This R-phycoerythrin (R-PE) conjugate is formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.4, 1% BSA and 0.09% sodium azide as a preservative.
Storage and Handling
This R-phycoerythrin (R-PE) conjugate is stable when stored at 2-8°C. Do not freeze.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
Next Day 2-8°C
Excitation Laser
Blue Laser (488 nm) and/or Green Laser (532 nm)/Yellow-Green Laser (561 nm)
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Cetuximab biosimilar antibody for staining cells in flow cytometry is ≤ 1.0 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Reported Applications For Relevant Conjugates ?
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 Cetuximab. Clone C225 recognizes human EGFR. This product is for research use only.
Background
EGFR is a 170 kD transmembrane glycoprotein that is part of the ErbB family of receptors within the protein kinase superfamily. EGFR is one of four closely related receptor tyrosine kinases: EGFR (ErbB-1), HER2/c-neu (ErbB-2), Her 3 (ErbB-3) and Her 4 (ErbB-4). EGFR is essential for various processes including controlling cell growth and differentiation and ductal development of the mammary glands. Ligand binding induces dimerization and autophosphorylation. It consists of a glycosylated extracellular domain which binds to EGF and an intracellular domain with tyrosine-kinase activity necessary for signal transduction. TGFα, vaccinia virus growth factor, and related growth factors can also bind to and signal through EGFR. Abnormal EGFR signaling has been implicated in inflammatory diseases such as psoriasis, eczema and atherosclerosis. Alzheimer's disease is linked with poor signaling of the EGFR and other receptor tyrosine kinases. Furthermore, over-expression of the EGFR is linked with the growth of various tumors. EGFR has been identified as an oncogene, a gene which in certain circumstances can transform a cell into a tumor cell, which has led to the therapeutic development of anticancer EGFR inhibitors. EGFR is a well-established target for both mAbs and specific tyrosine kinase inhibitors. Anti-Human EGFR (Cetuximab) utilizes the same variable regions from the therapeutic antibody Cetuximab making it ideal for research projects.
Antigen Distribution
EGFR is ubiquitously expressed and found in the plasma membrane.
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 Cetuximab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the basis for generating standard curves, which are essential for quantifying drug concentrations in serum samples.
Context and Supporting Details:
PK bridging ELISA is designed to measure the concentration of Cetuximab (reference and biosimilar versions) in biological matrices such as human serum, typically for bioequivalence or biosimilar characterization studies.
Calibration standards are prepared using known concentrations of the research-grade Cetuximab biosimilar, which are serially diluted and loaded into ELISA wells to generate a standard curve. This curve serves as the reference for quantifying unknown Cetuximab concentrations in sampled sera.
The use of a biosimilar as a calibration standard is considered best practice when the method has been demonstrated to quantify both reference and biosimilar products equivalently and with robust precision and accuracy. This approach reduces assay variability and regulatory risk by standardizing quantification across test products.
Assay kits or protocols, such as those described in product datasheets, often calibrate standards against both Erbitux™ (reference Cetuximab) and biosimilar drug, ensuring comparability and linearity (regression coefficients >0.9) for accurate measurement.
Reference controls (QC samples) may also be generated using either the biosimilar or reference compound at low, medium, and high concentrations, to monitor assay performance parameters such as accuracy, precision (%CV), and specificity.
In the typical PK bridging ELISA protocol for Cetuximab, anti-Cetuximab antibodies are used for capture and detection. The standard curve is generated using either the biosimilar or reference product, and sample concentrations are interpolated from this curve.
Biosimilars must be validated as suitable calibrators/control materials via method qualification studies that confirm their bioanalytical equivalency with the reference product in terms of recovery, linearity, and matrix effects.
Additional Relevant Information:
Both free and partially bound Cetuximab can be measured, depending on the antibody combination employed in the ELISA.
Strict documentation regarding batch, source, and calibration procedures must accompany each assay to ensure cross-study and regulatory consistency.
Calibration standard selection is especially critical in blinded clinical studies to avoid the need for separate analytical methods for biosimilar and reference products, facilitating regulatory acceptance of PK bridging data.
Summary Table: Cetuximab Biosimilar Use in PK Bridging ELISA
Role
Description
Calibration Standard
Used to generate standard curve for quantification of drug in serum
Reference Control (QC)
Monitors assay performance at defined concentrations
Equivalency Validation
Method must show reference & biosimilar are bioanalytically equivalent within assay
ELISA Protocol Usage
Standard curve typically uses biosimilar or reference Cetuximab, depending on equivalency
Regulatory Consistency
Single standard reduces variability and streamlines cross-study data
In summary, research-grade Cetuximab biosimilars are frequently employed as calibration standards and reference controls for PK bridging ELISAs, provided rigorous equivalency validation is performed to ensure accurate measurement of both biosimilar and reference drug concentrations in serum samples.
Standard flow cytometry protocols for assessing EGFR expression or binding capacity using conjugated Cetuximab biosimilars (e.g., PE or APC labeled) typically involve incubating target cells with the labeled antibody, followed by analysis to quantify cell-surface EGFR or antibody binding intensity.
