Anti-Human CD49D (Integrin alpha 4) (Natalizumab) [Hu114]

Anti-Human CD49D (Integrin alpha 4) (Natalizumab) [Hu114]

Product No.: LT1100

- -
- -
Product No.LT1100
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
B
,
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
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
This biosimilar antibody is aseptically packaged and formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.2 - 7.4 with no carrier protein, potassium, calcium or preservatives added. Due to inherent biochemical properties of antibodies, certain products may be prone to precipitation over time. Precipitation may be removed by aseptic centrifugation and/or filtration.
Product Preparation
Recombinant biosimilar antibodies are manufactured in an animal free facility using only in vitro protein free cell culture techniques and are purified by a multi-step process including the use of protein A or G to assure extremely low levels of endotoxins, leachable protein A or aggregates.
Pathogen Testing
To protect mouse colonies from infection by pathogens and to assure that experimental preclinical data is not affected by such pathogens, all of Leinco’s recombinant biosimilar antibodies are tested and guaranteed to be negative for all pathogens in the IDEXX IMPACT I Mouse Profile.
Storage and Handling
Functional grade preclinical antibodies may be stored sterile as received at 2-8°C for up to one month. For longer term storage, aseptically aliquot in working volumes without diluting and store at ≤ -70°C. Avoid Repeated Freeze Thaw Cycles.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Natalizumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Applications Reported In Literature ?
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 commonly used as calibration standards (analytical standards) or reference controls in PK bridging ELISA assays to accurately quantify Natalizumab concentration in serum samples over time.

Context and Supporting Details:

  • In PK bridging ELISA assays, a single analytical standard—often a research-grade biosimilar—is selected and validated for use in generating the standard curve. This curve enables quantification of Natalizumab (whether originator or biosimilar) in patient serum. The rationale for using a biosimilar as the standard is to minimize assay variability and maintain consistency, since all test samples (biosimilar and reference product) are measured against the same calibrator.

  • Calibration standard role:

    • The research-grade biosimilar is serially diluted in appropriate matrix (often human serum) to create known concentration levels, typically spanning a wide, validated range (e.g., from low ng/mL to several μg/mL).
    • These standards are run alongside patient samples. The resulting optical density values (from ELISA readout) for standards are plotted to generate the calibration curve.
    • Unknown sample concentrations are then interpolated from this calibration curve, ensuring precise quantification.
  • Reference control role:

    • Both biosimilar and reference Natalizumab products are used as quality controls to verify assay performance and bioanalytical equivalence.
    • During assay validation, samples spiked with both biosimilar and reference product at several concentrations are assayed and quantified against the biosimilar standard curve. Statistical comparability (e.g., 90% CI within predefined interval) confirms that both products are measured equivalently by the assay.
  • Assay performance validation:

    • Assays are rigorously validated for precision, accuracy, sensitivity, and specificity. For example, validation often includes concentrations from 50 ng/mL to 12800 ng/mL, with multiple analysts and days to assure robustness.
    • Industry guidelines and regulatory agencies recommend this single-method approach to reduce inter-method variability and streamline blinded clinical studies.

Summary of Use in PK ELISA:

  • Biosimilar Natalizumab as calibrator: Used to build the calibration curve, quantify both biosimilar and reference drug concentrations in all serum samples, and establish bioanalytical equivalency.
  • Reference controls: Both biosimilar and reference product samples are spiked into serum to verify equivalence and validate assay precision and accuracy.

This methodological approach ensures accurate, reproducible measurement of Natalizumab concentration in PK studies and supports the regulatory requirements for biosimilar drug development.

The primary in vivo models where a research-grade anti-CD49D antibody is administered to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models, such as MC38, often combined with adoptive cell transfer and other immunomodulatory treatments. Humanized models appear less commonly in the context of anti-CD49D intervention for TIL profiling based on available data.

Key experimental details:

  • Syngeneic mouse models (e.g., MC38 colon carcinoma in C57BL/6 mice) are widely used because they have fully functional murine immune systems, allowing direct study of immune cell dynamics—including TIL characterization—following immunotherapy.
  • In the referenced study, anti-CD49D antibody alone did not significantly influence tumor growth in the MC38 model. However, when combined with invariant natural killer T (iNKT) cell transfer and α-galactosylceramide (αGC) injection, anti-CD49D enhanced iNKT cell tumor infiltration and IFN-γ production, resulting in reduced tumor growth.
  • Tumor-infiltrating lymphocytes (TILs) analyzed included iNKT cells, CD8 T cells, and NK cells. Anti-CD49D antibody treatment promoted increased infiltration and functional activation (e.g., IFN-γ production) of these lymphocyte populations within tumors.

Model features and rationale:

  • Syngeneic models allow a controlled tumor-host interaction with murine antibodies (including anti-CD49D) and enable comprehensive TIL profiling by flow cytometry and functional assays.
  • These models are preferred for initial mechanistic studies due to reproducibility, immune system intactness, and well-characterized immune infiltrate profiles.
  • Humanized mouse models (engrafted with human immune cells and tumors) are rarely detailed in the context of anti-CD49D antibody administration for TIL studies in available literature; syngeneic mouse models remain the standard for such immunotherapy mechanistic research.

