Anti-Human α4β7 Integrin (Vedolizumab) [Clone LDP-02]

Anti-Human α4β7 Integrin (Vedolizumab) [Clone LDP-02]

Product No.: V204

- -
- -
Product No.V204
Clone
LDP-02
Target
α4β7 integrin
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Peyer patches-specific homing receptor LPAM-1 α4: CD49d
Isotype
Human IgG1κ
Applications
B
,
ELISA
,
FA
,
FC
,
LCI

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Rhesus Monkey
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Humanized version of the Act-1 monoclonal antibody. Immunogen unknown.
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.
State of Matter
Liquid
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
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
B,
ELISA,
FA,
FC,
LCI
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 Vedolizumab. LDP-02 (Vedolizumab) activity is directed against human, cynomolgus and rhesus monkey α4β7 integrin. Vedolizumab binds to the α 4 β 7 integrin, but not to the α 4 β 1 or α E β 7 integrins. Vedolizumab binds to a subset of human peripheral blood memory CD4 + T lymphocytes (25%) that includes gut-homing IL-17 T helper lymphocytes as well as to eosinophils, naïve T helper lymphocytes, naïve and memory cytotoxic T lymphocytes, B lymphocytes, natural killer cells and basophils but does not bind to the majority of CD4 + T lymphocytes (60%), neutrophils and most monocytes.
Background
Integrins are a large family of heterodimeric transmembrane molecules that mediate adhesion, migration, cell survival, and cell differentiation. The heterodimeric integrin receptor α4β7 is a cell surface glycoprotein that consists of β7 paired with α4 (CD49d) . The α4β7 integrin is a lymphocyte receptor for the mucosal vascular addressin MADCAM-11. MADCAM-1 is a cell adhesion leukocyte receptor expressed by mucosal venules that helps direct lymphocyte traffic into mucosal tissues and regulates the passage and retention of leukocytes2. α4β7 integrin preferentially mediates migration of lymphocytes into gastrointestinal tissue3. This interaction directs the migration of leukocytes into inflamed intestinal tissue4 and is a target for treating intestinal bowel disease.

Vedolizumab is a humanized version of the Act-1 monoclonal antibody that specifically recognizes the α4β7 integrin receptor3,4. Vedolizumab selectively blocks gut lymphocyte trafficking by inhibiting the migration of lymphocytes to the gastrointestinal mucosa during the inflammatory process5. Specifically, migration of memory T lymphocytes into inflamed gastrointestinal parenchymal tissue is inhibited. This is accomplished by blocking immune cell homing to the gut via disruption of α4β7 integrin adhesion on the cell surface of immune cells to its ligand MADCAM-1 on the intestinal endothelium6.

Vedolizumab selectively inhibits the adhesion of α 4 β 7 -expressing cells to MADCAM-1 and fibronectin but not VCAM-17. Consequently, vedolizumab does not induce the systemic immunosuppression seen with anti-α4 chain monoclonal antibodies. Rather, vedolizumab yields gut-selective anti-inflammatory activity8. Additionally, vedolizumab does not interfere with trafficking to the central nervous system3.

Antigen Distribution
α4β7 integrin has variable expression on circulating B and T lymphocytes.
Ligand/Receptor
Binds to HIV-1 gp120, MADCAM1, fibronectin, VCAM-1
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Adhesion
.
HIV
.
Immunology
.
Inflammatory Disease

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 Vedolizumab biosimilars are used as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA by serving as the analytical standard against which all serum sample measurements are quantified. This enables accurate determination of drug concentration in clinical samples and ensures comparability when bridging between biosimilars and reference products.

In a typical PK bridging ELISA for Vedolizumab biosimilars:

  • Calibration standards are prepared using the research-grade biosimilar at known concentrations in a human serum matrix. These standards establish a standard curve, which is used to interpolate Vedolizumab concentrations in unknown patient samples.
  • Reference controls—which may include both the biosimilar and the reference biologic—are included in the assay to verify bioanalytical equivalence. These controls are measured alongside standards and test samples to ensure the assay accurately detects both products within predefined equivalence criteria.
  • The single-assay approach is preferred, meaning the biosimilar (through its analytical standard preparation) is used for quantification of both biosimilar and reference (originator) Vedolizumab in bridging studies. This methodology minimizes variability and eliminates the need for multiple assays or crossover analyses.
  • Assay validation involves testing the precision, accuracy, specificity, and sensitivity of the method using sets of standards and quality control (QC) samples prepared from the biosimilar and/or reference product. For instance, validation studies may include several independent sets of calibrators and controls at different concentrations, analyzed over several days, to statistically confirm method performance.

Key technical points:

  • The ELISA kits typically use Vedolizumab-specific monoclonal antibodies for high specificity, ensuring no cross-reaction with other therapeutics or serum proteins.
  • Calibration standards made from the biosimilar are serially diluted and incubated in microtiter wells to generate a standard curve. Sample concentrations are interpolated from this curve.
  • Reference controls (biosimilar and/or reference Vedolizumab) are processed similarly to demonstrate analytic equivalence and validate the assay.

