Anti-Human Thymic Stromal Lymphopoietin (TSLP) (Tezepelumab) – Fc Muted™

Anti-Human Thymic Stromal Lymphopoietin (TSLP) (Tezepelumab) – Fc Muted™

Product No.: T-2045

- -
- -
Product No.T-2045
Clone
MEDI9929
Target
TSLP
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Thymic stromal lymphopoietin
Isotype
Human IgG2λ
Applications
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
Immunogen
Unknown
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,
FA
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 Tezepelumab. MEDI9929 (Tezepelumab; AMG157) activity is directed against human TSLP.
Background
Thymic stromal lymphopoietin (TSLP) is a pleiotropic, epithelium-derived cytokine that is characterized as a lymphocyte growth factor1. TSLP plays a key role in the inflammatory response of the lung, such as in asthma and COPD pathobiology. To function, TSLP engages a heteromeric complex of TSLP receptor chain and interleukin 7 receptor-α (IL-7Rα) to induce STAT5. TSLP can be expressed as either a long or short isoform. It is the long isoform that is specifically upregulated by inflammatory stimuli in human bronchial epithelial cells and macrophages. In contrast, the short isoform is constitutively expressed in bronchial and colonic epithelial cells, lung fibroblasts, macrophages, and keratinocytes. TSLP mediates airway structural cell and immune cell interactions 2 . TSLP activity is upstream of the asthma inflammatory cycle making it an effective target for treatment.

MEDI9929 (Tezepelumab) is a fully human monoclonal antibody developed for the treatment of asthma, COPD, chronic rhinosinusitis with nasal polyps, chronic spontaneous urticaria, and eosinophilic esophagitis2. Tezepelumab inhibits the action of TSLP by blocking interaction with its receptor TSLPR. Tezepelumab reduces T2 inflammatory biomarkers, including airway eosinophil counts, when administered in vivo3,4,5.
Antigen Distribution
TSLP is mainly expressed by bronchial epithelial cells. TSLP also selectively localizes to the cytoplasm of human lung macrophages (HLM) and can be released by HLMs. Additionally, TSLP is expressed by human dendritic cells, airway smooth muscle cells, mast cells, monocytes, granulocytes, and fibroblasts.
Ligand/Receptor
TSLP receptor (TSLPR) chain and interleukin 7 receptor-α
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Inflammatory Disease
.
Allergic Inflammation
.
Asthma
.
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 Tezepelumab biosimilars are commonly used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantitatively measure drug concentrations in serum samples during biosimilar development and comparability studies. They provide a standardized reference point for quantification and assure consistent assay performance.

Context and Application:

  • In a PK bridging ELISA, the quantitation of Tezepelumab (the reference monoclonal antibody) in serum is based on a standard curve constructed using known concentrations of a research-grade Tezepelumab biosimilar.
  • Biosimilar qualification: Regulatory and industry best practices recommend that, for biosimilar development, a single PK assay using a single analytical standard quantifies both the biosimilar and the reference product in the same assay. This approach reduces between-assay variability, eliminates the need for cross-validation, and ensures all test samples are measured under identical analytical conditions.
  • The research-grade Tezepelumab biosimilar is serially diluted in blank human serum to generate calibration standards at defined concentrations (e.g., 50–12,800 ng/mL). These standards are run alongside unknown serum samples.
  • Reference controls (prepared from either the biosimilar or the originator drug) are also included as quality control (QC) samples to monitor assay performance and verify method accuracy and precision.

Bridging the Innovator and Biosimilar:

  • Before using a biosimilar as the standard, bioanalytical equivalence between the biosimilar and innovator drug must be rigorously established within the assay. This involves side-by-side testing to ensure that both molecules behave equivalently in the ELISA, and statistical analysis (e.g., comparing precision and accuracy data, calculating 90% confidence intervals for the ratio of measured concentrations, and confirming they fall within pre-specified equivalence margins).

Assay Function:

  • The ELISA is typically configured so that Tezepelumab present in serum samples (or in prepared standards) binds to a capture antigen (e.g., immobilized human TSLP protein), and is then detected with a specific anti-human antibody. The readout is compared to the standard curve generated from the biosimilar calibrators to quantify drug levels in unknowns.

