Anti-Human PD-L1 (CD274) (Avelumab) [Clone MSB0010718C] — Fc Muted™

Anti-Human PD-L1 (CD274) (Avelumab) [Clone MSB0010718C] — Fc Muted™

Product No.: P705

- -
- -
Product No.P705
Clone
MSB0010718C
Target
PD-L1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Avelumab, PD-L1
Isotype
Human IgG1 L1
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 PD-L1
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 Avelumab. This product is for research use only.Avelumab blocks PD-L1 ligand from interacting with its receptors PD-1 and B7.1.
Background
Programmed cell death 1 ligand 1 (PD-L1; CD274; B7-H1) is a type I transmembrane glycoprotein widely expressed in many types of tissues that acts as a ligand for the immune inhibitory receptor programmed cell death 1 (PD-1; CD279)1, 2, 3 and B7.14. The PD-1 pathway is responsible for T cell activation, proliferation, and cytotoxic secretion, with PD-1/PD-L1 interaction triggering inhibitory signals that dampen T cell function. PD-L1 also plays a critical role in the differentiation of inducible regulatory T cells5.

In normal tissues, PD-L1/PD-1 ligation is crucial to maintaining homeostasis of the immune system and preventing autoimmunity during infection and inflammation5. In the tumor microenvironment, their interaction provides an immune escape mechanism for tumor cells by turning off cytotoxic T cells. As such, blocking the PD-L1/PD-1 interaction is a target of many anti-cancer immunotherapies.

Avelumab is a human IgG1 lambda monoclonal antibody that blocks the interaction between PD-L1 and its receptors PD-1 and B7.1, thereby enabling T cell activation and restoration of the adaptive immune response4. Avelumab can lyse a range of human tumor cells in the presence of peripheral blood mononuclear cells or natural killer cells6, 7. Avelumab engages both adaptive and innate immune functions and mediates antibody-dependent cell-mediated cytotoxicity by retaining a native Fc region6, 7. Avelumab binds to a functional epitope comprising residues Y56, D61, E58, E60, Q66, R113 and M115 as well as a conformational epitope comprising residues 54-66 and 12-122 of human PD-L18.
Antigen Distribution
PD-L1 is commonly expressed on the surface of antigen presenting cells (macrophages, activated B cells, dendritic cells), some epithelial cells under inflammatory conditions, some activated T cells, and several types of tumors as well as tumor infiltrating immune cells. PD-L1 can also exist in a soluble form (sPD-L1) in myeloid-derived cells (monocytes, macrophages, and dendritic cells) and several human cancer lines.
Ligand/Receptor
PD1 (CD279)
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immuno-Oncology
.
Immunology
.
Oncology

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 Avelumab biosimilars serve as critical calibration standards in pharmacokinetic (PK) bridging ELISAs through a comprehensive analytical framework designed to ensure accurate and reliable measurement of drug concentrations in serum samples.

Single Assay Methodology with Biosimilar Standards

The optimal approach for PK assays involves developing a single analytical method that uses one standardized calibration reference for quantifying both biosimilar and reference products. In practice, this means the biosimilar Avelumab is selected as the analytical standard for the unified method, creating a streamlined approach that eliminates variability associated with running multiple methods and removes the need for crossover analysis during blinded clinical studies.

Calibration Standard Preparation and Concentration Range

Research-grade Avelumab biosimilars are prepared as calibration standards in human serum across a wide concentration range. During method validation, nine independent sets of biosimilar standards are typically analyzed with nominal concentrations of 50, 100, 200, 400, 800, 1600, 3200, 6400, and 12800 ng/mL. This broad range ensures accurate quantification across the expected pharmacokinetic profile of the drug in patient samples.

ELISA Assay Configuration and Detection Principles

The bridging ELISA employs a sandwich assay principle where standards and serum samples are incubated in microtiter plates coated with specific reactants for Avelumab. The biosimilar standards create a calibration curve through the following process:

  • Capture Phase: Avelumab (both standards and samples) binds to the coated reactant on the well surface
  • Detection Phase: Horse radish peroxidase (HRP) conjugated probe binds to captured avelumab
  • Quantification: Chromogen-substrate addition produces color intensity proportional to avelumab concentration

Bioanalytical Comparability Assessment

Before implementing the single standard approach, a comprehensive method qualification study must demonstrate bioanalytical equivalence between the biosimilar and reference products. This involves:

Statistical Validation: Precision and accuracy datasets are generated for both biosimilar and reference products, with statistical analysis determining if test products are bioanalytically equivalent within the method.

Equivalence Criteria: The 90% confidence interval is compared to pre-defined equivalence intervals [0.8, 1.25], and bioanalytical equivalence is concluded by combining the totality of evidence.

Quality Control Implementation

Once bioanalytical comparability is established, the method validation proceeds using the biosimilar as the single analytical standard to quantify Quality Control (QC) samples prepared with both biosimilar and reference products. Validation QC samples are typically prepared at concentrations of 50, 150, 1250, 9600, and 12800 ng/mL and quantified against the biosimilar standard curve.

