Anti-Human PD-1 (Tislelizumab)

Product No.: P800

- -
- -
Product No.P800
Clone
BGB-A317
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anti-PD-1, PDCD1, CD279
Isotype
Human IgG4κ
Applications
ELISA
,
FA
,
FC
,
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-1
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
Applications and Recommended Usage?
Quality Tested by Leinco
ELISA,
WB
Additional Applications Reported In Literature ?
FA,
ELISA Cap,
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 Tislelizumab. This product is for research use only. Tislelizumab activity is directed against human PD-1 (CD274).
Background
Programmed cell death 1 (PD-1) is a transmembrane protein in the Ig superfamily1,2 that acts as an immune checkpoint receptor3, a T cell inhibitory receptor, plays critical roles in peripheral tolerance induction, autoimmune disease prevention, macrophage phagocytosis, tumor cell glycolysis, and dendritic cell survival2. PD-1 prevents uncontrolled T cell activity, leading to attenuation of T cell proliferation, cytokine production, and cytolytic activities. Additionally, the PD-1 pathway is a major mechanism of tumor immune evasion, and, as such, PD-1 is a target of cancer immunotherapy2. Programmed cell death 1 ligand 1 (PD-L1; CD274; B7H1) and programmed cell death 1 ligand 2 (PD-L2; CD273; B7DC) are ligands1.

Tislelizumab was developed by BeiGene as an immunotherapeutic for hematological cancers and advanced solid tumors4. Tislelizumab is a humanized mouse monoclonal antibody designed as a synthetic protein fusion of the 317-4B6 heavy chain VH fragment with human γ4 chain clone mut10 effector/constant domain fragment (disulfide with anti-human PD-1) and synthetic clone 317-4B6 light chain VL fragment with human κ chain constant region fragment, dimer4,5.

Tislelizumab binds to PD-1 with high specificity and affinity using the critical epitopes Gln75, Thr76, Asp77 and Arg86, blocking PD-1 and preventing ligand binding4. The epitope is located on the CC’ loop of the front β sheet face of PD-1 and causes stereospecific hindrance to PD-L1 binding6. Unlike other IgG4 anti-PD-1 blocking antibodies, the S228P mutation known to bind to Fc-γ receptor 1 (FcγRI) and induce antibody-dependent cellular phagocytosis of T cells is not present4 and several mutations in the Fc-hinge region render tislelizumab unable to bind to FcγRs generally6. Consequently, tislelizumab has low affinity for FcγRI and baseline antibody-dependent cellular phagocytosis relative to control antibodies4. Additionally, FcR-mediated effects such as antibody-dependent cell-mediated cytotoxicity or compliment-dependent cytotoxicity are not observed4,6.
Antigen Distribution
PD-1 is expressed on activated T cells, B cells, a subset of thymocytes, macrophages, dendritic cells, and some tumor cells and is also retained in the intracellular compartments of regulatory T cells (Tregs).
Ligand/Receptor
PD-1, CD279
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.

Research-grade Tislelizumab biosimilars serve as critical calibration standards and reference controls in pharmacokinetic bridging ELISAs designed to measure drug concentrations in serum samples. This approach enables accurate quantification of both biosimilar and reference drug products using a unified analytical method.

Single Assay Strategy for Biosimilar Measurement

The most optimal approach for PK assays involves developing a single analytical method using one calibration standard to quantitatively measure both the biosimilar and reference products in serum samples. This strategy offers significant advantages by reducing inherent variability associated with multiple methods and eliminating the need for crossover analysis during blinded clinical studies.

Calibration Standard Development

Research-grade biosimilars are typically calibrated against multiple reference points to ensure accuracy and reliability. These include calibration against international standards from organizations like the National Institute of Biologicals and Control (NIBSC), as well as against commercially sourced innovator drugs and alternative recombinant biosimilars. This multi-tiered calibration approach ensures a high degree of accuracy when measuring drug concentrations in biological samples.

ELISA Assay Design and Implementation

The bridging ELISA employs anti-idiotypic monoclonal antibodies in a sandwich assay format, which provides exceptional specificity and sensitivity for drug detection, even at low concentrations. For Tislelizumab specifically, the assay utilizes a competitive format with colorimetric detection, capable of measuring concentrations across a test range of 0.31 μg/ml to 5 μg/ml.

The assay typically includes:

  • Lyophilized standards prepared in human serum at multiple concentration levels
  • Quality control samples prepared at low, medium, and high concentrations
  • Validation across multiple analytical runs performed by different analysts over several days

Validation and Performance Characteristics

Bioanalytical Comparability Assessment: Before implementing a single-standard approach, comprehensive method qualification studies generate precision and accuracy datasets for both biosimilar and reference products. Statistical analysis determines if the test products are bioanalytically equivalent within the method, typically using a 90% confidence interval compared to pre-defined equivalence intervals of [0.8, 1.25].

