CLDN7 Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Introduction to Claudin-7 (CLDN7) and Its Monoclonal Antibody

Claudin-7 (CLDN7) is a transmembrane protein critical for forming tight junctions, which regulate paracellular permeability and maintain epithelial cell polarity . As a member of the claudin family, it is encoded by the CLDN7 gene (Gene ID: 1366) and plays dual roles in cancer biology: acting as a tumor suppressor in colorectal, lung, and breast cancers, while promoting invasiveness in ovarian malignancies . CLDN7 monoclonal antibodies (mAbs) are essential tools for studying its expression patterns, cellular localization, and functional roles in both normal physiology and disease states .

Key Features of CLDN7 Monoclonal Antibodies

CLDN7 mAbs are engineered for high specificity and reproducibility across applications. Below is a comparative analysis of commercially available CLDN7 monoclonal antibodies:

CloneHost SpeciesIsotypeReactivityApplicationsLocalization
EP399RabbitIgGHuman (FFPE, frozen)IHC, IFCytoplasmic, Membranous
EPR18073RabbitIgGHumanICC/IF, Flow Cytometry, IHC-PMembranous
60800-2-PBSMouseIgG1HumanELISA, Cytometric Bead ArrayCell Junctions

Notable Characteristics:

  • EP399: Validated in transitional cell carcinoma tissues; detects loss of CLDN7 in ~50% of breast cancers .

  • EPR18073: Demonstrates membranous staining in breast adenocarcinoma cells (MCF7) .

  • 60800-2-PBS: Conjugation-ready format for multiplex assays .

3.1. Diagnostic and Prognostic Utility

  • Colorectal Cancer (CRC): Serum CLDN7 levels are significantly lower in CRC patients (11.57 ng/mL vs. 26.64 ng/mL in controls), correlating with metastasis and poor differentiation .

  • Ovarian Cancer: CLDN7 is overexpressed in 87% of epithelial ovarian carcinomas (EOCs), promoting invasion via Erk/Raf-1 signaling . Knockdown reduces transepithelial resistance by 40% and increases permeability .

  • Lung Cancer: CLDN7 stabilizes integrin β1, enhancing cell-matrix adhesion. Suppression leads to tumor spheroid formation in vitro and accelerated growth in vivo .

3.2. Mechanistic Insights

  • Tumor Suppression: Loss of CLDN7 disrupts epithelial integrity, enabling metastasis in breast and colorectal cancers .

  • Pro-Invasive Role: In ovarian cancer, CLDN7 upregulation alters genes linked to cell migration (e.g., MMP9, VEGF) .

4.1. Immunohistochemistry (IHC)

  • FFPE Tissues: EP399 detects CLDN7 in breast, colon, and kidney tissues, with loss observed in poorly differentiated tumors .

  • Quantitative Analysis: Automated platforms use CLDN7 mAbs to score expression levels in biopsy samples .

4.2. Functional Studies

  • siRNA Knockdown: Reduces ovarian cancer cell invasion by 60% .

  • Co-Immunoprecipitation: Confirms CLDN7-integrin β1 interaction in lung cancer models .

4.3. Therapeutic Development

  • Antibody-Drug Conjugates (ADCs): Preclinical trials target CLDN7-overexpressing tumors using MMAE-linked mAbs .

Product Specs

Buffer
Liquid in phosphate-buffered saline (PBS) containing 50% glycerol, 0.5% bovine serum albumin (BSA), and 0.02% sodium azide.
Form
Liquid
Lead Time
Product shipment typically occurs within 1-3 business days of order receipt. Delivery times may vary depending on the purchase method and location. Please contact your local distributor for precise delivery estimates.
Synonyms
CLDN7; CEPTRL2; CPETRL2; Claudin-7; CLDN-7
Target Names
Uniprot No.

Target Background

Function

CLDN7 plays a critical role in the tight junction-mediated obliteration of intercellular space.

Gene References Into Functions

The following studies illustrate the diverse roles of CLDN7 in various physiological and pathological processes:

