Anti-Human IL-13 (Tralokinumab) [Clone CAT-354] — Fc Muted™

Anti-Human IL-13 (Tralokinumab) [Clone CAT-354] — Fc Muted™

Product No.: I-2115

- -
- -
Product No.I-2115
Clone
CAT-354
Target
IL-13
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Isotype
Human IgG4κ
Applications
B
,
ELISA
,
ELISA Cap
,
FA
,
N

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
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 ?
B,
ELISA,
ELISA Cap,
FA,
N
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 Tralokinumab. CAT-354 (Tralokinumab) specifically binds to the type 2 cytokine IL-13.
Background
IL-13 is an important mediator of allergic inflammation and disease1 that plays a role in the regulation of IgE synthesis, induction of adhesion molecules, and chemokine expression2. The functions of IL-13 overlap considerably with those of IL-41. IL-13 induces its effects through a multi-subunit receptor composed of the alpha chain of the IL-4 receptor (IL-4Rα) and an IL-13 binding subunit IL-13Rα1. IL-13 induces many features of allergic lung disease, including airway hyper-responsiveness, goblet cell metaplasia, and mucus hypersecretion, which all contribute to airway obstruction. IL-13 is also associated with the induction of chronic pulmonary eosinophilia and eosinophilic esophagitis3 and plays a key role in the pathogenesis of atopic dermatitis4. Although IL-13 is associated primarily with the induction of airway disease, it also has anti-inflammatory properties1.

CAT-354 (Tralokinumab) was isolated and optimized using antibody display technologies3. Tralokinumab acts as a neutralizing antibody2 that binds specifically and with high affinity to IL- 13, preventing interaction with the IL-13 receptor4. In mice administered human IL-13, tralokinumab blocks human IL-13-induced lung eosinophilia, significantly decreases airway hyper-responsiveness, and inhibits eosinophil recruitment to the esophagus3. Tralokinumab does not affect human IL-13-induced goblet cell metaplasia in mouse lungs. Additionally, phase 3 trials produced inconsistent benefits for the treatment of asthma and clinical evaluation was discontinued for this indication4.

Tralokinumab has been studied and approved for use in atopic dermatitis4.

Antigen Distribution
IL-13 is a cytokine secreted by many cell types but especially T helper type 2 (Th2) cells.
Ligand/Receptor
IL-4Rα, IL13RA1, IL13RA2
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Inflammatory Disease
.
Allergic Inflammation
.
Pro-Inflammatory Cytokines

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 Tralokinumab biosimilars are typically used as analytical standards (calibrators) and sometimes as reference controls in pharmacokinetic (PK) bridging ELISA assays to quantify Tralokinumab concentrations in serum samples.

Context and Supporting Details:

  • In PK bridging ELISA for biosimilars, the analytical standard is used to generate a standard curve, which is essential to determine the concentration of Tralokinumab in unknown serum samples by comparison.
  • Research-grade biosimilars, such as those from suppliers like ichorbio, are purified preparations for research use that closely match the originator Tralokinumab in terms of structure and function (e.g., IgG4P-kappa isotype, humanized, CHO expression).
  • In a well-validated PK ELISA, one analytical standard (either the biosimilar or the reference/originator) is selected and used to:
    • Prepare calibration standards (spiked at defined concentrations in matrix, e.g., serum).
    • Generate the standard curve against which test samples (serum from preclinical or clinical studies) are measured.
    • Serve as quality control material when prepared at different concentrations for assay validation (accuracy, precision, reproducibility assessments). Both reference drug and biosimilar may be analyzed in parallel as QC controls when establishing assay comparability and method validation.

Why Use the Biosimilar as Calibrator?

  • Regulatory and industry consensus recommends using a single analytical standard (often the biosimilar, after appropriate comparability assessment) for both the biosimilar and reference product in bridging ELISA to minimize variability and enable direct comparison across all tested samples.
  • Before adopting the biosimilar as the calibration standard, a method qualification study should demonstrate that the biosimilar and the reference produce bioanalytically equivalent results in the PK assay. This usually involves side-by-side validation using both products as controls and quantifying bias, accuracy, and precision.

Practical Workflow in the PK Bridging ELISA:

  • Prepare a series of known concentrations of research-grade Tralokinumab biosimilar in human serum (standard curve).
  • Include quality controls (QCs) made from either the reference product and/or the biosimilar at low, medium, and high levels.
  • Analyze unknown serum samples from PK studies, interpolate their concentrations using the standard curve created from the biosimilar.
  • For assay transfer or bridging, both reference and biosimilar drug are measured across the curve and in QCs to establish equivalency, before adopting the biosimilar as the ongoing calibrator.

