Anti-Human Thymic Stromal Lymphopoietin (TSLP) (Tezepelumab)

Anti-Human Thymic Stromal Lymphopoietin (TSLP) (Tezepelumab)

Product No.: T-2040

- -
- -
Product No.T-2040
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
Active
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 or reference controls in pharmacokinetic (PK) bridging ELISAs to quantitatively measure drug concentrations in serum samples for PK studies and bioanalytical comparability assessments.

Research-grade biosimilars that are structurally and functionally comparable to the reference Tezepelumab are produced for research use only, not for therapeutic or diagnostic administration.

Use in PK Bridging ELISA:

  • Calibration Standard: A research-grade Tezepelumab biosimilar can be employed as the analytical standard to create a standard curve spanning a range of concentrations (e.g., standards at 50–12,800 ng/mL in human serum). This standard curve allows for the quantification of Tezepelumab in unknown serum samples by interpolation.
  • Reference Control: To assess assay accuracy and comparability, both the biosimilar (test) and reference (originator/innovator) Tezepelumab are tested as controls at various concentrations, ensuring the assay equally detects both forms and validating its use for biosimilar studies.

Bioanalytical Strategy and Equivalence Assessment:

  • The current consensus is to develop a single PK assay (a "bridging ELISA"), using a single analytical standard—a research-grade biosimilar—to quantify both reference and biosimilar products in test samples.
  • The method's suitability is confirmed by testing that the assay measures both biosimilar and reference Tezepelumab with equivalent accuracy and precision. Bioanalytical comparability is established if the concentration measurements for the test and reference products fall within pre-defined equivalence criteria (e.g., 0.8–1.25 for the ratio of measured concentrations).

Assay Workflow Summary:

  • Coat ELISA plate with TSLP protein (antigen).
  • Add samples containing Tezepelumab (originator drug or biosimilar) at known concentrations or from study subjects' serum.
  • Detect bound drug with a detection antibody recognizing the human IgG2 Fc domain (Tezepelumab is IgG2).
  • Generate standard curve using the biosimilar standard and interpolate unknown concentrations against this curve.
  • Include quality control samples with both reference and biosimilar products to assess method performance and ongoing comparability.

These practices are standard for bioanalytical support of biosimilar PK studies and conform to regulatory expectations for bridging and comparability studies.

The primary in vivo models used to administer research-grade anti-TSLP antibodies for studies of tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) characterization are:

  • Humanized mouse xenograft models
  • Syngeneic mouse tumor models

Essential context and supporting details:

  • Humanized Xenograft Models:
    In these models, human tumor cells (e.g., breast or colon cancer lines) are implanted into immunodeficient mice that have been reconstituted with human immune cells. Anti-TSLP antibodies are administered to evaluate effects on tumor growth and human immune response within the tumor. For example, administration of anti-TSLP neutralizing antibodies in humanized mice with human breast cancer and human T cells resulted in significant tumor growth inhibition, allowing for characterization of TIL profiles, such as IL13^+^TNF^+^CD4^+^ T cells.

  • Syngeneic Tumor Models:
    These use immune-competent mice with mouse tumor cell lines (e.g., CT26, RENCA, B16F10). While they are not humanized, syngeneic models are valuable for immuno-oncology because they enable assessment of immune-mediated anti-tumor effects and detailed TIL profiling after antibody treatment. Key features include the requirement for cross-reactive or chimeric antibodies compatible with mouse TSLP or its receptor. Studies routinely analyze both TIL composition (CD8, CD4 T cells, myeloid-derived suppressor cells, etc.) and response to immunotherapy interventions—including anti-cytokine antibodies.
    The utility of syngeneic models for immunotherapy and TIL profiling is well documented in the literature.

Additional relevant information:

  • Technical requirements:
    In syngeneic models, anti-TSLP antibodies must recognize the murine target; fully human antibodies require either recombinant mice expressing human TSLP or chimeric agents.

  • Comparisons of immune context:
    Humanized xenograft models enable study of human immune cell tumor infiltration, whereas syngeneic models provide comprehensive analysis of mouse TILs and immune modulation within a fully functional immune system.

  • Tumor types modeled:
    Published studies document use of both colon cancer and breast cancer xenografts. Mouse syngeneic models most commonly use lines such as RENCA (renal), CT26 (colon), and B16F10 (melanoma).

  • Antibody specificity:
    Literature documents the use of both anti-TSLP and anti-TSLPR antibodies for neutralization studies in these models.

Summary Table of Model Use Cases

Model TypeTumor SourceImmune SystemAnti-TSLP Antibody RequirementTIL Characterization
Humanized XenograftHuman cell linesHuman (repop)Human-reactiveHuman TILs
Syngeneic MouseMouse cell linesMouseMouse-reactive/chimericMouse TILs

Key sources:

Researchers use Tezepelumab biosimilar—an antibody targeting Thymic Stromal Lymphopoietin (TSLP)—in combination with checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to study synergistic immunomodulatory effects in immune-oncology models.

