Anti-Human PD-1 (Spartalizumab) – Fc Muted™

Anti-Human PD-1 (Spartalizumab) – Fc Muted™

Product No.: P455

- -
- -
Product No.P455
Clone
PDR001
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD279, PD1, Anti-PD1, PDCD1
Isotype
Human IgG4κ
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-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.
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
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 Spartalizumab. This product is for research use only. Spartalizumab activity is directed against human PD-1.
Background
PD-1 is a transmembrane protein in the CD28/CTLA-4 subfamily of the Ig superfamily 1, 2 . When stimulated via the T cell receptor (TCR), Tregs translocate PD-1 to the cell surface 3. Programmed cell death 1 ligand 1 (PD-L1; CD274; B7H1) and programmed cell death 1 ligand 2 (PD-L2; CD273; B7DC) have been identified as PD-1 ligands 1. PD-1 is co-expressed with PD-L1 on tumor cells and tumor-infiltrating antigen-presenting cells (APCs) 2 . Additionally, PD-1 is co-expressed with IL2RA on activated CD4 + T cells 3 .

PD-1 is an immune checkpoint receptor that suppresses cancer-specific immune responses 4. Additionally, PD-1 acts as a T cell inhibitory receptor and plays a critical role in peripheral tolerance induction and autoimmune disease prevention as well as important roles in the survival of dendritic cells, macrophage phagocytosis, and tumor cell glycolysis 2. 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 immunotherapy 2.

Spartalizumab is a humanized IgG4 anti-PD1 antibody that has been tested for the treatment of various cancers 5, 6. Spartalizumab binds PD-1 with sub-nanomolar affinity and blocks interactions with ligands PD-L1 and PD-L2, leading to T cell activation 5.
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-L1, CD274
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.

Using research-grade Spartalizumab biosimilars as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA involves several key steps and considerations:

Role of Biosimilars in Calibration

  1. Calibration Standards: Biosimilars can be used as calibration standards in ELISA assays to ensure that the assay recognizes both the biosimilar and the reference product equally well. This is crucial for establishing a robust and reliable PK assay, as it helps in minimizing variability and ensuring that both products are measured accurately.

  2. Reference Controls: As reference controls, biosimilars are used to validate the assay's performance by comparing the biosimilar and reference product within the same assay. This approach helps in ensuring that the assay is capable of detecting both products with similar sensitivity and specificity, which is essential for PK studies.

Assay Development and Validation

  1. Assay Development: The development of a PK ELISA involves creating an assay that can quantify both the biosimilar and the reference drug. This is typically achieved by using a single analytical standard for calibration, ensuring that the assay is robust and capable of measuring both products accurately.

  2. Validation Process: The validation process involves determining the assay's precision, accuracy, and sensitivity. This includes evaluating intra- and inter-assay variability, assessing recovery and matrix effects, and ensuring that the assay meets regulatory guidelines such as those set by the FDA and EMA.

  3. Regulatory Compliance: The assay must comply with international standards and regulatory guidelines, such as those from the NIBSC and WHO, to ensure its validity and reliability.

Key Considerations

  • Bioanalytical Comparability: Demonstrating bioanalytical comparability between the biosimilar and reference product is critical. This involves statistical analysis to ensure that the assay treats both products similarly.
  • Scientific Rigor: Employing a scientifically rigorous approach ensures that the assay is suitable for its intended use in PK studies.
  • Orthogonal Techniques: While ELISA is a primary method, orthogonal techniques like chromatography or mass spectrometry can be used to verify results and ensure comprehensive analysis.

By using biosimilars as calibration standards and reference controls, researchers can develop a robust PK bridging ELISA that effectively measures drug concentration in serum samples and supports the development of biosimilar therapies.

The primary models in which research-grade anti-PD-1 antibodies are administered in vivo to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models (such as MC38 colon adenocarcinoma or B16 melanoma) and, for translational applications, humanized mouse models.

