Anti-Human IL-6R (CD126) (Satralizumab) – Fc Muted™

Anti-Human IL-6R (CD126) (Satralizumab) – Fc Muted™

Product No.: I-1275

- -
- -
Product No.I-1275
Clone
SA237
Target
IL-6R alpha
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-6R-alpha, IL-6RA, IL-6R 1, gp80, CD126
Isotype
Human IgG2κ
Applications
B
,
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Cynomolgus Monkey
Human
Host Species
Hamster
Expression Host
CHO 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 Satralizumab. SA237 (Satralizumab) is an antagonist of human and cynomolgus monkey IL-6R.
Background
IL-6 is a pleiotropic cytokine that promotes B cell and T cell proliferation and differentiation and is also involved in the inflammatory response1. In the ‘classic’ signaling paradigm, IL-6 binds to its membrane bound receptor IL-6R to initiate intracellular signaling pathways2. Alternatively, in ‘trans-signaling’, IL-6 binds to a soluble form of IL-6R. In both events, a complex set of interactions with membrane-bound or soluble β-receptor glycoprotein 130 (gp130) modulates the downstream signaling pathways. Additionally, IL-6 plays an inflammatory role in autoimmune diseases3 and high IL-6 levels are a feature of cytokine storm and cytokine release syndrome during COVID-19 infection1. IL-6 signaling can be inhibited by antibodies directed against IL- 6R3.

SA237 (Satralizumab) is a humanized monoclonal recycling antibody that was developed for the treatment of neuromyelitis optica spectrum disorder, a rare autoimmune disease of the central nervous system4. Satralizumab binds to both membrane and soluble IL-6R, thereby inhibiting IL-6 signaling as well as reducing inflammation and IL-6 mediated autoimmune T cell and B cell activation. Ultimately, B cell differentiation into AQP4-IgG-secreting plasmablasts is prevented. Satralizumab circulation in the body is extended via a novel recycling technology that allows for pH dependent dissociation from IL-6R in the endosome after cellular uptake. Satralizumab is then transported back to the plasma membrane via the recycling endosomal pathway and released into plasma for reuse.

Satralizumab is produced in Chinese hamster ovary cells using recombinant DNA technology4. Satralizumab does not cross react with rodent IL-6R5.

Antigen Distribution
IL-6R is mainly found on hepatocytes, some leukocytes, and epithelial cells. IL-6R also has a soluble form.
Ligand/Receptor
IL-6
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Inflammatory Disease
.
Innate Immunity
.
Neuroscience
.
Signal Transduction
.
Autoimmune Disease
.
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 Satralizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA to accurately measure drug concentration in serum samples by serving as the quantitative reference material against which both biosimilar and reference Satralizumab are compared.

Essential context:

  • In a PK bridging ELISA, it is best practice to establish a single PK assay using a single analytical standard (often the biosimilar), which acts as the calibrator for quantifying both biosimilar and reference products in serum samples.
  • For Satralizumab, recombinant or high-purity biosimilar is prepared at various known concentrations to generate a standard curve. Serum samples containing unknown Satralizumab concentrations are assayed, and their concentrations are interpolated from the standard curve.
  • Calibration standards are typically prepared in human serum matrix at multiple concentrations (e.g., 50–12,800 ng/mL) to reflect the expected range in clinical samples and are analyzed in parallel with quality control (QC) samples with known concentrations of both biosimilar and reference drugs.
  • Analytical equivalence between the biosimilar standard and the reference standard is rigorously tested during method qualification and validation. Statistical analysis (e.g., 90% confidence interval within [0.8, 1.25] range) ensures that measurement bias is minimized and both products are assessed comparably.
  • The method is fully validated according to regulatory guidelines (such as FDA), assessing parameters including accuracy, precision, stability, and sensitivity in the same serum matrix where clinical samples will be measured.

Additional details:

  • The selected biosimilar standard must demonstrate bioanalytical comparability (essentially, the antibody behaves identically in the assay to the reference drug), justifying its use as a calibrator for all PK bridging analysis.
  • Reference controls (samples of known concentration) are included to monitor assay performance and batch-to-batch consistency, further ensuring reliable quantification for PK studies.
  • The competitive ELISA setup (pre-coated target, competition with biotin-labeled antibody, colorimetric readout) is one format suitable for Satralizumab measurement, offering quantitative data in concentration ranges appropriate for clinical pharmacokinetic analysis.
  • The validated assay enables direct comparison of serum Satralizumab levels from different populations or products, supporting essential regulatory PK bioequivalence studies for biosimilar development.

