Anti-Human IL-13 (Tralokinumab) [Clone CAT-354]

Anti-Human IL-13 (Tralokinumab) [Clone CAT-354]

Product No.: I-2110

- -
- -
Product No.I-2110
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
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 ?
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 frequently employed as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately measure drug concentrations in serum samples. This use is central to developing bioanalytical methods that are both comparable and robust in quantifying both biosimilar and reference (originator) antibodies.

Mechanism and Rationale:

  • In a typical PK bridging ELISA, a standard curve is generated by spiking known concentrations of the Tralokinumab biosimilar into serum. This standard curve is then used to interpolate the concentrations of Tralokinumab present in test serum samples from clinical or preclinical studies.
  • Research-grade biosimilars (RUO) are produced to mirror the structure and bioactivity of the reference product, but are designated for research applications (not for human therapeutic use).
  • Analytical methods are validated using this biosimilar as the standard to ensure accurate, precise, and linear detection of Tralokinumab across a biologically relevant range of concentrations. The method is stressed to demonstrate bioanalytical equivalence between the biosimilar and reference standard within pre-defined statistical boundaries (typically, 0.8 to 1.25 for the 90% confidence interval of the ratio of the means).

Workflow Details:

  • Calibration Standard: The biosimilar is serially diluted in serum to create a concentration curve, typically covering a range such as 50–12,800 ng/mL, depending on the expected concentrations in study samples.
  • Reference Controls: Additional serum samples are spiked with known amounts of either biosimilar or reference Tralokinumab to serve as quality control (QC) samples for accuracy and precision checks within the assay run.
  • Single-Standard Approach: Industry best practice favors developing a single PK assay (using either the biosimilar or reference as the standard) to minimize variability. If the biosimilar and reference product are analytically equivalent within the assay, the biosimilar itself may be selected as the standard for routine quantification.

Regulatory and Scientific Rigor:

  • Comprehensive method qualification and validation studies are required to demonstrate that the assay provides consistent, reproducible quantification of both biosimilar and reference products.
  • The calibration and QC samples span the physiological and expected clinical range, and the same batch of research-grade Tralokinumab biosimilar is used throughout validation and sample testing to ensure assay integrity.

Summary Table

PurposeRole of Tralokinumab Biosimilar in ELISA
Calibration StandardUsed to establish the standard curve for quantifying unknown serum samples
Reference ControlSpiked into serum to generate QC samples for accuracy and precision
Analytical EquivalenceUsed to demonstrate assay can robustly detect both biosimilar & reference

This single-standard, bridging assay strategy optimizes accuracy and reduces assay variability, which is crucial for PK studies supporting biosimilar development and regulatory submission.

The primary in vivo models where a research-grade anti-IL-13 antibody is administered to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are:

  • Syngeneic mouse models (using immunocompetent mice and murine tumors).
  • Humanized mouse models (using immunodeficient mice reconstituted with human immune cells and engrafted with human tumors).

Supporting context and details:

  • Syngeneic models involve implanting mouse tumor cell lines into genetically matched, immunocompetent mice, preserving the native murine immune system. These models allow direct assessment of how anti-IL-13 antibody therapy affects both tumor growth and the murine TIL compartment, as the full spectrum of immune responses is intact. Multiple studies have used IL-13 inhibition in wild-type or genetically modified mouse strains (e.g., IL-4 KO, Stat6 KO, CD1d KO) to demonstrate reduced tumor growth and enhanced anti-tumor immunity, sometimes supported by IL-13 inhibitory antibodies.
  • Humanized mouse models are immunodeficient mice (e.g., NSG) engrafted with human hematopoietic stem cells or peripheral blood mononuclear cells, sometimes in combination with human tumor xenografts. These models more faithfully reproduce human immune-tumor interactions, and anti-IL-13 antibodies directed against human IL-13 can be evaluated for both tumor inhibition and effects on human TILs.
  • Patient-derived xenografts (PDX) models and standard xenograft models can also be used, but unless humanized, only the tumor cells are of human origin and the immune system is murine or severely immunodeficient, limiting TIL characterization to mouse lymphocytes or none at all. Thus, xenograft models without humanized immune systems are less preferred for TIL phenotyping.
  • Studies using anti-IL-13 antagonists or antibodies frequently observe delayed tumor growth, increased cytotoxic T lymphocyte (CTL) activity, and altered TIL composition in tumors, supporting the immunomodulatory effect of IL-13 blockade in the TME.

Summary of advantages:

ModelTumor originImmune systemSuitability for anti-IL-13/TIL analysis
SyngeneicMouseMouse (intact)High: allows TIL and tumor analyses
HumanizedHumanHuman (reconstituted)High: enables human-specific antibody testing/TILs
Xenograft/PDXHumanMouse (deficient)Low: poor for immune/TIL studies unless humanized

Key references:

  • For syngeneic and humanized models enabling TIL and tumor inhibition studies: .
  • For specific tumor studies using IL-13 inhibition: .

If you require protocols, tumor models, or antibody sources, please clarify which tumor types or antibody clones are of interest.

