Anti-Human VEGFR2 (Ramucirumab) (Clone IMC-1121B)

Anti-Human VEGFR2 (Ramucirumab) (Clone IMC-1121B)

Product No.: LT2700

- -
- -
Product No.LT2700
Clone
IMC-1121B
Target
VEGFR2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Ramucirumab, VEGFR-2, 947687-13-0
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human VEGFR2
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8° C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
WB,
IP,
FA,
FC
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 Ramucirumab. Ramucirumab activity is directed against human vascular endothelial growth factor receptor-2 (VEGFR-2; also known as kinase insert domain-containing receptor, KDR).
Background
Vascular endothelial growth factors (VEGF) and VEGF receptors (VEGFR) play an essential role in angiogenesis1. There are three VEGFRs: VEGFR-1, VEGFR-2, and VEGFR-3. VEGFR- 1 and VEGFR-2 are responsible for angiogenesis, and VEGFR-3 affects lymphogenesis. In the pathogenesis of diseases including diabetes mellitus, rheumatoid arthritis, and cancer, new blood vessel formation is highjacked. Changes at the VEGF/VEGFR-2 axis are particularly potent at allowing VEGF-induced proliferation, migration, and vascular endothelial cell differentiation during tumor angiogenesis. Additionally, VEGFR-2 is upregulated in tumor vascular endothelial cells, and VEGF levels are associated with poor prognosis and resistance to chemotherapy. Consequently, the VEGF/VEGFR axis is a prime anti-cancer target. Blocking VEGF/VEGFR-2 with Ramucirumab inhibits tumor growth in animal models and cancer patients2, 3, 4, and Ramucirumab is approved by the US Food and Drug Administration for treatment of various cancers5.

Ramucirumab blocks all known VEGFs from binding to VEGFR-24, 6. Ramucirumab specifically and potently inhibits VEGFR-2 by binding to the VEGF-binding domain at an epitope located within VEGFR-2 extracellular Ig domain 37. Ramucirumab inhibits VEGF/VEGF-2 interaction 1 , VEGFR-2 phosphorylation7, VEGF-induced VEGFR-2 activation1, VEGF-stimulated cellular migration6 and proliferation1 , and prolongs the survival of leukemia-inoculated mice6.

Ramucirumab (IMC-1121B) was fully humanized from chimeric antibody IMC-11211, which was constructed from a Fab fragment (Hu-1121 Fab) isolated by immunopanning against VEGFR-2 under stringent conditions using a VL-shuffled library and the VH gene segment Hu- 2C6 Fab6, 7. The original library was constructed from spleen cells of mice immunized with a soluble form of VEGFR-28. Ramucirumab was converted into a full length bivalent IgG1 antibody from the Fab fragment 11217.

Ramucirumab, clone IMC-1121B, a non-therapeutic biosimilar antibody for research use only was developed recombinantly and has the same variable regions as the original therapeutic.
Antigen Distribution
VEGFR-2 is widely expressed by vascular endothelial cells, some vascular tumors, carcinomas, malignant melanomas, and lymphomas. Certain leukemia cells express functional VEGFR on the cell surface.
Ligand/Receptor
VEGFA
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology

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.

Use of Research-Grade Ramucirumab Biosimilars as Calibration Standards and Reference Controls in PK Bridging ELISA

Background

Pharmacokinetic (PK) bridging ELISAs are essential for measuring drug concentrations (e.g., Ramucirumab) in serum samples during biosimilar development. These assays allow comparison between a biosimilar candidate and the reference product, ensuring that both exhibit similar exposure and clearance profiles in vivo. Calibration standards and reference controls are critical for assay accuracy and reproducibility.

How Ramucirumab Biosimilars Are Used

Calibration Standards

  • Assay Setup: In a typical PK ELISA, a series of calibration standards with known concentrations of the drug are prepared, usually in the same matrix as the samples (e.g., human serum). For Ramucirumab, these might range from very low (e.g., 0.125 mg/L) to high (e.g., 40 mg/L) concentrations, covering the expected therapeutic range.
  • Reference Material: When a research-grade Ramucirumab biosimilar is used as a reference, it serves as the primary standard for generating the calibration curve. This curve is then used to interpolate the concentration of Ramucirumab in unknown clinical samples.
  • Biosimilar vs. Reference Product: Ideally, a single PK assay is developed to quantify both the biosimilar and the original reference product. The biosimilar can be used as the analytical standard if it is demonstrated to be bioanalytically equivalent to the reference drug by rigorous statistical and validation criteria (e.g., equivalence of dose-response within a predefined confidence interval).
  • Validation: Method validation involves analyzing multiple sets of standards and quality controls (QCs) prepared from both the biosimilar and the reference product, ensuring the assay is precise, accurate, and robust across both materials.

