Anti-Human IL-12/23 (Ustekinumab) [Clone CNTO-1275] — Fc Muted™

Anti-Human IL-12/23 (Ustekinumab) [Clone CNTO-1275] — Fc Muted™

Product No.: I-1255

- -
- -
Product No.I-1255
Clone
CNTO-1275
Target
IL-12/IL-23 p40
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
IL-12: CLMF p40, IL-12 subunit p40, NKSF2
IL-23: IL-23-A, IL-23p19
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
N

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
p40 subunit of IL-12/23
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 ?
ELISA,
FA,
N
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 sequenceas the therapeutic antibody Ustekinumab. CNTO-1275 (Ustekinumab) activity is directed againstthe p40 subunit of IL-12 and IL-23.
Background
IL-12 and IL-23 play a role in the differentiation and proliferation of type 1 T-helper cells (Th1)1. IL-12 stimulates IFN-γ and TNF-α production via Th1 differentiation, whereas IL-23 causes activation of IL-17-producing T cells2. IL-12 and IL-23 are members of the IL-12 cytokine family3. Members of the IL-12 family form soluble heterodimers consisting of α and β subunits. IL-12 and IL-23 both have a p40 subunit, and either p40 can bind to the IL-12 β1 receptor1,2,4. IL-23 consists of IL-12p40 and IL-23p193. IL-12 consists of IL-12p40 and IL-12p35.

IL-23 is associated with various autoimmune inflammatory diseases and is particularly highly expressed in psoriasis skin lesions1,2. Furthermore, IL-23 is suspected to play a role in tumorigenesis3. Anti-IL12/23 p40 antibodies antagonize key pathways in inflammatory autoimmune diseases, such as arthritis and colitis1,2.

Ustekinumab was developed from transgenic mice as an antibody against the p40 subunits of IL- 12/231,2. Ustekinumab binds with high affinity and specificity to p40, preventing interaction with the IL-12 β1 receptor found on natural killer cells or T cells and blocking downstream signaling, differentiation, and cytokine production1,2,4. Additionally, ustekinumab inhibits up-regulation of cutaneous lymphocyte antigen, IL-2, IL-2Rα, and IL-12R as well as secretion of IFN-γ, TNF-α, and IL-17A.

Ustekinumab is used in clinical settings to treat plaque psoriasis, Crohn’s disease, and ulcerative colitis.

Antigen Distribution
IL-12 is produced by dendritic cells, macrophages, neutrophils, and human B-lymphoblastoid cells. IL-23 is mainly secreted by activated dendritic cells, macrophages, or monocytes. Both are produced by activated antigen-presenting cells.
Ligand/Receptor
IL-12 β1 receptor
NCBI Gene Bank ID
IL-12: M65272
IL-23: AF301620
UniProt.org
IL-12: P29460
IL-23: Q9NPF7
Research Area
Biosimilars
.
Inflammatory Disease
.
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 Ustekinumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA by serving as the known reference material to generate the standard curve, enabling accurate quantification of Ustekinumab concentrations in serum samples.

Essential context and supporting details:

  • Calibration standards in a PK ELISA are solutions of known concentration, typically prepared by serially diluting a reference-grade Ustekinumab biosimilar (or the original reference product) in a biological matrix similar to study samples (like pooled human serum).

  • These standards are run on each ELISA plate to generate a standard curve (OD versus concentration), against which unknown serum sample concentrations are interpolated.

  • According to the KRIBIOLISA™ Ustekinumab ELISA kit, the calibrators/standards used for calibration are typically validated and calibrated against a commercially sourced reference Ustekinumab product (such as STELARA™), ensuring equivalence between the measured biosimilar and the reference during quantification.

  • Biosimilar-grade material must have demonstrated high similarity in terms of binding and immunoreactivity to the reference product to be suitable as a standard or reference control.

  • In a PK bridging ELISA assay, serum samples are processed alongside the standards. The sandwich ELISA format uses a capture antibody specific to Ustekinumab (including biosimilars) and a detection antibody targeting the Fc region, ensuring that the standard curve derived from the biosimilar calibrator accurately reflects the characteristics of the Ustekinumab in patient samples.

  • Results calculation: The unknown sample concentrations are interpolated from the standard curve, which has been generated using the biosimilar-based calibration standards.

  • Controls: Reference controls may include samples with low, medium, and high concentrations of the biosimilar to monitor accuracy and precision across the working range.

Key points for use in PK bridging ELISA:

  • Use of a research-grade Ustekinumab biosimilar as a calibration standard ensures consistency, traceability, and comparability of results across studies and laboratories.
  • Kits and protocols often specify that calibration standards should match or be cross-validated against the reference product to confirm assay validity, especially given regulatory requirements for biosimilar comparability studies.
  • PK similarity studies may use both biosimilar and reference Ustekinumab (EU or US-sourced) as standards/controls to ensure assay detects no meaningful differences, further supporting biosimilarity.

