CYCJ18 Antibody

Shipped with Ice Packs
In Stock

Description

Overview of Cytokeratin 18 (CK18) Antibodies

Cytokeratin 18 (CK18) is a type I intermediate filament protein encoded by the KRT18 gene. It pairs with KRT8 to form heteropolymers that maintain epithelial cell integrity and resist mechanical stress . CK18 antibodies target specific epitopes of this protein for diagnostic and research purposes.

Key Features of CK18 Antibodies:

  • Cellular Localization: Cytoplasmic, expressed in simple epithelial tissues (e.g., liver, gastrointestinal tract) .

  • Reactivity: Human-specific; used to identify epithelial malignancies (e.g., adenocarcinomas) .

  • Clinical Relevance: Detects apoptosis-related fragmented CK18 (fCK18) in serum, a biomarker for liver diseases like non-alcoholic steatohepatitis (NASH) .

Monoclonal Antibody Generation

Recent studies have developed high-affinity CK18 monoclonal antibodies (mAbs) using advanced platforms:

Antibody CloneEpitope SpecificityAffinity (KD)Application
K18-624 C-terminal fCK18 (381R–397D)0.056 ng/mL (LoQ)CLEIA for fCK18 detection
K18-328 Mid-region CK18 (323S–340G)N/APaired with K18-624 in CLEIA
MSVA-118R Full-length CK18N/AImmunohistochemistry (IHC)
  • K18-624/K18-328: These mAbs enable a chemiluminescent enzyme immunoassay (CLEIA) with 7x higher sensitivity than commercial kits, detecting fCK18 at 0.056 ng/mL .

  • Next-Generation Sequencing (NGS): Identified six CK18-specific mAbs with the most frequent CDR3 sequences. One candidate showed a binding affinity of 9.424×1010 M9.424 \times 10^{-10}\ \text{M} .

Clinical Applications

  • Liver Disease: Serum fCK18 levels are elevated in NASH patients (p<0.01p < 0.01 vs. healthy controls) .

  • Pulmonary Fibrosis: Anti-CK18 antibody complexes are significantly higher in idiopathic pulmonary fibrosis (IPF) patients (0.81±0.310.81 \pm 0.31 vs. 0.45±0.060.45 \pm 0.06 in controls) .

  • Cancer Diagnostics: CK18 antibodies (e.g., clone DC10) distinguish epithelial tumors (e.g., pancreatic, ovarian) from non-epithelial malignancies (e.g., melanoma) .

  • Superior precision (CV <10%) and recovery rates.

  • Detects fCK18 in healthy individuals, enabling early disease monitoring .

Challenges and Future Directions

  • Epitope Specificity: Commercial antibodies often target poorly defined regions (e.g., 284T–396D), whereas engineered mAbs like K18-624 improve specificity .

  • Biomarker Validation: Large-scale studies are needed to standardize fCK18 thresholds for NASH and IPF .

Product Specs

Buffer
Preservative: 0.03% Proclin 300
Constituents: 50% Glycerol, 0.01M Phosphate Buffered Saline (PBS), pH 7.4
Form
Liquid
Lead Time
Made-to-order (14-16 weeks)
Synonyms
CYCJ18 antibody; At2g01905 antibody; T23K3Cyclin-J18 antibody
Target Names
CYCJ18
Uniprot No.

Q&A

What is the difference between Claudin 18.1 and Claudin 18.2 antibodies?

Claudin 18.1 and Claudin 18.2 are isoforms of the Claudin 18 tight junction protein with distinct tissue expression patterns and research applications. Claudin 18.1 is primarily expressed in lung tissue, while Claudin 18.2 demonstrates high specificity for gastric epithelial cells and is abnormally activated in gastric cancer . When selecting an antibody, researchers must ensure they target the correct isoform for their specific tissue of interest. For instance, HUABIO manufactures a Claudin 18.1 Rabbit Polyclonal Antibody suitable for Western blotting and flow cytometry applications with reactivity to both human and mouse samples .