In summary, research-grade Satralizumab biosimilars are quantitatively prepared as calibration standards to generate standard curves, qualify assay equivalence, and serve as reference controls in validated PK ELISA assays to ensure consistent and accurate measurement of drug in serum samples.

The primary models for in vivo administration of research-grade anti-IL-6R alpha antibodies to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are murine syngeneic tumor models.

Key details:

  • Syngeneic models involve the implantation of mouse-derived tumor cell lines into immunocompetent mice of the same genetic background, allowing for intact native murine immune responses, including the recruitment and analysis of TILs.
  • Commonly used syngeneic models in this context include:
    • MC38 (colon carcinoma, C57BL/6 background)
    • CT26 (colon carcinoma, BALB/c background)
    • EMT-6 (mammary carcinoma, BALB/c background)
    • Hepa1-6 (hepatocellular carcinoma, C57BL/6 background)
  • Anti-mouse IL-6R alpha antibodies (e.g., clone 15A7, commercially available) are used to directly target mouse IL-6R in these models, enabling evaluation of both anti-tumor efficacy and changes in the tumor immune microenvironment, including TIL populations and phenotypes.

Why syngeneic models are favored for these studies:

  • Tumor and host are fully mouse origin, so both the antibody and resulting immune responses (including TILs) are physiologically relevant to the mouse system.
  • These models allow for in-depth analysis of immune cell subsets (CD4+ T cells, CD8+ T cells, Tregs, macrophages, NK cells) via flow cytometry, immunohistochemistry, or single-cell technologies.

Humanized models:

  • Humanized mouse models, in which human immune cells or tissues are engrafted into immunodeficient mice, primarily allow the use of anti-human IL-6R antibodies (e.g., tocilizumab) and study of human TILs.
  • These models are more complex and expensive, typically reserved for situations where cross-reactivity to human IL-6R is required or when studying human-specific responses—less common for initial anti-IL-6R screening due to technical limitations and cost.

Supporting details:

  • Syngeneic models are well established for immune-oncology, including characterization of TILs and immunotherapy combinations.
  • Multiple commercial anti-mouse IL-6R antibodies are available for preclinical in vivo applications.

In summary:
The standard approach to studying tumor growth inhibition and TIL composition upon in vivo anti-IL-6R alpha antibody administration is to use murine syngeneic tumor models (e.g., MC38, CT26, Hepa1-6) with research-grade antibodies specific to mouse IL-6R. Humanized models are possible but far less frequently used for this specific purpose, particularly at the research-grade antibody stage.

Researchers studying Satralizumab biosimilars in combination with other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars primarily aim to evaluate synergistic immunomodulatory effects in complex immune-oncology models, particularly within experimental and preclinical settings.

Satralizumab is a humanized monoclonal antibody targeting the interleukin-6 receptor (IL-6R), inhibiting downstream IL-6-mediated signaling, a pathway implicated in immune regulation and cancer immunity. While most clinical data for satralizumab centers on autoimmune disorders (notably neuromyelitis optica spectrum disorder), its biosimilar form is available as a research reagent intended for in vitro and in vivo immune-oncology studies.

Research Approach for Combination Strategies

To study potential synergy, researchers use the following general strategies:

  • In vitro models: Tumor cell lines and primary immune cells are co-cultured and exposed to Satralizumab biosimilar along with checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3). The readouts include T cell activation, cytokine production, cytotoxicity assays, and immune cell phenotyping.
  • In vivo models: Humanized mouse models or syngeneic tumor models are treated with these combinations to observe effects such as tumor growth inhibition, immune cell infiltration, and survival benefit.
  • Mechanistic exploration: Blocking multiple immune pathways (e.g., IL-6R, CTLA-4, LAG-3) allows investigators to dissect how dual or triple checkpoint blockade can overcome compensatory immune resistance in the tumor microenvironment.

Rationale for Synergy

  • Checkpoint inhibitors disrupt immune-inhibitory pathways, restoring T cell activity. For example, anti-CTLA-4 acts in lymph nodes to expand activated T cells, while anti-LAG-3 (often combined with anti-PD-1) can further unleash T cell responses at the tumor site.
  • IL-6 blockade (Satralizumab biosimilar): IL-6 is known to promote chronic inflammation and immunosuppression in the tumor microenvironment, hindering anti-tumor immunity and favoring tumor growth. Blocking IL-6R can potentially reduce immune suppression, enabling checkpoint inhibitors to be more effective.
  • Combinatorial effect: Simultaneous inhibition of IL-6 signaling and checkpoint pathways may both activate effector immune cells and diminish suppressive cytokine signaling, thus yielding greater anti-tumor activity than monotherapy.

