Anti-Human VEGF-A (Ranibizumab) – Fc Muted™

Anti-Human VEGF-A (Ranibizumab) – Fc Muted™

Product No.: V203

- -
- -
Product No.V203
Clone
RG-3645
Target
VEGF-A
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Vascular permeability factor (VPF), L-VEGF, N-VEGF, VEGFA, VEGF
Applications
B
,
ELISA
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Derived from murine monoclonal anti-VEGF antibody, original 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,
WB
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 asthe therapeutic antibody Ranibizumab. RG-3645 (Ranibizumab) activity is directed againsthuman and cynomolgus monkey VEGF-A. The antibody targets all VEGF-A isoforms. This product is research use only.
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.

Ranibizumab is a high-affinity recombinant, humanized antibody that neutralizes all isoforms of VEGF-A8,9. Ranibizumab was derived from a murine monoclonal anti-VEGF antibody10and consists of a nonbinding human sequence and a high affinity murine binding epitope11. When injected intravitreously, ranibizumab can penetrate the internal limiting membrane and access the subretinal space. Ranibizumab reduces the proliferation of endothelial cells as well as vascular permeability and formation of new blood vessels by preventing VEGF-A from interacting with its receptors on the endothelial cell surface9. Ranibizumab is also able to inhibit human umbilical vein endothelial cell proliferation in vitro. Ranibizumab does not bind to mouse VEGF-A in vitro by Western blot, nor does it have a measurable biological effect after intraperitoneal or intravitreal injection in oxygen-induced-retinopathy mouse models12. Intravitreal injections of ranibizumab in cynomolgus monkeys reduces choroidal neovascularization11.

Ranibizumab is also known as RhuFab V2 in the literature10.
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, tissue repair, as well as during disease- related neovascularization.
Ligand/Receptor
Binds to: FLT1/VEGFR1 and KDR/VEGFR2 receptors, heparan sulfate and heparin, NRP1/neuropilin-1 receptor
UniProt.org
Research Area
Apoptosis
.
Biosimilars
.
Inflammatory Disease
.
Angiogenesis
.
Neovascularization
.
Ocular Disorders

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 Ranibizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately quantify drug concentrations in serum by serving as the basis for standard curves and quality controls. This facilitates consistent and reliable measurement of both biosimilar and reference (innovator) Ranibizumab in PK studies.

Essential context and supporting details:

  • Calibration Standards Role: In a PK bridging ELISA, quantitative measurement of Ranibizumab (whether biosimilar or reference product) in serum requires a standard curve constructed from known concentrations of a Ranibizumab biosimilar (research-grade standard). These standards are typically run in parallel with test samples in each assay to ensure accurate interpolation of unknown concentrations.

  • Reference Controls: Quality control (QC) samples are prepared using the same or similar biosimilar material and spiked into the relevant biological matrix (e.g., blank human serum) at multiple concentrations. These serve to monitor assay accuracy and precision across its measurement range.

  • Test System Equivalence: Regulatory and scientific best practices recommend that a single PK assay, using a single analytical standard (commonly the biosimilar), quantifies both the biosimilar and reference product to minimize variability and enable direct bioanalytical comparability. During assay validation, both the biosimilar and reference (innovator) Ranibizumab are analyzed against the biosimilar-based standard curve, and analytical comparability is formally established through statistical analysis of accuracy and precision.

  • Kit Practices: Commercial ELISA kits for Ranibizumab often use lyophilized biosimilar or recombinant Ranibizumab as their standards, with concentration ranges suitable for expected clinical serum levels. These standards may be validated against international reference preparations (e.g., NIBSC/WHO) or innovator drug lots to ensure comparability and traceability.

  • Rationale: Using biosimilars as standards/reference controls is necessary because they represent the test product in clinical studies, may be more consistently available than innovator batches, and ensure the assay is directly relevant for biosimilar PK studies.

Summary Table: Roles of Ranibizumab Biosimilars in PK Bridging ELISA

UseDescription
Calibration StandardSets the standard curve to measure unknown serum concentrations
Reference ControlSpiked into blank serum at known concentrations for QC, monitoring assay validity
Analytical EquivalenceBoth biosimilar and innovator compared using same standard and method

Additional notes:

  • Calibration and QC must be performed as per regulatory guidelines (FDA, EMA, ICH) for accuracy, precision, and robustness.
  • When conducting multi-region studies (e.g., US and EU reference products), the single-assay/single-standard approach is recommended to avoid bias and confusion.