What applications are Claudin 18 antibodies most commonly used for?

Claudin 18 antibodies are utilized across multiple research applications, with varying optimization levels for each technique:

ApplicationOptimization LevelCommon Antibody Types
Flow CytometryHighMonoclonal, conjugated
Western BlottingHighPolyclonal, monoclonal
ImmunohistochemistryMedium-HighMonoclonal
ELISAHighMonoclonal
ImmunoprecipitationMediumPolyclonal, monoclonal

Flow cytometry applications typically use conjugated antibodies (such as FITC-labeled anti-Claudin 18 antibodies) for direct detection without secondary antibodies . For detecting low-abundance targets, researchers often employ polyclonal antibodies due to their ability to recognize multiple epitopes of the same antigen .

How do I select the appropriate Claudin 18 antibody for my research?

Selection of the appropriate Claudin 18 antibody depends on several key factors:

  • Clonality: Monoclonal antibodies offer high specificity for a single epitope with minimal batch-to-batch variation, while polyclonal antibodies recognize multiple epitopes and may provide stronger signals for low-abundance targets .

  • Application compatibility: Verify that the antibody has been validated for your specific application (flow cytometry, Western blotting, etc.) through published validation data .

  • Species reactivity: Ensure the antibody reacts with your species of interest (human, mouse, rat, etc.) .

  • Epitope location: For specific isoform detection (Claudin 18.1 vs. Claudin 18.2), confirm the epitope location recognized by the antibody .

  • Conjugation: For flow cytometry, directly conjugated antibodies (FITC, PE, etc.) may simplify protocols, while unconjugated antibodies require secondary detection .

For reproducibility and long-term experimental consistency, recombinant monoclonal antibodies are recommended when available, as they offer secured supply with minimal batch-to-batch variation .

How can I validate the specificity of a Claudin 18.2 antibody for translational research?

Validating antibody specificity for Claudin 18.2 requires a multi-parameter approach:

  • Epitope mapping validation: Confirm the antibody recognizes the specific region of Claudin 18.2 rather than Claudin 18.1 or other claudin family members. For example, the K18-624 monoclonal antibody was validated to recognize an epitope in the 381R-397D region of Claudin 18 .

  • Western blotting with recombinant proteins: Compare reactivity against full-length Claudin 18 protein versus fragmented Claudin 18, as demonstrated in the development of the K18-624 antibody, which showed 8-fold higher reactivity compared to commercially available antibodies .

  • Immunoprecipitation followed by Western blotting (IP-WB): This technique can verify antibody sensitivity and specificity in complex biological samples such as patient serum .

  • Tissue cross-reactivity studies: Evaluate staining patterns across normal tissues where Claudin 18.2 should be minimally expressed versus tissues with known expression .

  • Knockout/knockdown validation: Use Claudin 18-deficient cell lines as negative controls to confirm specificity.

When developing therapeutic antibodies, additional validation through immunohistochemistry on patient samples is essential to confirm appropriate target expression before treatment, as demonstrated in clinical trials of anti-Claudin 18.2 monoclonal antibodies like ASKB589 .

What methodological approaches can improve the sensitivity of Claudin 18 detection in clinical samples?

Several methodological refinements can enhance Claudin 18 detection sensitivity:

  • Chemiluminescent enzyme immunoassay (CLEIA) development: As demonstrated with fragmented cytokeratin 18 (fCK18), developing a CLEIA with optimized antibody pairs can significantly improve detection sensitivity. The study by Nature Scientific Reports achieved a detection sensitivity of 0.056 ng/mL for fCK18, enabling reliable measurement in human serum samples .

  • Antibody pair optimization: Selection of complementary capture and detection antibodies recognizing different epitopes can enhance signal amplification. For instance, using K18-624 as a capture antibody and K18-328 as a detection antibody improved sensitivity compared to commercial options .

