Anti-Human CD49D (Integrin alpha 4) (Natalizumab) – APC

Anti-Human CD49D (Integrin alpha 4) (Natalizumab) – APC

Product No.: LT1103

- -
- -
Product No.LT1103
Clone
Hu114
Target
CD49D
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD49D; alpha 4 subunit of VLA-4 receptor; ITGA4; Integrin alpha-IV
Isotype
Human IgG4κ
Applications
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
RAMOS cell line injected into mice.
Product Concentration
0.2 mg/ml
Formulation
This Allophycocyanin (APC) 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 Allophycocyanin (APC) 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
Red Laser (650 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.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars
.
Cell Adhesion
.
Cell Biology
.
Immunology
.
Innate Immunity

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.

Research-grade Natalizumab biosimilars are used as standards (calibrators) or reference controls in pharmacokinetic (PK) ELISAs by serving as the quantifiable reference against which unknown drug concentrations in serum samples are measured. These standards are used to establish a reliable calibration curve that enables accurate determination of Natalizumab levels in test samples.

Essential Context and Details

  • Calibration Standards: In a PK bridging ELISA, serial dilutions of research-grade Natalizumab biosimilar are prepared at known concentrations and included on each assay plate. These calibrators provide the standard curve, against which the absorbance values of serum samples are compared to calculate unknown drug concentrations.

  • Choice of Standard: Assay developers may use research-grade biosimilar Natalizumab as the standard because it is structurally and functionally equivalent to the reference (branded) drug but is more practical or cost-effective to obtain for assay production and validation. Strong analytical comparability is established to confirm equivalence in assay response between the biosimilar and the reference product.

  • Reference Controls: Reference controls—serum or matrix samples spiked with known concentrations of biosimilar or reference product—are run alongside standards and test samples to monitor assay performance, precision, and accuracy.

  • Bioanalytical Validation: The method’s suitability to measure both biosimilar and reference Natalizumab is confirmed during assay development and validation. This includes statistical analyses to ensure that both act comparably as controls and calibrators within the assay’s dynamic range. Only if bioanalytical equivalence is demonstrated can the biosimilar standard be reliably used in clinical PK studies.

  • Example from Commercial Kits: Some Natalizumab ELISA kits explicitly state that their calibrators/standards are “calibrated against commercially sourced biosimilar research grade Natalizumab,” ensuring quantitative accuracy and traceability.

Workflow Summary

  1. Prepare Calibration Curve: Serial dilutions of research-grade biosimilar Natalizumab are pipetted into designated wells to create a standard curve.

  2. Run Serum Samples: Test serum samples are added to separate wells; these may contain unknown concentrations of Natalizumab.

  3. Assay Detection: A capture antibody (often anti-idiotypic) binds Natalizumab from standards or samples, and detection is achieved using an anti-human or anti-Natalizumab antibody conjugated to an enzyme (e.g., HRP).

  4. Calculate Concentrations: The resultant colorimetric signal is proportional to Natalizumab amount and is converted to concentration via the standard curve constructed from biosimilar calibrators.

  5. Quality Control: Reference controls (biosimilar or reference product at known concentrations) are also measured to confirm assay reliability.

Additional Notes

  • Assay Equivalence: Regulatory and industry standard practice mandates validation that the biosimilar and reference product yield equivalent results in the PK assay, ensuring that quantitative differences, if present, reflect true differences in patient samples, not assay artifacts.
  • Advantages: Using a single standard (biosimilar or reference) reduces inter-assay variability and is considered best practice for comparative PK studies of biosimilars and originators.

In conclusion, research-grade Natalizumab biosimilars serve as traceable, validated reference standards and controls, enabling robust, reliable measurement of drug concentrations in serum PK bridging ELISAs.

Standard flow cytometry protocols for validating CD49D (integrin α4, VLA-4) expression levels or binding capacity using a PE- or APC-conjugated Natalizumab biosimilar involve quantitative assays that measure both the expression of CD49D and its saturation by the therapeutic antibody.

Key steps and components typically include:

  • Sample Preparation

    • Use either whole blood or PBMCs (peripheral blood mononuclear cells).
    • Isolate cells quickly to reduce receptor modulation ex vivo.
  • Staining Protocol

    • Incubate samples with a Natalizumab biosimilar directly conjugated to a fluorochrome (e.g., PE or APC).
    • Include an antibody against CD49D (PE-conjugated anti-CD49D for total expression).
    • To assess bound vs. free CD49D:
      • Use a fluorescent anti-human IgG4 (Fc-specific, labeled with PE/APC) to detect Natalizumab bound to CD49D on the cell surface.
      • Optionally, incubate cells with an excess of unconjugated Natalizumab to saturate all available CD49D, then stain to determine total receptor capacity.
  • Controls

    • Isotype controls for fluorochrome-conjugated antibodies.
    • Unstained controls to set gates.
  • Flow Cytometric Analysis

    • Gate on lymphocyte populations (e.g., CD4^+^ T, CD8^+^ T, CD19^+^ B cells).
    • Quantitate:
      • CD49D molecules/cell (using fluorescence intensity calibration with standards or beads).
      • Natalizumab-bound CD49D (using anti-IgG4 or direct fluorochrome signal from conjugated Natalizumab).
      • CD49D saturation %: Calculate as the fraction of receptor occupied by Natalizumab.
    • Express results as mean fluorescence intensity (MFI), number of molecules/cell, and percentage saturation.

