Anti-Human IL-17A (Ixekizumab) – Fc Muted™

Anti-Human IL-17A (Ixekizumab) – Fc Muted™

Product No.: I-1235

- -
- -
Product No.I-1235
Clone
LY2439821
Target
IL-17A
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
CTLA8, interleukin 17A
Isotype
Human IgG4κ
Applications
ELISA
,
FA
,
FC
,
IF
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Humanized antibody from mouse clone 2321
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,
IF,
FC,
WB
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 Ixekizumab. This product is for research use only.

Ixekizumab has similar binding affinity to and neutralization of human andcynomolgus monkey IL-17A as well as weak binding to rabbit IL-17A. No binding is detected against rat or mouse IL-17A.
Background
IL-17 is a group of proinflammatory cytokines (IL-17A to IL-17F) released by T helper 17 (Th17) cells1. IL-17A is the key effector cytokine of the group 1 and is involved in normal inflammatory and immune responses2. Additionally, increased IL-17A plays an important role in the pathogenesis of ankylosing spondylitis (AS), a chronic autoimmune inflammatory disease that primarily affects the axial skeleton2, and in the progression of psoriatic arthritis1 and plaque psoriasis3, 4.

Ixekizumab was developed as an IL-17A inhibitor for the treatment of AS, psoriasis, and psoriatic arthritis and has been approved for the treatment of some patients with plaque psoriasis3, psoriatic arthritis, AS, and non-radiographic axial spondyloarthritis4. Ixekizumab is a fully humanized monoclonal antibody that binds selectively to IL-17A and inhibits its interaction with the IL-17 receptor, thereby inhibiting the release of proinflammatory cytokines and chemokines2, 3. Ixekizumab disrupts the proinflammatory cascade present in psoriasis4, resulting in decreased expression of cytokines from multiple T cell subsets as well as decreased keratinocyte proliferation and differentiation5.

Antigen Distribution
IL-17A is expressed by Th17 cells, mast cells, and neutrophils.
Ligand/Receptor
Il-17R
NCBI Gene Bank ID
UniProt.org
Research Area
Autoimmune
.
Biosimilars
.
Cancer
.
Immunology
.
Inflammatory Disease

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 Ixekizumab biosimilars are used as calibration standards or reference controls in a pharmacokinetic (PK) bridging ELISA by serving as the analytical standard curve against which the concentration of the drug in serum samples is quantified.

A PK bridging ELISA is designed to measure the concentration of monoclonal antibodies like Ixekizumab in biological matrices (e.g., serum). In such assays, biosimilar material—which has been shown to be analytically and functionally equivalent to the reference (originator) drug—can be used as the standard for the entire assay, enabling measurement of both the biosimilar and the reference product within the same run.

How biosimilars are used as standards and controls:

  • Single Analytical Standard Approach: The biosimilar Ixekizumab is qualified through a method validation process to demonstrate bioanalytical equivalence to the reference (originator) product. If equivalence is established, the biosimilar is selected as the sole calibrator standard for the assay, creating a standard curve from serial dilutions in serum matrix (e.g., 50–12,800 ng/mL).

  • Calibration Curve: Standard curve points are prepared using the biosimilar in serum and run alongside test samples. The serum samples from PK studies (containing either biosimilar or reference drug) are then interpolated against this curve to determine drug concentrations.

  • Quality Control Samples: Additional QC samples are prepared using both biosimilar and reference material at multiple concentrations. These controls are quantified against the biosimilar-based calibration curve to verify equivalency in assay performance and ensure consistent accuracy and precision.

  • Bridging Detection Format: The ELISA kit design uses a capture antibody specific to Ixekizumab on the plate and a detection antibody (often labeled), allowing the detection of any Ixekizumab (biosimilar or reference) present in the sample in a way that does not discriminate between the two, provided their antigenic epitopes are the same.

Significance of this approach:

  • Reduces variability: Using a single, well-characterized biosimilar standard eliminates the need for parallel (separate) assays and minimizes inter-assay variability.
  • Regulatory compliance: This method meets regulatory guidance and industry best practices for the demonstration of PK similarity, as long as equivalence between biosimilar and reference is rigorously validated.
  • Applicability: This practice is common for other monoclonal antibody therapies and their biosimilars in both research and regulated bioanalysis settings.

In summary, research-grade Ixekizumab biosimilars are first validated for equivalency and then serve as the calibration standard in PK bridging ELISAs, enabling the precise quantification of Ixekizumab (biosimilar or reference) in serum samples for pharmacokinetic studies.

