This DyLight® 488 conjugate 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 DyLight® 488 conjugate is stable when stored at 2-8°C. Do not freeze.
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Alemtuzumab 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.
Additional Reported Applications For Relevant Conjugates ?
FA WB B N
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 antibody uses the same variable region sequence as the therapeutic antibody Sarilumab. Sarilumab binds to the transmembrane and soluble forms of the IL-6 receptor. This product is for research use only.
Background
IL-6 and its signaling pathway play a part in immune response regulation, inflammation, and hematopoiesis.2 Sarilumab is a research-grade recombinant human monoclonal IL-6 receptor antagonist. It specifically binds to both the transmembrane and soluble forms of the IL-6 receptor, thus inhibiting IL-6–mediated cis and trans-signaling in a dose-dependent manner.1 Therapeutic Sarilumab, also known by the trade name Kevzara, is currently used to treat Rheumatoid Arthritis1, however, as of March 2020, The Feinstein Institute of Northwell Health publicized a study on "a human antibody that may prevent the activity" of IL-6 for the treatment of COVID-19.3 Anti-Human IL-6 (Sarilumab) utilizes the same variable regions from the therapeutic antibody Sarilumab making it ideal for research projects.
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 Sarilumab biosimilars are used in pharmacokinetic (PK) bridging ELISA assays as analytical standards (calibrators) and occasionally as reference controls to enable accurate quantification of Sarilumab (the drug) concentration in serum samples during bioanalytical and biosimilarity studies.
Essential context and process:
Calibration Standard Curve: The biosimilar is serially diluted in a matrix (often 10% human serum in PBST) to generate a standard curve—this curve is essential for quantifying unknown Sarilumab concentrations in test samples by interpolating their ELISA signal (e.g., optical density).
Reference Control: Reference controls, sometimes run in parallel, serve to verify assay performance and may be either the biosimilar or the reference product (originator Sarilumab), ensuring reliable detection across different assay runs.
Assay Format: In bridging ELISA formats specific for Sarilumab quantitation, a capture reagent (commonly human IL-6Rα or an anti-Sarilumab antibody) binds Sarilumab in the sample, and a detection antibody (often anti-human IgG Fc/κ or biotin-conjugated) labels the antibody–antigen complex, leading to a measurable colorimetric signal proportional to drug concentration.
Assay Qualification: Before routine use, the assay is validated to confirm that both biosimilar and reference forms of Sarilumab give bioanalytically comparable results—demonstrating similar precision and accuracy within predefined equivalence margins.
Methodological Justification:
The use of a single analytical standard (often the biosimilar) for both biosimilar and reference product quantification reduces variability, avoids crossover corrections, and aligns with regulatory and industry best practices, provided assay validation confirms equivalence.
Practical Implementation Example:
Prepare Sarilumab biosimilar standards at set concentrations in serum matrix, covering the expected sample range (e.g., 50 to 12,800 ng/mL).
Use these standards to construct the ELISA calibration curve for each assay.
Test serum samples from PK studies alongside standards; interpolate their absorbance readings from the standard curve to obtain drug concentrations.
Summary Table: Role of Sarilumab Biosimilar in PK Bridging ELISA
Application
Purpose
Rationale/Requirement
Calibration Standard
Generate standard curve to quantify unknowns
Must ensure calibrator is bioanalytically equivalent
Reference Control
Monitor assay performance across plates/runs
May use either biosimilar or originator for comparability
Assay Development
Validate method performance for both biosimilar and originator Sarilumab
Meet precision, accuracy, and equivalence criteria
Key requirements:
Biosimilar used as calibrator must be cross-validated against the reference in the assay to demonstrate no matrix or affinity differences that affect quantitation.
Regulatory and scientific consensus supports this unified calibration approach as best practice in biosimilar PK studies.
References used:
For standard and control roles, calibration workflow: , .
For typical ELISA formats and biosimilar specifics: , .
For broader methodological and regulatory context: .
Conjugated Sarilumab biosimilars (e.g., PE- or APC-labeled) are used in flow cytometry to assess IL-6R (CD126) expression or validate binding capacity on cells, typically using protocols similar to other antibody-based surface staining procedures. Standardized procedures generally follow these steps:
1. Cell Preparation:
Single-cell suspensions are prepared from the target cell type (primary cells or cell lines).
Wash cells with PBS containing 1% BSA and 0.09% sodium azide to minimize nonspecific binding and maintain cell viability.
Cell concentrations typically range from 1 × 10⁵ to 1 × 10⁶ cells per tube.
2. Blocking (Optional but Recommended):
Incubate cells with Fc receptor blocking reagent or excess human IgG to prevent non-specific Fc-mediated binding, particularly when working with immune cells.
3. Antibody Staining:
Add the conjugated Sarilumab biosimilar (e.g., APC- or PE-labeled) directly to the cell suspension.
Recommended concentrations vary by supplier, but typical usage is ≤ 1.0 µg antibody per 1 × 10⁶ cells in 100 µL staining volume; titration is advised for optimal signal-to-noise.
Incubate for 20–30 minutes at 2–8°C or at room temperature, protected from light.
4. Washing:
Wash cells 1–2 times with PBS/BSA buffer to remove unbound antibody.
5. Data Acquisition:
Resuspend cells in appropriate buffer (e.g., PBS with 1% BSA).
Acquire samples on a flow cytometer with the correct configuration:
PE: excitation at 488 nm (blue laser), emission ~575 nm.
