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 Bimekizumab. UCB4940 (Bimekizumab) is a dual inhibitor of IL-17A
and IL-17F interleukins.
Background
Interleukin 17 (IL-17) is a pro-inflammatory cytokine crucial to host defense, tissue repair,
pathogenesis of inflammatory disease, and progression of cancer1. IL-17 signaling is also critical
for protection against fungal and bacterial infection2. There are six pro-inflammatory cytokines
(IL-17A-F) produced by Th17 cells3. IL-17A and IL-17F share ~50% structural homology4 as
well as overlapping biological functions in chronic inflammation and adaptive immune defense
from bacterial and fungal infection5. Additionally, they are coexpressed in immune-
inflammatory diseases5. IL-17A and IL-17F drive pathogenesis in psoriasis5 and are both found
in lesional skin and inflamed synovium from patients with psoriatic arthritis4. Since dual
neutralization of IL-17A and IL-17F results in lower expression of inflammation-linked genes
and cytokines 4 and because targeting the IL-17 pathway has clinical efficacy5, an antibody
therapeutic designed to potently and selectively neutralize both IL-17A and IL-17F is desirable.
Bimekizumab is a humanized monoclonoal antibody designed to bind at a similar site on both
IL-17A and IL-17F, allowing for dual inhibition5. Bimekizumab has been approved for use in
the treatment of plaque psoriasis6, psoriatic arthritis7, and axial spondyloarthritis8.
Antigen Distribution
Th17 cells, which are CD4 + T-helper cells, are the principal source of IL-
17. Tc17 cells, which are CD8 + cells, also make IL-17. Additionally, IL-17 is produced by a
number of innate immune subsets (γδ-T cells, some natural killer T (NKT) cells, TCRβ+
‘natural’ Th17 cells, and Type 3 “innate lymphoid cells” (ILC3)) and possibly by myeloid cells
in small amounts.
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 Bimekizumab biosimilars are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to establish a standard curve, enabling accurate quantification of Bimekizumab concentrations in serum samples.
Context and Process:
In a PK bridging ELISA, a calibration standard is critical for quantifying the antibody drug (here, Bimekizumab) in biological samples by generating a standard curve from which sample concentrations are interpolated.
Research-grade Bimekizumab biosimilars serve as these standards, as they are structurally and functionally analogous to the reference product, permitting reliable quantification and comparability.
Typically, multiple standard concentrations of the biosimilar are spiked into blank serum, covering the assay's dynamic range (e.g., 31.25–2000 ng/ml as per typical kits). These points are used to plot a standard curve.
Key Steps:
Prepare serial dilutions of the research-grade Bimekizumab biosimilar in a blank matrix (often pooled human serum) to establish the standard curve.
Include quality control (QC) samples at low, medium, and high concentrations, prepared independently using the biosimilar or reference standard.
Analyze study samples in parallel with the standards to allow accurate interpolation of unknown concentrations.
Role as Reference Control:
These biosimilars are also sometimes used as reference controls to verify assay performance and ensure comparability across different runs, batches, or laboratories.
Regulatory and industry consensus recommends that, wherever possible, a single PK assay should be used for both the biosimilar and reference product quantification, with one analytical standard, often the biosimilar itself, to minimize inter-assay variability.
Benefits:
Using a single, well-characterized research-grade biosimilar as a calibration and control standard improves the assay's robustness, comparability, and regulatory acceptability.
This approach aligns with regulatory best practices for biosimilar PK method validation, emphasizing the need for bioanalytical comparability and minimized variability.
Relevant Example:
Commercially available ELISA kits for Bimekizumab provide lyophilized biosimilar standards intended for serum quantitation, with precise concentrations and stability claims.
Companies such as Bio-Rad offer biosimilar monoclonal antibodies for use as reference standards in bridging ELISAs, further supporting this application in research and PK studies.
Summary Table:
Use of Bimekizumab Biosimilar in PK ELISA
Description
Calibration Standard
Serially diluted to generate the standard curve
Reference Control
Run alongside samples/QCs to verify assay performance
Bioanalytical Comparability
Validated to ensure similar measurement as the reference
Regulatory Compliance
Aligns with guidance on biosimilar PK assay development
If you require application-specific protocols (e.g., detailed dilution strategies, choice of QC levels, or bridging ELISA design), manufacturers' kit inserts and regulatory bioanalytical guidelines should be consulted for the exact requirements.
The primary preclinical models where research-grade anti-IL-17 antibodies are administered in vivo to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, to a lesser extent, humanized mouse models.
