Anti-Human PD-L1 (CD274) (Durvalumab) [Clone MEDI4736]

Anti-Human PD-L1 (CD274) (Durvalumab) [Clone MEDI4736]

Product No.: P690

- -
- -
Product No.P690
Clone
MEDI4736
Target
PD-L1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Durvalumab, PD-L1, B7-H1
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
Recommended Isotype Controls
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.
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
Antagonist
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 Durvalumab. This product is for research use only. Durvalumab activity is directed against human PD-L1.
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 B7.1 4. 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 cells 5.

In normal tissues, PD-L1/PD-1 ligation is crucial to maintaining homeostasis of the immune system and preventing autoimmunity during infection and inflammation 5. 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.

Durvalumab was generated using IgG2 and IgG4 XenoMouse animals immunized with human PD-L1-Ig or CHO cells expressing human PD-L1 6. Hybridomas were screened for binding to human PD-L1-transfected HEK 293 cells and inhibition of PD-1 binding to PD-L1 expressing CHO cells. To avoid triggering antibody-dependent cellular cytotoxicity and complement-dependent cytotoxicity, the constant domain was then exchanged for a human IgG1 triple-mutant domain that reduces binding to C1q and Fc gamma receptors. Durvalumab binds specifically to PD-L1 and inhibits interaction with PD-1 and CD80. Durvalumab does not cross react with human PD-L2, B7-H3, or mouse PD-L1. Durvalumab has been investigated as an anti-tumor immunotherapeutic agent in various clinical trials and yields significant improvement in progression-free survival 7,8,9,10.
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
PD-1 (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 Durvalumab biosimilars are used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISAs to accurately quantify drug concentrations in serum samples and to validate the comparability of biosimilar and reference products within the assay.

In PK bridging ELISAs, the assay quantifies Durvalumab (or its biosimilar) in patient samples by comparing unknown samples to a calibration curve generated using known concentrations of the biosimilar or reference Durvalumab. The biosimilar is typically selected as the analytical standard for the assay after demonstrating bioanalytical equivalence between the biosimilar and the reference product through rigorous method qualification and validation steps. This involves:

  • Preparing a calibration curve with the biosimilar in human serum across a wide concentration range (e.g., 50 to 12,800 ng/mL).
  • Running parallel sets of quality control (QC) samples prepared with both the biosimilar and reference Durvalumab to ensure the assay accurately and precisely measures both molecules.
  • Applying statistical analyses to confirm that the biosimilar and reference show equivalent behavior in the assay, ensuring results are comparable regardless of which version is present in the clinical sample.

Reference controls (often biosimilars at specific concentrations) are used throughout assay validation to assess performance characteristics such as accuracy, precision, linearity, and selectivity. This confirms the assay's reliability across a range of expected concentrations found in patient serum during PK studies.

The use of a single, research-grade biosimilar standard streamlines assay processes, reduces variability (versus having multiple standards for reference and biosimilar), and facilitates blinded study designs by avoiding crossover analyses. Once validated, this approach enables high-throughput, sensitive, and selective quantitation of Durvalumab in clinical PK, therapeutic drug monitoring (TDM), or bioequivalence studies.

Key steps summarized:

  • Establish a calibration curve in serum using the biosimilar standard.
  • Validate assay equivalence using both biosimilar and reference controls.
  • Use the biosimilar as the standard and reference for quantification if equivalence is demonstrated.
  • Accurately determine Durvalumab concentrations in patient samples for PK or TDM studies.

This methodology is consistent with regulatory guidance for biosimilar PK assay development and supports bioequivalence and clinical monitoring requirements.

The primary in vivo models used to study anti-PD-L1 antibody-mediated tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models and various forms of humanized mouse models.

Key details:

  • Syngeneic Mouse Models:
    These involve transplanting murine tumor cell lines (such as NS-1 myeloma cells) into immunocompetent mice of the same genetic background, typically strains like BALB/c or C57BL/6. Administration of research-grade anti-PD-L1 antibodies in these models reliably inhibits tumor growth, showing effects such as delayed tumor progression and total regression in some mice. TILs in these models can be extensively profiled, and changes in immune cell populations and cytokine profiles (e.g., IFN-γ and TNF-α in CD8⁺ T cells) can be correlated to anti-PD-L1 therapy response.

  • Humanized Mouse Models:
    These models are immunodeficient mice engrafted with human immune cells or tissues to better recapitulate human immune-tumor interactions. Anti-PD-L1 antibodies (humanized forms) are administered to assess tumor growth inhibition and effects on human TIL populations. Humanized mice permit analysis of human-specific immune markers and checkpoint signaling, and are necessary for evaluating human antibody therapeutics before clinical trials, especially regarding PD-1/PD-L1 interactions in human tumors and immune cells.

  • Clinical Relevance and Characterization:
    The anti-PD-L1 therapy is known to modulate TIL phenotype, increasing activation markers, expanding specific T cell populations (notably CD8⁺), and reversing their exhausted phenotype. These changes are detected both in preclinical models and in human patient samples. Monitoring PD-1 and PD-L1 expression on TILs, their proliferation, cytokine secretion, and cytotoxicity is standard in these studies.

