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 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.
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 commonly employed as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs for quantifying drug concentrations in serum samples due to their demonstrated analytical equivalence to the originator product.
In a typical PK bridging ELISA:
Calibration standards are generated by serially diluting the Guselkumab biosimilar in a blank serum matrix to create a standard curve covering the expected range of concentrations in patient samples.
The biosimilar standard curve serves as the benchmark for quantifying Guselkumab levels in serum samples by comparing the assay signal (optical density) of unknowns against the curve.
Quality control (QC) samples and validation sets are also prepared using the biosimilar, ensuring assay precision and accuracy across runs.
Key considerations and supporting details:
Industry guidance and regulatory practice support the use of a single analytical standard—typically the biosimilar—for both biosimilar and originator products when bioanalytical comparability has been demonstrated (i.e., analytical equivalence for quantification of both products in serum matrix has been validated).
Validation studies must show that calibration curves generated with the biosimilar provide equivalent measurement accuracy and precision compared to reference product curves, often by statistical analysis of measured concentrations in QC samples (e.g., equivalence intervals of 0.8-1.25 for geometric mean ratios).
The use of a single standard reduces variability, avoids the need for multiple methods or crossover analyses, and streamlines blinded clinical study operations.
In practice, this approach has yielded results with acceptable recoveries (e.g., 89–107%), low coefficients of variation (CV ≤13%), and dilutional linearity, supporting the suitability of biosimilar standards for serum Guselkumab measurement.
Summary workflow:
Prepare serial dilutions of the Guselkumab biosimilar to establish calibration curves in blank serum.
Analyze patient serum samples using the same ELISA.
Quantify unknown Guselkumab concentrations by interpolation from the biosimilar-derived calibration standard curve.
Use biosimilar as a reference control and for QC samples during assay validation to ensure robustness and reliability.
By following this paradigm, PK bridging ELISA assays can accurately measure serum Guselkumab concentrations during biosimilar development or clinical monitoring, provided rigorous analytical validation confirms bioanalytical equivalency between biosimilar and reference products.
The primary in vivo models used to administer a research-grade anti-IL-23 antibody and study tumor growth inhibition as well as the characterization of tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and humanized mouse models.
Key details:
Syngeneic models are the most common for anti-IL-23 antibody administration in preclinical cancer studies. These involve implanting murine tumor cells into immunocompetent mice (such as C57BL/6 or BALB/c strains) bearing a fully functional and native immune system, which allows for the direct study of immune modulation—including TIL expansion and phenotyping—following administration of anti-IL-23 antibodies.
Example: In the B16F10 melanoma syngeneic mouse model, administration of anti-IL-23 monoclonal antibody has been shown to suppress metastasis and inhibit primary tumor growth, with the mechanism attributed to enhanced activity of NK cells and CD8+ T cells (components of TILs).
These models provide direct measurement of alterations in TIL composition, cytokine profile, and effector function within tumors following treatment.
Humanized mouse models are used for more translational studies, particularly when human-specific immune responses or antibodies are being tested. In these models, human immune cells (such as PBMCs, or hematopoietic stem cells) are engrafted into immunodeficient mice (e.g., NOG or NSG strains), sometimes alongside human tumor xenografts. This system permits the study of human TILs and their modulation by immunotherapeutics like anti-IL-23 antibodies.
These models recapitulate human immune-tumor dynamics, critical for evaluating therapies intended for clinical translation, particularly for human-specific antibodies.
Other models for TIL characterization:
Patient-derived xenografts (PDX) are sometimes employed for studying TILs within human tumors engrafted in immunodeficient mice, but without a humanized immune system, their application for anti-IL-23 therapy is limited in immunology-focused studies.
Characterizing TILs:
In both syngeneic and humanized models, researchers use flow cytometry, immunohistochemistry, and transcriptomic profiling to quantify and phenotype TILs after anti-IL-23 treatment, focusing on changes in CD8+ T cells, Tregs, and production of effector cytokines (e.g., IFNγ).
Syngeneic models are particularly useful for evaluating how IL-23 blockade destabilizes suppressive Treg populations within tumors, leading to increased recruitment and activation of effector CD8+ T cells and enhanced tumor clearance.
In summary:
Syngeneic mouse models are the primary choice for mechanistic studies of anti-IL-23 antibody effects on tumor growth and immune infiltration.
Humanized mouse models are essential for translational work and for analyzing effects on human TILs.
Research protocols in both model types enable quantification and functional characterization of TILs following anti-IL-23 antibody administration, with emphasis on both tumor growth control and immune cell dynamics.
Researchers use Guselkumab biosimilars—which target IL-23A—in complex immune-oncology models primarily as cost-effective, research-only tools to investigate the effects of IL-23 pathway modulation, often in combination with other checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 biosimilars. By combining these agents, scientists aim to understand potential synergistic anti-tumor effects and the interplay between different immune regulatory pathways.
