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

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

Product No.: LT245

- -
- -
Product No.LT245
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
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 (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.

Using Research-Grade Pembrolizumab Biosimilars in PK Bridging ELISA

Introduction to PK Bridging ELISA

Pharmacokinetic (PK) bridging studies are crucial for demonstrating the bioequivalence of biosimilars and reference drugs. In the context of Pembrolizumab (Keytruda), a monoclonal antibody used in cancer treatment, PK bridging ELISA plays a significant role. This section outlines how research-grade Pembrolizumab biosimilars are utilized as calibration standards or reference controls in such assays.

Role of Biosimilars as Calibration Standards

  1. Quantification and Calibration:

    • Purpose: In PK bridging studies, a single analytical standard is typically used to quantify both the biosimilar and the reference product. This ensures consistency and reduces variability in the assay results.
    • Biosimilar Use: Research-grade Pembrolizumab biosimilars can serve as calibration standards by providing a known concentration against which unknown samples are measured. These standards help in constructing a calibration curve necessary for quantifying drug concentrations in serum samples.
  2. Bioanalytical Equivalence:

    • Importance: Demonstrating that the biosimilar and reference product are bioanalytically equivalent is crucial. This involves comparing their concentration profiles using a single PK assay.
    • Testing Strategy: A comprehensive approach involves validating the PK assay using the biosimilar as an analytical standard. This ensures that the assay can accurately measure both products with consistent precision and accuracy.
  3. Cross-Reactivity and Specificity:

    • Cross-Reactivity: The assay should not react with other proteins that might interfere with the measurement, such as human PD-1 or other monoclonal antibodies like nivolumab.
    • Specificity: The bioanalytical method must be specific to Pembrolizumab, ensuring accurate measurement without cross-reactivity with irrelevant components in serum samples.

Implementation in ELISA

  1. ELISA Principle:

    • Sandwich ELISA: The assay typically employs a sandwich ELISA format, where the biosimilar standard (or reference) is captured by specific antibodies and quantified by detecting bound antibodies.
    • Calibration: Known concentrations of the biosimilar standard are used to create a calibration curve, allowing for the quantification of Pembrolizumab in serum samples.
  2. Precision and Accuracy:

    • Validation Parameters: The assay needs to be validated for precision (%CV), accuracy, specificity, and sensitivity. The use of a single analytical standard improves the reliability of these parameters across different samples.
    • Inter and Intra-Assay Variation: Both intra-assay and inter-assay variability should be minimized to ensure consistent results.
  3. Example Kits:

    • KRIBIOLISA: Kits like KRIBIOLISA offer a quantitative determination of anti-Pembrolizumab in human serum and plasma, using recombinant Pembrolizumab for capture and detection.
    • Assay Genie: Similarly, Assay Genie provides ELISA kits for Pembrolizumab quantification in serum and plasma, following a sandwich ELISA principle.

Conclusion

Research-grade Pembrolizumab biosimilars are crucial as calibration standards in PK bridging ELISA assays. They enable accurate quantification of drug concentrations in serum samples by providing a consistent reference point. The use of a single analytical standard enhances the reliability and comparability of data, supporting the demonstration of bioequivalence between biosimilars and reference products.

The primary in vivo models to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization following anti-PD-1 antibody administration are syngeneic mouse tumor models and, to a more limited but growing extent, humanized mouse tumor models.

Syngeneic Mouse Tumor Models:

  • The most widely used models are syngeneic, where tumor cell lines derived from the same genetic background (often C57BL/6 or BALB/c mice) are implanted into immunocompetent mice.
  • Examples include MC38 (colon adenocarcinoma), B16F10 (melanoma), and others.
  • These models allow for the administration of research-grade anti-mouse PD-1 antibodies, resulting in inhibition of tumor growth and robust analysis of TIL response, including changes in CD8+ T cell infiltration, myeloid populations, and TIL phenotype.
  • Syngeneic models are the standard for mechanistic studies of the immune response to checkpoint inhibitors, since they support a fully functional and manipulable murine immune system.

