± 0.1InsufficientE154, R158
IM136191.5 ± 0.5InsufficientE48, D68, R158
IM171182.1 ± 0.5InsufficientT33, N38, E48, A153, R158

Table adapted from : MAbs with >35-fold higher CLDN6 vs. CLDN9 reactivity.

Key Features:

  • High Affinity: IM302 demonstrates the strongest binding (EC50 = 1.0 nM) .

  • Cross-Reactivity Avoidance: Biosensor assays confirm negligible binding to CLDN9, CLDN3, or CLDN4 .

  • Humanized Variants: IM301 and IM302 (derived from IM171) show improved developability for clinical use .

CLDN6/9 Cross-Reactive Antibody (Cusabio)

The CLDN6/9 recombinant monoclonal antibody (Cusabio, CSB-RA005508MA1HU) is a research-grade tool for detecting both CLDN6 and CLDN9.

Therapeutic vs. Research Antibodies: Comparative Analysis

FeatureTherapeutic CLDN6 MAbsCLDN6/9 Research Antibody
SpecificityCLDN6-onlyCLDN6 + CLDN9
ApplicationsADCs, CAR-T therapiesELISA, Flow Cytometry, IHC
AffinityHigh (EC50 < 3 nM)Moderate (ELISA/FC validated)
Clinical StatusPhase I (e.g., DS-9606) Preclinical (research use only)
Cross-Reactivity RiskLow (rigorously deselected)High (intentional dual-targeting)

CLDN6-Specific ADCs

  • CLDN6–23-ADC: Humanized anti-CLDN6 antibody conjugated to MMAE.

    • Efficacy: Tumor regressions in CLDN6+ xenografts/PDX models (ovarian/endometrial cancers) .

    • Internalization Rate: Rapid uptake in CLDN6+ cells, enabling cytotoxic payload delivery .

  • DS-9606: PBD-based ADC in Phase I trials.

    • Early Data: Promising activity in CLDN6+ solid tumors, including germ cell tumors .

CLDN6/9 Antibody in Research

  • Validation: Reactivity confirmed against recombinant CLDN6/9 proteins .

  • Limitations: Cross-reactivity limits utility in therapeutic contexts but enables dual-target studies in vitro.

Challenges and Future Directions

  • Specificity Barriers: CLDN6 and CLDN9 differ by just 3 extracellular residues, requiring atomic-level epitope engineering .

  • Clinical Translation: Ongoing trials (e.g., DS-9606) aim to validate CLDN6 as a viable target in cancers like ovarian and endometrial carcinomas .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M PBS, pH 7.4
Description

CUSABIO's CLDN6/9 recombinant monoclonal antibody is meticulously produced through a robust and well-defined process. This antibody is generated using a recombinant human CLDN6/9 protein as an immunogen. The immunization process involves isolating B cells from the spleen of an immunized animal. RNA extraction from these B cells is followed by reverse transcription into cDNA. The gene encoding the CLDN6/9 antibody is then amplified using a degenerate primer and subsequently inserted into a suitable vector. This recombinant vector is then introduced into host cells through transfection, facilitating efficient antibody expression. The CLDN6/9 recombinant monoclonal antibodies are harvested from the cell culture supernatant and purified using affinity chromatography. Extensive validation, encompassing ELISA and FC testing, is meticulously performed to confirm the antibody's reactivity with human CLDN6 and CLDN9 protein, ensuring its accuracy and suitability for diverse applications.

Form
Liquid
Lead Time
Typically, we can dispatch products within 1-3 business days after receiving your order. Delivery time may vary depending on the purchase method and destination. Please consult your local distributor for specific delivery timelines.
Synonyms
Claudin-6 antibody; Claudin6 antibody; CLD6_HUMAN antibody; CLDN 6 antibody; CLDN6 antibody; OTTHUMP00000159248 antibody; Skullin 2 antibody; Skullin antibody; UNQ757/PRO1488 antibody
Target Names
CLDN6/9
Uniprot No.

Target Background

Function

CLDN6/9 plays a pivotal role in tight junction-specific obliteration of the intercellular space. It also serves as a receptor for hepatitis C virus (HCV) entry into hepatic cells, significantly contributing to microbial infection.

