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.
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 Omalizumab. Omalizumab (RG-3648) is a recombinant DNA-
derived humanized IgG1 monoclonal antibody that specifically targets immunoglobulin E
(IgE).
Background
IgE is a pivotal antibody in allergic responses, crucially involved in conditions like allergic
asthma. It binds to high-affinity receptors on mast cells and basophils, triggering the release
of inflammatory mediators. The pathogenic role of IgE in allergic inflammation is well-
documented, with multivalent allergens binding to allergen-specific IgEs on sensitized
effector cells, leading to effector cell activation and the release of potent inflammatory
mediators. Therapies targeting IgE, such as omalizumab, have shown efficacy in reducing
exacerbations, symptoms, and medication use in allergic asthma, highlighting the
significance of IgE in allergic diseases1,2.
RG-3648, commonly known as Omalizumab, is a humanized monoclonal antibody that
targets IgE, a key player in allergic responses. Approved for severe allergic asthma and
chronic spontaneous urticaria, Omalizumab binds to serum IgE, preventing its interaction
with cellular IgE receptors. By downregulating high-affinity IgE receptors on inflammatory
cells and reducing eosinophil numbers, Omalizumab improves respiratory symptoms, and
quality of life, and reduces asthma exacerbations. Despite generally being well-tolerated, rare
anaphylactic reactions have been reported. Omalizumab's efficacy extends to various
conditions like allergic rhinitis, atopic dermatitis, and nasal polyps, showcasing its
therapeutic versatility3,4.
Antigen Distribution
IgE is primarily found in the lungs, skin, and mucosal membranes.
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 Omalizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as quantifiable, well-characterized surrogate molecules that mimic the original (reference) Omalizumab in assay conditions.
These biosimilars are integrated into the assay protocol as follows:
Calibration Standards: The biosimilar is serially diluted to generate a standard curve. This curve represents known concentrations of Omalizumab, allowing quantification of drug levels in study serum samples by comparison to the response (typically absorbance) of each standard concentration. The response from unknowns is interpolated against this curve to determine their actual concentrations.
Reference Controls: Alongside the calibration standards, quality controls—often prepared using research-grade biosimilar—are included at low, medium, and high concentrations to verify assay performance within expected ranges for accuracy and precision.
Core principles in PK bridging ELISA with Omalizumab biosimilars:
A capture antibody specific to Omalizumab is coated onto the assay plate.
Calibrators prepared from the research-grade biosimilar (or, if available, reference material) are loaded into wells along with diluted serum samples.
Detection is facilitated by a secondary, labeled anti-Omalizumab antibody; typically, a sandwich ELISA design is used with colorimetric readout proportional to Omalizumab concentration.
The calibration standards thus "bridge" the quantification between test samples and known concentrations.
Why biosimilars can be used: When developed and validated, research-grade biosimilars are biochemically and functionally equivalent or highly similar to the reference product, making them suitable as surrogates for calibration and control in bioanalytical methods like ELISA. Regulatory guidance requires that biosimilar calibration be demonstrated as comparable to the reference, via rigorous analytical validation.
In summary: The use of research-grade Omalizumab biosimilars as calibration standards and reference controls in PK bridging ELISAs enables the accurate measurement of drug concentration in serum samples by establishing a known reference across the assay using material that closely matches the structure and function of clinical Omalizumab.
The primary in vivo models for studying the effects of research-grade anti-Human Immunoglobulin IgE antibodies on tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are humanized xenograft models and syngeneic mouse models.
Key details:
Humanized models: These involve implanting human tumor cells into immunodeficient mice that have been engrafted with human immune effector cells (peripheral blood mononuclear cells, PBMCs, or specific subsets). This allows for administration of anti-human IgE antibodies and assessment of both tumor growth inhibition and TIL composition relevant to the human immune context.
Example: In melanoma xenograft models engrafted with human effector cells, anti-tumor IgE promoted anti-cancer activity and was associated with enhanced immune cell infiltration, notably monocytes/macrophages and activation of pro-inflammatory/interferon/TNF signaling as well as MHC class I/II antigen presentation pathways.
Such models can involve direct antibody administration and analysis of human TILs, including gene expression and subtype assessment.
Syngeneic mouse models: These are models where murine tumor cell lines are implanted into genetically identical mice (fully immunocompetent), allowing study of immune mechanisms in a mouse context.
