Anti-Human IL-6R (CD126) (Satralizumab)

Anti-Human IL-6R (CD126) (Satralizumab)

Product No.: I-1270

- -
- -
Product No.I-1270
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
Active
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 in PK bridging ELISAs as calibration standards (analytical standards) or reference controls to enable quantitative measurement and cross-comparison of drug concentrations in serum samples across reference and biosimilar products. Their use ensures assay consistency, minimizes analytical bias, and supports regulatory expectations for bioanalytical comparability in biosimilar development.

To elaborate:

  • Single Assay Calibration: The current industry consensus is to use a single validated PK assay with a single analytical standard—commonly the biosimilar—to generate a calibration (standard) curve for measuring concentrations in all study samples, regardless of whether the sample contains reference Satralizumab (originator) or its biosimilar. This minimizes analytical variability and eliminates the need for separate assays or crossover analyses, critical for blinded studies and regulatory alignment.
  • Selection & Evaluation of Standard: Before designating a biosimilar as the calibration standard, the assay is qualified and validated to ensure that both biosimilar and reference Satralizumab behave equivalently within the ELISA system. This involves spiking known concentrations of both products into human serum, constructing standard curves, and statistically confirming that recovery and detection are comparable (within pre-defined bioanalytical equivalence boundaries, e.g., confidence intervals fitting within [0.8, 1.25]).
  • Reference Controls: Parallel quality control (QC) samples prepared with both biosimilar and reference products are run alongside sample analyses to monitor assay performance and ensure ongoing bioanalytical comparability during routine testing.
  • Practical Implementation: Research-grade biosimilars are widely marketed for research use specifically as reference or calibration standards in ELISAs—enabling measurement of antibody drug levels in PK, biosimilarity, and QC studies. Their performance in these roles underpins biosimilarity claims and regulatory submissions.
  • Example—Satralizumab: The PK ELISA for Satralizumab was validated to ensure accuracy and precision, stability in serum, and reliable quantification across its concentration range in line with FDA guidance. This same framework is applied to biosimilar assays, with the biosimilar preparation used as the standard if comparability is proven.

In summary:

  • The research-grade Satralizumab biosimilar is used as the calibrating standard in PK ELISAs to build the standard curve.
  • Both reference and biosimilar samples are quantified against this curve.
  • Assay equivalency and validation are required to confirm that calibration with the biosimilar yields accurate, precise, and unbiased quantification for both biosimilar and reference samples.
  • These procedures align with regulatory expectations for bioanalytical methods in biosimilar development.

This approach ensures robust, comparable PK measurements that directly support biosimilarity assessments and meet regulatory scrutiny.

The primary in vivo models used to study the antitumor effects of anti-IL-6R alpha antibodies and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models using murine-specific antibodies, and humanized or xenograft models for testing antibodies targeting human IL-6R.

Syngeneic Mouse Models:

  • Description: Syngeneic models involve engrafting mouse tumor cell lines into immunocompetent mice of the same genetic background, enabling the assessment of immune responses and TIL characterization.
  • Application: Research-grade (not clinical-grade) anti-mouse IL-6Rα antibodies (such as clone 15A7) have been used in vivo to block IL-6R signaling in tumors like 4T1 mammary carcinoma in BALB/c mice, allowing for evaluation of tumor growth inhibition and immune infiltrate (including TILs).
  • Immune Context: These models are ideal for studying the impact of IL-6R blockade on the native murine immune system and provide detailed insights into changes in TIL populations, activation states, and related biomarkers.

Humanized or Xenograft Models:

  • Description: These models use immunodeficient mice implanted with human tumor cells (xenograft) or reconstituted with components of the human immune system (humanized).
  • Application: Anti-human IL-6Rα antibodies like tocilizumab have been administered in vivo in mice bearing human tumor xenografts to study tumor growth inhibition, though TIL analysis is more limited unless a humanized immune system is present.
  • Immune Context: Xenograft models with standard immunodeficient mice do not enable meaningful TIL characterization unless "humanized" with human immune cells; when this is done, the effects of anti-IL-6R antibodies on human TILs can be assessed.
Model TypeAntibody Target SpeciesImmune ContextExample TumorsTILs Characterized?
SyngeneicMouseMurine4T1, CT26, B16Yes (murine TILs)
XenograftHumanNone (immunodeficient)Human OS, HNSCCLimited (no TILs)
HumanizedHumanHuman reconstitutionHuman cancer linesYes (human TILs, less common)

Key Points:

  • Syngeneic models are the most widely used for studying tumor immune responses and IL-6R blockade due to a functional mouse immune system, enabling robust TIL characterization.
  • Humanized/xenograft models are crucial for assessing the efficacy of clinical-grade anti-human IL-6R antibodies but require specialized techniques for TIL studies.
  • When the goal is mechanistic understanding of TIL modulation by IL-6R inhibition, syngeneic murine models with anti-mouse IL-6R antibodies remain standard.

References from search results:

  • Use of anti-mouse IL-6Rα (clone 15A7) in 4T1 syngeneic tumor models.
  • Use of tocilizumab in human osteosarcoma xenograft models.
  • Mathematical modeling of IL-6 involvement in human tumor xenografts in immunodeficient mice (restricted TIL analysis).

Researchers study synergistic effects in immune-oncology models by combining agents targeting distinct immune pathways—such as Satralizumab biosimilars (anti-IL-6R) with checkpoint inhibitors like anti-CTLA-4 and anti-LAG-3 biosimilars—to assess enhancement of anti-tumor immunity beyond monotherapy.

