Anti-Human CD279 (PD-1) (Nivolumab) [Clone 5C4.B8] — Fc Muted™

Anti-Human CD279 (PD-1) (Nivolumab) [Clone 5C4.B8] — Fc Muted™

Product No.: LT1205

- -
- -
Product No.LT1205
Clone
5C4.B8
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
PD1; PD-1; CD279; SLEB2; hPD-1; hPD-l; hSLE1
Isotype
Human IgG1κ
Applications
B
,
FA
,
FC
,
IHC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
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.
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 Nivolumab 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 ?
IHC
FA
B
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 Nivolumab. Clone 5C4.B8 binds to the extracellular portion of Human/Cynomolgus PD-1 and does not bind to other IgG superfamily proteins. This product is for research use only.
Background
Programmed cell death protein 1 (PD-1) is a protein on the surface of cells that plays a role in the maintenance of self-tolerance. PD-1 promotes self-tolerance via the down-regulation of the immune system which results in the suppression of T cell inflammatory activity. PD-L1 and PD-L2 are the two ligands known to bind PD-1. PD-L1 has increased expression in several cancers.1 PD-L2 has a more limited expression and is primarily expressed by dendritic cells and only some tumor lines. Inhibition of the interaction of PD-1 with its ligands can function as an immune checkpoint blockade through the improvement of In vitro T-cell responses and via the mediation of anti-tumor activity.2 Nivolumab disrupts the negative signal that is responsible for T-cell activation and proliferation by binding to PD-1 on activated immune cells to selectively block the interaction of the PD-1 receptor with its ligands.3 Emerging research suggests that combined blockade of PD-1 and CTLA-4, with nivolumab and ipilimumab respectively, could produce greater antitumor activity than blockade of either pathway alone.4 This cost-effective, research-grade Anti-Human CD279 (PD-1) (Nivolumab) utilizes the same variable regions from the therapeutic antibody Nivolumab making it ideal for research projects.
Antigen Distribution
PD-1 is expressed on a subset of CD4-CD8- thymocytes, and on activated T and B cells.
Ligand/Receptor
PD-L1 and PD-L2
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars
.
Costimulatory Molecules
.
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.

In a pharmacokinetic (PK) bridging ELISA, research-grade Nivolumab biosimilars are used as calibration standards or reference controls to measure drug concentration in serum samples in several key ways:

Calibration Standards

  1. Standard Preparation: Nivolumab biosimilars are prepared in a range of concentrations, typically spanning from low to high ng/mL levels, to serve as calibration standards. These standards are used to establish a calibration curve, which allows for the quantification of Nivolumab in serum samples.

  2. Calibration Curve Establishment: The calibration curve is often established using a mathematical model, such as a polynomial function, to ensure a good fit of the data. Parameters like the coefficient of determination (R²) and p-value are used to validate the model's accuracy.

Reference Controls

  1. Reference Material: Biosimilars can be used as reference products to compare with the test biosimilar. This ensures that the PK assay is capable of accurately measuring both the reference and test products.

  2. Pharmacokinetic Profiling: By using biosimilars as reference controls, researchers can assess the pharmacokinetic profiles of different formulations, comparing their absorption, distribution, metabolism, and excretion (ADME) characteristics. This is crucial for demonstrating bioequivalence between biosimilars and reference products.

Bioanalytical Strategy

  1. Single Assay Method: A single PK assay is preferred for measuring both biosimilar and reference products due to its reduced variability and the elimination of crossover analysis in clinical studies.

  2. Validation and Verification: The assay's performance is validated for accuracy, precision, and robustness, ensuring reliable concentration measurements. This validation is essential for regulatory compliance and for supporting PK bioequivalence assessments.

By employing Nivolumab biosimilars in this manner, researchers can accurately measure drug concentrations in serum, facilitate the development of PK assays, and ultimately support the approval of biosimilar drugs.

The primary syngeneic tumor models used for in vivo anti-PD-1 antibody research to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes include several well-established mouse cancer cell lines that maintain functional immune systems for immunotherapy evaluation.

Syngeneic Tumor Models

MC38 Colorectal Adenocarcinoma is one of the most extensively used models for anti-PD-1 research. This model demonstrates sensitivity to anti-PD-1 treatment and has been used to develop resistant variants through serial passaging in treated mice. The MC38 model is particularly valuable because it responds well to various cancer immunotherapies including anti-PD-1 antibodies.

