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

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

Product No.: LT2205

- -
- -
Product No.LT2205
Clone
REGN2810
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CD279, PD1, REGN-2810, 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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8° C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
WP,
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 Cemiplimab. This product is for research use only. Cemiplimab 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.

Cemiplimab is a fully human, hinge-stabilized (S228P) high affinity anti-PD-1 antibody that potently blocks PD-1 interaction with PD-L1 and PD-L2 ligands and enhances human primary T-cell responses in vitro5. Cemiplimab was generated using VelocImmune knock-in mice immunized with recombinant human PD-1-mFc protein containing the PD-1 extracellular domain (amino acids 1-167) and the Fc portion of mouse IgG2a. Splenocyte-derived hybridomas were screened for human monoclonal antibody reactivity to recombinant human PD-1-hFc (extracellular domain of human PD-1 fused to human IgG1 Fc).

Cemiplimab is the first approved treatment in the United States and EU for patients with locally advanced or metastatic cutaneous squamous cell carcinoma who are not candidates for curative surgery or radiotherapy6.
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.

In a pharmacokinetic (PK) bridging ELISA, using research-grade Cemiplimab biosimilars as calibration standards or reference controls involves several key steps to ensure accurate measurement of drug concentration in serum samples.

Using Biosimilars as Calibration Standards

  1. Development of a Single PK Assay: The optimal approach is to develop a single PK assay that uses a single analytical standard for both the biosimilar and reference products. This method reduces variability and eliminates the need for crossover analysis in clinical studies.

  2. Bioanalytical Comparability: Establishing bioanalytical comparability between the biosimilar and the reference product is crucial. This involves method qualification studies to ensure precision and accuracy, followed by validation using a single analytical standard.

  3. Calibration and Validation: The biosimilar can be selected as the analytical standard for the single method if bioanalytical equivalence is demonstrated. This involves comparing the 90% confidence interval to pre-defined equivalence intervals, such as [0.8, 1.25], to conclude bioanalytical equivalence.

Controls in ELISA for Drug Concentration Measurement

  • Negative Controls: Include serum samples that do not contain Cemiplimab to check for non-specific binding and false positives.
  • Standard Curve: Use a sample with a known concentration of Cemiplimab to generate a standard curve, allowing for accurate quantification of drug concentrations in serum samples.

Cross-reactivity Considerations

  • Cemiplimab ELISA: The assay may detect other anti-PD-1 mAbs like pembrolizumab and nivolumab due to cross-reactivity. Using anti-idiotypic blocking antibodies can minimize this issue by specifically blocking the binding of unwanted mAbs to PD-1.
  • Assay Specificity: Ensure that the ELISA is optimized to specifically quantify Cemiplimab when other anti-PD-1 therapies are present, using blocking antibodies if necessary.

By following these strategies, research-grade Cemiplimab biosimilars can effectively serve as calibration standards or reference controls in PK bridging ELISA assays, enabling accurate measurement of drug concentrations in serum samples.

Primary Models for Studying Anti-PD-1 Antibody Effects In Vivo

When investigating tumor growth inhibition and the characterization of tumor-infiltrating lymphocytes (TILs) using research-grade anti-PD-1 antibodies, two major types of in vivo models are commonly employed: syngeneic tumor models and humanized mouse models.

Syngeneic Mouse Tumor Models

Syngeneic models are widely used both for mechanistic studies and preclinical evaluation of immunotherapy combinations. These models involve implanting murine cancer cells (e.g., MC38 colorectal adenocarcinoma) into immunocompetent mice of the same genetic background, allowing for an intact immune system and physiologically relevant immune responses.

