Anti-Human IL-23A (p19) (Tildrakizumab) [Clone MK-3222] — Fc Muted™

Anti-Human IL-23A (p19) (Tildrakizumab) [Clone MK-3222] — Fc Muted™

Product No.: I-2175

- -
- -
Product No.I-2175
Clone
MK-3222
Target
IL-23A (p19)
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-23p19
Isotype
Human IgG1κ
Applications
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
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
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 Tildrakizumab. MK-3222 (Tildrakizumab) targets the p19 subunit of 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.

Tildrakizumab is a humanized monoclonal antibody that inhibits IL-23 interaction with the IL-23 receptor by selectively binding the IL-23 p19 subunit5,6. Tildrakizumab has been approved for the treatment of plaque psoriasis.

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
.
Immunology
.
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 Tildrakizumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by generating standard curves that allow for the accurate quantification of Tildrakizumab concentration in human serum samples.

Essential context and supporting details:

  • The biosimilar (e.g., research-grade Tildrakizumab) is typically supplied in a lyophilized (freeze-dried) form with a defined concentration and is reconstituted, then serially diluted in blank serum or a suitable matrix to prepare calibration standards spanning the expected concentration range in patient samples.
  • These standards are included in each ELISA plate run to create a standard curve—usually by plotting absorbance (colorimetric signal) versus drug concentration—enabling the concentration of Tildrakizumab in unknown samples to be interpolated from this curve.
  • Reference controls or quality control (QC) samples, often at low, medium, and high concentrations, are run alongside standards to assess assay accuracy and precision across the range of quantification.
  • Typically, the same biosimilar standard used for calibration is also used as a reference control for validation and monitoring assay performance, supporting comparability and traceability across studies and labs.
  • In PK bridging ELISAs developed for biosimilars, using a research-grade biosimilar as the standard ensures that the assay can reliably measure either the originator or biosimilar version, which is critical for bioequivalence and bridging studies.

Workflow summary:

  • Prepare a dilution series of the research-grade Tildrakizumab biosimilar in human serum or assay buffer to cover the desired analytical range (e.g., 0.1–100 μg/mL).
  • Apply standards, controls, and samples to ELISA plates, following validated protocols for incubation, washing, and detection (e.g., using HRP-conjugated antibodies and colorimetric readout).
  • Generate a standard curve using the known concentrations, and calculate the Tildrakizumab concentration in patient serum samples by interpolation using four-parameter logistic regression or similar.

This use of calibrated biosimilar in assay setup ensures consistency, traceability, and regulatory compliance for pharmacokinetic measurements in clinical and research settings, allowing for accurate comparison between biosimilar and originator products.

The primary in vivo models for studying anti-IL-23A (p19) antibody effects on tumor growth and tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models; use in humanized models is rarer, largely due to species-restricted IL-23A interactions and antibody specificity.

Syngeneic Mouse Tumor Models:

  • Common models: B16F10 melanoma, CT26 colon carcinoma, and RENCA renal carcinoma.
    • B16F10 melanoma is frequently used to examine mechanisms of immunosuppression and immune cell dynamics, including how the IL-23/IL-23R axis influences Treg identity and TIL composition.
    • RENCA is noted for a highly immune-infiltrated microenvironment, whereas B16F10 is poorly infiltrated, which allows researchers to compare antibody effects across different immune contexts.
    • CT26 features robust CD8+ T cell infiltration, making it suitable for studying cytotoxic responses after blocking IL-23A.

Experimental features:

  • Administration: A research-grade anti-IL-23A (p19) antibody is generally administered systemically (e.g., intraperitoneal), and tumor growth is monitored over time.
  • TIL profiling: These models permit extensive flow cytometry or single-cell RNA-seq analysis of TILs to assess how anti-IL-23A changes the abundance or activation state of Tregs, CD8+ T cells, myeloid-derived suppressor cells, and other immune subsets.
  • Insight: Such studies reinforce that IL-23 supports immunosuppressive Treg programs in tumors, and antagonism may enhance anti-tumor CD8+ T cell activity.