Essential context and supporting details:

  • Syngeneic models use immunocompetent mice implanted with mouse-derived tumor cell lines. Because host and tumor share the same genetic background, the immune system can be studied without rejection.

    • Examples: MC38 (colon adenocarcinoma), B16 (melanoma), and various other murine tumor lines.
    • Anti-PD-1 antibodies (murine-specific) are administered, resulting in measurable effects on tumor growth and TIL composition.
    • These models are widely adopted for mechanistic studies of T cell infiltration, effector function, and resistance mechanisms.
    • Serial passaging in these models can produce PD-1-resistant tumor variants for further study.
  • Humanized mouse models employ immunodeficient mice engrafted with human immune cells and implanted with human tumors. These allow testing of human therapeutic antibodies.

    • Used to validate human immune responses and therapeutic candidates such as pembrolizumab or nivolumab.
    • Enable characterization of human TILs, PD-1 expression, cytokine production, and antitumor efficacy in vivo.
    • Some experimental studies use variants such as NSG (NOD scid gamma) mice reconstituted with human immune cells for these investigations.

Key insights for TIL characterization:

  • Administration of anti-PD-1 in these models is used to modulate the immune microenvironment, often resulting in increased infiltration and activation of CD8⁺ T cells, reduction in myeloid-derived suppressor cells (MDSCs), and changes in macrophage polarization.
  • These models help characterize functional changes in TILs, including cytokine output (e.g., IFN-γ, TNF-α), exhaustion marker expression, and anti-tumor cytotoxicity.

Additional relevant information:

  • Syngeneic mouse models remain the gold standard for preclinical mechanistic studies due to their robust immune system and established tumor biology.
  • Humanized models are essential for translational research focused on mimicking human immunotherapy responses and testing human-specific antibodies that cannot cross-react in mouse systems.
  • Choice of model depends on the experimental aim: mechanism discovery (syngeneic) vs therapeutic validation (humanized).

In summary, syngeneic mouse tumor models (using murine anti-PD-1 antibodies) and humanized mouse models (for evaluation of human anti-PD-1 therapeutics) are the primary platforms for in vivo tumor growth inhibition and TIL studies.

Researchers study the synergistic effects of Spartalizumab biosimilar (anti-PD-1) in combination with other checkpoint inhibitors (such as anti-LAG-3 biosimilars) in complex immune-oncology models by conducting preclinical and clinical trials that measure anti-tumor activity, safety, toxicity, and immune biomarkers in various cancer types.

Key approaches and findings include:

  • Combination Therapy Rationale: Resistance to single-agent immune checkpoint therapy (such as anti-PD-1) is often linked to tumor-mediated upregulation of other inhibitory receptors, like LAG-3, which contributes to T-cell exhaustion and immune escape. Therefore, combining Spartalizumab (blocks PD-1) with anti-LAG-3 biosimilars aims to overcome these adaptive resistance mechanisms and restore effective anti-tumor immunity.

  • Model Systems: Researchers use advanced solid malignancies—including melanoma, NSCLC (non-small cell lung cancer), RCC (renal cell carcinoma), TNBC (triple-negative breast cancer), and mesothelioma—as clinical models to evaluate these combinations. Preclinical characterization is performed in humanized mouse models and ex vivo tumor samples to study immune cell interactions, gene expression profiles, and tumor microenvironment dynamics.

