Anti-Human Claudin-18.2 (Zolbetuximab)

Anti-Human Claudin-18.2 (Zolbetuximab)

Product No.: C3190

- -
- -
Product No.C3190
Clone
IMAB-362
Target
Claudin-18.2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Claudin-18
Isotype
Human IgG1κ
Applications
ELISA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Active
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
State of Matter
Liquid
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
ELISA
Each investigator should determine their own optimal working dilution for specific applications. See directions on lot specific datasheets, as information may periodically change.

Description

Description

Specificity
This non-therapeutic biosimilar antibody uses the same variable region sequence as the therapeutic antibody Zolbetuximab . Zolbetuximab (IMAB-362) is a chimeric IgG1 monoclonal antibody that specifically targets Claudin 18.2.
Background
CLDN18.2 is a protein that is overexpressed in various cancers, including gastric, pancreatic, and esophageal cancers, making it a valuable therapeutic target. It plays a key role in carcinogenesis, cell polarity, and intercellular adhesion. Monoclonal antibodies like zolbetuximab have shown potential in treating advanced cancers with minimal adverse effects, positioning CLDN18.2 as a promising focus for cancer therapy. A deeper understanding of CLDN18.2's functions is crucial for developing effective treatments, especially for patients with poor prognoses1-3.

Zolbetuximab (IMAB362) has demonstrated promise in treating advanced gastric and gastroesophageal junction cancers by targeting CLDN18.2. Clinical trials have shown that Zolbetuximab is well-tolerated and effective, inducing antitumor activity through antibody- dependent cellular cytotoxicity and complement-dependent cytotoxicity. Preclinical studies also support its use in combination with standard chemotherapy, further enhancing its potential as a cancer treatment4-6.

Antigen Distribution
Claudin 18.2 is a highly selective marker with limited expression in normal tissues but is often abnormally expressed in various primary malignant tumors, such as gastric cancer, pancreatic cancer, and others.
Ligand/Receptor
YAP1
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Tumor Suppressors

Leinco Antibody Advisor

Powered by AI: AI is experimental and still learning how to provide the best assistance. It may occasionally generate incorrect or incomplete responses. Please do not rely solely on its recommendations when making purchasing decisions or designing experiments.

Research-grade Zolbetuximab biosimilars are commonly used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to ensure accurate quantification of drug concentrations in serum during PK and bioequivalence studies for biosimilars.

Essential context and supporting details:

  • In PK bridging ELISA assays designed for biosimilars, a single assay with a single standard curve is typically established to simultaneously quantify both the biosimilar and the reference (originator) product in serum samples.
  • Research-grade Zolbetuximab biosimilars are selected as the assay standards because they are structurally and functionally equivalent to the therapeutic Zolbetuximab, ensuring that the assay quantifies both variants with equivalent accuracy.
  • During assay development, comparability studies are conducted. Both biosimilar and reference samples are spiked into human serum at known concentrations, and their recovery curves are compared when quantified using the biosimilar as the standard. If bioanalytical equivalence is confirmed (e.g., equivalence margins of 0.8–1.25), the biosimilar is validated as a reliable calibrator for the assay.
  • Once validated, a calibration curve is generated by preparing serial dilutions of the Zolbetuximab biosimilar in serum matrix at defined concentrations. This standard curve is run alongside study samples in each assay.
  • Test and Quality Control (QC) samples—containing either biosimilar or reference Zolbetuximab—are quantified against this standard curve, allowing direct assessment of serum drug levels, PK profile, and, ultimately, comparability between biosimilar and reference products.
  • The process must comply with regulatory guidance on ligand binding assays, emphasizing precision, accuracy, and robustness; biosimilar standards must enable equivalent quantification of reference and test products as a critical part of demonstrating PK similarity.

Additional points:

  • Commercially available research-grade Zolbetuximab biosimilars, such as those from Bioss, Bio X Cell, or Assay Genie, are suitable for ELISA calibration due to validated specificity and similar sequence/structure to the clinical product.
  • The validation of assay performance (precision, accuracy, specificity, linearity) ensures the selected biosimilar standard is appropriate for use as an analytical calibrator in regulated PK studies.

In summary, Zolbetuximab biosimilar antibodies serve as validated, reliable calibration standards and reference controls in PK bridging ELISA to accurately and comparably quantify drug levels in serum from clinical and preclinical biosimilar studies.

The primary preclinical models for evaluating a research-grade anti-Claudin-18.2 (CLDN18.2) antibody in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are:

  • Humanized transgenic (Knock-in) syngeneic mouse models expressing human CLDN18.2 in otherwise immunocompetent backgrounds.
  • Human tumor xenograft models, most commonly in immunodeficient mice bearing human tumors engineered for CLDN18.2 expression.

