Anti-Human IFNAR-1 (Anifrolumab) [Clone MEDI-546]

Anti-Human IFNAR-1 (Anifrolumab) [Clone MEDI-546]

Product No.: LT650

- -
- -
Product No.LT650
Clone
MEDI-546
Target
IFNAR1
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Anifrolumab, MEDI-546, IFNAR1, IFNAR, Interferon α/β Receptor 1, 1326232-46-5
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IHC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human IFNAR1
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8° C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
WP,
IP,
FA,
FC,
IHC
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 Anifrolumab. This product is for research use only. Anifrolumab activity is directed against Human IFNAR1.
Background
Type I interferon (IFN) receptor (IFNAR) plays a central role in anti-viral and anti-proliferative responses and its endocytic trafficking is tightly associated with control of JAK/STAT signaling1. IFNAR is composed of two subunits, IFNAR1 and IFNAR2, that are ubiquitously expressed at variable levels depending on the cell type. IFNAR1 plays a role in the pathogenesis of complex multisystem autoimmune diseases such as systemic lupus erythematosus (SLE)2 and systemic sclerosis3. Approximately 60-80% of adult patients with active SLE express elevated levels of type I IFN inducible genes in tissues and blood 4, known as an ‘IFN signature'2.

Anifrolumab is an IFNAR1-specific antagonist produced in mouse myeloma cells (NS0)4, 5 that prevents IFN from binding to IFNAR12 and suppresses the receptor-mediated biological activity of all type I IFNs3, including those implicated in SLE pathogenesis (IFN-α, IFN-β and IFN-ω)5. Anifrolumab binding leads to inhibition of downstream signaling activities4, 6, including IFN responsive gene expression2. Anifrolumab also normalizes the IFN gene signature in patients with systemic sclerosis6.

Anifrolumab clone AL 5, a non-therapeutic biosimilar antibody for research use only was developed recombinantly and has the same variable regions as the original therapeutic which binds to IFNAR1 with high specificity and affinity, sterically inhibiting the binding of IFN ligands7 and preventing the formation of the IFN/IFNAR1/IFNAR2 ternary signaling complex by blocking heterodimerization2, 7. Additionally, anifrolumab induces internalization of IFNAR1, reducing the levels of cell surface IFNAR1 available for complex assembly2, 4. Anifrolumab recognizes the SD3 subdomain of IFNAR1 with the critical residue R279 providing a dominant contribution7.

Anifrolumab is an Fc-modified version of the anti-IFNAR 9D4 antibody8. Anifrolumab’s constant domain contains the triple mutations L234F/L235E/P331S for reduced antibody Fc-mediated effector functions7 and causes decreased binding to human FcyRI (CD64), FcyRIIA (CD32A), FcyRIII (CD16), and Clq8.
Antigen Distribution
IFNAR1 is a plasma membrane protein widely expressed on most nucleated cells that undergoes endocytosis when activated.
Ligand/Receptor
IFNAR1
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Signal Transduction

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 Anifrolumab biosimilars can be utilized as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to measure drug concentration in serum samples by providing a reliable and consistent reference point. Here's how they are used:

Role of Anifrolumab Biosimilars in PK Bridging ELISAs

  1. Calibration Standards: Anifrolumab biosimilars, being fully human monoclonal antibodies, can serve as calibration standards by providing known concentrations of the drug. This allows the creation of a standard curve that can be used to quantify the concentration of Anifrolumab in serum samples based on the ELISA assay's absorbance readings.

  2. Reference Controls: These biosimilars can act as positive controls to ensure the accuracy and reliability of the assay. By using known concentrations of the biosimilar, researchers can validate the performance of the ELISA kit and ensure it is functioning correctly to measure drug concentrations.

  3. QC and Validation: The use of Anifrolumab biosimilars in quality control (QC) and validation processes ensures that the ELISA assay is properly calibrated and that the results are consistent across different batches and runs. This is crucial for maintaining the integrity of PK studies.

  4. Bridging Studies: In bridging studies, where the goal is to establish pharmacokinetic equivalence between original and biosimilar products, Anifrolumab biosimilars can be used to bridge the data by providing a common reference point. This helps in comparing the pharmacokinetic profiles (e.g., absorption, distribution, metabolism, and excretion) of the original drug and its biosimilar.

Key Considerations

  • Characterization: The molecular structure and post-translational modifications of Anifrolumab biosimilars should be well-characterized to ensure they mimic the original drug closely.
  • Immunogenicity: Assessment of immunogenicity, which is the ability of the drug to induce an immune response, is important. Biosimilars should have similar immunogenicity profiles to the reference product.
  • Regulatory Compliance: The use of biosimilars in PK studies must comply with regulatory guidelines, such as those set by the FDA or EMA, which often require rigorous analytical and clinical evaluations.

By using Anifrolumab biosimilars as calibration standards and reference controls, researchers can ensure the accuracy and reliability of PK bridging ELISAs, facilitating the development and validation of new therapeutic antibodies.

