Anti-Human IL-5Rα (CD125) (Benralizumab)

Anti-Human IL-5Rα (CD125) (Benralizumab)

Product No.: I-2200

- -
- -
Product No.I-2200
Clone
MEDI-563
Target
IL-5Rα (CD125)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-5R subunit alpha, CDw125, CD125, IL5R, HSIL5R3, IL5RA
Isotype
Human IgG1κ
Applications
B
,
ELISA
,
FA
,
IHC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
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 ?
B,
ELISA,
IHC,
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 Benralizumab. MEDI-563 (Benralizumab) is an antagonist of human and cynomolgus monkey interleukin-5 receptor alpha chain (IL-5Rα). MEDI-563 does not bind to murine IL-5Rα.
Background
Eosinophilic inflammation underlies certain asthma and chronic obstructive pulmonary disease(COPD) phenotypes1. IL-5 is the primary cytokine mediating eosinophil mobilization,maturation, activation and survival2. Neutralization of IL-5 in murine and nonhuman primatemodels of asthma results in reduced eosinophil counts and improved lung pathology. Therefore,strategies that deplete lung eosinophils and basophils have been sought to improve asthmacontrol. IL-5Rα was selected as a suitable target because its expression is restricted toeosinophils, basophils and their progenitors in bone marrow.

Benralizumab is a humanized, monoclonal antibody that binds to an epitope indomain 1 of IL-5Rα that overlaps with a portion of the IL-5 binding site1,2. Residue I61 is criticalfor benralizumab binding2. Binding inhibits IL-5 receptor signaling independent of the ligandand leads to depletion of eosinophils as well as inhibition of IL-5-mediated cell proliferation1.Additionally, in vitro, benralizumab potently induces apoptosis of eosinophils and basophils viaantibody-dependent cell-mediated cytotoxicity in the presence of NK effector cells2. Incynomolgus monkeys, benralizumab depletes blood and lung eosinophils as well as eosinophilprecursors present in bone marrow.

Benralizumab has been approved for the treatment of eosinophilic asthma and chronicobstructive pulmonary disease1. This product is for research use only.

Antigen Distribution
IL-5Rα is expressed exclusively on mature eosinophils and basophils as well as eosinophil and basophil progenitors in bone marrow.
Ligand/Receptor
IL-5, CSF2RB
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Inflammatory Disease
.
Cell Proliferation & Viability
.
Pro-Inflammatory Cytokines
.
Pulmonary Disease

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 Benralizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA by serving as the analytical standard against which drug concentrations in serum samples are quantified.

In a PK bridging ELISA developed for biosimilar drugs, including Benralizumab, the best practice is to establish a single assay using a single analytical standard—typically the biosimilar itself—for quantification of both biosimilar and reference product concentrations in human serum. This standard is used to prepare a calibration curve spanning the relevant concentration range in serum samples (e.g., 50–12,800 ng/mL for Benralizumab, as described in validated assays). Serum samples containing unknown drug concentrations are then run in parallel with these calibration standards; the assay output (colorimetric or fluorescence readings, depending on detection chemistry) is mapped onto the calibration curve to interpolate the Benralizumab concentration in each sample.

Reference controls, prepared with known concentrations of Benralizumab—either the biosimilar or the reference product—are included as quality control (QC) samples. These are essential for monitoring assay performance and ensuring accuracy and precision throughout the analytical run. During method validation, both biosimilar and reference products are prepared as QC samples and measured against the biosimilar calibration curve to confirm analytical equivalence, typically by evaluating whether the 90% confidence interval for paired measurements falls within pre-defined boundaries (such as 0.8–1.25). If equivalence is established, the biosimilar becomes the accepted calibrator for future PK studies.

Key points:

  • Biosimilar Benralizumab is typically used as the calibrator for the ELISA standard curve.
  • Reference controls (biosimilar and/or originator product at known concentrations) are used as QC samples to validate the accuracy and specificity of the method.
  • A validated ELISA method allows drug concentration in serum samples to be interpolated from the calibration curve, supporting PK analysis and bioequivalence assessments.
  • Using a single calibration standard across both biosimilar and reference products minimizes variability and supports direct comparison in bridging studies.

This approach is consistent with regulatory guidance and current industry consensus for bioanalytical methods supporting biosimilar development, ensuring robust, reproducible quantification for PK studies.

The primary in vivo models for administering a research-grade anti-IL-5Rα (CD125) antibody to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, for antibodies specific to human IL-5Rα, humanized xenograft models.

