Anti-Human VEGF-Bevacizumab [Clone A4.6.1]

Anti-Human VEGF-Bevacizumab [Clone A4.6.1]

Product No.: LT400

- -
- -
Product No.LT400
Clone
A4.6.1
Target
VEGF
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Vascular Endothelial Growth Factor; VEGF-A; VEGFA; Vascular Permeability Factor; VPF
Isotype
Human IgG1κ
Applications
B
,
ELISA
,
FC
,
IP
,
N
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Recombinant human VEGF.
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 Adalimumab 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.
WB
ELISA
Additional Applications Reported In Literature ?
B
N
IP
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 Bevacizumab. Bevacizumab recognizes both native and reduced human VEGF (isoform 165). This product is for research use only.
Background
Bevacizumab is a monoclonal antibody that specifically recognizes vascular endothelial growth factor (VEGF). VEGF is a growth factor that participates in angiogenesis, vasculogenesis, and endothelial cell growth. It facilitates endothelial cell proliferation, cell migration, and the permeabilization of blood vessels. In addition, VEGF inhibits apoptosis. Bevacizumab neutralizes the biological activity of VEGF by preventing the interaction of VEGF with its receptors on the surface of endothelial cells, resulting in the regression of tumor vascularization, normalization of remaining tumor vasculature, and inhibition of the formation of new tumor vasculature, thus inhibiting tumor growth.1 Anti-Human VEGF (Bevacizumab) utilizes the same variable regions from the therapeutic antibody Bevacizumab making it ideal for research projects.
Antigen Distribution
VEGF is widely expressed in the thyroid, prostate, and various other tissues.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars

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.

In the context of pharmacokinetic (PK) bridging ELISA for measuring drug concentration in serum samples, research-grade VEGF-Bevacizumab biosimilars are used as calibration standards or reference controls through a highly specific and sensitive analytical process. Here's how they are utilized:

Role of Biosimilars in ELISA

  • Binding Assays: Bevacizumab biosimilars are evaluated for their binding potency to VEGF using ELISA. This involves coating VEGF onto the ELISA plate, allowing the biosimilar to bind, and then detecting the bound antibody using additional antibodies labeled with enzymes like HRP (horseradish peroxidase).
  • Calibration Standards: The biosimilars with known concentrations are used as calibration standards. These standards allow the creation of a calibration curve that relates the optical density (OD) of the ELISA wells to the concentration of the biosimilar in the sample. This curve is essential for quantifying the concentration of the drug in serum samples.

ELISA Methodology for PK Analysis

  1. Sample Preparation: Serum samples containing bevacizumab (either the original drug or its biosimilars) are prepared and diluted appropriately for the assay.
  2. Capture Antibody: An anti-idiotypic monoclonal capture antibody specific to the complementarity-determining region (CDR) of bevacizumab is used to capture the drug in the serum sample. This ensures that only bevacizumab and its biosimilars are detected.
  3. Detection: A second monoclonal antibody labeled with HRP is used to detect the captured bevacizumab. This step is crucial for the colorimetric detection of the bound drug.
  4. Quantification: By comparing the OD from the sample wells to the calibration curve generated from the calibration standards, the concentration of bevacizumab in the serum sample can be determined.

Importance of Biosimilars as Reference Controls

  • Inter-assay Variability: Biosimilars used as reference controls help in minimizing inter-assay variability by providing consistent results across different batches of ELISA kits.
  • Validation of Assays: They are crucial for validating the specificity and sensitivity of the ELISA assays, ensuring that the assays accurately measure the drug concentration without significant interference from other serum components.

In summary, research-grade VEGF-Bevacizumab biosimilars play a critical role in PK bridging ELISA by serving as calibration standards and reference controls, enabling the precise measurement of drug concentrations in serum samples. This process ensures the accuracy and reliability of the pharmacokinetic data obtained from these assays.

The primary in vivo models used to administer a research-grade anti-VEGF antibody and study tumor growth inhibition with analysis of tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, for human-specific antibodies, humanized or transgenic mouse models. Syngeneic models are favored for detailed TIL characterization due to their intact murine immune systems, while humanized models enable study of human immune responses when investigating human-specific therapeutics.