Precision Requirements: The assays must meet stringent precision criteria with coefficient of variation (CV) values:

  • Intra-assay CV: <10% for low concentrations, <5% for medium and high concentrations
  • Inter-assay CV: <10% for low concentrations, <5% for medium and high concentrations

Quality Control and Standardization

The validation process follows established regulatory guidelines including FDA and EMA requirements, with validation conducted according to ICH/EMA guidelines under ISO 13485 certification. Performance parameters are established consistent with quantitative pharmacokinetic methods as described in bioanalytical FDA guidance documents.

Matrix Effects and Recovery: To ensure no matrix interference and reproducible dilutional linearity, extensive serum and plasma spiking experiments are conducted at various dilutions during optimization. This ensures that the biosimilar standards perform consistently across different biological matrices and concentration ranges.

The research-grade biosimilar ultimately serves as the analytical standard for the validated single method, enabling accurate quantification of drug concentrations in clinical samples while maintaining the analytical rigor necessary for regulatory submission and biosimilar development programs.

To study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) using research-grade anti-PD-1 antibodies, researchers primarily employ two types of in vivo models: syngeneic mouse models and humanized mouse models.

Syngeneic Mouse Models

Syngeneic models involve the use of immunocompetent mice with a genetically identical background, which allows for the study of immune responses in a fully intact immune system. These models are often used to study the effects of anti-PD-1 therapies on tumor growth and immune cell infiltration. Tumors in syngeneic models can be derived from mouse cell lines that are syngeneic with the host (e.g., MC38 colon adenocarcinoma in C57BL/6 mice), enabling researchers to investigate the dynamics of TILs and other immune cells in response to anti-PD-1 treatment.

Syngeneic models are particularly useful for studying the mechanisms of action of anti-PD-1 therapy in a complete immune environment and for identifying potential resistance mechanisms, as shown in studies where resistant tumor models are developed through serial passaging of tumors in mice treated with anti-PD-1 antibodies.

Humanized Mouse Models

Humanized mouse models involve the transplantation of human immune cells or tissues into immunodeficient mice. These models can be used to study the effects of anti-PD-1 antibodies in a setting more relevant to human immune responses. In humanized models, patient-derived tumor xenografts (PDX) can be used, allowing for the study of human TILs in response to anti-PD-1 therapy. This approach is particularly useful for understanding the complex interactions between human immune cells and tumor cells under the influence of checkpoint inhibitors like anti-PD-1 antibodies.

However, the search results do not specifically highlight the use of humanized models with anti-PD-1 in the context of studying TILs and tumor growth inhibition. Typically, humanized models are more often used to study specific aspects of human immune responses, such as the activity of human T cells or the efficacy of human-specific therapies.

In summary, syngeneic models are frequently used for studying anti-PD-1 therapy's effect on tumor growth inhibition and TIL characterization due to their complete immune system, whereas humanized models offer a more human-relevant context for studying immune responses, though they are less commonly highlighted in this specific research context.

Researchers studying the synergistic effects of Tislelizumab biosimilars with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) in complex immune-oncology models typically employ combination strategies in preclinical and early-phase clinical settings to evaluate enhanced antitumor responses and mechanisms of resistance.

Essential context and supporting details:

  • Rationale for Combination: Targeting multiple immune checkpoints, such as PD-1/PD-L1 (with Tislelizumab) and CTLA-4 or LAG-3, can amplify antitumor immunity because these checkpoints function at different regulatory points of the immune response. PD-1 blockade primarily works in the tumor microenvironment to restore T-cell effector functions, while CTLA-4 inhibitors act in lymphoid organs to support T-cell priming and expansion.

  • Preclinical and Translational Models:

    • Researchers often use mouse tumor models with humanized immune systems, or patient-derived tumor models, to test the efficacy and immunologic responses when combining these inhibitors.
    • In such studies, Tislelizumab biosimilar is administered alongside other checkpoint inhibitor biosimilars, and researchers monitor tumor growth, survival, immune cell infiltration, cytokine profiles, and other immunological endpoints to detect synergistic effects.
    • These models are leveraged to mimic clinical resistance, immune evasion, and variability in immunotherapy response.
  • Observed Effects and Insights:

    • The combination often shows greater antitumor activity compared to monotherapy, as seen with dual blockade of PD-1 and CTLA-4 in both preclinical and clinical studies, enhancing responses especially in tumors that are resistant to single-agent therapy.
    • Analysis involves measuring objective response rates (ORR), progression-free survival (PFS), overall survival (OS), and immune-related adverse events to understand both efficacy and safety.
    • Translational studies focus on immune profiling (e.g., T-cell activation status, exhaustion markers) and tumor microenvironment changes post-treatment, with particular attention to overcoming suppressive pathways.
  • Experimental Workflows:

    • Agents are given in various dose combinations and schedules to optimize the synergy while minimizing toxicity.
    • Advanced translational research integrates multi-omics (genomics, transcriptomics, proteomics) and advanced imaging/flow cytometry to dissect mechanistic underpinnings and therapeutic windows.