  • MC38 Colon Adenocarcinoma Model: This model is particularly prominent in immunotherapy research. Studies have demonstrated its sensitivity to anti-PD-1 treatment, with resulting tumor growth delay and immune cell infiltration that can be readily analyzed. The model has also been adapted to study resistance mechanisms by serially passaging tumors in the presence of anti-PD-1, generating lines refractory to treatment for further mechanistic exploration.
  • Melanoma Models: Syngeneic models such as B16 melanoma have been used to study the effects of anti-PD-1 antibodies, including the impact on TIL phenotypes, macrophage polarization, and myeloid-derived suppressor cell infiltration. For example, combination therapy with PPT1 inhibitors (e.g., hydroxychloroquine) and anti-PD-1 has been shown to enhance antitumor effects and alter the tumor immune microenvironment, allowing detailed characterization of TILs and myeloid populations.
  • Other Cancers: Syngeneic models are available for a range of cancer types, enabling researchers to assess the differential effects of anti-PD-1 therapy across tumor microenvironments.

In these models, tumor growth and TILs (including CD4+ and CD8+ T cell subsets, exhausted T cells, and immune checkpoint molecule expression) are routinely assessed by flow cytometry, immunohistochemistry, and functional assays.

Humanized Mouse Models

Humanized models (e.g., NSG/NOG mice engrafted with human immune cells and human tumor xenografts) are increasingly utilized to study human-specific immune responses and the efficacy of human-targeted immunotherapies.

  • Preclinical Evaluation of Human Antibodies: These models are essential for testing human anti-PD-1 antibodies (e.g., pembrolizumab, nivolumab) in a context that more closely mimics the human immune system. However, the search results provided do not detail specific studies using humanized models for anti-PD-1 research, likely because the focus of the referenced articles is on syngeneic systems.
  • Limitations and Advantages: Humanized models allow for the study of human TILs and their response to therapy but are more complex and expensive, with potential limitations in fully recapitulating human tumor-immune interactions.

Comparative Table: Syngeneic vs. Humanized Models

Model TypeImmune SystemTumor OriginPrimary Use CaseKey AdvantagesLimitations
Syngeneic (e.g., MC38, B16)Mouse (intact)MouseMechanistic studies, combination therapies, resistanceIntact immune system, reproducible, cost-effectiveLimited to murine biology
Humanized (e.g., NSG/NOG-Hu)Human (engrafted)HumanPreclinical testing of human-targeted antibodiesHuman immune components, translatabilityComplex, expensive, incomplete reconstitution

Summary

  • Syngeneic mouse tumor models (e.g., MC38 colon adenocarcinoma, B16 melanoma) are the primary in vivo systems for administering research-grade anti-PD-1 antibodies, studying tumor growth inhibition, and characterizing TILs in the context of an intact immune system.
  • Humanized mouse models are specialized systems for testing human antibodies but are not highlighted in the provided search results; their use is more common in industry and translational research where human-specific interactions are crucial.
  • Both model types enable the assessment of TIL phenotypes, functional states, and the broader tumor immune microenvironment, but syngeneic models remain the gold standard for basic and translational immunotherapy research in academic settings.

Researchers are exploring the use of Cemiplimab biosimilars in conjunction with other checkpoint inhibitors, such as anti-CTLA-4 or anti-LAG-3 biosimilars, to study synergistic effects in complex immune-oncology models. Here's how this works and the potential benefits:

Synergistic Effects

  1. Combining Checkpoint Inhibitors: The use of Cemiplimab (a PD-1 inhibitor) biosimilars alongside other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 can enhance anti-tumor immunity. While PD-1 inhibitors alone can block PD-1 interaction with its ligands (PD-L1 and PD-L2), thereby preventing T-cell inhibition, combining them with other inhibitors can further amplify immune responses.

  2. Mechanism of Action:

    • PD-1 Inhibitors: Block the interaction between PD-1 and its ligands, enhancing T-cell activation and proliferation.
    • CTLA-4 Inhibitors: Interfere with the CTLA-4 checkpoint to prevent it from inhibiting T-cell activation, thus enhancing immune responses.
    • LAG-3 Inhibitors: Target the LAG-3 receptor, which also inhibits T-cell activity, and their use can enhance anti-tumor effects when combined with PD-1 inhibitors.
  3. Synergistic Studies: In preclinical models, combining PD-1 with LAG-3 inhibitors has shown significant antitumor efficacy compared to using either inhibitor alone. This approach can potentially overcome resistance mechanisms and improve treatment outcomes in cancers like melanoma and squamous cell carcinoma.