Analytical Performance Specifications

Research-grade biosimilar standards enable robust analytical performance with specific technical parameters:

  • Detection Limit: 10 ng/mL sensitivity
  • Required Sample Volume: 10 μL
  • Spike Recovery: 85-115% accuracy range
  • Total Assay Time: 70 minutes

This standardized approach ensures that concentration data serves as a reliable foundation for PK bioequivalence assessment and dose-response profile characterization, meeting regulatory requirements for biosimilar drug development studies.

The primary in vivo models for studying anti-PD-L1 antibody-mediated tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are:

  • Syngeneic mouse tumor models (using immunocompetent mice with murine tumors)
  • Humanized mouse models (incorporating elements of human immunity or human tumor cells)

Here are details for each category, with representative models and key methods:


Syngeneic Models

These are the most widely used platforms for anti-PD-L1 studies due to their fully functional mouse immune systems.

  • MC38 (colon adenocarcinoma, C57BL/6 background): Common for studying anti-PD-L1 or anti-PD-1 therapy effects on tumor growth and TILs, including CD8+ and CD4+ subsets.
  • CT26 (colon carcinoma, BALB/c background): Allows assessment of tumor growth inhibition and flow cytometry-based analysis of TILs after anti-PD-L1 treatment.
  • EMT-6 (breast carcinoma, BALB/c background) and Hepa1-6 (liver cancer, C57BL/6 background): Used for evaluating the immune microenvironment and response heterogeneity following anti-PD-L1.
  • NS-1 (multiple myeloma, BALB/c): Used for tumor growth inhibition studies and shown responsive to anti-PD-L1, with ability to analyze tumor-infiltrating CD8+ T cells via flow cytometry and immunohistochemistry.

Assessment & Readouts:

  • Tumor volume measurements post-antibody administration track inhibition or regression.
  • Flow cytometry/IHC for TILs (especially CD8+ and CD4+ T cells) determines changes in immune cell infiltration post-treatment.
  • Transcriptome analysis/RNA-seq of TILs after anti-PD-L1 in selected syngeneic models for mechanistic insights.

Humanized Mouse Models

These models enable study of human-specific antibodies and tumor-immune interactions.

  • Humanized target knock-in (KI) mice with syngeneic tumors: Mice express human PD-1 or PD-L1, enabling use of anti-human antibodies; tumors are still murine, permitting analysis of TILs in a controlled background.
  • NSG (NOD scid gamma) mice with human immune cell engraftment and human tumor xenografts: Used for in vivo studies of anti-PD-L1 (or related checkpoint) therapies, analyzing effects on human TIL populations within the tumor.

Assessment & Readouts:

  • Tumor growth kinetics after antibody treatment.
  • Flow cytometry/IHC of human TILs (e.g., CD8+, CD4+, other human leukocytes) in tumor biopsies.

Summary Comparison Table

Model TypeExample TumorsImmune SystemTypical TIL AssaysAntibody Types
Syngeneic (mouse)MC38, CT26, EMT-6, NS-1Mouse (intact)Mouse CD45, CD8+, CD4+Anti-mouse PD-L1
Humanized knock-in mouseMC38 KI, etc.Mouse (human target)Mouse TILsAnti-human PD-L1
Humanized NSG modelPC-3, HCT-116 (human)Human (engrafted)Human CD45, CD8+, CD4+Anti-human PD-L1

In summary:

  • Murine syngeneic tumor models (e.g., MC38, CT26, EMT-6, NS-1) are the dominant platforms for administering research-grade anti-PD-L1.
  • Humanized mouse models are essential when assessing human-specific antibodies or human TILs.
  • In both, tumor growth inhibition and TIL composition (especially CD8+ T cells) are central endpoints.

Researchers investigate the synergistic effects of avelumab biosimilars combined with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) by using complex immune-oncology models—including in vitro co-culture systems, humanized mouse models, and clinical trials—to analyze enhanced immune activation and tumor response.

Key context and supporting details:

  • Mechanistic rationale: Avelumab blocks PD-L1, restoring T cell activity, and uniquely induces antibody-dependent cellular cytotoxicity (ADCC) via its IgG1 structure. Combination with anti-CTLA-4 or anti-LAG-3 targets distinct, non-overlapping checkpoints (CTLA-4 inhibits early T cell priming; LAG-3 downregulates activated T cells), theoretically leading to broader immune activation than monotherapy.

  • Preclinical models: In vitro studies assess effects on T cell proliferation, cytokine secretion, and tumor cell lysis using human peripheral blood mononuclear cells (PBMCs), natural killer (NK) cells, or specific immune cell subsets. Humanized mouse models (mice engrafted with human immune cells) allow direct study of both T cell checkpoint blockade and ADCC (when possible), as well as tumor growth suppression. Notably, avelumab's ADCC effects cannot be directly studied in standard mouse models due to species differences; efficacy here depends more on CD4+/CD8+ T cells.

