Anti-Human IL-23 (Guselkumab) [Clone CNTO-1959] — Fc Muted™

Anti-Human IL-23 (Guselkumab) [Clone CNTO-1959] — Fc Muted™

Product No.: I-2155

- -
- -
Product No.I-2155
Clone
CNTO-1959
Target
IL-23
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-23p19
Isotype
Human IgG1λ
Applications
FA
,
IF

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 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 ?
FA,
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 Guselkumab. CNTO1959 (Guselkumab) activity is directed against the p19 subunit of human IL-23.
Background
IL-23 is a member of the IL-12 family of proinflammatory and immunoregulatory cytokines1 and plays a key role in the differentiation and proliferation of type 17 helper T cells (Th17)2. IL- 23 exists as a heterodimer composed of the IL-12p40 subunit and a novel p19 subunit that is shared with IL-393. IL-23 activities lead to the production of Th17-derived pro-inflammatory cytokines IL-17 and IL-221. Additionally, IL-23 possesses potent anti-tumor and anti-metastatic activity in mouse models of cancer, suggesting a potential role for IL-23 in therapeutic treatment of cancer4. IL-23 also contributes to chronic inflammation of immune-mediated diseases including psoriasis and psoriatic arthritis2.

Guselkumab was developed using MorphoSys proprietary HuCAL ® antibody library technology2 to explore IL-23 blockade without interfering with IL-12 activity5. Guselkumab inhibits the biological activity of secreted IL-23 by preventing IL-23 from binding to cell surface receptors and consequently blocking the initiation of IL-23-receptor-mediated signaling.

Guselkumab has been approved for treatment of psoriasis and psoriatic arthritis. Guselkumab is also being tested for the treatment of ulcerative colitis6.

Antigen Distribution
IL-23 is secreted by activated dendritic cells, macrophages, and monocytes.
Ligand/Receptor
IL12B, IL12RB1, IL23R
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Inflammatory 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 Guselkumab biosimilars are used as calibration standards or reference controls in PK bridging ELISAs by serving as the quantitative reference to generate the standard curve against which serum drug concentrations are measured. This approach ensures the assay measures both biosimilar and reference (originator) products with comparable accuracy and precision, which is critical for pharmacokinetic (PK) bridging and comparability assessments.

Essential context and supporting details:

  • Researchers validate the use of the biosimilar as the analytical standard by demonstrating bioanalytical equivalence with the reference product within the assay system. This is achieved by assaying multiple sets of standards and quality control (QC) samples prepared from biosimilar and reference products, then performing statistical analyses to confirm their responses fall within a pre-defined equivalence interval (commonly 0.8–1.25).

  • How calibration works in PK ELISA:

    • The biosimilar Guselkumab is serially diluted to prepare a standard curve in human serum at known concentrations (e.g., 50, 100, up to 12800 ng/mL).
    • Patient serum samples, containing unknown concentrations of Guselkumab, are diluted appropriately and tested in parallel.
    • The ELISA detects drug using specific antibodies; sample concentrations are determined by interpolation from the biosimilar standard curve.
    • QC samples (often including both biosimilar and reference Guselkumab preparations at typical serum concentrations) are measured against the standard curve to validate assay accuracy, precision, and dilutional linearity.
  • Advantages of using biosimilar as standard:

    • A single PK assay using one analytical standard for both biosimilar and reference Guselkumab reduces assay variability, simplifies operations, and avoids cross-method comparability concerns.
    • The method must be scientifically validated to demonstrate the biosimilar mirrors the reference product in assay response, ensuring PK bridging data are robust and interpretable.
  • Implementation in published Guselkumab ELISA approaches:

    • Serum samples from patients under Guselkumab therapy are measured using an ELISA calibrated with known concentrations of Guselkumab, demonstrating that the assay can reliably quantify drug in clinical samples and QC materials after appropriate dilution.
    • Validation includes checks for dilutional linearity, parallelism (between calibration curve and patient samples), and matrix effects, confirming that results from biosimilar and reference materials are equivalent in the assay system.
  • Regulatory and scientific perspective:

    • Industry consensus and regulatory guidance recommend using a single, validated PK assay with one standard for biosimilar bridging studies, contingent on demonstrated equivalence in quantitative performance.
    • Equivalence assessment includes accuracy, precision, total error, and consistency in recovery and instrument response.

Summary table:

Assay ComponentRole of Biosimilar GuselkumabRegulatory/Validation Principle
Calibration standardProvides standard curve for quantifying drug concentration in serumMust show analytical equivalence with reference product
Reference control/QCUsed in QC samples to monitor assay performance and equivalenceValidate accuracy, precision, and recovery
PK bridging ELISAQuantifies unknowns using biosimilar standardMust parallel reference product in clinical matrix

Key requirements:

  • Biosimilar and reference Guselkumab must be bioanalytically comparable when used as standards.
  • Comprehensive assay validation is necessary, including multiple concentration levels, matrix types (human serum), and assessment of performance parameters.
  • Only after demonstrating comparability can the biosimilar be adopted as the universal standard for PK bridging studies.