Key Model Categories:

  • Syngeneic mouse models:
    These models use immunocompetent mice implanted with mouse tumor cell lines, such as MC38, Hepa1-6, CT26, RENCA, B16F10, and EMT-6, allowing measurement of tumor growth and immune cell infiltration after antibody administration.

    • Standard immunotherapy studies in these models typically use depleting antibodies against mouse leukocyte markers (e.g., anti-CD8, anti-NK1.1, anti-CD4), but research-grade anti-IL-5Rα antibodies could be deployed if available for the murine (mouse) IL-5Rα.
    • These models facilitate detailed profiling of TILs (e.g., T cells, myeloid-derived suppressor cells, macrophages) by flow cytometry and immunogenomic techniques, allowing mechanistic interrogation of immune responses after therapy.
  • Humanized xenograft models:
    When studying a human-specific anti-IL-5Rα antibody (such as clone REA705 or engineered antibodies like 5R65.7), human tumor cell lines are implanted in immunodeficient mice engrafted with a human immune system (“humanized mice”).

    • Humanized NSG or NOG mice are frequently used to reconstitute human hematopoiesis, supporting the study of human antibody interactions with human tumor and immune cells.
    • These models are necessary when the antibody does not cross-react with mouse IL-5Rα, as is the case for many engineered human or humanized antibodies.
    • They facilitate analysis of human TIL subsets after antibody treatment.

Applications and Limitations:

  • Syngeneic models provide robust platforms for tumor growth studies and immune landscape profiling, but they require mouse-reactive antibodies.
  • Humanized models are essential for evaluating human-specific antibody therapies and for detailed study of human TIL phenotypes and mechanisms, although they are more complex and costly.

Model Selection Considerations:| Model Type | Species | Antibody Specificity | TIL Characterization | Commonly Used Tumors ||-------------------------|-------------|----------------------------|----------------------|------------------------------|| Syngeneic | Mouse | Anti-mouse or cross-reactive| Murine | MC38, CT26, RENCA, EMT-6, B16F10, Hepa1-6 || Humanized xenograft | Human (in mouse) | Anti-human | Human | Human cell lines in NSG/NOG mice |

Summary of Model Usage:

  • Syngeneic models are the mainstay for general immunotherapy mechanism studies and allow TIL characterization by standard immunophenotyping techniques.
  • Humanized xenograft models are required for in vivo evaluation of human-specific anti-IL-5Rα antibodies, including those like benralizumab analogues or REA705, with downstream analysis of human TIL populations.

To summarize:

  • Syngeneic models (with mouse-reactive antibody): MC38, CT26, RENCA, EMT-6, B16F10, Hepa1-6.
  • Humanized xenograft models (with human-reactive antibody): human tumor cell lines in humanized NSG/NOG mice.
    Selection depends on antibody species specificity and the desired TIL immune phenotyping.

Researchers explore the use of Benralizumab biosimilars alongside other checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—in complex immune-oncology models to assess potential synergistic antitumor effects. This is primarily a research area aimed at understanding how targeting different aspects of the immune system can enhance anti-tumor responses.

Essential context and supporting details:

  • Benralizumab is a monoclonal antibody targeting the IL-5 receptor α on eosinophils and basophils, leading to their depletion via enhanced antibody-dependent cell-mediated cytotoxicity (ADCC). While clinically approved mainly for severe eosinophilic asthma, its immunomodulatory effects are of interest in oncology models where eosinophils can shape the tumor microenvironment.

  • Checkpoint inhibitors such as antibodies against CTLA-4 and LAG-3 modulate T-cell activity—primarily boosting the activation and proliferation of cytotoxic and helper T cells, which are crucial for anti-tumor immunity.

  • Combination rationale: Targeting multiple immune pathways is hypothesized to overcome resistance mechanisms and achieve a more robust anti-tumor effect. For example:

    • Anti-CTLA-4 and anti-LAG-3 biosimilars act at different stages or aspects of T-cell activation and regulation, leading to distinct immune signatures and complementary effects when combined.
    • Benralizumab can further modify the immune microenvironment by depleting eosinophils and basophils, potentially reducing immunosuppression, which might amplify the benefits of checkpoint blockade.

Synergy exploration in research models:

  • Mouse and preclinical tumor models are commonly used to test these combinations. For checkpoint inhibitors, studies have mapped how anti-CTLA-4 increases cytotoxic CD8 T cells, while anti-LAG-3's effect relies more on helper CD4 T cells and suppressing regulatory T cells (Tregs).