Overview of Syngeneic and Humanized Mouse Models in IFNAR1 Antibody Research

Syngeneic models are the primary in vivo systems used to study the effects of anti-IFNAR1 (interferon alpha/beta receptor 1) antibodies on tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization. These models involve implanting murine tumor cell lines into immunocompetent mice of the same genetic background, allowing researchers to evaluate how blockade of the Type I interferon (IFN-I) pathway—using anti-IFNAR1 antibodies—affects tumor progression, immune cell infiltration, and therapeutic response.

Humanized mouse models (where human immune cells or tissues are engrafted into immunodeficient mice) are less commonly used for this specific antibody class, primarily because the anti-IFNAR1 antibodies must target the murine IFNAR1 receptor to be effective in syngeneic systems. For humanized models, the antibody would need to target human IFNAR1, and the model would require engrafted human tumors alongside a functional human immune system—a technically complex and less standardized approach in this context.

Key Syngeneic Tumor Models

Several well-characterized syngeneic tumor models are employed to assess immunotherapy, including:

  • RENCA (Renal Cell Carcinoma): Highly immune-infiltrated, with abundant TILs that diminish as tumors progress.
  • CT26 (Colon Carcinoma): Moderately immune-infiltrated, with increasing CD8⁺ T cells as tumors grow, though these cells may be functionally restrained.
  • B16F10 (Melanoma): Poorly immune-infiltrated, often used to model "cold" tumors that are less responsive to immunotherapy.
  • Other models (e.g., MC38, 4T1) are also used, each with distinct immune profiles and responses to therapy.

These models are selected based on their differing baseline immune infiltrates, allowing researchers to dissect how IFNAR1 blockade modulates TIL composition and function across a spectrum of tumor immunogenicity.

Experimental Design and TIL Analysis

When a research-grade anti-IFNAR1 antibody is administered in vivo:

  • Tumor Growth Inhibition: The impact on tumor growth is monitored to determine if IFN-I signaling blockade promotes or inhibits tumor progression. The outcome often depends on the baseline immunogenicity of the tumor model and the timing of antibody administration relative to tumor establishment.
  • TIL Characterization: Flow cytometry, immunohistochemistry, and single-cell RNA sequencing are used to profile TILs (e.g., CD4⁺, CD8⁺ T cells, NK cells, myeloid-derived suppressor cells). Changes in the frequency, activation state, and functional status of these populations reveal how IFNAR1 blockade reshapes the tumor immune microenvironment.
  • Mechanistic Insights: Studies often correlate TIL dynamics with tumor outcomes, providing insights into how IFN-I signaling influences antitumor immunity, antigen presentation, and immune suppression.

Humanized Models: Current Limitations

While syngeneic models are standard, humanized models are theoretically possible but face significant hurdles:

  • Antibody Specificity: Most research-grade anti-IFNAR1 antibodies target the murine receptor. For humanized models, a human-specific anti-IFNAR1 antibody would be required.
  • Model Complexity: Humanized models require engraftment of both human tumors and immune components, making them more variable and less routinely used for this research question.
  • Translational Relevance: Syngeneic models are preferred for preclinical immunotherapy studies because they maintain a fully intact, functional immune system, enabling direct assessment of how immune modulation affects tumor control.

Summary Table: Model Comparison

Model TypeAnti-IFNAR1 TargetTIL ProfilingTumor Growth InhibitionCommon Use Case
SyngeneicMurine IFNAR1YesYesStandard for IFN-I pathway modulation
HumanizedHuman IFNAR1PossiblePossibleRare, due to technical and specificity challenges

Conclusion

Syngeneic mouse tumor models are the primary in vivo systems where research-grade anti-IFNAR1 antibodies are administered to study tumor growth inhibition and TIL characterization, owing to their immune competence, well-defined immune profiles, and the availability of murine-specific antibodies. Humanized models remain a niche option due to challenges in antibody specificity and model standardization. The choice of syngeneic model (e.g., RENCA, CT26, B16F10) depends on the desired tumor immune context and research question.

Researchers utilize the Anifrolumab biosimilar—which blocks type I interferon receptor signaling—together with other checkpoint inhibitor biosimilars (such as anti-CTLA-4 or anti-LAG-3) in preclinical immune-oncology models to investigate potential synergistic effects on tumor immunity and immune regulation.

Key context and supporting details:

  • Mechanistic Rationale: Anifrolumab biosimilars specifically inhibit IFNAR1-mediated type I interferon (IFN) signaling, impacting monocyte maturation, NETosis, and inflammatory cytokine production, all of which shape immune responses in the tumor microenvironment. Checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 act by blocking inhibitory pathways in T cells, thus enhancing their anti-tumor activity through distinct immune compartments—CTLA-4 mainly in lymph nodes and LAG-3/PD-1/PD-L1 in peripheral and tumor sites.

