Anti-Human IL-2Rα (CD25) (Basiliximab) [Clone Hu107] — Fc Muted™

Anti-Human IL-2Rα (CD25) (Basiliximab) [Clone Hu107] — Fc Muted™

Product No.: LT305

- -
- -
Product No.LT305
Clone
Hu107
Target
CD25
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-2-RA; IL2-RA; TAC antigen; p55; CD25
Isotype
Human IgG1κ
Applications
B
,
CyTOF®
,
FC
,
IF
,
IHC
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Rhesus Monkey
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human CD25
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 Basiliximab 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 ?
WB
B
IF
IHC
Additional Reported Applications For Relevant Conjugates ?
CyTOF®
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 Basiliximab. Basiliximab recognizes human CD25. This product is for research use only.
Background
CD25, a 55 kD type I transmembrane glycoprotein, has been shown to play roles in lymphocyte differentiation, activation, and proliferation. Many resting memory T cells constitutively express IL2Rα. It functions as the receptor for HTLV-1, resulting in its expression on neoplastic cells in adult T cell lymphoma/leukemia. CD25 (sIL-2R) has been used to track disease progression. Some additional clinical applications include Chagas disease, a disease characterized by a decline of CD25 expression on immune cells, and Multiple sclerosis, in which treatments with mAbs target CD25. Anti-Human IL-2R alpha (CD25) (Basiliximab) utilizes the same variable regions from the therapeutic antibody Basiliximab making it ideal for research projects.
Antigen Distribution
IL-2Rα is expressed on activated mature T and B lymphocytes, during early stages of thymocytes development, pre-B cells, and in activated CD4+ memory T-lymphocytes.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars

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.

Role of Basiliximab Biosimilars in Pharmacokinetic (PK) Bridging ELISA

Overview of Basiliximab Biosimilars

Basiliximab biosimilars are used in various analytical studies, including pharmacokinetic (PK) bridging assays. These biosimilars are designed to mimic the structure and function of the reference product, in this case, Basiliximab, which is a monoclonal antibody used for immunosuppression in kidney transplant patients.

Use in Pharmacokinetic (PK) Bridging ELISA

In PK bridging ELISAs, biosimilars can serve as calibration standards or reference controls to measure drug concentration in serum samples. Here's how they are utilized:

  1. Calibration Standards: Biosimilar antibodies, such as Basiliximab biosimilars, are used to create a calibration curve. This curve is essential for quantifying the concentration of the drug in serum samples. The standards are typically prepared in a matrix similar to the sample matrix, such as human serum.

  2. Reference Controls: These biosimilars are also used as reference controls to ensure the accuracy and reliability of the assay. They help in validating the ELISA method by providing known concentrations of the drug, which are used to compare with unknown sample concentrations.

  3. Assay Development: Biosimilars facilitate the development of ELISA assays by providing a cost-effective option for researchers. They enable the creation of bridging assays necessary for demonstrating the bioequivalence of a biosimilar product to its reference product.

  4. Pharmacokinetic Analysis: In the context of PK studies, these biosimilars aid in assessing how a drug is absorbed, distributed, metabolized, and eliminated by the body. This information is critical for establishing appropriate dosing regimens and ensuring therapeutic efficacy.

Implementation in PK Bridging ELISA

Step-by-Step Process:

  • Preparation of Standards: Purified biosimilar Basiliximab is prepared in different concentrations to serve as standards. These standards are usually in a range that covers the expected concentrations in serum samples, such as 0 to 2000 ng/ml.

  • Calibration Curve: The standards are used to create a calibration curve by plotting the concentration of the standards against the measured absorbance in the ELISA assay.

  • Sample Assay: Serum samples containing Basiliximab are assayed using the ELISA method. The absorbance of each sample is measured and compared to the calibration curve to determine the concentration of Basiliximab in the sample.

  • Validation: The assay is validated by ensuring that it accurately measures known concentrations of the drug using the biosimilar standards as controls.

By using Basiliximab biosimilars as calibration standards and reference controls, researchers can effectively measure drug concentrations in serum samples during PK bridging ELISA assays.

The primary in vivo models for administration of research-grade anti-CD25 antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, more recently, humanized mouse models.

Key models and their features:

  • Syngeneic Mouse Models
    These are the most extensively used and well-characterized platform for testing anti-CD25 antibodies in vivo. They involve implantation of murine tumors (e.g., MC38, A20, CT26) into immunocompetent mice of the same genetic background, allowing for assessment of immune modulation, including Treg depletion and subsequent effects on TILs and tumor growth.