  • Signal amplification techniques: Implementing tyramide signal amplification or polymer-based detection systems can enhance sensitivity for immunohistochemistry applications.

  • Recombinant calibrator development: Creating a recombinant protein calibrator, as demonstrated with recombinant fCK18 (rfCK18), provides a reliable standard for quantitative assays .

  • Sample preparation optimization: For serum/plasma samples, optimizing collection, storage conditions, and pre-analytical processing can preserve epitope integrity and improve detection.

These approaches are particularly important when analyzing clinical samples where target proteins may be present at low concentrations, as demonstrated in studies measuring serum biomarkers in patients with non-alcoholic steatohepatitis (NASH) .

What are the mechanisms behind adverse effects observed with therapeutic Claudin 18.2 antibodies?

Therapeutic monoclonal antibodies targeting Claudin 18.2 can induce adverse effects through several mechanisms:

  • Antibody-dependent cellular cytotoxicity (ADCC): Therapeutic antibodies like ASKB589 are designed to induce ADCC, recruiting immune cells to attack tumor cells expressing Claudin 18.2. This immune activation can lead to off-target effects when Claudin 18.2 is expressed at low levels in normal tissues .

  • Complement-dependent cytotoxicity (CDC): Similarly, CDC mechanisms can trigger inflammation and tissue damage through complement activation .

  • Tissue-specific adverse effects: A case report documented severe ascites (abnormal fluid accumulation in the peritoneal cavity) as an adverse effect of anti-Claudin 18.2 antibody treatment for advanced gastric cancer . The ascites gradually developed during therapy and resolved after treatment discontinuation.

  • Persistence of therapeutic effect: Interestingly, in the reported case, even after discontinuation of treatment due to severe ascites, the patient continued to show tumor reduction, achieving near complete response with a progression-free survival of at least 10 months .

The balance between therapeutic efficacy and adverse effects remains a critical consideration, requiring careful patient monitoring and potentially dose adjustment or treatment interruption when severe adverse effects occur. Further mechanistic research is needed to understand the precise pathways leading to specific adverse effects like ascites .

How can I optimize flow cytometry protocols for Claudin 18 detection?

Optimizing flow cytometry for Claudin 18 detection requires attention to several methodological details:

  • Antibody selection: Choose antibodies specifically validated for flow cytometry applications. For example, BPS Bioscience offers FITC-labeled Anti-Claudin-18 Isoform 2 Antibody specifically optimized for flow cytometry .

  • Cell preparation considerations:

    • For cell lines: Ensure complete dissociation into single-cell suspensions

    • For tissue samples: Optimize digestion protocols to preserve epitope integrity

    • For blood samples: Use appropriate lysis buffers to remove red blood cells

  • Fixation and permeabilization optimization:

    • As a tight junction protein, Claudin 18 is membrane-localized, requiring gentle fixation

    • Test different permeabilization reagents to optimize access to epitopes while maintaining cellular integrity

    • Consider comparing commercial fixation/permeabilization kits designed for membrane proteins

  • Titration of antibody concentration: Perform antibody titration experiments to determine optimal concentration, typically starting with manufacturer recommendations and testing 2-fold dilutions.

  • Multiparameter panel design: When including Claudin 18 antibodies in multicolor panels:

    • Select fluorophores based on expression level (brighter fluorophores for low-expression targets)

    • Include appropriate FMO (fluorescence minus one) controls

    • Consider spectral overlap and compensation requirements

  • Validation controls:

    • Positive control: Cell lines with known Claudin 18 expression (e.g., gastric cancer cell lines)

    • Negative control: Cell lines lacking Claudin 18 expression

    • Isotype controls: Matched to the Claudin 18 antibody's host species and isotype

These optimizations are essential for obtaining reliable flow cytometry data, particularly when analyzing clinical samples or rare cell populations expressing Claudin 18.

What are the critical factors in developing a highly sensitive immunoassay for Claudin 18?