Gene References Into Functions
  1. Studies have shown that antibodies recognizing native CLDN6 on cell surfaces, mediating complement-dependent cytotoxicity, are elicited in vaccinated animals. This suggests potential applications of CLDN6 displaying Virus-like particles in cancer immunotherapy. PMID: 29131519
  2. Research indicates that Helicobacter pylori lipopolysaccharide induces TLR2 expression in gastric adenocarcinoma cells, with prolonged exposure leading to increased expression of TLR2 on the cell membrane. This, in turn, results in enhanced expression of claudin-4, -6, -7 and -9. PMID: 29031421
  3. Evidence suggests that high CLDN6 expression confers chemoresistance in breast cancer, mediated by GSTP1, whose activity is regulated by p53. PMID: 29116019
  4. CLDN6 enhances chemoresistance to ADM by activating the AF-6/ERK signaling pathway and up-regulating cancer stem cell characteristics in MDAMB231 cells. PMID: 29159771
  5. Research demonstrates that downregulation of CLDN6 is regulated through promoter methylation by DNMT1, dependent on the SMAD2 pathway. CLDN6 is a key regulator in the SMAD2/DNMT1/CLDN6 pathway, inhibiting EMT, migration and invasion of breast cancer cells. PMID: 28867761
  6. Bioinformatics analysis provides insights for future studies aimed at better understanding CLDN6 regulation and functions. PMID: 28656265
  7. High CLDN 6 expression is observed in approximately 65% of myxofibrosarcomas, while benign soft tissue tumors do not exhibit high CLDN 6 expression. The expression of CLDN 6 in myxofibrosarcomas is significantly higher than other tumor specimens. High CLDN 6 expression in myxofibrosarcomas is correlated with high FNCLCC grades and high AJCC stages. PMID: 28476380
  8. Results indicate that DNA methylation down-regulates CLDN6 expression through MeCP2 binding to the CLDN6 promoter, deacetylating H3 and H4, and altering chromatin structure, ultimately promoting migratory and invasive phenotype in breast cancer cells. PMID: 27461117
  9. Cldn6 is decreased in alveolar type II-like epithelial cells (A549) and primary small airway epithelial cells upon exposure to cigarette smoke extract. PMID: 27982694
  10. Studies suggest that claudin-6 induces MMP-2 activation through claudin-1 membrane expression. PMID: 27914788
  11. Data reveal that claudin-6 (CLDN6) R209Q and occludin (OCLN) P24A mutations do not affect HCV pseudoparticles (HCVpp) entry. PMID: 26561856
  12. The expression of ASK1 is correlated with the level of claudin-6 in cervical carcinoma cells and tissues. PMID: 26191261
  13. High levels of CLDN6 are associated with non-small-cell lung cancer. PMID: 24710653
  14. The expression of claudin-6 is downregulated in gastric cancer tissue. PMID: 23919729
  15. Only certain hepatitis C virus strains effectively utilize CLDN6 for infection. PMID: 23775920
  16. This research provides a proof of concept for utilizing Claudin-6 to eliminate residual undifferentiated human pluripotent stem cells from culture. PMID: 23778593
  17. Although claudin-6 and claudin-9 can serve as entry factors in cell lines, hepatitis C virus infection into human hepatocytes is not dependent on claudin-6 and claudin-9. PMID: 23864633
  18. ASK1 signaling may play a positive role in the inhibitory effect of claudin-6 in breast cancer. PMID: 22925655
  19. Research findings show that claudin-6 protein is significantly down-regulated in breast invasive ductal carcinomas. PMID: 22455563
  20. CLDN6 is not a specific biomarker for atypical teratoid rhabdoid tumors as it is expressed in various other pediatric CNS and soft tissue tumors. PMID: 21989342
  21. 17beta-E2 potentially regulates the expression of claudin-6 and inhibits the proliferation and migration of MCF-7 cells. PMID: 20388399
  22. Increased expression of claudin-6, claudin-7, or claudin-9 enhances tumorigenic properties of a gastric adenocarcinoma cell line. PMID: 20874001
  23. CLDN6 may serve as a useful positive marker to assist in identifying atypical teratoid/rhabdoid tumors for diagnostic and treatment purposes. PMID: 19220299
  24. Claudins 6, 7, and 9 expressions are closely linked to gastric carcinogenesis. PMID: 19960275
  25. Up-regulation of claudin-6 expression in MCF-7 breast cancer cells suppresses their malignant phenotypes, correlated with the restoration of tight junction integrity. PMID: 20367941
  26. Claudin-6 downregulates the malignant phenotype of breast carcinoma. PMID: 20215972
  27. Claudin 6 is absent in epithelioid glioblastomas or rhabdoid glioblastomas. PMID: 20118769
  28. CLDN6 and CLDN9, but not CLDN1, are expressed in peripheral blood mononuclear cells, an additional site of HCV replication. PMID: 17804490
  29. Claudin-6 and claudin-9 expressed in CD81+ cells also facilitate the entry of HCV pseudoparticles derived from six of the major genotypes. PMID: 18234789
  30. CLDN6, clustered with CLDN9 at human chromosome 16p13.3, is a four-transmembrane protein with WWCC motif, characterized by W-X(17-22)-W-X(2)-C-X(8-10)-C. PMID: 12736707

