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.
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.
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Basiliximab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Reported Applications For Relevant Conjugates ?
CyTOF®
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 Basiliximab. Basiliximab recognizes human CD25. This product is for research use only.
Background
CD25, a 55 kD type I transmembrane glycoprotein, has been shown to play roles in lymphocyte differentiation, activation, and proliferation. Many resting memory T cells constitutively express IL2Rα. It functions as the receptor for HTLV-1, resulting in its expression on neoplastic cells in adult T cell lymphoma/leukemia. CD25 (sIL-2R) has been used to track disease progression. Some additional clinical applications include Chagas disease, a disease characterized by a decline of CD25 expression on immune cells, and Multiple sclerosis, in which treatments with mAbs target CD25. Anti-Human IL-2R alpha (CD25) (Basiliximab) utilizes the same variable regions from the therapeutic antibody Basiliximab making it ideal for research projects.
Antigen Distribution
IL-2Rα is expressed on activated mature T and B lymphocytes, during early stages of thymocytes development, pre-B cells, and in activated CD4+ memory T-lymphocytes.
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 Basiliximab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to quantify Basiliximab concentrations in serum samples by establishing a standard curve against which unknown sample concentrations are measured.
In detail:
Biosimilar antibodies are highly similar to FDA-approved therapeutic antibodies in structure and function, but produced specifically for research purposes and not clinical use. This allows researchers to use them in analytical assays without the cost or regulatory constraints associated with clinical-grade drugs.
PK bridging ELISA assay principle:
Wells are coated with an anti-Basiliximab antibody (capture antibody).
Serum samples and calibration standards (prepared from research-grade Basiliximab biosimilar at known concentrations) are added to wells.
Any Basiliximab present in samples/standards binds to the capture antibody.
A horseradish peroxidase (HRP)-conjugated anti-Basiliximab (detection antibody) is added to form a "bridge" between the drug and the detection system.
After washing, a substrate (e.g., TMB) is added; the resulting colorimetric change is proportional to Basiliximab concentration and is measured spectrophotometrically.
Calibration standards/standard curve:
Serial dilutions of the research-grade biosimilar Basiliximab are prepared to create a range of known concentrations, typically spanning the expected concentrations in patient serum (e.g., 0–2000 ng/mL).
These standards are run alongside patient serum samples.
The colorimetric (absorbance) readings for standards are used to generate a standard curve, against which sample values are interpolated to provide actual drug concentration in ng/mL.
Reference controls:
Research-grade biosimilars can also be used as positive controls to ensure assay consistency and accuracy from run to run—confirming that the assay correctly measures known amounts of Basiliximab.
Why biosimilars are used:
Therapeutic-grade Basiliximab may be expensive or hard to obtain in small quantities.
Research biosimilars closely mimic the clinical drug's binding and structure, making them valid analytical surrogates, provided their equivalency is confirmed.
Considerations:
The accuracy of PK data hinges on the biosimilar's verified similarity to the clinical drug in relevant analytical properties.
Standards should be handled and stored according to manufacturer guidelines, and the assay validated for the specific sample type (e.g., human serum).
Summary Table: Basiliximab Biosimilar in PK Bridging ELISA
Step
Role of Research-grade Basiliximab Biosimilar
Calibration standard preparation
Serial dilutions for standard curve
Reference (positive) control
Quality control for assay consistency
Analytical validation
Equivalency check to ensure results mirror clinical drug
Quantitation of patient serum samples
Standard curve interpolation gives drug concentration
This approach is standard in PK bridging ELISA and ensures reliable, regulated quantification of therapeutic antibodies in serum samples for preclinical and clinical research.
Primary Mouse Models for Anti-CD25 Antibody Studies
Syngeneic and humanized mouse models are the primary platforms for in vivo administration of research-grade anti-CD25 antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs). Their advantages, uses, and limitations are summarized below.
Syngeneic Mouse Models
Syngeneic models involve implanting murine tumor cell lines (e.g., MC38, A20, TC-1) into immunocompetent mice of the same genetic background, thereby preserving a fully functional immune system. These models are widely used to evaluate the efficacy of immune checkpoint inhibitors and other immunotherapies, including anti-CD25 antibodies, because they allow for the expansion, infiltration, and functional characterization of TILs in a physiological context.
Application Example: In one study, an implantable anti-CD25 antibody-immobilized mesh (CD25-PE) was placed subcutaneously near tumors in mice, effectively capturing regulatory T cells (Tregs) and suppressing tumor growth. TILs (including Tregs) were subsequently characterized by histology and immunostaining.
Strengths: Syngeneic models are robust for studying immune-tumor interactions, TIL dynamics, and the efficacy of immunomodulatory agents. They are fully characterized for baseline immune populations and therapeutic responses.
Limitations: These models use mouse tumors and immune cells, so findings may not always translate directly to human biology.
Humanized Mouse Models
Humanized models are created by engrafting human immune cells (often hematopoietic stem cells or peripheral blood mononuclear cells) into immunodeficient mice, sometimes alongside patient-derived xenografts (PDX). These models enable the study of human TILs and human-specific immune responses.
Application Example: A CD25×TIGIT bispecific antibody was tested in CD25-humanized mice, demonstrating enhanced anti-tumor activity without peripheral Treg toxicity. Such models allow evaluation of human anti-CD25 antibodies against human CD25-expressing tumors in the presence of a (partial) human immune system.
Strengths: Humanized models can test human-specific antibodies and therapies, providing insights that may be more directly relevant to clinical translation.
Limitations: The reconstituted human immune system is incomplete, and the tumor microenvironment may not fully recapitulate human disease. These models are also more complex and expensive to establish and maintain.
