Anti-Human IL-6 (Siltuximab) [Clone CNTO-328]

Anti-Human IL-6 (Siltuximab) [Clone CNTO-328]

Product No.: I-450

- -
- -
Product No.I-450
Clone
CNTO-328
Target
IL-6
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
BSF-2, CDF, Hybridoma growth factor (HPGF), IFN-beta-2, HSF
Isotype
Human IgG1κ
Applications
B
,
ELISA
,
FA
,
IF
,
RIA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
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 ?
FA,
ELISA,
B,
RIA,
IF
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 Siltuximab. CNTO-328 (Siltuximab) is a neutralizing monoclonal antibody specific against human IL-6.
Background
IL-6 is a pleiotropic 26 kD protein that can act as both a pro-inflammatory cytokine and an anti-inflammatory myokine, a form of cytokine produced in muscle cells that participates in tissue regeneration and repair, maintenance of healthy bodily functioning, and homeostasis within the immune system 1. IL-6 also plays a part in the endocrine, nervous, and hematopoietic systems, bone metabolism, regulation of blood pressure, and inflammation. Furthermore, IL-6 is an important mediator of fever and of the acute phase response which is the body's rapid attempt to restore homeostasis after tissue injury, infection, neoplastic growth, or immunological disturbance. In its role as an anti-inflammatory myokine, IL-6 precedes the appearance of other cytokines in the circulation, is notably elevated with exercise, and is mediated by both its inhibitory effects on TNF-α and IL-1, and activation of IL-1R⍺ and IL-10. IL-6 signals through a cell-surface type I cytokine receptor complex formed by the binding of IL-6 to IL-6R, which in turn combines with GP130 to transduce extracellular signaling via STAT3 activation. Hence, it is thought that blocking the interaction between IL-6 and GP130 may have therapeutic potential via the inhibition of the IL-6/GP130/STAT3 signaling pathway. Moreover, IL-6 initiates inflammatory and auto-immune processes in many diseases, including diabetes, atherosclerosis, depression, Alzheimer's disease, rheumatoid arthritis, and cancer. For example, multicentric Castleman’s disease is a rare lymphoproliferative disorder caused by dysregulation of IL-6 2. Thus, there is an interest in the therapeutic potential of anti-IL-6 mAbs.

CNTO-328 (Siltuximab) is a chimeric monoclonal antibody that was developed for the treatment of IL-6 related disorders 2,3,4,5. Siltuximab is associated with sustained reductions in IL-6 levels along with various other cytokines and markers 2. In vitro studies in ovarian cancer cells show that siltuximab inhibits IL-6 induced STAT3 activation, nuclear translocation, and downstream gene expression 6. Siltuximab also induces apoptosis 2,7 and reduces C-reactive protein levels 2.

Siltuximab has been approved for the treatment of multicentric Castleman’s disease in HIV-negative patients 1. Siltuximab does not bind to virally produced IL-6 (vIL-6).
Antigen Distribution
IL-6 is a pleiotropic cytokine produced by B lymphocytes, T lymphocytes, macrophages, microglia, fibroblasts, keratinocytes, mesangial cells, vascular endothelial cells, mast cells, and dendritic cells. Additionally, osteoblasts secrete IL-6 to stimulate osteoclast formation. Smooth muscle cells in the tunica media of many blood vessels produce IL-6 as a pro-inflammatory cytokine. IL-6 is also released into circulation in response to various stimuli including PAMPs (pathogen-associated molecular patterns) and cortisol, a hormone produced by the human body under psychologically stressful conditions.
Ligand/Receptor
IL6R
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cell Biology
.
Immunology
.
Inflammatory Disease
.
Innate Immunity
.
Neuroscience
.
Autoimmunity
.
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 Siltuximab biosimilars are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays, particularly to quantify Siltuximab concentrations in serum samples during biosimilar development and comparability exercises.

The typical approach involves the following:

  • Single analytical standard use: The industry consensus is to use a single, well-characterized biosimilar (often research-grade) as the analytical standard for developing the ELISA calibration curve. This standard is serially diluted in a matrix similar to study samples (e.g., human serum) to generate a standard curve that will be used for quantifying unknowns.
  • Bridging ELISA design: The assay is designed, often using anti-idiotypic antibodies, to equally recognize both the reference Siltuximab (innovator product) and the biosimilar, ensuring the standard curve is valid for measuring both.
  • Validation and equivalence testing: The method is validated with quality control (QC) samples prepared from both reference and biosimilar products, spiked into blank matrices at varying concentrations. The QC samples are quantified against the biosimilar-derived calibration curve. The bioanalytical equivalence between test items is established by demonstrating that PK parameters measured for both products fall within pre-set equivalence margins (e.g., 90% CI within 0.8–1.25 range) and that the assay performs comparably for both.

Additional details:

  • Reference control use: The biosimilar standard and sometimes reference Siltuximab are used as controls across plates to ensure assay consistency—allowing for plate-to-plate adjustments and monitoring assay drift or variation. This mirrors practices in standardized ELISA methods that advocate for plate-specific internal standards and positive/negative controls.
  • Calibration linkage: Standards are often calibrated against the innovator product and sometimes traceable to international standards (e.g., NIBSC), adding legitimacy to the quantification.