Show More

Hide All

Database Links

HGNC: 2048

OMIM: 615798

KEGG: hsa:9074

STRING: 9606.ENSP00000328674

UniGene: Hs.533779

Protein Families
Claudin family
Subcellular Location
Cell junction, tight junction. Cell membrane; Multi-pass membrane protein.
Tissue Specificity
Expressed in the liver, in peripheral blood mononuclear cells and hepatocarcinoma cell lines.

Q&A

What is CLDN6 and why is it considered a promising cancer target?

CLDN6 is a tight junction protein that has emerged as a promising cancer target due to its differential expression pattern - it is found on cancer cells while showing almost no expression in healthy adult tissues. This highly selective expression profile makes it an ideal candidate for targeted cancer therapies with potentially minimal off-target effects on normal cells . Specifically, CLDN6 belongs to the claudin family of membrane proteins that are critical components of tight junctions, and its aberrant expression in certain cancer types provides a unique opportunity for therapeutic intervention with highly specific monoclonal antibodies.

How are CLDN6 and CLDN9 structurally related, and why does this present challenges?

CLDN6 and CLDN9 represent a unique challenge in antibody development due to their extraordinary sequence similarity. These 4-transmembrane proteins are nearly identical, with only 3 amino acid differences in their extracellular domains . This high homology makes it exceptionally difficult to develop antibodies that can distinguish between them.

The therapeutic challenge arises because CLDN9 is widely expressed in normal tissues, while CLDN6 expression is largely restricted to cancer cells. Therefore, antibodies that cross-react with CLDN9 may cause significant off-target toxicity in clinical applications, which has led to the termination of several clinical trials involving non-specific anti-CLDN6 antibodies . The development of truly CLDN6-specific antibodies requires sophisticated approaches to target the minimal differences between these highly conserved proteins.

What experimental methods can be used to generate CLDN6-specific monoclonal antibodies?

Generating CLDN6-specific antibodies requires specialized methodological approaches:

How can you characterize the binding properties of CLDN6-specific antibodies?

Characterization of CLDN6-specific antibodies requires multiple complementary approaches:

  • Flow cytometry binding studies: Test antibodies against cells expressing CLDN6, CLDN9, and other claudin family members to determine binding EC50 values and calculate specificity ratios .

  • Binding kinetics analysis: Use biosensor-based techniques to determine binding constants (KD), association rates (kon), and dissociation rates (koff). This provides critical information about binding affinity and the kinetic mechanisms of antibody-antigen interactions .

  • Cross-reactivity assessment: Test antibodies against cell lines expressing each claudin family member to identify any off-target binding within the family .

The table below shows example binding kinetics data that might be obtained during characterization:

MAbCLDN6 KD (nM)CLDN6 kon (nM^-1s^-1)CLDN6 koff (s^-1)CLDN9 KD (nM)Specificity Ratio
IM302<0.0014.0×10^-4<1×10^-7No binding>1000
IM3010.5 ± 0.032.1×10^-41.2×10^-4No binding>500
IM1720.030 ± 0.0026.8×10^-42.1×10^-595 ± 4~3167

What epitope mapping strategies can identify the molecular determinants of CLDN6 antibody specificity?

Epitope mapping for membrane proteins like CLDN6 requires specialized approaches:

  • Shotgun mutagenesis epitope mapping: This comprehensive approach involves performing an alanine scan across the entire CLDN6 sequence (219 amino acids). Each residue is mutated to alanine (or serine for native alanines), and the mutants are expressed in cells and tested for antibody binding using high-throughput flow cytometry . This technique allows researchers to:

    • Identify specific residues critical for antibody binding (typically defined as mutations causing <35% binding relative to wild-type)

    • Distinguish between residues directly involved in binding versus those affecting protein folding or expression using control antibodies

    • Map the complete binding footprint of each antibody

  • Atomic-level epitope mapping: This approach builds on the results of shotgun mutagenesis to determine the precise structural mechanism of specificity. For example, research has identified that steric hindrance at the γ carbon on CLDN6 residue Q156 enables certain antibodies to differentiate between CLDN6 and CLDN9 .