  • TGF-β1 modulation of esophageal epithelial barrier function via claudin-7 attenuation in eosinophilic esophagitis. (PMID: 28832026)
  • Identification of CLDN7 as a novel candidate gene associated with corneal astigmatism, alongside CLDN7, ACP2, and TNFAIP8L3, through genome-wide association study and gene-based analysis. (PMID: 29422769)
  • Distinct expression profiles of claudin-5, 7, and 8 in gastric carcinoma versus non-neoplastic tissues, highlighting their association with metastasis and patient prognosis. (PMID: 29901188)
  • Claudin-7 as a potential marker for lung cancer identification using digital image cytometry for quantification of immunohistochemical staining. (PMID: 29512497)
  • Influence of cycling hypoxia on CLDN1 and CLDN7 expression in nasopharyngeal cancer cells, impacting P18 expression, cell invasion, and proliferation. (PMID: 28055967)
  • Functional link between EpCAM, HAI-2, matriptase, and claudin-7 in a pathway contributing to disease pathogenesis when dysregulated. (PMID: 28094766)
  • Membrane localization of claudin-1, -3, and -4 in cancer and colon polyps, with novel findings of nuclear mislocalization of claudin-3 in colon cancer and claudin-4 in adenomas. (PMID: 28295005)
  • Suppressive role of CLDN5, 7, and 18 expression on the PDK1-Akt interaction, and its impact on Akt phosphorylation and disordered proliferation in lung squamous carcinoma. (PMID: 27884700)
  • Dysregulated miRNA expression and high claudin-1 levels as potential biomarkers for a basal-like breast cancer subtype. (PMID: 26982264)
  • No significant difference in claudin-7 expression between cervical carcinoma and adjacent non-neoplastic tissues. (PMID: 26464708)
  • Hepatocyte nuclear factor 4α as a regulatory factor binding to the CLDN7 promoter and stimulating its activity in differentiating intestinal epithelial cells. (PMID: 26216285)
  • Claudin-7's role in regulating cell proliferation and epithelial cell attachment via integrin β1 engagement. (PMID: 26081244)
  • Association of the CLDN7 variant rs222857 with adiponectin levels following intensive lifestyle intervention. (PMID: 25759378)
  • CLDN-7 palmitoylation's inhibition of tight junction integration and its promotion of glycolipid-enriched membrane domain recruitment, supporting motility and invasion. (PMID: 26054340)
  • Association of increased claudin-1 expression with high pathologic grade, perineural invasion, vascular invasion, nodal metastasis, and advanced clinical stage in oral squamous cell carcinoma. (PMID: 25078758)
  • Decreased anchorage-independent growth in claudin-7 knockdown cells. (PMID: 25514462)
  • Role of claudin-7 loss in potentiating epithelial-to-mesenchymal transition and promoting colon cancer progression in a Rab25-dependent manner. (PMID: 25500541)
  • Correlation between increased claudin-7 expression and decreased survival in nasopharyngeal carcinoma. (PMID: 25778318)
  • Association of increased claudin-7 expression with liver cirrhosis and hepatocellular carcinoma. (PMID: 24696415)
  • Potential involvement of EpCAM, CD44v6, claudin-7, and ALDH1 in the aggressive phenotype of anaplastic thyroid carcinoma. (PMID: 24727741)
  • Claudin-1's role in promoting colon tumorigenesis, highlighting the dysregulated antigen-tumor interaction and inflammation. (PMID: 24997475)
  • Expression of claudin-3 and -7 in a subset of gastric cancer tissues and reduced claudin-18 expression in a majority of cases. (PMID: 24333468)
  • Aberrant claudin-7, α-catenin, and β-catenin expression/localization as potential markers for distinguishing localized from metastatic prostate cancer. (PMID: 24358122)
  • Association of the CLDN7 rs4562 genotype with lymph node involvement and tumor differentiation. (PMID: 24479816)
  • Lack of claudin-7 expression in tumor centers as a potential indicator of increased risk for regional recurrence. (PMID: 23953778)
  • Phosphorylation of claudin-7 at serine 204 by protein kinase C. (PMID: 23433123)
  • Trend toward better prognosis in hepatocellular carcinoma patients with claudin-7 overexpression. (PMID: 23146509)
  • Anti-apoptotic role of increased claudin-1 expression in TNF-α-induced apoptosis. (PMID: 22941467)
  • Association of CD24+ and claudin-7 positivity with reduced recurrence time in breast cancer. (PMID: 21956537)
  • Marked reduction of claudin-7 and tricellulin at all stages of tumor development, with no correlation between HPV infection and altered tight junction protein expression. (PMID: 21480761)
  • Significant upregulation of claudin-7 in epithelial ovarian cancer. (PMID: 21789222)
  • Down-regulation of Claudin-7 and overexpression of Slug in lung squamous cell carcinoma and adenocarcinoma. (PMID: 21645451)
  • Inhibition of cell migration and invasion by claudin-7 through ERK/MAPK signaling in human lung cancer cells. (PMID: 21641901)
  • Claudin-7 gene transcriptional activity not a useful marker for laryngeal tumors. (PMID: 21193919)
  • Early decrease in Claudin-7 mRNA levels during colorectal carcinogenesis, contributing to compromised epithelial barrier in adenomas. (PMID: 21310043)
  • Claudin-7 down-regulation as an important feature in oral squamous cell carcinoma. (PMID: 21083599)
  • Regulation of claudin-1 localization at the plasma membrane by proteasomes, with changes to Rab5a-mediated endosomal localization upon proteasomal inhibition. (PMID: 20926780)
  • Enhanced tumorigenic properties in a gastric adenocarcinoma cell line upon increased expression of claudin-6, 7, or 9. (PMID: 20874001)
  • Close relationship between claudins 6, 7, and 9 expression and gastric carcinogenesis. (PMID: 19960275)
  • Claudin-1 expression in all cases of Epstein-Barr virus-associated nasopharyngeal carcinoma studied. (PMID: 20204275)
  • Correlation of claudin-7 loss with histological grade in breast ductal carcinoma in situ and invasive ductal carcinoma, suggesting a role in cancer progression and dissemination. (PMID: 12673207)
  • Regulation of prostate-specific antigen expression in LNCaP cells by two claudin-7 isoforms (full-length and C-terminal truncated). (PMID: 14502431)
  • Strong prediction of disease recurrence and poor survival in stage II colon cancer based on claudin-1 expression loss. (PMID: 15475928)
  • Potential role of claudin-1 and -7 in cervical neoplasia tumor progression and as markers for malignant transformation. (PMID: 15790437)
  • Association of claudin-7 overexpression with gastric tumorigenesis. (PMID: 16049341)
  • Critical role of ELF3-induced claudin-7 expression in epithelial structure formation in biphasic synovial sarcoma. (PMID: 17060315)
  • Significant differences in CLDN7 expression between adenocarcinomas and squamous cell carcinomas. (PMID: 17418912)
  • Role of claudin-7-associated EpCAM in metastasis formation through complex formation with CO-029 and CD44v6 in tetraspanin-enriched membrane microdomains. (PMID: 17579117)
  • Changes in claudin-7 expression during gastric carcinogenesis and its implications in cancer characteristics. (PMID: 17611659)
  • Claudin-7 as a potential expression marker to differentiate chromophobe renal cell carcinoma from oncocytoma. (PMID: 17922590)
Database Links