Application context:

  • Used to assess pharmacodynamics in MS patients receiving Natalizumab therapy, especially for extended-interval versus standard-interval dosing regimens.
  • Assays are crucial for monitoring treatment efficacy, receptor occupancy (RO), and personalizing dosing strategies.

Typical Protocol References

  • Quantitative flow cytometry with labeled Natalizumab biosimilar and anti-CD49D antibody for receptor occupancy and expression.
  • Detection of cell-bound Natalizumab using fluorescent anti-IgG4 antibody, and assessment of free CD49D via saturation experiments.
  • Whole blood or PBMC-based staining, analysis via standard multiparametric cytometry.

Summary of the protocol’s workflow:

  1. Stain cells with PE/APC-Natalizumab biosimilar.
  2. Stain with PE/APC-anti-CD49D (for total receptor).
  3. Optionally, stain with fluorescent anti-human IgG4 to detect drug-bound receptor.
  4. Analyze by flow cytometry for:
    • Total CD49D expression.
    • Natalizumab Rx receptor occupancy.
    • Calculate % saturation.

This approach is validated for clinical and translational research settings to monitor drug-receptor pharmacodynamics and optimize individualized therapy in MS.

Biopharma companies typically use a comprehensive set of analytical assays to confirm the structural and functional similarity of a proposed biosimilar to the originator (reference) drug. These assays focus on both detailed molecular characterization and functional performance, ensuring that any differences do not impact clinical performance.

Essential Analytical Assays:

  • Structural Characterization:

    • Primary structure analysis: Peptide mapping, amino acid sequencing, and mass spectrometry to confirm the amino acid sequence identity and integrity.
    • Higher-order structure: Techniques such as circular dichroism, Fourier-transform infrared (FTIR) spectroscopy, and NMR are used to examine secondary and tertiary structures.
    • Post-translational modifications: Glycosylation profiling, charge variants, and disulfide bond mapping to ensure molecular consistency.
    • Purity and impurities: Size exclusion chromatography and capillary electrophoresis to evaluate purity, aggregates, fragments, and process-related impurities.
    • Product-related variants: Detection of aggregates, fragments, and other modified forms is crucial.
  • Functional Comparison:

    • Biological (cell-based) assays: Assess the biological activity and mechanism of action, often directly measuring receptor activation or cell proliferation/inhibition.
    • Binding assays: Examine affinity and kinetics with target antigens (e.g., Fc receptor binding for antibodies).
    • Potency assays: Functional activity is measured relative to a reference standard to confirm equivalence.
    • Enzyme kinetics: Applied if the biologic is an enzyme to compare catalytic activity.
  • Orthogonal methods: Multiple complementary (orthogonal) techniques are applied to provide robust confirmation of similarity and to sensitively detect potential differences.

Critical Quality Attributes (CQAs):All assays are focused on CQAs—molecular properties most closely related to biologic activity, pharmacokinetics, pharmacodynamics, safety, and immunogenicity. Demonstrating biosimilarity requires that any variations in these CQAs between the biosimilar and originator are within acceptable limits.

Regulatory and Strategic Considerations:Analytical similarity is the foundation for biosimilar approval, reducing the need for extensive clinical testing if enough analytical and functional data demonstrate high similarity. The standards for these comparisons are informed by ICH Q6B and regulatory agency guidelines.

Leinco Biosimilar Role:No specific information on the use of Leinco biosimilars in comparative analytical studies was found in the retrieved results. Generally, companies like Leinco supply reference standards, biosimilar controls, or critical reagents (such as monoclonal antibodies, proteins, or assay kits) used as controls or comparators in these assays. If Leinco produces biosimilars, their products would typically be applied as analytical comparators or controls to benchmark assay performance or for method development. Without more detailed, source-confirmed information, this is an informed inference based on industry practice.

Key insights:

  • Structural and functional similarity is established using an extensive panel of precise and orthogonal analytical assays.
  • Functional equivalence is as critical as structural sameness, and robust assay data support regulatory approval.
  • The specific role of Leinco biosimilars is not detailed in the available results, but these products are generally used as reference materials or analytical standards in biosimilar testing.

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.
Flow Cytometry

Certificate of Analysis

Formats Available

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