Supporting Details:

  • Humanized Syngeneic Models:
    Recent studies report the use of humanized transgenic mice with human immune system components combined with murine tumor cell lines engineered to express human CLDN18.2 (e.g., MC38^hCLDN18.2^ syngeneic colorectal carcinoma model). Mice can also be engineered to express human target genes or immune proteins (e.g., human 4-1BB), allowing administration of human-specific antibodies within a functional murine immune context. These models are explicitly used for immunophenotyping TILs by flow cytometry, including CD45^+, CD8^+ T cells, Tregs (Foxp3^+), and calculation of CD8/Treg ratios after anti-CLDN18.2 or bispecific antibody treatment.

  • Human Xenograft Models:
    Tumor growth inhibition is commonly tested in xenograft models, where human cancer cell lines (gastric, pancreatic, or engineered lines overexpressing CLDN18.2) are implanted into immunodeficient mice. Here, anti-CLDN18.2 antibodies (e.g., Zolbetuximab, hu7v3-FC) significantly impair tumor growth and can extend mouse survival. TIL analysis is limited due to lack of a fully competent murine immune system but can sometimes be achieved with reconstituted human immune cells in partially humanized mice.

  • Syngeneic models (Murine tumor/murine immune system):
    Fully murine models, with mouse tumor cells expressing human CLDN18.2 in mice possessing intact immune systems, are valuable for evaluating TIL dynamics and ADCC/CDC mechanisms. These models facilitate precise profiling of immune cell subsets (e.g., CD8^+ T cells, Tregs, macrophages) after antibody therapy.
    However, for anti-human CLDN18.2 antibodies, murine models are only informative if the antibody is cross-reactive (binds murine CLDN18.2) or the tumor cell line expresses the human protein. Use of engineered cell lines (e.g., MC38^hCLDN18.2^) in C57BL/6 mice is the typical approach for this scenario.

Model Comparison Table:

Model TypeCLDN18.2 ExpressionImmune SystemTIL Analysis FeasibilityExamples
Humanized SyngeneicMurine tumor cellsFunctionalHighMC38^hCLDN18.2^ in humanized transgenic mice
Xenograft (human tumor)Human tumor cellsDeficientLimitedBGC823CLDN18.2, AGSCLDN18.2 in nude mice
Humanized XenograftHuman tumor cellsHumanModerateHu immune-engrafted NSG mice with xenograft
Syngeneic (mouse tumor)Murine or humanizedFunctionalHigh, if hCLDN18.2 usedMC38^hCLDN18.2^ in C57BL/6 mice

Key Insights:

  • TIL characterization after anti-CLDN18.2 antibody administration is most robust in syngeneic or humanized syngeneic models, with immunophenotyping by flow cytometry.
  • Tumor growth inhibition is consistently demonstrated in both xenograft and syngeneic settings with CLDN18.2-directed antibody therapies.
  • Choice of model depends on antibody specificity (human or cross-reactive), tumor cell engineering, and the requirement for full immunologic context (essential for TIL studies).

Alternative/Additional Approaches:

  • Combination therapies (e.g., bispecific antibodies targeting CLDN18.2 and immune costimulatory molecules) and CAR-T cell products targeting CLDN18.2 are also tested in these models for advanced immunological characterization.
  • Antibody-drug conjugate versions can be evaluated for tumor growth inhibition using similar models.

In summary:
The most commonly used and informative models for in vivo administration of anti-CLDN18.2 antibodies to study tumor growth and analyze TILs are humanized syngeneic mouse models (e.g., MC38^hCLDN18.2^) and human tumor xenografts in immunodeficient mice, with the former being superior for deep immunophenotyping of TILs.

Researchers currently study zolbetuximab—a monoclonal antibody targeting Claudin 18.2—in combination with other immune-oncology drugs, most commonly checkpoint inhibitors such as pembrolizumab (an anti-PD-1 antibody), to explore potential synergistic effects in cancer models. However, there is no published evidence from the search results of studies using zolbetuximab specifically in combination with anti-CTLA-4 or anti-LAG-3 biosimilars in complex immune-oncology models.

Key approaches and findings include:

  • Combination with Immune Checkpoint Inhibitors: The most documented clinical investigation involves combining zolbetuximab with pembrolizumab and chemotherapy in patients with Claudin 18.2-positive, HER2-negative gastric and gastroesophageal junction cancers. The rationale is that zolbetuximab initiates an immune attack against tumor cells (via ADCC and CDC), while checkpoint inhibitors such as pembrolizumab relieve immune suppression, potentially yielding additive or synergistic anti-tumor effects.

  • Mechanistic Studies: Other studies examined the combination of zolbetuximab with immune-modulating agents like zoledronic acid and interleukin-2 to monitor enhanced activation, expansion, and cytotoxic function of immune cells (γδ T cells, NK cells), but not specifically with checkpoint inhibitors targeting CTLA-4 or LAG-3. While zolbetuximab alone robustly primes effector cells for ADCC, co-administration with these immune modulators did not show a clear enhancement of effect, although the sample size and design limited conclusive findings.