  • Clinical and translational trials: Combination approaches are being evaluated in the clinic—avelumab combined with anti-CTLA-4 (e.g., ipilimumab) has entered early-phase trials for difficult-to-treat cancers, aiming to leverage increased T cell activation while monitoring for immune-related toxicities.

  • Readouts and biomarkers: Researchers assess:

    • Tumor growth and regression.
    • Activation markers on T cells (e.g., CD69, IFN-γ release).
    • Myeloid and NK-cell function (unique to avelumab).
    • Expression of checkpoint ligands and immune infiltration via immunohistochemistry and flow cytometry.
    • Predictive biomarkers, including PD-L1 on tumor and immune cells, are actively researched, but remain controversial as response predictors.
  • Synergy quantification: Synergistic effects are characterized by greater antitumor efficacy or immune activation in combination versus monotherapies. This can involve:

    • Increased T cell proliferation or cytotoxicity in vitro.
    • Enhanced tumor regression and survival in preclinical models.
    • Additive or synergistic efficacy and manageable toxicity profiles in clinical trials.
  • Limitations: Not all combinations enhance outcomes, as some combos may lead to increased toxicity or diminished additive activity, depending on tumor type and immune landscape.

In summary, avelumab biosimilars are combined with other checkpoint inhibitors in advanced immune-oncology models to explore whether dual or multi-pathway blockade results in more robust immune responses and tumor suppression, with researchers tracking both mechanistic (immune activation) and clinical (survival, tumor regression) endpoints to define synergy.

A Avelumab biosimilar can be directly used as a capture and/or detection reagent in a bridging ADA ELISA to monitor a patient's immune response by detecting anti-drug antibodies (ADAs) generated against Avelumab therapy. In this assay format, the biosimilar serves as a structural and functional proxy for the original drug to bind patient-derived ADAs, making it suitable for immunogenicity testing as long as it mimics the reference antibody's epitopes and post-translational modifications.

Key Steps in the Bridging ADA ELISA Using a Avelumab Biosimilar:

  • Coating/Capture: The microplate is either directly coated with the Avelumab biosimilar or, for higher sensitivity, a biotinylated version of it is captured on streptavidin-coated plates.
  • Sample Incubation: Patient serum samples are incubated in the well. ADAs (if present) will bind to the immobilized Avelumab biosimilar via one binding arm of the antibody.
  • Detection: An enzyme-labeled (commonly HRP) or otherwise detectable form of the same Avelumab biosimilar is added. This labeled biosimilar binds to the other arm of the ADA, forming a “bridge” between the capture and detection molecules.
  • Readout: After washing away unbound molecules, a substrate is added (e.g., TMB for HRP) and colorimetric signal is measured. The intensity correlates with the amount of ADA present.

Why Use a Biosimilar as Reagent?

  • Biosimilars, when developed to be highly similar in structure, glycosylation, and function to the reference drug, present identical immunogenic epitopes as the originator.
  • They are suitable for ADA assays as they will bind anti-Avelumab antibodies generated in patients just as the original therapeutic would.
  • Using a biosimilar can be cost-effective and more accessible for research and regulatory studies when the originator is unavailable, so long as comparability is demonstrated.

Critical Assay Considerations:

  • The bridging format is preferred for its sensitivity and ability to detect bivalent ADAs. However, careful validation is needed to ensure comparable binding efficiencies between biosimilar and originator, as different sources or lots may have subtle biochemical differences affecting ADA binding or assay specificity.
  • Interfering substances in serum (e.g., circulating drug, soluble antigen, or rheumatoid factors) can complicate detection, so assay controls and validation procedures are vital.

Practical Example:Suppose you use a commercial Avelumab biosimilar (e.g., from Bio X Cell) in both biotinylated and HRP-conjugated forms. Both forms would be manufactured to preserve native structure so that patient-derived ADAs generated against Avelumab therapy would efficiently form the antibody "bridge" required for sensitive and specific detection.

In summary, a Avelumab biosimilar is suitable for use as both capture and detection reagent in bridging ADA ELISA as long as it faithfully mimics the therapeutic's structure, enabling an effective assessment of patient immunogenicity toward Avelumab therapy.

References & Citations

1. Freeman GJ, Long AJ, Iwai Y, et al. J Exp Med. 2000192(7):1027-1034. 2000.
2. Tsai KK, Zarzoso I, Daud AI. Hum Vaccin Immunother. 10(11):3111-3116. 2014.
3. Han Y, Liu D, Li L. Am J Cancer Res. 10(3):727-742. 2020.
4. Kim ES. Drugs. 77(8):929-937. 2017.
5. Dermani FK, Samadi P, Rahmani G, et al. J Cell Physiol. 234(2):1313-1325. 2019.
6. Boyerinas B, Jochems C, Fantini M, et al. Cancer Immunol Res. 3(10):1148-1157. 2015.
7. Collins JM, Gulley JL. Hum Vaccin Immunother. 15(4):891-908. 2019.
8. https://patents.google.com/patent/WO2013079174A1/en
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.