Summary of key workflow:

  • Serial dilutions of the Siltuximab biosimilar standard are prepared in serum or plasma to create a standard curve.
  • Serum samples (unknowns) and QC samples (biosimilar and reference spike-ins) are analyzed in parallel and referenced to this curve.
  • Controls (positive: known Siltuximab, negative: blank) are included on each plate to monitor for technical variability.
  • The method is stringently validated for accuracy, specificity, and equivalence in detection for both biosimilar and reference Siltuximab, minimizing cross-method variability and supporting regulatory expectations for biosimilar PK comparison.

In summary, research-grade Siltuximab biosimilars serve as the quantified standard for serum drug concentration analysis in PK bridging ELISAs by providing the reference calibration curve and critical controls for method comparability and regulatory validation.

Humanized xenograft models and syngeneic mouse models are the primary in vivo systems for evaluating the anti-tumor efficacy of anti-IL-6 antibodies and characterizing tumor-infiltrating lymphocytes (TILs). Humanized models—often based on immunodeficient mice implanted with human tumors—are the most widely used, as mouse IL-6 does not bind human IL-6 receptor, and humanized antibodies like siltuximab and tocilizumab specifically target human IL-6 or IL-6 receptor. Syngeneic models, while commonly used for immunotherapy research, have limited utility for directly testing human-specific anti-IL-6 antibodies due to species cross-reactivity issues, but may be used with appropriately engineered reagents or mouse-specific analogs.

Model details and relevant context:

  • Humanized Xenograft Models:

    • Immunodeficient mice (such as NOD-SCID or NSG) are engrafted with human tumor cells; these models preserve the production of human IL-6 by tumor cells, allowing for the assessment of anti-IL-6 antibody therapy (e.g., siltuximab, tocilizumab).
    • These models allow detailed study of how blocking IL-6 impacts human cancer stem cell self-renewal, proliferation, and tumor growth.
    • While these mice lack a full adaptive immune system, human immune cells (e.g., PBMCs or TILs) can be co-engrafted to allow for limited characterization of TILs in some setups.
  • Syngeneic Mouse Models:

    • These involve implantation of murine tumor cell lines into immunocompetent mice (e.g., MC38, CT26, B16F10, RENCA), enabling analysis of the endogenous mouse immune response and TIL populations.
    • Anti-IL-6 therapies can be studied using mouse-specific antibodies instead of humanized forms.
    • These models are ideal for mechanistic, immunoprofiling experiments, allowing robust characterization of changes in TILs following drug administration.
Model TypeTumor SourceImmune SystemAntibody CompatibilityTIL Characterization
Humanized XenograftHuman tumor cellsImmunodeficient or reconstitutedHuman-specific (siltuximab, tocilizumab)Possible if human immune cells engrafted
SyngeneicMouse tumor cellsFully functional (murine)Mouse-specific antibodiesRobust, with full immune context

Additional Considerations:

  • Humanized models best capture effects of human IL-6 and its blockade on tumor biology, but have limited immune/TIL readouts unless co-engrafted with human immune cells.
  • Syngeneic models are preferred for full immunophenotyping and immunotherapy evaluation, but require murine-optimized anti-IL-6 reagents.

For studies aiming to analyze both tumor growth inhibition and changes in TILs with anti-IL-6 antibodies, the experimental choice depends on species specificity of the antibody and the immunological endpoints required. Most published studies on research-grade anti-IL-6 antibodies and TIL analyses use these two model systems guided by the above considerations.

Researchers use the Siltuximab biosimilar in combination with other checkpoint inhibitors, such as anti-CTLA-4 or anti-LAG-3 biosimilars, primarily to investigate synergistic effects on tumor immune evasion and the modulation of the tumor microenvironment in immune-oncology models.

Combining these agents is based on their complementary mechanisms:

  • Siltuximab targets and neutralizes interleukin-6 (IL-6), a cytokine that promotes cancer progression, metastasis, immune suppression, and resistance to therapy. By inhibiting IL-6 signaling, siltuximab can reduce tumor-associated inflammation, help restore normal immune responses, and potentially make tumors more susceptible to other immunotherapies.
  • Checkpoint inhibitors (such as anti-CTLA-4 and anti-LAG-3 biosimilars) act by blocking inhibitory pathways on T cells, thereby reinvigorating anti-tumor immune responses.
    • Anti-CTLA-4 mainly acts in the lymph nodes to enhance T cell activation.
    • Anti-LAG-3 modulates T cell exhaustion and function at the tumor site.

Synergy in Complex Models:

  • In research settings, biosimilar antibodies (like siltuximab biosimilars) provide cost-effective, reproducible tools for non-clinical studies, allowing combination experiments to be conducted on a broader scale.
  • In complex immune-oncology models (such as humanized mouse models or ex vivo human tumor cultures), researchers administer the siltuximab biosimilar along with anti-CTLA-4 or anti-LAG-3 biosimilars to:
    • Block IL-6-mediated immune suppression
    • Release multiple inhibitory checkpoints simultaneously (e.g., CTLA-4, LAG-3)
    • Observe the impact on T cell activation, tumor infiltration, tumor growth, and immune cell profiles
  • By targeting distinct immunoregulatory pathways, these combinations can help overcome the limitations of monotherapies (such as incomplete responses due to alternative suppressive mechanisms) and may result in additive or synergistic anti-tumor effects.