Key contexts and supporting details:

  • Syngeneic Models:

    • These use mouse tumor cell lines implanted into immune-competent mice of the same genetic background, allowing for full characterization of TILs.
    • Anti-VEGF antibodies for mouse targets or chimeric/human-mouse antibodies can be administered to evaluate tumor growth inhibition and immune cell profiles in the tumor microenvironment.
    • Quantification of TILs is routine in these models, and diverse tumor types like MC38, TC-1, and others are well characterized for baseline TIL populations and therapeutic responses.
    • Studies using syngeneic models after anti-VEGF treatment typically assess TIL subsets (e.g., CD8+ T cells, regulatory T cells) to correlate immune changes with tumor growth inhibition.
  • Humanized and Transgenic Mouse Models:

    • Humanized models are required for in vivo evaluation of human-specific anti-VEGF antibodies, as standard syngeneic models do not permit direct interaction with the human immune system.
    • Transgenic mice engineered to express human antibody targets or immune cells, or mice bearing tumors that overexpress human antigens, allow the testing of therapeutics cross-reactive with human proteins.
    • These models enable both measurement of antitumor effects and analysis of human immune cell infiltration into tumors after antibody treatment.
    • Chimeric antibody agents may be used when full human antibody compatibility is needed in a murine system.

Alternative Model: Xenograft

  • Xenograft models (human tumor cells implanted into immunodeficient mice) are frequently used for testing anti-VEGF efficacy in tumor growth suppression. However, these models lack a functional immune system and are thus generally unsuitable for detailed TIL characterization since TILs are either absent or limited to human-derived cells introduced with the tumor.

Summary Table: Model Types and Uses for Anti-VEGF Antibody Studies

Model TypeImmune SystemTIL CharacterizationUse with Mouse Anti-VEGFUse with Human Anti-VEGF
SyngeneicMurine (intact)YesYesPossible (with chimeric/transgenic mice)
Humanized/TransgenicHumanized murineYes (human cells)NoYes
XenograftMurine (deficient)Limited/NoYes (for tumor growth only)Yes (for tumor growth only)
  • Syngeneic mouse models are the gold standard for TIL analysis with research-grade anti-VEGF antibodies targeting murine antigens or compatible therapeutics.
  • Humanized or transgenic models are essential for evaluating human-specific antibodies and human TIL dynamics.
  • Xenograft models do not support robust immune profiling due to their immunodeficiency.

For robust immunological studies involving TILs and tumor growth inhibition with anti-VEGF antibodies, syngeneic or appropriately engineered murine models are predominantly used, tailored by the species specificity of the antibody and the therapeutic hypothesis.

Researchers use the VEGF-Bevacizumab biosimilar in combination with immune checkpoint inhibitors—such as anti-CTLA-4, anti-PD-1, and potentially anti-LAG-3 antibodies—to study synergistic anti-tumor effects in advanced preclinical and clinical immune-oncology models. These combinations are designed to exploit the complementary mechanisms of angiogenesis inhibition (by blocking VEGF) and immune modulation (by checkpoint blockade), with the goal of enhancing durable anti-tumor immune responses beyond what either approach achieves alone.

Synergistic Mechanisms Studied

  • Bevacizumab (VEGF Inhibitor):
    • Neutralizes VEGF-A, reducing tumor angiogenesis, disrupting vascular formation, and modulating the tumor microenvironment to lower immunosuppression.
    • Normalizes tumor vasculature, improving infiltration and function of immune cells within tumors.
  • Checkpoint Inhibitors (e.g., anti-CTLA-4, anti-PD-1):
    • Relieve immune suppression by blocking inhibitory signaling on T cells, enhancing anti-tumor immune activity.

Combining these therapies leverages both altered vascular context and direct T-cell activation:

  • Enhanced Immune Cell Infiltration: Studies show co-administration leads to greater infiltration of CD8+ T cells and macrophages into tumors compared to checkpoint inhibitor monotherapy.
  • Tumor Vasculature Changes: Combined therapy induces changes in vascular endothelium (i.e., expression of E-selectin, formation of high endothelial venule-like structures), facilitating lymphocyte trafficking into tumors.
  • Circulating Memory T-Cell Expansion: Increases in memory CD4+ and CD8+ T cells are observed in peripheral blood following combined administration compared to checkpoint inhibitors alone.

Representative Experimental Approaches

  • Clinical Trials:

    • Phase I and II Trials: Ipilimumab (anti-CTLA-4) or Nivolumab (anti-PD-1) combined with Bevacizumab (biosimilar or reference) in cancers such as melanoma or ovarian cancer; endpoints include safety, efficacy, immune cell infiltration, and changes to tumor microenvironment.
    • Patients are treated with established dosing regimens of each drug concurrently and undergo serial biopsies and blood sampling to evaluate immune and vascular changes.
  • Preclinical Models:

    • Syngeneic mouse tumor models or humanized mouse systems often receive anti-VEGF-A (or biosimilar) alongside checkpoint inhibitors.
    • Readouts include tumor growth, immune cell phenotyping (flow cytometry/histology), and cytokine/chemokine profiling; these studies test for additive or synergistic anti-tumor activity and dissect underlying mechanisms.

