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 Belimumab. Belimumab is a monoclonal antibody that
specifically targets and inhibits B-lymphocyte stimulator (BLyS), also known as B-cell
activating factor (BAFF).
Background
CD257, commonly referred to as the BAFF, plays a crucial role in regulating B-cell survival,
maturation, and differentiation by interacting with specific receptors such as TACI, BCMA,
and BAFFR1. Through these interactions, BAFF exerts anti-apoptotic effects that promote
the survival of B cells, making it a key factor in the pathogenesis of autoimmune diseases
and B-cell malignancies. Elevated levels of BAFF have been associated with conditions like
systemic lupus erythematosus (SLE) and rheumatoid arthritis, where overactive B-cells
contribute to disease progression. Targeting CD257/BAFF has emerged as a promising
therapeutic strategy for managing such disorders, as it can disrupt the signaling pathways
that maintain abnormal B-cell activity1,2.
Belimumab, a monoclonal antibody, specifically inhibits BAFF (also known as BLyS). By
neutralizing BAFF, Belimumab reduces the survival and activity of B- cells, thereby
dampening the autoimmune response. It is primarily used to treat SLE and lupus nephritis,
conditions characterized by excessive B-cell activation and inflammation3,4.
Antigen Distribution
BAFF is distributed throughout the body, with high expression in B cell
lineage cells 3 . It is found in various tissues, including the blood, lymphatic tissues, and 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 Belimumab biosimilars are typically used as analytical standards or reference controls in pharmacokinetic (PK) bridging ELISAs to measure drug concentration in serum samples, enabling direct quantification and cross-comparison between biosimilar and reference products.
In practice:
Single Method, Single Standard Approach: Industry best practice supports developing a single PK assay using a single analytical standard—often the biosimilar itself—for quantitative measurement of both the biosimilar and the reference Belimumab in test samples. This minimizes variability and eliminates the need for multiple calibration curves.
Calibration Standard Preparation: Researchers prepare sets of standards by spiking the research-grade biosimilar into human serum at defined concentrations, covering the expected PK range (e.g., 50–12,800 ng/mL). These standards establish the calibration curve in the sandwich ELISA.
Reference Controls/Quality Controls (QCs): Alongside the calibration standards, quality control samples are prepared using both the biosimilar and reference products at various concentrations (low, medium, high), to validate assay performance and confirm bioanalytical equivalence between products.
Assay Validation & Bioanalytical Equivalence: Extensive method validation assesses precision, accuracy, and comparability. Quantitation of “unknown” serum samples (from clinical PK studies) is referenced to the biosimilar’s calibration curve. Analytical equivalence is confirmed statistically, ensuring the assay measures the biosimilar and reference product indistinguishably.
ELISA Principle: The assay typically employs a sandwich ELISA, where anti-Belimumab antibodies are precoated onto microwells, capturing Belimumab in standards, controls, and test samples alike. After the binding and detection steps, the concentration is interpolated from the calibration curve generated using the biosimilar standard.
Use case in bridging studies:
This approach is essential in biosimilar development, as regulatory agencies require direct measurement and demonstration of comparable PK profiles between the biosimilar and reference product. Using a research-grade biosimilar as the standard provides a robust, reproducible basis for quantitation and method validation across multiple test products.
In summary, research-grade Belimumab biosimilars are spiked into human serum to generate calibration curves and QC samples, serving as standards in validated PK ELISAs for quantifying drug concentrations and demonstrating bioanalytical equivalence with reference Belimumab in clinical studies.
The primary in vivo models where research-grade anti-BAFF antibodies are administered to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models. These models are favored because they utilize immunocompetent mice, allowing for proper assessment of immune response—including TILs—after antibody administration.
Key Points:
Syngeneic Models:
Consist of mouse tumor cell lines implanted into genetically identical (same strain) immunocompetent mice.
Facilitate detailed evaluation of immunotherapies (e.g., anti-BAFF antibody) and their effects on tumor growth and immune cell infiltration.