Key aspects of these combination studies include:
Mechanistic Dissection: Guselkumab biosimilars allow for precise blockade of the IL-23/Th17 axis, while anti-CTLA-4 and anti-LAG-3 biosimilars inhibit immune checkpoints that dampen T cell responses. Their combination enables dissection of how concurrent modulation of pro-inflammatory (IL-23) and inhibitory (checkpoint) signals alters anti-tumor immunity.
Synergistic Evaluation: Studies in preclinical immune-oncology models often test whether combining IL-23 inhibition with checkpoint blockade produces additive or synergistic increases in T cell activation, proliferation, or tumor infiltration compared to either agent alone. Dual or triple checkpoint inhibition (e.g., anti-PD-1, anti-CTLA-4, anti-LAG-3) has shown enhanced efficacy, and researchers use Guselkumab biosimilars to explore if further IL-23 axis modulation intensifies these effects.
Assay Approaches:
Cell-based functional assays: Measuring cytokine production, T cell proliferation, or tumor cell killing in response to various drug combinations.
In vivo tumor models: Assessing tumor growth, immune cell infiltration, and survival following treatment with combinations of biosimilars targeting IL-23, CTLA-4, or LAG-3.
Signal pathway studies: Elucidating how inhibition of IL-23/Th17 interrelates with immune checkpoint blockade at the molecular level.
Experimental Rationale: Evidence suggests that targeting multiple immune escape mechanisms (such as by combining checkpoint blockade with agents that modulate the tumor immune microenvironment, including IL-23) can overcome resistance and induce more robust anti-tumor responses. For example, combined blockade of CTLA-4 and LAG-3 has been shown to synergistically inhibit T cell proliferation and cytokine responses; adding IL-23 inhibition could amplify these effects by reducing immunosuppressive Th17 responses within the tumor.
Research Advantages:
Cost and accessibility: Biosimilars are more affordable for preclinical studies, enabling high-throughput screening and wider access.
Standardization: Consistent, research-grade biosimilars support reproducibility in multi-agent experiments.
Direct clinical application evidence for Guselkumab in oncology is still limited, but preclinical models using its biosimilars—sometimes in combination with checkpoint inhibitor biosimilars—aim to accelerate our understanding of immune-oncology synergy and to identify rational drug combinations for further clinical testing.
In summary, Guselkumab biosimilars are combined with other checkpoint inhibitor biosimilars in preclinical studies to elucidate synergistic mechanisms and optimize immunotherapy strategies, leveraging their accessibility for widespread, mechanistically rich immune-oncology research.
A Guselkumab 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 Guselkumab. In this assay format, the biosimilar serves as a functional equivalent to the original therapeutic drug, enabling detection of anti-Guselkumab antibodies in patient serum.
Context and Supporting Details:
Bridging ADA ELISA Principle: The assay detects patient-generated anti-drug antibodies by using the therapeutic (or its biosimilar) in two roles:
Capture: Guselkumab biosimilar is coated on the ELISA plate to "capture" any ADA present in the patient’s serum.
Detection: After washing away unbound components, the same (or a labeled) Guselkumab biosimilar is added to bind the captured ADA, forming a "bridge" between the two drug molecules via the ADA.
Why Use a Biosimilar?
Equivalence: Guselkumab biosimilars are designed to closely mimic the structure and function of the original drug, including the same IL-23A p19 subunit recognition, making them suitable proxies in analytical settings.
Accessibility and Consistency: Research-grade biosimilars offer consistent performance and easier access for large-scale or routine ADA testing in research labs.
Application in Immunogenicity: By using the biosimilar as both capture and detection reagents, any anti-Guselkumab antibodies (regardless of their isotype) present in a patient's sample will bridge the two biosimilar molecules, yielding a measurable ELISA signal. This method is standard for immunogenicity monitoring of biologics and their biosimilars.
Assay Considerations:
Sensitive Detection: The sensitivity and specificity of the ADA ELISA are influenced by the structural similarity of the biosimilar to the reference drug, reagent quality, and assay conditions.
Research Use Only: Commercially available Guselkumab biosimilars for this purpose are typically for research use, not clinical diagnostics or therapeutic interchange.
Clinical Context:
The appearance of ADA can affect drug efficacy and safety, so routine immunogenicity testing is valuable in both research and, where validated, clinical settings.
Factors such as concomitant medications, sample timing, and assay methodology can impact ADA detection rates.
Summary Table: Use of Guselkumab Biosimilar in Bridging ADA ELISA
Assay Role
Function
Relevance of Biosimilar
Capture reagent
Immobilizes to plate, binds ADA from patient sample
Mimics drug epitope for antibody capture
Detection reagent
(Often labeled) binds to ADA, completes the "bridge"
Structural equivalence ensures accuracy
By employing a Guselkumab biosimilar as the capture and/or detection reagent, the assay specifically recognizes anti-Guselkumab antibodies and quantifies immunogenicity in patient samples in a cost-effective and scalable manner for research.
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.