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.
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 calibration standards or reference controls in PK bridging ELISA assays to generate accurate and reproducible calibration curves that enable quantification of drug concentrations in serum samples. These calibration standards are essential to measure and compare pharmacokinetics (PK) of different drug products and ensure assay reliability.
Key points on how these biosimilars are used in a PK bridging ELISA:
Calibration Curve Preparation: Research-grade dinutuximab (or biosimilar) is serially diluted in human serum or plasma to produce a range of known concentrations (e.g., 7.45 to 200 ng/mL, or up to 12800 ng/mL). These standards are then run in the ELISA to establish a calibration (standard) curve.
Curve Fitting: The standard curve data are typically fitted using a five-parameter logistic model to relate ELISA signal (optical density) to dinutuximab concentration. This curve is then used to interpolate the concentrations of dinutuximab in unknown test samples.
Single Analytical Standard Approach: The current best practice is to use a single PK assay and a single analytical standard (typically the biosimilar) for quantifying both the reference and biosimilar samples to minimize assay variability and streamline regulatory comparison.
Reference Controls: Samples prepared with reference (innovator) and biosimilar dinutuximab at known concentrations are also analyzed as quality controls (QCs). These ensure the ELISA consistently and accurately quantifies both forms of the antibody and establishes method equivalence.
Bioanalytical Equivalence Testing: During assay development, precision and accuracy data are collected using both the reference and biosimilar across the calibration range. Statistical analysis is performed to demonstrate that both products behave equivalently in the assay, justifying use of the biosimilar standard for all sample quantitation.
Assay Validation: Full method validation is performed with multiple sets of standards and QC samples (including both biosimilar and innovator/reference) across low, medium, and high concentrations to confirm accuracy, precision, specificity, and robustness.
Matrix Matching: Calibration standards and QCs must be prepared in the same biological matrix (e.g., human serum) as the test samples to reflect matrix effects and ensure accurate measurement in clinical samples.
Summary Table: Application in PK Bridging ELISA
Use of Biosimilar Standard
Purpose/Function
Serial dilutions for standard curve
Defines calibration range and signal response
Served as single analytical standard
Minimizes variability, enables bridging
Reference controls (QC) included
Monitors assay performance & comparability
Equivalence testing with innovator
Confirms quantifiability across products
Matrix-matched preparation
Ensures clinical relevance of concentrations
This approach, required for rigorous PK bridging studies, is grounded in regulatory expectations for biosimilar development. It ensures the assay quantitatively measures drug concentrations in clinical and preclinical serum samples regardless of whether the origin is reference or biosimilar dinutuximab, supporting accurate PK comparison and regulatory submission.
The primary in vivo models for studying administration of a research-grade anti-GD2 antibody to assess tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are:
Syngeneic mouse tumor models (using immunocompetent mice with murine tumors engineered or selected to express GD2)
Humanized mouse models (immunodeficient mice engrafted with human immune cells and/or human tumor xenografts, though these are less frequently used for routine TIL assessment in the context of murine antibodies)
Key Details:
Syngeneic Models:
Predominantly used due to their fully functional immune system, which is essential for accurate study of antibody-related immune mechanisms, including TIL characterization and immune-mediated tumor inhibition.
Common models for GD2 studies include:
B78-D14 melanoma: Derived from B16-F1 murine melanoma, genetically modified to express GD2.
EL-4 lymphoma: GD2-expressing variant of a murine lymphoma.
These models allow for repeated measurement of tumor growth inhibition following in vivo administration of murine or chimeric anti-GD2 antibodies (e.g., 14.18).
Immune cell populations, including TILs, can be analyzed by flow cytometry or immunohistochemistry upon harvesting tumors from these models.
Example Evidence:
Anti-GD2 antibody administration in immunocompetent mice bearing B78-D14 melanoma or EL-4 lymphoma tumors showed significant tumor growth inhibition, validating the model's suitability.
Studies combine antibody therapy with other agents (e.g., NK cells, chemotherapy) and evaluate immune infiltration into the tumor, with ongoing assessments for immune cell composition by histology and flow cytometry.
