Anti-Human CD52 (Alemtuzumab) – Fc Muted™

Anti-Human CD52 (Alemtuzumab) – Fc Muted™

Product No.: LT205

- -
- -
Product No.LT205
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
CyTOF®
,
ELISA
,
FA
,
FC
,
IHC FF
,
IHC FFPE
,
WB

- -
- -
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
Muted
Immunogen
Human lymphocytes.
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.
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
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Alemtuzumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 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 ?
IHC FFPE (Formalin-fixed paraffin-embedded tissue)
IHC FF (Fresh Frozen)
FA
WB
ELISA
Additional Reported Applications For Relevant Conjugates ?
CyTOF®
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 are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to accurately quantify drug concentrations in serum samples by providing standardized reference points for assay calibration and validation.

In a PK bridging ELISA, calibration standards are essential for generating a calibration curve, which directly relates the ELISA signal to known concentrations of alemtuzumab. Typically, these standards are prepared from research-grade biosimilar alemtuzumab, ideally matched to the innovator (reference) molecule in terms of structure, purity, and binding properties.

Key steps and roles of biosimilar calibration standards in the assay:

  • Preparation: Calibration standards are prepared as serial dilutions of research-grade alemtuzumab biosimilar in a matrix that closely matches serum (e.g., 1% PBS/HSA, pooled human serum).
  • Comparison of Batches: To ensure consistency, multiple batches of the biosimilar are compared for concentration equivalence and accuracy using independent quantification. According to published validation, three different alemtuzumab calibration standards (from different batches) were compared and found to have accuracy within 91%–104% of theoretical values.
  • Calibration Curve Generation: The ELISA utilizes these standards to generate a standard curve, typically spanning a dynamic range appropriate for the expected serum concentrations (e.g., 0.78–25 ng/mL). The quality of this curve—its accuracy, precision, and linearity—is validated using repeated measurements.
  • Quality Control: The assay is validated for precision (coefficient of variation), accuracy, and sensitivity (lower limit of quantification, LLoQ) using these reference standards according to regulatory guidelines (EMA, FDA, ICH).
  • Reference Controls: Biosimilar reference controls serve to verify assay performance across runs, ensuring that test samples are reliably compared to a standardized reference. Multiple biosimilar batches may be used as controls to confirm batch-to-batch consistency and serve as quality benchmarks.

Why biosimilars can be used:

  • Biosimilar reference standards, if validated against innovator molecules and international standards (e.g., NIBSC/WHO), provide an accurate surrogate for the original reference drug.
  • They enable bridging studies between different assay formats, laboratories, and drug products, supporting regulatory and scientific requirements for drug monitoring and comparability.

Additional details:

  • Calibration standards are often lyophilized for improved stability and reproducibility, and are validated to ensure shelf life and consistency over time.
  • ELISA kits for therapeutic drug monitoring may be developed specifically for biosimilars, using anti-idiotypic antibodies for high specificity, and spiking experiments in human serum to address recovery and matrix effects.

In summary, research-grade alemtuzumab biosimilars provide the foundation for calibration and quality control in PK bridging ELISA assays, ensuring accurate and reproducible measurement of drug concentrations in serum samples. This process is rigorously validated to meet both scientific and regulatory standards.

The primary in vivo models for research-grade anti-CD52 antibody evaluation in tumor growth inhibition and TIL characterization are immunodeficient mouse xenografts (notably SCID mice) and murine syngeneic tumor models, with humanized mouse models also sometimes used.

Model Details:

  • Human Tumor Xenografts in Immunodeficient Mice:

    • SCID mouse models: Anti-CD52 monoclonal antibodies have been tested in SCID mice bearing human tumor cells (e.g., MC/CAR cell lines with high CD52 expression). These studies evaluate tumor inhibition and survival, often at doses up to 10 or 30 mg/kg of antibody. The SCID model features transplanted human tumors in an immunodeficient background, allowing direct assessment of anti-tumor effects but provides only limited immune characterization (as SCID mice lack functional T and B cells).
  • Syngeneic Mouse Tumor Models:

    • Murine syngeneic models involve implanting mouse tumor cell lines into immunocompetent mice of the same genetic background. These models are critical for evaluating the impact of immunotherapies, including anti-CD52 antibodies, within a fully functional immune system. Because syngeneic models allow for intact immune responses, researchers can perform detailed profiling of tumor-infiltrating lymphocytes (TILs) to study how anti-CD52 therapy modulates immune subsets inside tumors. Common syngeneic models for immunotherapy research include CT26, B16F10, MC38, and RENCA.
  • Humanized Mouse Models:

    • In some advanced studies, humanized mice (immunodeficient mice reconstituted with human immune cells) are used to enable both human tumor growth and assessment of human TILs after anti-CD52 treatment, though this specific application is less frequently described in published literature compared to the two models above.