Reference Controls

  • Quality Control Samples: QCs are samples with known concentrations of the drug, used to monitor assay performance. For Ramucirumab, QCs might be spiked at low, middle, and high concentrations (e.g., 0.2, 4, and 8 mg/L).
  • Biosimilar as Control: If the biosimilar and reference product are proved to be analytically equivalent, the biosimilar can also serve as the reference control, ensuring consistent performance across different batches and studies.
  • Assay Precision: Intra- and inter-assay precision (e.g., coefficient of variation <30%) is typically assessed using these controls to ensure reproducibility.

Considerations for Biosimilar-Based Calibration

  • Bioanalytical Equivalence: Before a biosimilar can be used as a standard, it must be shown to have equivalent binding properties and immunoreactivity to the reference product. This is assessed through a comprehensive comparability exercise, including statistical analysis of precision and accuracy data for both products in the assay.
  • Regulatory Compliance: The assay must comply with regulatory guidelines for ligand-binding assays (e.g., FDA, EMA), ensuring it is fit for purpose in demonstrating PK similarity between biosimilar and reference.
  • Minimizing Variability: Using a single standard (either biosimilar or reference) for both calibration and control minimizes assay variability, which is crucial for robust PK bridging studies.

Example Workflow

  1. Method Development: Develop a quantitative ELISA capable of detecting both the biosimilar and reference Ramucirumab in human serum.
  2. Standard Preparation: Prepare calibration standards using either the biosimilar or reference product, covering the relevant concentration range.
  3. QC Preparation: Prepare QCs at multiple concentrations using the same material as the standard.
  4. Validation: Validate the assay by analyzing standards and QCs prepared from both the biosimilar and reference product to confirm analytical equivalence.
  5. Sample Analysis: Use the validated assay to measure Ramucirumab concentrations in clinical serum samples, interpolating results from the calibration curve.
  6. Continuous Monitoring: Include QCs in each assay run to monitor performance over time.

Summary Table: Key Components of PK Bridging ELISA Using Biosimilar Standards

ComponentPurposeMaterial UsedValidation Requirement
Calibration StandardGenerate standard curveBiosimilar or referenceBioanalytical equivalence demonstrated
Quality ControlMonitor assay performanceBiosimilar or referenceConsistent with calibration standard
Clinical SampleMeasure drug concentrationPatient serumInterpolated from standard curve

Conclusion

Research-grade Ramucirumab biosimilars can serve as calibration standards and reference controls in PK bridging ELISAs, provided they are rigorously shown to be bioanalytically equivalent to the reference product. This approach reduces variability, streamlines assay validation, and supports robust demonstration of PK similarity during biosimilar development. The entire process must adhere to regulatory guidelines and include comprehensive validation to ensure accurate and reproducible measurement of Ramucirumab concentrations in serum samples.

The primary in vivo models where research-grade anti-VEGFR2 antibodies are administered to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic murine (mouse) tumor models.

Key details:

  • Syngeneic models: Most published studies use immunocompetent mice implanted with murine tumor cell lines, enabling the study of both anti-tumor effects and immune microenvironment changes. Two well-characterized examples are:

    • MC38 (murine colon adenocarcinoma)
    • EMT6-LM2 (murine breast carcinoma/metastatic model)
  • Anti-VEGFR2 antibody administration in these models results in:

    • Measurable tumor growth inhibition as a single agent or, more potently, in combination with immune checkpoint blockade (e.g., anti-PD-L1).
    • Flow cytometric analysis of tumors for TIL characterization, including frequency and activation states of CD8⁺ T cells and changes in myeloid-derived populations.
    • Investigation of the tumor immune microenvironment, such as effects on myeloid cells, antigen presentation (MHC-II upregulation), and checkpoint molecule expression (PD-L1) on TILs and myeloid cells.
  • Genetic models (VEGFR2 conditional knockout in myeloid compartment) further specify the immune contributions, but the fundamental approach remains the immunocompetent, syngeneic mouse model.