  • Biomarker-Driven Analysis: The synergistic effects are quantified by evaluating:

    • Clinical endpoints: Objective response rate (ORR), progression-free survival (PFS), and overall survival (OS).
    • Immune biomarkers: Expression of T-cell-inflamed gene signatures, LAG-3 gene and protein levels, and infiltration of immune cell subsets (CD3, CD8, CD20, etc.) in the tumor microenvironment.
    • Toxicity and safety: Dual checkpoint inhibitor blockade with Spartalizumab and anti-LAG-3 was generally well tolerated, with immune-mediated toxicities comparable to Spartalizumab alone—notably less than combinations involving anti-CTLA-4, which are known for higher toxicity.
  • Synergy Assessment: Studies demonstrated that the combination of Spartalizumab with ieramilimab (anti-LAG-3) led to:

    • Enhanced T-cell activation in the tumor microenvironment.
    • Durable anti-tumor responses in some cases, particularly in patients with high baseline T-cell-inflamed gene signature by RNA sequencing.
    • Increased LAG-3 gene expression upon combination treatment, suggesting upregulation of compensatory pathways and potential for deeper immune reactivation.
  • Limitations and Remaining Challenges: While LAG-3 gene expression in RNA sequencing showed some predictive value, protein-level LAG-3 expression by immunohistochemistry did not reliably predict benefit, highlighting the complexity of biomarker development for these combinations. The lack of single-agent efficacy of anti-LAG-3 in solid tumors further complicates interpretation and model optimization.

  • Extension to Other Checkpoints: Though most clinical synergy evidence is with anti-LAG-3, Spartalizumab (anti-PD-1) has also been tested in various complex models alongside other inhibitors; combinatorial approaches seek to dissect the interplay between multiple immune checkpoints and their collective impact on anti-tumor immunity.

In summary, combining Spartalizumab biosimilar with other checkpoint inhibitors enables the study of enhanced immune activation, improved tumor rejection, and the refinement of predictive biomarkers within complex immune-oncology models, advancing personalized immunotherapy strategies across diverse cancer types.

A Spartalizumab biosimilar can be used as either the capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA assay to monitor a patient’s immune response against the therapeutic drug by exploiting the bivalency of ADAs produced by the patient.

In this format:

  • One form of Spartalizumab biosimilar (usually biotinylated or directly immobilized) is attached to a solid phase (like a microtiter plate), where it serves as the capture reagent.
  • The patient’s serum is added; if anti-Spartalizumab antibodies (ADAs) are present, their two binding arms can simultaneously bind both the capture Spartalizumab and a second, labeled Spartalizumab (for instance, conjugated to HRP or a fluorescent dye), which acts as the detection reagent.

The generic workflow is:

  1. Coat the ELISA plate with Spartalizumab biosimilar (capture reagent).
  2. Add patient serum, allowing any ADAs to bind to the immobilized drug.
  3. Detect bound ADAs by adding a labeled Spartalizumab biosimilar (detection reagent) that binds the other arm of the ADA, forming a “bridging” complex.
  4. Read-out is generated via the label on the detection drug (colorimetric, chemiluminescent, etc.), correlating with the ADA levels present.

This design is appropriate for therapeutic protein drugs and their biosimilars because:

  • Bridging format exploits the fact that anti-drug antibodies are bivalent and can form a bridge between the capture and detection drug molecules.
  • Use of the biosimilar ensures specificity for antibodies generated against the actual therapeutic (which might be the biosimilar itself in clinical use).
  • Since ADAs may recognize both the reference product and biosimilar (depending on their sequence/structure similarity), a biosimilar is a valid reagent for both detection and capture roles.

Key Points:

  • The sensitivity and specificity of ADA detection rely on the quality and labeling of Spartalizumab biosimilar reagents.
  • Possible interference from circulating drug or target in patient serum must be minimized or accounted for.

In summary, a Spartalizumab biosimilar used as both capture and detection reagent in a bridging ADA ELISA allows specific, direct monitoring of a patient’s anti-Spartalizumab immune response during or after therapy.

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 Raimondi G, Shufesky WJ, Tokita D, et al. J Immunol. 176(5):2808-2816. 2006.
4 Pardoll DM. Nat Rev Cancer. 12(4):252-264. 2012.
5 Kaplon H, Reichert JM. MAbs. 11(2):219-238. 2019.
6 Dummer R, Long GV, Robert C, et al. J Clin Oncol. 40(13):1428-1438. 2022.
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.