Researchers use Tralokinumab biosimilar (an anti-IL-13 antibody) in combination studies with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to investigate possible synergistic effects on immune activation and tumor response in complex immune-oncology models.

Context and Experimental Approach:

  • Tralokinumab biosimilar neutralizes IL-13, a cytokine that biases the immune system toward a Th2 (allergic/inflammatory) profile, often associated with dampening of anti-tumor Th1 responses.
  • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) release inhibition (“brakes”) on T cells, enhancing their activation and the immune attack on cancer cells.
  • In combination, blocking IL-13 with Tralokinumab biosimilar may reduce immune suppression mediated by Th2 cytokines, potentially shifting the balance to a more robust Th1-driven anti-tumor response — which can be further amplified by checkpoint blockade.

Typical Experimental Designs:

  • In vivo studies: Researchers use humanized mouse models or syngeneic tumor models to assess tumor growth, immune cell infiltration, cytokine profiles, and overall survival when combining Tralokinumab biosimilar with checkpoint inhibitors.
  • Immune cell profiling: Flow cytometry and single-cell RNA sequencing are used to analyze activation states and subtypes of immune cells, such as CD8+ cytotoxic T cells and CD4+ helper T cells, in response to combination treatment.
  • Synergy assessment: Functional assays measure enhanced cytokine production, increased tumor cell killing, or reduction of regulatory T cell-mediated immunosuppression following dual antibody treatments.

Mechanistic Rationale for Synergy:

  • Anti-IL-13 (Tralokinumab biosimilar):
    • Counteracts pro-tumor Th2 immune bias.
    • Potentially boosts interferon-gamma (IFNγ) production and Th1 differentiation.
  • Checkpoint inhibitors (CTLA-4/LAG-3 biosimilars):
    • Release T cells from inhibitory signals, enhancing both helper and cytotoxic responses.
ComponentPrimary EffectSynergistic Potential
Tralokinumab biosimilarBlocks IL-13 (reduces Th2 bias)Increases Th1/IFNγ activity
Anti-CTLA-4 biosimilarBlocks CTLA-4 inhibitionEnhances T cell priming/activation
Anti-LAG-3 biosimilarBlocks LAG-3 inhibitionBoosts CD4+, CD8+ T cell activity

Application in Research:

  • Researchers typically use research-grade biosimilars of Tralokinumab alongside other checkpoint inhibitors in combination dosing protocols in preclinical models to measure tumor immune microenvironment modulation and therapeutic efficacy.
  • Detailed mechanistic studies evaluate how each agent affects distinct immune cell populations, signaling pathways (e.g., JAK/STAT), and resistance mechanisms within the tumor niche.

In summary: The use of Tralokinumab biosimilar with checkpoint inhibitors allows researchers to dissect how neutralizing Th2 cytokines and releasing T cell checkpoints can collaboratively enhance anti-tumor immune responses in advanced immune-oncology models.

A Tralokinumab biosimilar is commonly used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response to Tralokinumab therapy. In this assay, the biosimilar functions identically to the originator drug in its ability to bind anti-drug antibodies, ensuring the assay specifically detects antibodies directed against Tralokinumab itself.

Key steps and rationale:

  • In a standard bridging ADA ELISA, the bivalent nature of ADAs is leveraged: ADAs can bind to two molecules of the drug simultaneously.
  • The Tralokinumab biosimilar is:
    • Immobilized/coated on the plate as the capture antigen, or
    • Biotinylated/HRP-labeled and used as the detection reagent.

Assay workflow:

  • Patient serum is incubated in wells pre-coated with the Tralokinumab biosimilar (capture reagent).
  • If anti-Tralokinumab antibodies (ADAs) are present in the patient sample, they will bind to the coated drug.
  • After washing, a solution of labeled Tralokinumab biosimilar (detection reagent) is added. ADAs, if present, will "bridge" between the immobilized and labeled biosimilar, forming a drug-ADA-drug sandwich.
  • This complex is then detected via the associated label (e.g., HRP or biotin-streptavidin interaction), producing a quantifiable signal.

Significance of using a biosimilar:

  • Tralokinumab biosimilars are structurally and functionally comparable to the therapeutic drug, ensuring they bind the same ADA populations as the original drug.
  • Using a biosimilar preserves valuable clinical drug supply and can be more readily available in research-grade form for assay optimization.

Why is bridging format used?

  • The bridging format is highly specific for bivalent (IgG-type) ADAs.
  • It avoids detection of non-specific serum antibodies, and the use of the same (or biosimilar) drug in both capture and detection ensures specificity for antibodies formed against the therapeutic drug.

Summary Table: Use of Tralokinumab Biosimilar in Bridging ADA ELISA

Assay RoleFunction
Capture reagentImmobilized on plate to bind ADA from patient serum
Detection reagentLabeled form detects ADA by bridging with captured ADA (forms sandwich complex)

In conclusion, a Tralokinumab biosimilar can be used as either the capture or detection reagent in a bridging ADA ELISA, because it faithfully mimics the drug's immunological epitopes, enabling specific and sensitive detection of anti-drug antibodies in patient samples.

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.