Anti-Human α4β7 integrin (Vedolizumab) – Fc Muted™

Anti-Human α4β7 integrin (Vedolizumab) – Fc Muted™

Product No.: V205

- -
- -
Product No.V205
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
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
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 PK bridging ELISA by acting as the quantifiable analyte that establishes the standard curve and enables measurement of Vedolizumab concentrations in serum samples, supporting comparative PK analyses between biosimilar and reference products.

Context and Usage:

  • In PK bridging ELISA assays for biosimilar characterization, it is considered best practice to use a single analytical standard—often the biosimilar itself—for calibration. Using the biosimilar provides a direct and consistent reference for quantifying concentrations of both the biosimilar and the reference Vedolizumab present in serum samples.

  • The standard curve is prepared by serially diluting the research-grade biosimilar Vedolizumab in human serum or appropriate buffer. These standards cover a concentration range relevant to expected therapeutic levels in clinical samples (e.g., 50–12,800 ng/mL). Measured absorbance (such as OD450/620 nm) at each standard concentration is used to generate the calibration curve, against which unknown serum sample concentrations are interpolated.

  • The assay typically uses a monoclonal anti-Vedolizumab antibody that captures Vedolizumab in both standard and unknown samples. This ensures that biosimilar and reference Vedolizumab are detected equivalently if they exhibit similar analytical properties, which is essential for bioanalytical comparability and bridging studies.

  • Quality control (QC) samples prepared with both biosimilar and reference Vedolizumab are quantified against this biosimilar-generated calibration curve to confirm assay accuracy, precision, and equivalence. Analytical equivalence is statistically verified (e.g., by examining 90% confidence intervals and predefined equivalence margins).

  • For method validation, parameters such as specificity (no cross-reactivity with other antibodies), sensitivity (limit of quantification), intra/inter-assay precision, and recovery rates are assessed using these standards to ensure that the assay reliably reflects Vedolizumab concentrations in clinical matrices.

Summary of Essential Steps:

  • Serial dilution of research-grade Vedolizumab biosimilar to generate standard curve concentrations.
  • Application of common capture and detection reagents in ELISA, ensuring equal reactivity to biosimilar and reference products.
  • Quantitative measurement of unknown serum samples by interpolation from the biosimilar standard curve.
  • Verification of analytical equivalence and assay performance using standards and QC samples, supporting the regulatory requirement for PK bridging between biosimilar and reference Vedolizumab.

This approach ensures systematic and scientifically rigorous measurement of Vedolizumab concentrations in clinical PK studies, enabling direct assessment of pharmacokinetic similarity between biosimilar and reference products.

The primary in vivo models used to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) with research-grade anti-α4β7 integrin antibodies are syngeneic mouse tumor models, with some work also performed in humanized models depending on antibody species specificity and mechanistic aims.

Syngeneic Models:

  • Syngeneic models involve implanting murine tumor cell lines into immunocompetent mice of the same genetic background, allowing for full immunological evaluation, including TILs characterization after antibody treatment.
  • These models are widely used for immune checkpoint inhibitor studies, including integrin-targeted agents, because they provide a controlled system for evaluating both tumor growth and immune cell (including TIL) modulation in response to treatment.
  • Typical syngeneic tumor lines include MC38 (colon), B16 (melanoma), and 4T1 (breast), among others.
  • These models allow for detailed analysis of the impact of anti-α4β7 antibody administration on various TIL populations via flow cytometry or immunohistochemistry.

Humanized Models:

  • Humanized models, which involve engrafting human immune cells (and sometimes tissues) into immunodeficient mice, can be used if a research-grade anti-α4β7 integrin antibody is specific for the human antigen.
  • These models enable evaluation of human immune responses in vivo, though they are more complex and costly than syngeneic models and require antibodies reactive with human proteins.
  • The use of humanized models may be necessary for translational studies of clinically relevant antibodies such as AMG 181 or vedolizumab, which specifically target the human α4β7 integrin.

Key Details:

  • Research-grade murine or cross-reactive antibodies are required for syngeneic studies, while only human-reactive antibodies (and sometimes cross-reactive) can be tested in humanized systems.
  • Syngeneic models are preferable for mechanistic studies of mouse tumor-immune interactions and for initial evaluation of immunotherapy efficacy.
  • Humanized models provide valuable insights for translating findings to human cancer therapy but are less frequently used at the discovery stage due to complexity and cost.

In summary, syngeneic mouse tumor models are the primary system for in vivo testing of anti-α4β7 integrin antibodies regarding tumor growth inhibition and TIL analysis, while humanized models are selectively used for human-specific reagents or translational research.

Researchers investigating synergistic effects of vedolizumab biosimilars combined with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) in immune-oncology models typically design studies to evaluate how these agents interact to modulate immune responses, antitumor activity, and immune-related adverse events. The goal is to leverage differing mechanisms of action to enhance therapeutic efficacy while managing toxicity.

Context and Supporting Details:

  • Vedolizumab Mechanism: Vedolizumab is an IgG1 monoclonal antibody targeting α4β7 integrin, primarily used for gut-selective immunosuppression (notably in inflammatory bowel disease). In immune-oncology, it is also investigated to manage gastrointestinal immune-related adverse events (irAEs), especially those induced by checkpoint inhibitors such as anti-CTLA-4 and anti-PD-1.