Summary Table:

Model TypeTumor OriginImmune SystemUse CaseCharacterization of TILs
Syngeneic mouseMurine cell lineMouse (wild-type)Immunotherapy, mechanistic studiesMouse TILs, phenotype, cytokines
Humanized mouseHuman cell lineHuman immune cellsTranslational/therapeutic evaluationHuman TILs, checkpoint status

Additional points:

  • Several research-grade and clinical-grade anti-PD-L1 antibodies (e.g., BMS-936559, MEDI4736, MPDL3280A) are used in these models, depending on whether the focus is mechanistic (murine) or translational (humanized).
  • Studies typically measure tumor growth inhibition by caliper or bioluminescent imaging and analyze TILs via flow cytometry, immunohistochemistry, or single-cell sequencing.

In summary:
Both syngeneic mouse models (for mechanistic and immunological studies) and humanized mouse models (for translational relevance) are established in vivo systems to administer anti-PD-L1 antibodies for tumor growth inhibition studies and TIL characterization.

Researchers use biosimilars of Durvalumab (anti-PD-L1) in tandem with other checkpoint inhibitors, such as anti-CTLA-4 biosimilars, to investigate synergistic effects in complex immune-oncology models by evaluating changes in immune cell activation, cytokine production, and gene expression within tumor microenvironments.

  • In ex vivo studies of non-small-cell lung carcinoma (NSCLC), durvalumab was used alone and combined with tremelimumab (an anti-CTLA-4 antibody) to observe effects on immune response. Combination therapy led to:

    • Increased interferon gamma (IFN-γ) production, a marker of T cell activation, with synergy observed as D + T treatment elevated IFN-γ beyond levels seen with durvalumab alone.
    • Additional upregulation of Th1/Th2 pathways and enhanced reduction in genes associated with epithelial-mesenchymal transition (EMT), angiogenesis, and cancer stemness, suggesting potentiation of anti-tumor immune effects.
    • Greater reduction in immunosuppressive cytokine interleukin 10 (IL-10) with combination treatment, which further supports increased anti-tumor immunity.
    • Enhanced activation and proliferation of CD4+/CD8+ T cells and upregulation of activation markers, indicating heightened immune response.
  • In clinical trials (e.g., MYSTIC, NEPTUNE), combinations of durvalumab and tremelimumab have been tested in patients with advanced cancers to examine overall survival benefit and identify predictive biomarkers such as tumor mutational burden. While primary endpoints were not always met, exploratory analyses suggest certain patient subgroups may benefit more from combined checkpoint blockade.

  • Mechanistically, blockade of PD-L1 with durvalumab prevents inhibitory signaling between PD-L1 and PD-1/CD80, releasing the brake on T cell activation. When combined with CTLA-4 or other checkpoint blockade (e.g., anti-LAG-3 biosimilars), this results in multi-faceted relief of immune suppression, allowing for more robust anti-tumor responses.

  • Researchers use these biosimilar combinations in preclinical and clinical settings to study:

    • Gene and protein expression profiles after treatment, helping reveal the pathways most affected by dual checkpoint inhibition.
    • Functional outcomes, such as tumor shrinkage, immune cell infiltration, and changes in the tumor microenvironment, providing evidence for synergistic anti-tumor activity.

There is ongoing investigation into the optimal combinations, dosing, and patient selection criteria for synergistic checkpoint inhibitor therapy, and researchers pay close attention to immune-related adverse events due to increased immune activation. Use of additional checkpoint inhibitors such as anti-LAG-3 biosimilars is under exploration but not as well documented in published clinical combination studies as PD-L1 and CTLA-4.

In summary, by combining durvalumab biosimilars with other checkpoint inhibitors, researchers can model complex immune responses, identify synergistic effects, and refine strategies in immune-oncology for more effective cancer treatments.

A Durvalumab biosimilar is often used as both the capture and detection reagent in a bridging ADA ELISA to monitor a patient's immune response by detecting anti-drug antibodies (ADAs) generated against Durvalumab treatment.

In this assay:

  • Microtiter plate wells are coated with Durvalumab biosimilar to "capture" any ADAs present in the patient's serum or plasma.
  • The patient's sample is incubated, allowing ADAs to bind to the immobilized Durvalumab biosimilar.
  • After washing to remove unbound components, an HRP-conjugated (enzyme-labeled) Durvalumab biosimilar is added as the "detection" reagent.
  • Any ADA present forms a "bridge" between immobilized and enzyme-labeled Durvalumab biosimilars—essentially, the ADA simultaneously binds both drug molecules via their antigen-binding sites.
  • On addition of a chromogenic substrate (such as TMB), the degree of color change is proportional to the quantity of ADAs present in the sample.

Key details:

  • The biosimilar (instead of the reference drug) can be used in this role due to high structural similarity, ensuring immune recognition fidelity but potentially lower development costs and better reagent availability.
  • Since ADAs are bivalent (can bind two antigens), the bridging format is highly specific for these antibodies, minimizing false positives from monovalent binding.
  • The method is widely used in immunogenicity testing for monoclonal antibodies such as Durvalumab, as demonstrated in commercial and research protocols.

This design enables sensitive and specific detection of immune responses, quantifying patient ADAs that could affect drug efficacy or safety.

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. Stewart R, Morrow M, Hammond SA, et al. Cancer Immunol Res. 3(9):1052-1062. 2015.
7. Reichert JM. MAbs. 9(2):167-181. 2017.
8. Faiena I, Cummings AL, Crosetti AM, et al. Drug Des Devel Ther. 12:209-215. 2018.
9. Mathieu L, Shah S, Pai-Scherf L, et al. Oncologist. 26(5):433-438. 2021.
10. Melillo G, Chand V, Yovine A, et al. Adv Ther. 38(6):2759-2778. 2021.
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.