  • Comparative epitope analysis: By mapping epitopes of multiple CLDN6-binding antibodies with different specificity profiles, researchers can identify critical regions that confer selectivity. For example, residue R158 has been identified as critical for binding of multiple CLDN6-specific antibodies .

How does VH CDR3 length impact antibody specificity for challenging targets like CLDN6?

The length of the heavy-chain complementarity-determining region 3 (VH CDR3) plays a crucial role in determining antibody specificity, especially for challenging targets like CLDN6:

  • Correlation between VH CDR3 length and specificity: Highly specific CLDN6 antibodies often feature longer VH CDR3 regions (18-20 residues) compared to typical human- or mouse-derived antibodies . These longer CDR3 regions may provide additional contact points or conformational flexibility that enables discrimination between highly similar proteins.

  • Host-specific considerations: Chickens naturally produce antibodies with longer VH CDR3 regions than humans or mice, making them valuable hosts for generating antibodies against conserved targets . This characteristic can be exploited when designing immunization strategies for targets like CLDN6.

  • Structural implications: Longer VH CDR3 regions may create unique binding interfaces that can reach into recessed epitopes or create steric constraints that enhance specificity. For example, when comparing highly specific CLDN6 antibodies (IM136, IM171, IM172, IM173) with benchmark antibodies (IMAB027, IMAB206), the specific antibodies have VH CDR3 lengths of 18-20 residues versus 8 residues for the less specific benchmarks .

The following table illustrates this relationship:

AntibodyVH CDR3 LengthCLDN6 KD (nM)CLDN9 Cross-reactivityCritical Epitope Residues
IM1361911.7 ± 0.2MinimalE48, D68, R158
IM171183.00 ± 0.03MinimalT33, N38, E48, A153, E154, R158
IMAB02780.50 ± 0.01Significant (3.6 nM)F35, G37, S39
IMAB20684.30 ± 0.07Significant (2.1 nM)F35, G37, S39

What proteome-wide screening approaches can validate antibody specificity beyond the target family?

To thoroughly validate antibody specificity beyond the target family, comprehensive proteome-wide screening is essential:

  • Membrane Proteome Array (MPA) technology: This approach allows testing of antibodies against approximately 6,000 membrane proteins (representing 94% of the human membrane proteome) expressed in their native state in unfixed cells . The MPA provides several advantages:

    • Detects unexpected cross-reactivities that wouldn't be identified through targeted testing

    • Screens under conditions designed to maximize sensitivity (high antibody concentrations of 5 μg/mL)

    • Can detect even weak binding interactions (e.g., interactions with KD values in the high nanomolar range)

    • Allows testing under both permeabilized and non-permeabilized conditions to distinguish between intracellular and extracellular binding

  • Statistical analysis of hits: Positive hits are typically defined as more than three standard deviations above mean reactivity levels across the array . This statistical approach helps identify true off-target binding while minimizing false positives.

  • Follow-up validation: Potential off-target interactions identified in the MPA should be validated through secondary assays:

    • Binding to transfected cells expressing the potential off-target protein

    • Testing antibody binding to cells endogenously expressing the off-target

    • Comparison of binding under permeabilized versus non-permeabilized conditions

For example, when screening CLDN6 antibodies, some showed reactivity with the unrelated protein ABCC3. Follow-up experiments revealed this binding was weak and entirely intracellular (only detectable under permeabilized conditions), suggesting minimal risk for therapeutic applications targeting cell-surface CLDN6 .

How can the therapeutic potential of CLDN6-specific antibodies be optimized through antibody engineering?

Optimizing CLDN6-specific antibodies for therapeutic applications involves several engineering approaches:

  • Humanization: Converting chicken-derived antibodies to human-like sequences through CDR grafting while maintaining specificity. For example, the chicken antibody IM171 was successfully humanized to create IM301, retaining high affinity (0.5 nM) and specificity for CLDN6 .

