Anti-erbB-2 (Her-2/neu) (Margetuximab) [Clone MGAH22]

Anti-erbB-2 (Her-2/neu) (Margetuximab) [Clone MGAH22]

Product No.: LT220

- -
- -
Product No.LT220
Clone
MGAH22
Target
erbB-2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anti erbB-2, erbB2, HER2, CD340
Isotype
Human IgG1κ
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
Active
Immunogen
Human erbB2/EGFR2/CD340
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥98% 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 ?
FC,
ELISA,
WB,
IP,
FA
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 Margetuximab. This product is for research use only. Margetuximab activity is directed against Human erb-b2 receptor tyrosine kinase 2 (ERBB2; HER-2/neu).
Background
erbB-2 encodes a member of the epidermal growth factor (EGF) receptor family of receptor tyrosine kinases1. erbB-2 enhances kinase-mediated activation of downstream signaling pathways by forming a heterodimer with other ligand-bound EGF receptor family members. Dysregulation of erbB-2 contributes to tumorigenesis in breast, ovarian, gastric, and other cancers.

Margetuximab is a human/mouse chimeric anti-erbB-2 monoclonal IgG1 antibody derived from mouse clone 4D5, the precursor of trastuzumab2. Margetuximab has an Fc domain (MGFc0264) engineered for increased binding to both alleles of human activating Fcγ receptor IIIA (CD16A) and for reduced binding to CD32B. Compared with WT Fc domain, the optimized MGFc0264 domain demonstrates increased affinity for both alleles of human CD16A as well as human C1q but decreased binding to human CD32B (inhibitory FcγR) and the 131R allele of CD32A (human activating FcγR). Binding to the 131H allele is not substantially modified. The optimized Fc domain also confers improved antibody-dependent cell cytotoxicity against erbB-2-positive tumor cells, including low ERBB2 expressors, independent of the FcγR variant for the effector cells.

The MGFc0264 Fc domain was generated by mutating five sites: L235V, F243L, R292P, Y300L, and P396L2. The L235V mutation was inserted to reduce CD32B binding. The Fc domain modifications do not influence antigen recognition or anti-proliferative activity in the absence of effector cells.

In clinical trials, Margetuximab binds to erbB-2 with high affinity and produces direct growth suppression of erbB-2-expressing tumor cell lines3. Positive data from clinical trials led to US Food and Drug Administration approval for Margetuximab in the treatment of metastatic HER2-positive breast cancer in 20204.
Antigen Distribution
erbB-2 is an overexpressed cell-surface oncoprotein.
Ligand/Receptor
erbB-2/HER2/CD340, Receptor
PubMed
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 Margetuximab biosimilars are commonly used as calibration standards or reference controls in PK bridging ELISA assays to ensure accurate quantification of drug concentration in serum samples.

In these assays:

  • A single calibration standard—often the biosimilar—is selected to generate the standard curve for quantifying both the biosimilar and the reference product (e.g., the innovator drug) in test samples.
  • The reason for this approach is to minimize variability and to establish bioanalytical equivalence, ensuring the assay measures both the biosimilar and reference products with equal precision and accuracy.
  • The biosimilar standard is prepared at known concentrations in pooled human serum, enabling the creation of a dose-response curve throughout the assay’s dynamic range.
  • Quality control (QC) samples are also prepared using both biosimilar and reference material at multiple concentrations and quantified against the biosimilar standard curve.
  • This strategy enables direct, unbiased comparison of pharmacokinetic parameters between the biosimilar and reference products, supporting PK similarity and bioequivalence assessments required for regulatory submissions.

Supporting details:

  • The assay is fully validated for performance metrics like linearity, sensitivity, precision, and recovery as per FDA and industry guidelines.
  • Calibration standards typically span a wide, validated concentration range to ensure robust measurement across expected patient serum levels.
  • The biosimilar is selected for calibration only after demonstrating bioanalytical comparability between biosimilar and reference products in the assay context.
  • This approach aligns with regulatory best practice for ligand binding assays supporting biosimilar development, as it reduces inter-assay variability and streamlines clinical sample analysis.

Summary Table (for clarity):

Material UsedRole in ELISA PK Bridging AssayPurpose
Margetuximab BiosimilarCalibration StandardGenerate standard curve for quantifying drug concentration
Margetuximab ReferenceReference Control/QCValidate equivalence and assay performance
Human Serum MatrixMatrix for standards and QCsMimics patient sample environment

This approach ensures that serum drug concentrations measured in clinical samples are reliable and comparable for both biosimilar and branded Margetuximab products, a critical requirement for biosimilar approval and postmarketing surveillance.