Summary Table: Usage in a PK Bridging ELISA

ComponentMaterial UsedRole
Calibration standardsResearch-grade Tralokinumab biosimilarGenerate standard curve, quantify unknown samples
QC controlsReference drug and/or biosimilarMonitor accuracy/precision, compare products
Unknown samplesSerum from study subjectsDetermine Tralokinumab levels using above calibrators

Key References:

  • The approach of using a research-grade biosimilar as the standard is well-documented and aligns with regulatory and industry best practices for demonstrating bioanalytical comparability in biosimilar PK studies.
  • The described biosimilar (e.g., ichorbio’s research-grade Tralokinumab) matches the required specifications for analytical use in ELISA.

The primary models for evaluating the in vivo effects of anti-IL-13 antibodies on tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are mouse syngeneic tumor models, with some studies also employing humanized or xenograft models when assessing human-specific or receptor-targeted agents.

Key model types include:

  • Syngeneic mouse models: These are immunocompetent mice engrafted with mouse tumor cell lines of the same genetic background. Anti-IL-13 antibodies or IL-13 inhibitors are administered to evaluate effects on tumor growth and immune infiltration, allowing direct study of TIL populations and immune modulation. Common models reported in the literature for such studies include:

    • RENCA (renal cell carcinoma)
    • CT26 (colon carcinoma)
    • B16F10 (melanoma)
    • 4T1 (breast cancer)These models have been extensively used to profile therapeutic immune modulation, TIL infiltration, and changes in CD8+ T cells and myeloid populations after cytokine neutralization.
  • Humanized mouse models: Although less frequently used for anti-murine IL-13 targeting, humanized models facilitate testing human-specific anti-IL-13 antibodies. Human tumor xenografts in immunodeficient mice engrafted with components of the human immune system can be used to evaluate TIL responses and tumor growth inhibition, especially for agents directed at human IL-13 or human IL-13Rα2.

  • Xenograft models with IL-13 pathway targeting: Studies using antibodies against human or mouse IL-13Rα2—such as anti-IL-13Rα2 antibody-drug conjugates (ADC)—employ human tumor cell lines xenografted in immunodeficient mice to quantify tumor growth inhibition. Immune cell infiltration is less easily characterized unless the model is humanized for immune system components.

Model utility for TIL analysis:

  • Syngeneic models remain the gold standard for immune profiling of the tumor microenvironment following anti-IL-13 or IL-13 pathway blockade, as they possess intact, functional mouse immune systems and allow detailed analysis of lymphocyte populations via flow cytometry or single-cell sequencing.

  • Humanized models are used when the antibody is human-specific, but technical complexities and higher cost can limit their use for routine TIL characterization.

Summary Table:

Model TypeAntibody SpecificityTumor Growth InhibitionTIL AnalysisKey Notes
Syngeneic mouseMouse anti-IL-13 (or pan)YesRobustMost widely used for immunotherapy and TIL characterization
Humanized mouseHuman anti-IL-13Yes, if engraftedPossibleUsed if testing human-specific antibodies
XenograftIL-13Rα2 targetingYesLimitedImmune analyses limited unless immune system is humanized

References to experimental approaches:

  • Syngeneic model studies: Documented inhibition of tumor growth and increased anti-tumor immunity (via CD8 T cells and reduced suppressive myeloid cells) after IL-13 blockade in RENCA, CT26, and other models.
  • Xenograft (human tumor) models: Used for agents targeting IL-13Rα2, with significant tumor regression demonstrated, but limited assessment of TILs unless combined with humanized immune systems.

In sum, syngeneic mouse tumor models are the primary system used to study anti-IL-13 antibody effects on tumor growth and the immune landscape, with some research extending to humanized mice when necessary for human-specific antibodies.

Researchers use Tralokinumab biosimilars in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to model and analyze potential synergistic effects within complex immune-oncology settings, primarily in preclinical in vitro and in vivo systems. This approach aims to dissect how blocking multiple, distinct immunological pathways can result in more effective tumor immune responses.

Key Experimental Strategies:

  • Model Systems:

    • Researchers typically employ humanized mouse models or cell-based assays to mimic aspects of the human immune-tumor microenvironment.
    • Tralokinumab biosimilars, which neutralize the cytokine IL-13, are administered alongside checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars to study their combined roles in modulating immune activity against tumors.
  • Mechanistic Rationale:

    • Tralokinumab blocks IL-13, a cytokine implicated in driving a protumor, Th2-skewed immune environment (e.g., by dampening Th1 proinflammatory responses and supporting tumor-favorable immune suppression).
    • Checkpoint inhibitors (such as anti-CTLA-4, anti-LAG-3) release brakes on T cells, enhancing their anti-tumor cytotoxicity by counteracting immunosuppressive pathways exploited by tumor cells.
  • Assessing Synergy:

