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 asthe therapeutic antibody Dupilumab. REGN668 (Dupilumab) is an antagonist of human IL-4Rα,the α subunit of the IL-4 receptor. This product is research use only.
Background
IL-4R is a heterodimeric receptor composed of a common subunit (IL-4Rα) that pairs with distinct auxiliary subunits1 to create Type I and Type II IL-4 receptors as well as the IL-13 receptor system2. The IL-4/IL-13/IL-4R axis promotes T helper 2 (Th2) cell differentiation and mediates the pro-allergic adaptive immune response1. The IL‐4R pathway is central to allergic inflammation via its ligands IL-4 and IL-131, driving disease progression in atopic and allergic diseases3. IL‐4R activates effector pathways in target tissues where disease occurs1. Immunotherapies targeting IL-4R are being sought to interrupt the allergic inflammatory response.
REGN668 (Dupilumab) selectively binds to IL-4Rα and inhibits the signaling of type 2 (Th2) cytokines IL-4 and IL-13, thereby inhibiting the release of proinflammatory cytokines, chemokines, and IgE as well as reducing key Th2-associated biomarkers3. An increase in serum levels of IL-4 and IL-13 is seen following blockade. Additionally, dupilumab inhibits IgE production by ex vivo B cells induced by IL-4 treatment 1. Since IL-4Rα is targeted, dupilumab can block both Type I and Type II receptors. However, based on clinical data, dupilumab may not equally inhibit both receptor types1. IL-4 signaling is inhibited via the Type I receptor. While both IL-4 and IL13 signaling are inhibited via the Type II receptor.
Dupilumab has been approved for the treatment of atopic dermatitis and eosinophilic asthma.
Antigen Distribution
Type I IL-4R is composed of IL-4Rα and the γc chain and is expressed on
hematopoietic cells. Type II IL-4R is composed of IL-4Rα and IL-13Rα1 and is expressed on
both hematopoietic and non-hematopoietic cells, such as airway epithelium.
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 Dupilumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the quantitative benchmark to generate the standard curve against which the concentration of Dupilumab in test serum samples is measured.
Context and Supporting Details:
Single analytical standard: In PK bridging ELISA for biosimilars, the current industry best practice is to develop a single PK assay that uses one analytical standard (often the research-grade biosimilar itself) for quantifying both the biosimilar and the reference product in serum samples. This reduces variability and avoids the need for separate assays for each product.
Preparation of calibration curve: Serial dilutions of the research-grade Dupilumab biosimilar are spiked into the same matrix as test samples (usually blank human serum) to generate a calibration (standard) curve. These known concentrations are run in parallel with study samples in every assay.
Measurement process:
The ELISA plate typically contains a capture reagent (often an anti-Dupilumab antibody or anti-IgG4).
The serum samples and standard curve samples containing known Dupilumab concentrations are introduced.
After incubation and washing steps, a detection reagent (secondary antibody) is applied, and a substrate is added for color development.
The optical density (OD) at a specific wavelength (e.g., 450 nm) correlates with Dupilumab concentration.
Unknown concentrations in test samples are interpolated from the standard curve generated with the biosimilar standard.
Establishing comparability: Before using a biosimilar as the standard, the assay method is qualified and validated for precision, accuracy, and equivalence between the biosimilar and the reference product. Side-by-side analysis ensures that both are measured comparably in the assay.
Reference controls: Quality control (QC) samples are also prepared using either the biosimilar or reference product at multiple concentrations to ensure ongoing assay performance.
Summary Table: Use of Biosimilars as PK Bridging ELISA Standards
Purpose
Description
Calibration standard
Serial dilutions of biosimilar establish the standard curve for quantification
Reference controls
QC samples of known concentration ensure assay validity across runs
Equivalence validation
Both biosimilar and reference are tested to confirm comparable detectability and quantitation
Result calculation
Unknowns are interpolated against the biosimilar standard curve for serum concentration measurement
Key Points:
Using a single, well-characterized biosimilar as the standard is recommended to support PK bioequivalence assessments.
Method validation is required to demonstrate that the biosimilar and the originator produce equivalent signals across concentrations in the context of the assay.
This approach is aligned with regulatory expectations for minimizing assay variability in biosimilar development.
This procedure ensures accuracy, robustness, and regulatory compliance in measuring Dupilumab concentrations in clinical and preclinical PK studies.