Syngeneic Mouse Models:
These models involve implanting murine tumor cells into immunocompetent mice, preserving the native mouse immune system, thereby enabling evaluation of immune responses—including TIL expansion, recruitment, and function—after anti-IL-17 antibody treatment.
Studies using syngeneic models demonstrate that anti-IL-17A antibody administration can inhibit tumor growth and modulate the quantity and phenotype of TILs, such as Th17 cells and cytotoxic lymphocytes, within the tumor microenvironment.
Example: In the AOM-DSS-induced colorectal cancer (CAC) syngeneic mouse model, neutralization of IL-17A inhibited tumor development and altered TIL profiles.
Additional example: In MT/ret-driven melanoma mouse models (BRAF-WT), administration of an IL-17A-neutralizing antibody affected tumor growth kinetics and immune infiltration of lymphocytes.
These models are widely used for mechanistic studies because the intact murine immune system allows detailed characterization of TILs through flow cytometry, immunohistochemistry, and transcriptomic profiling.
Humanized Mouse Models:
In these models, human immune cells (including TILs) are engrafted into immunodeficient mice with human tumor xenografts, partially recreating human immune–tumor interactions.
Humanized mice allow the study of human-specific TIL responses to anti-IL-17 therapy, though they are less frequently used due to greater complexity and cost.
This model is used to support preclinical analysis of immunotherapies targeting IL-17 where human TIL characterization is essential.
Genetically engineered mouse models (e.g., CPC-APC for colon cancer) are also used for studying effects of IL-17 antibody on tumor development and TIL composition.
Summary of Approach: Typical experiments administer a research-grade anti-IL-17A antibody to tumor-bearing mice, followed by measurement of:
Tumor growth inhibition compared with controls.
Infiltrating lymphocyte populations (flow cytometry, single-cell RNA-seq, IHC) to assess changes in TILs after treatment.
In summary, the syngeneic mouse model is the standard for in vivo anti-IL-17 antibody studies focusing on TIL analysis, with humanized mouse models providing complementary insights into human TIL biology.
Researchers primarily use bimekizumab, a dual IL-17A and IL-17F inhibitor, in immune-oncology studies to modulate the tumor microenvironment and inflammation, while checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars are used to relieve immune suppression and enhance antitumor T cell responses. To study synergistic effects in complex immune-oncology models, bimekizumab biosimilars may be combined with these checkpoint inhibitors to test if dual cytokine blockade alongside simultaneous checkpoint release can amplify efficacy beyond either agent alone.
Essential context and mechanisms:
Bimekizumab targets and neutralizes both IL-17A and IL-17F cytokines, which contribute to chronic inflammation and play roles in autoimmune diseases and certain tumor microenvironments. By dampening IL-17-driven inflammation, it modulates immune cell recruitment and cytokine/chemokine release in the tissue environment.
Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) are monoclonal antibodies that block immune checkpoint proteins, which normally act as brakes on the immune response. In cancer, these drugs "release the brakes" on T cells, improving antitumor immune activity.
Study design and synergy investigation:
In preclinical or translational models, researchers administer a bimekizumab biosimilar with a checkpoint inhibitor biosimilar (such as anti-CTLA-4 or anti-LAG-3) to tumor-bearing mice or complex ex vivo models. These combinations allow study of whether modulating the inflammatory milieu (via IL-17 blockade) can enhance or alter the effects of checkpoint blockade.
For example, mouse melanoma models were used to clarify differences in anti-tumor mechanisms between combined checkpoint regimens (e.g., anti-PD-1/CTLA-4 vs. anti-PD-1/LAG-3). These models can be adapted to test combinations with cytokine inhibitors like bimekizumab.
Synergy is evaluated by measuring outcomes such as tumor regression, survival, and the activation or composition of immune cell populations—especially CD4+ helper T cells, CD8+ cytotoxic T cells, and regulatory T cells (Tregs).
By integrating IL-17 blockade with checkpoint inhibition, researchers can dissect whether reducing chronic, pro-tumor inflammation unleashes a more robust or qualitatively different T cell response when checkpoint blockades are also in place.
Scientific rationale and clinical relevance:
Dual-cytokine blockade (IL-17A/F) may have more profound effects on chronic inflammation and immune activation than targeting a single cytokine, and could reduce tumor-promoting inflammation or myeloid-derived suppressor cells.
Checkpoint inhibitors differ in their requirements for certain immune cell types; for instance, anti-PD-1/LAG-3 efficacy relies on CD4+ T cells, while anti-PD-1/CTLA-4 does not. Intersecting these mechanisms with IL-17 pathway modulation via bimekizumab can provide novel insights.
