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 ELISAs by preparing defined concentrations of the biosimilar in drug-free serum to generate a standard curve, against which unknown serum samples are quantified. This approach enables the accurate determination of Omalizumab concentration in preclinical and clinical samples even when the studied drug is a biosimilar, not the original reference product.
Essential protocol and context:
Calibration Standards: Research-grade Omalizumab (including biosimilars when necessary) is prepared at known concentrations by spiking it into pooled, drug-free serum to create a set of standards spanning the expected assay dynamic range (e.g., 0.1–12.8 μg/mL).
Quality Controls: Additional aliquots of research-grade Omalizumab are prepared at low, mid, and high concentrations within the validated range to serve as quality controls, confirming assay accuracy and precision.
Bridging ELISA Principle: In a PK bridging ELISA specific for Omalizumab, a capture antibody recognizes one epitope of Omalizumab (or its biosimilar), while a detection antibody binds another. The biosimilar is used identically to the originator to create the standard curve.
Result Quantification: Unknown serum samples from preclinical or clinical studies are run in parallel with the standards. After the immunoassay is developed, the optical densities (signal) of unknowns are interpolated against the standard curve generated with the research-grade biosimilar, enabling quantification in μg/mL or comparable units.
Biosimilar Suitability: The use of a biosimilar standard is appropriate and frequently necessary in biosimilar pharmacokinetic comparability studies (as the biosimilar may differ slightly in glycosylation or structure but must be quantitated equivalently). Regulatory and validation guidance requires ensuring that assay performance is equivalent whether using reference or biosimilar Omalizumab for calibration.
Technical details:
Calibration standards/controls are prepared in the same matrix (e.g., drug-free human or animal serum) as the test samples to minimize matrix effects.
The ELISA must be validated for accuracy, precision, linearity, specificity, and parallelism using the chosen biosimilar as the standard.
In competitive or bridging formats, free (unbound) Omalizumab is measured. Some assays use steps (e.g., acid dissociation) to ensure only free drug is detected.
Regulatory-compliant studies report back-calculation of standard and QC concentrations and demonstrate equivalence between the biosimilar and original product if both are quantified.
Summary Table: Use of Omalizumab Biosimilar in PK Bridging ELISA
Step
Description
Calibration Standard
Serial dilutions of research-grade biosimilar in drug-free serum
Reference Controls
Aliquots at low, medium, and high concentrations within validated range
ELISA Format
Bridging ELISA using anti-Omalizumab capture and detection antibodies
Validation
Accuracy, precision, linearity, specificity, parallelism per guidelines
Application
Quantification of Omalizumab (biosimilar or reference) in serum for PK assessment
This standardization ensures data from biosimilar and reference Omalizumab are comparable in pharmacokinetic analysis, supporting regulatory submissions and scientific rigor.
The primary in vivo models for administering a research-grade anti-Human Immunoglobulin (IgE) antibody to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are mostly humanized mouse models and patient-derived xenograft (PDX) models reconstituted with human immune effector cells.
Context and Supporting Details:
Humanized Mouse Models: In these models, immunodeficient mice are reconstituted with human immune cells (often peripheral blood mononuclear cells, PBMCs, or hematopoietic stem cells) to mimic human immune-tumor interactions. This approach enables the study of TIL composition and function following administration of anti-hIgE antibodies and allows for evaluation of antibody-mediated anti-tumor activity in a setting where human FcεRI-expressing effector cells (such as monocytes, macrophages, and basophils) can participate in immune responses.
Patient-Derived Xenograft (PDX) Models with Human Immune Effector Cells: PDX models involve engrafting human tumor tissues into immunocompromised mice. By further engrafting these mice with human immune cells, researchers can study tumor growth, inhibition, and immune cell infiltration, including TILs, in response to anti-hIgE therapy. These models have been specifically used to characterize immune activation, monocyte/macrophage infiltration, and transcriptomic changes associated with IgE antibody treatment.
Syngeneic Mouse Models: While syngeneic models use immunocompetent mice with mouse-derived tumors, they lack the ability to study human-specific IgE, since mouse immune cells do not express human Fcε receptors in the relevant configuration. These are not the primary models for evaluating human IgE antibodies but can be used for mechanistic studies with mouse IgE.
Typical Experimental Setup:
Recombinant human IgE antibodies (e.g., CSPG4 IgE or MOv18 IgE) are tested in human melanoma or other cancer xenografts.
Human immune effector cells are introduced (from healthy volunteers or cancer patients).
Tumor growth is monitored, and TILs are studied by flow cytometry, immunohistochemistry, and transcriptomics.
Tumor microenvironment changes are characterized, focusing on monocytes/macrophages, T cell priming, and pro-inflammatory signaling pathways.
Key Points:
Humanized xenograft and PDX models reconstituted with human immune cells are the standard for studying anti-hIgE antibody effects on tumor growth and TIL characterization.
Syngeneic models are not suitable for human-specific IgE antibody studies due to species barriers in Fcε receptor biology.
For rigorous study of human TILs in response to anti-hIgE antibodies, researchers typically select humanized or PDX-adapted mouse models. These provide physiologically relevant settings for evaluating both tumor immunology and therapeutic efficacy.
Researchers studying synergistic effects in complex immune-oncology models use combinations of immune-modulating agents—such as the Omalizumab biosimilar (an anti-IgE antibody) together with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars—to probe and enhance anti-tumor immune responses across various experimental contexts. While there are no direct, publicly available studies specifically detailing Omalizumab biosimilars used with anti-CTLA-4 or anti-LAG-3 agents in published preclinical or clinical immune-oncology models, several methodologies and rationales guide this area of research, and parallels can be drawn from established combination immunotherapy strategies.