Humanized Models:
Less common for standard anti-GD2 studies due to cost and complexity, but used when it's critical to test human immune responses to tumor and antibody interaction.
May involve engrafting immunodeficient (e.g., NSG) mice with human PBMCs or hematopoietic stem cells and implanting human GD2-positive tumors.
Essential when the research-grade antibody is specific to human GD2 or when assessing interactions with human immune cells, though limitations include partial reconstitution and incomplete recapitulation of TIL diversity.
Summary Table:
Model Type
Immune System
Example Tumors
GD2 Antibody Format
TIL Characterization?
Notable Studies
Syngeneic
Fully murine
B78-D14, EL-4
Murine/chimeric (14.18)
Yes (flow/IHC)
Humanized (PBMC/HSC)
Partially human
Human xenografts
Human or chimeric
Yes, but limited
Additional Notes:
Transgenic mice expressing human GD2 or "humanized" immune repertoires are rare but provide a bridge between mouse and fully humanized models.
The choice of model depends on whether the research aims to characterize mouse or human TILs and the species-specificity of the anti-GD2 antibody under study.
In summary, syngeneic mouse models (such as B78-D14 melanoma and EL-4 lymphoma) are the standard preclinical models for testing research-grade anti-GD2 antibodies and analyzing tumor-infiltrating immune responses in vivo. Humanized models are reserved for specific purposes where human immune interactions must be studied.
A Dinutuximab biosimilar can be used as both the capture and detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response by identifying antibodies formed against the therapeutic drug during treatment.
How a Bridging ADA ELISA Works in This Context:
In a bridging ELISA, the assay leverages the bivalent nature of ADA (antibodies are at least divalent), allowing one arm of an ADA in patient serum to bind to Dinutuximab biosimilar coated on the ELISA plate (capture reagent), while the other arm binds to a labeled form of the same biosimilar added in solution (detection reagent).
The biosimilar must be formatted for detection, often by labeling with an enzyme (e.g., horseradish peroxidase, HRP) or biotin so that a chromogenic reaction can reveal positive wells.
The signal generated correlates to the presence and, to some degree, the amount of ADA, thus reflecting the patient's humoral immune response against Dinutuximab.
Stepwise Mechanism:
Plate Coating: The ELISA plate is coated with the Dinutuximab biosimilar, providing the “capture” surface for ADAs.
Sample Incubation: Patient serum, which may contain anti-dinutuximab ADAs, is added and incubated so that ADAs (if present) will bind to the immobilized drug.
Detection: Labeled Dinutuximab biosimilar is added; it binds to the second antibody-binding site ("the other arm") of any captured ADA, creating a "bridge."
Signal Development: After washing to remove unbound reagents, a substrate is added for the label (e.g., TMB for HRP) to produce a measurable color change.
Interpretation: A positive signal indicates the presence of ADA targeting Dinutuximab, evidencing an immune response against the therapeutic.
Key Considerations:
Biosimilarity: Using a biosimilar is appropriate because antibodies raised against the original drug will generally also bind to the biosimilar if the structure is conserved.
Specificity Controls: It is crucial to confirm the specificity, ensuring that detected ADAs are against Dinutuximab and not unrelated antigens.
Clinical Relevance: Monitoring ADAs is critical, as their presence can affect both efficacy and safety (e.g., through loss of response or allergic reactions).
Assay Limitations: High circulating drug levels may interfere with detection due to competitive binding (drug tolerance issue).
Summary Table: Dinutuximab Biosimilar in Bridging ADA ELISA
Step
Role of Biosimilar
Purpose
Plate coating
Capture reagent
Binds ADA in patient sample
Patient sample
Contains potential ADA
Provides target for biosimilar
Detection reagent
Labeled biosimilar
Binds ADA, enabling signal detection
This approach enables sensitive detection of ADA against Dinutuximab by directly exploiting the drug itself as both the capture and detection reagent, providing an effective method for immunogenicity monitoring.
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.