Anti-Human PD-1 (Camrelizumab)

Product No.: P810

- -
- -
Product No.P810
Clone
SHR-1210
Target
PD-1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anti-PD-1, PDCD1, CD279
Isotype
Human IgG4κ
Applications
ELISA
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Recommended Isotype Controls
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.
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
ELISA,
WB
Additional Applications Reported In Literature ?
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 Camrelizumab. Camrelizumab activity is directed against human PD-1 (CD274).
Background
Programmed cell death 1 (PD-1) is a transmembrane protein in the Ig superfamily 1,2 that acts as an immune checkpoint receptor 3, a T cell inhibitory receptor, plays critical roles in peripheral tolerance induction, autoimmune disease prevention, macrophage phagocytosis, tumor cell glycolysis, and dendritic cell survival 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. Programmed cell death 1 ligand 1 (PD-L1; CD274; B7H1) and programmed cell death 1 ligand 2 (PD-L2; CD273; B7DC) are ligands 1. Camrelizumab is a humanized high-affinity monoclonal antibody developed by Jiangsu Hengrui Medicine Co. Ltd as a cancer immunotherapeutic 4 that is derived from murine hybridoma Mab005 5. Camrelizumab binds to and blocks PD-1 binding to PD-L1 and PD-L2, preventing activation of downstream signalling pathways and restoring immune function 4. Camrelizumab also has off-target binding to the vascular receptor VEGFR2 (KDR), frizzled class receptor 5 (FZD5), and UL16 binding protein 2 (ULBP2) due to activity in the complementarity-determining regions of the v-domains from its Mab005 parent 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.

Research-grade Camrelizumab biosimilars can be used as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA to measure drug concentration in serum samples through a process that ensures accurate and reliable assay performance. Here's how this can be achieved:

1. Assay Development and Optimization

  • Assay Format: The choice between sandwich or competitive ELISA formats depends on the specific requirements of detecting free or total drug in the serum. Sandwich ELISA typically involves a capture antibody and a detection antibody, while competitive ELISA involves competition between the drug and a labeled drug for binding sites on an antibody.
  • Reagent Selection: High-quality antibodies with specific binding affinity to Camrelizumab are crucial. These antibodies should be validated for their ability to bind both the biosimilar and reference products with similar affinity to minimize variability.
  • Optimization: Assay conditions such as dilution buffers, blocking buffers, and incubation conditions need to be optimized to enhance sensitivity and specificity.

2. Calibration and Standardization

  • Calibration Curve: A calibration curve is established using standards of known concentrations of the Camrelizumab biosimilar. This curve serves as a reference to quantify unknown concentrations in serum samples.
  • Reference Controls: Biosimilars are used as reference controls to validate the assay's performance. These controls help ensure that the assay can accurately measure the drug concentration across different batches and conditions.

3. Pharmacokinetic (PK) Studies

  • Single PK Assay Approach: Using a single PK assay that can quantify both the biosimilar and reference products reduces variability and simplifies the analysis. This approach requires a scientifically sound testing strategy to evaluate bioanalytical comparability.
  • Bioanalytical Equivalence: The assay must demonstrate bioanalytical equivalence by showing that the biosimilar and reference products are measured with similar precision and accuracy. This is typically done by comparing the 90% confidence intervals of the measured concentrations to a predefined equivalence interval.

4. Use of Research-Grade Biosimilars

  • Bridging ELISAs: Biosimilars can be used as reference standards or capture and detection reagents in bridging ELISAs. This helps to standardize the assay and ensure that it can accurately measure the drug concentration regardless of whether the sample contains the biosimilar or reference product.

By following these steps, research-grade Camrelizumab biosimilars can effectively be used as calibration standards or reference controls in PK bridging ELISAs, allowing for reliable and accurate measurement of drug concentrations in serum samples.

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

Research-grade anti-PD-1 antibodies are administered in vivo primarily using syngeneic and humanized mouse models to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs). Both systems have distinct advantages and are widely used in immunotherapy research.

Syngeneic Mouse Models

Syngeneic models involve transplanting cancer cell lines derived from a specific mouse strain into immunocompetent mice of the same genetic background. These models are especially valuable for studying the interplay between the immune system and tumors, and for evaluating the effects of anti-PD-1 antibodies on TILs.