In summary, biosimilar Ustekinumab is calibrated and validated against the reference product and used to generate standard curves and QC controls in PK ELISA, facilitating accurate drug quantitation in bridging and comparability studies.

The primary in vivo models where a research-grade anti-IL-12/IL-23 p40 antibody is administered to investigate tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models. Humanized models are less common for this specific application due to the species specificity of the p40 subunit recognized by such antibodies.

Key context and experimental details:

  • Syngeneic mouse models use immunocompetent mice implanted with mouse-derived tumors (e.g., B16 melanoma, MC38 colon carcinoma, MB49 bladder carcinoma), allowing for full immune interactions including TIL responses.
  • Anti-IL-12/IL-23 p40 antibodies such as clone C17.8 (or commercial equivalents from eBioscience, Bio X Cell, etc.) specifically block endogenous mouse IL-12 and IL-23, affecting both Th1 and Th17/Tc17 responses in the tumor microenvironment.
  • Studies using these models typically administer the anti-p40 antibody systemically (often IP or IV) during tumor establishment or growth, monitoring effects on tumor size and characterizing TIL populations (CD8+, CD4+, Tregs, myeloid cells) by flow cytometry at endpoint.

Supporting examples:

  • Application of anti-IL-12/IL-23 p40 neutralizing antibody in syngeneic mouse models, such as DMBA/TPA-induced skin tumors or orthotopic gliomas, has helped dissect the role of these cytokines in tumor immunity, modulating both tumor progression and TIL composition.
  • Syngeneic bladder and sarcoma mouse models have been used to show that blocking IL-23 (using p40 antibodies) enhances the antitumor efficacy of therapies that boost IL-12 or T cell responses.

About humanized models:

  • Use of anti-IL-12/IL-23 p40 antibodies in humanized mouse models (mice engrafted with human immune cells and human tumors) is rare for studies targeting endogenous cytokine signaling, because standard research antibodies for mouse p40 do not cross-react with human p40, and few studies have monitored both tumor control and TILs in this context.
  • Humanized models would require a human-specific anti-IL-12/IL-23 p40 antibody and appropriate humanized tumor/immune cell combinations, which is more typical in translational exploratory research or drug development and less in the standard academic setting.

In summary:

  • Syngeneic mouse tumor models (using murine anti-p40 antibodies) are the canonical approach for these studies.
  • These models facilitate full immune analyses, including effects on tumor-infiltrating lymphocyte populations under IL-12/23 blockade.
  • Humanized models have theoretical utility but far fewer empirical studies using anti-IL-12/IL-23 p40 blockade to dissect TIL phenotypes in vivo.

References for models and methodology:

Researchers investigating the synergistic effects of ustekinumab biosimilars in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) in immune-oncology models typically use preclinical studies and early-phase clinical trials to evaluate potential interactions and therapeutic benefits.

Context and Methodology:

  • Checkpoint Inhibitors and Mechanistic Rationale: The foundation of such combination studies lies in the distinct mechanisms of various checkpoint inhibitors. For example, anti-CTLA-4 agents mainly activate T cells in lymph nodes, while anti-PD-1/PD-L1 agents work at the tumor site to overcome local immunosuppression.
  • Ustekinumab’s Role: Ustekinumab (and its biosimilars) targets the IL-12 and IL-23 cytokine pathways, modulating the immune response in ways that complement checkpoint blockade mechanisms. By altering the tumor microenvironment and reducing immunosuppressive signals, ustekinumab may potentiate T-cell-mediated anti-tumor effects when used with checkpoint inhibitors.

Practical Study Designs:

  • Preclinical Models: Researchers use murine models bearing syngeneic or patient-derived tumors. They administer biosimilar ustekinumab with checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, or anti-PD-1/PD-L1 biosimilars) in various combinations and dosing regimens to map out synergistic effects.
  • Outcome Measures: They monitor tumor growth, survival, changes in tumor-infiltrating lymphocytes (especially CD8+ T cells and regulatory T cells), cytokine profiles, and markers of immune activation or exhaustion.
  • Mechanism Elucidation: These studies often employ flow cytometry, RNA-sequencing of tumor tissue, immunohistochemistry, and cytokine assays to detail the molecular and cellular mechanisms underlying any observed synergy.