The primary models where a research-grade anti-IL-4Rα antibody is administered in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models, particularly with murine tumor cell lines, and occasionally genetically engineered mouse models employing allografts.
Essential context:
Syngeneic mouse models involve implanting mouse tumor cells into immunocompetent mice of the same genetic background, allowing for full immune system analysis, including TIL profiling. Commonly used lines include B16F10 (melanoma), RENCA (renal carcinoma), CT26 (colon carcinoma), MC38 (colon adenocarcinoma), Hepa1-6 (liver carcinoma), and EMT-6 (breast carcinoma).
In published studies, anti-IL-4Rα antibodies such as RegN1103 (the murine homolog of the human therapeutic dupilumab) have been tested in genetically engineered or allograft models, such as the alveolar rhabdomyosarcoma (aRMS) orthotopic allograft in SHO mice and models derived from breast cancer cells.
Supporting details:
In the referenced study, murine SHO mice (deficient in B and T cells for allograft acceptance) were orthotopically implanted with murine aRMS tumor cells and treated with a research-grade anti-IL-4Rα antibody (RegN1103). Tumor growth and metastasis were assessed; TIL composition can be characterized as described in related syngeneic model studies.
Tumor-immune profiling of syngeneic models (e.g., B16F10, RENCA, CT26) allows immune cell composition—including TILs like CD8+ T cells, CD4+ T cells, NK cells, and myeloid-derived suppressor cells—to be studied in response to immunotherapies, which may include cytokine pathway inhibitors like anti-IL-4Rα.
Genetically engineered or allograft mouse models may be needed for humanized antibody testing, but true “humanized” models (with partially human immune systems, e.g., NSG mice reconstituted with human PBMCs or HSCs) are rarely used for mouse-specific IL-4Rα antibodies unless the antibody cross-reacts with mouse and human IL-4Rα.
Additional relevant information:
There is currently limited evidence of anti-IL-4Rα use specifically in humanized mouse models for TIL characterization due to receptor species specificity. Most published in vivo work employs syngeneic mouse models with anti-mouse IL-4Rα antibodies.
Key experimental endpoints commonly include tumor growth inhibition, survival, lung and lymph node metastasis rates, and immunophenotyping of TILs via flow cytometry and histology.
Summary Table: Common Model Types Used
Model Type
Example Tumor Cell Lines
Species for Antibody
TILs Characterized
Reference
Syngeneic mouse model
B16F10, RENCA, CT26
Mouse
Yes
Orthotopic allograft (mouse)
U48484 (aRMS), breast
Mouse
Yes
Genetically engineered (mouse)
Various
Mouse
Yes
Humanized mouse model
Rare, if cross-reactive
Human
Possible
-
In summary: Most in vivo studies administering research-grade anti-IL-4Rα antibodies to study tumor growth and TILs use syngeneic mouse models with murine tumor cell lines. Humanized models are seldom used unless the antibody is cross-reactive. TILs—including CD8+ T-cells, CD4+ T-cells, NK cells, and myeloid-derived suppressor cells—are routinely analyzed in these studies.
Researchers currently use Dupilumab biosimilars primarily to manage cutaneous immune-related adverse events (irAEs) such as eczema and bullous pemphigoid induced by checkpoint inhibitor therapies in cancer patients, rather than as direct agents for studying synergy in immune-oncology. There is limited evidence in the published literature regarding the use of Dupilumab biosimilars in combination with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to study synergistic antitumor effects in preclinical or clinical oncological models.
Context and Supporting Details:
Primary role of Dupilumab: Dupilumab, an antibody targeting the IL-4/IL-13 pathways, is mainly used to treat atopic dermatitis and pruritus, including in cancer survivors who develop these conditions as side effects of immune checkpoint inhibitor treatment. Its non-immunosuppressive profile makes it preferable to corticosteroids, as steroids could potentially interfere with the efficacy of checkpoint inhibitors.
Management of irAEs: In case series and reviews, Dupilumab has shown efficacy in treating immune-related cutaneous events triggered by therapies such as anti-CTLA-4, anti-PD-1, and anti-PD-L1. This has led to its consideration as a safer alternative to systemic steroids which could blunt antitumor immunity.