    • Anti-CD25 antibodies (typically rat anti-mouse CD25 such as PC61) are administered to deplete regulatory T cells (Tregs), after which changes in TIL phenotypes are quantified by methods such as flow cytometry and immunohistochemistry.
    • These models preserve the complexity of the intact mouse immune system, making them optimal for studying immune-tumor interactions and checkpoint modulation.
    • Example: In a syngeneic A20 lymphoma model, anti-mouse CD25 antibodies were evaluated for their impact on tumor growth and TIL composition.
  • Humanized Mouse Models
    Used when the goal is to evaluate human- or humanized anti-CD25 antibodies and their effects on human TILs, these models involve engrafting immunodeficient mice (such as NSG) with human immune cells and, frequently, patient-derived or engineered human tumors.

    • These models allow assessment of antibody specificity, cytotoxicity, and TIL phenotype in a human context.
    • Example: In a recent study, a CD25 x TIGIT bispecific antibody was tested in "CD25 humanized mice," which express human versions of these receptors, to evaluate Treg depletion and anti-tumor immunity.

Less commonly used or supplementary models:

  • Implantable anti-CD25 scaffolds
    One study used anti-mouse CD25 immobilized on mesh scaffolds (rather than systemic antibody administration) in tumor-bearing mice to locally capture Tregs and suppress tumor growth. While interesting, this is a variant of the syngeneic model and not standard practice for systemic antibody studies.

Summary Table

Model TypeHost SystemAnti-CD25 FormatPurpose/Advantages
Syngeneic mouseImmunocompetent mouseAnti-mouse CD25 abNative immune system, established tumor models, TIL study
Humanized mouseImmunodeficient mouse + human cellsHuman/humanized anti-CD25 abAllows evaluation of human-specific agents, human TILs

In both model types, TILs are isolated from tumors post-treatment and characterized with flow cytometry or histology for cell populations (CD8+, Tregs, etc.) and functional phenotypes.

Conclusion:
Syngeneic models are the standard for preclinical anti-CD25 studies involving tumor growth inhibition and TIL characterization, especially for murine anti-CD25 antibodies, whereas humanized models are increasingly used for evaluation of human or bispecific anti-CD25 reagents on human immune-tumor interactions.

Synergistic Effects of Basiliximab Biosimilar with Other Checkpoint Inhibitors in Immune-Oncology Research

Basiliximab Biosimilar: Mechanism and Role

Basiliximab biosimilars are monoclonal antibodies that selectively block the CD25 subunit of the interleukin-2 receptor (IL-2Rα), thereby inhibiting T-cell activation without broadly depleting T cells. This targeted approach is distinct from traditional immunosuppressants and is especially relevant in transplantation and, increasingly, in immuno-oncology research, where precise control of T-cell function can be advantageous. Biosimilars are particularly valuable in preclinical and translational research due to their cost-effectiveness and reproducibility.

Conceptual Basis for Combination Therapy in Immune-Oncology

In cancer immunotherapy, synergy is often sought by targeting multiple immune pathways simultaneously. For example, combining immune checkpoint inhibitors targeting CTLA-4 and PD-1/PD-L1 has shown enhanced anti-tumor efficacy in both preclinical and clinical settings, as these agents act at different stages of the immune response—CTLA-4 inhibitors enhance T-cell priming in lymph nodes, while PD-1/PD-L1 inhibitors block T-cell exhaustion at the tumor site. Similarly, combinations of LAG-3 or TIM-3 inhibitors with PD-1/PD-L1 inhibitors are being explored, with clinical trials demonstrating improved outcomes in certain cancers. The rationale is that each agent addresses a unique immune evasion mechanism, potentially overcoming the limitations of monotherapy.

Research Use of Basiliximab Biosimilar with Other Checkpoint Inhibitors

Current Landscape

While the primary clinical use of basiliximab is in transplantation, its biosimilars are increasingly employed in preclinical research to explore novel immunomodulatory strategies. The selective inhibition of IL-2 signaling by basiliximab biosimilars allows researchers to temporally modulate T-cell activation without full depletion, which may reduce toxicity risks compared to broad immunosuppressants. This property makes it an attractive candidate for combinatorial studies in complex immune-oncology models.

Potential Synergies with Other Checkpoint Inhibitors

  • With Anti-CTLA-4 Biosimilars: CTLA-4 inhibitors enhance T-cell activation in lymph nodes, while basiliximab biosimilars specifically block IL-2Rα on activated T cells. In theory, combining these could allow for fine-tuned control over T-cell proliferation and activation, potentially increasing the anti-tumor response while mitigating excessive immune toxicity. However, such combinations have not been widely reported in the oncology literature, as basiliximab is not a classical checkpoint inhibitor but rather a modulator of T-cell proliferation.
  • With Anti-LAG-3 Biosimilars: LAG-3 is a negative regulator of T-cell function, and its inhibition can further amplify T-cell responses, especially when combined with PD-1/PD-L1 blockade. Adding basiliximab biosimilar could theoretically allow researchers to investigate whether transiently limiting IL-2-driven T-cell expansion (via basiliximab) might reduce the risk of immune-related adverse events without completely abrogating the anti-tumor effect of LAG-3 inhibition.