HGNC: 2049

OMIM: 609131

KEGG: hsa:1366

STRING: 9606.ENSP00000353475

UniGene: Hs.513915

Protein Families
Claudin family
Subcellular Location
Cell membrane; Multi-pass membrane protein. Basolateral cell membrane. Cell junction, tight junction.
Tissue Specificity
Expressed in kidney, lung and prostate. Isoform 1 seems to be predominant, except in some normal prostate samples, where isoform 2 is the major form. Down-regulated in breast cancers, including ductal carcinoma in situ (DCIS), lobular carcinoma in situ (L

Q&A

What are the standard methods for detecting CLDN7 expression in tissue samples?

Detection of CLDN7 in tissue samples typically employs multiple complementary techniques to ensure reliable results. Immunohistochemistry (IHC) represents the primary approach for visualizing CLDN7 expression in formalin-fixed paraffin-embedded (FFPE) or frozen tissue sections. When properly optimized, IHC can reveal the cellular localization pattern of CLDN7, which typically appears cytoplasmic, nuclear, or membranous depending on the cellular context .

For quantitative assessment, enzyme-linked immunosorbent assay (ELISA) provides a sensitive method for measuring CLDN7 levels in patient samples and experimental contexts. The procedure involves using a double-antibody sandwich ELISA system with anti-CLDN7 antibodies, followed by detection with biotinylated secondary antibodies and streptavidin-HRP complexes . Western blotting offers another quantitative approach, requiring tissue homogenization with protease inhibitors, protein extraction, SDS-PAGE separation, and transfer to nitrocellulose membranes before probing with anti-CLDN7 primary antibodies and appropriate secondary antibodies .

For research requiring higher sensitivity, especially when detecting low CLDN7 expression, PCR-based methods may complement protein-level analyses. Each method has specific preparation requirements and controls that should be included to ensure validity of results.

How should researchers optimize immunohistochemistry protocols for CLDN7 detection?

Optimizing immunohistochemistry for CLDN7 detection requires systematic adjustment of multiple parameters. Begin with antigen retrieval optimization, testing both heat-induced epitope retrieval (HIER) methods with citrate buffer (pH 6.0) and EDTA buffer (pH 9.0) to determine which provides optimal signal-to-noise ratio for your specific tissue type. The choice between these buffers can significantly impact antibody binding efficiency to the extracellular domains of CLDN7.