Key supporting details from the literature:

  • The MC38 colon adenocarcinoma model is commonly used for anti-PD-1 studies; serial passage in the presence of anti-PD-1 can generate resistant lines for further investigation.
  • These models facilitate comprehensive analysis of TILs, examining factors such as phenotype, exhaustion markers, and functional status.
  • Therapeutic efficacy, resistance mechanisms, and synergistic effects with other agents are readily evaluated in this context.

Humanized Mouse Models:

  • Increasingly, humanized mouse models—immunodeficient mice engrafted with human hematopoietic stem cells or PBMCs, and/or implanted with human tumors—are used for studies with clinical-grade or cross-reactive anti-PD-1 antibodies.
  • Such models allow for the study of human TILs within a controlled in vivo environment, but current limitations include incomplete human immune system reconstitution and practical/ethical concerns.
  • These models are more often used for translational testing of clinical antibodies (like pembrolizumab or nivolumab) and for studying human-specific immune responses.
Model TypeTumor Type/SpeciesPD-1 Antibody TypeTILs Characterized?Key Benefits
Syngeneic mouseMurine tumor/mouseResearch-grade anti-mouse PD-1YesFull mouse immunity, high throughput
Humanized mouseHuman tumor/humanized mouseHuman or cross-reactive PD-1Yes (human TILs)Translational human relevance

Summary:
Most preclinical studies use syngeneic mouse models with murine tumors and anti-mouse PD-1 antibodies for robust TIL profiling and tumor growth monitoring. Humanized mouse models are employed for translational studies involving human tumors and antibodies, enabling analysis of human TILs, though they are technically more challenging and less standardized.

Researchers employ pembrolizumab biosimilars in combination with other checkpoint inhibitors to investigate synergistic immune responses through carefully designed experimental approaches that leverage the distinct mechanisms of action of different checkpoint pathways.

Mechanistic Rationale for Combination Studies

The foundation for combining pembrolizumab biosimilars with other checkpoint inhibitors stems from their complementary mechanisms of action. 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 operate predominantly at the periphery of tumor sites. This spatial and temporal separation creates opportunities for synergistic effects that researchers can explore using biosimilar compounds.

Pembrolizumab biosimilars use the same variable regions as the therapeutic antibody, making them ideal for research applications where investigators need to block PD-1 interactions with its ligands PD-L1 and PD-L2. This blocking action prevents the neutralization of cytotoxic T cells by PD-L1 expressing tumor and plasmacytoid dendritic cells in the tumor microenvironment.

Research Applications in Complex Models

Non-Small Cell Lung Cancer Studies

Researchers have demonstrated the potential for combination approaches in NSCLC models, where pembrolizumab combined with NK cell infusion significantly improved overall survival and progression-free survival compared to pembrolizumab alone. These studies revealed that pembrolizumab reversed high PD-1 expression, increased IFN-gamma secretion, and enhanced immune function while reducing circulating tumor cells and tumor markers.

Melanoma Research Models

The combination of CTLA-4 and PD-1/PD-L1 blockade has shown particular promise in melanoma research, where preclinical models demonstrated enhanced antitumor efficacy. Clinical translation of these findings, such as in the CheckMate 067 trial, revealed that patients with PD-L1-negative tumors showed longer progression-free survival when treated with combined ipilimumab and nivolumab compared to single-agent therapy.

Experimental Design Considerations

Overcoming Monotherapy Limitations

Researchers design combination studies based on the principle that targeting multiple checkpoints can overcome the limitations of individual therapies. The logic centers on the different mechanisms of action, where blockade of one pathway often results in increased activity and upregulation of other inhibitory pathways - an effect that combined therapy can potentially mitigate.

Biomarker-Driven Approaches

Studies incorporating pembrolizumab biosimilars often focus on PD-L1 expression as a predictive biomarker. Research in cervical cancer models has identified PD-1-positive T cells in tumor stroma and high PD-L1 levels on tumor cell surfaces, providing rationale for combination approaches. Similarly, hepatocellular carcinoma studies have associated treatment response with PD-L1 presence in patient subsets.

