Anti-Human CD52 (Alemtuzumab) – Biotin

Anti-Human CD52 (Alemtuzumab) – Biotin

Product No.: LT201

- -
- -
Product No.LT201
Clone
Campath-1H
Target
CD52
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
HE5; CDW52; EDDM5 CDW52; Cambridge pathology 1 antigen
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Rhesus Monkey
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human lymphocytes.
Product Concentration
0.5 mg/ml
Formulation
This Biotinylated antibody 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 biotinylated antibody 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
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Alemtuzumab 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 Applications Reported In Literature ?
FA
ELISA
Additional Reported Applications For Relevant Conjugates ?
CyTOF®
WB
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 Alemtuzumab. Clone Campath-1H recognizes human CD52. This product is for research use only.
Background
Clone Campath-1H is a monoclonal antibody that specifically binds to CD52, a protein present on the surface of mature lymphocytes. However, this protein is not present on the stem cells that generated these lymphocytes. Alemtuzumab is targets and destroys mature lymphocytes containing CD-52, and is used to treat chronic lymphocytic leukemia (CLL) and multiple sclerosis. Anti-Human CD52 (Alemtuzumab) utilizes the same variable regions from the therapeutic antibody Alemtuzumab making it ideal for research projects.
Antigen Distribution
CD52 is primarily expressed on the surface of mature lymphocytes. Additionally, CD52 is present on most lymphoid derived malignancies. However, variable expression on Myeloma cells should be noted.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars

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 alemtuzumab biosimilars serve as critical calibration standards and reference controls in pharmacokinetic bridging ELISA assays through a carefully validated approach that ensures accurate quantification of drug concentrations in serum samples.

Calibration Standard Preparation and Validation

In pharmacokinetic bridging ELISAs, alemtuzumab biosimilars are prepared as calibration standards using multiple batches to establish reliability. Research has demonstrated that three different alemtuzumab calibration standards from different batches are compared to exclude concentration differences between batches, with accuracy ranging from 91% to 104%. The calibration standards are typically prepared in neat pooled human serum to match the matrix of clinical samples.

Dynamic Range and Sensitivity Requirements

The calibration standards establish a dynamic range that accommodates the expected therapeutic concentration range. For alemtuzumab ELISA assays, the dynamic range typically spans from 0.78 to 25 ng·mL⁻¹, with a lower limit of quantification (LLoQ) of 0.5 ng·mL⁻¹. This sensitivity range is crucial for measuring both peak concentrations and concentrations below the lympholytic level (<0.1 mcg·mL⁻¹), requiring patient serum samples to be prediluted 20-400 times according to expected alemtuzumab concentrations.

Quality Control and Reproducibility

Batch-to-Batch Consistency: Multiple biosimilar batches are used as reference standards to validate consistency across different production lots. Complete calibration curves are measured multiple times over extended periods to analyze reproducibility based on precision (coefficient of variation) and accuracy compared to theoretical concentrations.

Performance Specifications: The calibration standards must meet stringent accuracy and precision criteria, with overall within-run accuracy between 96% and 105%, and within-run precision (coefficient of variation) between 3% and 9%. Between-run assessments provide overall accuracy between 86% and 95% and coefficient of variation between 5% and 14%.

Anti-Idiotypic Antibody Approach

Modern biosimilar ELISA assays employ anti-idiotypic monoclonal antibodies in a sandwich assay format, which provides exceptional specificity and sensitivity in drug detection, even at low concentrations. These assays are calibrated against innovator drugs to ensure high accuracy, with standards provided in kits that are calibrated against commercially sourced drugs and alternate recombinant biosimilars.

Regulatory Compliance and Validation

The use of biosimilar calibration standards follows strict regulatory guidelines, with validation performed according to European Medicines Agency (EMA) guidelines on bioanalytical method validation. Commercial assays are validated against ICH/EMA guidelines under ISO 13485 certification, ensuring inter and intra-assay coefficients of variation remain below 10% in accordance with FDA and EMEA requirements.

Matrix Effect Considerations

To ensure no matrix interference and reproducible dilutional linearity, multiple serum and plasma spiking experiments are conducted at various dilutions during optimization. This approach validates that the biosimilar standards perform consistently across different sample types and dilution factors, which is essential for accurate pharmacokinetic analysis in clinical samples.

The integration of research-grade alemtuzumab biosimilars as calibration standards enables precise therapeutic drug monitoring and pharmacokinetic analysis, providing clinicians with the accurate concentration data needed to optimize treatment strategies and dosing regimens.

Standard flow cytometry protocols using a conjugated Alemtuzumab biosimilar (such as PE, APC, or Alexa Fluor 647-labeled) to validate CD52 expression levels or binding capacity typically follow these structured steps:

  1. Sample Preparation

    • Use either human whole blood or a single-cell suspension (e.g., from bone marrow or tissue).
    • For whole blood, lyse erythrocytes with a commercial lysing buffer (such as BD Pharm Lyse™).
  2. Antibody Staining

    • Incubate the cells with fluorochrome-conjugated Alemtuzumab biosimilar (e.g., Alexa Fluor 647, PE, or APC) at a recommended concentration (see manufacturer's datasheet for optimal dilution).
    • Include an appropriate isotype control conjugated to the same fluorochrome to set background fluorescence.
  3. Controls and Calibration

    • Use isotype controls and compensation controls for multiparameter panels.
    • For quantitative binding capacity (antibody binding capacity, ABC), use quantitation beads (such as BD QuantiBRITE PE Beads) that contain known levels of fluorochrome; run these under the same settings as your samples to generate a standard curve for absolute quantitation.
  4. Washing and Acquisition

