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.
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.
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 are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as the standard material for generating a calibration curve, against which the concentration of tocilizumab in serum samples is quantitatively measured.
How PK Bridging ELISA Works with Reference Standards:
In a typical PK bridging ELISA for monoclonal antibodies like tocilizumab, the assay plate is coated with a target antigen (e.g., soluble IL-6 receptor, sIL-6R) to “capture” tocilizumab molecules from samples or standards.
After incubation and washing, bound tocilizumab is detected with an anti-tocilizumab antibody (often labeled for detection).
A calibration (standard) curve is established using serial dilutions of a well-characterized tocilizumab biosimilar or reference material in matrix-matched serum. This curve relates absorbance (signal) to known drug concentrations.
Role of Biosimilars as Calibration Standards:
Biosimilars, once shown to be highly similar in structure and function to the reference (originator) mAb, can be used as calibration standards. Their known and verified concentrations form the basis of the curve to interpolate unknown sample concentrations.
Using a research-grade biosimilar ensures the calibration curve is relevant to the analyte present in test samples and supports comparability studies between biosimilar and reference products (critical for PK bridging studies).
The calibration range, accuracy, and linearity are validated using these biosimilar standards to meet regulatory and methodological requirements.
Reference Controls:
Besides serving as calibrators, biosimilars are used as positive controls in each run to confirm assay performance (sensitivity, reproducibility) and reference response.
Guiding Principles and Examples:
For instance, in published studies of tocilizumab ELISA, standards are created by spiking known concentrations of tocilizumab (either originator or biosimilar) into blank human serum to mimic patient samples. These standards generate the reference curve used for quantification.
The similarity in PK parameters between biosimilars and originators (such as AUC and Cmax) is established in clinical PK bridging studies, but for the ELISA itself, the suitability of the biosimilar as a standard is determined by its demonstrated analytical similarity and validated performance in the assay system.
Summary Table: ELISA Reference Material Use
Material
Purpose in ELISA
Why Acceptable?
Reference (originator) Tocilizumab
Calibration and control
Gold standard, clinically validated
Research-grade Tocilizumab Biosimilar
Calibration and control
Demonstrated analytical and clinical similarity
PK bridging ELISA assays thus rely on biosimilar or originator tocilizumab as calibration standards and reference controls to ensure accurate, reproducible measurement of drug concentrations in serum. The biosimilar is chosen if it is proven analytically and functionally equivalent to the originator.
The primary models used for in vivo administration of research-grade anti-IL-6R antibodies to assess tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are:
Humanized xenograft models (often using immunodeficient mice engrafted with human tumors)
Syngeneic mouse tumor models (using immunocompetent mice with mouse-derived tumors, sometimes genetically modified or transgenic)
Humanized Xenograft Models:
These models use immunodeficient mice (such as SCID) engrafted with human tumor cell lines, as murine IL-6 does not bind human IL-6R. In one experiment, SCID mice were inoculated with human myeloma cells, and anti-human IL-6R antibody (e.g., PM1) was administered, which inhibited tumor growth when given soon after implantation.
These models allow study of human-specific pathways and direct effects of human-targeted antibodies, but lack a fully functional immune system, limiting characterization of TIL complexity, especially lymphocyte subsets.
Syngeneic Tumor Models:
These models use mouse tumor cell lines in immunocompetent mice. They provide a fully functional mouse immune system, enabling detailed in vivo study of TILs and immunotherapy responses.
Syngeneic models are widely utilized to profile changes in TIL populations, study immune checkpoint inhibitor efficacy, and model dynamic immune-tumor interactions resulting from drug administration.
Technical Considerations:
Cross-reactivity: Research-grade anti-IL-6R antibodies for syngeneic models typically require cross-reactivity with murine IL-6R, unless the model uses transgenic mice expressing human IL-6R or tumors overexpressing the human receptor.
Transgenic/Chimeric Models: Variants such as humanized mice with human IL-6R expression, or chimeric human/mouse antibodies, can be used to overcome species specificity, enabling characterization of immune responses (including TILs) in a more clinically relevant manner.
Syngeneic models (such as MC38 colon carcinoma, TC-1 lung carcinoma) are particularly useful for TIL characterization, providing detailed baseline immune profiling and allowing assessment of changes following antibody treatment.
Summary Table
Model Type
Immune System
IL-6R Targeting
TIL Characterization
Example Use Case
Humanized Xenograft
Immunodeficient
Human IL-6R
Limited
Myeloma xenograft in SCID mice
Syngeneic Mouse
Immunocompetent
Murine/Transgenic IL-6R
Detailed
MC38, TC-1 models, TIL profiling
Transgenic Syngeneic
Immunocompetent
Human IL-6R (transgenic)
Detailed
Human antigen-expressing syngeneic
Key Insights:
Human tumor xenograft models using immunodeficient mice are ideal for direct human IL-6R targeting and evaluating tumor growth inhibition.
