Anti-Human CD257 (BAFF) (Tabalumab) – Fc Muted™ Biotin

Anti-Human CD257 (BAFF) (Tabalumab) – Fc Muted™ Biotin

Product No.: LT1406

- -
- -
Product No.LT1406
Clone
Tabalumab
Target
BAFF
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Tabalumab, CD257, BAFF, TNFSF13b, BLYS
Isotype
Human IgG1κ

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Human
Rabbit
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Original antibody was raised against soluble human BAFF.
Product Concentration
0.5 mg/ml
Formulation
This Biotinylated antibody is formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.4, 1% BSA and 0.09% sodium azide as a preservative.
State of Matter
Liquid
Storage and Handling
This biotinylated antibody is stable when stored at 2-8°C. Do not freeze.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Reported Applications For Relevant Conjugates ?
N
IP
WB
IF
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 Tabalumab. Tabalumab neutralizes soluble human, cynomolgus monkey, and rabbit BAFF. Additionally, Tabalumab neutralizes membrane-bound BAFF. This product is for research use only.
Background
Tabalumab is a human monoclonal anti-B-cell activating factor (BAFF) antibody intended for the treatment of autoimmune diseases and B cell malignancies.1 BAFF is a membrane-bound, type II transmembrane protein that belongs to the tumor necrosis factor (TNF) ligand family and is the ligand for BR3, TACI, and BCMA. BAFF is an immunostimulant necessary for maintaining normal immunity. This cytokine has also been shown to play an important role in the proliferation and differentiation of B cells. An inadequate level of BAFF leads to immunodeficiency whilst an elevated level of BAFF causes unusually high antibody production that results in the development of autoimmune diseases such as systemic lupus erythematosus and rheumatoid arthritis. Additionally, BAFF has been found in renal transplant biopsies with acute rejection.2 Furthermore, BAFF may be a mediator of food-related inflammation, and is associated with multiple dietary ailments including celiac disease, insulin resistance, diabetes, and obesity.3 Interestingly, it is suspected that BAFF may be involved in non-IgE-mediated reactions because there is no known correlation between BAFF and IgE.4 More research is needed to unlock the enormous therapeutic potential for BAFF antagonists. This cost-effective, research-grade Anti-Human CD257 (BAFF) (Tabalumab) utilizes the same variable regions from the therapeutic antibody Tabalumab making it ideal for research projects.
Antigen Distribution
BAFF is expressed on various cell types including monocytes, dendritic cells and bone marrow stromal cells.
Ligand/Receptor
TACI, BCMA,APRIL ligand, BAFFR/BR3
PubMed
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Cell Biology
.
Costimulatory Molecules
.
Immuno-Oncology
.
Immunology
.
Signal Transduction
.
Stem Cell

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.

Use of Research-Grade Tabalumab Biosimilars as Calibration Standards in PK Bridging ELISA

Biosimilars are laboratory-made versions of existing biologic drugs designed to be highly similar, but not necessarily identical, to the reference product. In the context of pharmacokinetic (PK) studies—especially those supporting biosimilar development—calibration standards and reference controls play a crucial role in ensuring the accuracy, precision, and reliability of drug concentration measurements in biological matrices like serum.

Role of Biosimilars as Calibration Standards

  • Bioanalytical Comparability: For biosimilar development, it is considered best practice to use a single PK assay with a single analytical standard to quantify both the biosimilar and reference product in test samples. This minimizes variability and avoids the need for crossover analysis in blinded clinical studies.
  • Method Qualification: The process begins with a robust method qualification to generate precision and accuracy data for both the biosimilar and reference product. If bioanalytical comparability is demonstrated, the biosimilar can then serve as the analytical standard (calibrator) for the validated PK ELISA method.
  • Validation: During validation, multiple independent sets of standards—prepared in human serum at defined concentrations—are analyzed. These standards are used to construct a calibration curve against which test samples (serum samples from clinical studies) are quantified.
  • Regulatory Context: This approach aligns with regulatory guidance and industry best practices, where the goal is to show that the biosimilar and reference product can be measured equivalently, thus supporting PK similarity assessments required for regulatory approval.

Bridging ELISA for Drug Measurement

  • Bridging Assay Principle: The typical PK ELISA for monoclonal antibodies like tabalumab uses a "bridging format," where detection antibodies bind to different epitopes on the drug molecule. This format enhances specificity and is useful for detecting drug levels in complex matrices such as serum.
  • Reference Controls: Both calibrators (standard curve) and quality controls (QCs) are included in each assay run. Calibrators are precisely prepared drug solutions at known concentrations, while QCs are typically prepared at low, medium, and high concentrations to monitor assay performance.
  • Serum Matrix: Standards and QCs are prepared in the same biological matrix as the test samples (e.g., human serum) to account for matrix effects that could influence assay performance.
  • Limits of Quantification: For tabalumab, validated assays have defined lower and upper limits of quantification (LLOQ and ULOQ), ensuring that measurements are reliable within a specified concentration range.

