Anti-Human VEGF-A (Brolucizumab)

Anti-Human VEGF-A (Brolucizumab)

Product No.: V210

- -
- -
Product No.V210
Clone
RTH-258
Target
VEGF-A
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Vascular permeability factor (VPF), Vascular endothelial growth factor (VEGF)
Isotype
Human scFv
Applications
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Human
Host Species
Hamster
Expression Host
CHO Cells
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 ?
ELISA,
FA
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 Brolucizumab. RTH-258 (Brolucizumab) binds and inhibits the three major isoforms of VEGF-A (VEGF110, VEGF121, VEGF165). Brolucizumab also binds cynomolgus monkey VEGF-A.
Background
Vascular endothelial growth factor (VEGF), a potent proangiogenic cytokine1, is the key signal used by oxygen-hungry cells to promote the growth of blood vessels. VEGF binds to specialized receptors on the surfaces of endothelial cells and directs them to build new vessels2. VEGF are crucial regulators of vascular development during embryogenesis (vasculogenesis) and blood-vessel formation in the adult (angiogenesis). Abnormal VEGF function is associated with inflammatory diseases including atherosclerosis and hyperthyroidism3,4,5,6. VEGF-A is a member of the VEGF gene family, and several isoforms can be generated by alternative splicing7. Additionally, VEGF-A is a major mediator of angiogenesis and plays a key role in various ophthalmic conditions, including age-related macular degeneration8.

RTH-258 (Brolucizumab) is a low molecular weight, humanized single-chain variable antibody fragment that inhibits the three major isoforms of VEGF-A9. Brolucizumab prevents the interaction of VEGF-A with its receptors VEGFR-1 and VEGFR-2, thereby suppressing endothelial cell proliferation, neovascularization, and vascular permeability.

Brolucizumab was developed for the treatment of wet age-related macular degeneration and macular edema9. It is formerly known as ESBA100810.

Antigen Distribution
VEGF-A is a secreted protein produced by diverse cell types, including aortic vascular smooth muscle cells, keratinocytes, macrophages, and many tumor cells. Expression begins during embryogenesis and declines after birth. VEGF-A expression is relatively low in most adult organs, except for the brain choroid plexus, lung alveoli, kidney glomeruli, and heart vascular beds. VEGF-A is also up-regulated during the development of the endocrine corpus luteum in pregnancy, wound healing, and tissue repair as well as during disease related neovascularization.
Ligand/Receptor
Binds to FLT1/VEGFR1, KDR/VEGFR2, DEAR/FBXW7-AS1and NRP1/neuropilin-1 receptors, heparan sulfate and heparin
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cardiovascular Disease
.
Inflammatory Disease
.
Opthamology
.
Angiogenesis
.
Pro-Inflammatory Cytokines

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 Brolucizumab biosimilars are used as the calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the quantified analyte for the standard curve, against which unknown drug concentrations in serum samples are measured.

When measuring Brolucizumab (or its biosimilar) in a PK bridging ELISA:

  • A single PK assay is typically developed and validated to detect and quantify both the biosimilar and reference (innovator) product in serum samples, avoiding analytical variation from parallel assays for each product.
  • Research-grade Brolucizumab biosimilars are used to prepare serial dilutions (standards) in a matrix similar to test samples (e.g., human serum) to generate a standard curve.
  • The concentration-response relationship (standard curve) is established by measuring signal intensity (e.g., absorbance) at each known calibrator concentration, allowing the interpolation of unknown serum concentrations in study samples.
  • Reference controls: Both the biosimilar and reference (innovator) product can be used to spike quality control (QC) samples at predetermined concentrations, which are then measured in the assay as additional analytical controls to confirm assay precision, accuracy, and bioanalytical comparability.

Why biosimilars are suitable as standards:

  • Before using the biosimilar as a calibration standard, a robust comparability assessment is performed: both biosimilar and reference products are independently analyzed to ensure they produce equivalent assay signals at identical concentrations, meeting regulatory criteria for analytical similarity (usually within a predefined confidence interval, e.g., 0.8–1.25 for mean signal response ratios).
  • When equivalence is established, the biosimilar can become the single analytical standard for the assay, streamlining operations and minimizing cross-assay variability.
  • This approach is consistent with industry practice and regulatory expectations for PK similarity studies in biosimilar development.

Practically, the workflow includes:

  • Preparing a serial dilution series of the biosimilar in human serum to cover the assay range.
  • Running QC samples, sometimes spiked with both biosimilar and reference products, to assess performance.
  • Measuring patient or animal serum samples in parallel, then interpolating drug concentrations from the standard curve.

Summary Table: Use of Research-Grade Biosimilar in PK Bridging ELISA

RoleDescription
Calibration StandardSerial dilutions of biosimilar drugs form the standard curve in the assay matrix; drug levels in serum samples are interpolated using this standard curve.
Reference ControlsBiosimilar (for main curve) and sometimes reference product (as QC) are used to check accuracy, precision, and matrix effects in the ELISA.
Suitability ConfirmationPrior validation of assay response to both biosimilar and reference ensures equivalence, allowing a single standard for both products.

