Anti-Human PD-L1 (Atezolizumab) [Clone RG7446]

Anti-Human PD-L1 (Atezolizumab) [Clone RG7446]

Product No.: LT1750

- -
- -
Product No.LT1750
Clone
RG7446
Target
PD-L1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Programmed Death Ligand 1, B7-H1, PD-L1, CD274
Isotype
Human IgG
Applications
FC
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
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.
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
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Atezolizumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Applications Reported In Literature ?
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
Atezolizumab (RG7446; INN or code name MPDL3280A) activity is directed against PD-L1 (CD274) and B7.1 (CD80).
Background
Atezolizumab is a humanized, monoclonal immunoglobulin-G1 antibody that binds to programmed death ligand 1 (PD-L1; CD274) and B7.1 (CD80)1. PD-L1 is a transmembrane protein, widely expressed in many types of tissues, that acts as a ligand for the immune inhibitory receptor protein programmed death 1 (PD-1)2,3,4. Interaction between PD-1 and PD-L1 triggers inhibitory signals that dampen T cell function. PD-1 is expressed on activated T cells and is overexpressed on many human cancer cell types and on various tumor-infiltrating immune cells. B7.1 is a transmembrane glycoprotein present on dendritic cells, activated B cells, and macrophages that induces T cell proliferation and cytokine production. When atezolizumab prevents binding of PD-L1 to B7.1, the T-cell-mediated immune response is further enhanced4.

Atezolizumab was isolated by screening a human phage display library against a recombinant extracellular domain-Fc fusion of human PD-L11,5. A high-affinity antibody was selected from a single phage clone on a human IgG1 backbone. Because PD-L1 is expressed on activated T cells, the Fc region of atezolizumab was engineered to eliminate antibody-dependent cytotoxicity (ADCC) or complement-dependent cytotoxicity (CDC)1. An Asn to Ala change at position 298 was introduced in the CH2 domain of each heavy chain, rendering atezolizumab effectorless and incapable of binding to human Fcγ receptors1,5. Atezolizumab does not interfere with the interaction of PD-1 with ligand PD-L2 (CD273).

Atezolizumab is used in cancer immunotherapy and has been approved for some patients by the FDA to treat hepatocellular carcinoma, melanoma, non-small cell lung cancer, small cell lung cancer, urothelial cancer, and triple negative breast cancer6.
Antigen Distribution
PD-L1 is commonly expressed on the surface of antigen presenting cells (APC) and tumor cells. B7.1 is found on activated APCs including dendritic cells, macrophages, and activated B cells.
Ligand/Receptor
PD-1 (PDCD1)
Research Area
Biosimilars
.
Cancer
.
Costimulatory Molecules
.
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.

Role of Research-Grade Atezolizumab Biosimilars in PK Bridging ELISA

Atezolizumab biosimilars—developed and characterized for research purposes—can be employed as calibration standards or reference controls in pharmacokinetic (PK) bridging enzyme-linked immunosorbent assays (ELISAs) to measure drug concentration in serum samples. The process and rationale are as follows:

Calibration Standards

  • Calibration Curve Construction: Both innovator (Tecentriq™) and research-grade biosimilars can be used to prepare a set of standards (calibrators) at known concentrations within relevant serum matrices. These standards are used to generate a calibration curve, against which unknown sample concentrations are interpolated.
  • Reference Against Innovator: For maximum accuracy, standards are often calibrated against the commercially sourced innovator product (Tecentriq™) to ensure traceability and comparability to clinical material. Where available, international reference standards (e.g., from NIBSC) may also be used for calibration.
  • Matrix Compatibility: The performance of calibration standards in serum matrix (diluted to minimize interference) is verified using spiking and recovery experiments. Optimal recoveries are typically observed at intermediate dilutions (e.g., 1:1000 or 1:2000), confirming that the assay accurately quantifies atezolizumab in complex biological fluids.

Reference Controls

  • Quality Control (QC) Samples: In addition to calibration standards, reference controls—prepared with both the biosimilar and the innovator—are included in each assay run to monitor assay performance and ensure consistency across batches.
  • Bioanalytical Comparability: During method development and validation, both the biosimilar and innovator are tested in parallel to confirm that the assay provides equivalent results for both products. If bioanalytical equivalence is demonstrated, a single assay with a single standard (often the biosimilar for practical reasons) may be used for both products in clinical or nonclinical studies.
  • Statistical Equivalence: Regulatory guidance and industry best practices emphasize the need for rigorous statistical analysis (e.g., 90% confidence intervals compared to predefined equivalence margins) to confirm that the biosimilar and innovator are measured equivalently by the assay.

