This R-phycoerythrin (R-PE) 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 R-phycoerythrin (R-PE) conjugate is stable when stored at 2-8°C. Do not freeze.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
Next Day 2-8°C
Excitation Laser
Blue Laser (488 nm) and/or Green Laser (532 nm)/Yellow-Green Laser (561 nm)
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Natalizumab 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 ?
B
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 Natalizumab. Natalizumab binds to the alpha 4 subunit of α4β1 and α4β7 integrins. This product is for research use only.
Background
Natalizumab is characterized as a disease-modifying therapy for multiple sclerosis (a disease of the central nervous system (CNS)), and inflammatory bowel disease. It works by inhibiting the migration of leukocytes to inflammation sites. The VCAM-1 and α4β1-integrin interaction is necessary for leukocyte adhesion, firm attachment, and transmigration across the blood-brain barrier into the CNS. Natalizumab, a recombinant, humanized antibody, binds to α4β1 -integrin and blocks its interaction with VCAM-1. Hence, leukocyte migration into brain tissue is inhibited, thereby reducing inflammation and preventing the formation of multiple sclerosis lesions.1 Inflammation in the gut pertaining to inflammatory bowel disease can be controlled in a similar fashion. Blocking α4β7-integrin with a humanized, monoclonal antibody, specific to the α4β7 heterodimer inhibits the migration of leukocytes into the inflamed intestinal tissue, thus, reducing inflammation in the gut.2 This cost-effective, research-grade Anti-Human CD49D (Natalizumab) utilizes the same variable regions from the therapeutic antibody Natalizumab making it ideal for research projects.
Antigen Distribution
CD49D is a subunit of the integrin VLA-4, which is expressed on the cell surfaces of stem cells, progenitor cells, T and B cells, monocytes, natural killer cells, eosinophils, and neutrophils.
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 Natalizumab biosimilars are routinely used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantify Natalizumab in serum samples during biosimilar and reference product studies.
Key points on their application in PK bridging ELISAs:
Single Analytical Standard Approach: The industry consensus and best practice are to use a single PK assay with a single analytical standard—often the biosimilar—for quantification of both originator (reference) and biosimilar drug concentrations in test samples. This reduces assay variability, simplifies validation, and supports direct comparability required for biosimilar development and regulatory acceptance.
Standard Curve Preparation: Calibration standards are prepared by spiking the research-grade Natalizumab biosimilar into drug-free serum at defined concentrations (e.g., 50–12,800 ng/mL). This standard curve then allows quantification of unknown Natalizumab concentrations in patient serum by comparison.
Comparability Validation: Before the biosimilar is established as the calibration standard, proof of bioanalytical equivalence between reference and biosimilar Natalizumab in the ELISA format is required. This involves demonstrating that the assay measures both forms with equivalent accuracy and precision, typically evaluated by analyzing sample sets containing both product types and statistically confirming equivalence (e.g., 90% CI within 0.8–1.25 equivalence margins).
Quality Controls (QCs): Quality controls (QCs) at different concentrations are also prepared from both reference and biosimilar products when validating the method to assure recovery, precision, and accuracy across assay runs.
How the assay works:
The ELISA wells are coated with reagents that capture Natalizumab from serum samples; after washing, an HRP-conjugated probe detects bound Natalizumab, and color development is proportional to drug concentration. The unknowns and QC samples are quantified by interpolation from the biosimilar-based standard curve.
Examples:
Commercial ELISA kits (e.g., Assay Genie, KRIBIOLISA™) specifically state that their standards/calibrators are calibrated against commercially sourced biosimilar research-grade Natalizumab, rather than directly from the branded/originator product.
Validated methods used in clinical pharmacokinetic studies have employed biosimilar or originator as the reference standard provided that cross-reactivity and equivalence are demonstrated.
In summary, research-grade biosimilar Natalizumab is used as the assay reference standard in PK bridging ELISA after rigorous validation of analytical equivalence with the reference product, enabling accurate concentration measurements and comparability between biosimilar and reference serum samples.
Standard flow cytometry protocols using conjugated Natalizumab biosimilars (e.g., PE or APC-labeled) to validate CD49D (α4-integrin) expression or binding capacity typically involve quantitative assessment of both CD49d surface levels and antibody binding (receptor occupancy) in immune cells, most often T and B lymphocytes.
Essential protocol elements include:
Sample Preparation: Either whole blood or peripheral blood mononuclear cells (PBMCs) are isolated from treated or untreated patient samples.
Staining Panel:
Conjugated anti-CD49d antibody (e.g., PE-labeled): Used to directly quantify the surface expression of CD49d.
Natalizumab (or biosimilar) labeled with a fluorochrome (e.g., PE, APC): Used to assess unoccupied (free) CD49d sites or, when used in a competitive binding protocol, to determine the proportion of CD49d already bound by endogenous or therapeutic antibody.
Cell subset markers (e.g., anti-CD4, anti-CD8, anti-CD19) to gate on specific lymphocyte populations.
Viability dye is often included to exclude dead cells.
Incubation: Cells are incubated with the appropriate antibody mix at 4°C to prevent receptor modulation and antibody internalization.
Washing: Cells are washed to remove unbound antibody.