  • When a Benralizumab biosimilar is added, researchers measure:

    • Changes in immune cell populations within the tumor (e.g., enhanced T cell infiltration, altered cytokine profiles).
    • Tumor growth inhibition and survival benefits beyond what is observed with single agents.
    • Effects on eosinophil-mediated tumor-promoting inflammation or immunosuppression, which might alter how well checkpoint inhibitors work.

Limitations and considerations:

  • Most published data focus on Benralizumab's immunological mechanism and checkpoint inhibitor synergy in preclinical and early clinical studies; robust clinical evidence for this specific triple approach is still emerging.
  • The specific design and readouts of such studies depend on tumor type, immune context, and animal model used.

In summary, researchers use Benralizumab biosimilars with CTLA-4 or LAG-3 checkpoint inhibitors in complex models to study whether depleting eosinophils alters the tumor immune landscape in a way that enhances the efficacy of T-cell-targeted immunotherapies. Model-specific endpoints include tumor size, immune cell profiling, and functional immune assays. Direct evidence in peer-reviewed studies using this exact combination is currently limited, but the conceptual framework aligns with the broader field of combination immunotherapy.

A Benralizumab biosimilar can be used as either the capture reagent (plate-bound) or the detection reagent (labeled form) in a bridging ADA ELISA to detect anti-drug antibodies (ADAs) in patient samples, thus monitoring the immune response against Benralizumab therapy.

In a bridging ADA ELISA:

  • The assay typically relies on the bivalent nature of ADAs, which can simultaneously bind to two identical antigen molecules (in this case, Benralizumab or its biosimilar).
  • One form of Benralizumab biosimilar is immobilized on the assay plate to capture ADAs present in patient serum.
  • After incubation, a detection reagent—often the same Benralizumab biosimilar, but labeled with an enzyme (e.g., horseradish peroxidase, HRP) or a tag (e.g., biotin)—is added to bind to the opposite arm of the ADA, forming a "bridge".

Use of Biosimilar Reagent:

  • A Benralizumab biosimilar is structurally similar to the originator drug and can be used as the capture and/or detection reagent to measure patient ADAs that recognize Benralizumab or its biosimilar.
  • Using the biosimilar ensures that the assay detects ADAs against epitopes shared by both the biosimilar and the reference product, allowing comparative immunogenicity assessment.
  • The choice to use the biosimilar as the assay reagent is particularly common in biosimilar development and post-marketing surveillance to compare ADA profiles between biosimilar and reference products.

Assay Workflow Example:

  1. Plate Coating: Benralizumab biosimilar is coated on the ELISA plate to capture any ADA present in the patient serum.
  2. Sample Addition: Patient serum is added, allowing any ADAs (if present) to bind to the plate-bound drug.
  3. Detection: A labeled Benralizumab biosimilar is added to bind to the other idiotype of the ADA, forming a sandwich.
  4. Signal Development: The detection reagent’s label (e.g., HRP) enables signal generation upon substrate addition, measuring ADA levels.

Why use biosimilar as reagent?

  • Ensures equivalence of ADA detection against both biosimilar and originator.
  • Facilitates regulatory comparison and monitoring of immunogenicity in clinical practice.

Critical Points:

  • Optimizing assay conditions to ensure sensitivity and specificity for both biosimilar and reference product.
  • Assay must be validated for equivalence if comparative immunogenicity is the aim.

In summary, the Benralizumab biosimilar serves as an antigen to selectively capture and detect anti-Benralizumab ADAs using a bridging ELISA format, providing data on a patient’s immunogenic response to the therapeutic drug.

References & Citations

1 Markham A. Drugs. 78(4):505-511. 2018.
2 Kolbeck R, Kozhich A, Koike M, et al. J Allergy Clin Immunol. 125(6):1344-1353.e2. 2010.
3 Busse WW, Katial R, Gossage D, et al. J Allergy Clin Immunol. 125(6):1237-1244.e2. 2010.
4 Laviolette M, Gossage DL, Gauvreau G, et al. J Allergy Clin Immunol. 132(5):1086-1096.e5.2013.
5 FitzGerald JM, Bleecker ER, Nair P, et al. Lancet. 388(10056):2128-2141. 2016.
6 Park HS, Kim MK, Imai N, et al. Int Arch Allergy Immunol. 169(3):135-145. 2016.
7 Bleecker ER, FitzGerald JM, Chanez P, et al. Lancet. 388(10056):2115-2127. 2016.
8 Ferguson GT, FitzGerald JM, Bleecker ER, et al. Lancet Respir Med. 5(7):568-576. 2017.
B
Indirect Elisa Protocol
FA
IHC

Certificate of Analysis

Formats Available

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