Melanoma Models represent another critical category, with multiple variants used in anti-PD-1 studies. These include established melanoma cell lines that allow researchers to evaluate the antitumor efficacy of anti-PD-1 antibodies and study combination therapies. Melanoma models have been particularly useful for investigating how PPT1 inhibition can enhance anti-PD-1 antibody effectiveness.

Bladder Cancer Models such as MB49 and MBT2 have been developed with acquired resistance to anti-PD-1 and/or PD-L1 antibodies through in vivo serial treatment cycles. These models provide insights into resistance mechanisms and allow characterization of immune infiltrate changes.

Additional Models include RENCA kidney cancer model and various other tumor types. The search results also mention Hepa1-6, CT26, and EMT-6 as part of selected models showing distinctive tumor-immunity interactions.

Model Characteristics and Applications

These syngeneic models are fully characterized for gene expression profiles, baseline tumor-infiltrating lymphocyte populations, and responses to common immune checkpoint inhibitors. The models enable researchers to evaluate how anti-PD-1 treatment affects various immune cell populations within the tumor microenvironment.

Resistance Model Development has been a key application, where researchers create resistant variants by subjecting sensitive models like MC38 to serial treatment and reimplantation cycles. These resistant models show altered tumor immune microenvironments with changes in specific lymphoid and myeloid subpopulations.

Immune Microenvironment Analysis using these models reveals how anti-PD-1 treatment influences tumor-associated macrophage polarization, myeloid-derived suppressor cell infiltration, and T cell functionality. Spectral cytometry and RNA sequencing are commonly used to characterize these immune changes.

The syngeneic approach is preferred over humanized models for anti-PD-1 research because it maintains a fully functional immune system while allowing for reproducible tumor growth and immune response characterization. These models provide predictive value for clinical efficacy and serve as robust platforms for optimizing therapeutic strategies before advancing to clinical trials.

Rationale for Combination Therapy

Researchers are actively investigating the use of nivolumab biosimilars—molecules highly similar to the originator drug nivolumab, targeting the PD-1 immune checkpoint—in combination with other checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) to assess synergistic effects in immune-oncology models. The central hypothesis is that targeting multiple immune checkpoints can overcome the limitations of single-agent immunotherapy, such as primary and acquired resistance, and improve clinical outcomes for patients with complex cancers.

Scientific Basis and Mechanisms

  • CTLA-4 and PD-1/PD-L1 Blockade: CTLA-4 inhibition primarily acts in lymphoid tissues to enhance T-cell priming, while PD-1/PD-L1 blockade primarily functions at the tumor site to prevent T-cell suppression by the tumor microenvironment. Combining these agents can thus amplify immune activation at both the induction and effector phases of the immune response.
  • Preclinical and Clinical Evidence: In melanoma, combination therapy with the original nivolumab (anti-PD-1) and ipilimumab (anti-CTLA-4) demonstrated greater efficacy than either agent alone, especially in patients with PD-L1-negative tumors. However, toxicity (grade 3–4 adverse events) is significantly higher with combination therapy, necessitating careful management.
  • Emerging Targets: While less mature, combinations with anti-LAG-3 agents are also being explored. The logic remains similar—blocking multiple inhibitory signals may further unleash anti-tumor immunity, but this must be balanced against increased risk of immune-related side effects.

Use of Biosimilars in Research

Preclinical Models

  • In Vitro and In Vivo Synergy Studies: Biosimilars of nivolumab, such as BA1104, are rigorously compared to the reference product for pharmacokinetics, safety, and efficacy in both animal models and early-phase human trials. Once biosimilarity is established, these agents can be used interchangeably in complex immune-oncology models.
  • Combination Screening: Researchers use biosimilars in high-throughput screens to test combinations with other checkpoint inhibitors (e.g., CTLA-4 or LAG-3 biosimilars), cytokine therapies, or targeted agents. These screens help identify which combinations are most synergistic and which might have overlapping or unique toxicities.

Clinical Trials

  • Phase III Trials: Biosimilar nivolumab is entering late-stage clinical trials in combination with chemotherapy and, by extrapolation, could be combined with other checkpoint inhibitors in analogous studies.
  • Indicator Extrapolation: According to regulatory guidelines, once biosimilarity is proven in key clinical trials, the biosimilar can be approved for all indications of the reference product, including combination regimens.
  • Comparative Studies: Ongoing trials compare biosimilar nivolumab + ipilimumab (anti-CTLA-4) versus the originator combination to assess non-inferiority in efficacy and safety, ensuring that cost savings do not compromise patient outcomes.

