Anti-Human GD2 (Dinutuximab) – Fc Muted™

Anti-Human GD2 (Dinutuximab) – Fc Muted™

Product No.: G165

- -
- -
Product No.G165
Clone
APN311
Target
GD2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
human ganglioside GD2, disialoganglioside
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Recommended Isotype Controls
Immunogen
Humanized antibody derived from mouse clone ch14.18
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
Additional Applications Reported In Literature ?
ELISA
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 Dinutuximab. This product is for research use only. Dinutuximab activity is directed against human disialoganglioside (GD2).
Background
Neuroblastoma is an extracranial childhood cancer that accounts for 12% of cancer deaths in children1. Neuroblastomas highly express the antigen GD2, a glycolipid which assists in the attachment of tumor cells to the extracellular matrix2.

Dinutuximab is a mouse-human chimeric monoclonal antibody to disialoganglioside (GD2) 3. Engagement of dinutuximab with GD2 triggers antibody dependent cell cytotoxicity and complement dependent cytotoxicity, the effectiveness of which is increased by coadministration with granulocyte-macrophage colony stimulating factor (GM-CSF), interleukin-2 (IL-12), and 13-cis retinoic acid (isotretinoin).

Dinutuximab, also known as ch14.18, was developed as an IgG1 human/mouse chimeric switch variant of murine monoclonal antibody 14.18 4. Dinutuximab is composed of the variable heavy- and light-chain regions of the murine anti-GD2 mAb 14.18 and the constant regions of human IgG1 heavy-chain and κ light-chain. Dinutuximab is produced in the murine myeloma cell line SP2/O and has an approximate molecular weight of 150 kDa 5. Studies have also tested the efficacy of dinutuximab grown in Chinese hamster ovary (CHO) cells (ch14.18/CHO)6,7.
Antigen Distribution
GD2 is a cell surface glycolipid present in low concentrations on skin, neural or peripheral nerve cell surfaces. GD2 is overexpressed on neuroblastoma cells, most melanoma, and some other tumors.
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 Dinutuximab biosimilars are used as reference standards in PK bridging ELISA assays by serving as the calibration curve standards and quality control samples, enabling accurate quantification of drug concentration in serum and comparison between biosimilar and reference products.

  • Calibration Standards: Serial dilutions of the research-grade biosimilar are prepared at known concentrations (e.g., 7.45 to 200 ng/mL, or higher ranges depending on assay sensitivity). These standards create the calibration curve against which unknown serum sample concentrations are measured.
  • Reference Controls (Quality Control Samples): Biosimilars are also spiked into serum at predefined concentrations for quality control (QC), ensuring assay accuracy and precision.
  • Bridging ELISA Purpose: In PK bridging ELISAs for biosimilars, it is best practice to use a single analytical standard—typically the biosimilar itself—to quantify both biosimilar and reference (originator) products in study samples. This approach reduces assay variability and improves quantitative comparability.
  • Validation and Comparability: Before using the biosimilar as the calibration standard, its bioanalytical equivalence to the reference product is rigorously established. This includes demonstrating comparable accuracy, precision, and recovery for both products in the assay. Selectivity studies confirm that human IgG or other serum components do not interfere with the measurement.
  • Curve Fitting: Calibration curves in dinutuximab PK ELISAs typically use a five-parameter logistic curve fit to map known biosimilar concentrations against ELISA signal, allowing for reliable interpolation of drug concentrations in patient serum samples.
  • Assay Standardization: Commercial kits, such as KRIBIOLISA, offer ready-to-use calibration standards spanning relevant clinical PK ranges; research-grade biosimilars serve as the material for these standards.

In summary, research-grade dinutuximab biosimilars function both as calibration standards for quantification and as reference controls to validate PK assay performance, enabling robust bridging between biosimilar and reference drug concentration measurements in serum samples.

Research-grade anti-GD2 antibodies are primarily studied in vivo using syngeneic mouse models to evaluate tumor growth inhibition and characterize the resulting tumor-infiltrating lymphocytes (TILs). Here are the key aspects of these models and how they are used:

Syngeneic Mouse Models

  • B78-D14 Melanoma Model: This model is used to assess the antitumor efficacy of anti-GD2 antibody-drug conjugates (ADCs). ADCs targeting GD2 effectively inhibit tumor growth in mice with subcutaneous B78-D14 melanoma tumors, highlighting the potential of GD2 as a target for cancer therapy.
  • EL-4 Lymphoma Model: Similar to the B78-D14 model, the EL-4 model is used to evaluate the efficacy of anti-GD2 ADCs. Despite more aggressive tumor growth in this model, treatments like ch14.18-MMAF show significant antitumor effects.