Summary Table: Use of Tezepelumab Biosimilar as PK ELISA Standard

StepPurposeDetails
Biosimilar prepares std. curveCalibration for quantificationSerial dilutions in serum to build a standard concentration-response curve
Reference controlsQC and assay performance checkAliquots of biosimilar or originator at known concentrations
Equivalence testingConfirm suitability as universal standardPrecision & accuracy comparisons; statistical equivalence checks
Sample quantitationMeasure Tezepelumab in test serumCompare unknowns to standard curve; report concentration using biosimilar standard

Key points:

  • Use of a single, research-grade biosimilar standard is now the consensus best practice for PK bridging ELISA in biosimilar programs.
  • The biosimilar is not for clinical or diagnostic use—it is exclusively for research and assay development.
  • Independent validation and rigorous comparability testing are required before routine use as a calibrator.

This strategy supports regulatory submissions by standardizing PK measurements and ensuring robust comparison between biosimilar and reference product concentrations in preclinical and clinical matrices.

The most common models for studying research-grade anti-TSLP antibody administration in vivo for tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are:

  • Humanized xenograft models
  • Syngeneic mouse models

Model Details

1. Humanized Xenograft Models

  • These use immunodeficient mice engrafted with human tumor cells and human immune cells.
  • Anti-TSLP antibodies have been administered to study the inhibition of tumor growth and immune cell infiltration, particularly T and myeloid cell populations.
  • Example: In breast cancer, humanized mice reconstituted with cancer cells and human T cells received anti-TSLP antibodies, resulting in inhibition of tumor development and altered TIL profiles.

2. Syngeneic Mouse Models

  • These involve transplanting murine tumor cells into genetically identical, immune-competent mice.
  • They allow investigation of how anti-TSLP antibodies modulate a fully functional mouse immune system, including TILs.
  • These require antibodies cross-reactive to mouse TSLP or use genetically engineered mice expressing human TSLP or TSLP receptors.
  • Common models include TC-1 and MC38 (colon cancer), which are fully characterized for TIL populations and immune responses.

Considerations

  • Syngeneic models are vital for immunotherapy research as they better reflect the complexity of immune cell-tumor interactions, including TIL composition and response to immunomodulatory agents like anti-TSLP antibodies.
  • Humanized models are essential when studying therapies targeting human-specific antigens and antibody responses, especially if the anti-TSLP antibody does not cross-react with the murine protein.
  • Both models can be tailored using genetically engineered or chimeric antibodies that recognize either human or mouse TSLP for improved utility in immune profiling studies.

In summary: The primary in vivo models for anti-TSLP antibody evaluation in tumor growth and TIL characterization are humanized xenograft systems and syngeneic mouse models—each selected based on species-specific reactivity and research goals.

Researchers utilize the Tezepelumab biosimilar—a non-therapeutic, research-grade antibody that targets thymic stromal lymphopoietin (TSLP)—in combination studies with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to investigate potential synergistic effects on anti-tumor immunity within immune-oncology models.

Context and Rationale:

  • TSLP is an epithelial-derived cytokine central to regulating immune responses, influencing dendritic cell (DC) function, Th2 differentiation, antibody production, Treg development, and other immune processes relevant to both allergy and cancer immunobiology.
  • Checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 antibodies act on different immunological axes, primarily by releasing inhibition of T cells—thereby facilitating more robust anti-tumor responses.

Experimental Approaches:

  • Co-administration in Murine or Humanized Models: Tezepelumab biosimilar is administered alone or together with agents like anti-CTLA-4 or anti-LAG-3 in in vivo cancer models (e.g., melanoma, lung, or solid tumor xenografts).
  • Immune Profiling: Researchers track changes in tumor-infiltrating lymphocytes, such as CD8+ cytotoxic T cells, CD4+ helper T cells, and regulatory T cells (Tregs), to distinguish the individual and combined immunomodulatory effects of TSLP blockade and checkpoint inhibition.
  • Synergy Assessment: Synergistic effects are assessed by monitoring enhanced tumor rejection, increased T cell activation, changes in cytokine profiles, and reduced Treg-mediated suppression. For example, anti-LAG-3 and anti-CTLA-4 exploit different mechanisms: the former increases CD4 helper T cell activity upstream, while the latter enhances proliferation and activation of cytotoxic T cells at different immune checkpoints.