TIL characterization highlights after anti-CD49D treatment:

  • Enhanced infiltration of iNKT, CD8, and NK cells.
  • Increased production of effector cytokines (e.g., IFN-γ).
  • Analysis typically performed by flow cytometry, cytokine profiling, and functional assays on extracted tumor tissue.

In summary:

  • Syngeneic mouse tumor models—particularly MC38 in C57BL/6 mice—are the primary platforms for in vivo studies of anti-CD49D antibody effects on tumor growth and TIL characterization.
  • Humanized models are not prevalent for this application based on current public research.
  • Experiments often combine anti-CD49D antibody with adoptive cell transfer to maximize immune-mediated tumor control and enable TIL analysis.

To date, there is no published research or clinical evidence indicating the use of the natalizumab biosimilar (or reference natalizumab) in combination with other immune checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars for synergistic effects in complex immune-oncology models. The available literature focuses exclusively on natalizumab’s role in multiple sclerosis (MS) treatment, where it is compared head-to-head with its biosimilar for efficacy, safety, and immunogenicity in MS patients. No studies have been conducted or reported that explore the use of natalizumab (biosimilar or originator) in oncology settings, let alone in combination with checkpoint inhibitors.

Background on Natalizumab and Biosimilars

  • Natalizumab is a monoclonal antibody targeting the α4-integrin, primarily used for relapsing-remitting multiple sclerosis (RRMS). Its mechanism—blocking immune cell migration across the blood-brain barrier—is distinct from that of immune checkpoint inhibitors like anti-CTLA-4 or anti-PD-1, which modulate T-cell activation in the tumor microenvironment.
  • Biosimilar natalizumab has been rigorously tested against the reference product in MS but has not been evaluated (nor is it approved) for use in oncology or in combination with other immunotherapies.

Current Practices in Immune-Oncology Combination Therapies

  • Combination therapies in immune-oncology typically involve drugs that target different immune checkpoints (e.g., anti-CTLA-4 plus anti-PD-1) or combine checkpoint inhibitors with chemotherapy, targeted therapy, or cellular therapies to enhance anti-tumor immune responses.
  • These strategies are based on the hypothesis that targeting multiple immune pathways can overcome resistance and improve clinical outcomes, but they come with increased risk of immune-related adverse events.
  • There is no rationale or precedent for combining natalizumab (a CNS-targeted adhesion molecule inhibitor) with peripheral T-cell checkpoint inhibitors, as their mechanisms and indications are entirely different.

Conclusion

Researchers have not used natalizumab biosimilars in conjunction with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 to study synergistic effects in immune-oncology models. The clinical development and testing of natalizumab biosimilars have been confined to neurological diseases, particularly MS, with no overlap into oncology or combination immunotherapy trials. Any speculation about such combinations would be theoretical and unsupported by current evidence. Future research in this direction would require a clear biological rationale, which is presently lacking.

In the context of immunogenicity testing, particularly for a Natalizumab biosimilar, a bridging ELISA can be employed to monitor a patient's immune response by detecting anti-drug antibodies (ADAs) against the therapeutic drug. Here's how a Natalizumab biosimilar could be used as the capture or detection reagent in a bridging ADA ELISA:

Basic Principle of Bridging ELISA

  1. Capture Reagent: The Natalizumab biosimilar is biotinylated and used as the capture reagent by immobilizing it on streptavidin-coated plates. This allows the capture of ADAs present in a patient's serum sample.

  2. Detection Reagent: A NATALIZUMAB BIOSIMILAR conjugated with a label such as horseradish peroxidase (HRP) is used as the detection reagent. This labeled biosimilar binds to the captured ADAs, forming a "bridge" between the two arms of the assay.

  3. Detection: After adding a substrate for HRP, the color change is measured, which is proportional to the concentration of ADAs in the sample.

Steps for Using Natalizumab Biosimilar in Bridging ELISA

Preparation of Reagents

  • Biotinylated Capture Reagent: Prepare the biotinylated Natalizumab biosimilar according to the manufacturer's instructions.
  • HRP-Labeled Detection Reagent: Conjugate the Natalizumab biosimilar with HRP to create the detection reagent.

Assay Setup

  1. Coat Plates: Add streptavidin to the ELISA plates and incubate to create a surface for capturing biotinylated reagents.
  2. Capture: Add the biotinylated Natalizumab biosimilar to the plates and incubate to allow it to bind to the streptavidin.
  3. Sample Addition: Add patient serum samples to the plates.
  4. Detection: Add the HRP-labeled Natalizumab biosimilar to bind to any ADAs that have bound to the captured drug.
  5. Signal Development: Add a suitable substrate for HRP to develop a colorimetric signal proportional to ADA concentration.

Data Interpretation

  • ADA Detection: The presence and concentration of ADAs are determined by measuring the optical density of the color change.

This method allows for the sensitive detection of ADAs against Natalizumab, providing valuable information on the patient's immune response to the drug.

Advantages

  • High Sensitivity: The bridging ELISA format is highly sensitive, allowing for the detection of low levels of ADAs.
  • High Throughput: Suitable for screening large numbers of samples.

Limitations

  • Specificity Concerns: The presence of other matrix components in serum may affect assay specificity; thus, high-quality reagents and blocking solutions are crucial.

Implementing this approach requires careful optimization of assay conditions to ensure accurate and reliable results.

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.
B
Flow Cytometry

Certificate of Analysis

Formats Available

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