Essential protocol steps:
Cell preparation: Cells expressing EGFR (e.g., A431 or CRC cell lines) are harvested, washed with ice-cold PBS, and blocked to prevent non-specific binding (often with serum or commercial blocking reagents).
Primary antibody incubation: The conjugated Cetuximab biosimilar (e.g., PE or APC) is added at a defined concentration (commonly ranging from 1 to 100 nmol/L), incubated for 30–60 minutes on ice or at 4°C to maintain cell integrity and minimize internalization.
Washing: Excess antibody is removed by repeated washing with PBS to reduce background signal.
Viability staining (optional): Propidium iodide (PI) or similar viability dyes may be added to exclude dead cells from analysis.
Flow cytometry analysis: Samples are run on a cytometer. Fluorescence intensity in the specific channel (PE/APC/FITC) reflects EGFR expression or Cetuximab binding capacity. Isotype controls and non-conjugated Cetuximab, or EGFR-negative cells, provide background reference.
Controls and validation:
Use isotype-matched antibodies conjugated to the same fluorophore to define non-specific binding.
Include negative cell lines lacking EGFR, and positive controls with known high EGFR expression (e.g., A431).
Compare histograms and cytograms (e.g., FITC vs PE) to assess relative EGFR expression.
Secondary detection (for non-directly labeled Cetuximab) involves biotinylation and streptavidin-fluorophore conjugates, but direct conjugation (PE, APC) skips this step.
Data Interpretation:
EGFR level is generally reported as median or mean fluorescence intensity (MFI), which is proportional to antibody binding and thus to receptor density.
Multiple cell lines are evaluated side-by-side for direct quantitation and comparative assessment.
Notes:
Depending on the conjugate, incubation and washing times may vary.
Cell permeabilization (methanol, detergents) is only required for intracellular EGFR detection and is not standard for surface binding unless activated EGFR or internalized antibody is being measured.
For biosimilar validation, specific binding to EGFR versus off-target binding should be quantitatively assessed in parallel with wild-type and mutated receptor variants.
This protocol is broadly applicable to the use of PE/APC-conjugated Cetuximab biosimilars for EGFR expression validation and binding studies by flow cytometry.
Biopharma companies typically perform a comprehensive suite of analytical assays to confirm the structural and functional similarity of a proposed biosimilar to its originator drug, focusing on critical quality attributes (CQAs). Leinco biosimilars are often used as reference standards or controls in these comparative studies when available or applicable, aiding in establishing assay specificity and benchmarking the performance of other biosimilars, though the exact use depends on the molecule in question and assay design.
Key analytical assays include:
Physicochemical characterization: Primary structure analysis (including peptide mapping and sequencing), secondary and tertiary structure assessment (such as circular dichroism, differential scanning calorimetry, NMR spectroscopy), and higher-order structure analysis are performed to confirm that the biosimilar matches the originator at the molecular level.
Post-translational modification profiling: This includes glycosylation analysis, oxidation, deamidation, and other modifications using mass spectrometry and chromatographic methods.
Functional bioassays: Relevant cell-based assays, binding assays, and enzyme kinetics are employed to confirm the biological activity of the biosimilar matches the originator. These test whether any observed structural differences affect function in ways that would influence clinical efficacy or safety.
Impurity and aggregate analysis: Rigorous assessment of purity and impurity profiles, including product-related impurities like aggregates, fragments, and process-related impurities, is performed as a sensitive measure of process comparability.
Orthogonal and complementary methods: Multiple, complementary (“orthogonal”) techniques are used for each CQA to ensure robust characterization and to increase sensitivity for detecting minor differences.
Regulatory context: FDA and EMA biosimilar approval is highly dependent on these analytical comparability studies, with particular emphasis on properties that significantly affect pharmacokinetics, pharmacodynamics, immunogenicity, and clinical outcomes.
Leinco biosimilar products:
Leinco Technologies supplies biosimilar antibodies and proteins that can be used as controls or reference materials in assay development and validation. The purpose is to offer a characterized source of biosimilar for method validation, calibration, and benchmarking during comparative analysis. Their role is not to serve as the reference listed drug (originator), but rather to facilitate assay standardization and reproducibility where relevant.
In the context of biosimilarity studies, such control biosimilars from Leinco would be included alongside the proposed candidate and the originator to ensure specificity and accuracy in performance metrics. This may involve, for example, benchmarking binding affinity, purity, or potency across test runs.
Summary of workflow:
Define critical quality attributes based on the originator’s mechanism of action and clinical relevance.
Perform head-to-head, validated comparative analytical testing using sophisticated, orthogonal methods.
Evaluate and confirm that the biosimilar is highly similar to the reference, with any differences shown not to impact safety or efficacy.
Use reference standards (including Leinco biosimilars if relevant) for assay validation and performance benchmarking.
This analytic process is essential for demonstrating biosimilarity before proceeding to clinical comparability and regulatory approval.
References & Citations
1. Tortora, G. et al. (1999) Clin Cancer Res. 5(4):909-16.
2. Myers, J. et al. (2006) Clin Cancer Res. 12(2): 600–607.