Key context and supporting details:

  • Mechanistic Rationale: Combining different immune checkpoint inhibitors (ICIs) addresses non-overlapping immune suppression mechanisms. CTLA-4 and LAG-3 inhibit T-cell activation at different stages and locations, while IL-6R inhibition (Satralizumab) further modulates the immune microenvironment by blocking pro-inflammatory cytokine signaling. This multi-targeted approach is hypothesized to improve activation, proliferation, and persistence of anti-tumor T cells.

  • Experimental Design:

    • In complex immune-oncology models (often murine xenografts or syngeneic tumor models), researchers:
      • Administer Satralizumab biosimilars in combination with checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars).
      • Measure endpoints such as tumor growth inhibition, survival, immune cell infiltration, and cytokine profiles versus single-agent controls.
      • Analyze synergistic effects by assessing whether the combination yields superior anti-tumor responses than the additive effects of each agent alone.
    • These studies sometimes involve "triple combinations"—e.g., PD-1, CTLA-4, and LAG-3 blockade—given the unique but overlapping roles these targets play in suppressing the anti-cancer immune response.
  • Synergy Evidence:

    • Preclinical and clinical studies show that dual (or even triple) checkpoint blockade can synergistically augment T-cell activity, sustain tumor regression, and overcome resistance seen with monotherapy. The incorporation of IL-6 blockade (Satralizumab) is thought to further reduce tumor-promoting inflammation and regulatory mechanisms that compromise immunotherapy effectiveness, although most published clinical combinations to date focus on checkpoint inhibitors without IL-6R agents.
    • Studies cited have used tetravalent antibodies or combination regimens in animal models to demonstrate that dual blockade (e.g., CTLA-4 + LAG-3) dramatically reduced immune tolerance and enhanced anti-tumor responses.
  • Clinical Pipeline:

    • Clinical trials are ongoing to assess multi-checkpoint combinations, often with standard agents (e.g., ipilimumab [CTLA-4], relatlimab [LAG-3]) and sometimes exploring next-generation biosimilars. Inclusion of agents like Satralizumab biosimilars is still mainly in experimental or preclinical phases, with research-grade biosimilars available for laboratory studies.
    • Notably, most combination clinical trials in oncology have to date focused on anti-PD-1/LAG-3/CTLA-4 agents, with IL-6R combinations more commonly explored in preclinical settings.

Summary Table: Combination Strategy in Immune-Oncology Models

AgentRole/TargetCombination RationaleStudy Model
SatralizumabIL-6R blockadeModulates inflammatory TMEMouse, cell lines
Anti-CTLA-4T-cell checkpoint blockadeRestores T-cell activationMouse, human
Anti-LAG-3Inhibits exhausted T-cellsSynergizes with CTLA-4 blockadeMouse, human

Limitations and Future Directions:

  • Research is ongoing to clarify the full mechanistic interplay, optimize dosing/scheduling, and assess pharmacodynamics of these combinations.
  • Most direct evidence for Satralizumab biosimilar synergy is preclinical, with limited published clinical trial data.
  • Combinatorial immune-oncology remains an active, evolving research field, with new biosimilars advancing into laboratory and early clinical testing.

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 against Satralizumab by specifically binding antibodies that recognize epitopes on the therapeutic or its biosimilar. This format allows for the sensitive detection of ADAs directed against the drug, which are of clinical interest due to potential impact on efficacy or safety.

Context and Protocol:

  • In a bridging ELISA for ADA detection, the key principle is that ADA molecules can bind to two (or more) identical antigen molecules: one immobilized on the plate (capture), and another that is labeled for detection.
  • For Satralizumab (or its biosimilar), a typical protocol involves:
    • Coating the plate with Satralizumab biosimilar as the capture reagent.
    • Patient serum is added; any anti-Satralizumab ADAs present will bind to the immobilized biosimilar.
    • After washing, a biotinylated or enzyme-labeled Satralizumab biosimilar is added as the detection reagent—it binds the other arm of the ADA, forming a “bridge.”
    • A streptavidin-HRP (if using biotin label) or chromogenic substrate is used to develop a signal, which is proportional to the amount of ADA present.

Why Use a Biosimilar?

  • The biosimilar is generally epitope-matched to the originator (i.e., it has the same antibody-binding sites), so it binds the same ADAs as the originator would.
  • Using a biosimilar as both capture and detection allows monitoring the patient’s immune response regardless of slight differences, and may be more available or cost-effective compared to the reference product.

Assay considerations:

  • Sensitivity and specificity depend on the integrity and labeling method of the biosimilar, and the design must avoid non-specific signals (such as by pre-treating to dissociate immune complexes).
  • The bridging design favors the detection of bivalent ADAs that can crosslink two drug molecules, and can detect all isotypes/subtypes that can bridge (IgG, IgM, etc.).
  • The ELISA signal is interpreted as antibody positivity for anti-drug immunogenicity monitoring during or after treatment.

Additional notes:

  • This principle is standard for immunogenicity testing of monoclonal antibodies (including biosimilars).
  • The clinical significance of ADA detection should be interpreted with functional (e.g., neutralization) assays and in clinical context.
  • ADA incidence can be considerable for Satralizumab (reported as >50% in some studies).

Summary:Using a Satralizumab biosimilar as both capture and detection reagent in a bridging ADA ELISA enables the specific detection of patient-generated ADAs directed against the therapeutic, following a standardized, regulatory-accepted approach used for monoclonal antibody and biosimilar immunogenicity monitoring.

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.