  • Humanized models: There is little evidence that humanized mouse models (NSG or NOG mice engrafted with human immune cells) are typically used for research-grade anti-VEGFR2 antibodies, as these antibodies often target mouse VEGFR2 and not the human ortholog. Most anti-VEGFR2 studies focus on mouse-specific settings to understand mechanistic aspects of tumor immune regulation.

    If humanized tumor and immune cell models are used, they tend to require cross-reactive antibodies or fully human analogues for relevance, which is less common in preclinical mechanistic VEGFR2 antibody research.

Summary Table: Model Systems for In Vivo Anti-VEGFR2 Antibody Studies

Model TypeExample Tumor LinesAntibody ActivityTIL CharacterizationReference Use
Syngeneic (mouse)MC38, EMT6-LM2Yes (research-grade, mouse-specific)Yes (flow cytometry, function)Most common
Humanized mouse(Rarely used)Needs cross-reactivityPossible, but not typicalNot standard

Conclusion:
Syngeneic immunocompetent mouse models (e.g., MC38, EMT6-LM2) are the primary and best-characterized systems for in vivo administration of research-grade anti-VEGFR2 antibodies to study both tumor growth inhibition and TIL phenotype/function. Humanized models are generally not used unless specifically studying cross-reactive or human-specific antibodies.

Use of Ramucirumab Biosimilars in Immune-Oncology Research

Researchers are increasingly interested in combining angiogenesis inhibitors like Ramucirumab with immune checkpoint inhibitors (ICIs) to enhance antitumor immunity and overcome therapy resistance in complex cancer models. Here’s how this approach is being explored:

Mechanism and Rationale

  • Ramucirumab is a monoclonal antibody targeting vascular endothelial growth factor receptor 2 (VEGFR2), inhibiting tumor angiogenesis and potentially normalizing the tumor vasculature, which may improve immune cell infiltration and function within the tumor microenvironment (TME).
  • Checkpoint inhibitors such as anti-CTLA-4, anti-PD-1/PD-L1, or anti-LAG-3 antibodies block inhibitory signals on T cells, reinvigorating the immune response against tumors.
  • Biosimilars of these agents, including Ramucirumab, are used in preclinical and translational research to replicate the effects of originator drugs at reduced cost, enabling broader experimentation with novel combinations.

Research Applications

  • Preclinical Models: Ramucirumab biosimilars allow researchers to test combinations with various checkpoint inhibitors in cell lines, organoids, and animal models. The goal is to identify synergistic effects that may not be evident with monotherapy, such as enhanced tumor regression or prolonged survival.
  • Translational Studies: By using biosimilars, labs can scale up combination screening and mechanistic studies without the high cost of originator biologics. This is crucial for exploring the biological effects of combining angiogenesis inhibition with multiple immune checkpoint pathways (e.g., CTLA-4 + PD-1 + VEGFR2 blockade).
  • Clinical Correlation: Findings from preclinical models can inform clinical trial design. For example, a randomized phase II trial showed that combining Ramucirumab (VEGFR2 inhibitor) with pembrolizumab (PD-1 inhibitor) improved overall survival in patients with advanced NSCLC who had progressed on prior immunotherapy, suggesting potential clinical synergy.

Advantages of Using Biosimilars

  • Cost-Effectiveness: Biosimilars reduce the financial barrier to large-scale combination studies, enabling more labs to participate in immune-oncology research.
  • Regulatory Compliance: High-quality biosimilars undergo rigorous testing to ensure structural and functional equivalence to the reference drug, maintaining experimental validity.
  • Flexibility: Researchers can explore novel combinations, such as Ramucirumab with anti-CTLA-4 or anti-LAG-3 biosimilars, to mechanistically dissect interactions between angiogenesis and multiple immune checkpoint pathways.