  • Combination Rationale:

    • Multiple checkpoint inhibitors (e.g., anti-CTLA-4 and anti-PD-1/PD-L1) are combined in oncology for synergistic antitumor effects because they modulate immune activation at different stages and compartments (lymph nodes vs tumor microenvironment).
    • Combining vedolizumab with checkpoint inhibitors is mainly studied to counteract the adverse effects (notably colitis) caused by the heightened immune activation, enabling safer continuation of potent immunotherapies.
    • Preclinical models and some clinical studies use vedolizumab to limit immune-mediated tissue damage (in the GI tract) triggered by systemic checkpoint inhibition, thereby allowing for higher dosing or combined use of checkpoint agents.
  • Study Design Approaches:

    • Preclinical Models: Mouse models (often humanized) are treated with combinations of checkpoint inhibitors and vedolizumab biosimilars to measure:
      • Tumor regression/response rates
      • Immune cell infiltration into tumors and other tissues
      • Cytokine profiles and other immune activation markers
      • Incidence and severity of immune-mediated tissue toxicity (e.g., colitis)
    • Clinical Trials: In some clinical contexts, patients receiving combined checkpoint therapy (e.g., anti-CTLA-4 + anti-PD-1) and developing colitis are treated with vedolizumab to assess:
      • Efficacy of vedolizumab for irAE resolution
      • Impact on ongoing tumor control
      • Possible immunological synergies or antagonism between GI-specific immunosuppression and systemic antitumor immunity.
  • Checkpoint Biosimilars: While the use of anti-CTLA-4 or anti-LAG-3 biosimilars—as opposed to originator monoclonals—is less detailed in current literature, the experimental logic remains the same: combinations test whether selective blockade or immune modulation can improve the efficacy and safety profile over monotherapy or dual-checkpoint inhibition alone.

  • Key Considerations/Findings:

    • Dual checkpoint inhibition markedly increases immune-related toxicity such as colitis.
    • Vedolizumab provides a gut-selective approach with a more favorable safety profile for colitis than anti-TNFα agents.
    • The primary research focus is not just antitumor synergy, but also the management of checkpoint-induced toxicity to facilitate uninterrupted cancer treatment.

There is limited published evidence on the use of vedolizumab specifically in combination with anti-LAG-3 biosimilars in preclinical or clinical synergy studies, likely due to LAG-3 inhibitors' more recent development and slower adoption in routine combination immune-oncology protocols. However, the same immunological principles apply to new checkpoint pathways.

In summary:Researchers use vedolizumab biosimilars alongside other checkpoint inhibitors principally to study if gut-specific immunosuppression can reduce immune-related toxicity, enabling more potent or prolonged combinatory checkpoint therapy, and to interrogate potential synergistic effects on immune activation and tumor control in complex oncology models.

A Vedolizumab biosimilar can be used as both the capture and detection reagent in a bridging anti-drug antibody (ADA) ELISA to detect and monitor a patient’s immune response—specifically the presence of anti-Vedolizumab antibodies (AVAs)—against the therapeutic drug.

Mechanism and Role in Bridging ELISA:

  • Bridging ELISA is a commonly used immunogenicity assay format for ADA detection. The principle is based on the bivalent nature of antibodies: a patient’s ADA can simultaneously bind two identical molecules of Vedolizumab (or its biosimilar), one immobilized and one labeled, creating a "bridge" across the solid-phase and the detection system.
  • In this context, Vedolizumab biosimilar is used in two forms:
    • Capture Reagent: The plate is coated with Vedolizumab biosimilar (unconjugated or sometimes biotinylated).
    • Detection Reagent: A separately labeled (e.g., peroxidase- or ruthenium-conjugated) Vedolizumab biosimilar is used as the detection reagent in the assay.

How the Assay Works:

  1. Patient Serum is Added: The plate-bound Vedolizumab biosimilar captures any ADAs (anti-Vedolizumab antibodies) present in the patient's serum.
  2. Detection Reagent: After a wash, the labeled Vedolizumab biosimilar is added. If ADA is present, it bridges between the solid-phase Vedolizumab and the labeled Vedolizumab, forming a complex.
  3. Signal Development: Binding of the detection reagent reveals the presence of ADAs through a measurable signal (e.g., color change for peroxidase, chemiluminescence for ruthenium labeling).

Why Use a Biosimilar?

  • Biosimilars are highly similar to the originator product regarding structure and immunogenic epitopes; thus, a biosimilar is functionally equivalent to the originator Vedolizumab for ADA capture and detection.
  • This format ensures specificity for anti-Vedolizumab antibodies and allows monitoring of immunogenicity during biosimilar or reference product therapy.

Advantages and Considerations:

  • Drug Tolerance: Standard ELISAs using the bridging format are often drug-sensitive (i.e., they may not detect ADA if a high amount of drug is present in the sample). Modified, drug-tolerant assays may include an acid dissociation step to release ADA from drug complexes before the ELISA step.
  • Clinical Relevance: Bridging ELISAs are useful for routine monitoring. They can inform on loss of response to therapy, as high ADA titers may correlate with lower drug levels and treatment failure.

Example from Literature:

  • An analogous bridging ADA ECL/ELISA assay for infliximab utilizes biotinylated drug as capture and ruthenium-labeled drug as detector. For Vedolizumab, a similar approach can be applied: use of the biosimilar (instead of the innovator) in both steps, assuming appropriate labeling and validation.

Summary Table: Vedolizumab Biosimilar in Bridging ADA ELISA

FunctionReagentRole
CaptureVedolizumab biosimilar (unlabeled/biotinylated)Binds ADA from patient sample
DetectionVedolizumab biosimilar (labeled, e.g. HRP/ruthenium)Binds to other arm of ADA, enables signal detection

This setup allows the sensitive and specific detection of anti-Vedolizumab antibodies, enabling clinicians and researchers to monitor immunogenicity in patients receiving Vedolizumab or its biosimilars.

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.