Model Comparison Table
Model Type
Immune System
Tumor Origin
TIL Characterization
Anti-CD25 Antibody Example
Key References
Syngeneic
Mouse
Mouse cell lines
Mouse TILs
Implanted anti-CD25 mesh captures Tregs
Humanized
Human (partial)
Human or mouse
Human TILs
CD25×TIGIT bispecific in humanized mice
Summary
Syngeneic models are the most commonly used for initial in vivo characterization of anti-CD25 antibodies in oncology research, offering a fully functional (mouse) immune system and enabling detailed study of TIL dynamics and therapy response.
Humanized models are employed when the goal is to test human-specific antibodies or to model human immune-tumor interactions more closely, despite their technical and interpretational challenges.
Both models are critical for preclinical immunotherapy development, with syngeneic models providing broad immune-tumor interaction data and humanized models offering human-relevant insights for clinical translation.
Researchers use Basiliximab biosimilars to selectively inhibit the IL-2 signaling pathway via CD25 blockade, and when combined with other immune checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars), they study synergistic effects on T-cell activation, proliferation, and anti-tumor immune responses in complex immune-oncology models.
Context and Supporting Details:
Basiliximab Mechanism: Basiliximab biosimilars block the CD25 subunit of the IL-2 receptor on activated T-cells, reducing their proliferation while sparing overall T-cell populations, which makes them an attractive tool for targeted immunomodulation without broad immunosuppression.
Checkpoint Inhibitor Mechanisms:
Anti-CTLA-4 inhibitors (e.g., ipilimumab biosimilars) block CTLA-4, a negative regulator predominantly active in lymph nodes, enabling enhanced induction and expansion of activated T-cells.
Anti-LAG-3 biosimilars inhibit LAG-3, which is often co-expressed with PD-1 on exhausted T-cells, potentially restoring their function and further enhancing anti-tumor immunity.
Combination Strategies:
Researchers hypothesize that combining Basiliximab biosimilars with other checkpoint inhibitors targets multiple immune regulatory pathways, leading to greater synergistic augmentation of anti-tumor responses than single agents alone.
For example, blocking CD25 (IL-2RA) with Basiliximab can modulate T-cell activation, while simultaneous blockade of CTLA-4 or LAG-3 can further lift inhibitory brakes, potentially resulting in more robust and sustained cytotoxic T-cell activity against tumor cells.
These combinations are often studied in preclinical models (e.g., murine cancer models) to assess effects on tumor growth, immune cell infiltration, cytokine profiles, and survival outcomes.
Technical Approaches:
Experimental Design typically involves administering Basiliximab biosimilars alongside checkpoint inhibitors in vitro (cell-based assays) and in vivo (animal models) to monitor:
T-cell proliferation and activation
Cytokine secretion profiles
Tumor immune microenvironment changes
Tumor regression and survival rates
Synergistic Effects are evaluated by comparing combination therapy outcomes versus each monotherapy, with end-points including enhanced tumor cell clearance and altered patterns of immune-related gene expression.
Advantages of Biosimilars for Research:
Lower cost and high consistency allow broad experimental use, facilitating large-scale and repeated combinatorial studies in controlled settings before progressing to clinical trials.
Safety and Toxicity:
Combination approaches can increase efficacy but often also raise the risk for immune-related adverse events, such as enhanced autoimmunity or cytokine storms, which are systematically recorded and analyzed in these models.
In summary, researchers leverage the targeted T-cell activation inhibition of Basiliximab biosimilars in tandem with checkpoint blockade (anti-CTLA-4, anti-LAG-3, etc.) to dissect and enhance the complex, multifaceted immune responses in oncology, aiming to optimize therapeutic synergies while carefully monitoring safety profiles.
A Basiliximab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient's immune response against Basiliximab by exploiting the bivalent nature of IgG anti-drug antibodies produced in response to the therapeutic.
In a bridging ADA ELISA:
The plate is typically coated with Basiliximab (or its biosimilar).
Patient samples, which may contain anti-Basiliximab antibodies, are then added; if present, these ADAs will bind to the Basiliximab on the plate.
Next, a detection reagent—often Basiliximab conjugated to a reporter (such as biotin or an enzyme)—is added. ADA molecules, via their two antigen-binding arms, “bridge” between the capture Basiliximab on the plate and the labeled detection Basiliximab.
This “bridging” format requires that the anti-drug antibodies are at least bivalent (typically IgG), allowing each ADA molecule to bind both the immobilized and the labeled Basiliximab simultaneously.
Key points about this setup:
Biosimilars of Basiliximab are especially useful as capture or detection reagents because they closely mimic the structure of the original drug, ensuring relevant immune recognition and consistent assay performance for research and clinical monitoring.
Using a Basiliximab biosimilar in both capture and detection steps increases specificity for anti-Basiliximab antibodies, minimizing interference from other serum antibodies.
The assay detects total binding antibodies (regardless of whether they are neutralizing or not), providing a sensitive method for monitoring immunogenicity in patients.
Supporting context:
The bridging ELISA is a common method for ADA detection across multiple biologics (such as adalimumab and others), and its principle remains the same for biosimilars: a drug or biosimilar-coated surface captures drug-specific antibodies, and labeled drug/biosimilar serves as detection to form the "bridge".
The use of biosimilars can make such assays more cost-effective and broadly available for research and clinical surveillance, provided the biosimilar is rigorously characterized for equivalency to the reference product in binding and structure.
In summary, a Basiliximab biosimilar as both capture and detection reagent in a bridging ADA ELISA enables sensitive detection of anti-Basiliximab antibodies, facilitating monitoring of immunogenicity in patients or study participants exposed to the therapeutic drug.