APC: excitation at 633–650 nm (red laser), emission ~660 nm.
Collect at least 10,000 events for robust analysis.
6. Analysis:
Gate on single, viable cells based on forward/side scatter and viability dye if included.
Assess IL-6R expression as the proportion of positive cells and/or as median fluorescence intensity (MFI).
7. Controls:
Use isotype controls (e.g., APC- or PE-conjugated human IgG1) to account for nonspecific staining.
Include unstained and single-color controls for compensation and gating.
Validation Purposes:
To quantitate IL-6R expression: Stain IL-6R–expressing and negative cell populations for comparison.
To assess binding capacity: Include competition or saturation assays, pre-incubating with excess unconjugated antibody to demonstrate specificity.
Example Protocols/References:
Leinco Technologies: APC-conjugated Sarilumab biosimilar, “≤ 1.0 µg per 10⁶ cells in 100 µl” for surface staining in flow cytometry. Titration and optimal working dilutions are advised for each application.
ichorbio: APC-conjugated Sarilumab biosimilar, for use in flow cytometry and IHC, with 1% BSA and 0.09% sodium azide as part of the staining buffer, titration advised per application.
R&D Systems: Demonstrates use of Sarilumab biosimilar for flow cytometric detection of IL-6R alpha (CD126) on engineered HEK293 cells, following standard protocols for fluorescent antibody staining.
Key Details:
Detection sensitivity depends both on the fluorochrome and on antibody affinity.
The same basic workflow applies to other fluorescent labels (PE, FITC, etc.).
Protocols may require customization based on cell type and expression levels.
No available literature describes novel or unique steps specific to Sarilumab biosimilar conjugates compared to standard antibody–antigen staining in flow cytometry. If used to functionally validate IL-6R engagement, blocking/competition assays are added, but core protocols remain as above.
Biopharma companies utilize a comprehensive suite of analytical assays—including advanced structural and functional characterization—to confirm the similarity of a proposed biosimilar to the originator drug. These assessments target critical quality attributes (CQAs) encompassing physiochemical, structural, and biological potency features, using multiple validated and often orthogonal methods.
Higher order structure (e.g., circular dichroism, NMR, X-ray crystallography)
Post-translational modifications (e.g., glycosylation analysis via mass spectrometry)
Aggregate and impurity assessment (e.g., size-exclusion chromatography, electrophoresis)
Functional Characterization:
Binding assays (e.g., antigen or receptor binding, Fc-receptor assays)
Cell-based potency assays (e.g., in vitro bioactivity)
Enzyme kinetics (if relevant)
Biological activity panels covering all known mechanisms of action
Comparative analysis of activity, pharmacokinetics/dynamics, safety, efficacy, and immunogenicity attributes
Impurity and Variant Profiling:
Detailed impurity fingerprinting (process-related and product-related)
Sensitive comparison of aggregates, fragments, and other product variants
Orthogonal and Multi-Lot Analysis:
Manufacturers typically deploy complementary (orthogonal) methods and analyze multiple lots of both biosimilar and reference product to ensure robust comparability and sensitive detection of differences.
Critical Quality Attribute Focus:
Analyses are prioritized based on the CQAs most likely to impact clinical activity, PK/PD, safety, or immunogenicity.
Leinco Biosimilar Usage in Comparative Studies
Leinco Technologies produces biosimilar antibodies and proteins that are applied in these comparative analytical studies, typically as benchmark molecules or assay controls. In biosimilar development workflows, a Leinco biosimilar may serve as:
A reference standard in binding assays, potency assays, or impurity characterizations to evaluate the analytical methods and ensure accuracy and reproducibility.
An alternate comparator for analytical platforms to validate orthogonal techniques or cross-verify assay sensitivity and specificity.
Typically, Leinco biosimilars are used when there is a need for high-quality, well-characterized material to enable head-to-head studies against both the proposed biosimilar and the originator drug. This helps confirm assay performance, calibrate results, and validate the detection of structural or functional differences in critical quality attributes. While Leinco products themselves are not approved substitutes for the originator in regulatory submissions, they are regularly cited in peer-reviewed studies as part of assay validation and method development prior to formal biosimilarity assessment.
If further details on a specific Leinco biosimilar's role are required, refer to published assay validation data or Leinco’s technical documentation, as the precise application varies with the biosimilar product and targeted biological activity.
Summary Table: Structural and Functional Assay Categories
Attribute Evaluated
Representative Assays
Role of Leinco Biosimilar
Primary & Higher Order Structure
Peptide mapping, CD, NMR, MS
Reference standard for comparison
Post-Translational Modifications
Glycosylation, phosphorylation analysis
Calibration/control in MS assays
Biological Potency/Function
Cell-based potency/binding assays
Assay control or benchmark
Impurities/Aggregates
Chromatography, electrophoresis
Validation sample, process control
These methods ensure robust and sensitive comparison between the biosimilar and the originator, supporting regulatory acceptance upon confirmation of high analytical similarity.
References & Citations
1. Kevzara (sarilumab) injection [prescribing information]. Tarrytown, NY: Regeneron Pharmaceuticals; Bridgewater, NJ: sanofi-aventis U.S.; May 2017.
2. Yoshida Y, Tanaka T. Interleukin 6 and rheumatoid arthritis. Biomed Res Int. 2014;2014:698313.
3. "Northwell Health Initiates Clinical Trials of 2 COVID-19 Drugs". 21 March 2020.