Antibody concentration requires careful titration, typically starting with 1:100 to 1:1000 dilutions of commercial CLDN7 monoclonal antibodies. The optimal concentration will produce clear membrane staining in positive control tissues (breast, colon, fallopian tube, pancreas, kidney, or transitional cell carcinoma ) while minimizing background.

Incubation conditions significantly influence results—compare overnight incubation at 4°C versus 1-2 hours at room temperature to determine optimal conditions for your specific antibody. Additionally, a blocking step using 3-5% BSA or serum from the species of the secondary antibody is essential to reduce non-specific binding.

For detection systems, compare the sensitivity of standard avidin-biotin complex (ABC) methods versus polymer-based detection systems, as the latter often provides superior results for membrane proteins like CLDN7. Always include appropriate positive and negative controls, including tissues known to express CLDN7 and serial sections with primary antibody omitted.

What controls should be included when using CLDN7 antibodies in experimental settings?

Rigorous control selection is critical for experiments employing CLDN7 antibodies. Tissue positive controls should include breast, colon, fallopian tube, pancreas, kidney, or transitional cell carcinoma samples, which naturally express CLDN7 at detectable levels . If working with cell lines, include both high-expressing (like certain breast cancer lines) and low-expressing models.

Negative controls should be multi-tiered: (1) technical negative controls where primary antibody is omitted but all other steps remain identical; (2) isotype controls using non-specific IgG of the same isotype as the CLDN7 antibody to assess non-specific binding; and (3) biological negative controls using CLDN7-deficient cell lines or tissues with known low expression.

For advanced applications, consider including CLDN7 knockdown/knockout controls generated via siRNA or CRISPR-Cas9 methods. These genetic controls provide the most stringent validation of antibody specificity. Additionally, peptide competition assays, where the antibody is pre-incubated with purified CLDN7 peptide before application to samples, can confirm binding specificity.

How can researchers distinguish between technical artifacts and true biological variation when analyzing CLDN7 expression patterns across cancer subtypes?

Distinguishing between technical artifacts and genuine biological variation in CLDN7 expression requires implementation of a multi-faceted validation strategy. First, researchers should employ standardized tissue processing protocols across all samples being compared, as variations in fixation time, processing methods, or storage conditions can significantly alter antibody performance with membrane proteins like CLDN7.

Cross-platform validation is essential—confirm IHC findings with quantitative methods like Western blotting or ELISA to ensure consistency across detection platforms . For serum CLDN7 detection, be aware that levels can be extremely low in some cancer patients (below 0.01 pg/mL), necessitating highly sensitive detection methods and careful statistical analysis incorporating these low values .

When analyzing differential expression across cancer subtypes, implement tissue microarrays containing multiple samples from each subtype processed simultaneously to minimize batch effects. Additionally, use receiver operating characteristic (ROC) curve analysis to establish optimal cut-off values for defining "high" versus "low" CLDN7 expression within your specific experimental context .

For advanced studies, single-cell methods can help determine whether observed variations reflect true biological heterogeneity within tumor populations rather than technical variability. Finally, correlation with clinical parameters and other molecular markers can help validate the biological relevance of observed CLDN7 expression patterns.

What methodological approaches allow for studying the functional consequences of CLDN7 dysregulation in cancer models?

Investigating functional consequences of CLDN7 dysregulation requires sophisticated experimental designs spanning in vitro and in vivo systems. Begin with generating stable cell line models with manipulated CLDN7 expression: use lentiviral or CRISPR-Cas9 systems to create CLDN7 knockdown/knockout lines from high-expressing cells, and overexpression models from low-expressing backgrounds. Validate expression changes via Western blot and immunofluorescence to confirm proper localization.

For tight junction integrity assessment, transepithelial/transendothelial electrical resistance (TEER) measurements provide quantitative data on barrier function, while paracellular permeability assays using fluorescent tracers can reveal functional changes in cellular barriers. These functional assays should be complemented with co-immunoprecipitation studies to identify protein-protein interactions between CLDN7 and other junction components or signaling molecules.

Migration and invasion phenotypes can be assessed through wound healing assays, transwell migration/invasion chambers, and 3D spheroid invasion assays. For signaling pathway analysis, examine integrin signaling networks and epithelial-to-mesenchymal transition markers that may be affected by CLDN7 levels .

In vivo studies should employ orthotopic xenograft models using the engineered cell lines, with particular attention to metastatic potential. Patient-derived xenografts (PDXs) with varying CLDN7 expression provide more clinically relevant models. Therapeutic testing might include evaluating response to standard chemotherapeutics in the context of varying CLDN7 levels, as altered tight junctions may affect drug penetration and efficacy.