This process ensures the PK bridging ELISA is robust, reproducible, and scientifically sound for accurate quantification and regulatory comparability studies during biosimilar development.

The primary models where a research-grade anti-α4β7 integrin antibody is administered in vivo to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) characterization are predominantly syngeneic mouse models and, to a lesser extent, humanized mouse models.

Syngeneic Models:

  • Syngeneic models involve implanting murine tumor cells into genetically identical immunocompetent mice. This setup allows for detailed in vivo assessment of immuno-oncology agents, including immune-modulating antibodies such as anti-α4β7, since the host's immune system is fully functional and mouse-specific immune interactions are preserved.
  • These models are optimal for:
    • Tumor growth inhibition studies: allowing the evaluation of anti-tumor efficacy of immune-targeting antibodies.
    • Tumor-infiltrating lymphocyte analysis: since the immune responses are robust, researchers can profile how compounds like anti-α4β7 antibodies alter lymphocyte subsets and their function within the tumor microenvironment.
  • Common syngeneic tumor models used in immunotherapy research include strains such as MC38 (colon carcinoma), TC-1 (lung carcinoma), and others, which have been well characterized for baseline TILs and are responsive to various immunomodulators.

Humanized Mouse Models:

  • In some preclinical settings, humanized mouse models—in which mice are engrafted with a human immune system or human PBMCs—are also used, especially when evaluating antibodies that are species-specific (e.g., a human anti-α4β7 mAb).
  • These models are suitable for:
    • Studying antibodies that recognize only the human α4β7 integrin (as often observed with clinical candidates).
    • Characterizing how human TILs respond to tumor challenge and antibody therapy—noting that the tumor and immune environment are less physiologic than in syngeneic models.

Key Points:

  • Syngeneic models are the standard for murine anti-α4β7 studies due to their intact mouse immune system, which enables robust TIL analysis and tumor inhibition experiments.
  • Humanized models are employed when the anti-α4β7 antibody is specific for the human target or when cross-reactivity is essential; these models use immunodeficient mice engrafted with human PBMCs to create a surrogate for the human immune context.
  • The choice between model types depends on the species specificity of the antibody and the aspect of immune response or tumor biology under study.

Summary Table:

Model TypeImmune System SourceTumor TypeAntibody Target CompatibilityMain Uses
SyngeneicMouse (inbred)Mouse-derivedMouse α4β7Tumor inhibition, murine TIL profiling
Humanized MouseHuman (PBMC/HSC)Human xenograftHuman α4β7Human TIL characterization, human Ab study

Although clinical anti-α4β7 antibodies are mostly human-specific, murine analogs or surrogate antibodies are generated for use in syngeneic models, allowing the study of TILs and tumor inhibition in an immune-competent setting relevant to the native murine system.

Researchers primarily use the Vedolizumab biosimilar in immune-oncology models to study the mechanisms and management of immune-related adverse events (irAEs)—notably, enterocolitis—resulting from combination checkpoint inhibitor therapy such as anti-CTLA-4, anti-PD-1, and increasingly, anti-LAG-3 antibodies. There is currently no direct evidence that Vedolizumab or its biosimilar is used to directly synergize with these checkpoint inhibitors for anti-tumor effects; rather, it is studied to manage and understand the immune toxicities that emerge when these agents are used together.

Essential context and current research use:

  • Mechanistic studies: Vedolizumab is an anti-integrin α4β7 antibody that selectively blocks lymphocyte trafficking to the gut, mitigating gastrointestinal irAEs (especially colitis) without broadly suppressing systemic anti-tumor immunity.

  • Preclinical research: Biosimilar forms of Vedolizumab (for example, as available from research suppliers) are chosen because they possess the same variable region as the clinical therapeutic but are designed for laboratory use, allowing for controlled immunophenotyping or functional assays. These biosimilars enable:

    • Modeling of immune cell localization and trafficking in animal or in vitro systems.
    • Studies in combination with checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) to observe the immune-modulatory effects on both toxicity and tumor immunity.
  • Checkpoint inhibitor synergy: The intended immune-oncology model often involves dual checkpoint inhibition (e.g., anti-CTLA-4 and anti-PD-1, sometimes extended to anti-LAG-3) to enhance anti-tumor immune activity, which unfortunately also increases the risk for immune toxicities, especially in the gut.

  • Research focus:

    • Vedolizumab or its biosimilar is added to such models not to boost anti-tumor effects, but to selectively modulate the immune environment—preventing or treating colitis while examining any potential impact on overall anti-tumor efficacy.
    • The use of Vedolizumab enables distinction between gut-specific vs systemic immune modulation, helping researchers separate toxicity management from loss of anti-tumor response.
  • Synergy investigations: Recent systematic reviews and trial data highlighted that most improved outcomes from combination checkpoint blockade are additive, not synergistic, suggesting that combining with a gut-specific agent like Vedolizumab can help parse out whether gut toxicity management improves overall therapy tolerability without truly synergizing on tumor response.

