Anti-Human PD-1 (Pembrolizumab) [Clone MK-3475]

Anti-Human PD-1 (Pembrolizumab) [Clone MK-3475]

Product No.: LT240

- -
- -
Product No.LT240
Clone
MK-3475
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anti PD-1, PDCD1, CD279, lambrolizumab
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
Active
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
FC,
FA,
ELISA,
WB,
IP
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 Pembrolizumab. This product is for research use only. Pembrolizumab (lambrolizumab) activity is directed against human PD-1.
Background
PD-1 is a transmembrane protein in the CD28/CTLA-4 subfamily of the Ig superfamily1, 2. When stimulated via the T cell receptor (TCR), Tregs translocate PD-1 to the cell surface3. 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 ligands1. 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 cells3.

PD-1 is an immune checkpoint receptor that suppresses cancer-specific immune responses4. 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 glycolysis2. 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.

Pembrolizumab was generated as a humanized monoclonal antibody by grafting the variable region sequences of a mouse anti-human PD-1 antibody onto a human IgG4-κ isotype framework containing a stabilizing S228P Fc mutation5, 6. Pembrolizumab shows high affinity for the PD-1 receptor and prevents PD-1 binding to ligands PD-L1 and PD-L2. Additionally, pembrolizumab strongly inhibits PD-L1 and PD-L2 and has robust activity in a functional ex vivo T cell modulation assay using human donor blood cells.

Pembrolizumab is used in adult and pediatric patients to treat unresectable or metastatic solid tumors with certain genetic abnormalities7. Binding of pembrolizumab to PD-1 does not engage Fc receptors or activate complement and therefore is devoid of cytotoxic activity8.
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.

Use of Research-Grade Pembrolizumab Biosimilars in PK Bridging ELISA

Research-grade pembrolizumab biosimilars can play a crucial role as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs designed to measure drug concentration in serum samples, particularly when bridging biosimilar and reference product (Keytruda®) PK data.

Calibration Standards in PK ELISAs

In a typical PK ELISA, calibration standards are solutions with known concentrations of the analyte (here, pembrolizumab or its biosimilar) used to generate a standard curve. This curve allows quantification of unknown concentrations in patient serum samples based on the optical density (OD) signal generated in the assay. The accuracy and precision of the assay depend heavily on the quality and consistency of the calibration standards.

Advantages of Using a Single Biosimilar Standard

Current best practice in biosimilar development is to use a single PK assay with a single analytical standard for quantifying both the biosimilar and reference product in serum samples. This approach reduces variability that would arise from using separate assays or standards for each product and eliminates the need for crossover analysis in blinded clinical studies. The biosimilar is first rigorously compared to the reference product in qualification studies to ensure bioanalytical equivalence; if equivalence is demonstrated, the biosimilar can serve as the analytical standard for the assay.

Validation and Quality Control

The PK assay is validated for precision, accuracy, sensitivity, and specificity using a comprehensive set of calibration standards (e.g., concentrations ranging from 50 to 12,800 ng/mL in human serum). Quality control (QC) samples are prepared using both the biosimilar and reference product and quantified against the biosimilar standard curve to confirm analytical equivalence. The statistical evaluation of these data, typically comparing the 90% confidence interval to a predefined equivalence interval (e.g., [0.8, 1.25]), ensures the assay is fit for purpose and minimizes confounding variability in PK similarity studies.

Practical Implementation

Steps in a PK Bridging ELISA:

  1. Qualification: Compare the biosimilar and reference product within the assay to confirm bioanalytical equivalence.
  2. Validation: Validate the assay using the biosimilar as the primary calibration standard, testing performance across a wide concentration range in human serum.
  3. Sample Analysis: Quantify unknown patient samples against the biosimilar-generated standard curve.
  4. Quality Control: Use both biosimilar and reference product QC samples to monitor assay performance over time.

Specificity and Cross-Reactivity

The specificity of the assay is critical: the calibration standard (biosimilar) should not cross-react with irrelevant proteins or other therapeutic antibodies (e.g., nivolumab, trastuzumab, human IgG4, human PD-1). The pembrolizumab biosimilar used must closely mimic the structure and binding properties of the reference product to avoid bias in quantification.

Regulatory Considerations

Assays must comply with regulatory guidelines (e.g., EMA/FDA, ICH) for biological assays, ensuring adequate sensitivity, precision (intra- and inter-assay CVs <15%), and robustness. The limit of quantification (LOQ) and limit of detection (LOD) must be established and meet clinical requirements for the intended use.