What considerations are important when developing new monoclonal antibodies against CLDN7?

Developing effective monoclonal antibodies against CLDN7 requires careful consideration of multiple factors. The immunization strategy is critical—traditional approaches using purified protein may be less effective than cell-based immunization using CLDN7-overexpressing cells that present the protein in its native conformation . This approach preserves the natural epitopes, particularly within the extracellular loops that are accessible for therapeutic targeting.

For hybridoma screening, implement a comprehensive selection strategy using both CLDN7-positive and CLDN7-negative cells to identify clones with high specificity . Include testing against related claudin family members (particularly CLDN3, CLDN4, and CLDN1) to assess cross-reactivity, as the claudin family shares high sequence homology in certain domains .

Epitope mapping is essential—determine whether your antibodies recognize the extracellular loop 1 (EL1), extracellular loop 2 (EL2), or intracellular domains of CLDN7. This information is critical for applications like blocking experiments or therapeutic development. The high sequence homology between claudin family members in the EL1 domain (often >90% between closely related members) necessitates careful design to achieve specificity .

For therapeutic potential assessment, evaluate both antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) in relevant cell models expressing CLDN7 . Additionally, assess internalization kinetics if the antibody is intended for antibody-drug conjugate development.

How can contradictory findings regarding CLDN7 expression in different cancer types be reconciled methodologically?

Reconciling contradictory findings about CLDN7 expression requires systematic methodological approaches. First, standardize antibody validation across studies—implement a validation panel including Western blot, flow cytometry, and immunoprecipitation to confirm specificity before comparative analyses. When possible, use the same validated antibody clone across different cancer types to eliminate clone-specific variations.

Consider context-dependent post-translational modifications that might affect antibody recognition. CLDN7 undergoes phosphorylation and palmitoylation that can influence both its localization and antibody binding characteristics. Employ phospho-specific antibodies or depalmitoylation treatments to assess whether these modifications explain apparent contradictions.

The apparent paradox that CLDN7 is downregulated in some cancers (colorectal, lung, breast) but overexpressed in others (ovarian) may reflect tissue-specific functional roles . Implement transcriptional analysis alongside protein detection to determine whether discrepancies occur at mRNA or protein levels. Additionally, analyze subcellular localization patterns—CLDN7 may maintain normal expression levels but display aberrant localization away from tight junctions in certain cancers.

Establish quantitative thresholds appropriate for each cancer type rather than applying universal cutoffs. For instance, the diagnostic significance of serum CLDN7 levels in colorectal cancer was most evident when accounting for cases with extremely low levels that were nearly undetectable by standard ELISA methods .

What are the most effective protocols for using CLDN7 monoclonal antibodies in flow cytometry applications?

Optimizing CLDN7 antibody use for flow cytometry requires addressing the challenges associated with membrane protein detection. Begin with proper sample preparation—use gentle cell dissociation methods (like Accutase or non-enzymatic dissociation buffers) rather than harsh trypsinization which can cleave extracellular domains of membrane proteins like CLDN7.

For fixation and permeabilization, compare paraformaldehyde (1-4%) fixation followed by detergent permeabilization (0.1-0.5% saponin or Triton X-100) against commercially available fix/perm kits designed for membrane proteins. The optimal protocol will preserve CLDN7 epitopes while allowing antibody access to all cellular compartments. Since CLDN7 localization can be cytoplasmic, nuclear, or membranous , permeabilization is often necessary for comprehensive detection.

Antibody titration is essential—test concentrations between 0.1-10 μg/mL to determine the optimal signal-to-noise ratio specific to your cell type. Include blocking steps with 2-5% serum or BSA to reduce non-specific binding. For multi-parameter analyses, select fluorophores with minimal spectral overlap with other markers in your panel, and include compensation controls.

Critical controls include: unstained cells, isotype controls, FMO (fluorescence minus one) controls, and both positive control cells (known CLDN7 expressors) and negative control cells (CLDN7-deficient). For quantitative applications, include calibration beads to convert fluorescence intensity to absolute molecule numbers.

What are common pitfalls when interpreting CLDN7 localization in cancer tissues?

Fixation artifacts frequently confound analysis—overfixation can mask antigens while underfixation may allow protein translocation during processing. Standardize fixation protocols (typically 10% neutral buffered formalin for 24 hours) and validate with multiple fixation conditions when establishing new protocols.

The heterogeneity of CLDN7 expression within tumors represents another challenge—focal expression patterns may be missed in small biopsies or tissue microarrays. Analyze multiple regions when possible and consider quantifying the percentage of positive cells rather than using simple positive/negative designations.