Current Knowledge and Limitations:

  • There is extensive support for combining checkpoint inhibitors (e.g., anti-CTLA-4 and anti-PD-1) in both preclinical and clinical settings, where such combinations have increased efficacy but also higher toxicity.
  • For ustekinumab biosimilars specifically, most available research focuses on biosimilar equivalency, pharmacokinetics, and approvals for autoimmune diseases rather than direct immune-oncology combinations. Translational and preclinical oncology studies with ustekinumab (originator or biosimilar) in conjunction with checkpoint inhibitors are an active area of research, but large-scale, published results on these specific combinations are currently limited.

Summary Table: Mechanistic Rationale for Combination

Agent/ClassMechanismRationale in Combination Studies
Ustekinumab biosimilarIL-12/23 blockade, reduces immunosuppressionMay increase T-cell activation indirectly
Anti-CTLA-4 biosimilarReleases T-cell priming/proliferation brakesAmplifies T-cell pool for anti-tumor action
Anti-LAG-3 biosimilarReverses T-cell exhaustion in tumorSustains and strengthens T-cell activity

Conclusion:
Researchers use ustekinumab biosimilars in combination with checkpoint inhibitors, exploiting their complementary mechanisms to enhance anti-tumor immunity in complex preclinical models, often using murine systems, deep immunophenotyping, and functional assays to establish synergy and inform future clinical trials. Direct clinical or real-world data with ustekinumab biosimilars in these specific combinations for oncology are still emerging, and most evidence is derived from analogous combinations or mechanistic studies.

In a bridging ADA ELISA to monitor a patient’s immune response against ustekinumab, a biosimilar version of ustekinumab can be used as either the capture or detection reagent due to its structural similarity to the reference drug. The typical process is as follows:

  • Capture phase: Microtiter plate wells are first coated with the ustekinumab biosimilar, which acts as the capture reagent to bind any anti-drug antibodies (ADAs) present in the patient's serum that recognize ustekinumab.

  • Detection phase: After incubation and washing, a labeled form of ustekinumab (for example, horseradish peroxidase [HRP]-conjugated or biotinylated biosimilar ustekinumab) is added to the wells. This labeled drug acts as the detection reagent and will also bind to the captured patient ADAs due to its identical epitope presentation, thus forming a "bridge" via bivalent binding (one arm of the ADA binds to plate-coated biosimilar, and the other arm to the labeled detection biosimilar).

This sandwich or "bridging" format is particularly suitable for the detection of bivalent antibodies – especially IgG-class ADAs – which must simultaneously bind to two identical antigen molecules (here, the biosimilar ustekinumab).

Key details for this format:

  • The biosimilar ustekinumab is functionally interchangeable with the reference product in the assay, provided it is shown to be analytically highly similar, especially in relevant epitopes recognized by ADAs.
  • Using the biosimilar as both the capture and detection reagent is acceptable so long as there is no clinically meaningful difference in immunogenic epitopes between biosimilar and reference.
  • The assay can detect the presence and sometimes the specificity (e.g., domain mapping) of ADAs produced in response to therapy, which may impact drug efficacy and safety.

Summary Table: Use of Ustekinumab Biosimilar in Bridging ADA ELISA

StepRole of Biosimilar UstekinumabPurpose
CapturePlate coating (unlabeled biosimilar)Binds ADAs from patient serum
DetectionLabeled biosimilar (e.g., HRP- or biotin-labeled)Detects captured ADAs via signal amplification

In this context, the biosimilar's use is justified by stringent analytical and functional comparability requirements, ensuring results reflect the true immune response to the therapeutic drug rather than assay artifacts. This approach provides sensitive and specific detection of patient anti-drug antibodies in pharmacovigilance and clinical monitoring settings.

References & Citations

1. Reich K, Yasothan U, Kirkpatrick P. Nat Rev Drug Discov. 8(5):355-356. 2009.
2. Cingoz O. MAbs. 1(3):216-221. 2009.
3. Floss DM, Moll JM, Scheller J. Cells. 9(10):2184. 2020.
4. Kauffman CL, Aria N, Toichi E, et al. J Invest Dermatol. 123(6):1037-1044. 2004.
5. Toichi E, Torres G, McCormick TS, et al. J Immunol. 177(7):4917-4926. 2006.
6. Reddy M, Davis C, Wong J, et al. Cell Immunol. 247(1):1-11. 2007.
7. Krueger GG, Langley RG, Leonardi C, et al. N Engl J Med. 356(6):580-592. 2007.
8. Feagan BG, Sandborn WJ, Gasink C, et al. N Engl J Med. 375(20):1946-1960. 2016.
9. Brok HP, van Meurs M, Blezer E, et al. J Immunol. 169(11):6554-6563. 2002.
Indirect Elisa Protocol
FA
N

Certificate of Analysis

Formats Available

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