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.
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.
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 commonly used as calibration standards and reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately quantify Benralizumab concentrations in serum samples. This ensures consistent and comparable measurement of both biosimilar and reference biologic across analytical runs.
Use as Calibration Standards:
Research-grade Benralizumab biosimilars can serve as the analytical standard for generating the calibration curve in a PK ELISA bridging assay.
Calibration curves are created using serial dilutions of the biosimilar standard in human serum at known concentrations, e.g., 50–12,800 ng/mL.
The signal generated from patient samples is compared against this standard curve to determine drug concentration in unknowns.
Use as Reference Controls:
During assay development and validation, both the biosimilar (test) and the reference (originator) products are used to assess bioanalytical comparability.
Quality control (QC) samples are prepared using both biosimilar and reference Benralizumab at different concentration levels to validate accuracy and precision of the method across product sources.
If equivalence is demonstrated (typically by ensuring response ratios between 0.8 and 1.25 for both products), a single standard—often the biosimilar—can be used for routine quantification.
Bridging ELISA Principle:
In a sandwich ELISA, pre-coated anti-Benralizumab antibodies bind drug from the sample (which may be the reference or biosimilar form).
A horseradish peroxidase (HRP)-conjugated anti-Benralizumab antibody detects the bound drug.
The colorimetric signal is proportional to the amount of Benralizumab present, which is interpolated from the biosimilar-based calibration curve.
PK Bridging Strategy:
Assaying both the biosimilar and reference using the same assay and standard minimizes variability and supports robust, regulatory-compliant PK bioequivalence assessment.
The approach includes comprehensive analytical validation: parallel curve qualification, cross-QC sample measurement, and statistical assessment of comparability before adopting a biosimilar as the universal standard in the assay.
In summary, research-grade Benralizumab biosimilars are essential in PK ELISAs as well-characterized calibration standards and reference controls, enabling accurate, reproducible quantitation and cross-comparisons between biosimilar and originator products in serum PK studies.
The primary in vivo models for testing anti-IL-5Rα (CD125) antibodies in the context of tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are humanized mouse models and syngeneic mouse tumor models, though direct evidence for anti-IL-5Rα specifically is limited.
1. Syngeneic Mouse Models
Definition: These involve implantation of mouse tumor cell lines into immunocompetent mice of the same genetic background, enabling a functional murine immune system.
Use for Immunotherapy: Syngeneic models such as CT26 (colon), MC38 (colon), B16 (melanoma), RENCA (renal), EMT-6 (breast), and Hepa1-6 (liver) are frequently applied to study responses to many immunotherapies by depleting or modulating TIL populations with depleting antibodies against immune markers or relevant targets.
Anti-IL-5Rα Relevance: In the literature, while syngeneic models are widely used for TIL analysis and antibody-based functional depletion of immune populations (e.g., CD4+, CD8+, NK cells), there is no direct evidence of published in vivo administration of research-grade anti-IL-5Rα (CD125) antibodies in standard mouse syngeneic tumor models due to the species specificity of most such antibodies. IL-5Rα is present on murine eosinophils and some lymphocytes, but most anti-human IL-5Rα antibodies (such as those modeled after benralizumab) do not cross-react with mouse IL-5Rα.
2. Humanized Mouse Models
Definition: Mice genetically engineered to express human immune system components, either through genetic humanization of the Il5ra locus or through engraftment with human hematopoietic cells expressing human IL-5Rα.
Use for Therapeutic Testing: These models support the in vivo study of human-specific antibodies against human IL-5Rα, such as benralizumab analogs, by allowing for direct engagement with human IL-5Rα+ immune cells and subsequent monitoring of tumor growth and detailed TIL characterization.
Anti-IL-5Rα in Humanized Mice: The development and characterization of potent humanized anti–IL-5Rα antibodies (e.g., 5R65.7) has been described, with in vitro validation for specificity, binding, and functional activity; preclinical testing for such antibodies for tumor immunity would require humanized mouse models expressing human IL-5Rα. These models enable both tumor xenografting and immunoprofiling of human immune cells post-antibody treatment.
3. Key Considerations
Availability of Cross-Reactive Antibodies: Most anti-IL-5Rα (CD125) antibodies are human-specific and do not recognize the murine ortholog, limiting their utility in wild-type or syngeneic mouse studies unless the mouse is genetically engineered to express human IL-5Rα.
Assessment of TILs: Both syngeneic and humanized models allow comprehensive TIL profiling by flow cytometry, RNA-seq, and mass cytometry. However, the mechanism of anti-IL-5Rα effects on the TME and TILs (especially targeting eosinophils or other IL-5Rα+ populations) can only be directly modeled in humanized systems if using human-mAb.
Summary Table: Anti-IL-5Rα Models for Tumor Immunology
Model Type
Use with Anti-IL-5Rα
IL-5Rα Compatibility
Typical Study Goals
TIL Characterization Possible
Syngeneic Mouse
Limited*
Mouse IL-5Rα (antibody-specific)
Immune cell depletion/immune mechanism study
Yes
Humanized Mouse
Yes
Human IL-5Rα
Efficacy, mechanism, immune dynamics
Yes
*Only possible if mouse-specific anti-IL-5Rα antibody is available.
Conclusion: Humanized mouse models are the primary choice for preclinical testing of research-grade anti-IL-5Rα (CD125) antibodies with reliable evaluation of tumor growth inhibition and TILs, due to antibody specificity requirements for human IL-5Rα. Syngeneic models are broadly used for TIL studies with other immunotherapies and, in rare cases, may be adapted for study with appropriate cross-reactive or mouse-specific reagents.