When comparing different studies, be aware that various antibody clones may recognize different CLDN7 epitopes, potentially leading to discrepant results. Additionally, verify that nuclear CLDN7 staining represents genuine nuclear localization rather than cross-reactivity with other nuclear proteins by confirming with alternative detection methods or antibody clones.

How should researchers design experiments to investigate the relationship between CLDN7 expression and metastatic potential?

Investigating CLDN7's relationship with metastatic potential requires a carefully structured experimental design spanning in vitro, in vivo, and clinical analyses. Begin with a comprehensive cell line panel representing primary tumors and matched metastatic derivatives from the same patients. Quantify CLDN7 expression in these paired models using multiple methods (Western blot, qPCR, immunofluorescence) and correlate expression with established metastatic phenotypes.

In vitro functional assays should include:

  • Invasion assays using Matrigel-coated transwells

  • 3D organoid models to assess invasive projections

  • Cell-matrix adhesion assays to evaluate interactions with extracellular matrix components

  • Assessment of epithelial-mesenchymal transition markers

For in vivo metastasis models, implement both experimental and spontaneous metastasis approaches. Experimental metastasis involves tail-vein injection of cells with manipulated CLDN7 levels to assess colonization efficiency. Spontaneous metastasis models use orthotopic implantation and allow tumors to progress naturally, providing insights into the entire metastatic cascade.

In clinical samples, design a tissue microarray containing matched primary tumors and metastatic lesions from the same patients. Quantify CLDN7 expression and correlate with clinical outcomes including time to metastasis, metastatic site tropism, and post-metastasis survival. Multivariate analyses should adjust for known prognostic factors to isolate CLDN7's independent contribution to metastatic potential.

What experimental approaches can help resolve antibody specificity issues when working with closely related claudin family members?

Resolving antibody specificity issues with claudin family members requires systematic validation strategies. Begin with sequence-based epitope analysis—align the amino acid sequences of CLDN7 with other claudins (particularly CLDN1, CLDN3, and CLDN4) to identify regions of high homology versus unique segments. This information guides epitope selection for antibody generation or commercial antibody selection.

For experimental validation, implement a cell line panel overexpressing individual claudin family members. Transfect expression vectors for CLDN1, CLDN3, CLDN4, CLDN5, CLDN6, CLDN7, and CLDN9 into a claudin-negative cell line, then test antibody binding to each transfectant by flow cytometry and Western blot . This approach directly assesses cross-reactivity with related claudins.

Peptide competition assays provide another validation layer—pre-incubate the antibody with synthetic peptides corresponding to the presumed epitope from CLDN7 and related claudins. If signal diminishes only with the CLDN7 peptide, this confirms specificity. For monoclonal antibodies recognizing conformational epitopes, implement immunoprecipitation followed by mass spectrometry to identify all proteins captured by the antibody.

Knockout/knockdown models provide the most stringent validation—compare antibody reactivity in wild-type versus CLDN7-knockout cells. Ideally, implement CRISPR-Cas9 knockout of CLDN7 in a naturally expressing cell line rather than relying solely on overexpression systems that may not recapitulate natural epitope presentation .

What statistical approaches are most appropriate for analyzing CLDN7 expression data in patient cohorts?

Analyzing CLDN7 expression in patient cohorts requires sophisticated statistical approaches tailored to the data type and research questions. For immunohistochemistry scoring, implement standardized quantification methods—either H-score (intensity × percentage of positive cells), Allred score, or digital pathology quantification with machine learning algorithms for objective assessment. Establish inter-observer concordance between multiple pathologists to ensure scoring reliability.

When analyzing serum CLDN7 levels by ELISA, be aware that some cancer patients may have values below detection limits. Statistical approaches must properly handle these cases—either by setting them to the lower detection limit (e.g., 0.01 pg/mL) or using specialized statistical methods for left-censored data . Receiver operating characteristic (ROC) curve analysis should be used to determine optimal diagnostic cutoff values, with area under the curve (AUC) calculations to assess diagnostic performance .

For correlating CLDN7 with other biomarkers, use appropriate correlation coefficients (Pearson for normally distributed continuous data, Spearman for non-parametric data) and correct for multiple comparisons using methods like Benjamini-Hochberg procedure.

How can CLDN7 monoclonal antibodies be effectively utilized in therapeutic development?