  • Affinity maturation: Further engineering humanized antibodies to improve binding characteristics. The affinity-matured variant IM302 derived from IM301 achieved sub-picomolar affinity (<0.001 nM) while maintaining specificity .

  • Epitope optimization: Engineering antibodies to target epitopes distinct from existing clinical candidates that have shown cross-reactivity issues. Mapping studies have revealed that novel CLDN6 antibodies (IM136, IM171, IM172, IM173, IM301, IM302) target different epitopes than clinical benchmarks (IMAB027, IMAB206, AE49-11), potentially explaining their improved specificity .

  • Developability assessment: Evaluating antibody candidates for properties that will enable successful development, including thermostability, resistance to aggregation, and manufacturability .

What experimental strategies can identify the precise structural mechanism of antibody specificity between highly homologous proteins?

Understanding the precise structural mechanisms that enable antibody discrimination between highly homologous proteins like CLDN6 and CLDN9 requires sophisticated experimental approaches:

  • Comprehensive mutation analysis: Perform alanine scanning mutagenesis across the entire protein sequence to identify all residues involved in antibody binding . Then focus on the residues that differ between CLDN6 and CLDN9 to determine which ones are critical for specificity.

  • Reciprocal mutations: Create CLDN6 mutants where specific amino acids are changed to their CLDN9 counterparts and vice versa. Test if these mutations can convert binding specificity, which would identify key determinants of selectivity .

  • Atomic-level epitope mapping: Use high-resolution techniques to identify precise molecular contact points. For example, research has identified that the γ carbon on CLDN6 residue Q156 serves as a critical determinant for specificity, with steric hindrance at this position preventing certain antibodies from binding to CLDN9 .

  • Structural comparisons of antibody-antigen complexes: Analyze the binding interfaces of antibodies with different specificity profiles (CLDN6-specific vs. CLDN6/9 cross-reactive) to identify key structural features that confer selectivity.

What expression systems are optimal for generating CLDN6 proteins for antibody discovery?

Generating properly folded membrane proteins like CLDN6 for antibody discovery requires specialized expression systems:

  • Lipoparticle technology: Virus-like particles incorporating structurally intact membrane proteins provide significant advantages for antibody discovery:

    • Allows isolation of CLDN6 at protein concentrations 10-100 fold higher than typical membrane preparations

    • Preserves native protein structure and orientation

    • Can be used directly for immunization, panning, and screening

    • Enables efficient deselection strategies against highly similar proteins (CLDN9)

  • Mammalian cell expression: For final validation, stable cell lines expressing individual claudin family members should be established to confirm antibody specificity across the family .

  • Transient transfection systems: For high-throughput applications like epitope mapping, transient transfection in HEK-293T cells can be used, with appropriate controls to ensure proper protein expression and folding .

How should researchers design deselection strategies to isolate CLDN6-specific antibodies?

Effective deselection strategies are crucial for isolating antibodies that specifically recognize CLDN6 over the highly similar CLDN9:

  • Sequential panning rounds: Perform multiple rounds of phage display selection, incorporating deselection steps against CLDN9. For example, allow the phage library to bind to wells coated with CLDN6 for positive selection, while removing phages that bind to CLDN9-coated wells .

  • Competitive binding approaches: Include soluble CLDN9 or CLDN9-expressing cells during selection to competitively remove cross-reactive antibodies from the pool .

  • Negative screening: Following initial identification of CLDN6 binders, implement high-throughput screening against both CLDN6 and CLDN9 to identify clones with the highest specificity ratios .

  • Multi-parameter selection: Combine binding strength (affinity) and specificity criteria when selecting antibody candidates, rather than focusing solely on affinity for CLDN6 .

In one successful approach, researchers performed three consecutive rounds of phage panning with CLDN6 Lipoparticles while deselecting against CLDN9 Lipoparticles, resulting in 1,891 unique reactive scFv fragments. Through further screening, they identified 159 fragments with strong CLDN6 binding (signal-to-background ratio >5) and ultimately selected 5 monoclonal antibodies with >35-fold higher reactivity to CLDN6 compared to CLDN9 .

What control experiments are essential when validating CLDN6 antibody specificity?

Rigorous validation of CLDN6 antibody specificity requires multiple control experiments:

  • Cross-claudin family testing: Test antibodies against all 24 members of the claudin family to ensure specificity within the family .

  • Control antibodies for epitope mapping: When performing mutagenesis-based epitope mapping, include control antibodies to distinguish mutations that affect antibody binding from those that disrupt protein expression or folding .