In summary, research-grade Guselkumab biosimilars can serve reliably as calibration standards or reference controls in PK bridging ELISAs following rigorous validation to ensure bioanalytical equivalence to the originator, allowing accurate measurement of drug concentration in serum samples for PK assessment.

The primary preclinical models used to study the effects of research-grade anti-IL-23 antibody administration on tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, to a lesser extent, humanized mouse models.

Key details:

  • Syngeneic Mouse Models

    • These involve implanting murine tumor cell lines (such as B16F10 melanoma) into immunocompetent mice, which preserves the native mouse immune system and allows robust investigation of TIL responses to anti-IL-23 therapy.
    • Example: A neutralizing anti-IL-23 monoclonal antibody inhibited both tumor growth and early experimental lung metastases in the B16F10 syngeneic mouse model, with effects mediated by innate immune cells and CD8+ T cells.
    • These models are commonly used to characterize changes in TIL populations—including CD8+ cytotoxic T cells and regulatory T cells (Tregs)—after antibody treatment.
  • Humanized Mouse Models

    • These models are generated by engrafting immunodeficient mice with human immune cells (such as PBMCs or CD34+ stem cells), and then implanting either patient-derived tumor xenografts (PDXs) or established human cancer cell lines.
    • They allow the study of anti-IL-23 therapy effects on human TILs within a human-like immune microenvironment. However, data on direct anti-IL-23 administration in humanized systems are primarily prospective; most extensively characterized in vivo studies still use syngeneic systems.
    • Humanized models are especially valuable for understanding the interaction between human-specific TILs and therapeutic antibodies, supporting the translation of findings to clinical contexts.

Model Comparison Table

Model TypeTumor/Immune OriginAntibody TestedKey UseImmune System IntactTIL Analysis?
SyngeneicMouse/mouseAnti-mouse IL-23Tumor growth inhibition, TIL functionYesYes
HumanizedHuman/humanAnti-human IL-23Human TIL dynamics, translational studiesPartially (human)Yes
PDX (non-humanized)Human/mouseLess informativeTumor growth (lacks functional immune response)NoLimited

Summary of findings from in vivo studies:

  • Anti-IL-23 antibody administration in syngeneic models inhibits tumor progression and alters TIL composition, often increasing CD8+ T cell infiltration and reducing suppressive Treg phenotypes.
  • Treg-specific IL-23R targeting (via knockout or antibody blockade) in tumor-bearing mice led to destabilization of intratumoral Tregs, enhanced recruitment of effector CD8+ T cells, and promoted pro-inflammatory (Th1-like) immune responses in the tumor microenvironment.
  • Combination approaches, such as using anti-IL-23 with immunostimulatory antibodies (like anti-GITR), have demonstrated synergistic effects in tumor control via modulation of TILs.

References to direct administration of anti-IL-23 in humanized mice exist primarily in prospective or translational contexts, with syngeneic mouse models forming the backbone of current mechanistic insights into how anti-IL-23 therapy shapes TIL composition and function.

Researchers use Guselkumab biosimilars in preclinical immune-oncology models to examine the effects of blocking the IL-23/Th17 axis, often in combination with other immune checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars, to investigate synergistic anti-tumor effects and immune modulation.

Context and Application:

  • Guselkumab biosimilars are affordable, research-grade antibodies that target IL-23A (p19 subunit), which is pivotal for Th17 cell differentiation and inflammatory cytokine production. In immune-oncology studies, blocking IL-23 signaling is relevant because this pathway may influence immune cell infiltration, tumor microenvironment inflammation, and immune evasion.
  • Checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 are well-established in cancer immunotherapy for releasing brakes on T cells, promoting anti-tumor immunity by neutralizing suppressive signals.

Synergistic Combinations in Complex Models:

  • Combined blockade of IL-23 (using Guselkumab biosimilars) with checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) is hypothesized to:

    • Promote T cell activation and proliferation by releasing multiple inhibitory signals.
    • Alter the tumor microenvironment, reducing regulatory T cell (Treg) activity and increasing effector immune cell infiltration.
    • Disrupt tumor-promoting inflammation while simultaneously boosting anti-tumor responses.
  • Experimental approach:

    • Researchers utilize in vitro assays (such as co-culture systems of tumor cells and immune cells) and in vivo models (e.g., syngeneic mouse tumor models) to administer combinations of Guselkumab biosimilars and checkpoint inhibitors.
    • Outcomes measured include T cell proliferation, cytokine release, immune cell composition within tumors, tumor growth kinetics, and overall survival.
  • Mechanistic rationale:

    • IL-23 blockade may decrease the recruitment and maintenance of immunosuppressive cells (such as pathogenic Th17 and Treg subpopulations) within the tumor.
    • Checkpoint inhibition (CTLA-4/LAG-3) boosts cytotoxic T cell function and reverses T cell exhaustion, with evidence from preclinical studies and clinical trials showing that dual or triple checkpoint blockade yields synergistic suppression of tumor progression by affecting complementary arms of immune regulation.