Experimental Considerations

  • Model Systems: Researchers are likely to employ in vivo tumor models or ex vivo human immune cell cultures to assess the impact of combining basiliximab biosimilar with other checkpoint inhibitors. These models can help determine whether transient T-cell inhibition with basiliximab can enhance the therapeutic index of checkpoint blockade by reducing toxicity without compromising efficacy.
  • Outcome Measures: Key readouts include tumor growth kinetics, immune infiltration profiles, cytokine release, and the incidence of immune-related adverse events. The goal is to identify dosing regimens and timing that maximize synergy while minimizing side effects.
  • Mechanistic Insights: Such studies can provide insights into the temporal dynamics of T-cell activation and exhaustion, helping to refine combination immunotherapies for cancer.

Challenges and Future Directions

  • Lack of Clinical Data: The vast majority of evidence for basiliximab’s use in oncology is preclinical. Its clinical safety and efficacy in combination with checkpoint inhibitors remain to be established.
  • Complex Immune Interactions: The immune system’s redundancy and compensatory mechanisms mean that blocking one pathway (e.g., IL-2Rα) may unexpectedly affect others, necessitating careful experimental design and validation.
  • Biosimilar Variability: While biosimilars are rigorously standardized, minor differences could influence experimental outcomes, requiring rigorous quality control.

Summary Table: Potential Combinations and Research Questions

CombinationHypothesized SynergyKey Research Questions
Basiliximab + Anti-CTLA-4Enhanced T-cell priming with controlled expansionDoes IL-2Rα blockade reduce CTLA-4 inhibitor toxicity?
Basiliximab + Anti-LAG-3Amplified T-cell function with reduced exhaustionCan transient IL-2 inhibition improve safety of LAG-3 blockade?
Basiliximab + Anti-PD-1/PD-L1Combined blockade of T-cell inhibition and proliferationWhat is the optimal timing for IL-2Rα inhibition?

Conclusion

Researchers use basiliximab biosimilars alongside other checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) to explore whether selective, transient inhibition of T-cell proliferation can enhance the safety and efficacy of immune checkpoint blockade in complex oncology models. This approach is largely preclinical at present, with the goal of uncovering novel combinatorial strategies that maximize anti-tumor immunity while minimizing toxicity. The growing availability of biosimilars supports such innovative research, though clinical translation will require rigorous validation.

A Basiliximab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against basiliximab by detecting antibodies generated against this therapeutic monoclonal antibody.

Context and Mechanism:

  • In a bridging ADA ELISA, the assay leverages the bivalent nature of antibodies: anti-drug antibodies from patient serum are "bridged" between a capture and a detection arm of the drug (or its biosimilar).
  • For basiliximab, the biosimilar version—having the same antigenic structure as the reference biologic—can substitute for the original antibody as both the solid-phase (plate-bound) capture and the labeled detection reagent.

How it works:

  • Plate Coating: The ELISA plate is coated with basiliximab biosimilar (“capture”).
  • Sample Addition: Patient serum is added; any anti-basiliximab antibodies (ADAs) present will bind to the immobilized biosimilar.
  • Detection: After washing, a labeled basiliximab biosimilar (“detection reagent,” often biotinylated or enzyme-conjugated) is added. If ADA is present, it forms a “bridge” between the capture and detection biosimilars, creating a detectable complex.
  • Readout: Detection is performed using a substrate (e.g., TMB for HRP-labeled reagents), providing a readout proportional to ADA concentration.

Why use a biosimilar instead of the reference product?

  • Biosimilars are structurally and functionally equivalent to the reference product, ensuring assay specificity for antibodies directed against basiliximab (detecting immune responses targeting the administered drug).
  • They are often more accessible and cost-effective for research and assay development, making them preferable for large-scale immunogenicity studies.

Summary Table: Basiliximab Biosimilar in Bridging ADA ELISA

StepRole of Basiliximab BiosimilarPurpose
CaptureCoats plate (solid phase)Captures anti-basiliximab antibodies
DetectionLabeled biosimilar (e.g., biotinylated)Detects “bridged” patient ADAs
ReadoutEnzyme or tag on detection reagentMeasures immune response (ADA level)

Key Points:

  • This approach specifically detects circulating anti-basiliximab antibodies, not those generated against other components.
  • Using a biosimilar ensures the assay targets the correct epitope/structure present in the therapeutic formulation.

Limitations and Considerations:

  • The biosimilar must be validated to have matching binding characteristics to the original basiliximab to ensure the clinical relevance and accuracy of immunogenicity measurements.
  • Acid dissociation may be used prior to the assay to dissociate immune complexes and improve drug-tolerant ADA detection, especially where circulating drug may interfere.

This method is widely used for post-marketing immunogenicity monitoring of both innovator and biosimilar biologics.

References & Citations

B
CyTOF®
Flow Cytometry
IF
IHC
General Western Blot Protocol

Certificate of Analysis

Formats Available

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