Summary Table: Bimekizumab vs. Checkpoint Inhibitors in Combination Studies
Agent/Class
Mechanism
Outcome in Combination Studies (Immune-Oncology Model)
Bimekizumab
Dual IL-17A/F blockade
Reduces pro-inflammatory signaling and tissue damage, may decrease tumor-supportive inflammation
Anti-CTLA-4
Blocks CTLA-4 on T cells
Promotes T cell activation; in models, works mainly via CD8+ T cell activation
Anti-LAG-3
Blocks LAG-3 on T cells
Alters Treg and CD4+ helper T cell activity, leading indirectly to enhanced CD8+ T cell responses
Combination
Both pathways targeted
Tests synergy: heightened antitumor immunity if inflammation suppression and immune "brake" release are complementary
Important note: Direct experimental evidence specifically combining bimekizumab biosimilars and checkpoint inhibitor biosimilars in immune-oncology models is not present in search results, but the described rationale and methodologies are consistent with current translational immuno-oncology research practices. Where specific preclinical data is missing, this answer is justified as an informed synthesis.
References:Mechanism and uses of bimekizumab, its effect on cytokine blockade, and immune modulation.Immune checkpoint combination mechanisms, T cell requirements, and preclinical experimental designs.
A Bimekizumab biosimilar is used in a bridging anti-drug antibody (ADA) ELISA by serving as both the capture reagent (coated on the plate) and the detection reagent (labeled with a reporter) to detect patient antibodies generated against the therapeutic drug.
How the bridging ADA ELISA works:
Capture step: The ELISA plate is coated with Bimekizumab biosimilar, which will bind to any anti-bimekizumab antibodies (ADAs) in the patient's sample.
Bridging/detection step: After the sample and any ADAs bind to the immobilized drug, a second, labeled form of the Bimekizumab biosimilar (commonly conjugated with an enzyme such as HRP or with biotin) is added. This binds to another epitope on the ADA, forming a "bridge": biosimilar–ADA–biosimilar.
Signal development: The plate is washed to remove unbound components, and a substrate for the enzyme label (e.g., TMB for HRP) is added. A measurable signal indicates the presence of ADAs that recognize and can simultaneously bind two Bimekizumab molecules.
Key points in the context of Bimekizumab:
The method can utilize either the reference Bimekizumab or a biosimilar with identical or highly similar structure and epitopes, ensuring that patient-generated ADAs to the therapeutic are detected with similar sensitivity.
The assay detects free (unbound) ADAs in patient plasma/serum and can be adapted to assess immune complexes or even specific antibody subclasses with additional reagents.
Why use a biosimilar as reagent?
Biosimilars allow detection of ADAs against regions present in the marketed therapeutic without using the original, potentially expensive material.
They ensure that the immunogenic profile assessed reflects what the patient was exposed to, assuming structural equivalency.
Application: This approach is routinely used to monitor a patient's immune response during clinical trials or treatment with Bimekizumab, helping assess immunogenicity risk and understand potential impacts on safety or efficacy.
Summary Table: Bridging ADA ELISA with a Bimekizumab Biosimilar
Step
Reagent Used
Role
Plate coating
Bimekizumab biosimilar (unlabeled)
Captures anti-Bimekizumab ADA
Detection
Bimekizumab biosimilar (labeled)
Binds to other ADA epitope
Signal development
Enzyme substrate (e.g., TMB)
Detects bridging complex
This methodology is a standard for monitoring immunogenicity of monoclonal antibody therapeutics, including Bimekizumab.
References & Citations
1 Li X, Bechara R, Zhao J, et al. Nat Immunol. 20(12):1594-1602. 2019.
2 Amatya N, Garg AV, Gaffen SL. Trends Immunol. 38(5):310-322. 2017.
3 Golbari NM, Basehore BM, Zito PM. Brodalumab. [Updated 2023 Aug 8]. In: StatPearls [Internet]. Treasure Island (FL): StatPearls Publishing; 2024 Jan-. Available from: https://www.ncbi.nlm.nih.gov/books/NBK470324/
4 Papp KA, Merola JF, Gottlieb AB, et al. J Am Acad Dermatol. 79(2):277-286.e10. 2018.
5 Glatt S, Helmer E, Haier B, et al. Br J Clin Pharmacol. 83(5):991-1001. 2017.
6 Kaplon H, Chenoweth A, Crescioli S, et al. MAbs. 14(1):2014296. 2022.
7 Nie T, Shirley M. Drugs. 84(5):587-598. 2024.
8 Baraliakos X, Deodhar A, van der Heijde D, et al. Ann Rheum Dis. 83(2):199-213. 2024.