Key Findings and Challenges

  • Enhanced Efficacy: Combination therapy often yields higher response rates and prolonged progression-free and overall survival compared to monotherapy in select cancers, as seen in melanoma studies.
  • Increased Toxicity: The main drawback is the higher incidence of severe immune-related adverse events, which must be managed with corticosteroids or other immune modulators.
  • Patient Selection: Researchers are working to identify biomarkers (e.g., tumor mutational burden, PD-L1 status) that predict which patients are most likely to benefit from combination therapy, thereby optimizing risk-benefit ratios.

Future Directions

  • Novel Combinations: Trials are exploring nivolumab biosimilar + anti-LAG-3, or triple combinations (e.g., PD-1 + CTLA-4 + LAG-3 blockade), to further augment anti-tumor immunity.
  • Personalized Approaches: Integration of biosimilars into adaptive trial designs allows for real-time assessment of combinatorial efficacy and safety in diverse patient populations.
  • Global Access: The development of biosimilars may democratize access to these expensive but potentially life-saving combinations, particularly in resource-limited settings.

In summary, researchers use nivolumab biosimilars in combination with other checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) to study synergistic anti-tumor effects in complex immune-oncology models, leveraging both preclinical and clinical trial frameworks. These studies aim to optimize efficacy, understand and manage toxicity, and expand access to advanced cancer immunotherapies worldwide.

In bridging ADA ELISA assays for monitoring immune responses against nivolumab, the biosimilar serves as both the capture and detection reagent in a specialized immunoassay format designed to detect anti-drug antibodies (ADAs) in patient samples.

Bridging ELISA Principle for Nivolumab ADA Detection

The bridging ELISA format utilizes nivolumab (or its biosimilar) in a dual capacity to create a "bridge" that captures and detects bivalent anti-drug antibodies. In this assay design, biotinylated nivolumab is captured on streptavidin-coated plates, and anti-drug antibodies in patient samples bind to this captured drug. The detection is then accomplished using a dye or HRP-labeled version of the same nivolumab molecule.

Assay Configuration and Methodology

Capture Phase: The biotinylated nivolumab biosimilar is immobilized on streptavidin-coated microtiter plates, creating a solid-phase capture surface. When patient serum or plasma samples are added, any anti-nivolumab antibodies present will bind to the immobilized drug.

Detection Phase: After washing away unbound components, a peroxidase-labeled nivolumab conjugate is added to each well. This labeled version of the drug acts as the detection reagent, binding to any captured anti-nivolumab antibodies that have at least two binding sites (bivalent antibodies), effectively creating a "bridge" between the capture and detection reagents.

Signal Generation: The bound enzymatic activity is detected through the addition of tetramethylbenzidine (TMB) chromogen-substrate, where the intensity of the reaction color is directly proportional to the concentration of anti-nivolumab antibodies in the sample.

Clinical Significance and Applications

This bridging format is particularly valuable because the formation of anti-drug antibodies induced by nivolumab treatment has been associated with loss of response, hypersensitivity reactions, and severe therapy-limiting side effects. The assay allows for quantitative determination of anti-nivolumab antibodies in human serum and plasma as an analytical tool for monitoring immunogenicity.

Advantages and Considerations

The bridging ELISA technique offers high sensitivity and allows high-throughput sample screening. However, the specificity may be limited in complex matrices like human serum due to matrix components, soluble target molecules, or residual drug components that can interfere with the assay. Therefore, using high-quality assay reagents and appropriate blocking solutions is crucial for obtaining meaningful results in clinical immunogenicity testing.

This methodology represents an innovative approach where the same therapeutic molecule (nivolumab or its biosimilar) serves dual functions, making it an efficient and specific method for detecting immune responses against the drug in treated patients.

References & Citations

1. Minato, N. et al. (2002) Proc Natl Acad Sci U S A. 99(19): 12293–97.
2. Korman, AJ. et al. (2014) Cancer Immunol Res. 2(9):846-56.
3. Li, Y. et al. (2016) MAbs. 8(5):951-60.
4. Wolchok, JD. et al. (2013) N Engl J Med 369(2):122-33.
B
FA
Flow Cytometry
IHC

Certificate of Analysis

Formats Available

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