Underlying Scientific Logic:

  • TSLP as an Upstream Target: Blocking TSLP may suppress immunosuppressive microenvironment conditions (e.g., Th2-dominated inflammation, Treg induction), potentially making tumors more susceptible to the effects of checkpoint inhibitors.
  • Checkpoint Blockade Synergy: Anti-CTLA-4, anti-LAG-3, and anti-PD-1/PD-L1 inhibitors work at different immunological “checkpoints”—combining these with TSLP blockade could counteract multiple, non-redundant immunosuppressive pathways in the tumor microenvironment, yielding additive or synergistic anti-tumor effects.

Relevant Experimental Tools and Products:

  • Commercial research-grade Tezepelumab biosimilar antibodies are available to specifically block human TSLP in in vitro or in vivo systems, facilitating the study of these combination regimens for research purposes rather than direct clinical use.

Current Limitations:

  • Most data on synergy come from preclinical mouse models with engineered immune systems or ex vivo human cell assays, due to the non-clinical nature of available biosimilar reagents.
  • Detailed, published examples specifically combining Tezepelumab biosimilars with checkpoint inhibitors in immune-oncology animal models remain limited, and ongoing research is elucidating the best models and dosing regimens for combination therapy.

In summary, researchers co-administer Tezepelumab biosimilar with checkpoint inhibitor biosimilars in complex immune-oncology models to dissect the interplay of epithelial-initiated cytokine signaling and adaptive immune inhibition, probing whether dual or multi-pathway blockade enhances anti-tumor immune responses compared to monotherapies.

In immunogenicity testing, a Tezepelumab biosimilar can serve as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA, enabling the monitoring of a patient’s immune response against Tezepelumab.

Bridging ADA ELISAs are designed to detect anti-Tezepelumab antibodies (i.e., ADAs) generated by the patient in response to treatment. The general principle involves the drug itself (or a biosimilar with identical variable regions to the therapeutic drug) being used on both sides of the immunoassay "bridge":

  • As a capture reagent, Tezepelumab biosimilar is immobilized (e.g., on a microtiter plate or via biotin-streptavidin interaction) and binds to one arm of any ADA present in the patient’s serum sample.
  • After a wash step, a detection reagent—typically Tezepelumab biosimilar conjugated to an enzyme (e.g., HRP) or tagged with biotin or a dye—binds the other arm of the ADA, thus forming a "bridge": plate-bound Tezepelumab biosimilar – ADA – labeled Tezepelumab biosimilar.
  • The presence of this bridge is then detected through colorimetric or luminescent substrate conversion by the enzyme label, indicating ADAs in the sample.

Key technical characteristics:

  • The biosimilar must have identical antigen-binding regions (“variable regions”) as the therapeutic Tezepelumab, ensuring specific recognition of ADA against the drug.
  • Using a biosimilar (as opposed to the clinical drug substance) allows for high-throughput, reproducible, and cost-effective ADA screening strictly for research purposes, without impacting clinical supply.
  • Both capture and detection reagents may be the biosimilar, differing only by their tag or method of plate attachment.

Practical example:

  • Tezepelumab biosimilar is coated onto a plate (capture). Patient serum containing ADA is incubated, allowing ADAs to bind (if present).
  • Next, another portion of Tezepelumab biosimilar, conjugated with HRP (detection), is added.
  • Color change after incubation and substrate addition reflects ADA presence and quantity.

This ELISA format is highly sensitive for bivalent ADAs and is widely used in immunogenicity assessments for monoclonal antibody therapeutics. Assay specificity depends heavily on reagent purity and the similarity of the biosimilar to the clinical drug. Blocking steps and optimized buffers are critical to minimize background noise from serum matrix components.

In summary, Tezepelumab biosimilar is used as both a capture and detection reagent in ADA bridging ELISAs, directly facilitating quantitation of patient immune responses (ADAs) to the drug via the specificity of the biosimilar’s antigen-binding regions.

References & Citations

1 Canè L, Poto R, Palestra F, et al. Eur J Intern Med. 124:89-98. 2024.
2 Hoy SM. Drugs. 82(4):461-468. 2022.
3 Gauvreau GM, O'Byrne PM, Boulet LP, et al. N Engl J Med. 370(22):2102-2110. 2014.
4 Diver S, Khalfaoui L, Emson C, et al. Lancet Respir Med. 9(11):1299-1312. 2021.
5 Corren J, Pham TH, Garcia Gil E, et al. Allergy. 77(6):1786-1796. 2022.
6 Zheng Y, Abuqayyas L, Megally A, et al. Clin Ther. 43(1):142-155.e5. 2021.
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

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