Anti-Human OX40L (Oxelumab) [Clone R4930] — Fc Muted™

Anti-Human OX40L (Oxelumab) [Clone R4930] — Fc Muted™

Product No.: LT1305

- -
- -
Product No.LT1305
Clone
R4930
Target
TNFSF4
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
TNFSF4; GP34; CD252; TXGP1; CD134L; TNLG2B
Isotype
Human IgG1κ
Applications
B
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Original antibody raised against Human OX40L
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.
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Oxelumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Applications Reported In Literature ?
FA
B
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 Oxelumab. Clone R4930 binds to Human OX40L (TNFSF4). This product is for research use only.
Background
Oxelumab is a human monoclonal antibody designed for the treatment of autoimmune diseases.3 Oxelumab recognizes human OX40L (TNFSF4). OX40L is a member of the tumor necrosis family and is the ligand for OX40 . The OX40/OX40L interaction generates an optimal T cell response and plays a significant role in determining the amount of memory T-cells remaining after the immune response.1 Therapeutic treatments with antibodies against TNFSF can sometimes result in serious side effects.2 More research is needed to understand the precise molecular mechanism of TNF inhibition. This cost-effective, research-grade Anti-Human OX40L (Oxelumab) utilizes the same variable regions from the therapeutic antibody Oxelumab making it ideal for research projects.
Antigen Distribution
OX40L is expressed in various cell types including antigen presenting cells, T-cells, vascular endothelial cells, mast cells, and natural killer cells.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars
.
Costimulatory Molecules
.
Immunology

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.

To use research-grade Oxelumab biosimilars as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA for measuring drug concentration in serum samples, several steps and considerations are involved:

Principle of the Assay

  • Quantitative Competitive Enzyme Immunoassay: This method involves pre-coating a microplate with a specific protein, such as recombinant human CD252, if relevant to Oxelumab's mechanism. Standards or samples are premixed with a biotin-labeled antibody and pipetted into the wells. Oxelumab in the sample competitively binds to the pre-coated protein, and after washing, Streptavidin-HRP is added to detect bound Oxelumab. The color development, inversely proportional to the amount of Oxelumab bound, is measured colorimetrically.

Preparation and Use as Calibration Standards:

  1. Biosimilar Preparation: The biosimilar Oxelumab is prepared in a serum matrix at various concentrations to create a standard curve. This curve serves as a reference for quantifying unknown samples.
  2. Calibration and Reference Control: The biosimilar is used as both a calibration standard and a reference control. It is crucial to ensure that the biosimilar is highly similar to the reference product, as established through analytical biosimilarity studies.
  3. Assay Validation: The PK assay is validated by assessing precision and accuracy using multiple runs of standards and quality control samples. This validation ensures that the biosimilar can serve as a reliable analytical standard.

Key Considerations:

  • Bioanalytical Equivalence: It is essential to establish bioanalytical equivalence between the biosimilar and the reference product. This involves demonstrating that both products behave similarly within the assay.
  • Single Assay Methodology: Using a single PK assay for both biosimilar and reference products helps reduce variability and eliminates the need for complex crossover analyses.
  • Regulatory Compliance: The development and validation of PK assays must comply with regulatory guidelines, ensuring that the methods are precise, accurate, and robust.

By following these considerations and using a well-validated assay, research-grade Oxelumab biosimilars can effectively serve as calibration standards and reference controls in PK bridging ELISAs to measure drug concentrations in serum samples.

To study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) using a research-grade anti-TNFSF4 (OX40L) antibody, researchers often employ both syngeneic and humanized models. Here's an overview of these models and their use in tumor growth studies:

Syngeneic Models

Syngeneic models involve transplanting tumor cells from one mouse into another mouse of the same genetic background. These models are particularly useful for studying anti-cancer immunotherapies because they allow for the evaluation of immune responses in a fully functional immune system.

  • Key Features: Syngeneic models are well-characterized for gene expression, baseline TIL populations, and responses to common immune checkpoint inhibitors. They enable detailed analysis of immunotherapy efficacy and provide insights into how treatments interact with the immune system to target cancer cells.
  • Models Used: Commonly used syngeneic models include MC38, CT26, EMT6, RENCA, and B16F10. These models vary in their immune infiltration profiles, which helps in understanding how different immunotherapies work across various tumor types.
  • OX40L Inhibition: Inhibition of the OX40-OX40L interaction could be explored in these models to study its effects on tumor growth and TILs, although specific studies on anti-TNFSF4 antibodies in these models are limited.

Humanized Models

Humanized models, typically in immunodeficient mice, involve engrafting human immune cells or tissues. These models are crucial for studying human immune interactions with tumors.

  • Key Features: Humanized models allow for the study of human-specific immune responses and the interaction between human immune cells and tumors. They are particularly useful for evaluating therapies that target human-specific epitopes or immune cells.
  • Use in Tumor Growth Studies: While humanized models are widely used for studying tumor growth and immune responses, specific studies on the administration of anti-TNFSF4 antibodies for tumor growth inhibition are not extensively documented in the provided literature. However, humanized models are essential for evaluating the efficacy of novel immunotherapies, including those targeting the OX40-OX40L pathway.

In summary, while syngeneic models are more commonly used for studying tumor growth inhibition and TIL characterization due to their fully functional immune systems, humanized models offer insights into human-specific immune interactions. Both models are crucial for preclinical studies, but specific research on anti-TNFSF4 antibodies in these contexts may require further investigation.

