Anti-Human IL-6R (Tocilizumab) [Clone RG-1569]

Anti-Human IL-6R (Tocilizumab) [Clone RG-1569]

Product No.: I-1240

- -
- -
Product No.I-1240
Clone
RG-1569
Target
IL-6R
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL6R, CD126, IL6RA
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Humanized antibody derived from mouse clone PM-1
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 ?
ELISA,
FA,
WB
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 Tocilizumab. This product is for research use only. Tocilizumab activity is directed against 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 response 1 . In the ‘classic’ signaling paradigm, IL-6 binds to its membrane bound receptor IL-6R to initiate intracellular signaling pathways 2. 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 diseases 3 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-6R 3.

Tocilizumab is the first IL-6R inhibitor3 . It competitively inhibits the binding of IL-6 to both soluble and membrane bound IL-6R 3, 4, and thereby prevents IL-6 signal transduction to inflammatory mediators that summon B and T cells 3.

Tocilizumab is a chimeric murine-human antibody engineered by CDR grafting of the antigen binding regions of murine antihuman IL-6R antibody PM-1 to the human IgG1 framework 3, 5. Tocilizumab binds to the IL-6 binding site and neutralizes IL-6 activity 5, 6, 7. Tocilizumab has been studied for the treatment of many autoimmune diseases including rheumatoid arthritis, systemic juvenile idiopathic arthritis, Castleman disease, and Crohn’s disease 7 as well as for the treatment of severe COVID-19 1 .
Antigen Distribution
IL-6R is expressed on hepatocytes and certain subpopulations of leukocytes.
Ligand/Receptor
IL-6 & ciliary neurotrophic factor
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology

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 tocilizumab biosimilars serve as critical calibration standards and reference controls in pharmacokinetic bridging ELISA assays through several key mechanisms that ensure accurate quantification of drug concentrations in serum samples.

Calibration Standard Applications

Research-grade tocilizumab biosimilars are utilized to create concentration curves that span the expected range of drug levels in patient serum samples. These biosimilars undergo rigorous characterization to demonstrate structural and functional similarity to the reference tocilizumab, making them suitable as analytical standards. The concentration of tocilizumab in serum is determined using a sandwich ELISA format, where the biosimilar standards provide the quantitative backbone for measurement.

In pharmacokinetic studies, these calibration standards are essential for establishing bioequivalence parameters. Studies have demonstrated that biosimilars like CT-P47 and MSB11456 show pharmacokinetic equivalence with reference tocilizumab, with 90% confidence intervals for geometric least-squares mean ratios falling within the 80-125% equivalence margin. This equivalence validation makes research-grade biosimilars reliable calibration references.

Reference Control Functions

Quality control applications represent a major use of tocilizumab biosimilars in PK bridging assays. These biosimilars serve as reference controls during the development and manufacturing phases, helping characterize biosimilar products in terms of their structural and functional similarities to reference monoclonal antibodies. They enable assessment of critical parameters including purity, potency, and stability throughout the analytical process.

Biosimilarity assessment is facilitated through these reference controls by comparing binding kinetics, affinity, and other relevant properties between the biosimilar and reference product. This comparative analysis is essential for regulatory submissions, as biosimilar ELISA kits generate the quantitative and qualitative data required to demonstrate biosimilarity to regulatory authorities.

PK Bridging Methodology

The sandwich ELISA format typically employed uses anti-tocilizumab antibodies as capture reagents, with the research-grade biosimilar standards providing concentration references across the analytical range. Blood samples for pharmacokinetic analysis are collected at multiple time points, and the biosimilar standards enable accurate quantification of drug levels throughout the elimination phase.

Immunogenicity monitoring is another critical application where biosimilar standards help assess anti-drug antibodies (ADAs) and neutralizing antibodies that may develop during treatment. Studies have shown comparable ADA positivity rates between biosimilars and reference products, with research-grade standards enabling consistent measurement of these immune responses.

The use of research-grade tocilizumab biosimilars as calibration standards and reference controls ensures that PK bridging ELISA assays maintain the analytical rigor necessary for regulatory compliance while providing cost-effective alternatives to reference products for routine analytical applications.

The research literature reveals several primary models where anti-IL-6R antibodies have been administered in vivo to study tumor growth inhibition, though specific TIL characterization data is more limited in the available studies.