  • Common tumor types: Syngeneic models have been established for a wide range of cancers, including melanoma, colon, breast, bladder, kidney, pancreatic, non-small cell lung cancers, and lymphomas.
  • TIL characterization: Flow cytometry and immunohistochemistry (IHC) are used to quantify and characterize infiltrating immune cells, especially CD8⁺ T cells, before and after anti-PD-1 treatment.
  • Key findings: In these models, anti-PD-1 therapy often leads to increased TILs, particularly CD8⁺ T cells, which are associated with tumor regression. The efficacy of PD-1 blockade can depend on the baseline presence of TILs and tumor mutation burden (TMB).
  • Advantages: The intact immune system allows researchers to study the full spectrum of immune responses, including T-cell priming, exhaustion, and functional reinvigoration.

Example: Studies have shown pronounced antitumor effects of PD-1/PD-L1 inhibitors in syngeneic models, with tumor growth inhibition closely associated with CD8⁺ T-cell infiltration and PD-L1 expression.

Humanized Mouse Models

Humanized mouse models involve engrafting human immune systems (often hematopoietic stem cells or peripheral blood mononuclear cells) into immunodeficient mice, followed by implantation of human tumor cells (xenografts). These models are used to study human-specific immune responses and the effects of humanized anti-PD-1 antibodies.

  • Clinical relevance: Humanized models are critical for preclinical testing of therapeutic antibodies designed for human use, such as pembrolizumab and nivolumab.
  • TIL analysis: While TIL characterization is more challenging due to the chimeric nature of the immune system, these models can still provide insights into human T-cell responses against tumors in the presence of humanized anti-PD-1 therapy.
  • Limitations: The human immune system in these mice is not fully reconstituted, and the tumor microenvironment may not perfectly mimic the human setting.

Example: Approved anti-PD-1 drugs (e.g., pembrolizumab, nivolumab) and research-grade humanized antibodies (e.g., HX008) are tested in humanized models to confirm their antitumor efficacy and immune effects.

Comparison Table

Model TypeImmune SystemTumor OriginTIL CharacterizationKey AdvantagesLimitations
SyngeneicMouse (intact)Mouse cell linesFlow cytometry, IHCFull immune response, T-cell dynamicsLimited to mouse antigens
HumanizedHuman (engrafted)Human cell linesFlow cytometry (chimeric)Tests human-specific antibodies/effectsIncomplete immune reconstitution

Summary

  • Syngeneic models are the primary choice for mechanistic studies of anti-PD-1 antibodies, offering a robust platform to analyze TIL dynamics and tumor growth inhibition in an immunocompetent host.
  • Humanized models are essential for preclinical evaluation of humanized anti-PD-1 antibodies, bridging the gap between mouse studies and clinical trials.
  • Both models are complementary, with syngeneic systems providing mechanistic insights and humanized systems validating clinical candidates.

Researchers often use syngeneic models for discovery and mechanism-of-action studies and humanized models for translational and preclinical development of anti-PD-1 immunotherapies.

Researchers use Camrelizumab biosimilar, a humanized anti–PD-1 antibody, in combination with other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to investigate potential synergistic effects in complex immune-oncology models, primarily in preclinical and clinical studies focused on various cancer types.

Context and Supporting Details:

  • Combination Rationale: Camrelizumab (anti–PD-1) targets the PD-1 receptor to prevent inhibition of T cells by tumor cells, thus restoring T cell–mediated antitumor activity. Other checkpoint inhibitors, such as anti-CTLA-4 or anti-LAG-3 antibodies, work through distinct yet complementary mechanisms. Anti-CTLA-4 primarily acts at the priming phase of T-cell activation, while LAG-3 modulates T cell function and exhaustion, providing different nodes for immune modulation.

  • Preclinical Models: Synergistic effects are often assessed using complex animal models:

    • In studies with advanced melanoma, combinations such as anti–PD-1/CTLA-4 and anti–PD-1/LAG-3 have shown distinct patterns of immune cell activation and different mechanisms of action. For example, anti–PD-1/LAG-3 combinations require CD4+ T cell help for efficacy and tend to increase CD8+ cytotoxic T cell activity by reducing Treg-mediated suppression. In contrast, anti–PD-1/CTLA-4 combinations more directly enhance CD8+ cytotoxic T cell numbers and activity.
  • Efficacy Assessment: Researchers measure:

    • Tumor growth inhibition
    • Immune profiling (e.g., flow cytometry of tumor-infiltrating lymphocytes to assess T cell subsets, activation, exhaustion)
    • Survival outcomes
    • Assessment of immunogenicity and anti-drug antibody development, especially relevant for biosimilars
  • Clinical Application: Camrelizumab is currently used in combination with various agents (e.g., chemotherapy, antiangiogenic agents) in clinical settings, with ongoing or planned trials to further differentiate the contributions of each agent, including comparisons to anti–PD-1 monotherapy. While direct clinical studies of camrelizumab biosimilars in combination with anti-CTLA-4 or anti-LAG-3 biosimilars are less common in published literature, combinations of PD-1 inhibitors with these other checkpoint antibodies are well-studied with other agents.