In Vivo Models for Studying Anti-ErbB-2 Antibody Effects on Tumor Growth and TILs

Anti-ErbB-2 monoclonal antibodies (mAbs) are a cornerstone of research and therapy for HER2-positive cancers, with both xenograft (humanized/immunocompetent) and syngeneic (mouse-derived, immunocompetent) models used to study direct tumor growth inhibition and immune-mediated effects, including characterization of tumor-infiltrating lymphocytes (TILs).

Xenograft (Humanized) Models

  • Human ErbB-2+ Tumor Cells in Athymic Mice: Classic studies have used athymic (nude) mice engrafted with human tumor cell lines overexpressing ErbB-2 (e.g., gastric cancer line N87). Researchers then systemically administer research-grade anti-ErbB-2 mAbs (e.g., L26, N12, L431) and monitor tumor growth. In these models, antibody combinations have been shown to synergistically inhibit tumor growth, even leading to complete tumor eradication in some cases.
  • Focus: The primary outcome in these models is tumor growth inhibition via direct antibody targeting, with less emphasis on TIL characterization due to the lack of a functional mouse immune system. These studies primarily address mechanisms of antibody-induced ErbB-2 downregulation (e.g., endocytosis), not immune-mediated effects.

Syngeneic (Immunocompetent) Models

  • NDLUCD Breast Cancer Model: A notable syngeneic model is the NDLUCD cell line, derived from mammary tumors in FVB/N-Tg(MMTV-ErbB2*)NDL2-5Mul transgenic mice. These cells express high levels of ErbB-2 and PD-L1 and are transplantable into immunocompetent FVB/N mice. In this model, tumors elicit a robust stromal immune response, detectable via histology, immunophenotyping, and gene expression analysis.
  • Applications: Syngeneic models like NDLUCD are critical for studying the complex interplay between antibody therapy and the host immune system, including TIL characterization. Such models allow evaluation of not only direct antibody effects but also secondary immune responses (e.g., checkpoint blockade, T-cell infiltration).
  • Broad Utility: While not all syngeneic models are ErbB-2-specific, organizations like TD2 offer a range of characterized syngeneic tumor models suitable for immunotherapy research, though ErbB-2 overexpression is not always specified. The NDLUCD model is uniquely tailored for HER2-positive cancer research with an intact immune system.

Comparative Summary Table

Model TypeExample SystemKey FeaturesTIL CharacterizationUtility for Anti-ErbB-2 mAb Studies
XenograftHuman N87 in athymic miceDirect tumor inhibition, mechanistic studiesLimitedAntibody mechanism, tumor growth inhibition
SyngeneicNDLUCD in FVB/N miceImmune-competent, PD-L1+, ErbB-2+RobustImmune response, TILs, combination therapies

Conclusions

  • Xenograft models (human tumor cells in immunodeficient mice) are the standard for studying direct tumor growth inhibition by anti-ErbB-2 antibodies, with limited application for TIL analysis due to the lack of a functional immune system.
  • Syngeneic models, especially the NDLUCD breast cancer model, are emerging as powerful tools for studying both tumor inhibition and the immune microenvironment, including TIL dynamics, in the context of HER2-targeted therapy.
  • For mechanistic studies of antibody action (e.g., receptor downregulation), xenografts are preferred. For immunotherapy and TIL characterization, syngeneic models are essential.

In summary: Research-grade anti-ErbB-2 antibodies are most commonly studied in vivo using human tumor xenografts in immunodeficient mice for direct growth inhibition and mechanism studies, and in syngeneic, immune-competent models (notably NDLUCD) for comprehensive immune response and TIL characterization.

Researchers are exploring the use of margetuximab, a novel anti-HER2 monoclonal antibody, in conjunction with other checkpoint inhibitors to study synergistic effects in immune-oncology models. While margetuximab itself is not a checkpoint inhibitor, its mechanism of enhancing antibody-dependent cellular cytotoxicity (ADCC) through engineered Fc domains can complement immunotherapies. Here’s how researchers might approach combining margetuximab with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3:

Combining Margetuximab with Checkpoint Inhibitors

Mechanism of Action

  • Margetuximab: Targets HER2-positive cancer cells, enhancing ADCC through increased affinity for activating Fcγ receptors (CD16A) and decreased affinity for inhibitory Fcγ receptors (CD32B).
  • Checkpoint Inhibitors: Target immune checkpoints like CTLA-4, PD-1, or LAG-3 to prevent cancer cells from evading the immune response.

Synergistic Effects

Combining margetuximab with checkpoint inhibitors could potentially offer synergistic benefits by:

  • Enhancing Immune Response: Margetuximab increases ADCC, which could be further enhanced by checkpoint inhibitors that activate more T cells.
  • Targeting Multiple Pathways: Simultaneously targeting HER2 and immune checkpoints can attack cancer cells from multiple angles, potentially overcoming resistance mechanisms.