Syngeneic vs. Humanized Models: Comparison

Model TypeSpecies/TumorImmune SystemTIL CharacterizationTypical Use with Anti-CD52
SyngeneicMouse/MouseFully competent (mouse)Yes, murine TIL analysisEvaluates immune response, TIL profiles, tumor inhibition
SCID xenograftMouse/HumanImmunodeficientMinimal (murine)Direct tumor inhibition, limited TIL data
Humanized mouseMouse/HumanHuman immune cellsYes, human TIL analysisBest for human TIL and anti-CD52 studies, but less common
  • Syngeneic models are preferred for mechanistic immunotherapy studies requiring TIL analysis due to their intact immune compartments, allowing robust characterization post-treatment.
  • SCID xenograft models are commonly used for direct anti-tumor effect evaluations, particularly where human tumor lines expressing CD52 are administered, but are limited in immune profiling.

No results show direct use of anti-CD52 antibodies in humanized mouse models specifically for TIL profiling in cancer studies, but this remains an emerging area.

Summary:

  • Syngeneic mouse tumor models are the gold standard for in vivo immunotherapy studies integrating tumor growth inhibition and detailed TIL characterization with anti-CD52 administration.
  • SCID xenograft models are widely used for tumor growth inhibition analyses in the context of anti-CD52, though immune characterization is limited.
  • Humanized mice offer potential for human-specific TIL analysis but are less commonly utilized in published anti-CD52 studies.

Researchers studying synergistic effects of Alemtuzumab biosimilars with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) in complex immune-oncology models generally use preclinical in vivo and in vitro systems to illuminate combined anti-tumor efficacy and immune modulation.

Approach and Model Selection:

  • Alemtuzumab biosimilars (e.g., Mab-TH) are first rigorously tested for equivalence to the originator in in vitro cytotoxicity and in vivo tumor models (often using severe immunodeficient mice transplanted with human leukemia cell lines).
  • The effectiveness is confirmed by tumor regression and improvements in animal survival rates at defined dosing regimens.
  • Complex models, such as humanized mice or tumorgraft systems, may be used to assess immune cell dynamics when combining Alemtuzumab with immune checkpoint inhibitors.

Combination Strategies:

  • These biosimilars are combined with checkpoint inhibitors (like anti-CTLA-4, anti-PD-1, anti-LAG-3) because the targets are mechanistically distinct—Alemtuzumab depletes CD52+ lymphocytes, affecting both malignant and immune cells, whereas checkpoint inhibitors release brakes on T cell activation.
  • Researchers design experiments where tumor-bearing mice are co-treated with Alemtuzumab (or its biosimilar) and checkpoint inhibitor biosimilars, assessing:
    • Tumor growth inhibition
    • Immune cell infiltration and activation within the tumor
    • Peripheral immune cell profiles
    • Cytokine release and other biomarkers of immune activation.

Rationale for Combination:

  • The synergistic hypothesis is that depleting immunosuppressive cells (via CD52 targeting) can enhance the efficacy of checkpoint blockade, resulting in greater anti-tumor activity than either agent alone.
  • This approach mirrors strategies used with CTLA-4 and PD-1 combinations, where targeting different regulatory points of the immune response amplifies overall therapeutic effect.

Endpoints and Mechanistic Studies:

  • Primary readouts include survival, tumor volume, histological analysis of immune cell populations, and toxicity assessments.
  • Researchers often perform mechanistic experiments to investigate changes in T cell subsets, myeloid infiltration, and cytokine/chemokine signatures.

Example Evidence and Experimental Outcomes:

  • Preclinical studies with anti-CD52 (Alemtuzumab or biosimilars) have shown dose-dependent tumor inhibition in grafted mouse models.
  • Combination studies (not limited to Alemtuzumab, but generalizable) demonstrate that dual checkpoint inhibition or pairing with cell-depleting antibodies can produce durable responses, though often at the cost of increased immune-mediated toxicities.

Key Considerations:

  • Models must be chosen with care, as CD52 is not universally expressed in all animal substrates—often requiring primate or humanized models for translational relevance.
  • Dosing regimens for combination are optimized based on pharmacodynamics and safety in single-agent studies before combinatorial use.