Key Techniques and Readouts:

  • Use of multi-arm, randomized trials or preclinical experiments with defined control and combination groups to isolate the effect of each checkpoint inhibitor.
  • Detailed immune monitoring: quantifying changes in CD4+, CD8+, and Treg cell populations, cytokine release, and molecular markers of T cell activation, exhaustion, or proliferation.
  • Integration of biomarkers such as tumor mutational burden (TMB) and anti-drug antibody (ADA) levels to evaluate correlation with efficacy or safety.

Summary Table: Mechanistic Differences in Checkpoint Inhibitor Combinations

CombinationRequired Cell TypesMain MechanismTypical Readouts
Camrelizumab + anti-CTLA-4CD8+ T cellsDirect CD8+ T cell activationTumor size, CD8+ T cell infiltration
Camrelizumab + anti-LAG-3CD4+ & CD8+ T cellsCD4+ help, reduced Treg, CD8+ boostTumor size, Treg/CD8/CD4 ratios

Conclusion:
Researchers design combination regimens using camrelizumab biosimilars with anti-CTLA-4 or anti-LAG-3 biosimilars to dissect and optimize synergistic immune responses, leveraging complementary mechanisms in complex immune-oncology models, with experimental evaluation focusing on detailed immune profiling and efficacy against tumors.

In the context of immunogenicity testing for a Camrelizumab biosimilar, a bridging ADA ELISA utilizes the biosimilar as both the capture and detection reagent to monitor anti-drug antibodies (ADAs) that patients may develop against the therapeutic drug.

Bridging ELISA Design for Camrelizumab Biosimilar

The bridging ELISA format represents an innovative assay approach specifically designed to detect bivalent anti-drug antibodies with high sensitivity. In this configuration for a Camrelizumab biosimilar, the drug serves a dual role in the assay system.

Capture Phase: The Camrelizumab biosimilar is biotinylated and immobilized onto streptavidin-coated plates, where it functions as the capture reagent. When patient serum samples containing potential ADAs are added to the wells, any anti-Camrelizumab antibodies present will bind to the immobilized biosimilar drug.

Detection Phase: A second portion of the Camrelizumab biosimilar, labeled with either a fluorescent dye or horseradish peroxidase (HRP), serves as the detection reagent. This labeled biosimilar binds to the other arm of the bivalent ADAs that are already captured on the plate, creating a "bridge" formation that gives the assay its name.

Mechanism of ADA Detection

The bridging format is particularly effective because it exploits the bivalent nature of most clinically relevant ADAs. When ADAs are present in the patient sample, they bind to the immobilized Camrelizumab biosimilar through one binding site while maintaining their second binding site available for the labeled detection reagent. This creates a sandwich-like complex that can be quantified through colorimetric, fluorescent, or chemiluminescent detection methods.

The signal intensity correlates with the concentration of ADAs in the patient sample, allowing for both qualitative detection and semi-quantitative assessment of the immune response against the therapeutic drug.

Clinical Significance and Sensitivity

Bridging ELISAs have demonstrated high sensitivity in detecting ADAs against various therapeutic monoclonal antibodies, including adalimumab, infliximab, and etanercept. The format's advantage lies in its ability to perform high-throughput screening while maintaining the sensitivity necessary to detect clinically relevant immune responses that could impact therapeutic efficacy or patient safety.

However, the specificity of bridging ELISA assays can be challenged by complex serum matrix components, soluble target molecules, or residual drug components, making the use of high-quality assay reagents and appropriate blocking solutions crucial for obtaining meaningful results. For Camrelizumab biosimilar monitoring, careful optimization of assay conditions would be essential to ensure reliable detection of treatment-induced ADAs while minimizing false-positive results from matrix interference.

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. Pardoll DM. Nat Rev Cancer. 12(4):252-264. 2012.
4. Markham A, Keam SJ. Drugs. 79(12):1355-1361. 2019.
5. Finlay WJJ, Coleman JE, Edwards JS, et al. MAbs. 11(1):26-44. 2019.
6. Huang J, Xu B, Mo H, et al. Clin Cancer Res. 24(6):1296-1304. 2018.
7. Huang J, Mo H, Zhang W, et al. Cancer. 125(5):742-749. 2019.
Indirect Elisa 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.