In summary: Research-grade Ranibizumab biosimilars provide the quantitative anchor and control framework in PK bridging ELISAs, allowing sensitive, specific, and standardized measurement of drug concentration in serum samples, and supporting reliable PK comparison between biosimilar and innovator products.

The primary in vivo models for studying the effects of research-grade anti-VEGF-A antibodies on tumor growth and the characterization of tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models and, more recently, humanized mouse models.

Key points about these models:

  • Syngeneic mouse tumor models are the most widely used for this purpose. In these models, murine tumor cell lines (e.g., CT26, RENCA, B16F10) are implanted into immunocompetent mice of the same genetic background, allowing for the assessment of both tumor growth inhibition and immune microenvironment changes, including TIL profiling. These models have well-characterized immune infiltrates and are amenable to immunotherapy studies, including anti-VEGF-A antibody administration.

    • For example, treatment with anti-VEGF-A antibodies such as mouse-specific monoclonals (e.g., G6-31) in models like Apc^min, CT26, B16F10, and RENCA has been shown to inhibit tumor growth and modify the tumor microenvironment, making it possible to evaluate TIL composition.
  • Humanized mouse models—in which immunodeficient mice are engrafted with human hematopoietic cells—are increasingly used when the objective is to study human-specific immune-TIL interactions with humanized or cross-reactive anti-VEGF-A antibodies. However, these are more complex, more expensive, and less commonly used than syngeneic models due to technical difficulties and the need for human-specific reagents. They are favored when precise recapitulation of human immune interactions or the use of human tumor xenografts is required.

  • Xenograft models (implantation of human tumor cells in immunodeficient mice) are more suitable for assessing tumor growth inhibition but not for evaluating TILs, as these mice lack a competent immune system.

Notable syngeneic models for anti-VEGF-A + TIL studies include:

  • CT26 (colon carcinoma)
  • RENCA (renal cell carcinoma)
  • B16F10 (melanoma)
  • Apc^min mice (intestinal adenoma model; recapitulates angiogenesis and allows antibody efficacy evaluation).

These models allow:

  • Administration of research-grade anti-VEGF-A antibody.
  • Monitoring of tumor growth inhibition.
  • Characterization of TILs via flow cytometry, immunohistochemistry, or transcriptomic analysis.

Summary Table:

Model TypeTumor Growth?TIL Analysis?Typical Antibody UsedNotes
Syngeneic mouse modelsYesYesMouse-specific anti-VEGF-AWidely used for immunotherapy studies
Humanized mouse modelsYesYesHuman/cross-reactive antibodyModels human immune responses
Human tumor xenograft modelsYesNoHuman-specific antibodyNot suited for TIL analysis

In practice, the majority of published in vivo anti-VEGF-A and TIL studies are performed in syngeneic mouse tumor models using mouse-specific antibodies. Humanized models are used for translational studies but are less standard for basic TIL characterization.

References:

  • For broad tumor-immune profiling in syngeneic systems:
  • For tumor inhibition and microenvironment modulation by anti-VEGF-A:
  • For experimental details using anti-VEGF-A antibodies in vivo:

There is no evidence in current literature that ranibizumab biosimilars are used in conjunction with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to study synergistic effects in immune-oncology models. Ranibizumab and its biosimilars are anti-VEGF agents primarily used for treating ophthalmologic diseases like neovascular age-related macular degeneration, not as cancer immunotherapies.

Researchers investigating synergistic effects of multiple checkpoint inhibitors generally focus on combinations like anti-CTLA-4 (e.g., ipilimumab) with anti-PD-1 (e.g., nivolumab) or emerging targets such as anti-LAG-3. These studies use preclinical tumor models and clinical trials to assess additive or synergistic immune responses by blocking different inhibitory pathways in T cell activation and tumor microenvironment. The rationale is that these checkpoints regulate immunity at different sites; for example, anti-CTLA-4 enhances T cell priming in lymph nodes, while anti-PD-1 and anti-LAG-3 act predominantly within the tumor.

