Anti-Human IL-4Rα (CD124) (Dupilumab) – Fc Muted™

Anti-Human IL-4Rα (CD124) (Dupilumab) – Fc Muted™

Product No.: I-1265

- -
- -
Product No.I-1265
Clone
REGN668
Target
IL-4Rα
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-4R-alpha, CD124, IL4R
Isotype
Human IgG4κ
Applications
B
,
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Unknown
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
≤ 1.0 EU/mg as determined by the LAL method
Purity
≥95% by SDS Page
≥95% monomer by analytical SEC
Formulation
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.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
B,
ELISA,
FA
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 Dupilumab. REGN668 (Dupilumab) is an antagonist of human IL-4Rα, the α subunit of the IL-4 receptor.
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.
Ligand/Receptor
IL-4, IL-13
NCBI Gene Bank ID
UniProt.org
Research Area
Apoptosis
.
Biosimilars
.
Cell Biology
.
Cell Death
.
Immunology
.
Inflammatory Disease
.
Signal Transduction
.
Transcription Factors
.
Tumor Suppressors
.
Pro-Inflammatory Cytokines

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 Dupilumab biosimilars are routinely used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately measure drug concentrations in serum samples. In a PK bridging ELISA, a single analytical standard—typically the biosimilar itself—is chosen to calibrate the assay for quantification of both biosimilar and reference product concentrations in clinical samples.

This approach involves several steps and key considerations:

  • Assay Calibration: The biosimilar standard, prepared at known concentrations in human serum, is used to generate the standard curve that correlates optical density (OD) measurements to drug concentration.
  • Measurement of Unknowns: Serum samples from subjects are run in parallel. The concentration of Dupilumab—whether originator or biosimilar—is inferred from the standard curve by comparing the OD values of unknown samples to those of the calibrator.
  • Quality Control & Method Validation: Method validation includes extensive testing to ensure biosimilar and reference product are bioanalytically equivalent within the same assay; results are validated by generating precision and accuracy datasets and statistical analysis to confirm both products are measured reliably and without bias.
  • Regulatory and Scientific Rationale: Using a single biosimilar calibrator standard reduces assay variability and avoids the need for crossover analysis, which is crucial for robust bioequivalence assessments and minimization of confounding factors.

Reference controls (often batches of biosimilar and reference drug at fixed concentrations) are included to monitor assay consistency and performance during validation and sample testing.

Key ELISA implementation details:

  • Microplates are typically precoated with anti-Dupilumab antibodies.
  • After sample addition and incubation, unbound components are washed away.
  • Detection is accomplished via HRP-conjugated secondary antibodies, followed by substrate development and colorimetric readout at 450 nm.
  • The standard curve is used for concentration calculations.

Summary Table: Dupilumab Biosimilar Use in PK Bridging ELISA

PurposeMaterial UsedHow UsedValidation Requirement
Calibration standardResearch-grade biosimilarSerial dilutions to create standard curve in human serumAnalytical equivalence for calibration
Reference controlsBiosimilar & referenceQC samples at defined concentrations, tested in parallelPrecision, accuracy, robustness

This strategy ensures drug levels (originator/reference or biosimilar) are measured comparably in all clinical serum samples, supporting pharmacokinetic equivalence and regulatory requirements for biosimilar approval.

The primary in vivo models for administering research-grade anti-IL-4Rα antibodies to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models, often utilizing well-characterized murine tumor cell lines implanted into immunocompetent mice.

Context and Supporting Details:

  • Syngeneic Models:
    These are the most commonly used platforms because they permit the study of tumor-immune interactions within a fully functional mouse immune system. Frequently used syngeneic models for immunotherapy studies include RENCA (renal cell carcinoma), CT26 (colon carcinoma), MC38 (colon carcinoma), EMT-6 (breast cancer), Hepa1-6 (hepatoma), and B16F10 (melanoma). Their TIL composition and immune microenvironments have been extensively characterized, allowing detailed assessment of TIL phenotypes after therapeutic intervention.

  • Anti-IL-4Rα Antibody Use:
    Studies have used murine analogues of clinical anti-IL-4Rα antibodies (such as RegN1103, the mouse homologue of dupilumab) in allograft or genetically engineered mouse models. For instance, in an orthotopic allograft model of alveolar rhabdomyosarcoma (aRMS), immunodeficient mice were treated with an anti-IL-4Rα antibody to evaluate effects on tumor metastasis and immune parameters. While these allograft models use immunodeficient mice, the antibody is applied to manipulate the tumor microenvironment.

  • Assessment of TILs:
    Detailed immunophenotyping of TILs (such as CD8^+^ T cells, CD4^+^ T cells, Tregs, myeloid-derived cells, and macrophages) is routinely performed in these models using flow cytometry and immunohistochemistry after treatment.

  • Humanized Models:
    While not the most common system for anti-IL-4Rα antibody work, humanized mouse models (engrafted with human hematopoietic cells or tissues) can be used when antibodies require human IL-4Rα specificity. These are primarily for translational work or where the antibody does not cross-react with murine IL-4Rα. Syngeneic models remain preferred for mechanistic studies due to their robust immune contexture.