Advantages of Biosimilars in Research:

  • Cost-effective and standardized: Biosimilars of Guselkumab provide consistent, scalable reagents for bench research, eliminating the cost and access barriers of clinical-grade biologics.
  • Facilitates combination screening: Their availability enables systematic, high-throughput evaluation of combinatory immunotherapy strategies, including checkpoint blockade and cytokine-targeted intervention.

Limitations and Further Research:

  • Use of these combinations in preclinical rather than clinical settings is critical—biosimilars used in research are not approved for human therapy.
  • There remains a need for extensive mechanistic and translational research to clarify the roles of IL-23 within varying tumor microenvironments and how its inhibition affects responsiveness to checkpoint therapy.

In summary, investigators use Guselkumab biosimilars alongside anti-CTLA-4 or anti-LAG-3 biosimilars to dissect and optimize multi-targeted immune-oncology strategies, leveraging their affordability and reproducibility to map complex immune interactions and discover synergistic anti-tumor effects in preclinical models.

In immunogenicity testing, a Guselkumab biosimilar is used as either the capture or detection reagent in a bridging ADA ELISA to identify anti-drug antibodies (ADAs) in patient samples, thus monitoring immune responses against Guselkumab therapy.

Essential context and supporting details:

  • Bridging ADA ELISA Principle:
    The assay detects antibodies developed by patients in response to the therapeutic drug. It utilizes the bivalency of IgG ADAs: one arm binds to an immobilized form of the drug (capture reagent), and the other to a labeled form (detection reagent), forming a "bridge".

  • Role of Guselkumab Biosimilar:

    • A biosimilar to Guselkumab, having the same variable regions, is ideal for research applications and can function interchangeably with the therapeutic drug in the assay.
    • The biosimilar can be immobilized (e.g., biotinylated and bound to a streptavidin-coated plate) as the capture reagent, allowing ADAs in the patient’s serum to bind.
    • For detection, the biosimilar can be conjugated (e.g., HRP-labeled) to provide a measurable signal when complexed with serum ADAs.
  • Protocol Example:

    • Capture: Biotinylated Guselkumab biosimilar binds to a streptavidin-coated ELISA plate.
    • Sample: Patient serum containing possible ADAs is added; ADAs bind to immobilized Guselkumab-related protein.
    • Detection: HRP-conjugated Guselkumab biosimilar is added; it binds to the other antigen-binding site on the ADA, forming a sandwich complex.
    • Measurement: Signal generated by HRP activity reflects the quantity of ADA present.
  • Key Considerations:

    • The biosimilar must match the variable regions of Guselkumab to ensure immunoreactivity with patient-generated ADAs.
    • Assay sensitivity and specificity are influenced by the quality of the biosimilar used, assay setup, and controls to avoid interference from serum matrix or residual drug.
  • Research Usage:
    Guselkumab biosimilar reagents make it possible to run sensitive, customizable ADA bridging ELISAs in preclinical or clinical research settings without using the actual therapeutic product.

Additional information:

  • The bridging ELISA format is the industry standard for ADA detection due to its ability to detect bivalent antibodies and its adaptability for various biotherapeutics.
  • The use of biosimilars helps avoid using precious clinical-grade material and ensures supply consistency for large-scale screening.

In summary, a Guselkumab biosimilar in ADA bridging ELISA serves as both the target and probe for patient-generated anti-Guselkumab antibodies, enabling sensitive, reliable immunogenicity monitoring during therapy.

References & Citations

1 Korn T, Oukka M, Kuchroo V, et al. Semin Immunol. 19(6):362-371. 2007.
2 Markham A. Drugs. 77(13):1487-1492. 2017.
3 Deodhar A, Gottlieb AB, Boehncke WH, et al. Lancet. 391(10136):2213-2224. 2018.
4 Wertheimer T, Zwicky P, Rindlisbacher L, et al. Nat Immunol. 25(3):512-524. 2024.
5 Zhuang Y, Calderon C, Marciniak SJ Jr, et al. Eur J Clin Pharmacol. 72(11):1303-1310. 2016.
6 Hanžel J, D'Haens GR. Expert Opin Biol Ther. 20(4):399-406. 2020.
7 Sofen H, Smith S, Matheson RT, et al. J Allergy Clin Immunol. 133(4):1032-1040. 2014.
8 Blauvelt A, Papp KA, Griffiths CE, et al. J Am Acad Dermatol. 76(3):405-417. 2017.
9 Reich K, Armstrong AW, Foley P, et al. J Am Acad Dermatol. 76(3):418-431. 2017.
10 Mease PJ, Rahman P, Gottlieb AB, et al. Lancet. 395(10230):1126-1136. 2020.

Formats Available

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