Commonly used models include MC38 (colon carcinoma), B16F10 (melanoma), RENCA (renal carcinoma), and CT26 (colon carcinoma), which are well-characterized for immune compositions and responses.
Allow for robust analysis of TIL populations due to the presence of a fully functional murine immune system.
Humanized Models:
Less commonly used in the anti-BAFF context for TIL characterization, primarily due to their complexity and cost.
Humanized models involve engraftment of human immune cells (or hematopoietic stem cells) into immunodeficient mice, which are then implanted with human tumor cells.
These are valuable for testing human-specific antibodies (e.g., clinical-grade belimumab) when murine surrogates are not available or do not cross-react, but their use in anti-BAFF antibody oncology studies is not as widely documented as syngeneic models.
Relevance to Anti-BAFF Research:
Studies have shown that BAFF signaling plays a role in B cell survival and function, making it a target of interest in both autoimmunity and oncology domains.
In cancer immunotherapy research, syngeneic models are used to evaluate changes in B cell and TIL populations following BAFF blockade, and to study resultant effects on the tumor microenvironment.
Humanized mice have been leveraged predominantly when anti-BAFF antibodies under investigation are specific to human BAFF, or when translation of findings to human immune context is needed.
In summary:
Syngeneic mouse tumor models are the standard system for in vivo administration of research-grade anti-BAFF antibodies to study both tumor growth inhibition and TIL phenotype.
Humanized mouse models are used more selectively, especially for human-specific therapeutic antibodies, but are not as commonly referenced in the specific context of in vivo anti-BAFF and TIL research compared to syngeneic models.
If a particular tumor type or BAFF antibody specificity is of interest, researchers choose between these models based on species compatibility of the antibody and the immunological questions being addressed.
Researchers use Belimumab biosimilars in combination with other immune checkpoint inhibitors, such as anti-CTLA-4 or anti-LAG-3 biosimilars, to investigate potential synergistic effects on anti-tumor immunity and immune regulation in complex immune-oncology models.
Context and Supporting Details:
Belimumab Mechanism: Belimumab and its biosimilars target BAFF (B-cell activating factor), also known as BLyS, which is critical for B cell survival and function. By blocking BAFF, Belimumab inhibits the survival of immature, naïve, and autoreactive B cells, thereby modulating humoral immunity and autoantibody production.
Checkpoint Inhibitors: In immune-oncology, checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3, anti-PD-1) function by releasing the brakes on T cell activation. Anti-CTLA-4 mainly restores the induction and proliferation of activated T cells within lymph nodes, while anti-PD-1 and anti-LAG-3 act more peripherally in the tumor microenvironment, preventing functional exhaustion of cytotoxic T cells.
Research Applications of Combination Therapy:
By combining Belimumab biosimilars (which modulate B cell activity) with checkpoint inhibitors (which enhance T cell responses), researchers can dissect how B cell-mediated processes (including antigen presentation, antibody production, and immune regulation) interact with T cell-driven anti-tumor immunity.
Such combinations allow researchers to test whether dual targeting of both humoral (B cell–mediated) and cellular (T cell–mediated) immune checkpoints produces additive or synergistic anti-tumor effects, and whether this approach overcome limitations of single-agent therapies.
These studies are typically performed in preclinical tumor models or early-phase clinical trials, allowing systematic evaluation of immune cell infiltration, cytokine profiles, tumor regression, and immune-related adverse events.
Researchers employ biosimilars for Belimumab due to their cost-effectiveness, consistency, and accessibility for laboratory models, enabling the broad mechanistic investigation needed for translational research.
Additional Notes:
Most publicly documented preclinical combination strategies have focused on PD-1/PD-L1 and CTLA-4 blockade together, but extending this framework to include BAFF/BLyS blockade (Belimumab biosimilar) is supported by its documented role in regulating B cell and, by extension, T cell function.
Belimumab's effect on B cell modulation is particularly relevant in tumor models where B cells play a significant immunoregulatory or suppressive role, providing a rationale for evaluating such combinations.