Summary Table: Key Aspects of Biosimilar Use in PK Bridging ELISA

AspectBiosimilar as StandardRegulatory Requirement
Calibration CurveBiosimilar used to generate standard curveMust demonstrate bioanalytical equivalence
QC SamplesBoth biosimilar and reference product used for QCMonitor assay performance
SpecificityNo cross-reactivity with irrelevant proteins/antibodiesValidated per regulatory guidelines
PrecisionIntra- and inter-assay CVs <15%Meets ICH/FDA/EMA standards
SensitivityLOQ ~8.75 ng/mL, LOD <7.95 ng/mL (example values)Fit for clinical PK studies

Conclusion

Research-grade pembrolizumab biosimilars are used as calibration standards or reference controls in PK bridging ELISAs after demonstrating bioanalytical equivalence to the reference product. This single-standard approach minimizes variability, streamlines PK similarity assessment, and supports regulatory submissions for biosimilar development. The biosimilar must be rigorously validated for specificity, precision, and sensitivity, and the assay must meet all regulatory requirements for quantitative bioanalysis.

Primary Models for Studying Anti-PD-1 Antibody Effects

To study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) following the administration of a research-grade anti-PD-1 antibody, researchers commonly use two main in vivo models: syngeneic models and humanized models.

Syngeneic Models

  1. Definition: Syngeneic models involve transplanting tumor cells from the same species into a host animal, ensuring genetic identity between the tumor and the host's immune system. This allows for a fully functional immune response against the tumor.

  2. Tumor Models: These models include various cancer types such as melanoma, colon adenocarcinoma (e.g., MC38), and others. The MC38 model is particularly useful for studying resistance to anti-PD-1 treatment, as it can be passaged to develop resistance, simulating clinical scenarios where tumors become unresponsive to therapy.

  3. Advantages: Syngeneic models enable the study of immune checkpoint therapies like anti-PD-1 in a controlled, immunocompetent environment. They are widely used for preclinical studies to assess the efficacy of novel immunotherapies.

  4. Example Application: A study using a syngeneic model demonstrated that inhibiting palmitoyl-protein thioesterase 1 (PPT1) can enhance the antitumor activity of anti-PD-1 antibodies in melanoma by promoting T cell-mediated killing and reducing immune suppressive cells in the tumor microenvironment.

Humanized Models

  1. Definition: Humanized models are used to study human cancers in a more relevant context. These models typically involve transplanting human tumor cells into immunocompromised mice (e.g., SCID or NOD-SCID mice) to allow the growth of human tumors.

  2. Tumor Models: Human tumors from various types of cancer, such as breast, lung, and colon, can be xenografted into these mice. However, to study human immune responses, additional human immune cells need to be introduced into the model.

  3. Advantages: Although these models lack a fully functional native immune system, they can be modified to include human immune cells, allowing researchers to study human tumor interactions with human immune components.

  4. Example Application: While not specifically mentioned in the search results, humanized models are critical for studying the effects of anti-PD-1 therapies on human tumors with human immune cells. This approach helps in understanding how human TILs interact with tumors under the influence of checkpoint inhibitors.

Both syngeneic and humanized models are essential tools for investigating the effects of anti-PD-1 therapies on tumor growth and the immune microenvironment.

Researchers employ pembrolizumab biosimilars in combination with other checkpoint inhibitor biosimilars to investigate synergistic mechanisms and overcome the limitations of monotherapy approaches in immune-oncology research. These combination strategies are based on the understanding that different checkpoint inhibitors target distinct pathways in the immune system, potentially enhancing overall anti-tumor efficacy.

Mechanistic Rationale for Combination Studies

The combination of pembrolizumab biosimilars with other checkpoint inhibitors operates on the principle that multiple immune checkpoints have different mechanisms of action and sites of activity. Anti-CTLA-4 agents primarily function in the lymph node compartment, where they restore the induction and proliferation of activated T cells, while anti-PD-1 agents like pembrolizumab biosimilars mainly act at the periphery of the tumor site, preventing the neutralization of cytotoxic T cells by PD-L1-expressing tumor and plasmacytoid dendritic cells in the tumor microenvironment.

PD-1/CTLA-4 Combination Research

Researchers utilize pembrolizumab biosimilars alongside anti-CTLA-4 biosimilars to study how these agents can increase each other's activity and overcome individual monotherapy limitations. The combination has demonstrated significant antitumor efficacy in preclinical models. In research applications, scientists use pembrolizumab biosimilars that contain the same variable regions as the therapeutic antibody, making them ideal for investigating these synergistic mechanisms.