    • Researchers track changes in the subtypes of immune cells activated or suppressed by these therapies (e.g., CD4+ T helper cells, CD8+ cytotoxic T cells, regulatory T cells [Tregs]).
    • For example, in melanoma mouse models, combinations like anti-PD-1/LAG-3 versus anti-PD-1/CTLA-4 have shown distinct effects: anti-LAG-3 regimens reduce Treg activity and promote CD4+ T cell help, while anti-CTLA-4 regimens more directly boost CD8+ cytotoxic responses.
    • Combining Tralokinumab biosimilars with these agents allows investigators to evaluate whether attenuating Th2/IL-13 signaling (which can suppress anti-tumor immunity) potentiates the checkpoint inhibitor-driven activation of effector T cells, leading to more robust tumor rejection.

Experimental Readouts Typically Include:

  • Tumor growth rates in vivo.
  • Immune cell infiltration and phenotype via flow cytometry or immunohistochemistry.
  • Cytokine profiles in the tumor microenvironment.
  • Functional assays (e.g., cytotoxicity, cytokine release).

Context and Limitations:

  • The use of biosimilars (non-therapeutic antibodies that reproduce binding without clinical use) allows for controlled experimentation without direct clinical confounders, making them ideal for mechanism-focused studies in models rather than in patients.
  • While single checkpoint inhibitor regimens are well-characterized, research into their combinatorial effects with agents like Tralokinumab is largely preclinical and aims to identify new therapeutically relevant synergies and underlying immune dynamics.

In summary, by using Tralokinumab biosimilars together with checkpoint inhibitors in complex immune-oncology models, researchers are able to probe how targeting both cytokine signaling (IL-13 pathway) and T-cell checkpoints may yield enhanced anti-tumor immune responses compared to either approach alone, and to dissect the cellular and molecular mechanisms of these combinatorial effects.

A Tralokinumab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against the therapeutic drug by mimicking the binding properties of the original drug, allowing for detection of antibodies generated against Tralokinumab in patient serum.

In a typical bridging ADA ELISA procedure:

  • Capture Reagent: A Tralokinumab biosimilar (which contains the same variable regions as the therapeutic antibody) is immobilized on the ELISA plate (often via biotin-streptavidin interaction). This biosimilar captures ADA from the patient's sample if present.
  • Detection Reagent: After washing, the same Tralokinumab biosimilar conjugated to a detection enzyme (e.g., HRP) is added. If ADA has bound to the immobilized biosimilar, the detection reagent binds to the other "arm" of these bivalent antibodies, forming a "bridge".
  • Result Interpretation: The presence of ADAs is detected via a colorimetric or luminescent reaction produced when the detection reagent interacts with its substrate. The signal intensity correlates with the amount of ADA in the sample.

Using a biosimilar in this manner offers several advantages:

  • It provides a reagent that is structurally and functionally equivalent to the therapeutic Tralokinumab, ensuring high assay specificity for ADAs while avoiding depletion of clinically used drug supply.
  • Because the biosimilar contains identical variable regions, it enables detection of patient antibodies that recognize the original drug's epitope.
  • This approach is standard in immunogenicity testing of monoclonal antibodies, as described in bridging ELISA protocols, which are highly sensitive and allow for high-throughput screening.

Special considerations:

  • The assay may be affected by sample matrix components or soluble drug targets in serum, so high-quality biosimilar reagents and rigorous controls are essential for accurate results.
  • The structural integrity of the biosimilar must be confirmed to accurately replicate antigenicity; Tralokinumab biosimilars are typically purified and characterized using multi-step affinity chromatographies to ensure this.

In summary, a Tralokinumab biosimilar serves as the critical reagent in bridging ADA ELISA assays by acting as both the capture and detection component, enabling sensitive and specific monitoring of ADA responses in patients treated with Tralokinumab.

References & Citations

1 Wynn, TA. et al. (2003) Annu Rev Immunol. 21: 425
2 Kaur D, Hollins F, Woodman L, et al. Allergy. 61(9):1047-1053. 2006.
3 Blanchard C, Mishra A, Saito-Akei H, et al. Clin Exp Allergy. 35(8):1096-1103. 2005.
4 Duggan S. Drugs. 81(14):1657-1663. 2021.
5 Oh CK, Faggioni R, Jin F, et al. Br J Clin Pharmacol. 69(6):645-55. 2010.
6 Singh D, Kane B, Molfino NA, et al. BMC Pulm Med. 10:3. 2010.
B
Indirect Elisa Protocol
Elisa Sandwich Protocol
FA
N

Certificate of Analysis

Formats Available

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