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 frequently used as analytical standards (calibrators) and quality control (QC) reference materials in pharmacokinetic (PK) bridging ELISA assays to measure drug concentration in serum samples.
In PK bridging ELISA:
A single analytical standard, typically the biosimilar, is chosen as the calibration standard for quantitation of both the biosimilar and the reference originator antibody in serum samples.
The biosimilar standard is used to generate standard curves over a defined concentration range (e.g., 50–12,800 ng/mL prepared in human serum), and all test samples (including those dosed with reference or biosimilar) are measured against this curve.
QC samples are prepared separately with both the biosimilar and reference product to ensure assay accuracy and precision across products.
Why use a biosimilar calibrator?
Use of a single research-grade biosimilar minimizes assay variability and eliminates the need for multiple calibration curves or crossover corrections, enabling a direct, head-to-head bioanalytical comparison of the pharmacokinetics for the reference and biosimilar product.
Validation studies ensure the biosimilar is analytically equivalent to the reference product for assay performance, typically by comparing precision and accuracy between biosimilar and reference QC samples. When equivalence is confirmed, the biosimilar standard is relied upon in all study samples.
Handling and characterization details:
Research-grade biosimilar Naxitamab is produced under controlled conditions (e.g., CHO cell expression, affinity chromatography), provided at known concentrations (e.g., 5 mg/mL), high purity (>95%), and in validated buffer formulations suitable for ELISA and PK analytics.
The biosimilar should have matched antigen specificity (Ganglioside GD2 for Naxitamab), isotype, and host cell (CHO) as the originator to ensure comparable assay binding profiles.
Summary: In PK bridging ELISA, a thoroughly characterized and validated research-grade Naxitamab biosimilar is employed as the common standard to generate calibration curves and serve as a reference control, thereby enabling reliable and equivalent quantification of drug levels for both biosimilar and originator products in serum matrices. This approach aligns with industry and regulatory recommendations to maximize accuracy and comparability in biosimilar PK studies.
Primary Preclinical Models for Anti-GD2 Antibody Tumor Studies
Syngeneic Mouse Models
Syngeneic mouse models are the primary in vivo system for studying the effects of research-grade anti-GD2 antibodies on tumor growth inhibition and for characterizing tumor-infiltrating lymphocytes (TILs). These models use immunocompetent mice and tumors derived from the same genetic background, enabling the evaluation of both direct antibody effects and host immune responses, including TIL dynamics.
B78-D14 Melanoma and EL-4 Lymphoma Models: These are established GD2-positive syngeneic models where anti-GD2 antibody-drug conjugates (ADCs) have demonstrated significant tumor growth inhibition compared to controls. The B78-D14 model, in particular, has been used to show that anti-GD2 ADCs (e.g., ch14.18-MMAE, ch14.18-MMAF) can reduce tumor volume by severalfold, with detailed analysis of antitumor efficacy at multiple time points.
MC38 and TC-1 Models: While not specifically cited for anti-GD2 studies in the provided results, MC38 is a commonly used syngeneic model for immunotherapy research, highlighting the flexibility of these systems for various cancer types. The presence of a functional immune system in syngeneic models allows for robust assessment of TIL populations and their correlation with treatment outcomes.
Humanized Mouse Models
Humanized mouse models—where human immune cells or tissues are engrafted into immunodeficient mice—are not highlighted in the provided results as a primary system for anti-GD2 antibody studies. Most preclinical data for anti-GD2 therapies, especially those focused on TIL characterization, derive from syngeneic rather than humanized systems. The syngeneic approach is preferred when the goal is to study the integrated effects of the antibody and the host immune system, including endogenous TIL responses.
Other Applications
CAR T-Cell Studies: While GD2 is a target for CAR T-cell therapy in neuroblastoma, these studies often use first-generation human CAR constructs in xenograft models, not syngeneic systems, for initial proof-of-concept. Subsequent refinement, including TIL analysis, may transition to syngeneic or transgenic models.
Combination Therapies: Syngeneic models have also been used to test anti-GD2 antibodies in combination with other immunotherapies (e.g., NK cell therapies), providing insights into cooperative mechanisms of tumor control and changes in the tumor immune microenvironment.