  4. Research Use: The Cemiplimab biosimilar, which mimics the variable regions of the therapeutic Cemiplimab, is ideal for research purposes to study these synergies in a controlled setting.

Conducting Synergistic Studies

To study these synergistic effects, researchers typically follow these steps:

  • Model Selection: Choose appropriate preclinical models that mimic human cancers, such as melanoma or squamous cell carcinoma.
  • Treatment Design: Design experiments where animals or human samples are treated with either single checkpoint inhibitors or combinations of them (e.g., PD-1 + LAG-3 or PD-1 + CTLA-4).
  • Outcome Analysis: Evaluate the efficacy of these treatments by analyzing tumor growth, survival rates, and immune response markers.
  • Data Interpretation: Compare the outcomes of combination therapies with monotherapies to identify synergistic effects.

Potential Benefits

  • Enhanced Efficacy: Combination therapies can lead to higher response rates and better disease control compared to single-agent treatments.
  • Overcoming Resistance: By targeting multiple checkpoints, these combinations may help overcome resistance mechanisms that limit the effectiveness of single checkpoint inhibitors.
  • Tailored Treatment Approaches: Understanding synergies can help develop personalized treatment strategies based on the specific immune landscape of individual patients' tumors.

Overall, using Cemiplimab biosimilars in combination with other checkpoint inhibitors offers a promising strategy for enhancing anti-tumor immunity and improving treatment outcomes in immune-oncology research.

In a bridging ADA ELISA for monitoring immunogenicity against cemiplimab (or its biosimilar), the cemiplimab biosimilar can be used as both capture and detection reagent to specifically measure anti-drug antibodies (ADA) in patient samples.

Key assay steps and rationale:

  • Bridging Assay Format:
    Patient serum potentially containing ADA is incubated with two forms of the drug:

    • One form (the biosimilar) is immobilized on the plate (capture reagent).
    • The other form (also the biosimilar), but labeled (e.g., with biotin or HRP), is added as the detection reagent.
  • ADA bridging:
    If ADA against cemiplimab is present in the sample, it can bind both the immobilized and the labeled drug, effectively "bridging" them to generate a detectable signal.

  • Why Use the Biosimilar:
    Regulatory and scientific best practice recommends using the biosimilar itself as both capture and detection reagent to ensure that any immune response measured represents antibodies specific to the biosimilar (and by extension, to cemiplimab’s unique or shared epitopes), assessing the true immunogenic potential of the marketed product.

  • Interpretation:
    The signal indicates the presence of antibodies in the patient sample capable of binding the drug. Care is taken to validate assay specificity to avoid cross-reactivity from antibodies against similar anti-PD-1 drugs, as variable regions differ among products and most ADAs are directed against these regions.

Summary Table: Cemiplimab Biosimilar Use in ADA Bridging ELISA

RoleReagentPurpose
Capture (plate-bound)Cemiplimab biosimilarBinds one arm of patient ADA
Detection (labeled)Cemiplimab biosimilarBinds other arm of ADA; label produces signal if ADA bridges reagents

Additional considerations:

  • This approach ensures detection of any novel immunogenic epitopes present on the biosimilar that may not exist on the reference, which is particularly important for biosimilar approval and monitoring.
  • The bridging format detects all isotypes of ADA, provided they can bind two drug molecules simultaneously.
  • The assay should be validated for specificity to cemiplimab-like antibodies, particularly in settings where patients might have received similar anti-PD-1 antibodies, but studies show cross-reactivity in the bridging ADA assay is minimal for anti-cemiplimab antibodies.

In summary, a cemiplimab biosimilar serves as both capture and detection reagent in a bridging ADA ELISA to accurately monitor the patient’s immune response against the therapeutic drug, in accordance with regulatory expectations.

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. Burova E, Hermann A, Waite J, et al. Mol Cancer Ther. 16(5):861-870. 2017.
6. Lee A, Duggan S, Deeks ED. Drugs. 80(8):813-819. 2020.
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.