This approach harmonizes data comparability, reduces inter-assay variation, and aligns with established regulatory guidelines for biosimilar PK evaluation.

The primary models used for in vivo administration of a research-grade anti-VEGF-A antibody to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are mouse syngeneic models and genetically engineered mouse models (GEMMs), with humanized models becoming increasingly relevant but less commonly used due to technical challenges and cost.

Essential contexts and supporting details:

  • Syngeneic mouse models (e.g., using murine cell lines such as CT26 (colon), B16F10 (melanoma), RENCA (renal), or EMT6 (breast)) allow repeated study of the immune microenvironment after anti-VEGF-A treatment because the host mouse is immunocompetent. These models are standard for characterizing changes in tumor-infiltrating lymphocytes (such as CD8+ T cells, regulatory T cells, and myeloid-derived suppressor cells) post-antibody therapy.

  • Example of model and intervention: Anti-VEGF-A antibodies administered to Apc +/min mice (a GEMM for intestinal adenoma) showed profound tumor growth inhibition and significant reduction in vascular density after antibody treatment. Although this particular study did not explicitly chart TILs, this GEMM approach is directly relevant for mechanistic immune studies using anti-VEGF-A readouts.

  • Profiling TILs: Syngeneic tumor models such as CT26 and RENCA have been carefully characterized for their tumor-infiltrating lymphocyte populations, and these profiles are used to assess immunological responses to in vivo drug treatments (including targeted antibodies and checkpoint inhibitors).

  • Humanized models: Although humanized mouse models—immunodeficient mice engrafted with human hematopoietic cells—are powerful systems for studying human immune interactions, in vivo characterization of TILs after anti-VEGF-A administration is less frequently reported in these systems, due to their expense and technical limitations. However, they are increasingly used in translational studies with human antibodies.

Summary Table: Key In Vivo Models for Anti-VEGF-A Therapeutic Studies

Model TypeHostTumor SourceImmune SystemTIL CharacterizationCommon Usage
SyngeneicMurine (inbred)Mouse cell linesMouse (intact)ExtensiveStandard
GEMMMurine (genetically)Spontaneous (e.g., Apc+/min)Mouse (intact)VariableMechanistic
HumanizedImmunodeficientHuman cell lines/xenograftsHuman (engrafted)IncreasingTranslational
  • Syngeneic models are preferred for immunological effects, including TIL profiling.
  • Genetically engineered models (e.g., Apc+/min with anti-VEGF-A) provide mechanistic validation for tumor growth inhibition.
  • Humanized models bridge preclinical to human studies but are less commonly used for anti-VEGF-A antibody/TIL characterization due to technical barriers.

Alternative interpretations: While some xenograft models use human tumors in immunodeficient mice for anti-VEGF-A pharmacology, these models are unsuitable for comprehensive study of TILs due to lack of functional mouse immunity.

Conclusion:
Syngeneic mouse models are the primary in vivo system for administering research-grade anti-VEGF-A antibodies to study both tumor growth inhibition and to extensively profile resulting TILs, with engineered models and humanized mice representing specialized, complementary approaches.

Researchers have not widely reported the use of Brolucizumab biosimilar in conjunction with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars in complex immune-oncology models, as Brolucizumab and its biosimilars are primarily utilized as VEGF-A inhibitors for ophthalmic indications like neovascular age-related macular degeneration, not directly in cancer immunotherapy.

Essential context:

  • Brolucizumab biosimilars target VEGF-A isoforms to inhibit vascularization and endothelial proliferation. While VEGF inhibition is a recognized strategy in oncology to limit tumor angiogenesis, Brolucizumab's clinical application and published research focus on eye diseases rather than cancer immune-oncology models.
  • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) are used in oncology to promote anti-tumor immunity by blocking inhibitory signals on T cells. These have demonstrated efficacy but can lead to synergistic toxicity and require careful modeling and combination strategies.
  • Synergistic effects in oncology commonly involve combining angiogenesis inhibitors (such as those targeting VEGF/VEGFR) with checkpoint inhibitors. The rationale is that VEGF blockade can modulate the tumor microenvironment to enhance T-cell infiltration and function, thereby improving checkpoint inhibitor efficacy. Most published combinations have used agents like bevacizumab (another VEGF inhibitor), not Brolucizumab.

Support and additional detail:

  • Numerous studies on combination immunotherapy use multiple checkpoint inhibitors (CTLA-4 and PD-1/PD-L1). These studies highlight enhanced efficacy through dual pathway blockade, but Brolucizumab biosimilar is not mentioned in the published combinations.
  • Preclinical models for combination therapy typically involve established immune-oncology agents. If Brolucizumab biosimilar were considered for use, researchers would likely employ it:
    • To suppress angiogenesis and modulate the tumor microenvironment.
    • Alongside checkpoint inhibitors to study changes in immune cell infiltration and anti-tumor activity.
    • Using functional assays such as ELISA, neutralization, and bioanalytical PK and ADA assays, as are described for Brolucizumab in other disease models.