Example: Murine IgE was targeted to tumors using a biotin-avidin bridge in C57BL/6 mice bearing MC38-CEA-2 colon carcinoma tumors. IgE treatment reduced tumor growth, prolonged survival, and induced TIL recruitment—including eosinophils, CD4, and CD8 T cells—which were individually shown to be critical for the anti-tumor effect.
Syngeneic models remain a pivotal platform for in vivo immunotherapeutic testing, enabling detailed functional analysis and immune cell profiling.
Model Comparison Table:
Model Type
Immune System
Tumor Origin
IgE Antibody Used
TIL Characterization
Cited Source
Humanized xenograft
Human (engrafted)
Human
Human or research-grade
Human TILs
Syngeneic mouse
Mouse (fully intact)
Mouse (syngeneic)
Murine or engineered
Mouse TILs
Additional notes:
Both models have demonstrated that anti-tumor IgE can recruit and activate multiple immune populations (monocytes/macrophages, dendritic cells, T cells, NK cells) and that effective anti-tumor responses rely on cross-talk, antigen presentation, and pro-inflammatory signaling.
Humanized models allow direct testing of anti-human IgE antibodies, whereas syngeneic models typically use murine IgE but can include engineered approaches to target responses.
These models are foundational for both mechanistic in vivo studies and preclinical evaluation prior to clinical translation.
In summary, the most relevant systems are humanized xenograft models (for human IgE antibody and human TIL study in vivo) and syngeneic mouse models (for mechanistic, immunocompetent analysis primarily with murine IgE but occasionally engineered antibodies). Both allow detailed study of tumor inhibition and TIL response following anti-IgE antibody administration.
Researchers use Omalizumab biosimilars primarily in preclinical and translational studies to modulate the IgE pathway in immune-oncology models, often in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars), to study potential synergistic effects on antitumor responses.
Key mechanisms and approaches:
Omalizumab biosimilars, such as those using the same variable regions as the therapeutic antibody, are designed for research use and specifically bind free human IgE and membrane-bound IgE on B cells without cross-linking mast cell-bound IgE; thus, they do not trigger histamine release. This allows researchers to selectively inhibit the IgE signaling pathway in vitro or in animal models expressing human immune components.
Checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 antibodies target distinct steps in T cell activation and tolerance. Anti-CTLA-4 mainly enhances T cell priming and proliferation in lymphoid organs, while anti-LAG-3 affects T cell exhaustion at tumor sites or in the tumor microenvironment (TME). Combining these with omalizumab biosimilars enables exploration of how IgE-dependent inflammation interacts with or modulates the T cell–mediated antitumor immunity fostered by checkpoint blockade.
Research models: Studies often use humanized mice (mice engrafted with human immune cells or tissues) or ex vivo cultures of human immune cells and tumor cells to examine the combinatorial impact. In such models:
Omalizumab biosimilars are administered to neutralize human IgE, altering the activity of IgE-expressing B cells and, by extension, the infiltration and activation of other immune cells such as mast cells and basophils.
Concurrently, checkpoint inhibitors are used to relieve immune suppression by tumor-associated checkpoints (CTLA-4, LAG-3), allowing for enhanced T cell–driven cytotoxicity against tumor cells.
Researchers monitor changes in immune cell phenotypes, cytokine production, tumor burden, and survival to assess synergy (greater effect than individual agents alone).
Rationale for studying combinations:
Synergistic potential: Since Omalizumab targets IgE-mediated pathways (more relevant in allergic and inflammation-driven tumor microenvironments) and checkpoint inhibitors relieve adaptive immune suppression, their joint use can interrogate whether dampening allergic/inflammatory signals while activating cytotoxic T lymphocytes produces stronger or different antitumor effects.
Distinct mechanisms: For example, anti-CTLA-4 and anti-LAG-3 have different effects on the T cell compartment; combining these with Omalizumab could reveal additive or synergistic modulation of both innate/allergic and adaptive immune components.
Study design considerations:
Researchers utilize non-therapeutic biosimilars (research-grade antibodies) rather than clinical formulations, allowing precise dosing and timing for mechanistic studies without safety concerns relevant to humans.
Readouts include tumor growth, flow cytometry of immune infiltration, cytokine panels, IgE levels, and gene expression profiling in tumors and immune organs.
Current limitations:
While the outlined approach is supported by the mechanistic logic and available preclinical tools, published studies directly combining Omalizumab biosimilars with checkpoint inhibitors in cancer models remain limited; most direct evidence comes from studies combining checkpoint inhibitors with each other, not specifically with IgE-targeting agents.
