Claudin-18.2 Recombinant Monoclonal Antibody

Shipped with Ice Packs
In Stock

Description

Production and Validation

The antibody is produced via recombinant DNA technology:

  1. B Cell Isolation: Spleen-derived B cells from immunized animals are used as a source of antibody genes.

  2. Gene Amplification: Reverse transcription and PCR amplify CLDN18.2-specific variable regions.

  3. Expression: Genes are inserted into expression vectors (e.g., HEK293 cells) for protein production.

  4. Purification: Affinity chromatography (e.g., Protein A/G) yields ≥90% pure antibody .

Validation includes:

  • ELISA: EC50 values of 2.623–14.06 ng/mL for human CLDN18.2 detection .

  • Flow Cytometry: Effective binding to CLDN18.2-expressing gastric mucosa cells .

  • IHC: High specificity in human, mouse, and rat tissues (e.g., EPR19202 clone) .

Monoclonal Antibodies

Drug NamePhaseTargetMechanismStatus
AB011Phase ICLDN18.2+ solid tumorsADCC/CDC inductionOngoing
M108Phase IGastric/esophagogastricTumor cell lysisOngoing
ZolbetuximabPhase IIPancreatic/gastric cancerADCC/CDC; combination with chemotherapyCompleted

Source:

Antibody-Drug Conjugates (ADCs)

Drug NamePhasePayloadKey Features
IBI343Phase ITopoisomerase IBystander killing; tolerable safety profile
LM-302Phase IMMAEOrphan drug designation for pancreatic cancer

Source:

Mechanisms of Action

  1. Antibody-Dependent Cellular Cytotoxicity (ADCC):

    • Recruits immune cells (e.g., NK cells) to lyse CLDN18.2+ tumor cells .

  2. Complement-Dependent Cytotoxicity (CDC):

    • Activates the complement system, causing direct cell membrane disruption .

  3. ADC Bystander Effect:

    • IBI343 and LM-302 release cytotoxic payloads (e.g., topoisomerase inhibitors) after internalization, killing adjacent tumor cells .

Expression Patterns and Prognostic Relevance

Cancer TypeCLDN18.2 OverexpressionPrognostic Impact
Gastric46.5% (stages I-III) Poor survival
Pancreatic65.7% (metastases) Disease progression
Breast/ColonLimited dataUnder investigation

Source:

Challenges and Future Directions

  • Heterogeneity: CLDN18.2 expression varies, limiting efficacy in low-expressing tumors. ADCs with bystander effects (e.g., IBI343) address this .

  • Combination Therapies: AB011 trials explore synergy with chemotherapy or immunotherapy .

  • CAR-T Therapies: Emerging approaches target CLDN18.2 with engineered T cells, though clinical data remain pending .

Product Specs

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

The Claudin-18.2 Recombinant Monoclonal Antibody is meticulously engineered to ensure high quality and specificity. The production process begins with the isolation of B cells from the spleen of an immunized animal, using recombinant human Claudin-18.2 protein as the immunogen. RNA extracted from these B cells is then reverse transcribed into cDNA, allowing for the amplification of Claudin-18.2 antibody genes using specific primers targeting the antibody constant regions. These amplified genes are subsequently inserted into an expression vector and transfected into host cells, enabling the production of the Claudin-18.2 recombinant monoclonal antibody. Following a period of cell culture, the antibody is collected from the cell culture supernatant and purified through affinity chromatography, resulting in a highly purified form suitable for diverse applications. Rigorous characterization assays, including ELISA and flow cytometry (FC) analysis, are conducted to validate the antibody's specificity and functionality in detecting human Claudin-18.2 protein. This meticulous production process guarantees the development of a reliable and effective Claudin-18.2 recombinant monoclonal antibody, playing a critical role in various Claudin-18.2-related research endeavors.

Form
Liquid
Lead Time
Generally, we can ship the products within 1-3 business days of receiving your order. Delivery times may vary depending on the mode of purchase or location. For specific delivery timeframes, please consult your local distributors.
Synonyms
Claudin-18, CLDN18
Target Names
Uniprot No.

Customer Reviews

Overall Rating 5.0 Out Of 5
,
B.A
By Anonymous
★★★★★

Applications : FC

Sample type: cell

Review: Normal assay process. The prodcuts performes well in the whole assay.

