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 Naxitimab. Clone hu3F8 recognizes Human GD2. This product is for research use only.
Background
Neuroblastoma is an extracranial childhood cancer that accounts for 12% of cancer deaths in children 1. Neuroblastomas highly express the antigen GD2, a glycolipid that assists in the attachment of tumor cells to the extracellular matrix 2.
Naxitamab (hu3F8) is a humanized IgG1 anti-GD2 monoclonal antibody developed for the treatment of neuroblastoma, osteosarcoma, and other GD2-positive cancers 3. Naxitamab was engineered by grafting the complementarity determining regions of murine 3F8 (m3F8) onto human IgG1 frameworks and optimizing the sequences for stability, binding kinetics to GD2, and efficiency in antibody dependent cell-mediated cytotoxicity (ADCC) 4. Naxitamab demonstrates direct cytotoxicity against neuroblastoma cell line LAN-1 in vitro and induces complement dependent cytotoxicity as well as ADCC 3,4. Naxitamab has low level cross-reactivity with the ganglioside GD1b and no cross-reactivity with human N-CAM 4.
The chemical name for naxitamab is immunoglobulin G1, anti-(ganglioside GD2) (humanized mus musculus clone hu3f8 gamma1-chain), disulfide with humanized mus musculus clone hu3f8 light-chain, dimer 3.
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 Naxitamab biosimilars are used as calibration standards or reference controls in PK bridging ELISA assays to enable accurate and comparable measurement of drug concentrations in serum samples across biosimilar and reference products. This ensures that both the biosimilar and reference (originator) drugs can be quantified reliably in a single standardized method, supporting pharmacokinetic (PK) equivalence studies.
Essential Context and Supporting Details
Calibration Standard in Single-Assay Approach: In PK bridging ELISA, a single PK assay often uses the biosimilar (here, research-grade Naxitamab) as the analytical standard (calibrator) for quantifying both biosimilar and reference product concentrations in serum samples. This approach is considered best practice because it minimizes inter-assay variability and ensures data consistency across clinical studies comparing the biosimilar and reference (originator) drugs.
Establishing Analytical Equivalence: Before using the biosimilar as the calibrator, analytical equivalence between the biosimilar and reference product must be demonstrated within the assay. This involves:
Preparing precision and accuracy datasets for both biosimilar and reference products.
Applying statistical analysis (often comparing the 90% confidence interval to a predefined equivalence margin, e.g., 0.8–1.25).
If equivalence is established, the biosimilar standard is used throughout method validation and sample analysis.
Method Validation and Use: During method validation:
Standard curves are generated using the biosimilar in human serum at a range of concentrations, e.g., 50–12,800 ng/mL in the referenced study.
Quality Control (QC) samples containing either the biosimilar or reference product are quantified using the biosimilar-based standard curve.
Assay parameters such as sensitivity, specificity, accuracy, and precision must meet regulatory and industry guidance for PK bioanalytical methods.
Role in Bridging ELISA: The biosimilar's critical quality attributes (CQAs) are closely matched to the reference product, supporting the bridging ELISA design, where antibodies and assay conditions are chosen to capture and detect both products equally. This ensures comparable analytical performance, a regulatory expectation for biosimilar PK studies.
Additional Information
Formulation and Suitability: Research-grade Naxitamab biosimilars intended for such use are highly purified (e.g., >95% by SDS-PAGE), formulated without interfering additives, and supplied at high concentration and purity to serve as reliable calibrators and QC materials.
Regulatory and Industry Standards: This approach aligns with modern regulatory expectations and industry consensus for ligand binding assays in biosimilar development, as it facilitates direct demonstration of PK similarity and method robustness.
Reference Control Use: In addition to being used as calibration standards in standard curve preparation, research-grade biosimilars can also function as consistent reference controls in assay runs to confirm long-term assay reliability.
In summary, research-grade Naxitamab biosimilars are central to the analytical validity of PK bridging ELISA by serving as calibration standards and reference controls, provided their equivalence to the reference product is validated within the assay according to regulatory and scientific best practices.
Syngeneic mouse models are the primary in vivo models used for administering research-grade anti-GD2 antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs), due to their fully functional murine immune systems that allow assessment of immune-mediated tumor responses.
Supporting details:
Syngeneic Models: These use mouse tumor cells transplanted into immunocompetent mice of the same genetic background, ensuring intact and relevant mouse immune responses. Examples include B78-D14 melanoma and EL-4 lymphoma models, where anti-GD2 antibodies lead to significant inhibition of tumor growth. Such models facilitate characterization of TILs following antibody administration because all immune cell populations are present and functional.
Model features for TIL analysis: Well-characterized syngeneic models allow researchers to evaluate both the efficacy of anti-GD2 therapies and detailed immune cell infiltration. These models routinely measure baseline TIL populations and immune responses, enabling post-treatment characterization of immune changes within the tumor microenvironment. This is essential for understanding the mechanism of tumor control and the immunological effects of the therapy.
Humanized Models: While syngeneic models are the standard for murine antibody and immune response studies, humanized mouse models (immunodeficient mice engrafted with human immune cells and/or human tumor xenografts) are used when specifically evaluating a humanized anti-GD2 antibody’s effects on human immune cells and TIL composition. However, limitations exist due to incomplete human immune system reconstitution.
Summary Table:
Model Type
Immune System
Tumor Origin
Study Focus
Reference
Syngeneic Mouse
Murine (intact)
Mouse
Tumor inhibition, TIL analysis
Humanized Mouse
Human (incomplete)
Human xenograft
Human TILs, human antibody effect
Most common antibodies and targets: Research often utilizes murine, chimeric (e.g., ch14.18), or humanized anti-GD2 antibodies in these models to inhibit tumor growth and study immune responses.