Novel synergy hypotheses are being explored, but confirmatory clinical data are not yet available.
Summary Table: Research Use of Omalizumab Biosimilar with Checkpoint Inhibitors
Agent
Target/Pathway
Primary Immune Mechanism
Typical Use in Models
Omalizumab Biosimilar
IgE
Blocks free IgE and membrane-bound B cell IgE
Suppresses IgE/allergic inflammation
Anti-CTLA-4
CTLA-4
Enhances T cell priming and proliferation
Promotes adaptive immunity
Anti-LAG-3
LAG-3
Reverses T cell exhaustion, especially CD4 cells
Restores effector T cell functions
Researchers design combination protocols with these agents to test whether concurrent IgE blockade and checkpoint inhibition lead to amplified immune-oncology responses (including tumor rejection), enhanced immune cell activation, or reprogramming of the tumor microenvironment.
If further technical details are needed (e.g., dosing strategies or readouts), published preclinical protocols or antibody supplier datasheets (such as those for InVivoSIM anti-human IgE) provide additional specificity for experimental design.
A biosimilar of Omalizumab can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response against the therapeutic drug by leveraging the antibody’s ability to bind free ADA present in patient samples.
Context and Mechanism:
In a bridging ADA ELISA for Omalizumab, you need two forms of Omalizumab (the innovator drug, a biosimilar, or both)—one labeled (e.g., with HRP for detection), and one unlabeled (typically as the capture reagent coated on the plate).
Patient serum potentially containing ADAs (anti-Omalizumab antibodies developed by the immune system) is added. If ADAs are present, they act as a “bridge”: one arm binds the Omalizumab attached to the plate, and the other arm binds the labeled Omalizumab added later.
Detection: Only if the patient sample contains ADAs will the detection reagent (e.g., HRP-labeled Omalizumab biosimilar) become bound to the plate, generating a signal after substrate development.
Why Use a Biosimilar?
An Omalizumab biosimilar can substitute the innovator drug in this assay, provided its structure and epitope presentation are highly similar (a requirement for biosimilarity).
Using a biosimilar as either the capture or detection reagent circumvents issues related to original product availability, intellectual property, or reagent cost, and supports continued monitoring after the innovator supply is discontinued.
Assay Steps (summarized from protocols):
Coat microplate wells with Omalizumab or its biosimilar.
Add patient serum or plasma; if ADAs are present, they bind to the coated Omalizumab.
Add HRP-labeled Omalizumab biosimilar; ADAs “bridge” and link both the coated and labeled Omalizumab.
Wash to remove unbound reagents.
Add chromogenic substrate, measure colorimetric change, reflecting ADA concentration.
Key details:
The bridging format is sensitive and relies on the bivalency of the ADA, therefore only free ADAs (not complexed with Omalizumab in circulation) are detected.
The use of a biosimilar is valid as long as it mimics the original's ADA epitopes sufficiently to ensure assay performance and reliability.
Summary Table: Bridging ADA ELISA Key Components
Step
Reagent
Purpose
Plate coating
Omalizumab (or biosimilar)
Captures ADAs in patient sample
Sample incubation
Patient serum/plasma
Provides possible anti-Omalizumab IgG
Detection
Labeled Omalizumab biosimilar
Binds ADA, completes “bridge”
Readout
Chromogenic/fluorogenic substrate
Measures ADA presence by signal
This approach allows for ongoing, cost-effective, and specific monitoring of immunogenicity in patients treated with Omalizumab, utilizing a biosimilar as a core reagent.
References & Citations
1. Yamazaki T, Inui M, Hiemori K, et al. J Biol Chem. 2019;294(17):6659-6669.
2. Sn K, P K, Dh J, et al. Microbiology spectrum. 2013;1(1).
3. Pelaia G, Gallelli L, Renda T, et al. J Asthma Allergy. 2011;4:49-59.
4. G P, T R, P R, Mt B, R M. Therapeutic advances in respiratory disease. 2008;2(6).
5. Anti Omalizumab Antibody, clone AbD20669. Bio-Rad. Accessed October 5, 2024. https://www.bio-rad-antibodies.com/monoclonal/omalizumab-antibody-abd20669-hca236.html
6. Omalizumab mAb-Based ELISA Assay. Eagle Biosciences. Accessed October 5, 2024. https://eaglebio.com/product/omalizumab-mab-based-elisa-assay/