Utilizing CLDN7 monoclonal antibodies for therapeutic development requires a comprehensive development pipeline addressing multiple aspects of antibody functionality. Begin with epitope selection—antibodies targeting the extracellular loops (particularly EL1) of CLDN7 have the greatest therapeutic potential as these domains are accessible in intact cells . The antibody generation strategy should employ cell-based immunization with CLDN7-overexpressing cells to present the protein in its native conformation, similar to approaches used for other claudin family antibodies .

Functional screening should assess both direct and immune-mediated mechanisms. Direct mechanisms include the antibody's ability to disrupt tight junction formation or induce conformational changes in CLDN7. Immune-mediated effects should evaluate antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) against CLDN7-expressing cancer cells . Creating mouse-human chimeric antibodies (with human IgG1 Fc domains) can enhance these effector functions for translational development .

For antibody-drug conjugate (ADC) development, assess internalization kinetics using fluorescently-labeled antibodies and live-cell imaging. Optimal ADC candidates demonstrate rapid internalization upon CLDN7 binding. Additionally, evaluate the impact of CLDN7 expression heterogeneity on potential therapeutic efficacy using patient-derived samples representing the target cancer types.

Pre-clinical evaluation should include both cell line-derived xenograft models and patient-derived xenografts with varying CLDN7 expression levels to determine the relationship between expression and response. Pharmacokinetic studies must assess tissue penetration, particularly into solid tumors with complex microenvironments.

What methodologies can detect dynamic changes in CLDN7 expression during cancer progression or treatment?

Detecting dynamic CLDN7 expression changes requires implementing complementary methodologies across multiple time points. For clinical samples, design longitudinal collection protocols obtaining matched biopsies at diagnosis, during treatment, and at progression/recurrence. Implement consistent processing protocols across all time points to minimize technical variation.

For real-time monitoring in experimental models, develop reporter systems—create CLDN7 promoter-driven fluorescent or luminescent reporter cell lines that allow non-invasive tracking of CLDN7 transcriptional activity. These reporters can be incorporated into in vitro drug screening assays or in vivo models using window chambers or whole-animal imaging to monitor expression changes in response to treatments.

Single-cell RNA-sequencing provides powerful insights into CLDN7 expression heterogeneity and dynamic changes within tumor populations that bulk analyses might miss. This approach can identify specific cellular subpopulations where CLDN7 expression changes most dramatically during disease progression or treatment.

For liquid biopsy applications, optimize methods for detecting circulating tumor cells (CTCs) expressing CLDN7 or quantifying CLDN7 protein in plasma samples . Highly sensitive ELISA methods capable of detecting low pg/mL concentrations are necessary, as serum CLDN7 levels may be extremely low in some cancer patients .

Finally, multiplexed imaging approaches combining CLDN7 detection with markers of tumor progression (e.g., proliferation, invasion, or stemness markers) can reveal the contextual changes in CLDN7 expression relative to other cancer hallmarks. Technologies like cyclic immunofluorescence, imaging mass cytometry, or spatial transcriptomics provide comprehensive spatial context for these dynamic changes.

How can researchers design high-throughput screening assays to identify compounds that modulate CLDN7 function?

Designing high-throughput screening (HTS) assays for CLDN7 modulation requires functional readouts that reflect CLDN7's biological roles. Begin with cell line engineering—create paired isogenic cell lines with and without CLDN7 expression, ideally in multiple cancer contexts, to serve as screening platforms. For reporter-based primary screens, integrate luciferase or fluorescent proteins under control of CLDN7-responsive promoters or design FRET-based sensors that detect CLDN7 protein-protein interactions.

Functional screening assays should focus on tight junction integrity—measure transepithelial/transendothelial electrical resistance (TEER) in cell monolayers using electrode arrays compatible with 96- or 384-well formats. Alternatively, implement high-content imaging assays tracking paracellular permeability of fluorescent tracers in real-time. These assays can be miniaturized and automated for HTS applications.

For targeting CLDN7 in cancer contexts, design phenotypic assays measuring invasion through extracellular matrix barriers, cell-matrix adhesion, or anoikis resistance. These endpoints reflect key processes influenced by CLDN7 in cancer progression. Consider multiplexed assays that simultaneously measure CLDN7 expression levels and relevant phenotypic outcomes.

Counter-screens must be implemented to eliminate non-specific hits—test candidate compounds against other claudin family members, particularly those with high sequence homology to CLDN7. Additionally, cytotoxicity assays should distinguish between specific CLDN7 modulation and general cytotoxic effects.

For hit validation, implement dose-response analyses across multiple cell models with varying CLDN7 expression levels. Secondary assays should confirm target engagement through direct binding assays, thermal shift assays, or cellular thermal shift assays (CETSA) specific to CLDN7 protein.