  • Species cross-reactivity: Test antibodies against orthologous proteins from relevant species (mouse, cynomolgus monkey) to assess conservation of the epitope and predict translational relevance .

  • Binding format controls: Test antibodies in multiple formats (scFv, IgG) to ensure that specificity is maintained when the antibody is reformatted .

  • Membrane proteome screening: Validate specificity against the entire human membrane proteome using technologies like the Membrane Proteome Array to identify any unpredicted cross-reactivities .

  • Permeabilization controls: When evaluating potential off-target binding, test under both permeabilized and non-permeabilized conditions to distinguish between intracellular binding (generally lower risk for therapeutic antibodies) and cell-surface binding .

How should researchers interpret weak cross-reactivity detected in proteome-wide screening?

Interpreting weak cross-reactivity detected in proteome-wide screening requires careful consideration of multiple factors:

  • Context of detection: The Membrane Proteome Array (MPA) is typically screened using high antibody concentrations (5 μg/mL) to maximize sensitivity for off-target binding. This can detect very weak interactions that may not be biologically relevant at therapeutic concentrations .

  • Quantitative analysis: Compare the affinity (KD) of the primary target interaction versus the off-target interaction. Large differences (e.g., sub-nanomolar vs. high nanomolar) typically indicate minimal risk .

  • Subcellular localization: Determine if off-target binding occurs only intracellularly (requiring cell permeabilization) or on the cell surface. For therapeutic antibodies targeting cell-surface proteins, intracellular cross-reactivity presents minimal risk since antibodies generally cannot access intracellular compartments in vivo .

  • Expression pattern: Evaluate the tissue expression pattern of any potential off-target proteins to assess overlap with the intended treatment indication .

For example, when screening CLDN6 antibodies, some showed reactivity with the unrelated protein ABCC3. Follow-up experiments demonstrated this binding was weak and entirely intracellular (only under permeabilized conditions), suggesting minimal risk for therapeutic applications targeting cell-surface CLDN6 .

What approaches can resolve conflicting data between different antibody specificity assays?

Resolving conflicting data between different antibody specificity assays requires systematic troubleshooting:

  • Assay sensitivity differences: Different assays have varying sensitivity thresholds. For example, biosensor analysis might detect weak interactions that are below the detection limit of flow cytometry. Understanding these differences helps interpret seemingly conflicting results .

  • Protein conformation effects: The conformation of the target protein can vary between assays. Lipoparticles, cell-surface expression, and recombinant proteins may present different epitopes, leading to apparent discrepancies in binding results .

  • Hierarchical validation approach: Implement a hierarchical approach where initial high-throughput screening is followed by progressively more detailed characterization using orthogonal methods:

    • Initial screening by flow cytometry

    • Detailed binding kinetics analysis by biosensor

    • Epitope mapping to understand molecular binding determinants

    • Proteome-wide specificity assessment

    • Cellular functional assays

  • Quantitative comparison: When different assays show varying degrees of cross-reactivity, quantify the specificity ratio (affinity for target vs. off-target) rather than making binary assessments of specificity .

By implementing this systematic approach, researchers can develop a comprehensive understanding of antibody specificity that accounts for the strengths and limitations of each assay format.

What novel approaches might further improve the specificity of antibodies targeting highly conserved membrane proteins?

Several innovative approaches hold promise for further improving antibody specificity against highly conserved targets like CLDN6:

  • Structure-guided antibody engineering: Using atomic-level understanding of the epitope-paratope interface to introduce mutations that enhance specificity. For example, knowledge that the γ carbon on CLDN6 residue Q156 enables specificity through steric hindrance could guide rational design of even more selective antibodies .

  • Combinatorial epitope targeting: Developing bispecific antibodies that simultaneously engage two distinct epitopes on CLDN6, requiring both to be present for high-affinity binding. This would dramatically reduce the probability of cross-reactivity .

  • Alternative scaffold proteins: Exploring non-antibody binding proteins that may offer different binding geometries and potentially access epitopes that are challenging for conventional antibodies .

  • High-throughput epitope binning: Comprehensive epitope binning of large antibody panels to identify unique epitopes that confer maximal specificity .

  • Advanced computational design: Applying computational antibody design tools to predict modifications that would enhance specificity based on structural models of CLDN6 and CLDN9 .

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 2024 Thebiotek. All Rights Reserved.