Extension to Other Checkpoints (anti-LAG-3, etc.)

While published clinical and translational data primarily describe combinations of Bevacizumab with anti-CTLA-4 or anti-PD-1/PD-L1 antibodies, the same research framework is being extended to new checkpoint inhibitors (e.g., anti-LAG-3 biosimilars). Researchers hypothesize similar immunomodulatory and synergistic benefits when combining VEGF pathway inhibition with agents targeting novel immune checkpoints, although explicit published studies with anti-LAG-3 plus bevacizumab biosimilar are limited as of now.

Biosimilars in Combination

  • Bevacizumab biosimilars retain the relevant VEGF-blocking functionality and have been used interchangeably with the reference product in combination regimens, providing cost-effective and accessible alternatives for large-scale or global research and clinical trials.

In summary:Researchers study the synergy between Bevacizumab biosimilars and checkpoint inhibitors by combining their complementary effects—disrupting tumor angiogenesis and reversing immune suppression—then measuring enhanced immune cell responses and improved anti-tumor activity in preclinical and clinical models, generating early evidence for superior outcomes over monotherapy.

In immunogenicity testing for anti-drug antibodies (ADAs) against bevacizumab biosimilars, the biosimilar (VEGF-bevacizumab) is typically used as both the capture and detection reagent in a bridging ELISA format to assess the patient's immune response to the therapeutic antibody.

Context and details:

  • In a bridging ELISA for ADA detection, patient serum is incubated with both labeled and unlabeled forms of the therapeutic antibody (here, the bevacizumab biosimilar). ADA present in the patient sample "bridges" between these two forms, allowing complex formation and subsequent detection.
  • Usually, the bevacizumab biosimilar is biotinylated (to serve as the capture reagent, typically bound to a streptavidin-coated plate) and ruthenylated (or otherwise labeled, to serve as the detection reagent) in assay platforms such as Meso Scale Discovery’s ECL system.
  • If the patient has developed ADAs, these antibodies bind simultaneously to both the capture and detection forms of the drug, forming a "bridge" that can be quantified.

Application to VEGF-bevacizumab and ADA testing:

  • As bevacizumab (and biosimilars) target VEGF-A, there is a risk that circulating soluble VEGF in human serum could bind to the therapeutic antibody reagents in the assay, leading to interference or false positives. To prevent this, specific additives are incorporated to block or neutralize soluble VEGF during the assay incubation steps.
  • The process generally involves:
    • Acid dissociation: Patient serum is acid-treated to dissociate any immune complexes present in vivo (e.g., ADA-drug complexes).
    • Neutralization and incubation: Samples are neutralized, then incubated with biotinylated and labeled versions of the biosimilar.
    • Complex capture and detection: After incubation, the formed ADA-drug-bridged complexes are captured on the plate and detected through the label, yielding a signal proportional to ADA concentration.

Key technical considerations:

  • The use of both biotinylated and labeled (e.g., ruthenium-tagged) forms of the biosimilar drug ensures the assay specifically detects bridging by human ADA, since only antibodies capable of binding two drug molecules simultaneously generate a signal.
  • To ensure clinical relevance and avoid false signals due to VEGF interference, validated methods incorporate steps/additives to neutralize or block VEGF, which has been demonstrated as essential for robust ADA measurement in bevacizumab biosimilar programs.

Summary Table: Use of Biosimilar as Reagent in Bridging ADA ELISA

Assay StepReagentPurpose
CaptureBiotinylated VEGF-bevacizumab biosimilarImmobilizes complexes on streptavidin plate
DetectionLabeled VEGF-bevacizumab biosimilarEnables signal detection via bridging
BlockingAdditive to block soluble VEGFPrevents interference/false positives
Acid Treatment--Dissociates in vivo ADA-drug complexes

References:

  • Detailed assay methodology, rationale for biosimilar use, and interference mitigation are described in Eurofins documentation.

Alternative approaches:

  • While bridging ELISA is standard, some labs may use similar principles with different detection systems (e.g., ECL, radioimmunoassay) for comparable ADA monitoring.

Conclusion:
The VEGF-bevacizumab biosimilar functions as both capture and detection reagent in bridging ELISA for ADA monitoring, with critical technical adaptations to remove interference by soluble VEGF, ensuring accurate detection of immune responses to the therapeutic drug.

References & Citations

1. Pazdur, R. et al. (2018) Clin Cancer Res. 24(18):4365-70.
B
Indirect Elisa Protocol
Flow Cytometry
Immunoprecipitation Protocol
N
General Western Blot Protocol

Certificate of Analysis

Formats Available

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