What are the most rigorous approaches for validating CLDN7 as a biomarker in multicenter clinical studies?

Validating CLDN7 as a clinical biomarker requires a systematic approach following the REMARK (REporting recommendations for tumor MARKer prognostic studies) guidelines. Begin with analytical validation—establish standardized protocols for CLDN7 detection across participating centers, including pre-analytical variables (sample collection, processing, storage), analytical methods (antibody clone, detection system, scoring criteria), and quality control measures. Implement centralized training and proficiency testing for technicians and pathologists.

For biospecimen considerations, design prospective collection protocols with appropriate consent for biomarker studies. When using archival tissues, document pre-analytical variables that might affect CLDN7 detection. Create centralized biospecimen repositories with standardized processing to minimize batch effects between centers.

Statistical validation requires predetermined analytical plans with clearly defined endpoints. Power calculations should determine minimum sample sizes needed to detect clinically meaningful differences in outcomes between CLDN7-positive and CLDN7-negative patients. Implement internal cross-validation (training/validation cohorts) within multicenter studies before advancing to independent external validation.

Reproducibility assessment is crucial—conduct ring studies where identical sample sets are analyzed across multiple participating laboratories to quantify inter-laboratory variation. Additionally, evaluate inter-observer and intra-observer concordance in CLDN7 assessment through blinded repeat scoring by multiple pathologists.

Contextual validation should assess CLDN7's performance within multimarker panels rather than in isolation. This requires sophisticated statistical approaches like decision tree analysis or multivariate models that can determine CLDN7's incremental value beyond established biomarkers and clinicopathological variables.

What emerging technologies will advance CLDN7 monoclonal antibody research in the next decade?

The next decade of CLDN7 monoclonal antibody research will likely be transformed by several emerging technologies. Antibody engineering approaches will become increasingly sophisticated, with computational design methods enabling the creation of highly selective antibodies that can distinguish between closely related claudin family members despite high sequence homology. Structure-guided antibody design, informed by recent advances in structural biology techniques like cryo-EM for membrane proteins, will allow targeting of specific functional epitopes within CLDN7's extracellular domains.

Single-cell technologies will revolutionize our understanding of CLDN7 heterogeneity in tumors. Single-cell proteomics methods capable of measuring CLDN7 alongside hundreds of other proteins at single-cell resolution will reveal previously unappreciated subpopulations and functional states. These approaches, combined with spatial transcriptomics and multiplexed imaging, will map CLDN7 expression within the complex tumor microenvironment context.

For therapeutic applications, novel antibody formats beyond traditional IgG—including bispecific antibodies targeting CLDN7 and immune cells, antibody fragments with enhanced tumor penetration, and antibody-drug conjugates with novel payload mechanisms—will expand the therapeutic potential. Additionally, intrabodies (intracellular antibodies) designed to modulate CLDN7 function within cancer cells may become feasible as delivery technologies improve.

In translational research, liquid biopsy technologies will advance to permit reliable monitoring of circulating CLDN7 protein or CLDN7-expressing circulating tumor cells as minimally invasive biomarkers. The integration of artificial intelligence approaches for image analysis will standardize CLDN7 assessment in tissue samples, reducing inter-observer variability and enhancing reproducibility across clinical studies.

How can researchers integrate CLDN7 studies with broader cancer biology frameworks?

Integrating CLDN7 research into broader cancer biology frameworks requires multidisciplinary approaches connecting tight junction biology with established cancer hallmarks. Systems biology approaches should position CLDN7 within protein-protein interaction networks and signaling pathways, particularly focusing on its connections to integrin signaling, epithelial-to-mesenchymal transition pathways, and cell adhesion complexes . Network analyses can reveal how CLDN7 alterations propagate effects throughout cellular systems.

Multi-omics integration will be essential—combine transcriptomic data on CLDN7 expression with proteomic analyses of tight junction complexes, phosphoproteomic studies of downstream signaling events, and metabolomic consequences of altered cellular barrier function. These integrated datasets can identify mechanistic links between CLDN7 status and broader cancer phenotypes.

For clinical translation, incorporate CLDN7 assessment into existing molecular classification systems for cancers. Determine whether CLDN7 status adds value to established molecular subtypes in breast cancer (luminal, HER2-enriched, basal-like) or colorectal cancer (CMS1-4). This requires large-scale retrospective studies of existing cohorts with comprehensive molecular characterization.

Therapeutic strategies should consider CLDN7 in the context of precision oncology frameworks—identify synthetic lethal interactions where CLDN7 status sensitizes tumors to specific targeted agents or immune therapies. This approach requires systematic combination screening in models with varying CLDN7 expression.

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.