Developing a highly sensitive immunoassay for Claudin 18 requires addressing several critical factors:

  • Antibody pair selection: The selection of complementary capture and detection antibodies is crucial:

    • The capture antibody should have high affinity and specificity for Claudin 18

    • The detection antibody should recognize a different epitope to avoid competition

    • For isoform-specific detection, at least one antibody should target unique epitopes of Claudin 18.1 or 18.2

  • Recombinant protein standard development: Creating a well-characterized recombinant protein standard enables accurate quantification:

    • Express the protein in an appropriate system (bacterial, mammalian)

    • Purify to high homogeneity

    • Verify by mass spectrometry and other quality control methods

    • Create a standard curve covering the physiological concentration range

  • Assay format optimization:

    • Signal amplification strategy (e.g., chemiluminescence vs. colorimetric)

    • Incubation times and temperatures

    • Blocking reagents to minimize background

    • Sample dilution protocols

  • Analytical validation parameters:

    • Detection limit: The lowest concentration that can be reliably distinguished from background

    • Within-run and between-day coefficients of variation (CVs) should be <10%

    • Recovery rates within 15% of expected values

    • Linearity across the measurement range

This approach has been successfully applied to other proteins, as demonstrated in the development of a highly sensitive CLEIA for fragmented cytokeratin 18, achieving a detection sensitivity of 0.056 ng/mL with CVs below 10% and recoveries within 15% .

How can I differentiate between Claudin 18 isoforms in experimental samples?

Differentiating between Claudin 18 isoforms (particularly 18.1 and 18.2) requires specific methodological approaches:

  • Isoform-specific antibody selection:

    • Identify antibodies targeting unique epitopes in each isoform

    • Verify specificity through Western blotting against recombinant proteins representing each isoform

    • For example, BPS Bioscience offers Anti-Claudin-18 Isoform 2 Antibody specifically targeting Claudin 18.2

  • PCR-based detection:

    • Design primers spanning unique exon junctions or sequences of each isoform

    • Perform RT-PCR or qPCR to quantify isoform-specific mRNA expression

    • Include appropriate controls (tissues known to express specific isoforms)

  • Mass spectrometry approaches:

    • Identify unique peptide sequences for each isoform

    • Develop selective reaction monitoring (SRM) or parallel reaction monitoring (PRM) methods

    • Use isotopically labeled peptide standards for absolute quantification

  • Sequential immunoprecipitation:

    • First immunoprecipitate with pan-Claudin 18 antibody

    • Perform subsequent immunoprecipitations with isoform-specific antibodies

    • Analyze by Western blotting to confirm isoform identity

  • Tissue expression pattern analysis:

    • Claudin 18.1 is predominantly expressed in lung tissue

    • Claudin 18.2 is predominantly expressed in gastric epithelial cells

    • Compare expression patterns to reference tissues to infer isoform identity

These approaches can be combined for increased confidence in isoform identification, particularly important in cancer research where Claudin 18.2 is being explored as a therapeutic target in gastric and pancreatic cancers .

What are the emerging applications of Claudin 18.2 antibodies in cancer research?

Claudin 18.2 antibodies are being increasingly utilized in cancer research, with several emerging applications:

These applications highlight the translational potential of Claudin 18.2 antibodies, particularly for patients with gastric cancer who lack other targetable biomarkers (HER2-negative, low PD-L1 expression) .

How can I troubleshoot inconsistent results when using Claudin 18 antibodies in research?