The primary in vivo models in which a research-grade anti-IL-17A antibody is administered to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models. In these studies, researchers typically implant mouse-derived tumor cells into immunocompetent mice of the same genetic background and treat them with anti-IL-17A antibodies to evaluate effects on tumor progression and immune cell composition.

Key models and approaches:

  • Syngeneic mouse tumor models (e.g., EO771 mammary carcinoma, MC38 colon carcinoma, B16 melanoma) are the gold standard for this type of study because they preserve the native murine immune microenvironment. In these models, anti-IL-17A antibody is administered after tumor cell inoculation to quantify effects on tumor growth and survival, as well as to assess TIL phenotypes using flow cytometry or immunohistochemistry.

    • For example, in the EO771 model, anti-IL-17A antibody treatment led to significantly lower tumor burden and alterations in TIL populations, such as an increase in CD8^+^ T cells and a decrease in regulatory T cells (Tregs).
  • Manipulation Methods:

    • Most studies use systemic (intraperitoneal) or local (intratumoral) administration of anti-IL-17A antibody to block IL-17A function in vivo.
    • Some studies use genetic or viral tools (such as intratumoral delivery of siRNA against IL-17A) to locally inhibit IL-17A as an alternative, but antibody blockade remains the standard when characterizing TIL changes relevant to antibody therapy.
  • Endpoints:

    • Tumor growth inhibition (measured by tumor volume and/or survival).
    • TIL characterization: Frequencies of cytotoxic T cells (CD8^+^), regulatory T cells (FoxP3^+^), and other immune subsets are quantified within tumor tissues post-treatment.
  • Humanized mouse models (bearing human tumor xenografts and reconstituted with a human immune system) are not commonly used for anti-IL-17A antibody studies aimed at TIL assessment, due to cost and complexity, and species cross-reactivity concerns with mouse anti-IL-17A antibodies. Reports of in vivo anti-IL-17A antibody administration in humanized models with full TIL characterization are scarce in the literature surveyed.

Model typeIL-17A BlockadeTumor cell linesTIL analysisReference
Mouse syngeneicAntibody/siRNAEO771, MC38, B16Yes (CD8, Tregs etc.)
Humanized mouseNot standardHuman xenograftRare/No

In summary:
Syngeneic mouse tumor models—such as EO771, MC38, and B16—in immunocompetent mice are the primary preclinical models where anti-IL-17A antibody is administered in vivo to analyze tumor growth inhibition and TIL profiles, with robust protocols for immune cell phenotyping directly from tumor tissue. Humanized or xenograft models are not widely used for this specific application.

Researchers use the Ixekizumab biosimilar—a monoclonal antibody targeting IL-17A—in combination with other checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) to investigate synergistic immune effects in complex immune-oncology models, especially in preclinical or translational studies. This approach enables the study of how simultaneous blockade of adaptive immune checkpoints and inflammatory cytokines can impact tumor immunity or modulate immune-related toxicity.

Essential context and methodological details:

  • Ixekizumab biosimilar function: Neutralizes human IL-17A, an inflammatory cytokine produced by Th17 and other immune cells, thereby modulating immune and inflammatory responses. IL-17A plays complex roles in immune regulation, including both promoting anti-microbial defense and involvement in chronic inflammation or autoimmune disease.
  • Checkpoint inhibitors: Agents such as anti-CTLA-4 and anti-LAG-3 block inhibitory pathways that "brake" T cell activity, thereby enhancing antitumor immune responses. These drugs boost distinct aspects of T cell activation—CTLA-4 mainly affects T cell priming, while LAG-3 regulates effector and exhausted T cells in peripheral tissues or tumors.
  • Rationale for combination:
    • Targeting multiple, non-redundant pathways (e.g., inflammatory cytokines plus adaptive checkpoints) may overcome resistance seen with single-agent therapies and induce more potent or durable antitumor responses.
    • In preclinical tumor models (often syngeneic mouse models engineered to express human targets), the biosimilar is administered alongside anti-checkpoint antibodies to parse out additive or synergistic effects on immune cell populations, cytokine environments, and tumor regression.

Experimental Design:

  • Researchers typically administer the Ixekizumab biosimilar with anti-CTLA-4, anti-LAG-3, or placebo in immune-competent mouse models that recapitulate aspects of human cancer and immunity.
  • They monitor immune subset composition (CD4, CD8, Tregs), cytokine profiles, and tumor growth.
  • Outcomes include modulation of T helper cells (notably Th17 and associated IL-17A production), cytotoxic T cell activation, and regulatory cell depletion or inactivation. For instance, IL-17A blockade may decrease tumor-promoting inflammation or limit off-tumor autoimmunity, while checkpoint inhibition broadens T cell effector function and memory formation.
  • The combinations are also evaluated for adverse effects, since combining immunomodulatory therapies risks increasing immune-mediated toxicity.