Researchers investigate the synergistic effects of immune-oncology drug combinations—including Benralizumab biosimilars (targeting IL-5Rα, thereby depleting eosinophils) and checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars)—by co-administering them in complex preclinical immune-oncology models. The core aim is to evaluate how these combinations modulate different arms of the immune system to enhance antitumor efficacy.
Key Approaches and Rationale:
Mechanistic Foundation: Checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, anti-PD-1/PD-L1) enhance T-cell-mediated antitumor immunity by releasing inhibitory brakes on immune responses, but tumors often evade these mechanisms using suppressive immune cells like eosinophils. Benralizumab, by depleting eosinophils, may reduce immunosuppression in the tumor microenvironment, potentially enhancing the efficacy of checkpoint blockade therapies.
Experimental Design:
Preclinical Models: Scientists typically utilize immune-competent mouse models engrafted with syngeneic tumors. These models enable the assessment of combination immunotherapies on tumor growth, immune cell infiltration, and cytokine profiles.
Treatment Regimens: Mice are treated with biosimilar Benralizumab (to deplete eosinophils) and checkpoint inhibitor biosimilars (e.g., anti-CTLA-4 or anti-LAG-3), either alone or in combination, to compare therapeutic outcomes.
Immune Profiling: Detailed flow cytometry and immunohistochemistry analyses are conducted to profile changes in immune cell subsets—especially T cell activation (CD8+ cytotoxic and CD4+ helper T cells), the reduction of regulatory T cells (Tregs), and other suppressive populations.
Outcome Measures: Endpoints include tumor volume reduction, survival, immune infiltration, and mechanistic readouts of T cell activation and exhaustion.
Synergy Assessment:
Studies often focus on whether combining eosinophil-depletion (via Benralizumab biosimilars) with checkpoint inhibition results in more pronounced tumor regression than either approach alone.
The combinations may target different immune escape mechanisms: for example, reducing eosinophil-mediated protection of tumor cells and simultaneously unleashing exhausted T-cell populations.
Data also reveal that the exact mechanisms depend on the specific checkpoint inhibitor:
Anti-PD-1/LAG-3 combinations preferentially require and activate CD4+ T cells, modulate Tregs, and subsequently boost CD8+ responses.
Anti-PD-1/CTLA-4 combinations more directly stimulate CD8+ cytotoxic T cells and may modulate immune cell infiltration differently.
Combining agents like Benralizumab biosimilars with different checkpoint inhibitors provides a way to dissect cooperative or redundant mechanisms across immune cell subsets.
Clinical and Translational Considerations:
While published studies detail combinations of checkpoint inhibitors (e.g., anti-CTLA-4 with anti-PD-1/PD-L1 and anti-LAG-3) and their mechanistic interplay, the literature directly describing Benralizumab (or IL-5Rα-directed agents) in these models is limited.
The rationale for using biosimilars rather than original biologics is primarily for cost and access in translational and preclinical research, ensuring equivalence without introducing confounding variables associated with batch or product variation.
Translational studies may investigate peripheral blood and tumor microenvironment changes as companion biomarkers, guiding the design of future clinical trials.
Summary Table: Experimental Strategy
Agent
Target/Mechanism
Role in Immune Modulation
Potential Synergy with Checkpoint Inhibitors
Benralizumab biosimilar
IL-5Rα (Eosinophil depletion)
Reduces suppressive eosinophil activity
Lowers immune suppression, improves T-cell effect
Anti-CTLA-4 biosimilar
T-cell checkpoint
Increases T-cell priming, CD8 activation
Direct cytotoxic response, more robust tumor kill
Anti-LAG-3 biosimilar
T-cell checkpoint
Modulates CD4/Treg balance, CD8 activation
Increases helper T cell support for cytotoxic response
Currently, most mechanistic detail comes from studies with canonical biologics (rather than biosimilars) or checkpoint inhibitor combinations. The extension to using biosimilars in bench or translational models follows standard immunological paradigms, using biosimilar antibodies to ensure broader access and reproducibility without altering the fundamental study design.
No direct study combining Benralizumab biosimilars with anti-CTLA-4 or anti-LAG-3 biosimilars appears in the provided sources, but the experimental approach is clear from established immune-oncology methodologies and documented checkpoint inhibitor combination studies.
A Benralizumab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA by mimicking the structure of the original therapeutic (Benralizumab), allowing detection of ADAs in patient samples that specifically recognize the drug.
Core methodology in a bridging ELISA for ADA detection:
Benralizumab biosimilar is coated onto the ELISA plate as the capture reagent. Patient serum is added; if it contains ADA, these antibodies will bind to the immobilized biosimilar.
For detection, a labeled (e.g., biotin- or HRP-conjugated) Benralizumab biosimilar is added. This will bind the other arm of the ADA, “bridging” between capture and detection molecules.
Mechanistic details:
ADAs are bivalent (they have two antigen-binding sites). One binding site recognizes the plate-bound Benralizumab biosimilar, the other binds the labeled biosimilar. Only true bridging (by bona fide ADAs) results in a signal.
The readout (commonly colorimetric via HRP and TMB substrate) is proportional to the amount of ADA present in the sample.
Using a biosimilar (as opposed to innovator Benralizumab) is acceptable as long as the biosimilar has biochemically and immunologically equivalent epitopes, which is the case for research-grade biosimilars designed for assay development.
Key points for assay design:
Specificity: Assay detects only antibodies against Benralizumab, not other proteins.