This antibody was produced by phage display technology.
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.
Storage and Handling
This biotinylated antibody is stable when stored at 2-8°C. Do not freeze.
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Briakinumab biosimilar antibody for staining cells in flow cytometry is ≤ 1.0 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application. ELISA
Additional Reported Applications For Relevant Conjugates ?
B N WB IF IP
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 Briakinumab. Briakinumab recognizes both human IL12 and IL23 via IL-12/23p40. This product is for research use only.
Background
Briakinumab is a human monoclonal antibody targets the p40 subunit shared by interleukins 12 and 23. IL-12 associates with IL-23α to form the heterodimeric cytokine IL-23. IL-23 is associated with various autoimmune inflammatory diseases, and is particularly highly expressed in psoriasis skin lesions. In addition, IL-23 is suspected to play a role in tumorigenesis. Briakinumab binds to and neutralizes human IL-12 and IL-23 (via their shared p40 subunit) and is being investigated for the treatment of rheumatoid arthritis, inflammatory bowel disease, and multiple sclerosis. Anti-Human IL 12/23 (Briakinumab) utilizes the same variable regions from the therapeutic antibody Briakinumab making it ideal for research projects.
Antigen Distribution
IL-12 is produced by dendritic cells, macrophages, neutrophils, and human B-lymphoblastoid cells. IL-23 is mainly secreted by activated dendritic cells, macrophages or monocytes.
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 Briakinumab biosimilars can be used as calibration standards (analytical standards) or reference controls in a pharmacokinetic (PK) bridging ELISA to measure drug concentration in serum samples by serving as quantifiable, well-characterized proteins that establish the assay’s standard curve and verify analytical performance.
Essential context and supporting details:
Calibration Standards:
Research-grade Briakinumab biosimilars are prepared at known concentrations and used to generate a standard curve within the ELISA assay, allowing quantitation of Briakinumab in test serum samples.
The biosimilar is typically chosen as the analytical standard for both the biosimilar and reference (innovator) drug concentrations, provided that analytical equivalence (bioanalytical comparability) has been validated within the method.
This approach reduces assay variability versus using multiple standards and simplifies cross-comparisons in biosimilar PK studies.
Reference Controls:
In addition to standards, research-grade biosimilars can serve as positive controls or QC samples to monitor assay accuracy and precision across runs.
Quality control samples are prepared at relevant concentrations using the biosimilar and (sometimes) reference drug, and both are quantified within the same validated method to confirm the method's reliability throughout clinical sample analysis.
How this works in a PK bridging ELISA:
The PK bridging ELISA typically uses capture and detection antibodies specific to Briakinumab or its unique epitopes.
The assay is qualified and validated for serum/plasma matrices, and the standard curve is created by spiking serial dilutions of research-grade Briakinumab biosimilar into blank serum, covering a relevant concentration range (e.g., 50 ng/mL–12,800 ng/mL).
Unknown samples from preclinical or clinical studies are then quantified by interpolating their signal off the biosimilar-derived standard curve.
In a properly validated method, both innovator (reference) and biosimilar formulations of Briakinumab should yield comparable results when measured against the biosimilar standard, if bioanalytical equivalence is demonstrated.
Key points from regulatory and industry practice:
Current best practice is to use a single, well-validated PK assay and a single analytical standard (often a research-grade biosimilar) for measuring both biosimilar and reference Briakinumab drug concentrations in bridging ELISA studies.
Before establishing the biosimilar as the standard, rigorous testing (precision, accuracy, statistical comparability) must demonstrate bioanalytical equivalence between the biosimilar used as the standard and the product(s) being measured in real samples.
Regulatory guidance emphasizes such validation as critical for the integrity of PK similarity (bioequivalence) assessments for biosimilars.
Additional technical details:
Commercial suppliers offer research-grade Briakinumab biosimilars purified to high quality (often >95%, low endotoxin, and free of preservatives/carriers), suitable for calibration and as assay controls in ELISAs.
These biosimilars are expressly designated For Research Use Only and must not be repurposed for clinical application, but are standard in drug monitoring method development.
Summary table: Use of Research-grade Briakinumab Biosimilars in PK Bridging ELISA
Role in ELISA
Purpose
Condition for Use
Calibration Standard
Generate standard curve to quantify drug in serum
Must be validated as bioanalytically equivalent
Reference/QC Control
Monitor accuracy, precision across assay runs
Used at low/medium/high concentrations during batch runs
This standardization and validation enable confident and robust measurement of Briakinumab concentrations in serum samples from both preclinical and clinical studies, facilitating accurate PK analysis and supporting regulatory biosimilarity claims.
The standard Flow Cytometry protocol for using a conjugated Briakinumab biosimilar (e.g., PE- or APC-labeled) to assess expression or binding of the IL-12/IL-23 p40 target follows established staining procedures for monoclonal antibodies, with application-specific titrations and controls. Below are the key steps and considerations based on published protocols and product datasheets.
Key Steps in Flow Cytometry Using Conjugated Briakinumab Biosimilar:
Antibody Selection & Preparation: Use a Briakinumab biosimilar directly conjugated to PE, APC, or another fluorochrome, matching your cytometer’s laser configuration.
Suggested Starting Concentrations:
PE-conjugate: ≤ 1.0 μg per 10⁶ cells in 100 μL staining volume.
APC-conjugate: Empirically recommended, e.g., 0.2 mg/mL stock; titration to determine optimal staining concentration is strongly advised.
A lower concentration (e.g., 0.25 μg per 10⁶ cells) may be sufficient for certain biosimilar preparations.
