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 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.
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 pharmacokinetic (PK) bridging ELISAs to quantitatively measure drug concentration in serum samples by serving as the assay calibrator for both originator and biosimilar drugs.
In PK bridging ELISA development for biosimilars:
The biosimilar is established as the analytical standard (calibrator). This means that serial dilutions of the biosimilar are used to create a standard curve spanning the relevant concentration range in serum (e.g., 50–12,800 ng/mL), which enables precise quantification of drug levels in patient samples.
All samples—containing either the reference (originator) drug or the biosimilar—are quantified against the biosimilar-based standard curve using validated ligand-binding systems with antibodies capable of recognizing both the originator and biosimilar forms equally.
This approach reduces assay variability that could occur if separate calibration standards were used for each drug, and it ensures a direct comparability of PK data between reference and biosimilar products.
During method validation:
Comparable accuracy and precision are evaluated by measuring test samples containing known concentrations of both biosimilar and reference drugs against the biosimilar standard curve. Regulatory and industry standards require demonstration of “bioanalytical equivalence” within predefined margins (typically a 90% confidence interval within [0.8, 1.25]).
Controls and quality control samples made from both biosimilar and reference compounds are analyzed to ensure the assay’s reliability and comparability.
For ELISA format:
Plates are coated with capture antibodies that bind Ixekizumab, and detection relies on secondary antibodies (e.g., biotinylated polyclonal anti-Ixekizumab) plus enzymatic signal development.
The colorimetric signal’s intensity following substrate addition is proportional to the drug concentration, as determined via the biosimilar calibration curve.
Summary of use:
The biosimilar standard enables consensus, robust, and comparable PK data generation for bioequivalence studies.
It ensures regulatory expectations for accuracy, matrix comparability, and bridging between reference and biosimilar products are met.
In essence, the research-grade Ixekizumab biosimilar functions as the quantitative benchmark and assurance of assay equivalence for measuring serum drug concentrations in PK bridging ELISAs.
The primary in vivo models for administering a research-grade anti-IL-17A antibody to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models, sometimes using transgenic mice to enable recognition of human antibodies, and less frequently, humanized models for cross-reactivity or translational research.
Key model types:
Syngeneic mouse tumor models:
These involve implantation of murine tumor cell lines (such as B16 melanoma or MC38 colon carcinoma) into immunocompetent mice, allowing the study of host immune response, including TILs.
Anti-IL-17A antibodies with cross-reactivity to murine IL-17A, or chimeric/human/mouse antibody variants, can be used in these models.
These models are preferred because they retain a fully functional immune system and are widely used for assessing immunotherapies and profiling TILs.
Example from literature: Intratumoral blockade of IL-17A in B16 and MC38 syngeneic models led to tumor growth inhibition and changes in TIL populations, including increased activation of cytotoxic T lymphocytes.
Transgenic syngeneic models:
Mice engineered to express humanized targets (such as human IL-17A or its receptor) permit the use of research-grade human antibodies for in vivo testing.
These allow for testing of antibodies that would not otherwise bind to mouse IL-17A, supporting translational studies for clinical candidates.
Humanized mouse models (less common for anti-IL-17A studies):
These models have functional human immune systems, generated by engrafting human hematopoietic stem cells into immunodeficient mice.
They are used mainly when the antibody is strictly human-specific and cross-reactivity cannot be engineered, though they are less frequently cited in studies specifically using anti-IL-17A to study TILs and tumor growth inhibition.
Most studies focus on syngeneic or transgenic syngeneic models due to practical advantages and established protocols.
Tumor types frequently studied in these models:
Melanoma (B16-F10 murine melanoma, human A375 melanoma in appropriate hosts).
Colon carcinoma (MC38 murine colon carcinoma).
Others: Neuroblastoma, head and neck cancer models (as reported for IL-17 studies, though not always with anti-IL-17A antibody administration).
TIL Characterization:
Studies typically use flow cytometry, immunohistochemistry, or RNA profiling to analyze changes in TIL subsets (e.g., CD8+ cytotoxic T cells, neutrophils, regulatory T cells) following anti-IL-17A treatment.
Summary Table: Syngeneic vs. Humanized Models for anti-IL-17A Tumor Studies
Model Type
Main Features
Antibody Requirements
Immune System Status
Key TIL Analysis
Syngeneic
Mouse tumor lines in immunocompetent mice
Mouse-reactive/chimeric/human
Fully murine/functional
Flow, IHC, gene profiling
Transgenic
Humanized targets (IL-17A/hIL-17R) in mice
Human or human/mouse hybrids
Functional, may express human targets
Same as above
Humanized
Mice engrafted w/human stem cells, human tumors
Human-specific
Human hematopoietic compartment
Human TILs, limited accessibility
In conclusion, syngeneic (immunocompetent mouse) tumor models—often with transgenic modifications for human antibody compatibility—are the primary preclinical platform for evaluating anti-IL-17A antibody-mediated tumor growth inhibition and TIL characterization. Humanized mice are used less frequently but remain an option for translational research.