Humanized Models

While humanized models are not frequently mentioned in the context of anti-GD2 antibody studies for tumor growth inhibition in the provided texts, they are invaluable for studying xenografts or humanized immune systems in mice. Humanized models would be more relevant for studying the effects of anti-GD2 antibodies in a human-like immune context, especially when combined with other therapies such as CAR T cells or natural killer cells, as reported in studies involving dinutuximab (ch14.18) in triple-negative breast cancer. However, the primary focus for anti-GD2 antibody studies in terms of syngeneic models is on evaluating immune interactions and antitumor effects in an immunocompetent murine environment.

Tumor-Infiltrating Lymphocytes (TILs)

While the primary focus of syngeneic models is on evaluating the direct antitumor effects of anti-GD2 antibodies, these models also facilitate the study of TILs. By analyzing the immune response within these models, researchers can gain insights into how anti-GD2 antibodies influence the tumor microenvironment and modulate TIL populations, which is crucial for developing effective immunotherapies. However, specific studies focusing on TIL characterization in the context of anti-GD2 therapy are less detailed in the provided sources.

Researchers investigating synergistic effects in complex immune-oncology models often use Dinutuximab biosimilars (such as dinutuximab beta) in combination with other checkpoint inhibitors—including anti-CTLA-4 or anti-LAG-3 biosimilars—to understand how these agents can work together to enhance anti-tumor immunity.

Essential context and supporting details:

  • Dinutuximab biosimilars target the GD2 antigen found on neuroblastoma and some other tumors and are approved for use in combination with cytokines and chemotherapy.
  • While most clinical studies to date have paired dinutuximab primarily with cytokines (e.g., GM-CSF, IL-2), and chemotherapeutic agents (e.g., temozolomide, irinotecan), the broader strategy in immune-oncology is to combine monoclonal antibodies with immune checkpoint inhibitors to potentiate antitumor T-cell responses.
  • Checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 act on T-cell regulation pathways. Combining these with dinutuximab can:
    • Directly target tumor cells via antibody-dependent mechanisms.
    • Release inhibitory "brakes" on T-cell mediated immune responses, facilitating a more robust attack against tumor cells.
    • Address multiple aspects of tumor immune evasion: checkpoint inhibitors can reinvigorate exhausted T cells, while dinutuximab can drive antibody-dependent cellular cytotoxicity (ADCC)—often engaging effector cells such as NK cells and macrophages.

Synergy in immune-oncology models:

  • In preclinical studies (typically mouse models), researchers administer dinutuximab biosimilars with checkpoint blockers, observing effects on tumor growth, immune cell activation, and overcoming resistance mechanisms.
  • For anti-CTLA-4 or anti-LAG-3 combinations, recent research demonstrates mechanistic differences in how these regimens activate the immune system:
    • Anti-PD-1/LAG-3 combinations rely on CD4 T-cell activity, reducing regulatory T cells (Tregs) and increasing helper T-cell activity—ultimately enhancing CD8 cytotoxic T-cell activation.
    • Anti-PD-1/CTLA-4 combinations showed more direct accumulation and activation of cytotoxic CD8 T-cells—less dependent on CD4 T-cell presence.
  • Applying these approaches with dinutuximab biosimilars may allow researchers to:
    • Dissect how antibody therapies and checkpoint inhibition can complement each other to maximize immune-mediated killing of tumor cells.
    • Identify biomarkers and immune subtypes most likely to benefit from combinatorial approaches (e.g., assessing GD2 expression status for dinutuximab responsiveness, and T-cell infiltration/exhaustion for checkpoint inhibitor efficacy).

Additional relevant information:

  • Current ongoing trials in neuroblastoma and other tumors explore various combinations with immunomodulators, including multi-antibody regimens potentially targeting other antigens (e.g., B7-H3).
  • The choice of checkpoint inhibitors may be tailored depending on tumor immune context—since anti-CTLA-4 and anti-LAG-3 differ in their mechanisms and the immune cell populations they affect.
  • Dinutuximab's effectiveness can depend on tumor GD2 expression, and combining it with checkpoint blockade may help overcome resistance in low-antigen-expressing tumor subpopulations.