SCID Mouse-Human Myeloma Model

The most well-documented model involves SCID mice inoculated with human myeloma cells. In this system, researchers used the S6B45 myeloma cell line, where human IL-6 functions as an autocrine growth factor. The anti-human IL-6R antibody PM1 was administered intraperitoneally (100 micrograms every 48 hours for 10 doses), demonstrating strong tumor growth inhibition when treatment began 24 hours after tumor inoculation. This model also showed comparable results with anti-human IL-6 antibody MH166, providing validation for IL-6 pathway targeting.

The effectiveness of this model was timing-dependent, with significant inhibitory effects observed only when treatment started early (within 24 hours), while administration beginning 5 or more days post-inoculation showed no significant effect. This humanized model system is particularly valuable because murine IL-6 does not bind to human IL-6R, ensuring that only human tumor cell-derived IL-6 contributes to the signaling pathway.

Head and Neck Squamous Cell Carcinoma (HNSCC) Model

Another important model involves primary human cancer stem cells transplanted into IL-6+/+ immunodeficient mice. This system uses ALDH^HIGH^CD44^HIGH^ cancer stem cells from head and neck squamous cell carcinoma. Mathematical modeling of this system revealed that tumor cell-secreted IL-6 significantly influences tumor growth dynamics, with tumors unable to produce IL-6 showing approximately 45% smaller final sizes.

The model demonstrates that even low IL-6 secretion rates (ρ=7×10^-7 fMol/cell×day) substantially influence tumor growth, and a fractional occupancy of just 12% of IL-6R on cancer stem cells is sufficient to drive the experimentally observed tumor growth rates.

Syngeneic Models for Immunotherapy Studies

While specific anti-IL-6R studies in syngeneic models are less documented in the provided literature, several well-characterized syngeneic tumor models are available for immunotherapy research that could be adapted for IL-6R targeting studies. These include:

EMT6, CT26, and RENCA models, which are described as immune-infiltrated with unique tumor microenvironment features. These models have been characterized for baseline tumor-infiltrating lymphocyte populations and responses to immune checkpoint inhibitors. The RENCA model, in particular, shows strong immune infiltration and responsiveness to immunotherapies like anti-OX40 treatment.

MC38 and TC-1 models are also mentioned as established syngeneic systems, with MC38 being commonly used and TC-1 noted as more challenging to develop but successfully implemented.

Limitations in TIL Characterization

While these models provide robust platforms for studying tumor growth inhibition with anti-IL-6R antibodies, specific characterization of tumor-infiltrating lymphocytes following IL-6R blockade is not extensively detailed in the available literature. The syngeneic models are described as being "fully characterized for gene expression, TIL baseline populations, and response to common immune checkpoint inhibitors", but the specific effects of IL-6R targeting on TIL composition and function would require dedicated studies in these systems.

The mathematical modeling work suggests that IL-6 signaling significantly affects cancer stem cell self-renewal and survival, which could have downstream effects on immune cell infiltration and activation, but direct TIL analysis following anti-IL-6R treatment remains an area requiring further investigation across these model systems.

Researchers investigate synergistic effects of Tocilizumab biosimilars with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 by combining these agents in complex immune-oncology models to study both enhanced anti-tumor responses and the management of immune-related adverse events (irAEs).

Preclinical models—often murine systems of cancer (e.g., lung cancer) and immune toxicity (e.g., PD-1 inhibitor-associated myocarditis)—are used to assess:

  • Tumor growth and regression when checkpoint inhibitors are administered with Tocilizumab biosimilars, evaluating synergy in anti-tumor activity.
  • Immunopathology, particularly the mitigation of toxicity (e.g., myocarditis) induced by checkpoint blockade, with Tocilizumab exerting its effect by inhibiting the IL-6/JAK2/STAT3 pathway in activated immune cells.
  • Mechanistic studies measuring cytokine levels (e.g., IL-6), immune cell profiling (such as macrophage polarization), and downstream signaling in tissue samples to elucidate how the biosimilar modulates immune activation or durability.

Research combining checkpoint inhibitors with agents like Tocilizumab biosimilars typically addresses:

  • Improved efficacy: The combination can result in slower tumor progression than checkpoint inhibitors alone, attributed to anti-inflammatory effects and modulation of immune pathways.
  • Enhanced immunogenicity risk: Combined checkpoint inhibitor therapies heighten immunogenicity and anti-drug antibody (ADA) rates, prompting careful immunogenicity monitoring in preclinical and clinical studies, especially as biosimilars may have variable immune profiles in different diseases.
  • Safety and tolerability: Biosimilars such as those for Tocilizumab show comparable safety, immunogenicity, and efficacy to the original drugs in controlled settings, supporting their use in combination regimens.