Step-by-Step Application in PK Studies

  1. Assay Development: Select or develop a bridging ELISA that can detect both the biosimilar and reference product with equivalent sensitivity and specificity.
  2. Standard Preparation: Prepare calibration standards using the research-grade biosimilar at defined concentrations (e.g., 50, 100, 200, ... 12,800 ng/mL).
  3. Quality Controls: Prepare QCs at multiple concentrations within the assay range.
  4. Validation: Validate the assay by analyzing multiple replicates of standards and QCs to establish precision, accuracy, and robustness.
  5. Sample Analysis: Quantify drug concentrations in clinical serum samples by interpolating their signal against the biosimilar-derived calibration curve.
  6. Data Analysis: Compare the PK profiles of the biosimilar and reference product to assess bioequivalence, using the same calibration standard for both to ensure comparability.

Scientific and Regulatory Considerations

  • Minimizing Variability: Using a single set of biosimilar-derived standards reduces inter-assay variability and facilitates direct comparison between biosimilar and reference product PK data.
  • Stringent Criteria: Regulatory expectations require demonstration of bioanalytical equivalence between the biosimilar and reference product within the assay, often using statistical criteria (e.g., 90% confidence intervals) to confirm comparability.
  • Documentation: Comprehensive method validation and documentation are essential to support regulatory submissions and ensure confidence in the PK data generated.

Summary Table

StepPurposeUse of Biosimilar
Calibration StandardConstruct standard curve for quantificationBiosimilar as primary calibrator
Quality ControlMonitor assay precision and accuracyBiosimilar as QC material
Sample AnalysisMeasure drug in clinical samplesQuantify using biosimilar-based curve
Data ComparisonAssess PK similarityDirect comparison enabled by same std

Conclusion

Research-grade tabalumab biosimilars are used as calibration standards and reference controls in PK bridging ELISAs by serving as the primary material for constructing the assay's standard curve and quality controls. This ensures that the pharmacokinetic profiles of both the biosimilar and reference product can be compared directly and accurately within the same assay system, supporting robust bioequivalence assessments required for regulatory approval. This approach minimizes variability, enhances data reliability, and is aligned with current industry and regulatory best practices for biosimilar development.

Standard flow cytometry protocols using a conjugated Tabalumab biosimilar (e.g., PE or APC-labeled) for validating BAFF expression levels or binding capacity typically involve direct staining of cells expressing BAFF and analysis of fluorescence intensity to quantify binding or expression.

Essential protocol steps and context:

  • Sample Preparation:

    • Cells naturally expressing membrane-bound BAFF (e.g., activated monocytes) or cell lines engineered to express human BAFF (e.g., HEK293 transfectants) are prepared, often using enzyme-free dissociation to preserve surface proteins.
    • Suspend cells in a FACS buffer (e.g., D-PBS, 2% FBS, 0.1% sodium azide) to minimize non-specific staining.
  • Blocking Non-specific Binding:

    • Pre-incubate cells with an irrelevant IgG (e.g., goat IgG at 1 mg/mL) to block Fc receptor-mediated background staining.
    • If using an Fc-mutated biosimilar, such as N279A/N297A variants, non-specific Fc binding is reduced, improving signal clarity.
  • Antibody Staining:

    • Incubate cells with fluorochrome-labeled Tabalumab biosimilar (e.g., directly conjugated to PE or APC) or use a biotinylated Tabalumab followed by streptavidin-PE/APC.
    • Typical concentrations for binding assays may be optimized (e.g., 5–50 μg/mL Tabalumab, 18–300 ng/mL biotinylated BAFF), with incubation on ice for 15–20 minutes to limit internalization and maintain surface expression.
  • Washing:

    • Wash cells with FACS buffer to remove unbound antibody and reduce background signal.
  • Secondary Detection (if indirect):

    • If Tabalumab is not directly conjugated, use secondary detection reagents (e.g., streptavidin-PE for biotinylated Tabalumab).
  • Flow Cytometry Acquisition:

    • Acquire data using a flow cytometer equipped for the chosen fluorochrome (e.g., PE, APC).
    • Fluorescence intensity correlates with BAFF expression or antibody binding capacity.
  • Data Analysis:

    • Analyze mean fluorescence intensity (MFI) or percent positive cells to validate BAFF expression or antibody binding.
    • Use overlays and appropriate controls for comparison (e.g., isotype controls, unstained cells).