Technical and Validation Considerations

  • Assay Specificity: The use of anti-idiotypic monoclonal antibodies ensures high specificity for atezolizumab, minimizing cross-reactivity and matrix effects.
  • Sensitivity and Precision: The assay’s limit of detection (LoD), sensitivity, and precision (intra- and inter-assay variability) are determined during validation, typically with both innovator and biosimilar. Precision is expressed as %CV, with values <10% generally accepted.
  • Guideline Compliance: The entire process—from calibration to validation—is conducted according to EMA/FDA/ICH guidelines and, where applicable, ISO 13485 certification, ensuring regulatory compliance and scientific rigor.

Bridging Strategy

The bridging concept refers to the use of a single, validated assay platform to measure both innovator and biosimilar concentrations, demonstrating that the biosimilar is measured equivalently to the innovator in serum samples. This is essential for PK similarity studies in biosimilar development, where differences in assay performance could confound the interpretation of PK data.

Summary Table: Key Steps in Using Biosimilars as Standards/Controls

StepPurposeExample/Reference
Calibration curve generationQuantify unknown samples
QC sample preparationMonitor assay performance
Bioanalytical comparabilityConfirm equivalent measurement of innovator/biosimilar
Matrix recovery experimentsVerify accuracy in serum
Statistical equivalenceDemonstrate regulatory compliance

Conclusion

Research-grade atezolizumab biosimilars are used as calibration standards and reference controls in PK bridging ELISAs by generating calibration curves, serving as quality controls, and demonstrating bioanalytical comparability to the innovator. This approach enables accurate, specific, and regulatory-compliant quantification of atezolizumab in serum, supporting both PK similarity studies and therapeutic drug monitoring.

The primary preclinical models used to study in vivo tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) following administration of a research-grade anti-PD-L1 antibody are:

  • Syngeneic mouse tumor models (using immunocompetent mice and mouse-derived tumor lines)
  • Humanized mice with xenografted human tumors (typically employing human immune system components and human tumor lines)

Syngeneic Models

Syngeneic models are the most commonly used platforms for such studies because they preserve an intact, functional murine immune system. These models use mouse tumor cell lines implanted into genetically identical (or nearly identical) immunocompetent mice, allowing direct assessment of immune-mediated antitumor effects of anti-PD-L1 antibodies. Examples of commonly used syngeneic tumor models include:

  • MC38 (colon carcinoma, C57BL/6 background)
  • CT26 (colon carcinoma, BALB/c background)
  • EMT-6 (breast cancer, BALB/c background)
  • Hepa1-6 (liver cancer, C57BL/6 background)
  • NS-1 (myeloma, BALB/c background)

These models reliably enable evaluation of tumor growth inhibition and quantitative and qualitative characterization of TILs (e.g., CD8^+ T cells) by flow cytometry, immunohistochemistry, and transcriptomic analyses. For example, in the MC38 and Hepa1-6 models, anti-PD-1/PD-L1 treatment, sometimes combined with depletion of CD8^+ or CD4^+ T cells, is used both to study tumor growth and detailed immune populations within the tumor microenvironment. Mouse anti-PD-L1 antibodies are typically used, ensuring cross-reactivity and functional blockade of the murine PD-L1 pathway.

Humanized and Human Knock-in Models

Humanized mouse models are used for translational studies with anti-human PD-L1 antibodies, which do not cross-react with murine PD-L1. These involve:

  • Immunodeficient mice (like NSG mice) engrafted with human hematopoietic cells (producing a human immune system), and
  • Engraftment of human tumor cell lines (e.g., PC-3, HCT-116).

Alternatively, newer “human knock-in” (KI) mice express human PD-1 or PD-L1 genes in otherwise syngeneic (immunocompetent) backgrounds, allowing use of research-grade anti-human PD-L1 antibodies in a more immunologically intact system.

These humanized models allow for:

  • Tumor growth inhibition studies with anti-human PD-L1 mAbs.
  • Direct investigation of the effect on human TILs such as CD8^+ and CD4^+ T cells.
  • More direct assessment of antibodies designed for clinical use in humans.

Model Comparison Table

Model TypeTumorImmune SystemAntibody UsedTypical Applications
SyngeneicMouseMouseAnti-mouse PD-L1Tumor inhibition, mouse TIL analysis, dosing/toxicology studies
Humanized xenograftHumanHuman (HSC)Anti-human PD-L1Tumor inhibition, human TIL analysis, translational evaluation
Human PD-L1 KI mouseMouseMouseAnti-human PD-L1Bridging mouse & human assays, mechanistic/efficacy studies

Essential Context

  • Syngeneic models predominate due to their practicality, cost-effectiveness, and preserved immunity. They support TIL profiling, cytokine analysis, and investigation of response/resistance to immunotherapy.
  • Humanized models are essential for testing antibodies with strict human specificity and for translational studies but are more technically demanding and expensive.
  • Both model types are often used in parallel to complement mechanistic murine studies with translational human data.