Summary Table
Model Type
Example Tumors
Anti-GD2 Antibody Use
TIL Characterization
Notes
Syngeneic
B78-D14, EL-4, MC38
Direct tumor growth inhibition
Yes
Host immune system intact, robust TIL data
Humanized
Not specified
Not highlighted
Limited
Less commonly used for anti-GD2/TIL studies
Xenograft (CAR T)
Neuroblastoma
CAR T targeting GD2
Possible
Focus on CAR T efficacy, not native TILs
Key Points
Syngeneic mouse models (e.g., B78-D14, EL-4) are the primary platform for in vivo evaluation of research-grade anti-GD2 antibodies, enabling direct assessment of tumor growth inhibition and comprehensive TIL profiling.
Humanized models are not prominently featured in the current literature for anti-GD2 antibody studies with a focus on TILs.
Syngeneic systems are critical for understanding how anti-GD2 therapies interact with the host immune system, including NK cells and T cells, to mediate tumor control.
These models are essential for preclinical validation before clinical translation, especially for combination immunotherapies and biomarker discovery.
In summary, syngeneic mouse tumor models are the workhorse for preclinical anti-GD2 antibody research, offering a physiologically relevant context to study tumor growth inhibition and the resulting immune infiltrate, including TILs.
Current Use of Naxitamab in Immune-Oncology Models
Naxitamab is an anti-GD2 monoclonal antibody (mAb) primarily used in clinical and preclinical studies for neuroblastoma, a pediatric solid tumor. It targets GD2, a disialoganglioside overexpressed on the surface of neuroblastoma cells, and it is typically combined with immune-stimulating agents such as granulocyte-macrophage colony-stimulating factor (GM-CSF) to enhance antibody-dependent cellular cytotoxicity (ADCC) in vivo. In preclinical models, the cytotoxic effect of anti-GD2 mAbs like naxitamab includes both direct cell death and activation of complement-dependent cytotoxicity (CDC), but a significant component of the antitumor effect in vivo is attributed to immune cell-mediated responses such as ADCC and antibody-dependent phagocytosis (ADP).
Synergistic Strategies in Preclinical Research
Combination with Checkpoint Inhibitors: Currently, there are no published studies—in the provided search results or in the wider literature summarized here—directly testing naxitamab in combination with checkpoint inhibitors targeting CTLA-4 or LAG-3 in neuroblastoma or other cancers. Most research on synergy with checkpoint inhibitors discusses combination regimens involving anti-PD-1/PD-L1 and anti-CTLA-4 antibodies, sometimes with other immunomodulatory or chemotherapeutic agents, but not with anti-GD2 mAbs.
General Principles from Preclinical Models: When researchers study synergistic effects in complex immune-oncology models, they combine agents that act on different immune pathways—for example, a checkpoint inhibitor (e.g., CTLA-4 or LAG-3 blockade) to reinvigorate T-cell responses and a tumor-targeting antibody (e.g., anti-GD2) to direct immune effector cells to the tumor. The rationale is that checkpoint inhibitors may overcome tumor-induced immune suppression, while the mAb recruits immune cells to the tumor site, potentially amplifying the combined effect. Preclinical evidence supports that targeting multiple immune checkpoints can have additive or synergistic antitumor effects compared to monotherapy.
Translational and Clinical Context: While naxitamab combinations are still mostly explored with GM-CSF and chemotherapy (e.g., irinotecan, temozolomide), the field is moving toward testing novel combinations with immunomodulators, including checkpoint inhibitors, especially in adult solid tumors. In neuroblastoma, clinical trials focus on chemoimmunotherapy, but preclinical studies could explore the addition of checkpoint inhibitors to anti-GD2-based regimens. Such studies would aim to determine if checkpoint blockade enhances the recruitment, activation, or persistence of immune cells engaged by naxitamab, and whether this translates to improved tumor control and survival in animal models.
Potential Experimental Design
In a typical synergistic preclinical study, researchers might:
Treat tumor-bearing mice (or humanized mouse models) with naxitamab (or its biosimilar), alone and in combination with anti-CTLA-4 or anti-LAG-3 biosimilars.
Monitor tumor growth, survival, and immune cell infiltration (e.g., CD8+ T cells, NK cells, macrophages).
Evaluate adverse effects to determine the therapeutic index of the combination.
Compare results to single-agent and control arms to quantify synergy (e.g., using Bliss or Loewe additivity models).
Such experiments could reveal whether dual immune engagement—targeting both the tumor (via anti-GD2) and the immune suppressive microenvironment (via checkpoint blockade)—leads to superior antitumor immunity.
Gaps and Future Directions
No direct evidence: There is currently no published evidence in the provided sources that naxitamab has been combined with CTLA-4 or LAG-3 inhibitors in preclinical models.
Rationale exists: The scientific rationale for such combinations is strong, given the different mechanisms of action and complementary immune effects.