Humanized Mouse Models:

  • Use of humanized mice (with a reconstituted human immune system) for anti-IL-23A, p19 studies is possible but far less common, primarily due to the need for cross-reactive antibodies and the cost/complexity involved.
    • Most existing published research on IL-23/p19 blockade in tumors focuses on mouse syngeneic models, while humanized models are more often used for other checkpoint therapies.
    • Antibodies such as BI 655066 (humanized anti-IL-23 mAb) have been generated for clinical purposes, but in vivo tumor experiments with these reagents in humanized mice are not well documented in the preclinical literature.

Key summary table:

Model typeTypical tumor linesAnti-IL-23A use in vivoTIL characterizationNotes
Murine syngeneicB16F10, CT26, RENCARoutineExtensive (by FACS, scRNA-seq)Directly applicable for mouse reagents and mechanistic studies
Humanized (immunodeficient mice engrafted with human immune cells)VariedRare and technically challengingLess documented; possible with cross-reactive mAbUsed mostly for translational/clinical agent validation, not routine for mechanistic IL-23 studies

Conclusion:
Murine syngeneic models—notably B16F10, CT26, and RENCA—are the principal systems used to study anti-IL-23A administration, tumor growth inhibition, and TIL analysis in vivo. Use of humanized models for this purpose is far less common due to practical constraints and is not yet standard for mechanistic IL-23A/p19 tumor immunology investigations.

Researchers studying synergistic effects in immune-oncology models often use combinations of checkpoint inhibitors, such as anti-CTLA-4, anti-LAG-3, and biosimilars, but there is currently limited specific evidence of direct use of the Tildrakizumab biosimilar (anti-IL-23A) with other checkpoint inhibitors in published preclinical or clinical synergy studies. The underlying combination rationale is to target distinct immune pathways—checkpoint inhibition (CTLA-4/LAG-3/PD-1) unleashes T cell responses, while cytokine-targeted antibodies like Tildrakizumab modulate antigen-presenting cell and T cell differentiation and potentially reduce suppressive signals in the tumor microenvironment.

Context and Supporting Details:

  • Checkpoint inhibitor combination logic: Combining inhibitors of multiple immune checkpoints (e.g., CTLA-4, PD-1/PD-L1, LAG-3) is established as a strategy to increase anti-tumor immune activity, overcome resistance to monotherapy, and broaden the impact on T cell activation and exhaustion. Distinct mechanisms, such as CTLA-4 blockade enhancing T cell priming in lymph nodes and PD-1/PD-L1 blockade acting at peripheral tumor sites, can yield additive or synergistic effects.

  • Role of IL-23 inhibition (Tildrakizumab): Tildrakizumab targets the IL-23/Th17 axis, which can contribute to tumor progression and immune suppression. By modulating cytokine signals, IL-23 blockade may alter the tumor microenvironment, potentially making it more permissive for T cell-mediated attack induced by checkpoint inhibitors. Preclinical rationale supports combining cytokine-targeted agents with ICIs to amplify anti-tumor immunity, though specific experimental proof for Tildrakizumab with anti-CTLA-4 or anti-LAG-3 biosimilars in cancer models is not described in current literature.

  • Practical use in models: In preclinical studies, researchers typically:

    • Administer biosimilars (e.g., Tildrakizumab, anti-CTLA-4, anti-LAG-3) together in mouse tumor models or engineered cell lines.
    • Assess tumor growth inhibition, immune cell activation (especially T cells and dendritic cells), cytokine profiles, and survival outcomes.
    • Use flow cytometry, immunohistochemistry, and gene expression profiling to characterize changes in immune cell subsets and signaling.
    • Monitor safety (adverse events, autoimmunity markers, immunogenicity).
  • Challenges and caveats: Major hurdles are increased risk of immune-related toxicities (noted in past combinations like anti-CTLA-4 plus anti-PD-1), and sometimes unpredictable interactions that may not always yield synergy. Data on combination therapy with cytokine inhibitors (e.g., anti-IL-23A) and checkpoint blockade are mostly extrapolated from related agents; robust cancer data on tildrakizumab biosimilar combinations is currently lacking.