Supporting evidence:

  • Studies have shown that different combinations of checkpoint inhibitors (e.g., anti-PD-1/CTLA-4 vs anti-PD-1/LAG-3) activate distinct T cell subsets and alter tumor microenvironment dynamics in unique ways. Adding IL-17A blockade can further shift immune balance, potentially improving efficacy or safety profiles.
  • Ixekizumab biosimilars are designed for research—not for direct clinical use—allowing mechanistic dissection of these pathways without therapeutic intent.

Key Insights:

  • Combining Ixekizumab biosimilar with other checkpoint inhibitors in research provides a powerful tool to study immune synergy and tumor microenvironment modulation.
  • These models can reveal new biomarkers for response, mechanisms of resistance, or optimal therapeutic combinations for translation to clinical trials.

No direct clinical data exist yet for these combinations in patients; findings are primarily at the preclinical (animal or cell-based) research level.

A Ixekizumab biosimilar can be used as either the capture or detection reagent in a bridging ADA ELISA to specifically detect anti-drug antibodies (ADAs) in patient samples directed against the therapeutic drug ixekizumab. This is possible because a biosimilar with identical or matched variable regions to ixekizumab will bind the same patient-derived ADAs as the therapeutic drug itself, enabling accurate monitoring of immunogenic responses.

How a Bridging ADA ELISA Using an Ixekizumab Biosimilar Works:

  • Coating (Capture): The ELISA plate is coated with the ixekizumab biosimilar. This immobilized antibody will specifically bind any circulating ADA in the clinical sample that recognizes ixekizumab.
  • Sample Incubation: The patient’s serum is then incubated on the plate. If the patient has developed ADAs against ixekizumab, those antibodies will bind to the immobilized biosimilar.
  • Detection: A labeled (e.g., HRP-conjugated or biotinylated) form of the ixekizumab biosimilar is then added. This labeled antibody will bind to the ADA that has already formed a complex with the plate-bound biosimilar, forming a “bridge” between the two drug molecules via the bivalent ADA, which is specific to ixekizumab.
  • Signal Development: Substrate for the label (e.g., TMB substrate for HRP) is added, and a colorimetric signal is produced, proportional to the amount of ADA present in the serum.

Importance of Using a Biosimilar as Reagent:

  • Biosimilars developed for research use have the same variable regions as the therapeutic drug but are non-therapeutic and can be produced at scale specifically for assay purposes.
  • Using a biosimilar instead of the therapeutic-grade drug allows for high consistency, avoids using clinical material, and ensures the assay targets patient antibodies generated against the specific variable region of ixekizumab.

Key Features of the Method:

  • The method quantifies anti-drug antibodies that might neutralize ixekizumab or alter its efficacy.
  • A bridging format is highly specific because a “bridge” can only form if patient serum contains ADAs capable of binding two ixekizumab (or biosimilar) molecules simultaneously.
  • The use of a biosimilar reagent reduces cost and logistical challenges as compared to using the original therapeutic material.

This bridging ELISA format for ADA detection is established for other monoclonal antibody drugs and is directly applicable to ixekizumab and its biosimilars, provided the biosimilar retains matched antigenicity.

References & Citations

1 Aboobacker S, Kurn H, Al Aboud AM. Secukinumab. [Updated 2023 Jun 20]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK537091/
2 Blair HA. Drugs. 79(4):433-443. 2019.
3 Markham A. Drugs. 76(8):901-905. 2016.
4 Preuss CV, Quick J. Ixekizumab. [Updated 2023 Jul 4]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2023 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK431088/
5 Krueger JG, Fretzin S, Suárez-Fariñas M, et al. J Allergy Clin Immunol. 130(1):145-54.e9. 2012.
6 Genovese MC, Van den Bosch F, Roberson SA, et al. Arthritis Rheum. 62(4):929-939. 2010.
7 Wang CQF, Suárez-Fariñas M, Nograles KE, et al. J Invest Dermatol. 134(12):2990-2993. 214.
8 Beerli RR, Bauer M, Fritzer A, et al. MAbs. 6(6):1608-1620. 2014.
9 Clarke DO, Hilbish KG, Waters DG, et al. Reprod Toxicol. 58:160-173. 2015.
Indirect Elisa Protocol
FA
Flow Cytometry
IF
General Western Blot Protocol

Certificate of Analysis

Formats Available

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