Context and Rationale:
Immune checkpoint inhibitors (ICIs) such as anti-CTLA-4 and anti-LAG-3 work by releasing brakes on T-cell activation. Combining multiple checkpoint inhibitors is already a validated strategy to enhance anti-tumor immunity, leveraging their distinct but complementary effects on immune subsets.
Omalizumab targets immunoglobulin E (IgE), modulating pathways primarily in allergy and asthma but also in the tumor microenvironment, where IgE and associated effector cells can regulate inflammation and immune activity. The availability of Omalizumab biosimilars like CT-P39 (Omlyclo) increases feasibility for research and combination protocols due to improved access and affordability.
Research Approaches in Complex Models:
In preclinical mouse models, combinations of checkpoint inhibitors (e.g., anti-PD-1 + anti-CTLA-4 or anti-PD-1 + anti-LAG-3) are evaluated for their effects on various immune cell populations (CD4+ helper T cells, CD8+ cytotoxic T cells, Tregs).
Synergistic potential is determined by:
Measuring tumor growth inhibition and survival outcomes.
Profiling immune infiltrates (via flow cytometry or single-cell RNA sequencing).
Assessing cytokine/chemokine changes in the tumor microenvironment.
Mechanistic studies reveal that while anti-CTLA-4 and anti-LAG-3 can potentiate T cell responses differently, their combination (or combination with other immunomodulators) can overcome resistance mechanisms and amplify anti-tumor effects.
While not yet detailed in the literature, a plausible extension is combining Omalizumab biosimilar (to reduce IgE-driven immunosuppression or inflammatory skewing) with checkpoint inhibitors, then measuring additive or synergistic effects on:
Immune cell activation
Antigen presentation
Reduction of suppressive myeloid or mast cell populations
Key Experimental Strategies:
Parallel treatment arms in syngeneic or humanized murine tumor models, comparing each agent (Omalizumab biosimilar, anti-CTLA-4, anti-LAG-3) alone and in combination.
Biomarker assessment to identify enhanced immune signatures (e.g., increased CD8+ T cell infiltration, IFN-γ production) attributable to combinatorial therapy.
Safety and tolerability profiling, since combining immunomodulators increases the risk of immune-related adverse events; attention to cytokine release or allergic manifestations is monitored in these models.
Current Evidence and Knowledge Gaps:
There are currently no published outcomes from clinical or preclinical models explicitly combining Omalizumab biosimilars with checkpoint inhibitors. Most published combination strategies with checkpoint blockade focus on other immunotherapies (e.g., PD-1/PD-L1, CTLA-4, LAG-3, or small molecule agents).
However, the established methods for testing combinations in immune-oncology are directly applicable, and the recent entry of biosimilar Omalizumab is likely to spur such studies, especially given preclinical and translational evidence of IgE's role in the tumor microenvironment.
Summary Table: Mechanistic Features Studied in ICI Combination Models
Agent
Target
Primary Mechanism
Measured Synergy in Models
Omalizumab Biosimilar
IgE
Reduces IgE-mediated immunosuppression; possible effects on tumor-associated inflammation
Not directly published with ICIs, anticipated mechanisms
Anti-CTLA-4
CTLA-4
Enhances T cell priming in lymph nodes, increases cytotoxic T cell pool
Proven synergy with anti-PD-1
Anti-LAG-3
LAG-3
Modulates T cell exhaustion, especially CD4+ T cells
Distinct synergy compared to anti-CTLA-4
In summary, while studies on the use of Omalizumab biosimilar and checkpoint inhibitors together are not yet detailed publicly, the research framework from combination immunotherapy studies guides how researchers would assess synergistic effects—by deploying immune-oncology models, measuring tumor response, and dissecting immune mechanism shifts using modern immunoprofiling methods.
In a bridging ADA (anti-drug antibody) ELISA to monitor immunogenicity against omalizumab, a biosimilar omalizumab can be used as both the capture and detection reagent due to its structural similarity to the therapeutic drug.
Essential context and supporting details:
In this assay format, patient serum (potentially containing anti-omalizumab antibodies) is added to a plate coated with omalizumab (either the originator or a biosimilar).
If ADAs are present in the serum, they bind to the immobilized omalizumab on the plate.
After washing, a second layer of biotinylated (or otherwise labeled) omalizumab biosimilar is added as the detection reagent. This forms a "bridge," since the patient's ADA binds to omalizumab on the plate and simultaneously to the labeled omalizumab in solution.
Detection is accomplished by adding a reporter (such as streptavidin-HRP for biotin-labeled detection omalizumab) and a chromogenic substrate, generating a signal proportional to the amount of ADA in the sample.
This "bridging" format is highly specific because both capture and detection require the ADA to be bivalent (multivalent antibodies), drawing a bridge between the immobilized and labeled omalizumab molecules.
Additional relevant information:
The use of a biosimilar omalizumab as the reagent is permitted if it is shown to be structurally and functionally equivalent to the originator, as biosimilars are designed to have nearly identical amino acid sequences and binding characteristics.
One advantage is that validated biosimilar reagents can provide consistent, scalable supply for assay development and quality control activities, which is particularly relevant after patent expiration of the branded therapeutic.
Controls should always be included to ensure specificity and that detection is not due to cross-reactivity or nonspecific binding.
The bridging format may not detect IgG4 or monovalent ADAs as efficiently, as such antibodies may only bind one omalizumab molecule and fail to "bridge" capture and detection reagents.
In summary, omalizumab biosimilars used as both capture and detection reagents in a bridging ADA ELISA enable the sensitive and selective detection of anti-omalizumab antibodies in patient samples, providing critical information about immunogenicity development against the therapeutic.
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/