In summary: Most tumor growth and TIL characterization studies using research-grade anti-PD-L1 antibodies employ syngeneic mouse models (such as MC38, CT26, EMT-6, NS-1) or, for human reactivity, humanized or human knock-in mice with relevant human tumors and immune cells.

Use of Atezolizumab Biosimilars in Combination with Other Checkpoint Inhibitor Biosimilars

Atezolizumab is an anti-PD-L1 monoclonal antibody, and its biosimilars are utilized in research to emulate its mechanism of action at lower cost while maintaining the biological specificity and activity of the original drug. Researchers employ these biosimilars—alongside other checkpoint inhibitor biosimilars, such as those targeting CTLA-4 or LAG-3—to investigate synergistic effects in complex immune-oncology models. This approach is increasingly important as combination immunotherapy emerges as a promising strategy to overcome resistance and enhance tumor responses.

Experimental Rationale and Design

  • Synergy Testing: Scientists combine atezolizumab biosimilars with biosimilars of other checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) to test whether dual or multiple checkpoint blockade produces a stronger anti-tumor effect than monotherapy.
  • Mechanistic Studies: These combinations aim to target different immune checkpoints simultaneously, exploiting their unique biological roles. For instance, CTLA-4 inhibitors primarily act in lymph nodes to enhance T-cell priming, while PD-1/PD-L1 inhibitors (like atezolizumab) act mainly in the tumor microenvironment to prevent T-cell exhaustion.
  • Overcoming Resistance: Preclinical studies suggest that compensatory upregulation of alternative inhibitory checkpoints (e.g., LAG-3, TIM-3) can limit the efficacy of single-agent therapy. Dual or triple blockade may reverse T-cell exhaustion and improve tumor control.

Technical Approach

  • Biosimilar Production: Atezolizumab biosimilars are generated in mammalian cell systems (e.g., CHO cells), purified, and validated for binding and functional equivalence to the originator drug. Similar processes are used for other checkpoint inhibitor biosimilars.
  • In Vitro and In Vivo Models: Researchers use these biosimilars in co-culture systems (e.g., immune cells with tumor cells) and animal models (e.g., humanized mice with patient-derived xenografts) to assess tumor growth, immune cell infiltration, cytokine profiles, and survival outcomes.
  • Endpoint Analysis: Key readouts include tumor volume measurement, immune cell phenotyping (e.g., CD8+ T cells, Tregs), cytokine secretion (e.g., IFN-γ), and gene expression profiling to assess changes in the tumor microenvironment.

Advantages of Using Biosimilars in Research

  • Cost-Effectiveness: Biosimilars reduce the financial burden of high-throughput or large-scale studies, allowing more extensive exploration of combination regimens.
  • High Fidelity: They accurately replicate the biological activity of originator drugs, ensuring that experimental results are relevant to clinical scenarios.
  • Versatility: Biosimilars can be used across a range of preclinical applications, from basic mechanistic studies to translational research.

Clinical and Translational Relevance

While these studies are primarily preclinical, their findings inform the design of clinical trials testing combination immunotherapies. For example, the combination of anti-CTLA-4 and anti-PD-1/PD-L1 agents has already shown improved outcomes in several cancers and is FDA-approved for certain indications. Ongoing research with biosimilars continues to explore novel combinations (e.g., anti-PD-L1 + anti-LAG-3) to identify new therapeutic opportunities, especially in tumors resistant to single-agent immunotherapy.

“The logic of combining multiple checkpoint inhibitors is that they have different mechanisms of action...thereby overcoming each monotherapy’s limitations.”

Summary Table: Research Applications of Atezolizumab Biosimilars in Combination Studies

ApplicationPurposeExample CombinationsKey Outcomes Measured
Synergy TestingEnhance anti-tumor immunityAnti-PD-L1 + anti-CTLA-4/LAG-3Tumor growth, survival
Mechanistic StudiesElucidate immune cell dynamicsMulti-checkpoint blockadeT-cell activation, cytokine levels
Resistance OvercomingReverse T-cell exhaustionAnti-PD-L1 + anti-TIM-3/VISTAImmune cell infiltration
Biosimilar ValidationEnsure equivalence to originator drugsN/ABinding, functional assays

Conclusion

Researchers leverage atezolizumab biosimilars—alongside other checkpoint inhibitor biosimilars—to systematically study the synergistic potential of combination immunotherapies in complex immune-oncology models. These studies are critical for identifying effective multi-agent regimens, understanding mechanisms of action and resistance, and guiding the development of next-generation cancer immunotherapies. Biosimilars play a pivotal role in making such research more accessible and reproducible, accelerating the translation of findings from bench to bedside.