Researchers studying synergistic effects in immune-oncology models combine the Ixekizumab biosimilar with other immune checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—to interrogate interactions between IL-17A blockade and inhibition of T cell checkpoints central to tumor immunity.
Key experimental approaches:
Model Systems: Most studies employ mouse tumor models (e.g., melanoma), as well as humanized in vitro immune contexts.
Combination Strategy: The Ixekizumab biosimilar neutralizes human IL-17A, blocking cytokine-driven proinflammatory and immune-modulating effects. Checkpoint inhibitor biosimilars (e.g., anti-CTLA-4, anti-LAG-3) block attenuation signals on T cells, enhancing cytotoxic T lymphocyte activity against tumors.
Rationale for Combination: IL-17A is implicated in shaping the tumor microenvironment, impacting immune cell infiltration and function. Combining IL-17A neutralization with checkpoint inhibition seeks to both diminish immunosuppressive inflammation and maximally activate antitumor T cells.
Distinct Pathways: Anti-CTLA-4 acts in lymph nodes, promoting early T cell activation, while anti-LAG-3/PD-1 mainly act within the tumor, sustaining cytotoxic activity.
Cellular Crosstalk: For example, anti-PD-1/LAG-3 combinations rely on CD4+ T cells for tumor control, which may also be influenced by shifts in inflammation, such as those mediated by IL-17A blockade.
Immune Modulation by IL-17A: IL-17A blockade by Ixekizumab can reduce inflammatory cell recruitment and myeloid-driven immunosuppression, possibly creating a more permissive microenvironment for checkpoint therapies to activate effector T cells.
Experimental Controls and Validation:
Researchers utilize biosimilars (non-therapeutic, research-grade analogues) in parallel arms (single therapy vs. combinations) to distinguish combinatorial vs. additive effects.
Additional endpoints include pharmacokinetics and immunogenicity testing to ensure absence of cross-reactivity or unexpected immune activation.
Limitations and Ongoing Work:
Many findings derive from preclinical models; translation to human cancers is ongoing.
Risk of increased toxicity is evaluated due to broad immune activation in combination arms.
In summary, the Ixekizumab biosimilar is used in combination with checkpoint inhibitors to investigate how targeting IL-17A-driven inflammation and relieving T cell inhibition can jointly enhance antitumor immunity, with distinct cellular and molecular mechanisms under active investigation using complex preclinical immune-oncology models.
In immunogenicity testing using a bridging ADA ELISA, a biosimilar of Ixekizumab can be employed as either the capture or detection reagent to sensitively monitor a patient's immune response against the therapeutic drug, specifically the formation of anti-drug antibodies (ADAs).
Key steps and rationale:
Bridging format: The ADA bridging ELISA detects bivalent ADAs in patient serum by their ability to "bridge" between two molecules of the drug—one immobilized as the capture reagent and the other conjugated (often labeled with HRP or biotin) as the detection reagent.
Use of biosimilar: A biosimilar Ixekizumab (not for therapeutic use, but with identical variable regions to the original drug) is ideal for research protocols, as it will bind any ADAs formed against the therapeutic Ixekizumab in treated patients without competing with the original clinical supply.
Assay protocol:
Step 1: The plate is coated with the Ixekizumab biosimilar (capture reagent).
Step 2: Patient serum is added; if ADAs are present, their Fab regions bind to the immobilized biosimilar.
Step 3: An enzyme- or dye-labeled Ixekizumab biosimilar (detection reagent) is added. The other Fab arm of an ADA binds to this labeled biosimilar, forming a "bridge".
Step 4: The reaction is developed (e.g., via HRP and TMB substrate), and the signal intensity correlates with ADA concentration.
Advantages:
The bridging format is highly sensitive and suitable for high-throughput screening.
Using a biosimilar for both capture and detection ensures that the assay detects ADAs against all epitopes present on the original drug without cross-reactivity or depletion of clinical drug supply.
This approach is a standard for ADA measurement in the context of therapeutic monoclonal antibodies.
Additional context:
The biosimilar must be validated to ensure it recapitulates the binding profile of the therapeutic drug, especially since subtle structural differences could affect ADA recognition.
Common challenges include matrix interference (from patient serum components) and interference from free drug in circulation, which can mask ADA detection.
The immunogenicity detected in this way is clinically relevant because ADAs can neutralize drug efficacy or provoke hypersensitivity reactions.
In summary, an Ixekizumab biosimilar used in both capture and detection roles in ADA bridging ELISA enables monitoring of patient immune responses against the drug by quantifying ADAs that can bind two molecules of Ixekizumab simultaneously, thus providing sensitive and specific immunogenicity data.
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.