    • Wash cells to remove unbound antibodies.
    • Acquire data on a flow cytometer compatible with the chosen fluorochrome (e.g., BD LSRFortessa™ for Alexa Fluor 647).
  5. Gating Strategy

    • Use forward scatter (FSC) and side scatter (SSC) to identify viable lymphocytes or leukocytes.
    • Additional markers (e.g., CD45, CD3) may be included to gate specific subsets (for malignant samples, use markers relevant to the malignancy, such as CD25 for ATLL).
  6. Data Analysis

    • For qualitative validation: Compare fluorescence intensity of the Alemtuzumab biosimilar antibody against isotype control to confirm CD52 expression.
    • For quantitative assessment: Use the geometric mean fluorescence intensity (MFI) of the stained sample and convert MFI to antibody binding capacity (ABC) using quantitation beads and analysis software (e.g., FlowJo™, QunatiCALC).

Key technical details drawn from protocols:

  • Incubation time is typically 20–30 minutes at 4°C or on ice to avoid internalization of surface antigens.
  • Exclusion of dead cells is often done by light scatter gating or viability dyes (not always required for whole blood if using lyse-no-wash protocols).
  • To compare different samples (e.g., patient and control), keep instrument settings standardized and run calibration beads with each batch for quantitation.

Fluorochrome Options:

  • Alexa Fluor™ 647, PE, and APC are commonly used for their compatibility with standard flow cytometer lasers and distinct emission spectra.

Use-case examples:

  • Assessing variability and suitability for therapy: Quantify CD52 on T- and NK-cell malignancy samples with Alemtuzumab biosimilar-PE and QuantiBRITE beads to determine whether CD52 expression is sufficient for targeted therapy.
  • Routine immunophenotyping: Confirmation of CD52 presence/absence using a conjugated Alemtuzumab biosimilar antibody in clinical or research panels.

References to standard protocols and datasheets:

  • BD Biosciences provides detailed protocols using Alexa Fluor 647-conjugated Alemtuzumab biosimilar for multiparameter flow cytometric analysis of CD52.
  • Quantitative methods using QuantiBRITE beads for ABC determination are described in peer-reviewed studies and by manufacturers.

In summary:
Use a fluorochrome-labeled Alemtuzumab biosimilar for direct staining, include isotype controls and quantitative beads as needed, acquire on a compatible cytometer, apply proper gating, and use analysis software to measure either relative or absolute CD52 expression or binding capacity.

Biopharma companies perform a comprehensive suite of analytical assays to demonstrate that a proposed biosimilar is structurally and functionally similar to its originator drug. These include rigorous comparative studies of physicochemical properties, structural features, post-translational modifications, impurity profiles, and a range of functional (biological) assays.

Analytical Assays for Biosimilar Characterization

Structural Characterization:

  • Primary structure: Peptide mapping (mass spectrometry) and amino acid sequencing ensure the biosimilar’s sequence matches the reference.
  • Higher-order structure: Techniques like circular dichroism (CD), nuclear magnetic resonance (NMR), and differential scanning calorimetry (DSC) verify secondary, tertiary, and quaternary structures.
  • Post-translational Modifications: Glycosylation profiling (using HPLC or MS), deamidation, oxidation, and other modifications are compared, given their impact on efficacy and immunogenicity.
  • Purity and Impurity Profiles: Size-exclusion chromatography, capillary electrophoresis, and mass spectrometry detect aggregates, fragments, and process-related impurities.

Functional Characterization:

  • Biological Activity: Cell-based potency assays confirm the biosimilar’s bioactivity matches the reference, often involving measurement of target cell response (e.g., proliferation, cytotoxicity).
  • Binding Assays: ELISA, SPR (surface plasmon resonance), or BLI (biolayer interferometry) determine binding affinity to targets such as antigens or receptors.
  • Fc-Function Assays: For antibodies, effector functions like FcγR binding or ADCC (antibody-dependent cell-mediated cytotoxicity) are measured to ensure matching functional performance.

Orthogonal and Risk-Based Approaches:

  • Multiple, complementary (orthogonal) methods increase confidence in detected similarities or differences, focusing particularly on attributes (referred to as critical quality attributes, or CQAs) that most influence clinical outcomes.
  • Results are expected to fall within the predefined range observed for the originator, accounting for potential lot-to-lot variability.

Role of Leinco Biosimilars in Analytical Studies

Leinco Technologies provides research-grade biosimilar antibodies and reagents often used as surrogates or standards within biosimilar analytical comparability programs.

  • In biosimilar development, Leinco biosimilars may serve as reference materials for calibration, assay development, or benchmarking, especially in preclinical and early-stage analytical method validation.
  • They can be used in head-to-head testing against the originator biologic to optimize assay sensitivity, characterize binding, or demonstrate equivalency of functional activity, though these materials are not themselves used for direct clinical biosimilarity assessment or regulatory submission.
  • Leinco’s biosimilar reagents are employed to streamline method development and serve as control reagents for the evaluation of binding, potency, and specificity in various analytical platforms.

In summary, the typical analytical strategy combines extensive physicochemical, structural, and functional testing with sensitive, validated methods; Leinco biosimilars play a supporting role in assay calibration and development but are not a substitute for head-to-head analytical assessment with the regulatory reference (originator) product.

References & Citations

Indirect Elisa Protocol
FA
Flow Cytometry

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.