Role of Atezolizumab Biosimilar in ADA Bridging ELISA

Anti-drug antibody (ADA) bridging ELISA is a widely used method to monitor a patient’s immune response (immunogenicity) to therapeutic proteins, including monoclonal antibodies such as atezolizumab. The principle of this assay involves detecting antibodies produced by the patient against the therapeutic drug (in this case, atezolizumab) by bridging two drug molecules—one used as a capture reagent on the plate and another as a detection reagent, both typically requiring the same or highly similar drug structure.

Application of Atezolizumab Biosimilar

A biosimilar is designed to be highly similar to the reference product (here, atezolizumab), particularly in the variable regions that determine antigen binding, while manufacturing processes and some structural features may differ slightly. The use of an atezolizumab biosimilar as the capture or detection reagent in a bridging ADA ELISA is not standard practice for approved clinical immunogenicity assays, but in research settings, a biosimilar can be used to functionally mimic the therapeutic for antibody detection.

Capture Strategy

  • Capture Reagent: The biosimilar atezolizumab (or the reference drug itself, if available) is immobilized onto a solid phase, such as a microtiter plate, often via biotinylation and streptavidin coating to ensure efficient and uniform capture.
  • Sample Addition: Patient serum, which may contain ADAs, is added. If anti-atezolizumab antibodies are present, they will bind to the immobilized atezolizumab (or biosimilar) on the plate.

Detection Strategy

  • Detection Reagent: The same or a highly similar atezolizumab molecule (biosimilar or reference drug), conjugated to a detectable label (e.g., horseradish peroxidase, HRP), is added.
  • Formation of Complex: Bivalent ADAs can bridge the immobilized drug and the labeled drug, enabling detection via enzymatic reaction and subsequent signal measurement.
  • Signal Interpretation: The intensity of the signal correlates with the amount of ADA present in the patient’s serum.

Rationale for Using Biosimilars

  • Functional Equivalence: Since the critical component for ADA binding is the antigenic epitope, a biosimilar that preserves the variable regions of atezolizumab should theoretically bind ADAs with similar affinity and specificity as the reference drug.
  • Research Applications: Biosimilars are particularly useful for research, where the reference drug may be cost-prohibitive or unavailable in large quantities for assay development.
  • Validation Requirements: Any biosimilar used in an ADA assay must be rigorously validated to ensure it behaves identically to the reference drug in terms of ADA binding, to avoid false positives or negatives.

Limitations and Considerations

  • Exact Epitope Matching: Minor differences in glycosylation or other post-translational modifications between biosimilar and reference drug could theoretically affect ADA binding, potentially impacting assay sensitivity and specificity.
  • Assay Development: The use of biosimilars in clinical immunogenicity testing is not standard unless equivalence has been formally demonstrated to regulatory standards. For research, however, biosimilars can be a practical alternative.
  • Matrix Effects: Complex biological matrices can interfere with ADA detection, so robust assay optimization and validation are essential.

Summary Table: Roles of Atezolizumab and Biosimilar in ADA Bridging ELISA

Assay StepTypical ReagentPossible Biosimilar UsePurpose
CaptureReference atezolizumabAtezolizumab biosimilarImmobilize drug to capture patient ADAs
DetectionReference atezolizumab (labeled)Atezolizumab biosimilar (labeled)Bind ADAs to generate detectable signal

Conclusion

An atezolizumab biosimilar can serve as both the capture and detection reagent in a bridging ADA ELISA to monitor patient immune responses against the therapeutic, provided it demonstrates equivalent binding characteristics to ADAs as the reference drug. This approach is especially valuable in research settings, but requires rigorous validation for clinical use to ensure reliability and regulatory compliance. The bridging ELISA format remains a sensitive and specific tool for immunogenicity assessment, with the choice of reagent (reference or biosimilar) depending on availability, cost, and validation data.

References & Citations

1. Herbst RS, Soria JC, Kowanetz M, et al. Nature. 515(7528):563-567. 2014.
2. Freeman GJ, Long AJ, Iwai Y, et al. J Exp Med. 2000192(7):1027-1034. 2000.
3. Tsai KK, Zarzoso I, Daud AI. Hum Vaccin Immunother. 10(11):3111-3116. 2014.
4. NCI Dictionaries. https://www.cancer.gov/publications/dictionaries/cancer-drug/def/atezolizumab
5. Irving H, Chiu H, et al, inventors; F Hoffmann La Roche AG, assignee. Anti-PD-L1 antibodies, compositions and articles of manufacture. US Patent US 8,217,149B2. July 10, 2012.
6. A to Z List of Cancer Drugs. https://www.cancer.gov/about-cancer/treatment/drugs/atezolizumab
Flow Cytometry
General Western Blot Protocol

Certificate of Analysis

Formats Available

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