In summary, Alemtuzumab biosimilars are integrated into preclinical immune-oncology models, in combination with other checkpoint inhibitor biosimilars, by co-administering them in tumor-bearing animals or in vitro immune cell assays to assess if synergistic anti-tumor effects or enhanced immune activation occur. The experimental readouts guide further translational research, paving the way for potential clinical combination therapies.

Use of Alemtuzumab Biosimilar in Bridging ADA ELISA for Immunogenicity Testing

Bridging anti-drug antibody (ADA) ELISA is a widely used method to detect and quantify antibodies that patients may develop against therapeutic biologics, such as monoclonal antibodies (mAbs) like alemtuzumab or its biosimilars. These assays are critical in immunogenicity assessment because anti-drug antibodies can neutralize drug activity, accelerate clearance, or trigger adverse effects—ultimately impacting therapeutic efficacy and safety.

Mechanism of Bridging ADA ELISA

A typical bridging ELISA for ADA detection involves the following steps:

  • Biotinylation of the Drug (Alemtuzumab/Biosimilar): The therapeutic mAb (e.g., alemtuzumab or a biosimilar like Mab-TH) is biotinylated to enable capture onto streptavidin-coated plates.
  • Sample Application: Patient serum, potentially containing anti-alemtuzumab antibodies (ADAs), is added to the plate. ADAs bridge between the captured biotinylated drug and a labeled version of the same drug.
  • Detection: The labeled drug (e.g., HRP-conjugated alemtuzumab/biosimilar) binds any ADAs, forming a detectable immune complex. The signal intensity correlates with ADA concentration in the sample.

Role of Alemtuzumab Biosimilar in the Assay

  • Capture Reagent: The biotinylated biosimilar (e.g., Mab-TH, which is highly similar to alemtuzumab in structure and binding activity) can be immobilized on streptavidin-coated plates to capture ADAs from patient serum.
  • Detection Reagent: The biosimilar (or sometimes the originator drug, alemtuzumab) is labeled with a detection moiety (e.g., HRP) and used to detect captured ADAs, completing the “bridge”.
  • Ensuring Specificity: Since the biosimilar (Mab-TH) is highly similar to alemtuzumab and binds CD52 with comparable affinity and specificity, it is suitable for both capture and detection steps, ensuring that the assay detects antibodies reactive to the therapeutic agent itself.

Why Use a Biosimilar?

  • Preclinical Demonstration of Similarity: Biosimilars like Mab-TH have been shown to have highly comparable binding and functional activities to alemtuzumab in preclinical models, supporting their use as substitutes in assay development.
  • Regulatory Pathway: Regulatory agencies (e.g., NMPA, FDA, EMA) require biosimilars to demonstrate similar efficacy, safety, and immunogenicity profiles to the originator, which includes validation in analytical and clinical testing.
  • Consistency and Supply: Use of a biosimilar can ensure consistent reagent supply for ADA assays, especially as patents expire and biosimilars become more widely available.

Considerations and Challenges

  • Interference from Drug: High levels of circulating drug in patient samples can compete with ADAs for binding to the capture reagent, potentially causing false-negative results. Assay optimization (e.g., acid dissociation steps) may be needed to mitigate this.
  • Matrix Effects: Human serum components may interfere with the assay, so proper blocking and controls are crucial for specificity and reproducibility.
  • Assay Validation: The assay must be validated to confirm that the biosimilar behaves equivalently to the originator in the ADA ELISA format, especially for regulatory submissions.

Summary Table: Role of Alemtuzumab Biosimilar in Bridging ADA ELISA

StepReagent UsedPurposeRationale
CaptureBiotinylated biosimilar (e.g., Mab-TH)Immobilize drug on plate to capture ADAsBiosimilar is structurally and functionally similar to originator
DetectionLabeled biosimilar or originatorDetect captured ADAs via bridgingEnsures detection of antibodies reactive against the therapeutic agent
Quality ControlBiosimilar characterizationValidate assay performanceSupports regulatory approval and clinical relevance

Conclusion

Alemtuzumab biosimilars, such as Mab-TH, can be effectively used as both capture and detection reagents in bridging ADA ELISAs to monitor patient immune responses against the therapeutic drug. Their high similarity to the originator in terms of binding and functional activity ensures the assay’s relevance for immunogenicity assessment in clinical settings. Proper assay design and validation are essential to address potential interferences and ensure reliable ADA detection.

References & Citations

CyTOF®
Indirect Elisa Protocol
FA
Flow Cytometry
IHC FF
IHC FFPE
General Western Blot Protocol

Certificate of Analysis

Formats Available

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