Anti-Human VEGFR2 (Ramucirumab) – Fc Muted™

Anti-Human VEGFR2 (Ramucirumab) – Fc Muted™

Product No.: LT2705

- -
- -
Product No.LT2705
Clone
IMC-1121B
Target
VEGFR2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Ramucirumab, VEGFR-2
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
Muted
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.

The primary models used to study anti-VEGFR2 antibody effects on tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models, particularly the MC38 (colon carcinoma) and EMT6 (breast carcinoma) models in immunocompetent mice.

Supporting details:

  • Syngeneic models involve implantation of mouse tumor cell lines into immunocompetent mice of the same genetic background, preserving a functional immune system that allows for the characterization of TILs and immune modulation.
  • MC38 (colon carcinoma) and EMT6 (breast carcinoma/EMT6-LM2 metastatic variant) are among the most widely used models for administering research-grade anti-VEGFR2 antibodies. These models have shown that anti-VEGFR2 monotherapy can inhibit tumor growth and increase intra-tumoral T cell infiltration. Importantly, combining anti-VEGFR2 with anti-PD-L1 antibodies further enhances tumor inhibition and TIL infiltration.
  • Other syngeneic models such as MMTV-PyMT–derived breast tumor and models of murine glioma have also been used to genetically or pharmacologically dissect VEGFR2 function on both endothelial and myeloid cells, allowing detailed analysis of TIL phenotype and function.

Humanized models:

  • There are limited examples in the provided search results of humanized mouse models (where human immune cells are engrafted) being used specifically to study anti-VEGFR2 antibody effects on TILs. Most preclinical research cited focuses on syngeneic mouse models.
  • Humanized models can, in principle, be used for such studies, but suitable reagents (cross-reactive humanized anti-VEGFR2 antibodies) and careful selection of tumor systems and donor immune cells are required to achieve reliable TIL characterization. This remains more technically challenging and less commonly reported than syngeneic systems.

Summary Table: Syngeneic vs. Humanized Models for Anti-VEGFR2/TIL Studies

Model TypeExamplesFeaturesUsage for Anti-VEGFR2/TILs
Syngeneic MurineMC38, EMT6Murine tumor cells in immunocompetent mice; intact immune microenvironmentMost frequently used; direct phenotypic/functional TIL analysis
HumanizedN/A in citationsHuman immune cells in immunodeficient mice; allows human-specific responsesLimited data in current literature; technically complex

Conclusion:
Murine syngeneic models, specifically MC38 and EMT6, represent the gold standard for in vivo studies of anti-VEGFR2 antibodies with tumor-infiltrating lymphocyte characterization due to their intact, functional immune systems that allow direct assessment of immune-tumor interactions. Humanized models are possible but are less frequently utilized and not cited in the major relevant literature above.

Researchers are actively exploring the use of Ramucirumab biosimilars in combination with various checkpoint inhibitors to investigate synergistic effects in complex immune-oncology models, though this represents an emerging area of investigation with significant potential.

Mechanistic Rationale for Combination Approaches

The combination of Ramucirumab with checkpoint inhibitors is based on the understanding that VEGF and its receptor modulate the tumor immune microenvironment. Ramucirumab, as an anti-VEGFR2 monoclonal antibody, works by blocking angiogenesis pathways, while checkpoint inhibitors target different aspects of immune regulation. This dual approach aims to address both the vascular and immune components of the tumor microenvironment simultaneously.

The mechanistic basis for combining multiple therapeutic approaches stems from the recognition that targeting multiple checkpoints can increase the activity of each other and thereby overcome each monotherapy's limitations. Different checkpoint inhibitors have distinct mechanisms of action - for instance, anti-CTLA-4 agents primarily act in the lymph node compartment to restore induction and proliferation of activated T cells, while anti-PD-1/PD-L1 agents mainly function at the tumor periphery to prevent neutralization of cytotoxic T cells.

Current Research Applications Using Biosimilars

Ramucirumab biosimilars are being utilized in research settings to investigate new combinations with immunotherapies and targeted treatments. These biosimilar versions maintain the same structural and functional characteristics as the original drug, demonstrating comparable efficacy in blocking VEGFR-2-mediated signaling pathways while offering researchers more accessible options for studying complex combination effects.