  • Complex Model Design:

    • Researchers combine these agents in in vitro (e.g., co-culture of immune cells and tumor cells) and in vivo (e.g., murine tumor xenografts) systems.
    • By using variant isotypes of Anifrolumab biosimilars—with different Fc effector functions—they can parse out the effects of receptor blockade versus antibody-mediated cytotoxicity.
  • Synergy Assessment:

    • Multiple checkpoint pathway inhibition (such as combining anti-CTLA-4 with Anifrolumab biosimilars) allows researchers to observe enhanced T cell activation, interferon-blockade-mediated reduction of immunosuppressive signals, and improved overall anti-tumor immune responses when compared to monotherapies.
    • When combined with biosimilars targeting LAG-3 (or TIM-3), the models test how IFN pathway inhibition may modulate exhaustion marker expression or synergize with further checkpoint blockade.
  • Experimental Readouts:

    • Tumor growth inhibition
    • Immune cell infiltration and activation markers
    • Cytokine profiles
    • Gene expression analyses (e.g., IFN signatures, checkpoint markers)
  • Utility of Biosimilars: Using biosimilars rather than clinical-grade mAbs enables mechanistic studies (e.g., isotype comparison, mutational analysis) to optimize therapeutic combinations and distinguish Fc-mediated effects from receptor blockade.

Additional notes:

  • While direct clinical evidence for the Anifrolumab biosimilar’s synergy with checkpoint inhibitors in oncology is still emerging, these combinatorial strategies are heavily explored in preclinical and translational research aiming to overcome monotherapy limitations and immune resistance.
  • Toxicity profiles and immune activation are closely monitored, as combinatorial regimens often increase risk for higher-grade immune-related adverse events.

In summary, researchers leverage these biosimilars in combination studies to dissect and optimize immune activation, balancing the blockade of immunosuppressive pathways (type I IFN, CTLA-4, LAG-3) for more robust anti-tumor responses in complex models.

An Anifrolumab biosimilar can be used as either a capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response (immunogenicity) against therapeutic Anifrolumab. In this assay, the biosimilar is functionally equivalent to the original drug and exploits the bivalent nature of ADAs, which can bind two identical Anifrolumab molecules—one for capture, one for detection.

Context and Methodology:

  • Bridging ADA ELISA Principle: This method detects ADAs using the therapeutic antibody (here, Anifrolumab or its biosimilar) labeled in two forms: one immobilized (capture) and one labeled (detection, e.g., HRP or biotin). Anti-drug antibodies from a patient's serum “bridge” these two molecules, forming a detectable complex.

    • The capture reagent (often biotinylated biosimilar) is coated onto a streptavidin plate.

    • Patient serum is added; any ADA present will bind to the capture agent.

    • The detection reagent (usually enzyme- or dye-labeled biosimilar) is then added; if ADA is present, it will bind both arms, generating a signal.

  • Why Use a Biosimilar?

    • A biosimilar with matched variable domains ensures correct native antigen recognition, maximizing sensitivity and specificity to the anti-Anifrolumab ADA responses.
    • It is important that the Fc functionality of the biosimilar does not interfere with ADA binding, which is ensured by using versions with appropriate Fc modifications or isotypes.
  • Advantages:

    • This bridging format is highly sensitive and allows for detection of bivalent ADAs in patient samples, regardless of isotype.
    • It is customizable for high-throughput screening and compatibility with complex matrices like serum.
  • Limitations:

    • Sensitivity can be affected by free or circulating drug, soluble antigen, or matrix interference, and may require optimization of blocking reagents and controls.
    • Only detects bridging (bivalent) antibodies, so some isotypes or monovalent fragments may not be efficiently detected.

Summary Table:

Role in bridging ELISAHow Anifrolumab biosimilar is used
CaptureCoated on plate (often biotinylated) to bind ADA from patient sample
DetectionLabeled with enzyme/dye, binds other ADA arm captured on plate

Key consideration:

  • Both capture and detection reagents must use the same (matched) Anifrolumab biosimilar for specificity to anti-Anifrolumab ADAs, and they must be functionally labeled or immobilized in ways that do not hinder antibody recognition.

This approach is well-established for other biologics and was used in clinical trials to assess the immunogenicity of Anifrolumab, showing a low incidence of ADA development.

References & Citations

1. Zanin N, Viaris de Lesegno C, Lamaze C, et al. Front Immunol. 11:615603. 2021.
2. Deeks ED. Drugs. 81(15):1795-1802. 2021.
3. Goldberg A, Geppert T, Schiopu E, et al. Arthritis Res Ther. 16(1):R57. 2014.
4. https://www.accessdata.fda.gov/drugsatfda_docs/label/2021/761123s000lbl.pdf
5. Kaplon H, Chenoweth A, Crescioli S, et al. MAbs. Jan-Dec;14(1):2014296. 2022.
6. Wang B, Higgs BW, Chang L, et al. Clin Pharmacol Ther. 93(6):483-492. 2013.
7. Peng L, Oganesyan V, Wu H, et al. MAbs. 7(2):428-439. 2015.
8. https://patents.google.com/patent/WO2017031288A1/en
9. Guo X, Higgs BW, Bay-Jensen AC, et al. J Invest Dermatol. 135(10):2402-2409. 2015.
Indirect Elisa Protocol
FA
Flow Cytometry
IHC
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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