Clinical relevance: If preclinical synergy is demonstrated, this could lead to clinical trials in refractory neuroblastoma, and possibly other GD2-expressing cancers, to address unmet needs in immunotherapy resistance.
Summary Table: Status of Naxitamab and Checkpoint Inhibitor Combinations
Combination
Preclinical Evidence
Clinical Evidence
Rationale for Synergy
Naxitamab + GM-CSF
Yes
Yes
Enhanced ADCC/ADP
Naxitamab + Chemotherapy
Yes
Yes
Direct cytotoxicity + immune recruitment
Naxitamab + CTLA-4/LAG-3 inhibitors
No (in sources)
No
Potential to overcome immune suppression
Conclusion
Researchers have not yet reported using naxitamab (or its biosimilars) in combination with anti-CTLA-4 or anti-LAG-3 checkpoint inhibitors in published preclinical models, but the rationale for such studies is compelling based on the complementary mechanisms of these agents. Future experiments should directly test these combinations in complex immune-oncology models to determine if synergistic antitumor effects can be achieved, potentially informing new clinical strategies for GD2-positive cancers.
Use of Naxitamab Biosimilar in a Bridging ADA ELISA for Immunogenicity Testing
Patients treated with the monoclonal antibody Naxitamab can develop anti-drug antibodies (ADAs), which may affect drug efficacy and safety. To monitor these immune responses, a bridging ADA (anti-drug antibody) enzyme-linked immunosorbent assay (ELISA) is commonly employed. The design of such an assay—specifically, how a Naxitamab biosimilar may be used as the capture or detection reagent—requires careful consideration of assay format and reagent specificity.
Bridging ADA ELISA: Core Principle
In a bridging ELISA, both the capture and detection steps use the drug of interest—here, Naxitamab or a biosimilar—to specifically recognize ADAs in patient serum. The assay works as follows:
Capture: The drug (Naxitamab or biosimilar) is immobilized on the plate, often via biotin–streptavidin coupling if the drug is biotinylated.
Incubation: Patient serum is added. Any ADAs present will bind to the immobilized drug.
Detection: A labeled version of the drug (e.g., HRP-conjugated Naxitamab or biosimilar) is added. If ADAs are present, they will “bridge” the immobilized and labeled drug, leading to a detectable signal.
Signal Generation: A chromogenic substrate reveals the presence and quantity of ADAs.
Role of the Naxitamab Biosimilar
A Naxitamab biosimilar is a non-therapeutic antibody with the same variable region sequence as the therapeutic Naxitamab, designed to recognize the same antigen (GD2) and mimic the therapeutic’s binding properties. In the context of ADA testing:
Capture Reagent: The biosimilar can be coated directly onto the plate (or biotinylated and captured via streptavidin) to bind ADAs from patient serum. This ensures that the assay detects antibodies that recognize the therapeutic’s unique epitopes.
Detection Reagent: The biosimilar can also be conjugated to a detection enzyme (e.g., horseradish peroxidase, HRP) and used to reveal bound ADAs, completing the “bridge” and allowing signal detection.
Specificity: The biosimilar must be nearly identical to the therapeutic in its antigen-binding regions to ensure that the ADA response measured is relevant to the actual drug administered to patients.
Assay Design Considerations
High Sensitivity: Bridging ELISAs are highly sensitive and can detect low levels of ADAs, which is critical for monitoring immunogenicity in patients receiving biologics.
Potential Interferences: Serum matrix effects, soluble drug, and target can interfere. Careful assay optimization and blocking are required to minimize false positives/negatives.
Validation: The biosimilar must be thoroughly characterized to confirm it behaves identically to the therapeutic in the assay, ensuring that the ADA results are clinically meaningful.
Summary Table: Biosimilar Roles in Bridging ADA ELISA
Reagent Role
Purpose
Example in Naxitamab ADA Assay
Capture (Immobilized)
Binds ADAs from patient serum
Naxitamab biosimilar coated on plate
Detection (Labeled)
Binds ADAs already captured, enables signal
HRP-conjugated Naxitamab biosimilar
Conclusion
In immunogenicity testing, a Naxitamab biosimilar can serve as both the capture and detection reagent in a bridging ADA ELISA, enabling sensitive and specific monitoring of patient immune responses against the therapeutic. The biosimilar must closely match the therapeutic’s binding properties to ensure the assay accurately reflects the patient’s ADA profile, which is critical for assessing clinical risk and guiding treatment decisions.
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.