Q&A

What is Claudin-18.2 and what is its normal physiological function?

Claudin-18.2 is a tight junction protein primarily found in gastric epithelial cells. It serves a critical physiological function by binding cells in a tight formation that prevents stomach acids from penetrating into surrounding stomach tissue. Unlike many cancer biomarkers, CLDN18.2 does not appear to have inherent oncogenic properties. Current research indicates that higher CLDN18.2 expression levels do not correlate with accelerated cancer growth or poorer prognosis in patients . This characteristic makes it a unique target as its overexpression in cancer represents a positional change rather than a functional one in malignant transformation.

Which cancer types show significant Claudin-18.2 overexpression?

CLDN18.2 overexpression has been documented across multiple cancer types with varying prevalence rates. The current literature identifies the following cancer types with significant CLDN18.2 expression:

  • Gastric cancer

  • Gastroesophageal junction adenocarcinoma

  • Pancreatic ductal adenocarcinoma (PDAC)

  • Non-small cell lung cancer (in a subset of cases)

  • Ovarian cancer

The abnormal expression pattern in these malignancies has established CLDN18.2 as an attractive therapeutic target, particularly in gastric cancers where expression is frequently high . The expression pattern suggests epithelial cancers originating from tissues with gastric-type differentiation are most likely to exhibit CLDN18.2 positivity.

What methodologies should researchers employ for reliable Claudin-18.2 detection in tumor specimens?

Accurate CLDN18.2 detection requires standardized methodologies to ensure consistency across studies. The current gold standard is immunohistochemistry (IHC) with validated antibodies specific to the CLDN18.2 isoform. Critical methodological considerations include:

  • Specimen preparation: Formalin-fixed paraffin-embedded (FFPE) tissue samples are typically used, with standardized fixation times to prevent antigen masking.

  • Validation protocol: Implementation of centralized validation using clinical trial assays to ensure consistent interpretation .

  • Scoring criteria: The field lacks standardized positivity thresholds, with studies using different cutoffs ranging from ≥40% to ≥75% of tumor cells showing membrane staining .

  • Controls: Inclusion of appropriate positive controls (normal gastric tissue) and negative controls to validate assay specificity.

  • Multi-region sampling: To account for potential intratumoral heterogeneity in CLDN18.2 expression.

Researchers should be aware that variations in detection protocols significantly impact the reported prevalence of CLDN18.2 positivity, which has implications for therapeutic targeting strategies.

How does Claudin-18.2 expression correlate with clinicopathological features in cancer?

Meta-analyses of the relationship between CLDN18.2 expression and clinicopathological parameters have yielded important insights for researchers. Current evidence indicates:

These findings suggest that CLDN18.2 expression represents an independent biomarker rather than a surrogate for established prognostic factors. This independence enhances its value as a therapeutic target since it potentially identifies a patient population not currently captured by existing biomarkers . The relationship between CLDN18.2 positivity and clinicopathological features appears highly dependent on the specific threshold used to define CLDN18.2 positivity, underscoring the need for standardized assessment criteria.

What are the primary mechanisms of action for Claudin-18.2 targeted antibodies in the tumor microenvironment?

CLDN18.2-targeted antibodies exert anti-tumor effects through multiple immune-mediated mechanisms of action:

  • Antibody-Dependent Cellular Cytotoxicity (ADCC): After binding to CLDN18.2 expressed on tumor cell surfaces, the Fc portion of the antibody recruits natural killer cells and other immune effectors that recognize the antibody and subsequently eliminate the antibody-coated tumor cells.

  • Complement-Dependent Cytotoxicity (CDC): Antibody binding activates the complement cascade, leading to formation of the membrane attack complex and tumor cell lysis.

In vitro studies with humanized VHH-based recombinant antibodies like hu7v3-Fc have demonstrated both ADCC and CDC activity against CLDN18.2-positive tumor cells . The relative contribution of each mechanism may vary depending on antibody structure, the tumor microenvironment, and patient-specific factors such as immune competence. Understanding these mechanisms is crucial for rational design of combination treatment strategies.

How do different Claudin-18.2 antibody formats compare in terms of tumor penetration and efficacy?