Summary Table:

Model TypeFeaturesExample TumorsNotes regarding anti-IL-4Rα studies
SyngeneicMurine tumors in immunocompetent miceB16F10, CT26, MC38Standard for TIL profiling after immunotherapy
AllograftTransplanted murine tumors (often in immunodeficient)aRMS orthotopicUsed for anti-IL-4Rα effect on metastasis
HumanizedHuman immune system components in immunodeficient micePatient-derived xenograftsRare for murine IL-4Rα antibodies

The prevailing approach for studying anti-IL-4Rα–mediated tumor inhibition and TIL characterization is the use of syngeneic mouse models with murine-specific, research-grade antibodies, sometimes augmented with genetically engineered or allograft systems for specific mechanistic questions.

Researchers have not documented the use of Dupilumab biosimilars in combination with immune checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to study synergistic anti-tumor effects in preclinical or translational immune-oncology models based on currently available published evidence. Current research focuses instead on dupilumab’s application for managing immune-related adverse events (irAEs), particularly cutaneous toxicities, associated with checkpoint inhibitor therapy rather than direct synergy in cancer killing.

Context and Supporting Details:

  • Dupilumab is generally used for atopic dermatitis and other type 2 inflammatory conditions; it is a monoclonal antibody against the IL-4 receptor alpha, impeding IL-4 and IL-13 signaling. It is explicitly not classified as immunosuppressive, which presents an advantage for treating dermatologic conditions in cancer patients.
  • Studies have shown its safety profile in patients with cancer or history of malignancy, but published clinical trials or preclinical models have not evaluated dupilumab or its biosimilars as synergistic agents to enhance the efficacy of checkpoint inhibitors.
  • Checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, anti-PD-1, anti-PD-L1) have been studied in various combinations (e.g., anti-PD-1 + anti-CTLA-4 or anti-PD-1 + anti-LAG-3) to improve anti-tumor immune responses, with mechanistic studies showing distinct effects on T cell subtypes. However, dupilumab is not typically included in such synergy studies targeting tumor immunity.

Current Combination Strategies in Immuno-Oncology:

  • Studies have elucidated that combining checkpoint inhibitors (such as anti-PD-1/CTLA-4 or anti-PD-1/LAG-3) activates different immune cell populations, offering insight into personalized or resistance-overcoming strategies in advanced melanoma.
  • The focus of these studies is on maximizing tumor-targeted immune activity, whereas dupilumab’s role is primarily as a supportive therapy to mitigate immune-related skin toxicity in patients undergoing checkpoint blockade.
  • Research documenting direct synergy in antitumor effect from combining dupilumab (or biosimilars) with immune checkpoint inhibitors is not currently available in the literature or preclinical reports.

Additional Relevant Information:

  • Alternative Mechanisms: The rationale for combining checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, etc.) lies in their non-redundant immune activation pathways.
  • Research Gaps: If synergy studies with dupilumab biosimilars and checkpoint inhibitors exist, they are either unpublished, in private industry pipelines, or ongoing and not yet reported.

In summary, dupilumab (and biosimilars) are investigated in oncology primarily for the management of immune-mediated side effects and not as direct partners in combination immune-oncology regimens for synergistic tumor targeting. Combination studies addressing synergy in anti-tumor effect currently focus on pairings among canonical checkpoint inhibitors, not type 2 cytokine antagonists like dupilumab.

A Dupilumab biosimilar can be used as the capture and/or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor patient immune responses against Dupilumab by exploiting the bivalent nature of anti-drug antibodies. In this assay, biosimilar Dupilumab (functionally equivalent to the original drug) is immobilized on the plate (capture reagent), and a labeled form of biosimilar Dupilumab is used for detection.

Essential context and supporting details:

  • Principle: In bridging ADA ELISA, endogenous anti-Dupilumab antibodies from patient serum act as a bridge between the immobilized biosimilar Dupilumab (capture) and the labeled biosimilar Dupilumab (detection). The presence of bound complex indicates the patient has developed antibodies against the therapeutic drug.
  • Why biosimilar: Using a biosimilar instead of the original biologic for reagents is appropriate as long as the biosimilar is analytically similar and retains the relevant epitopes. This allows interchangeable use in immunogenicity assays.
  • Detection strategy: The detection reagent can be conjugated with biotin (for streptavidin-based colorimetric readout) or an enzyme such as horseradish peroxidase (HRP). High-quality biosimilar reagents ensure sensitivity and specificity.
  • Assay workflow:
    • Plate is coated with biosimilar Dupilumab (capture).
    • Patient serum is added; any anti-Dupilumab antibodies present bind to the immobilized drug.
    • Labeled biosimilar Dupilumab is introduced; it binds to the other arm of the patient antibody.
    • The complex is detected via substrate conversion by the label, with signal strength proportional to ADA level.

Additional information:

  • The bridging format is highly sensitive and suitable for high-throughput screening, but may require careful optimization due to potential interference from serum components or soluble drug.
  • Both IgM and IgG ADAs can be detected with this assay design.
  • Biosimilar reagents must be validated as equivalent for immunoassay use to ensure reliable results.
  • This format provides the primary screening for anti-drug antibodies, after which confirmatory and titer assays may follow.
  • Regulatory guidance typically accepts biosimilar reagents if comparability to the reference is demonstrated.

In summary, a Dupilumab biosimilar—properly validated for equivalent epitope content—can substitute for the reference drug as capture and detection reagent in a bridging ADA ELISA, enabling sensitive, specific quantification of anti-Dupilumab immune responses in patient serum.

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.
B
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

- -
- -
Disclaimer AlertProducts are for research use only. Not for use in diagnostic or therapeutic procedures.