Troubleshooting inconsistent results with Claudin 18 antibodies requires systematic evaluation of several factors:

  • Antibody quality and specificity issues:

    • Verify antibody validation data from manufacturer

    • Consider switching to recombinant monoclonal antibodies for better consistency

    • Compare results from different antibody clones/suppliers

    • For example, when investigating fragmented CK18, researchers found that new monoclonal antibodies (K18-624) showed 8-fold higher reactivity compared to commercial antibodies

  • Protocol optimization by application:

    ApplicationCommon IssuesTroubleshooting Approaches
    Flow CytometryCell clumping, poor fixationOptimize cell preparation, titrate antibody
    Western BlottingWeak signal, multiple bandsAdjust lysis buffer, blocking conditions
    IHC/ICCBackground staining, weak signalOptimize antigen retrieval, detection system
    ELISAMatrix effects, hook effectTest different sample dilutions, blocking reagents
  • Sample preparation considerations:

    • Protein degradation during storage/handling

    • Inefficient extraction of membrane proteins

    • Epitope masking during fixation

    • For tight junction proteins like Claudin 18, use specialized lysis buffers containing detergents suitable for membrane proteins

  • Biological variability assessment:

    • Verify Claudin 18 expression in your experimental system

    • Consider isoform-specific expression patterns (18.1 vs 18.2)

    • Account for cancer heterogeneity in clinical samples

  • Cross-reactivity with related proteins:

    • Claudin family comprises multiple members with structural similarity

    • Verify specificity against other claudins, particularly Claudin 3 and Claudin 4

    • Include appropriate positive and negative controls

Implementing a systematic troubleshooting approach can significantly improve reproducibility and reliability when working with Claudin 18 antibodies across various research applications.

What potential mechanisms explain the continued therapeutic effect of anti-Claudin 18.2 antibodies after treatment discontinuation?

The observed phenomenon of continued therapeutic effect after discontinuation of anti-Claudin 18.2 antibody treatment, as reported in a clinical case , suggests several potential mechanisms:

  • Extended antibody half-life:

    • Humanized monoclonal antibodies like ASKB589 typically have serum half-lives of 2-3 weeks

    • This enables continued target engagement even after treatment cessation

    • The pharmacokinetic profile may support a prolonged presence in the circulation

  • Immune memory induction:

    • Antibody-dependent cellular cytotoxicity (ADCC) relies on immune effector cells

    • Initial exposure may trigger expansion of immune cell populations

    • These expanded populations may persist and continue surveillance

  • Epitope spreading phenomenon:

    • Initial immune response against Claudin 18.2 might spread to recognize additional tumor antigens

    • This diversification of the immune response could sustain anti-tumor effects

  • Tumor microenvironment remodeling:

    • Anti-Claudin 18.2 therapy may alter the immunosuppressive tumor microenvironment

    • These changes could permit continued immune-mediated tumor control

  • Induction of adaptive immunity:

    • Beyond innate immune activation through ADCC and CDC mechanisms

    • Potential activation of T-cell responses against tumor antigens

    • Development of tumor-specific memory T cells

The case report describing near complete tumor response with progression-free survival of at least 10 months after treatment discontinuation highlights the need for further research into these mechanisms. Understanding these phenomena could inform optimal treatment schedules, potentially allowing intermittent dosing strategies that maintain efficacy while reducing adverse effects such as ascites.

How might novel antibody engineering approaches enhance Claudin 18-targeted therapies?

Emerging antibody engineering approaches offer several promising avenues to enhance Claudin 18-targeted therapies:

  • Bispecific antibody development:

    • Creating antibodies that simultaneously bind Claudin 18.2 and:

      • Immune effector cells (T cells, NK cells) to enhance tumor killing

      • Other tumor antigens to overcome heterogeneity and resistance

      • Immune checkpoint molecules to combine targeting with immunomodulation

  • Antibody-drug conjugate optimization:

    • Coupling potent cytotoxic payloads to anti-Claudin 18.2 antibodies

    • Engineering cleavable linkers optimized for tumor microenvironment conditions

    • Exploring novel payloads beyond traditional chemotherapeutics

  • Fc engineering for enhanced effector functions:

    • Modifying the Fc region to enhance:

      • ADCC activity through improved FcγRIIIa binding

      • CDC through enhanced C1q recruitment

      • Half-life extension via enhanced FcRn binding

    • Potentially reducing adverse effects while maintaining efficacy

  • Intrabody development:

    • Engineering antibody fragments that function intracellularly

    • Targeting Claudin 18 biosynthesis or trafficking pathways

    • Delivering these constructs via gene therapy approaches

  • Antibody fragment approaches:

    • Developing smaller formats (scFv, Fab, nanobodies)

    • Improving tumor penetration, particularly for solid tumors

    • Reducing immunogenicity and improving biodistribution

These innovative approaches could address current limitations of anti-Claudin 18.2 therapies, including adverse effects like severe ascites documented in clinical cases , while potentially expanding the patient population that might benefit from these targeted therapies.

What methodological advances are needed to improve the sensitivity and specificity of Claudin 18 detection in diverse biological samples?

Several methodological advances could significantly improve Claudin 18 detection:

  • Enhanced epitope retrieval techniques:

    • Development of specialized antigen retrieval protocols for tight junction proteins

    • Optimization of tissue fixation methods to preserve epitope structure

    • Novel enzymatic treatment approaches to expose hidden epitopes

  • Digital pathology and artificial intelligence integration:

    • Machine learning algorithms for automated scoring of Claudin 18 expression

    • Standardization of expression threshold determination

    • Improved reproducibility in patient stratification for clinical trials

  • Single-cell analysis technologies:

    • Flow cytometry with spectral unmixing for improved multiparameter analysis

    • Mass cytometry (CyTOF) incorporation of Claudin 18 antibodies

    • Single-cell sequencing coupled with protein detection

  • Proximity-based detection methods:

    • Proximity ligation assays to study Claudin 18 protein interactions

    • CODEX multiplexed tissue imaging for spatial relationship analysis

    • In situ sequencing approaches for simultaneous detection of multiple markers

  • Liquid biopsy refinements:

    • Highly sensitive CLEIA development for serum/plasma Claudin 18 detection

    • Similar to achievements with fragmented cytokeratin 18 detection (sensitivity of 0.056 ng/mL)

    • Exosome isolation and analysis for Claudin 18 detection

These methodological advances would enable more precise patient selection for targeted therapies, improved monitoring of treatment response, and better understanding of Claudin 18 biology in both normal physiology and disease states.

How can researchers better predict and mitigate adverse effects associated with therapeutic Claudin 18.2 antibodies?

Improving prediction and mitigation of adverse effects from therapeutic Claudin 18.2 antibodies requires multi-faceted approaches:

  • Comprehensive tissue cross-reactivity studies:

    • Expanded analysis of Claudin 18.2 expression across normal tissues

    • Quantitative assessment of expression levels rather than binary presence/absence

    • Correlation of expression patterns with observed toxicities

  • Mechanistic understanding of adverse events:

    • Investigation of ascites development mechanisms reported with ASKB589

    • Analysis of whether adverse effects are on-target (related to Claudin 18.2 binding) or off-target

    • Examination of antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) contributions to toxicity

  • Biomarker development for toxicity prediction:

    • Identification of early biomarkers predicting severe adverse effects

    • Genetic polymorphisms affecting antibody metabolism or immune activation

    • Baseline immune status assessment to predict excessive immune activation

  • Dosing optimization strategies:

    • Exploration of alternative dosing schedules

    • Pharmacokinetic/pharmacodynamic modeling to identify optimal therapeutic window

    • Investigation of continued anti-tumor response despite treatment discontinuation

  • Combination approach refinement:

    • Careful evaluation of toxicity profiles when combining with chemotherapy

    • Pre-treatment strategies to mitigate anticipated adverse effects

    • Sequential rather than concurrent therapy approaches

These strategies could help maximize the therapeutic benefit of anti-Claudin 18.2 antibodies while minimizing adverse effects, potentially expanding their clinical utility for patients with gastric, pancreatic, and other Claudin 18.2-expressing cancers.

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.