Experimental Workflow Example:

  • Tumor-bearing immune-oncology models are established.
  • Siltuximab biosimilar is administered to inhibit IL-6 signaling.
  • Checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-LAG-3) are co-administered.
  • Tumor growth rates and immune cell populations (e.g., T cell activation, regulatory T cell depletion, cytokine levels) are monitored.
  • The data are compared to monotherapy controls to assess synergistic vs. additive effects.

Current Status and Limitations:

  • While these approaches are widely used in preclinical research, the siltuximab biosimilar provided by commercial suppliers is for research use only and not approved for clinical application.
  • Most data about such combinations come from animal models or in vitro studies; there are few published results from clinical trials of siltuximab combined with checkpoint inhibitor therapy as of 2024.

Summary Table: Mechanisms in Combination Studies

AgentTarget/MechanismEffect in Combination
Siltuximab biosimilarNeutralizes IL-6 (immune modulation)Reduces tumor-driven immune suppression
Anti-CTLA-4 biosimilarBlocks CTLA-4 on T cellsEnhances T cell priming/activation
Anti-LAG-3 biosimilarBlocks LAG-3 on exhausted T cellsFurther restores T cell function

This combination approach enables researchers to systematically dissect immune regulatory networks in cancer and explore rational immunotherapy strategies for future translational studies.

A Siltuximab biosimilar can be used as both the capture and detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor a patient's immune response against Siltuximab by exploiting the bivalent nature of patient-derived ADAs that bind simultaneously to two molecules of the biosimilar.

In a typical bridging ADA ELISA:

  • Capture reagent: The Siltuximab biosimilar is immobilized (commonly via biotinylation and binding to streptavidin-coated wells), which captures circulating anti-Siltuximab antibodies (ADAs) present in the patient sample.
  • Detection reagent: A differently labeled version of the same biosimilar (e.g., conjugated with HRP or a dye) is used to detect bound ADAs by bridging to their other Fab arm.

Essential details:

  • The ADA in patient serum forms a “bridge” between the plate-bound (capture) Siltuximab biosimilar and the labeled (detection) Siltuximab biosimilar, producing a signal correlated to ADA concentration.
  • Both capture and detection use the biosimilar rather than the originator, ensuring assay consistency, specificity, and relevant matrix effects for clinical comparability studies.
  • This format provides high sensitivity for ADA detection, essential for monitoring immunogenicity profiles during therapy.
  • The strength and reliability of the assay depend on the quality, purity, and reproducibility of the biosimilar reagent prepared for ELISA.
  • Similar approaches have been validated for multiple monoclonal antibodies and their biosimilars, showing that immunogenicity profiles for biosimilars and reference drugs are highly comparable when using this method.

In summary, a Siltuximab biosimilar used as capture and detection reagent in a bridging ADA ELISA allows the sensitive, specific quantification of anti-Siltuximab antibodies in patient samples, thereby providing critical immunogenicity monitoring during therapy.

References & Citations

1. Trikha M, Corringham R, Klein B, et al. Clin Cancer Res. 9(13):4653-4665. 2003.
2. Markham A, Patel T. Drugs. 74(10):1147-1152. 2014.
3. van Zaanen HC, Koopmans RP, Aarden LA, et al. J Clin Invest. 98(6):1441-1448. 1996.
4. van Zaanen HC, Lokhorst HM, Aarden LA, et al. Br J Haematol. 102(3):783-790. 1998.
5. van Zaanen HC, Lokhorst HM, Aarden LA, et al. Leuk Lymphoma. 31(5-6):551-558. 1998.
6. Guo Y, Nemeth J, O'Brien C, et al. Clin Cancer Res. 16(23):5759-5769. 2010.
7. Hunsucker SA, Magarotto V, Kuhn DJ, et al. Br J Haematol. 152(5):579-592. 2011.
8. Voorhees PM, Chen Q, Kuhn DJ, et al. Clin Cancer Res. 13(21):6469-6478. 2007.
9. Cavarretta IT, Neuwirt H, Zaki MH, et al. Adv Exp Med Biol. 617:547-555. 2008.
10. Voorhees PM, Chen Q, Small GW, et al. Br J Haematol. 145(4):481-490. 2009.
11. Karkera J, Steiner H, Li W, et al. Prostate. 71(13):1455-1465. 2011.
12. Kurzrock R, Voorhees PM, Casper C, et al. Clin Cancer Res. 19(13):3659-3670. 2013.
13. van Rhee F, Wong RS, Munshi N, et al. Lancet Oncol. 15(9):966-974. 2014.
14. van Rhee F, Rosenthal A, Kanhai K, et al. Blood Adv. 6(16):4773-4781. 2022.
B
Indirect Elisa Protocol
FA
IF
RIA

Certificate of Analysis

Formats Available

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