Although clinical data for Belimumab combination with checkpoint inhibitors in oncology is limited, ongoing translational and mechanistic studies are exploring these synergies in autoimmune and cancer models.
In summary, researchers utilize Belimumab biosimilars alongside other checkpoint inhibitors in complex immune-oncology models to explore combined effects on both B cell and T cell immunity, aiming to enhance anti-tumor responses beyond what is achievable with single-agent therapies.
A Belimumab biosimilar is used as a capture or detection reagent in a bridging ADA ELISA to specifically detect anti-drug antibodies (ADAs) in patient samples by mimicking the therapeutic drug's structure, particularly its antigen-binding sites.
Context and Methodology:
Bridging ELISA Principle: In immunogenicity testing for monoclonal antibody drugs like Belimumab, a "bridging" ELISA is commonly used to monitor the patient's immune response against the drug (i.e., the development of ADAs).
Reagent Role: The Belimumab biosimilar, which has the same variable regions as the therapeutic antibody, is used interchangeably with the original drug for these assays due to its structural similarity.
Assay Workflow:
Step 1: Microtiter plates are coated with Belimumab biosimilar (drug or biosimilar acts as the capture reagent).
Step 2: Patient serum is added; if the patient has generated ADAs, these antibodies "bridge" between the coated biosimilar on the plate and an added, labeled (e.g., HRP- or biotin-conjugated) Belimumab biosimilar that acts as the detection reagent.
Step 3: After washing, the detection signal is developed (commonly using chromogenic substrate for the HRP enzyme), indicating the presence of bridging complexes—i.e., patient-derived ADAs bound to both capture and detection biosimilar molecules.
Why Use a Biosimilar?
Structural Equivalence: The biosimilar's identical variable region ensures all ADAs recognizing the original therapeutic will also bind the biosimilar, ensuring sensitivity and specificity.
Non-therapeutic Use: Biosimilars are often formulated for laboratory studies and not for therapeutic use, preventing analytics from impacting therapeutic supply or patient risk.
Summary Table: Belimumab Biosimilar in Bridging ADA ELISA
Step
Reagent
Role
Plate Coating
Belimumab biosimilar (unlabeled)
Capture patient ADAs
Serum Addition
Patient sample
Source of potential ADAs
Detection
Labeled Belimumab biosimilar (e.g., HRP or biotin)
Binds to another ADA epitope
Signal Development
Chromogenic substrate
Quantifies the ADA-bridged complex
Key Points:
Both capture and detection reagents are typically the biosimilar, labeled or unlabeled, taking advantage of the ADA's ability to "bridge" between two drug molecules.
This design ensures detection of bivalent antibodies (the most common format for ADAs) in patient sera.
The approach is widely established and accepted for the immunogenicity monitoring of monoclonal antibody therapeutics and their biosimilars.
Supporting Reference Examples:
Bridging ELISAs detect ADAs by capturing them with one drug molecule and detecting with a labeled version of the same or biosimilar drug molecule.
Belimumab biosimilars, with the same variable region as the original, are suited for assay use since they faithfully mimic the therapeutic for immune detection purposes.
This method provides a robust, drug-specific tool to assess and monitor immune responses against Belimumab or its biosimilar in clinical immunogenicity studies.
References & Citations
1. Xu H, He X, Zhao W, et al. Clin Lab. 2012;58(5-6):411-418.
2. Ye W, Zhou Y, Xu B, et al. Medicine (Baltimore). 2019;98(51):e18407.
3. Figgett WA, Deliyanti D, Fairfax KA, Quah PS, Wilkinson-Berka JL, Mackay F. J Autoimmun. 2015;61:9-16.
4. Auyeung-Kim DJ, Devalaraja MN, Migone TS, Cai W, Chellman GJ. Reprod Toxicol.2009;28(4):443-455.
5. Belimumab Recombinant Monoclonal Antibody (MA5-41860). Accessed September 28,
2024. https://www.thermofisher.com/antibody/product/Belimumab-Antibody-Recombinant-Monoclonal/MA5-41860