Cell Preparation: Harvest and count the relevant cell population (e.g., PBMCs, dendritic cells, macrophages, or engineered cell lines expressing human IL-12/23 p40).
Fc Blocking: To reduce nonspecific binding, pre-incubate cells with human IgG or a commercial Fc block, especially for cells with high Fc-receptor expression.
Antibody Staining:
Resuspend 10⁶ cells in 100 μL staining buffer (PBS + 1% BSA or FBS).
Add conjugated Briakinumab to the desired final concentration.
Incubate for 20–30 min at 4 °C, protected from light.
Wash Steps: Wash cells with 1–2 mL of staining buffer, centrifuge at 300–400g for 5 min, and discard supernatant. Repeat wash if needed.
Viability Stain (optional): Include a viability dye to distinguish live/dead cells.
Fixation (optional): For fixed-cell analysis, fix cells in 1–2% paraformaldehyde for 10–20 min after staining.
Acquisition: Analyze samples using flow cytometry, collecting at least 10,000 events per sample. Use the appropriate laser and filter for the fluorochrome (e.g., red laser for APC).
Controls to Include:
Isotype control: Human IgG1 of matching species/fluorochrome to determine background.
Unstained and single-stained controls: For setting compensation.
Negative/positive cell controls: Cells known not to express (or express) IL-12/23 p40.
Data Analysis: Assess mean fluorescence intensity (MFI) or percent positive cells to quantify expression level or antibody binding capacity.
Notes on Target Validation and Protocol Optimization:
Briakinumab specifically binds the p40 subunit shared by human IL-12 and IL-23, thus detecting either cytokine where p40 is present.
Titration of antibody for your specific cell type and biosimilar lot is essential for optimal results—monitor for background and non-specific staining.
Product datasheets and Certificate of Analysis (CoA) from suppliers should be checked for any special instructions or changes in protocol for specific conjugates or lots.
Summary Table: Typical Protocol Steps
Step
Description/Consideration
Antibody Concentration
≤ 1.0 μg/10⁶ cells (PE), titrate for optimal; ≤ 0.25 μg/10⁶ cells effective in some cases
Staining Volume
100 μL per 10⁶ cells
Incubation Time/Temp
20–30 min at 4 °C, protected from light
Wash Buffer
PBS + 1% BSA or FBS
Analysis
By flow cytometry, use proper compensation controls and gating strategies
Controls
Isotype, unstained, positive/negative targets
References for Protocol Specificity:
The datasheets from Leinco Technologies (PE-conjugate) and iChorBio (APC-conjugate) provide current and widely accepted protocol guidance.
Titration and optimization are universally recommended.
For target-expressing cells, see biosynthetic or stimulated primary immune cell models.
If you have a specialized application (e.g., cell-line development, competitive binding analysis), protocols may require further adaptation. Always review the most recent product datasheet for your specific conjugate.
Biopharma companies perform a comprehensive panel of analytical assays to confirm both the structural and functional similarity of a proposed biosimilar to its reference (originator) product. These assays focus on critical quality attributes (CQAs), which are properties shown to affect clinical performance, such as safety and efficacy.
Higher-order structures: Techniques like circular dichroism (CD), differential scanning calorimetry (DSC), and NMR to analyze secondary/tertiary structure.
Post-translational modifications: Glycosylation profiling by mass spectrometry or chromatography.
Purity and impurities: Methods such as size-exclusion chromatography (SEC) to detect aggregates, fragments, or other variants.
Physicochemical Properties:
Isoform/charge variants: Isoelectric focusing or capillary electrophoresis to profile charge differences.
Molecular weight: Mass spectrometry or SDS-PAGE.
Functional Characterization:
Binding assays: Assess the biosimilar's affinity for its molecular target (e.g., antigen, receptor).
Biological activity assays: Cell-based assays to measure the product’s intended biological effect or “potency”.
Mechanism-of-action bioassays: For antibodies, may include antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), or Fc receptor binding.
Enzyme kinetics, if applicable.
Stability Studies:
Assess product behavior under stress (e.g., temperature, light) to verify that the biosimilar degrades or aggregates similarly to the reference.
These assays are typically performed in a head-to-head comparison between the biosimilar and multiple lots of the originator to account for batch-to-batch variability. Orthogonal methods (distinct technologies measuring the same attribute) are employed for robust confirmation and to address any subtle differences.
Leinco Biosimilar Use in Analytical Similarity Studies:
While the term "Leinco biosimilar" refers to biosimilar reference materials offered by Leinco Technologies, these are typically used as well-characterized benchmark controls in biosimilar analytical studies. When developing or validating assays, companies use reference biosimilars from suppliers like Leinco to:
Benchmark analytical performance: Ensure assay sensitivity, specificity, and reproducibility using highly characterized biosimilar standards.
Compare candidate molecules: Verify that test biosimilars match both the originator and established reference standards (like Leinco’s biosimilars) in structure and function.
Facilitate assay development: Provide a non-clinical but highly similar material for method standardization and troubleshooting, especially during early-stage development.
The direct role of Leinco biosimilars is largely as controls or reference comparators in assay validation, especially in laboratories without access to the original branded drug or when large-scale, non-clinical controls are needed for routine testing. Their use helps ensure robust, reproducible analytics throughout the biosimilar development pipeline.
In summary, biopharma companies use a suite of advanced, validated structural and functional assays—including orthogonal methods—to confirm biosimilarity, and Leinco biosimilar products serve as well-characterized reference controls to support assay development and benchmarking.
References & Citations
1. Vsn, M. et al. (2016) VALUE IN HEALTH 19 PSS5:A123