Checkpoint inhibitor combinations: Synergistic antitumor effects are more rigorously studied with combinations of checkpoint inhibitors themselves (e.g., anti-PD-1/CTLA-4, anti-PD-1/LAG-3), with research focusing on their different mechanisms of immune activation and effects on populations such as CD4 and CD8 T cells. These studies often use animal models to characterize how these combinations modulate specific immune cell subsets and resistance mechanisms.
Limitations and Inference:
There is no reported use, from available sources, of Dupilumab biosimilars combined directly with anti-CTLA-4 or anti-LAG-3 biosimilars in immune-oncology models to study synergistic efficacy against tumors. Instead, Dupilumab is used to control adverse cutaneous side effects arising from checkpoint inhibitor treatment, enabling patients to continue immunotherapy with reduced toxicity.
The absence of data about Dupilumab's synergy with checkpoint inhibitors in tumor models likely reflects its role in affecting Th2-driven pathways, rather than direct modulation of antitumor immunity (which is driven by Th1 and cytotoxic responses targeted by checkpoint inhibitors).
Summary Table: Use of Dupilumab vs. Checkpoint Inhibitor Combinations in Immuno-Oncology Research
In conclusion, Dupilumab biosimilars are valuable for managing immunotherapy-induced skin side effects in oncology patients, supporting continued checkpoint inhibitor therapy, but are not currently used experimentally in combination with CTLA-4 or LAG-3 biosimilars to investigate antitumor synergy in immune-oncology models.
A Dupilumab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response by taking advantage of the bivalent nature of ADAs generated against the therapeutic drug.
Key steps in a bridging ADA ELISA:
Coating or labeling: The Dupilumab biosimilar is either immobilized on the ELISA plate (capture) or conjugated with a detection label (such as biotin or HRP).
Sample incubation: Patient serum samples, which may contain anti-Dupilumab ADAs, are incubated so that the ADAs can bind to the immobilized Dupilumab biosimilar.
Detection: A labeled Dupilumab biosimilar is added as the detection reagent, which binds to the remaining free antigen-binding sites of the patient's ADA, forming a "bridge" between capture and detection reagents via the ADA in the sample.
Signal generation: The signal from the detection reagent is proportional to the amount of ADA present in the patient sample.
This assay design is termed "bridging" because the bivalent ADAs simultaneously bind to both the capture and detection molecules, producing a measurable signal only if patient anti-drug antibodies are present. Biosimilars are appropriate for this use because they possess the same or highly similar antigenic epitopes as the reference Dupilumab, ensuring ADAs generated against the drug bind equivalently to the biosimilar forms used in the assay.
Typical workflow example:
Biotinylated Dupilumab biosimilar is captured via streptavidin on the plate.
Patient serum (potentially containing ADAs) is added.
HRP-labeled Dupilumab biosimilar is applied; if ADAs are present, a sandwich forms between the immobilized and HRP-conjugated biosimilars via the patient's bivalent ADA.
Addition of chromogenic substrate leads to a color change proportional to ADA concentration.
Assay advantages:
High specificity for patient-generated ADAs.
Sensitivity suitable for both IgM and IgG responses.
Relevance: The biosimilar, being highly similar to the originator product, is ideal for capturing a clinically meaningful immune response.
Take-home point: Dupilumab biosimilars provide a reliable and equivalent antigenic source compared to the reference drug, allowing them to serve as either the capture or detection agent in a bridging ELISA, ensuring robust and specific detection of patient anti-Dupilumab antibodies in immunogenicity monitoring.
References & Citations
1. Harb H, Chatila TA. Clin Exp Allergy. 50(1):5-14. 2020.
2. Kovalenko P, DiCioccio AT, Davis JD, et al. CPT Pharmacometrics Syst Pharmacol. 5(11):617-624. 2016.
3. Shirley M. Drugs. 77(10):1115-1121. 2017.
4. Hamilton JD, Suárez-Fariñas M, Dhingra N, et al. J Allergy Clin Immunol. 134(6):1293-1300. 2014.
5. Jonstam K, Swanson BN, Mannent LP, et al. Allergy. 74(4):743-752. doi: 2019.
6. Blauvelt A, de Bruin-Weller M, Gooderham M, et al. Lancet. 389(10086):2287-2303. 2017.
7. Rabe KF, Nair P, Brusselle G, et al. N Engl J Med. 378(26):2475-2485. 2018.
8. Wollenberg A, Beck LA, Blauvelt A, et al. Br J Dermatol. 182(5):1120-1135. 2020.