Studies have revealed that the effectiveness of PD-1/CTLA-4 combinations varies based on PD-L1 expression levels. In patients with PD-L1-negative tumors, the combination showed superior progression-free survival (11.2 months) compared to PD-1 monotherapy (5.3 months), while patients with PD-L1-positive tumors showed similar outcomes regardless of combination therapy.

LAG-3/PD-1 Biosimilar Combinations

Researchers are increasingly exploring combinations of pembrolizumab biosimilars with anti-LAG-3 agents to study more targeted immune responses. LAG-3 is a co-inhibitory receptor expressed on exhausted tumor-infiltrating lymphocytes (TILs) with reduced effector functions. The combination strategy focuses on the observation that LAG-3 and PD-1 are often co-expressed at high levels on infiltrating TILs, making them attractive targets for combination therapy.

Advantages of LAG-3/PD-1 Combinations

This combination approach offers several research advantages. LAG-3 blockade may produce milder side effects compared to currently used checkpoint inhibitors, as demonstrated in preclinical models where autoimmunity development was slower and less penetrant than with CTLA-4 deficient models. The high-level co-expression of LAG-3 and PD-1 on tumor-infiltrating lymphocytes suggests that combination therapy may encourage tumor-specific responses while avoiding non-specific or self-antigen specific immune responses.

Research Applications and Model Systems

Researchers employ pembrolizumab biosimilars in various experimental settings to study these synergistic effects. The biosimilars react with human PD-1 (CD279), a 50-55 kDa cell surface receptor that belongs to the CD28 family of the immunoglobulin superfamily. These research-grade biosimilars enable scientists to investigate how PD-1 binding blocks the interaction with PD-L1 and PD-L2, releasing PD-1 pathway-mediated inhibition of immune responses and restoring T-cell immune surveillance of tumors.

In complex immune-oncology models, researchers use these combinations to study how different checkpoint pathways interact within the tumor microenvironment, evaluate biomarkers for patient selection, and assess the balance between enhanced efficacy and increased toxicity. The research has shown that targeting multiple checkpoints can overcome resistance mechanisms that limit single-agent therapies, providing valuable insights for translating these findings into clinical applications.

In the context of immunogenicity testing, a Pembrolizumab biosimilar can be utilized as a capture or detection reagent in a bridging ADA ELISA to monitor a patient's immune response against the therapeutic drug, Pembrolizumab. Here's how it is used:

Bridging ADA ELISA Overview

Bridging ELISA is a method used to detect anti-drug antibodies (ADAs) by employing the drug itself as both the capture and detection reagents. This technique is highly specific and sensitive, allowing for the detection of ADAs against therapeutic proteins like Pembrolizumab.

Using a Pembrolizumab Biosimilar

A Pembrolizumab biosimilar, being structurally and functionally similar to the original Pembrolizumab, can serve as a reliable substitute in the ELISA. This is particularly useful in research contexts where the therapeutic version might not be available or is not suitable for the assay.

Steps in the Bridging ELISA:

  1. Coating the Plate: The microtiter plate is coated with one form of the Pembrolizumab biosimilar, which captures the ADAs present in the patient's serum.

  2. Sample Addition: The patient's serum is added to the plate, allowing ADAs to bind to the biosimilar if present.

  3. Detection: A second form of the Pembrolizumab biosimilar, this time labeled with a reporter like HRP (horseradish peroxidase), is added. If ADAs are present, they will be sandwiched between the two forms of the biosimilar, allowing for detection.

  4. Detection via Chromogenic Substrate: A chromogenic substrate, such as TMB, is added, which reacts with the HRP enzyme to produce a color change proportional to the amount of ADAs present in the sample.

Advantages of Using a Biosimilar:

  • Cost-Effective: Biosimilars are often less expensive than the therapeutic version, making them more accessible for research purposes.
  • Similarity to Therapeutic Drug: The biosimilar can mimic the therapeutic drug's interaction with ADAs, ensuring that the assay accurately reflects the patient's immune response.

Conclusion

The use of a Pembrolizumab biosimilar in bridging ADA ELISAs provides a reliable and efficient method for monitoring a patient's immune response to Pembrolizumab, by leveraging the structural similarity of the biosimilar to the therapeutic drug.

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. Hamid O, Robert C, Daud A, et al. N Engl J Med. Jul 11;369(2):134-144. 2013.
6. Patnaik A, Kang SP, Rasco D, et al. Clin Cancer Res. 21(19):4286-4293. 2015.
7. Marcus L, Fashoyin-Aje LA, Donoghue M, et al. Clin Cancer Res. 27(17):4685-4689. 2021.
8. Kwok G, Yau TC, Chiu JW, et al. Hum Vaccin Immunother. 12(11):2777-2789. 2016.
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.