Current Evidence and Limitations

  • Animal and early-stage studies suggest that combining multiple checkpoint inhibitors (such as anti-CTLA-4 plus anti-PD-1, or anti-LAG-3 combinations) can synergistically enhance anti-tumor responses—but also increase the risk of immune-related adverse events.
  • The safety and mechanistic profile of combining IL-6R inhibitors like Satralizumab with checkpoint inhibitors in cancer is less well characterized than established combinations (such as anti-PD-1 plus anti-CTLA-4).
  • Clinical application and mechanistic details of Satralizumab with checkpoint inhibitors remain an area of active research; most evidence is preclinical.

Summary Table: Mechanistic Rationale

AgentPrimary TargetSite of ActionExpected Effect in Combination
Satralizumab biosimilarIL-6RSystemic & tumor microenvironmentReduces pro-tumor inflammation and immunosuppression
Anti-CTLA-4CTLA-4Lymph nodesExpands activated T cells
Anti-LAG-3LAG-3Tumor siteReleases T cell inhibition (especially with PD-1 co-blockade)

Key point: Researchers leverage Satralizumab biosimilar and other checkpoint inhibitor biosimilars in combination to dissect and potentially enhance anti-tumor immunity, focusing on both synergistic activation of effector cells and reduction of tumor-driven immune suppression, though such strategies are mostly explored in preclinical or early translational studies.

A Satralizumab biosimilar 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 by exploiting the ability of bivalent or multivalent ADAs to bridge two identical drug molecules—one immobilized on the plate (capture), and one labeled for detection.

Detailed context and supporting details:

  • In a bridging ELISA for ADA detection:

    • Microtiter plates are coated with the therapeutic antibody or its biosimilar (here, Satralizumab biosimilar) to serve as the capture reagent.
    • Patient serum is added; if ADAs specific to Satralizumab are present, they bind to the immobilized drug.
    • A labeled form (e.g., HRP-conjugated) of the same biosimilar is then added as the detection reagent, binding to another site on the captured ADA and creating a "bridge".
    • Detection is achieved through substrate conversion and signal measurement; the presence and level of signal is proportional to ADA in the sample.
  • The use of a biosimilar (rather than the original biologic drug) is appropriate if:

    • The biosimilar is structurally and immunologically equivalent to the reference product. It ensures that any ADA response detected is truly directed against the active moiety and not a product-specific impurity or modification.
    • The biosimilar's use is fully validated in the context of the assay (to demonstrate equivalent binding and performance).
  • Why a biosimilar is suitable:

    • Regulatory agencies accept the use of biosimilars in immunogenicity assays once they are proven equivalent in structure, post-translational modifications, and binding properties.
    • Using a biosimilar can reduce cost and availability issues, especially for large-scale or long-term monitoring.
  • Typical workflow in ADA detection:

    • Coat plate with Satralizumab biosimilar.
    • Add patient serum (potentially containing anti-Satralizumab ADAs).
    • Add labeled Satralizumab biosimilar.
    • Add substrate and measure signal; positive signal indicates ADA presence bridging the two biosimilar molecules.
  • Alternative formats:

    • The biosimilar can be biotinylated or enzymatically labeled for detection, or used in different formats depending on assay design.

This approach is the standard for biotherapeutics such as monoclonal antibodies, including Satralizumab, and is supported in the analytical literature for ADA assay development. It’s essential that assay validation demonstrates the equivalence of the biosimilar to the reference molecule in its ability to detect clinically relevant ADAs.

References & Citations

1 Cortegiani A, Ippolito M, Greco M, et al. Pulmonology. 27(1):52-66. 2021.
2 Wolf J, Rose-John S, Garbers C. Cytokine. 70(1):11-20. 2014.
3 Sebba A. Am J Health Syst Pharm. 65(15):1413-1418. 2008.
4 Heo YA. Drugs. 80(14):1477-1482. 2020.
5 Katagiri R, Ishihara-Hattori K, Frings W, et al. Birth Defects Res. 109(11):843-856. 2017.
6 Yamamura T, Kleiter I, Fujihara K, et al. N Engl J Med. 381(22):2114-2124. 2019.
7 Traboulsee A, Greenberg BM, Bennett JL, et al. Lancet Neurol. 19(5):402-412. 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.