  • Synergistic Mechanisms: The theoretical basis for synergy with checkpoint inhibitors (whether anti-PD-1, anti-CTLA-4, or anti-LAG-3) is that:

    • Zolbetuximab marks and destroys CLDN18.2-positive tumor cells, unlocking immune cell engagement.
    • Checkpoint inhibitors prevent immune exhaustion or suppression, allowing sustained or augmented anti-tumor activity by the endogenous immune system.
    • Combining these agents could enable more durable and broader immune-mediated tumor regression, especially in immunologically "cold" tumors.
  • Evidence Gap on anti-CTLA-4 and anti-LAG-3: No available studies directly address the effect of zolbetuximab combined with anti-CTLA-4 or anti-LAG-3 biosimilars as of the search date. The reported combinations are limited to anti-PD-1 (pembrolizumab) and various immune activators. There is a need for future preclinical or clinical trials testing these specific combinations to clarify their synergistic potential.

Experimental Models: In such studies, researchers would typically use:

  • In vivo mouse xenograft or patient-derived tumor models expressing CLDN18.2.
  • Ex vivo immune cell assays (e.g., peripheral blood mononuclear cells or tumor-infiltrating lymphocytes) to quantify cytotoxicity, cytokine profiles, and immune cell activation in the presence of antibody combinations.

Conclusion: While zolbetuximab's combination with immune checkpoint inhibitors (notably anti-PD-1) is under active clinical investigation and supported by a synergistic mechanistic rationale, published data on its co-administration with anti-CTLA-4 or anti-LAG-3 biosimilars is lacking. Most research thus far prioritizes the anti-PD-1 axis; expansion to other checkpoint inhibitor combinations is expected in future translational and clinical studies.

A Zolbetuximab biosimilar is used as a key reagent in a bridging anti-drug antibody (ADA) ELISA to detect anti-Zolbetuximab antibodies that might develop in patients, indicating an immune response against the therapeutic drug.

In the bridging ADA ELISA format:

  • Zolbetuximab biosimilar is used both as the capture reagent and the detection reagent. The biosimilar is structurally and functionally similar to the therapeutic mAb and is suitable because it has the same variable region as Zolbetuximab.

Typical process:

  • Plate coating: Zolbetuximab biosimilar is immobilized on the microtiter plate to capture any anti-Zolbetuximab antibodies (ADAs) present in the patient serum.
  • Bridging complex formation: If ADAs are present in the sample, they will bind to the immobilized Zolbetuximab biosimilar via one binding site, while their other binding site will capture a second molecule of labeled Zolbetuximab biosimilar (e.g., HRP-labeled Zolbetuximab) added subsequently.
  • Detection: Formation of this "bridge" between two Zolbetuximab biosimilar molecules, via the ADA, is then detected with a substrate for the label (e.g., TMB for HRP), producing a measurable color change proportional to ADA concentration.

Critical points:

  • The biosimilar is preferred for research and method development because it mimics the therapeutic but is often available without proprietary restrictions.
  • Detection can utilize various Zolbetuximab formats (biotinylated, HRP-conjugated) as long as the biosimilar maintains affinity for the patient ADA.
  • The bridging format is advantageous for detecting bivalent antibodies (i.e., typical IgG responses) and is the standard for most ADA assays for monoclonal antibodies.

Example analogy (from metuzumab and other mAbs):

  • The same approach has been used for other monoclonal antibodies: the target drug or its biosimilar is coated onto a plate to capture ADAs, and a labeled version is used for detection, relying on the "bridging" capacity of the ADA molecule between two drug molecules.

In summary:
A Zolbetuximab biosimilar acts both as the immobilized capture reagent and as the labeled detection reagent in a bridging ADA ELISA, enabling the detection of anti-Zolbetuximab antibodies in patient samples, thus allowing monitoring of the patient's immune response to the therapeutic drug.

References & Citations

1. Zhu G, Foletti D, Liu X, et al. Sci Rep. 2019;9(1):8420.
2. Sahin U, Koslowski M, Dhaene K, et al. Clin Cancer Res. 2008;14(23):7624-7634.
3. Jiang H, Shi Z, Wang P, et al. J Natl Cancer Inst. 2019;111(4):409-418.
4. Sahin U, Schuler M, Richly H, et al. Eur J Cancer. 2018;100:17-26.
5. H BM, U E, Cr S, et al. Oncoimmunology. 2023;12(1).
6. Türeci Ӧzlem, Mitnacht-Kraus R, Wöll S, Yamada T, Sahin U. Oncoimmunology.2019;8(1):e1523096.
7. Zolbetuximab Chimeric Recombinant Monoclonal Antibody (MA5-42158). Accessed October 4, 2024. https://www.thermofisher.com/antibody/product/Zolbetuximab-Chimeric-Antibody-Recombinant-Monoclonal/MA5-42158
Indirect Elisa Protocol

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.