Additional information:

  • The success of combinations, such as anti-PD-1 with anti-CTLA-4, in improving durable responses has led to ongoing interest in adding further checkpoints like LAG-3 to counteract adaptive resistance.
  • The primary limitation is increased immune-related adverse events, which necessitates balancing efficacy with safety, especially as combinations move into clinical trials.
  • While Tislelizumab has demonstrated meaningful results as monotherapy in various cancers, its full synergistic potential with other biosimilar checkpoint inhibitors continues to be explored and optimized in ongoing preclinical and early-clinical studies.

In summary, researchers use Tislelizumab biosimilars in combination with other checkpoint inhibitor biosimilars in sophisticated preclinical immune-oncology models to dissect and enhance antitumor responses, employing a range of translational approaches to measure synergy, resistance mechanisms, and safety.

A Tislelizumab biosimilar can be effectively utilized in a bridging ADA ELISA assay to monitor patient immune responses by serving as both the capture and detection reagent in this highly sensitive immunogenicity testing format.

Bridging ELISA Design for Tislelizumab

In a bridging ADA ELISA designed to detect anti-Tislelizumab antibodies, the biosimilar functions through a dual-reagent system. The biotinylated Tislelizumab biosimilar is first captured on streptavidin-coated microtiter plates, creating a solid-phase antigen surface. Patient serum samples are then added, allowing any anti-drug antibodies present to bind to the captured Tislelizumab biosimilar. For detection of bivalent anti-drug antibodies, a second Tislelizumab biosimilar molecule labeled with a detection marker (such as HRP or fluorescent dye) is introduced.

Mechanism of ADA Detection

The assay operates on the principle that anti-drug antibodies in patient samples will simultaneously bind to both the capture and detection forms of the Tislelizumab biosimilar, creating a "bridge" between the solid phase and the detection reagent. This bridging formation occurs only when bivalent ADAs are present, as they possess multiple binding sites that can interact with both forms of the drug simultaneously. The signal generated is directly proportional to the concentration of anti-Tislelizumab antibodies in the patient sample.

Advantages and Considerations

The bridging ELISA format offers high sensitivity and enables high-throughput sample screening, making it particularly valuable for monitoring multiple patients over extended treatment periods. The technique has been successfully applied to detect ADAs against various monoclonal antibody-based therapeutics, including adalimumab, infliximab, and etanercept.

However, the specificity of bridging ELISA assays can be challenged by complex serum matrices, soluble target molecules, or residual drug components that may interfere with the assay. For Tislelizumab monitoring, this is particularly relevant since PD-1 targeting drugs can have complex interactions with circulating immune components.

Optimization Strategies

To maximize assay performance when using Tislelizumab biosimilar reagents, several optimization approaches can be employed. Acid dissociation pretreatment can be combined with the bridging ELISA to minimize interference from high drug concentrations, which is especially important during active treatment periods. Additionally, the use of high-quality blocking solutions and careful optimization of reagent concentrations are crucial for obtaining meaningful results in the complex matrix of human serum.

The detection limit and linear range of the assay will depend on the specific labeling efficiency and binding characteristics of the Tislelizumab biosimilar reagents used, but similar bridging ELISAs have achieved detection limits as low as 0.39 ng/mL with linear ranges spanning 0.39-50 ng/mL.

References & Citations

1. Matsumoto K, Inoue H, Nakano T, et al. J Immunol. 172(4):2530-2541. 2004.
2. Zhao Y, Harrison DL, Song Y, et al. Cell Rep. 24(2):379-390.e6. 2018.
3. Pardoll DM. Nat Rev Cancer. 12(4):252-264. 2012.
4. Lee A, Keam SJ. Drugs. 80(6):617-624. 2020.
5. https://searchusan.ama-assn.org/usan/documentDownload?uri=/unstructured/binary/usan/tislelizumab.pdf
6. Zhang L, Geng Z, Hao B, et al. Cancer Control. 29:10732748221111296. 2022.
7. Zhang T, Song X, Xu L, et al. Cancer Immunol Immunother. 67(7):1079–90. 2018.
8. Zhang T, Song J, Li Y, et al. Cancer Research Conference: 107th AACR Annual Meeting 2016;76(Suppl 14).
9. Desai J, Deva S, Lee JS, et al. J Immunother Cancer. 8(1):e000453. 2020.
10. Song Y, Gao Q, Zhang H, et al. Leukemia. 34(2):533-542. 2020.
11. Hong Y, Feng Y, Sun H, et al. FEBS Open Bio. 11(3):782-792. 2021.
Indirect Elisa Protocol
FA
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.