Limitations and speculation (explicit):

  • There is no direct evidence or reported studies for using Brolucizumab biosimilar specifically with checkpoint inhibitors (anti-CTLA-4 or anti-LAG-3) in immune-oncology models in the available literature. Most combination studies in oncology use other anti-VEGF agents, and further research would be needed to establish protocols involving Brolucizumab biosimilar.
  • If adapted for oncology research, protocols would likely extrapolate from similar VEGF blockade combinations, examining immune cell dynamics, tumor growth inhibition, and toxicity profiles.

In summary, Brolucizumab biosimilar is not currently established in published immune-oncology models involving checkpoint inhibitors, but the conceptual framework for combining anti-angiogenic therapies with immune checkpoint inhibition exists and is being explored with other agents.

A Brolucizumab biosimilar can be used as both the capture and detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient’s immune response against the therapeutic drug. In this assay format, the biosimilar is presented in two labeled forms (often biotinylated for capture and HRP- or enzyme-labeled for detection) that together "bridge" any ADA present in the patient’s sample.

Context and supporting details:

  • Bridging ADA ELISA Principle: Bridging ELISA exploits the bivalency of human anti-drug antibodies, allowing them to simultaneously bind two identical or highly similar drug molecules. The assay typically involves immobilizing one form of the drug (e.g., biotinylated Brolucizumab biosimilar) on a streptavidin-coated plate. When patient serum is added, any ADAs present will bind to this immobilized drug. After washing, a labeled form of the same biosimilar (e.g., HRP-conjugated Brolucizumab biosimilar) is added, which will bind the other arm of the ADA, forming a "bridge".

  • Use of Biosimilar as Reagent: The Brolucizumab biosimilar can be used for capture (biotinylated form) and detection (enzyme- or dye-labeled form) because it is structurally and antigenically nearly identical to the reference drug, so it effectively interacts with anti-Brolucizumab antibodies generated in patients.

  • Immunogenicity Monitoring: This format allows detection of ADAs generated in response to Brolucizumab therapy—a critical component of biosimilar development, since immune responses can impact both efficacy and safety. Immunogenicity studies for biosimilars, especially those with higher immunogenic potential (as referenced for Brolucizumab), must incorporate such assays to comprehensively monitor and compare immune responses between biosimilar and originator products.

Typical Protocol Steps:

  • Coat streptavidin microplate with biotinylated Brolucizumab biosimilar (capture reagent)
  • Add patient serum; ADAs, if present, bind the immobilized drug
  • Add enzyme-labeled Brolucizumab biosimilar (detection reagent), which binds the second epitope on ADA
  • Add substrate, develop color, and measure absorbance proportional to ADA concentration

Additional Notes:

  • The method can quantify both total and specific anti-Brolucizumab ADAs, including neutralizing antibodies when paired with confirmatory or competitive inhibition steps.
  • The assay's specificity may be affected by high drug levels, matrix effects, or soluble target (VEGF), so appropriate controls and validation steps are required.

Summary Table: Key Reagents in ADA Bridging ELISA

Reagent / ComponentRole in Bridging ELISA
Biotinylated Brolucizumab biosimilarCapture on plate (binds ADA first site)
Patient serumProvides ADAs to be detected
Enzyme-labeled Brolucizumab biosimilarDetection reagent (binds ADA second site)
SubstrateSignal development

Using a biosimilar in both roles ensures that the assay detects responses against the clinical product and is suitable for both biosimilar and reference monitoring, making it the standard approach for ADA immunogenicity testing in biosimilar development.

References & Citations

1. Fainaru O, Adini I, Benny O, et al. FASEB J. 22(10):3728-3735. 2008.
2. Goodsell DS. Oncologist. 7(6):569-570. 2002.
3. Matsumoto T, Mugishima H. J Atheroscler Thromb. 13(3):130-135. 2006.
4. Shibuya M, Claesson-Welsh L. Exp Cell Res. 312(5):549-560. 2006.
5. Cross MJ, Dixelius J, Matsumoto T, et al. Trends Biochem Sci. 28(9):488-494. 2003.
6. Hicklin DJ, Ellis LM. J Clin Oncol. 23(5):1011-1027. 2005.
7. Holmes DI, Zachary I. Genome Biol. 6(2):209. 2005.
8. Ferrara N, Damico L, Shams N, et al. Retina. 26(8):859-870. 2006.
9 Markham A. Drugs. 79(18):1997-2000. 2019.
10 Holz FG, Dugel PU, Weissgerber G, et al. Ophthalmology. 123(5):1080-1089. 2016.
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

- -
- -
Prod No.
Description
V210
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.