Research comparing different antibody formats has revealed important differential characteristics:

Antibody FormatSizeTumor PenetrationTumor UptakeIn Vivo EfficacyExample
Conventional monoclonal antibodies~150 kDaModerateSlowerDemonstrated clinical efficacyZolbetuximab
VHH-based recombinant antibodies~80 kDaEnhancedFasterSuperior in mouse xenograft modelshu7v3-Fc

In vivo biodistribution studies using zirconium-89 (89Zr) labeled antibodies provide compelling evidence that hu7v3-Fc exhibits better tumor penetration and faster tumor uptake compared to conventional antibodies like Zolbetuximab. This advantage is attributed to its smaller size and higher binding affinity. In mouse xenograft models, hu7v3-Fc demonstrated significantly more potent anti-tumor efficacy than Zolbetuximab, suggesting that engineering smaller antibody formats may overcome limitations in targeting solid tumors where penetration is challenging .

What criteria define Claudin-18.2 positivity in research and clinical contexts?

The definition of CLDN18.2 positivity varies across studies, creating challenges for comparison and interpretation. Current approaches include:

  • Percentage-based thresholds: Studies employ different cutoffs for the percentage of tumor cells with membrane staining:

    • Moderate threshold: ≥40% positive tumor cells

    • High threshold: ≥70-75% positive tumor cells

  • Intensity-based assessment: Some protocols incorporate both the percentage of positive cells and the intensity of staining (weak, moderate, strong).

  • Centralized validation: Clinical trials typically determine CLDN18.2 expression using centralized laboratory testing with validated assays to ensure consistency .

Meta-analyses have demonstrated that the relationship between CLDN18.2 expression and clinical outcomes depends significantly on the threshold used to define positivity . This variability highlights the critical need for standardized criteria in both research and clinical settings to enable meaningful cross-study comparisons and optimal patient selection for targeted therapies.

What challenges exist in current Claudin-18.2 detection methodologies?

Researchers face several methodological challenges in CLDN18.2 detection that may impact study outcomes:

Addressing these challenges requires rigorous validation protocols, consideration of multiple sampling sites, and development of complementary detection methods to enhance reliability.

How does the humanized VHH-based anti-CLDN18.2 recombinant antibody compare mechanistically to conventional monoclonal antibodies?

The humanized VHH-based anti-CLDN18.2 recombinant antibody (hu7v3-Fc) demonstrates several mechanistic advantages over conventional monoclonal antibodies:

Parameterhu7v3-Fc (VHH-based)Zolbetuximab (Conventional mAb)Significance
Tumor penetrationEnhancedStandardImproved access to poorly vascularized tumor regions
Tumor uptake kineticsFasterSlowerPotentially faster onset of therapeutic effect
Binding affinityHigherStandardMore efficient target engagement
Xenograft efficacyMore potentLess potentPotentially improved clinical outcomes
SizeSmaller (~80 kDa)Larger (~150 kDa)Contributes to superior tissue penetration

In vivo biodistribution studies using 89Zr-labeled antibodies demonstrate that hu7v3-Fc achieves better tumor penetration and faster tumor uptake compared to Zolbetuximab, advantages attributed to its smaller size and higher target affinity. These properties translate to significantly more potent anti-tumor efficacy in mouse xenograft models. Additionally, the modular nature of the hu7v3 component makes it a versatile building block for developing novel CLDN18.2-targeted therapeutics with diverse mechanisms of action .

What therapeutic modalities beyond monoclonal antibodies are being investigated for Claudin-18.2 targeting?

Research into CLDN18.2-targeted therapies has expanded beyond conventional antibodies to explore multiple innovative approaches:

  • T cell antigen coupler (TAC) technology: TAC01-CLDN18.2 represents an autologous T-cell product where T cells are modified ex vivo to express CLDN18.2-specific TAC receptors. This approach co-opts the natural T cell receptor to enable cytotoxicity against tumor cells while demonstrating a potentially safer profile than chimeric antigen receptor T cells .

  • Antibody-drug conjugates (ADCs): These combine CLDN18.2-targeting antibodies with cytotoxic payloads, potentially enhancing efficacy against tumors with heterogeneous CLDN18.2 expression.

  • Bispecific antibodies: These simultaneously engage CLDN18.2 on tumor cells and immune effector cells to enhance immune-mediated tumor killing.

  • CAR-T therapies: Multiple chimeric antigen receptor T cell approaches targeting CLDN18.2 are in development globally, leveraging cellular immunity against CLDN18.2-positive tumors.