The biosimilar approach provides several advantages for researchers studying combination effects. They enable researchers to expand access to high-quality biologic agents for preclinical and translational studies while helping to reduce research costs while maintaining experimental reliability. This cost-effectiveness is particularly important when designing complex multi-drug combination studies that require extensive dose-finding and optimization work.

Clinical Evidence Supporting Combination Strategies

Recent clinical evidence demonstrates the potential of combining VEGFR inhibition with checkpoint blockade. A randomized phase II study showed that ramucirumab plus pembrolizumab led to improved overall survival compared with standard of care in patients with advanced NSCLC previously treated with chemotherapy and immunotherapy. The study found a median overall survival of 14.5 months for the combination versus 11.6 months for standard of care, representing the first trial in the ICI-acquired resistance setting to demonstrate potential survival benefit.

Research Model Development and Optimization

Researchers use biosimilar combinations to study synergistic effects through various approaches:

Preclinical Model Systems: Complex immune-oncology models often involve multiple cell types including tumor cells, immune cells, and vascular components. Ramucirumab biosimilars allow researchers to systematically study how VEGFR2 blockade enhances the activity of checkpoint inhibitors in these multi-component systems.

Dose and Schedule Optimization: The availability of cost-effective biosimilars enables extensive dose-finding studies to identify optimal ratios and timing for combination treatments. This is particularly important since the major disadvantage of combination immunotherapy is the increase in grade 3 or grade 4 toxicities.

Biomarker Discovery: Researchers can use biosimilar combinations to identify predictive biomarkers for synergistic responses, similar to findings that certain patient populations benefit more from specific combinations based on PD-L1 expression status.

Future Directions and Limitations

While the potential for synergistic effects is promising, researchers must carefully design studies to demonstrate true synergy rather than additive effects. It has been hypothesized that the addition of the combination drug could enhance tumor immunogenicity, and therefore improve overall response, but proving synergistic rather than merely additive effects requires sophisticated experimental design and statistical analysis.

The research use of these biosimilar combinations remains strictly confined to preclinical and translational studies, as Ramucirumab biosimilars are designated for research purposes only and are not intended for clinical use. This regulatory framework ensures that researchers can explore innovative combination approaches while maintaining appropriate safety and regulatory compliance standards.

A Ramucirumab biosimilar can be used as the capture and/or detection reagent in a bridging ADA ELISA to detect anti-drug antibodies (ADAs) generated in a patient’s serum against Ramucirumab or its biosimilar. When monitoring a patient’s immune response, the biosimilar must be highly similar in structure and epitopes to the originator, ensuring that all clinically relevant ADAs—whether they react to the reference or biosimilar—are detectable.

Context and protocol details:

  • In a typical bridging ELISA, the drug (Ramucirumab biosimilar) is immobilized onto a plate to act as the capture reagent. Patient serum is then added, and any ADAs present bind to the biosimilar drug on the plate. After washing, a second molecule of the biosimilar drug—labeled with a detection enzyme (such as HRP) or biotin—is added. This forms a "bridge" via the ADA, linking the capture and detection reagents.
  • The bridging format is highly specific for bivalent antibodies, where both Fab arms of the ADA bind to identical or highly similar epitopes on Ramucirumab.
  • For reliable results, the biosimilar’s quality, purity, and structural similarity to the originator must be validated, as differences in glycosylation, Fc region, or epitope structure could affect ADA detection.

Summary of key steps in bridging ADA ELISA using a Ramucirumab biosimilar:

  • Plate coating: Immobilize the biosimilar as the capture reagent.
  • Sample addition: Add patient serum; ADAs (if present) bind the biosimilar.
  • Detection: Add labeled biosimilar as the detection reagent, so it can bind to the other arm of bivalent ADAs.
  • Signal development: Detection reagent is typically enzyme-linked (e.g., HRP), allowing quantification by signal generation.

Additional notes:

  • This assay is sensitive and high-throughput but requires careful consideration of possible interference from free drug, soluble target (VEGFR-2), or matrix components in patient serum.
  • The method is widely used for biosimilars because they are designed to be immunologically indistinguishable from the originator, ensuring that “total ADA” (against both reference and biosimilar forms) are measured.

In essence, utilizing Ramucirumab biosimilar in bridging ELISA ensures the monitoring of immune responses against both the biosimilar and the originator product, supporting pharmacovigilance and immunogenicity assessment in clinical studies.

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.