Key details:

  • Practical application: Researchers simulate complex clinical scenarios by exposing preclinical models to anti-CTLA-4 or anti-LAG-3 biosimilars (alongside a Vedolizumab biosimilar) to:

    • Quantify immune infiltration, cytokine profiles, and T-cell function in tumor and gut tissues.
    • Assess whether Vedolizumab can preserve anti-tumor immunity while preventing or treating gastrointestinal irAEs.
  • Current limitations: Most reports and preclinical studies focus on colitis mitigation and the preservation of checkpoint inhibitor efficacy, not on immune-oncology synergy in tumor responses when adding Vedolizumab. Mechanistic studies use biosimilars because of accessibility for in vivo and in vitro work not directly tied to patient care.

In summary, the Vedolizumab biosimilar is incorporated into immune-oncology combination models with checkpoint inhibitors to investigate the balance between irAE control and retention of anti-tumor immunity, not to specifically generate or investigate direct synergistic anti-tumor effects.

In a bridging anti-drug antibody (ADA) ELISA for monitoring immune responses to vedolizumab, a vedolizumab biosimilar can be used as either the capture or detection reagent—both forms labeled differently—to specifically detect antibodies the patient’s immune system generates against the therapeutic drug.

How the bridging ADA ELISA works with a vedolizumab biosimilar:

  • Principle: The bridging format employs two versions of the drug (the biosimilar), one attached to the plate (capture), and the other labeled for detection.
  • Patient serum is incubated so that if anti-vedolizumab antibodies (ADAs) are present, each ADA molecule can simultaneously bind both immobilized and labeled vedolizumab, forming a “bridge.”
  • Capture step: For vedolizumab, the biosimilar is often attached to the ELISA plate (via standard protein-protein or biotin-streptavidin interactions). This immobilizes the biosimilar antibody.
  • Detection step: A second batch of the vedolizumab biosimilar, now labeled (e.g., with horseradish peroxidase, biotin, or ruthenium depending on detection system), is added. If ADA is present, it bridges between the immobilized and labeled biosimilar, generating a measurable detection signal (colorimetric for ELISA, chemiluminescent in ECL).
  • Readout: The final signal is proportional to the amount of ADA in the patient serum capable of binding to two molecules of vedolizumab (one capture, one detection).

Use of biosimilar vedolizumab specifically:

  • Biosimilars have virtually identical antigen-binding regions to reference vedolizumab, so they will bind the same ADAs as the original drug.
  • Using a biosimilar as the reagent in the assay ensures assay reagents are available without relying only on originator drug sources—which is important for long-term pharmacovigilance and for sponsors developing biosimilar products.
  • The choice of biosimilar for both capture and detection ensures that the detected ADA is specific to the therapeutic and is not confounded by irrelevant reactivity.

Context and advantages:

  • Drug-tolerant bridging ELISAs can detect ADA even when vedolizumab is present in the serum, depending on the dissociation procedure.
  • Bridging format requires ADAs to be bivalent, which is typical for IgG (the main ADA isotype of concern).
  • Assays using biosimilars as reagents are validated to ensure equivalent sensitivity and specificity for ADAs as assays using the originator, as regulatory guidance requires functional equivalency of biosimilar and reference product in all analytical contexts.

Example description (by analogy):
A similar bridging ELISA for infliximab uses biotinylated infliximab (capture) and ruthenium-labeled infliximab (detection) as the paired reagents, which is directly applicable to how vedolizumab biosimilar could be used.

Summary table:

Reagent roleBiosimilar formFunction in assayKey Point
CapturePlate-bound vedolizumabBinds one arm of ADAAnchors ADA if present in serum
DetectionLabeled vedolizumabBinds other ADA armEnables detection via bridging mechanism

This bridging design is the main immunogenicity screening method for biologics, including vedolizumab and its biosimilars, enabling quantitative monitoring of patient ADA responses during therapy.

References & Citations

1 Berlin C, Berg EL, Briskin MJ, et al. Cell. 74(1):185-195. 1993.
2 Schiffer SG, Day E, Latanision SM, et al. Biochem Biophys Res Commun. 216(1):170-176. 1995.
3 Haanstra KG, Hofman SO, Lopes Estêvão DM, et al. J Immunol. 190(5):1961-1973. 2013.
4 Feagan BG, Rutgeerts P, Sands BE, et al. N Engl J Med. 369(8):699-710. 2013.
5 Poole RM. Drugs. 74(11):1293-1303. 2014.
6 Schneider I, Allner C, Mühl L, et al. Transl Res. 253:8-15. 2023.
7 Soler D, Chapman T, Yang LL, et al. J Pharmacol Exp Ther. 330(3):864-875. 2009.
8 Fedyk ER, Wyant T, Yang LL, et al. Inflamm Bowel Dis. 18(11):2107-2119. 2012.
9 Chaparro M, Garre A, Ricart E, et al. Aliment Pharmacol Ther. 48(8):839-851. 2018.
B
Indirect Elisa Protocol
FA
Flow Cytometry
LCI

Certificate of Analysis

Formats Available

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