Combination studies typically involve:

  • Immunocompetent murine tumor models for mechanistic insights and efficacy evaluation, sometimes using humanized mice for translational relevance.
  • Assessment of tumor growth, immune cell infiltration, cytokine production, and survival outcomes to delineate synergy or additive effects.
  • Phase I–III clinical trials validating safety, dosing, and antitumor efficacy in humans.

There is no evidence from current research that anti-VEGF biosimilars like ranibizumab have been systematically combined with immune checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, or their biosimilars) in complex immune-oncology models. Reports of anti-VEGF/immune checkpoint inhibitor combinations exist for other anti-VEGF agents (notably bevacizumab) in oncology, but not for ranibizumab or its biosimilars.

In summary:

  • Ranibizumab biosimilars are not currently investigated with checkpoint inhibitors in immune-oncology models.
  • Synergy studies with checkpoint inhibitors focus on anti-CTLA-4, anti-PD-1, and anti-LAG-3, usually in combination with each other and in cancer settings, not with anti-VEGF agents like ranibizumab.

Any reference to ranibizumab in immune-oncology is likely a confusion with other anti-VEGF agents used in cancer research.

A Ranibizumab biosimilar can be used as the capture or detection reagent in a bridging ADA ELISA assay to monitor a patient’s immune response against Ranibizumab by specifically detecting anti-drug antibodies (ADAs) that develop after exposure to the therapeutic. This approach relies on the biosimilar's structural equivalence to originator Ranibizumab, ensuring immune recognition and assay validity.

Context and Supporting Details:

  • In a typical bridging ELISA for ADA detection, the therapeutic drug (or its biosimilar) is immobilized on the ELISA plate, serving as the capture reagent. This allows any ADAs present in the patient’s serum to bind to the drug on the plate.
  • After washing to remove unbound material, a solution containing the biosimilar drug, chemically labeled (often with biotin, HRP, or similar tags), is added as the detection reagent. ADAs—due to their bivalency—can simultaneously bind both the immobilized (capture) and labeled (detection) Ranibizumab molecules, forming a "bridge."
  • The presence of bridged complexes is detected via the label on the biosimilar, providing a quantitative readout of ADA levels. Generally, a colorimetric or fluorescent substrate is used to visualize and quantify the response.

Why Use a Biosimilar?

  • Structural identity: Ranibizumab biosimilars, such as SB11, are engineered to closely match originator Ranibizumab in their amino acid sequence and tertiary structure, making them immunologically indistinguishable for ADA detection. This ensures that all patient-generated ADAs against Ranibizumab—whether exposed to the original drug or its biosimilar—can be detected using biosimilar reagent in the ELISA.
  • Practicality and robustness: If the biosimilar is more readily available, stable, or cost-effective, it can replace the originator drug for both capture and detection steps, provided the ADA epitopes are preserved.

Assay Principle Example:

| Step                     | Reagent Used                 | Function                     ||--------------------------|------------------------------|------------------------------|| Plate coating            | Ranibizumab biosimilar (capture) | Immobilizes drug for ADA binding || Sample incubation        | Patient serum                | ADAs (if present) bind drug   || Detection                | Labeled Ranibizumab biosimilar (detection) | Bridges with ADA and captured drug || Signal readout           | Substrate (HRP/TMB, etc)     | Quantifies ADA-bridged complexes |

Key Insights:

  • Using a biosimilar as both capture and detection reagent ensures that the assay measures immunogenicity against both originator and biosimilar Ranibizumab.
  • Assay specificity relies on the biosimilar's structural match and must be validated to confirm it detects clinically relevant ADAs.
  • Studies show low immunogenicity rates for both Ranibizumab and its biosimilar, and these rates do not appear to affect safety or efficacy outcomes in patients.

This approach is standard for therapeutic antibody immunogenicity testing and facilitates reliable monitoring of ADA development in patients throughout treatment.

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. Blick SK, Keating GM, Wagstaff AJ. Drugs. 67(8):1199-1206. 2007.
10. Eter N, Krohne TU, Holz FG. BioDrugs. 20(3):167-179. 2006.
11. Krzystolik MG, Afshari MA, Adamis AP, et al. Arch Ophthalmol. 120(3):338-346. 2002.
12. Ichiyama Y, Matsumoto R, Obata S, et al. PLoS One. 17(12):e0278951. 2022.
B
Indirect Elisa Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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