Acquisition: Samples are analyzed with flow cytometry, collecting enough events for reliable quantification of each major lymphocyte subset.
Data analysis:
Expression is quantified as molecules of equivalent soluble fluorochrome (MESF) per cell, or as mean/median fluorescence intensity (MFI).
Receptor occupancy (RO) or saturation can be calculated as the proportion of CD49d molecules occupied by Natalizumab (or biosimilar), often using dual-antibody approaches: one detects all CD49d and the other detects only unbound or bound sites, depending on clone specificity.
Results may also report percentage of CD49d+ cells and degree of saturation in each lymphocyte subset.
Example Protocol for Binding/Occupancy Assessment:
Incubate cells with labeled Natalizumab biosimilar (PE- or APC-conjugated), possibly after blocking Fc receptors.
Optionally, use a different fluorochrome-conjugated anti-CD49d antibody that binds only free (unbound) CD49d to distinguish total and available receptor pools.
Stain for major cell markers.
Wash, fix (if needed), acquire, and analyze.
Key Controls:
Isotype controls for each fluorochrome.
Unstained and FMO (fluorescence minus one) controls.
Cells from untreated donors as negative binding controls.
Applications in Literature:
Quantitation of CD49d molecules and bound Natalizumab per lymphocyte population in monitoring therapy, receptor saturation, and receptor occupancy in relapsing-remitting multiple sclerosis (RRMS) patients.
Direct validation of biosimilar binding to CD49d by competitive (blocking) binding assay with labeled biosimilar.
Common Readouts:
Surface expression: PE/APC fluorescence (linked to number of CD49d molecules or biosimilar binding per cell).
Occupancy/Saturation: Ratio of bound vs. unbound receptor, calculated via dual labeling or competitive binding.
Protocols can vary in exact antibody-fluorochrome combinations, sample type (whole blood versus PBMCs), and use of commercial versus biosimilar Natalizumab, but the outlined workflow and analytical rationale are consistent in published studies.
References to illustrative protocols and analysis:
[Quantitative flow cytometry for CD49d expression and Natalizumab binding, including use of PE-labeled reagents and assessment of saturation]
[Monitoring of CD49d expression and receptor occupancy in clinical studies using flow cytometry]
Biopharma companies confirm the structural and functional similarity of a proposed biosimilar to the originator drug through a suite of advanced analytical assays, focusing on multiple molecular characteristics. The Leinco biosimilar—assuming this refers to Leinco Technologies' biosimilar antibody reagents—is commonly used as a well-characterized comparator or as a control/reference standard to validate these analytical assays, but it is not an approved biosimilar drug and thus serves primarily for assay development, method qualification, and benchmarking purposes.
Analytical Assays Performed for Biosimilar Characterization:
Primary structure confirmation: Sequence analysis (such as peptide mapping and mass spectrometry) to confirm identical amino acid sequence.
Higher-order structure characterization: Techniques like circular dichroism (CD) and nuclear magnetic resonance (NMR) spectroscopy, as well as differential scanning calorimetry, to assess folding, aggregation, and overall three-dimensional shape.
Post-translational modifications: Analysis of glycosylation and other modifications to ensure comparable patterns between biosimilar and originator.
Purity and impurity analysis: Assessment using chromatography (HPLC, SEC) and capillary electrophoresis to detect and quantify aggregates and process-related impurities.
Binding assays: ELISA, surface plasmon resonance (SPR), and other ligand-binding studies to evaluate the biosimilar's ability to bind its target antigen or Fc receptors (critical for biologics such as monoclonal antibodies).
Biological/functional assays: Cell-based bioassays that measure mechanism-of-action (MoA) related activities (e.g., ADCC, CDC, neutralization assays) to confirm equivalent biological function and potency.
Immunogenicity assessment: Predictive in vitro assays and, eventually, in vivo studies to gauge the risk of immune responses.
Stability studies: Forced degradation and real-time stability tests to confirm product integrity under stress and over shelf-life.
Role of Leinco Biosimilars in Analytical Studies:
Leinco Technologies provides biosimilar antibody references and assay standards, which are used in R&D and quality control laboratories as analytical comparators or positive controls but are not themselves licensed therapeutic biosimilars.
These biosimilar-like reagents help standardize and validate analytical methods, ensuring accuracy and repeatability when characterizing genuine biosimilars and their reference products. Their main application is in assay development, pharmacokinetics studies, and bridging methodological gaps where the originator may be inaccessible (research-only use).
Key Principles:
The “totality of evidence” approach requires orthogonal analytical methods to cover structure, function, and safety aspects, supported by in vitro and in some cases in vivo comparability.
Method validation relies on rigorous controls and standards, for which well-characterized biosimilar-like reagents play a practical, supporting role when regulatory-grade reference products are unavailable.
These analytical efforts and the use of reference standards are foundational to gaining regulatory approval for a biosimilar, as they establish critical quality attributes and confirm there are no clinically meaningful differences in safety, purity, and potency compared to the originator.
References & Citations
1. Hutchinson, M. (2007) Ther Clin Risk Manag. 3(2):259-68.
2. Vandervoort, M. et al. (2005) N Engl J Med 352:2499-507.