In sum, researchers deploy Tocilizumab biosimilars with checkpoint inhibitors by:

  • Establishing tumor and toxicity models (mouse or cell-based).
  • Administering each agent singly and in combination.
  • Comparing readouts for tumor suppression, immune activation, and side effect management.
  • Evaluating immunogenicity to ensure biosimilar performance aligns with originator products in multi-agent settings.

In immunogenicity testing for tocilizumab and its biosimilars, the bridging ADA (anti-drug antibody) ELISA format utilizes the biosimilar drug as both capture and detection reagent to monitor patients' immune responses against the therapeutic. This sophisticated assay design enables sensitive detection of anti-tocilizumab antibodies that may develop during treatment.

Bridging ELISA Principle and Setup

The bridging assay format employs tocilizumab (either originator or biosimilar) in a dual capacity - as a capture antibody pre-coated onto the microplate wells and as a detection reagent labeled with either biotin or enzyme tags. This approach creates a "sandwich" configuration where anti-tocilizumab antibodies present in patient serum can simultaneously bind to both the immobilized drug and the labeled drug, forming a detectable bridge complex.

For tocilizumab biosimilar testing specifically, the assay utilizes biotinylated tocilizumab as the capture reagent and sulfo-tag-labeled tocilizumab as the detection reagent. The bound complex (biotin-TC-ADA-sulfo-tag-labeled-TC) generates an electrochemiluminescent signal when read buffer is added, with signal intensity proportional to the concentration of anti-tocilizumab antibodies present.

Biosimilar Integration and Antigenic Equivalence

A critical aspect of using tocilizumab biosimilars in immunogenicity assays is demonstrating antigenic equivalence between the biosimilar and originator products. This equivalence enables the use of a single assay format where biosimilar drugs can serve interchangeably for both coating and detection purposes. This approach streamlines the immunogenicity assessment process and ensures that immune responses detected are relevant regardless of whether patients received the originator tocilizumab or its biosimilar.

Sample Processing and Interference Mitigation

The immunoassay protocol begins with acid dissociation of immune complexes using 300 mM acetic acid at high minimum required dilution (1/80) to separate any pre-formed drug-antibody complexes. This step is crucial because circulating tocilizumab in patient serum can interfere with antibody detection by masking binding sites.

Following dissociation, the assay incorporates interleukin-6 (IL-6) supplementation at 5 μg/ml to address a significant analytical challenge - high levels of serum IL-6 receptor in clinical samples can generate false positive results in the bridging format. The addition of IL-6, which is the natural ligand for the IL-6 receptor, helps quench excess IL-6 receptor and maintains assay specificity while preserving sensitivity and precision.

Assay Validation and Confirmation

The immunogenicity testing protocol includes both screening and confirmatory assay formats. The confirmatory step incorporates a competitive displacement approach where excess unlabeled tocilizumab (50 μg/ml) is added during pre-incubation. This confirmatory drug competes with the labeled detection reagent for antibody binding sites, helping to distinguish specific anti-tocilizumab antibodies from non-specific binding interactions.

Quality control samples are prepared using commercially sourced or custom-synthesized anti-tocilizumab antibodies, typically generating standard curves ranging from 500 ng/ml to 7.8 ng/ml. These controls provide quantitative reference signals that enable determination of anti-tocilizumab antibody levels in unknown patient samples.

The bridging ELISA approach for tocilizumab biosimilar immunogenicity testing represents a sophisticated analytical framework that accounts for the complex interactions between therapeutic antibodies, endogenous targets, and patient immune responses, ensuring reliable monitoring of treatment-emergent immunogenicity throughout therapy.

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 Mihara M, Kasutani K, Okazaki M, et al. Int Immunopharmacol. 5(12):1731-1740. 2005.
5 Sato K, Tsuchiya M, Saldanha J, et al. Cancer Res. 53(4):851-856. 1993.
6 Tsunenari T, Koishihara Y, Nakamura A, et al. Blood. 90(6):2437-2444. 1997.
7 Yao X, Huang J, Zhong H, et al. Pharmacol Ther. 141(2):125-139. 2014.
Indirect Elisa Protocol
FA
General Western Blot Protocol

Certificate of Analysis

Formats Available

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