Researchers utilize oxelumab biosimilars in combination with other checkpoint inhibitors to investigate complex immune interactions and potential synergistic therapeutic effects in immune-oncology research. The strategic use of these biosimilar combinations provides valuable insights into how different immune pathways can be modulated simultaneously to enhance anti-tumor responses.

Understanding Oxelumab's Mechanism in Combination Research

Oxelumab biosimilars target the OX40L-OX40 interaction, which plays a crucial role in T cell activation and immune memory formation. This human monoclonal antibody blocks the binding between OX40L (CD252) and its receptor OX40 (CD134) on T cells, effectively modulating immune responses by preventing T cell activation and proliferation. The mechanism involves disrupting key signaling pathways including the NF-κB pathway and PI3K/Akt signaling, which are essential for T cell survival and function.

Combination Strategies with Different Checkpoint Inhibitors

Research demonstrates that different checkpoint inhibitor combinations operate through distinct mechanisms of action, making them valuable for studying synergistic effects. The combination approaches typically focus on:

Anti-PD-1/CTLA-4 Combinations: These regimens work primarily through direct activation of cytotoxic CD8 T cells, leading to increased accumulation of these tumor-killing cells without requiring CD4 T cell presence. When combined with oxelumab biosimilars, researchers can study how blocking the OX40L-OX40 pathway affects this direct CD8 T cell activation.

Anti-PD-1/LAG-3 Combinations: These combinations require CD4 T cell presence and work by decreasing regulatory T cell (Treg) activity while increasing CD4 helper T cell activity, ultimately leading to CD8 T cell activation. The addition of oxelumab biosimilars to this regimen allows researchers to investigate how simultaneous modulation of the OX40L pathway influences the CD4-dependent immune response.

Research Applications in Complex Models

Investigators use these biosimilar combinations in sophisticated experimental systems, particularly mouse models of melanoma and melanoma brain metastases, to identify the exact immune cell populations that become activated during treatment. The research focuses on understanding how different checkpoint inhibitor combinations affect various T cell subsets, including CD8 cytotoxic T cells, CD4 helper T cells, and regulatory T cells.

The cost-effective nature of research-grade biosimilars, such as the oxelumab biosimilar that utilizes the same variable regions as the therapeutic antibody, makes them ideal for comprehensive research projects investigating multiple pathway interactions. This allows researchers to systematically study how blocking the OX40L-OX40 costimulatory pathway synergizes with other checkpoint inhibitors to enhance overall anti-tumor immune responses.

Mechanistic Insights and Future Directions

Current research indicates that targeting multiple checkpoints can increase the activity of each other, potentially overcoming individual pathway limitations. By combining oxelumab biosimilars with other checkpoint inhibitors, researchers can investigate whether simultaneous modulation of costimulatory (OX40L-OX40) and inhibitory (PD-1, CTLA-4, LAG-3) pathways creates synergistic effects that are superior to single-agent therapies.

The growing focus on combination immunotherapy agents targeting multiple pathways reflects the understanding that complex immune responses require sophisticated intervention strategies. These biosimilar-based research approaches provide crucial mechanistic insights that inform the development of more effective combination therapies for cancer treatment.

In a bridging ADA ELISA for immunogenicity testing of Oxelumab biosimilars, the biosimilar drug is used as either the capture or detection reagent to monitor a patient’s immune response (specifically, the presence of anti-drug antibodies or ADAs) against the therapeutic drug.

Essential context and details:

  • In the bridging ADA ELISA, both the capture and detection reagents are usually the therapeutic drug (in your case, the Oxelumab biosimilar), but labeled differently. One form (e.g., biotinylated Oxelumab biosimilar) is immobilized on the plate and serves to capture ADAs present in a patient's serum sample. The other form (e.g., HRP- or dye-conjugated Oxelumab biosimilar) is used to detect the ADAs that have bound to the immobilized drug.

  • The principle is based on the bivalent nature of ADAs: an ADA molecule can bind two identical epitopes. Thus, ADAs can "bridge" between the capture and detection reagents, forming a detectable complex. Oxelumab biosimilar in both roles (capture and detection) provides a matched pair for this bridging to occur.

  • By using the biosimilar version (rather than the originator drug), the assay specifically measures the immune response against the biosimilar itself, which is crucial for monitoring immunogenicity in patients treated with biosimilars.

  • This approach is highly sensitive but requires careful assay optimization, as serum matrix components, target molecules, or free drug can interfere with the specificity of ADA detection.

  • The measured signal (e.g., via a chromogenic substrate for HRP) reflects the level of ADA present in the patient’s sample that can recognize and bind the Oxelumab biosimilar, thus indicating immunogenicity.

Additional relevant information:

  • Similar approaches are used for other therapeutic mAbs: the biosimilar can precisely replicate the originator’s structure, so ADAs detected by the biosimilar reagent generally reflect the patient’s immune response to the active drug, allowing for head-to-head immunogenicity comparison.

  • Regulatory guidance recommends the use of sensitive and specific assay formats (such as bridging ELISA) to accurately assess ADA incidence and impact in biosimilar development.

In summary, an Oxelumab biosimilar is used in both capture and detection roles in a bridging ADA ELISA so that the assay specifically detects antibodies the patient’s immune system may have produced against the biosimilar therapeutic agent, supporting immunogenicity monitoring in clinical and post-marketing studies.

References & Citations

1. Mahmood,T. and Yang, P. (2012) N Am J Med Sci. 4(11): 533–536
2. Spicer, P. and Runkel, L. (2019) Expert Opin Investig Drugs. 28(2):99-106
3. Heo, YS. et al. (2014) Bio Design 2(2):55-61
B
FA
Flow Cytometry

Certificate of Analysis

Formats Available

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