Research Approaches

  1. Preclinical Models: Researchers use animal models to study how margetuximab and checkpoint inhibitors interact synergistically to enhance antitumor activity.
  2. Clinical Trials: Pilot or phase I studies might be conducted to assess safety and efficacy when combining these agents in patients with HER2-positive cancers.

Challenges and Opportunities

  • Challenges: Combining therapies can increase toxicity, so careful dosing and patient selection are crucial.
  • Opportunities: This approach could offer new treatment options for patients with HER2-positive cancers, especially those who have failed other treatments.

While there are no specific studies directly combining margetuximab with biosimilars of anti-CTLA-4 or anti-LAG-3, the concept of using such combinations to enhance immune-oncology treatments is an active area of research. The use of biosimilars could potentially make these therapies more accessible due to their lower cost compared to innovator molecules.

Example Combination

A hypothetical approach could involve using margetuximab with a bispecific antibody like MGD013, which targets both PD-1 and LAG-3, to attack both the tumor and immune evasion mechanisms simultaneously. This combination could potentially increase the effectiveness of margetuximab by enhancing the immune system's ability to recognize and destroy cancer cells.

Overall, while there are no direct studies mentioned on using a margetuximab biosimilar with checkpoint inhibitor biosimilars, the concept aligns with ongoing efforts to combine targeted therapies and immunotherapies to improve cancer treatment outcomes.

In the context of immunogenicity testing, a Margetuximab biosimilar can be used as the capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient's immune response against the therapeutic drug through the following steps:

  1. Capture Reagent Preparation:

    • Biotinylation: The Margetuximab biosimilar is biotinylated using appropriate biotinylation kits. This allows it to bind specifically to streptavidin-coated plates, which are typically used in ELISA assays to capture the biotinylated drug.
  2. ELISA Setup:

    • Plate Preparation: Streptavidin-coated microtiter plates are prepared. The biotinylated Margetuximab biosimilar is added to the plates and allowed to bind, creating a high-density antigen surface that captures ADAs from serum samples.
  3. Detection Reagent Preparation:

    • HRP Labeling: A separate portion of the Margetuximab biosimilar is labeled with horseradish peroxidase (HRP) to serve as the detection reagent. This HRP-labeled Margetuximab will bind to the captured ADAs, facilitating detection.
  4. ADA Detection:

    • Sample Addition: Serum samples from patients are added to the plate. Anti-Margetuximab antibodies (ADAs) in the samples bind to the captured biotinylated Margetuximab.
    • Detection: The HRP-labeled Margetuximab detection reagent is then added and allowed to bind to the captured ADAs. A chromogenic substrate like TMB is added to react with the HRP, producing a color change proportional to the amount of ADAs present.
    • Measurement: The color change is quantitatively measured at a specific wavelength using a spectrophotometer, providing a measure of the patient's immune response against the therapeutic drug.

This approach allows for sensitive detection of ADAs, which is crucial for assessing the immunogenicity of therapeutic drugs and monitoring potential immune responses that could impact treatment efficacy and safety.

General Steps in Bridging ADA ELISA:

# Simplified steps in ADA ELISAdef detect_adas():    # Step 1: Prepare capture reagent    # - Biotinylate Margetuximab biosimilar    # Step 2: Set up ELISA    # - Coat plates with streptavidin    # - Add biotinylated Margetuximab to capture ADAs    # Step 3: Prepare detection reagent    # - HRP-label Margetuximab biosimilar    # Step 4: Detect ADAs    # - Add patient serum samples    # - Add HRP-labeled detection reagent    # - Add chromogenic substrate (e.g., TMB)    # - Measure color change (OD at specific wavelength)    return "ADA levels measured"

Key Considerations:

  • Sensitivity and Specificity: The use of sensitive assays is crucial for detecting ADAs, as they can be present at low levels but still significantly impact treatment efficacy.
  • Interference from High Drug Concentration: Techniques such as acid dissociation and solid-phase extraction can help minimize interference from high drug levels in serum samples.
  • Regulatory Guidelines: Studies should adhere to regulatory guidelines for similar biological medicinal products, such as those from the EMA, to ensure appropriate testing and interpretation of immunogenicity data.

References & Citations

1. https://www.ncbi.nlm.nih.gov/gene/2064
2. Nordstrom JL, Gorlatov S, Zhang W, et al. Breast Cancer Res. 13(6):R123. 2011.
3. Bang YJ, Giaccone G, Im SA, et al. Ann Oncol. 28(4):855-861. 2017.
4. https://www.accessdata.fda.gov/drugsatfda_docs/label/2020/761150s000lbl.pdf
5. Catenacci DVT, Kang YK, Park H, et al. Lancet Oncol. 21(8):1066-1076. 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.