Syngeneic and transgenic mouse models are essential for analyzing changes in the native immune microenvironment and TILs after anti-IL-6R therapy, provided cross-reactivity or human IL-6R expression is achieved.
Detailed TIL characterization (phenotyping, quantification) is most feasible in fully immunocompetent syngeneic settings, increasing translational relevance for immune response studies.
Researchers use Tocilizumab biosimilars in combination with checkpoint inhibitors (ICIs) such as anti-PD-1 in preclinical and clinical immune-oncology models to explore potential synergistic effects on both anti-tumor efficacy and the management of immune-related adverse events (irAEs). The published research has primarily focused on the dual impact of this combination, addressing both tumor progression and immune toxicity in complex models.
Key strategies and findings include:
Synergistic anti-tumor effect: In murine models, Tocilizumab (an IL-6 receptor inhibitor) combined with checkpoint inhibitors—specifically PD-1 inhibitors—resulted in significantly slower tumor growth than PD-1 inhibitors alone. The mechanism involves inhibition of the IL-6–JAK2–STAT3 pathway, which is implicated in tumor proliferation and immune-related inflammation.
Model systems: C57BL/6J mice were treated with checkpoint inhibitors (e.g., anti-PD-1) alone and in combination with Tocilizumab. Outcomes included measurement of tumor growth, immune cell infiltration, cytokine levels, and adverse event profiles such as myocarditis (a known immune-related complication of ICIs).
Mechanistic exploration: The combination was found to alleviate immune-mediated myocardial injury (a representative irAE) by inhibiting M1 macrophage polarization and reducing IL-6-driven STAT3 activation. Researchers tracked IL-6 levels in blood and tissue, as patients with high IL-6 often experience worse outcomes and more severe irAEs.
Implications for broader checkpoint blockade combinations: Although the detailed research publicly available primarily involves PD-1 inhibitors, the general approach can theoretically extend to other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars. The rationale is that all these agents can drive robust immune activation and auto-inflammatory toxicities, which may be mediated through overlapping cytokine pathways including IL-6. Combining Tocilizumab could thus modulate excessive IL-6 signaling, mitigating these side effects while possibly enhancing anti-tumor efficacy when used with various ICIs.
Biosimilar use justification: Studies show that Tocilizumab biosimilars display comparable efficacy, safety, and immunogenicity to the reference product (Actemra/RoActemra), which supports their use in experimental and clinical studies seeking cost-effective, scalable solutions in these complex models.
In summary: Researchers use Tocilizumab biosimilars to dampen IL-6-mediated inflammation, reduce immune-related side effects, and potentially augment the anti-tumor response from checkpoint inhibitor regimens in immune-oncology models. Most data so far covers PD-1 inhibitor combinations, but the mechanistic insights suggest potential for combination with other checkpoint targets (e.g., CTLA-4, LAG-3), though specific preclinical or clinical reports are less common for those specific pairings as of 2024.
A Tocilizumab biosimilar can be used as either the capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor immune responses against the therapeutic drug, leveraging its antigenic equivalence to the originator Tocilizumab. In a typical bridging ADA ELISA format, both the capture and detection reagents are forms of the therapeutic—here, the Tocilizumab biosimilar or its labeled derivatives—enabling detection of patient anti-Tocilizumab antibodies, which form a bridge between the two drug molecules.
Essential Context and Technical Details:
The biosimilar is used in the same way as the originator Tocilizumab due to demonstrated antigenic equivalence. This allows for a single assay format to be validated for both biosimilar and originator products, supporting regulatory expectations for immunogenicity assessment.
In the bridging assay:
Biotinylated Tocilizumab biosimilar (capture reagent) is immobilized on a streptavidin-coated plate.
Sulfo-tag-labeled Tocilizumab biosimilar (detection reagent) is used to detect the presence of anti-drug antibodies in patient samples. If a patient's serum contains anti-Tocilizumab antibodies, these antibodies bind to both the capture and detection reagents, forming a "bridge".
The immune complex generated by this binding results in a measurable signal (commonly ECL, electrochemiluminescence), indicating the presence of anti-Tocilizumab antibodies.
For biosimilars, it's crucial that the assay can detect antibodies against both the reference and biosimilar molecules, confirming that immunogenicity profiles are comparable and no new ADA risk is introduced by the biosimilar.
Assay developers must account for interference by serum IL-6R (Tocilizumab’s natural target), which can affect results. High concentrations of IL-6 (the natural ligand) are added to quench excess receptor, ensuring specific ADA detection.
Additional Relevant Information:
Regulatory and scientific literature recommends assay formats using biosimilar drug for capture/detection to facilitate direct immunogenicity comparisons and support biosimilar registration.
Clinical studies on Tocilizumab biosimilars (e.g., BAT1806, MSB11456) consistently show comparable immunogenicity profiles to the reference product, validating the use of biosimilar as assay reagent.
In summary, Tocilizumab biosimilars serve as precise substitutes for the originator drug in the bridging ADA ELISA, enabling robust immune monitoring and direct comparison of immunogenicity between biosimilar and original products.
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.