Challenges and Considerations

  • Toxicity: Combining multiple immunomodulatory agents can increase the risk of severe adverse events, a finding observed in clinical trials of checkpoint inhibitor combinations.
  • Mechanistic Complexity: The biological interplay between angiogenesis inhibition and immune checkpoint blockade is not fully understood. Preclinical models using biosimilars help unravel these mechanisms.
  • Clinical Translation: Not all combinations that show synergy in vitro or in animal models will be effective or safe in patients. Careful preclinical validation is essential before advancing to clinical trials.

Example Approach in the Lab

1. **Model Selection**: Choose an immunocompetent mouse model of a solid tumor.2. **Treatment Groups**: Administer Ramucirumab biosimilar alone, checkpoint inhibitor biosimilar(s) alone, and their combination.3. **Outcome Measures**: Assess tumor growth, immune cell infiltration (e.g., by flow cytometry or immunohistochemistry), and survival.4. **Mechanistic Studies**: Evaluate changes in the TME, such as vascular normalization, T-cell activation, and cytokine profiles.5. **Data Analysis**: Compare outcomes across groups to identify synergistic, additive, or antagonistic effects.

Conclusion

Researchers use Ramucirumab biosimilars—often in combination with biosimilars of checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3—to systematically study potential synergistic effects in complex immune-oncology models. These studies aim to uncover mechanisms of action, optimize combination strategies, and identify candidates for clinical testing, all while leveraging the cost and accessibility advantages of biosimilars. However, the translation of these findings to the clinic requires careful consideration of efficacy and safety, as not all preclinical synergies will prove beneficial in patients.

A Ramucirumab biosimilar is used as a reagent in a bridging ADA ELISA (anti-drug antibody enzyme-linked immunosorbent assay) to detect whether a patient has developed antibodies against the therapeutic drug, signaling an immune response. In this assay format, the biosimilar serves as either the capture or detection reagent because it is structurally and functionally highly similar to the original Ramucirumab, ensuring recognition by any ADAs that have developed against the therapeutic.

In a typical bridging ELISA for ADA detection:

  • The assay plate is coated with the Ramucirumab biosimilar (capture reagent).
  • Patient serum is added; if ADAs against Ramucirumab are present, they will bind to the capture reagent.
  • A labeled (biotinylated or HRP-conjugated) Ramucirumab biosimilar (detection reagent) is then added, which binds to the other arm of the ADA, forming a “bridge” between the two drug molecules.
  • The signal generated by the label is proportional to the amount of ADA present.

This design relies on the ability of ADAs to bind simultaneously to two molecules of Ramucirumab—typically targeting the same or overlapping epitopes on both the biosimilar and the originator drug. Using a biosimilar as the reagent is valid so long as it retains the same immunoreactive epitopes as the original drug, thus allowing the assay to monitor the immune response against either the biosimilar or the reference Ramucirumab.

Bridging ELISA formats are widely used for other therapeutic monoclonal antibodies and biosimilars; this approach is considered robust for ADA detection in clinical monitoring. The presence and amount of ADAs detected can impact drug efficacy and patient safety, so sensitive ADA assays are required during biosimilar development and post-marketing surveillance.

Key technical points:

  • Ramucirumab biosimilar is interchangeable with the originator for reagent use.
  • Bridging ELISA requires two drug molecules for ADA “bridging.”
  • This method detects ADAs specific to Ramucirumab, regardless of minor biosimilar variations, as long as epitope similarity is maintained.

There is no evidence in the search results suggesting increased immunogenicity risk when a biosimilar is used as the reagent. Response characteristics, detection limits, and cross-reactivity must still be validated for the particular ELISA setup.

References & Citations

1. Spratlin J. Curr Oncol Rep. 13(2):97-102. 2011.
2. Posey JA, Ng TC, Yang B, et al. Clin Cancer Res. 9(4):1323-1332. 2003.
3. Spratlin JL, Cohen RB, Eadens M, et al. J Clin Oncol. 28(5):780-787. 2010.
4. Wilke H, Muro K, Van Cutsem E, et al. Lancet Oncol. 15(11):1224-1235. 2014.
5. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/125477s034lbl.pdf
6. Zhu Z, Hattori K, Zhang H, et al. Leukemia. 17(3):604-611. 2003.
7. Lu D, Shen J, Vil MD, et al. J Biol Chem. 278(44):43496-43507. 2003.
8. Zhu Z, Rockwell P, Lu D, et al. Cancer Res. 58(15):3209-3214. 1998.
Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation 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.