Additional Relevant Information:

  • Most combination immune-oncology studies currently focus on dual or triple checkpoint blockade (PD-1/LAG-3, CTLA-4/PD-1), with cytokine blockers like Tildrakizumab primarily investigated in autoimmunity and not widely reported in oncology synergy experiments.
  • Tildrakizumab biosimilar is available for research use and could be utilized to probe IL-23's role in the tumor microenvironment alongside checkpoint inhibition, but peer-reviewed synergy results are not yet reported.
  • Safety/tolerability of Tildrakizumab is established in other indications, but immune-related adverse events should be carefully monitored when used in combination, based on prior evidence in other antibody combinations.

In summary, while the conceptual basis and methodology for combining Tildrakizumab biosimilar with checkpoint inhibitors in immune-oncology models is well described, there is a lack of direct published data showing their synergistic effects. Existing literature primarily explores multi-checkpoint combinations and preclinical rationales; actual experimental details with Tildrakizumab biosimilar plus anti-CTLA-4 or anti-LAG-3 must still be developed and published.

A Tildrakizumab biosimilar is used as both capture and detection reagent in a bridging ADA (anti-drug antibody) ELISA to monitor and compare a patient's immune response against the biosimilar and the original therapeutic drug. In this assay format, the biosimilar molecule is labeled in two ways: one portion is typically biotinylated (for capture on a streptavidin-coated plate), and another is labeled (often with HRP or a similar enzyme) for detection.

Key points on assay setup and rationale:

  • Bridging Assay Principle: In the ADA ELISA, patient serum containing anti-drug antibodies (if present) will bind to both the capture (biotinylated) and detection (enzyme-labeled) forms of the biosimilar. This forms a "bridge" between the two labeled drug molecules, generating a measurable signal.
  • Use of the Biosimilar: Using the biosimilar as the reagent (rather than the originator) ensures the assay detects antibodies reactive to the biosimilar's specific molecular structure, which is central for assessing biosimilar immunogenicity.
  • Assay Validation: Before routine use, the assay must demonstrate that the biosimilar and originator have antigenic equivalence—meaning they both bind similarly to a panel of control antibodies. This testing assures that anti-drug antibodies against either version (biosimilar or originator) can be reliably detected in patient samples.
  • Regulatory Recommendation: Current guidance recommends a single, biosimilar-based assay for comparative immunogenicity because it simplifies validation and provides a consistent platform for monitoring patient immune responses across both biosimilar and reference treatments, provided cross-reactivity and equivalence are shown.
  • Interpretation of Results: The appearance of a signal indicates the presence of anti-drug antibodies against Tildrakizumab (the biosimilar and, by validated equivalence, the reference product). High circulating drug levels can interfere with this assay ("drug tolerance" limitation), potentially leading to false negatives if the drug concentration in the serum exceeds the assay’s tolerance threshold.

Summary Table—ADA Bridging ELISA Using Tildrakizumab Biosimilar

Reagent FunctionMolecule UsedPurpose in ADA ELISA
CaptureBiotinylated BiosimilarBinds ADA on plate
DetectionEnzyme-labeled BiosimilarDetects bound ADA for signal
Patient SampleSerumMay contain anti-drug antibody

This approach allows for sensitive and specific detection of anti-Tildrakizumab antibodies in patient samples and is central to demonstrating biosimilar immunogenicity comparability as required by regulatory agencies.

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 Markham A. Drugs. 78(8):845-849. 2018.
6 Khalilieh S, Hodsman P, Xu C, et al. Basic Clin Pharmacol Toxicol. 123(3):294-300. 2018.
7 Papp K, Thaçi D, Reich K, et al. Br J Dermatol. 173(4):930-939. 2015.

Formats Available

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