Typical experimental endpoints: Besides tumor growth inhibition, studies include survival analysis, toxicity monitoring, immune histology, and detailed flow cytometric phenotyping of TILs.
Researchers select syngeneic mouse models for direct assessment of anti-GD2 efficacy and immune modulation due to their intact immune systems, with humanized models reserved for studies prioritizing human-specific immune interactions and TIL characterization.
Researchers investigating synergistic effects in immune-oncology models typically use naxitamab biosimilars—which target GD2, a surface antigen found in several cancers—in combination with other immune checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to enhance anti-tumor immunity and overcome resistance seen in monotherapy.
In preclinical and translational studies, these combinations are designed to:
Target multiple immune regulatory pathways: For example, naxitamab enhances antibody-dependent cellular cytotoxicity (ADCC) against tumor cells expressing GD2, while checkpoint inhibitors such as anti-CTLA-4 act to relieve T cell suppression, each operating through distinct immune mechanisms.
Model synergistic effects: Utilizing complex in vitro co-culture systems and in vivo murine models, researchers examine the additive or synergistic impact of combining tumor cell–directed antibodies (like naxitamab) with agents that block T cell regulatory pathways (like CTLA-4 or LAG-3). These studies assess outcomes such as tumor regression, immune cell infiltration, and cytokine profiles.
Mechanistic studies: Combination regimens are used to understand tumor microenvironment changes. For instance, anti-CTLA-4 increases the priming and proliferation of T cells, while naxitamab augments targeted tumor cell killing; their concurrent use seeks to maximize both arms of the anti-tumor immune response.
Early phase clinical testing: After promising preclinical synergy, phase I/II clinical trials may test these combinations for safety, toxicity, and efficacy. Notably, combining multiple checkpoint inhibitors (such as anti-CTLA-4 plus anti-PD-1) has demonstrated improved responses in some cancers, though with increased toxicity risk.
Checkpoint inhibitor biosimilars (e.g., ipilimumab for CTLA-4, relatlimab for LAG-3) are often sourced for research from commercial antibody suppliers. Researchers optimize dosing schedules, assess toxicity profiles, and perform immune monitoring in these models to determine the best combination regimens.
Direct evidence for naxitamab biosimilar combinations with checkpoint inhibitors is still emerging. Most published data highlight combination regimens involving chemotherapy, naxitamab, and immune stimulants such as GM-CSF, especially in relapsed/refractory neuroblastoma. The logic, extrapolated from checkpoint inhibitor literature, is that combining naxitamab with inhibitors of CTLA-4 or LAG-3 holds promise for superior anti-tumor effects through reciprocal immune activation.
In summary:
Naxitamab biosimilars are combined with checkpoint inhibitors in complex immune-oncology models to study synergistic immune activation, tumor targeting, and resistance mechanisms.
Research strategies include in vitro and in vivo experimentation, mechanistic immunology studies, and subsequent clinical translation.
Published clinical evidence with checkpoint inhibitor combinations specifically with naxitamab is still limited, but general principles from combination immunotherapy inform ongoing and future work.
A Naxitamab biosimilar is used as a capture and/or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response by detecting antibodies that the patient may develop against the therapeutic drug naxitamab.
In a bridging ADA ELISA:
The assay takes advantage of the bivalent nature of anti-drug antibodies, which can simultaneously bind two identical drug molecules.
Naxitamab biosimilar can be used in two key roles:
As the capture reagent: The biosimilar is immobilized on the ELISA plate to "capture" any anti-naxitamab antibodies present in the patient’s serum.
As the detection reagent: Typically, a labeled form of the naxitamab biosimilar (e.g., biotinylated or HRP-conjugated) is added after patient serum incubation to "detect" if ADA has formed a bridge between the capture and detection molecules.
Basic workflow:
Plate coating: The ELISA plate is coated with naxitamab biosimilar.
Sample incubation: Patient serum, which may contain ADAs, is added; ADAs will bind to the immobilized naxitamab via one of their antigen-binding sites.
Detection: Labeled naxitamab biosimilar is added. If ADA is present, the other antigen-binding site of the ADA binds the labeled naxitamab, forming a “bridge.”
Signal generation: A substrate is added if the detection reagent is enzyme-labeled, leading to a measurable signal proportional to the ADA concentration.
Why use a biosimilar?
Research-grade naxitamab biosimilars are structurally and functionally similar to the original drug, ensuring that any patient-generated ADA will recognize both.
Using a biosimilar can minimize interference from drug-related impurities present in commercial formulations, and helps standardize and scale research assays.
Key technical considerations:
The biosimilar must be highly pure and free from aggregates or contaminants, as these can cause non-specific binding.
Both capture and detection reagents must present the therapeutic’s antigenic epitopes in a conformation recognized by potential ADA.
Blocking steps and controls are necessary to reduce background and distinguish specific ADA from non-specific interactions.
Application:
This assay enables highly sensitive detection of ADAs in patient samples during naxitamab therapy, supporting immunogenicity monitoring and therapeutic decision-making.
This approach is fundamental in clinical trials and routine clinical labs for safety and efficacy assessments of therapeutic antibodies like naxitamab.
ADA bridging ELISAs are widely used for anti-drug antibody detection across therapeutic monoclonal antibodies and are the cornerstone of immunogenicity testing due to their sensitivity and specificity.
References & Citations
1. Aust Prescr. 43(6):212-213. 2020.
2. Hoy SM. Target Oncol. 11(2):247-253. 2016.
3. Markham A. Drugs. 81(2):291-296. 2021.
4. Cheung NK, Guo H, Hu J, et al. Oncoimmunology. 1(4):477-486. 2012.