Key considerations:

  • Biosimilar antibodies from vendors like R&D Systems, BD Biosciences, and others are validated for use in flow cytometry and are available conjugated to fluorochromes such as PE, APC, Alexa Fluor dyes, enabling multiplexing and expanded applications.
  • Fc-mutated biosimilars (e.g., N297A) are preferred for minimizing background staining, especially when analyzing Fc receptor-expressing cells.
  • Controls include isotype-matched negative controls and, where relevant, cells not expressing BAFF.

Typical applications for these protocols include:

  • Validation of BAFF expression on cell surfaces during immune activation or after genetic modification.
  • Assessment of Tabalumab binding affinity and specificity for BAFF.
  • Development and validation of biosimilar antibody candidates for therapeutic applications or assay development.

If using a labeled Tabalumab biosimilar directly (PE/APC conjugate), the protocol is streamlined by eliminating secondary detection steps, maintaining sensitivity and specificity in flow cytometric assays.

Biopharma companies perform a comprehensive battery of analytical assays to establish the structural and functional similarity of biosimilars to their reference products. This analytical similarity assessment is the foundation of biosimilar development and regulatory approval.

Structural Characterization Assays

The structural assessment encompasses multiple levels of protein analysis. Primary structure evaluation includes amino acid sequencing and peptide mapping to confirm the correct protein sequence. Secondary and higher-order structure analysis utilizes techniques such as circular dichroism spectroscopy and nuclear magnetic resonance to assess protein folding patterns. Mass spectrometry and chromatographic techniques are extensively deployed to evaluate post-translational modifications, including glycosylation patterns, which can significantly impact protein function.

Physicochemical characterization involves determining molecular weight, charge variants, and thermal stability profiles. These properties are ranked by their potential risk of impact on the product's activity, pharmacokinetics, pharmacodynamics, safety, efficacy, or immunogenicity.

Functional Assessment Methods

Functional assays serve as the crucial bridge between structural data and clinical expectations, answering whether observed structural differences translate into functional significance. The functional characterization program typically includes two main categories of assays.

Binding assays evaluate the biosimilar's ability to interact with its intended targets, including antigen binding for monoclonal antibodies and receptor binding studies. For antibody-based biosimilars, Fc receptor binding assays assess interaction with immune receptors like FcγRIIIa, which is critical for antibody-dependent cellular cytotoxicity.

Cell-based biological assays measure the actual biological activity of the molecule in relevant cellular systems. These include potency assays that assess the functional activity of the biosimilar compared to the reference product, enzyme kinetics studies, and bioassays that measure specific biological responses.

Purity and Impurity Profiling

The analytical assessment includes rigorous comparison of purity and impurity profiles, which serves as a sensitive fingerprint of the entire manufacturing process. This analysis focuses on product-related variants such as aggregates, fragments, precursors, and other modified forms that may arise during manufacturing or storage.

Process-related impurities are also evaluated, including host cell proteins, DNA, and residual solvents that could impact safety or efficacy. The impurity profile comparison provides indirect evidence that the biosimilar developer has successfully replicated critical manufacturing process controls.

Analytical Strategy and Implementation

Manufacturers conduct head-to-head comparisons of the proposed biosimilar's properties against the reference product, with results required to fall within appropriate limits, ranges, or distributions. The approach often employs multiple complementary or orthogonal methods to better characterize product properties and more sensitively assess potential differences.

The analytical studies utilize highly sensitive analytical methods that allow measurement of molecular properties across multiple lots of both the biosimilar and reference product. This extensive characterization of the reference product establishes the standards against which the proposed biosimilar is evaluated.

Critical Quality Attributes Focus

The analytical similarity assessment involves identification of critical quality attributes (CQAs) that are relevant to clinical outcomes. The objective is to demonstrate a highly similar profile with respect to variations in these CQAs, with variations required to lie within the range established by the innovator product.

Regarding the specific question about Leinco biosimilars, the search results do not contain any information about Leinco or how their biosimilars are specifically used in analytical similarity studies. The information available focuses on general analytical approaches and regulatory requirements for biosimilar assessment rather than specific company products or applications.

References & Citations

1. Manetta, J. et al. (2014) J Inflamm Res. 7: 121–131
2. Clatworthy, MR. et al. (2013) Transplantation. 96(4): 413–420.
3. Lied, GA. and Berstad, A. (2011) Scand J Immunol. 73(1):1-7.
4. Büchler, JR. and Cano, MN. (1986) Jpn Heart J. 27(1):117-22.

Formats Available

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