Tezepelumab blocks TSLP, a cytokine signaling molecule that influences multiple aspects of inflammation, notably driving Th2 differentiation, regulating Treg development, and supporting B cell class switching. In immune-oncology models, modulating TSLP can affect the tumor microenvironment by decreasing type 2 inflammatory signatures, eosinophilia, and excessive immune suppression through Treg activation.

Checkpoint inhibitors like anti-CTLA-4 and anti-LAG-3 are typically used to unleash T cell responses against tumors by preventing immune escape mechanisms. Anti-CTLA-4 primarily acts in lymph nodes to enhance activation and proliferation of T cells, while anti-LAG-3 modulates T cell exhaustion and function, often in synergy with PD-1/PD-L1 blockade.

Synergistic Immune Modulation

  • Combining Tezepelumab biosimilar with checkpoint inhibitors allows researchers to test whether reducing type 2 inflammation (Th2) and Treg-mediated immune suppression via TSLP blockade can enhance the efficacy of T cell activation from checkpoint inhibitors.
  • Studies have shown that combining different checkpoint inhibitors (e.g., anti-CTLA-4 and anti-PD-1, or anti-LAG-3 and anti-PD-1) activates distinct immune cell subtypes and can yield improved antitumor responses compared to monotherapies.
  • Experimental models often use mouse models of cancer (e.g., melanoma) to systematically manipulate immune pathways. By administering Tezepelumab biosimilar and checkpoint inhibitors either concurrently or sequentially, researchers can assess changes in:
    • Tumor infiltration by immune cells (CD8 cytotoxic T cells, CD4 helper T cells, Tregs).
    • Cytokine profiles and inflammatory biomarkers.
    • Tumor regression and progression-free survival.
  • Combining Tezepelumab biosimilar may augment checkpoint inhibitor efficacy by diminishing immune suppression and amplifying antigen-specific T cell responses within the tumor microenvironment.

Experimental Considerations

  • The non-therapeutic Tezepelumab biosimilar is specifically designed for research, allowing controlled modulation of TSLP without therapeutic confounders.
  • Immune-oncology models leverage combinations to parse mechanistic interactions, such as:
    • Whether TSLP inhibition increases cytotoxic T cell infiltration subsequent to checkpoint blockade.
    • How shifts in Treg and Th2/Th1 ratios affect overall anti-tumor immunity.

Limitations and Future Directions

  • There are not yet large-scale published studies directly combining Tezepelumab biosimilar with checkpoint inhibitors in advanced cancer models; the approach is a logical extrapolation from studies showing additive or synergistic effects when targeting distinct inflammatory and immune escape pathways.
  • Most synergy research is conducted in early-phase preclinical models and will require validation in human systems and clinical trials.

In summary, researchers use Tezepelumab biosimilar in combination with checkpoint inhibitors to dissect how blocking TSLP-mediated inflammation interacts with T cell activation, seeking to overcome tumor immune escape and boost antitumor responses in complex oncology models.

A Tezepelumab biosimilar can be used as the capture and/or detection reagent in a bridging ADA ELISA to detect anti-drug antibodies (ADAs) in patient samples, monitoring the immune response against the therapeutic drug.

In a typical bridging ADA ELISA:

  • The biosimilar Tezepelumab antibody—which shares the same variable regions as the therapeutic Tezepelumab—can be used for either the capture or detection stage, or both, because it binds to the same epitopes on anti-Tezepelumab antibodies as the clinical drug.

Key steps in the assay:

  • Capture reagent: The Tezepelumab biosimilar is immobilized on an ELISA plate (often via direct coating or via biotin-streptavidin if biotinylated).
  • Patient sample: Serum or plasma containing potential anti-Tezepelumab antibodies (ADAs) is added. If present, these ADAs bind to the immobilized biosimilar.
  • Detection reagent: A labeled (e.g., HRP-conjugated or biotinylated) Tezepelumab biosimilar is added, which will bind to the other arm of the ADA, thus “bridging” the two biosimilar molecules through the bivalent nature of the antibody.

This design ensures:

  • Only bivalent antibodies (i.e., the patient's ADAs against Tezepelumab) will create the bridge between the capture and detection biosimilar reagents, giving a specific readout for anti-Tezepelumab antibodies.
  • The biosimilar is preferred over the therapeutic Tezepelumab to avoid cross-reactivity with the original drug present in patient samples, and because its matching variable regions ensure equivalent epitope recognition.

Additional details:

  • Use of high-quality biosimilar reagents is crucial for specificity and sensitivity, as the ADA ELISA can be affected by matrix factors and presence of circulating drug.
  • The chromogenic substrate (such as TMB) is then added to react with the HRP-enzyme on the detection reagent, allowing quantification based on colorimetric change.

In summary, the Tezepelumab biosimilar acts as a surrogate reagent in the bridging ADA ELISA, providing selective detection of patient immune responses (ADAs) directed against the therapeutic Tezepelumab.

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.