Challenges and Optimization Strategies

Toxicity Management

A critical consideration in combination studies is the increased toxicity profile, particularly grade 3 or 4 adverse events associated with multi-agent checkpoint inhibition. Researchers must carefully balance efficacy gains against safety concerns when designing experimental protocols.

Patient Selection and Treatment Sequencing

Research indicates that combination benefits may be most pronounced in specific patient populations. For instance, patients with PD-L1-negative tumors appear to derive greater benefit from combination therapy compared to those with PD-L1-positive disease. This finding guides researchers in developing more targeted experimental approaches.

The use of pembrolizumab biosimilars in combination research continues to evolve as investigators explore novel checkpoint targets and optimize treatment sequences to maximize therapeutic synergy while maintaining acceptable safety profiles in complex immune-oncology models.

In immunogenicity testing for pembrolizumab therapy, biosimilar antibodies serve as critical reagents in bridging ELISA assays designed to detect anti-drug antibodies (ADAs) that patients may develop against the therapeutic drug. This approach leverages the identical binding characteristics of biosimilars while providing a cost-effective and reliable method for monitoring immune responses.

Bridging ELISA Design for Pembrolizumab ADA Detection

The bridging ELISA format represents the gold standard for ADA detection, utilizing pembrolizumab biosimilars in both capture and detection roles. In this configuration, the biosimilar antibody is immobilized on the microtiter plate surface to capture any ADAs present in patient serum samples. The captured ADAs are then detected using a labeled version of the same pembrolizumab biosimilar, creating a "bridge" formation where the ADA simultaneously binds to both the capture and detection antibodies.

Capture Phase Implementation

During the capture phase, pembrolizumab biosimilar is coated onto ELISA plate wells at optimized concentrations. Patient serum samples are then added, allowing any anti-pembrolizumab antibodies to bind specifically to the immobilized biosimilar. The biosimilar's identical variable regions to the therapeutic pembrolizumab ensure that it captures the same spectrum of ADAs that would interact with the actual therapeutic drug.

Detection and Signal Generation

For detection, a labeled pembrolizumab biosimilar (typically conjugated with horseradish peroxidase or biotin) is added to bind to the captured ADAs. When ADAs are present, they form a bridge between the capture and detection antibodies, enabling signal generation through chromogenic substrates like 3,3',5,5'-tetramethylbenzidine (TMB). This bridging configuration provides high specificity since only antibodies capable of binding to pembrolizumab will generate a positive signal.

Advanced Applications and Modifications

Immune Complex Analysis

Modern immunogenicity testing has evolved beyond detecting free ADAs to include circulating immune complexes formed between pembrolizumab and patient antibodies. Bridging ELISAs can be modified to capture these drug-ADA complexes, providing a more comprehensive view of the patient's immune response. This approach is particularly valuable since immune complexes may persist longer in circulation and provide different clinical implications compared to free ADAs.

Drug Tolerance Strategies

High concentrations of circulating pembrolizumab can interfere with ADA detection by competing with the capture antibody for ADA binding sites. To address this challenge, acid dissociation pretreatment can be combined with the bridging ELISA to separate drug-bound ADAs from free drug, enabling accurate quantification even in the presence of high therapeutic drug concentrations.

Clinical Monitoring Applications

The pembrolizumab biosimilar-based bridging ELISA provides quantitative measurements of ADA levels with typical detection limits around 0.39 ng/mL and linear ranges extending to 50 ng/mL. This sensitivity enables clinicians to monitor the development of immunogenicity over the course of treatment and correlate ADA levels with clinical outcomes such as reduced therapeutic efficacy or increased adverse reactions.

The assay format also allows for characterization of ADA specificity, determining whether patient antibodies target the antigen-binding regions, constant domains, or other specific epitopes of pembrolizumab. This information proves valuable for understanding the mechanism of immunogenicity and potential clinical implications for individual patients receiving pembrolizumab 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. 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.