Summary Table: Mechanistic Contribution in Combination Models

AgentImmune MechanismKey Synergy with Dinutuximab Biosimilars
Dinutuximab biosimilarsADCC via effector cells, targets GD2+ tumor cellsMay provide direct cytotoxicity against neuroblastoma, synergizes with immune activation
Anti-CTLA-4 biosimilarsReleases CTLA-4 mediated inhibition on T-cellsPromotes broader T-cell activation, increases CD8 cytotoxic cells
Anti-LAG-3 biosimilarsInhibits LAG-3-mediated T-cell exhaustionEnhances CD4 T-cell helper activity, reduces Tregs

The full mechanistic synergy and best combinatorial strategies remain active areas of preclinical and translational research, with studies increasingly focusing on immune cell subtypes, tumor antigenicity, and resistance mechanisms.

In the context of immunogenicity testing, a bridging ELISA can be adapted to use a therapeutic drug like dinutuximab (or its biosimilar) as the capture or detection reagent to monitor a patient's immune response. Here's how this can be done:

Bridging ELISA Setup for ADA Detection

  1. Capture Reagent: The therapeutic drug or its biosimilar (e.g., dinutuximab) is immobilized (coated) onto the surface of microtiter plates. This step captures anti-drug antibodies (ADAs) from patient serum samples.

  2. Detection Reagent: A labeled form of the therapeutic drug (e.g., HRP-labeled dinutuximab) is added to bind the captured ADAs. The label (e.g., horseradish peroxidase, or HRP) is then used for detection by reacting with a chromogenic substrate (like 3,3’,5,5’-tetramethylbenzidine, or TMB).

  3. Detection Process: The reaction between the labeled drug and the chromogenic substrate changes the color of the solution, indicating the presence of ADAs in the patient's serum. The intensity of the color change is proportional to the amount of ADAs present.

  4. Data Analysis: The results are typically quantified using a standard curve generated from calibrator samples with known concentrations of ADAs. This allows for the measurement of the ADA concentration in patient samples.

How Dinutuximab Biosimilar Acts as Capture or Detection Reagent

  • Capture Reagent: Using the biosimilar as the capture reagent involves immobilizing it onto the ELISA plate. It acts as an antigen to capture ADAs from the patient's serum sample.

  • Detection Reagent: The biosimilar is labeled with a detectable tag (e.g., HRP) and used to bind the captured ADAs. This step is crucial for visualizing and quantifying the ADA response.

Using a biosimilar of dinutuximab in this manner helps ensure that the assay is sensitive and specific for detecting ADAs against the therapeutic drug, as biosimilars are structurally similar to the original drugs and can accurately mimic their immunogenic properties.

Challenges and Considerations

  • Interference from High Drug Concentrations: High levels of circulating drug can interfere with ADA detection in ELISAs. Techniques like acid dissociation or using solid-phase extraction can help mitigate these issues.

  • Assay Validation: The ELISA must be thoroughly validated to ensure accuracy and reliability in detecting ADAs. This includes testing for specificity, sensitivity, and reproducibility.

In summary, using a dinutuximab biosimilar in a bridging ELISA allows for the detection and quantification of ADAs against the therapeutic drug, providing valuable insights into a patient's immune response over time.

References & Citations

1. Aust Prescr. 43(6):212-213. 2020.
2. Hoy SM. Target Oncol. 11(2):247-253. 2016.
3. Dhillon S. Drugs. 75(8):923-927. 2015.
4. Mueller BM, Romerdahl CA, Gillies SD, et al. J Immunol. 144(4):1382–1386. 1990.
5. https:// www.unituxin.com/full-prescribing-information.pdf
6. Ladenstein R, Weixler S, Baykan B, et al. MAbs. 5(5):801-809. 2013.
7. Ladenstein R, Pötschger U, Valteau-Couanet D, et al. Lancet Oncol. 19(12):1617-1629. 2018.
8. Barker E, Mueller BM, Handgretinger R, et al. Cancer Res. 51(1):144–149. 1991.
9. Yu AL, Gilman AL, Ozkaynak MF, et al. N Engl J Med. 363(14):1324–1334. 2010.
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.