  • Combination approaches: CLDN18.2-targeted agents are being evaluated in combination with chemotherapy, immune checkpoint inhibitors, and other targeted therapies to enhance efficacy .

Phase I clinical trials with TAC01-CLDN18.2 have shown promising early safety data, with no dose-limiting toxicities in the first two dose cohorts and only low-grade TAC-related adverse events .

What strategies can researchers employ to investigate and overcome resistance to Claudin-18.2 targeted therapies?

Developing strategies to address resistance to CLDN18.2-targeted therapies requires systematic investigation of potential resistance mechanisms:

  • Expression dynamics analysis:

    • Serial tumor biopsies before, during, and after treatment to monitor CLDN18.2 expression changes

    • Single-cell RNA sequencing to detect resistant subpopulations

    • Proteomic analysis to identify compensatory signaling pathways

  • Resistance modeling approaches:

    • Development of resistant cell lines through prolonged exposure to CLDN18.2-targeted agents

    • Patient-derived xenografts from treatment-resistant tumors

    • CRISPR-Cas9 screens to identify genes mediating resistance

  • Therapeutic strategies to overcome resistance:

    • Multi-epitope targeting using antibodies binding different regions of CLDN18.2

    • Combination with agents targeting potential resistance pathways

    • Alternating therapeutic modalities (e.g., antibodies followed by cellular therapies)

    • Development of antibody-drug conjugates that require lower CLDN18.2 expression for efficacy

  • Biomarker development for early resistance detection:

    • Liquid biopsy approaches to monitor circulating tumor DNA and potential CLDN18.2 alterations

    • Imaging biomarkers correlating with developing resistance

This systematic approach to understanding and addressing resistance mechanisms will be crucial for maximizing the long-term efficacy of CLDN18.2-targeted therapies across multiple cancer types.

What experimental models are most appropriate for preclinical evaluation of Claudin-18.2 targeted therapies?

Selection of appropriate experimental models for CLDN18.2-targeted therapy evaluation requires careful consideration of the specific research questions:

Model TypeAdvantagesLimitationsBest Applications
Cell line-derived xenograftsReproducible, well-characterized, cost-effectiveLimited heterogeneity, immunodeficient hostInitial efficacy screening, mechanism studies
Patient-derived xenograftsPreserve tumor architecture and heterogeneityExpensive, variability between samples, immunodeficient hostTranslation studies, heterogeneity assessment
Syngeneic modelsIntact immune system, evaluate immune-mediated effectsRequire murine versions of human therapeuticsImmunotherapy combinations, ADCC/CDC studies
Humanized immune system miceHuman immune components, evaluation of human-specific therapiesIncomplete immune reconstitution, expensiveT cell-engaging therapies, advanced immunotherapy
Genetically engineered modelsPhysiological expression patterns, intact microenvironmentTime-consuming, expensive, may not fully recapitulate human diseaseSafety studies, on-target/off-tumor effects

The mouse xenograft model has been successfully used to demonstrate the superior anti-tumor efficacy of hu7v3-Fc compared to conventional antibodies like Zolbetuximab . For cellular therapies like TAC01-CLDN18.2, humanized immune system models may provide more relevant preclinical data. The ideal approach often involves using multiple complementary models to address different aspects of therapeutic development.

What protocols should researchers implement for comprehensive pharmacokinetic and biodistribution assessment of Claudin-18.2 antibodies?

Comprehensive pharmacokinetic and biodistribution assessment of CLDN18.2 antibodies requires multi-modal approaches:

  • Advanced imaging methodologies:

    • Radiolabeling with positron emitters (e.g., zirconium-89) for PET imaging to quantitatively track whole-body distribution

    • This approach has been successfully applied to compare biodistribution profiles of hu7v3-Fc and Zolbetuximab

    • Near-infrared fluorescence imaging for high-resolution preclinical studies

    • Multimodal imaging combining anatomical and functional information

  • Quantitative tissue analysis protocols:

    • Standardized collection of multiple tissues at defined timepoints

    • Quantification of antibody concentration using ELISA or mass spectrometry

    • Correlation of tissue levels with CLDN18.2 expression patterns

    • Assessment of antibody integrity and potential metabolites

  • Tumor-specific distribution analysis:

    • Multiplex immunohistochemistry to simultaneously visualize antibody penetration, CLDN18.2 expression, and microenvironmental factors

    • 3D reconstruction techniques to assess penetration gradients relative to vasculature

    • Microdialysis for dynamic measurement of antibody concentrations in tumor interstitium

  • Pharmacokinetic modeling approaches:

    • Physiologically-based pharmacokinetic (PBPK) modeling to predict tissue distribution

    • Population PK analysis to account for inter-individual variability

    • Integration of imaging and sampling data for comprehensive PK/PD relationships

These methodologies provide complementary information about antibody behavior in vivo and should be adapted based on the specific antibody format and research question.

How should in vitro functional assays be designed to evaluate the efficacy of Claudin-18.2 targeted antibodies?

Robust in vitro evaluation of CLDN18.2-targeted antibodies requires carefully designed functional assays:

  • Cell model selection and validation:

    • Panel of cell lines with varying CLDN18.2 expression levels quantified by flow cytometry and Western blot

    • Engineered isogenic cell lines (CLDN18.2-positive/negative pairs) to control for non-target variables

    • Primary tumor cells from patient samples when feasible

    • 3D spheroid cultures to better represent solid tumor architecture

  • Immune effector function assays:

    • ADCC assays using:

      • Isolated NK cells at varying effector:target ratios

      • PBMCs from multiple donors to account for Fc receptor polymorphisms

      • Flow cytometry and real-time cell analysis for dynamic assessment

    • CDC assays with human complement using colorimetric or luminescent readouts

    • Phagocytosis assays with macrophages and dendritic cells

  • Direct effect assessment:

    • Proliferation assays (various timepoints: 24h, 48h, 72h, 7d)

    • Apoptosis detection (Annexin V/PI staining, caspase activation)

    • Cell migration and invasion assays

    • Tight junction integrity analysis

  • Mechanistic interrogation:

    • Antibody binding kinetics via surface plasmon resonance

    • Internalization studies using pH-sensitive fluorophores

    • Signaling pathway analysis via phosphoprotein arrays

    • Combination studies with other therapeutic agents

These comprehensive assays provide a multi-dimensional assessment of antibody functionality and mechanism of action, facilitating the selection of optimal candidates for in vivo evaluation .

What considerations are essential when designing clinical trials for Claudin-18.2 targeted therapies?

Clinical trial design for CLDN18.2-targeted therapies requires careful consideration of several factors:

The Phase I/II study design for TAC01-CLDN18.2 exemplifies these considerations, employing a 3+3 dose escalation design in Phase I followed by expansion cohorts across multiple tumor types in Phase II, with the option for redosing based on predefined clinical and safety criteria .

What approaches should researchers use to investigate biomarkers of response to Claudin-18.2 targeted therapies?

Comprehensive biomarker investigation for CLDN18.2-targeted therapies requires multi-omic approaches:

  • Tissue-based biomarker analysis:

    • Beyond CLDN18.2 expression level, assess:

      • Spatial distribution within tumor

      • Co-expression with immune checkpoint molecules

      • Immune infiltrate characterization (multiplex IHC)

      • Epitope accessibility and membrane localization

    • Sequential biopsies to monitor dynamic changes during treatment

  • Liquid biopsy approaches:

    • Circulating tumor DNA analysis for genetic alterations

    • Exosomal CLDN18.2 as potential surrogate marker

    • Immune monitoring (cytokine profiles, immune cell phenotyping)

    • Circulating tumor cells for CLDN18.2 expression on metastatic cells

  • Functional imaging biomarkers:

    • FDG-PET for early metabolic response

    • Novel tracers targeting CLDN18.2 directly

    • Diffusion-weighted MRI for changes in tumor cellularity

  • Integrative analysis methods:

    • Machine learning approaches to identify complex biomarker signatures

    • Correlation of molecular features with imaging and clinical outcomes

    • Systems biology approaches to model response pathways

  • Resistance biomarker exploration:

    • Identification of bypass pathways activated in non-responding tumors

    • Epitope alterations that may affect antibody binding

    • Immune escape mechanisms in the tumor microenvironment

This comprehensive biomarker strategy enables identification of patients most likely to benefit from CLDN18.2-targeted therapies and provides mechanistic insights to guide combination approaches and address resistance.

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.