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 Rituximab. Clone 10F381 recognizes human CD20. This product is for research use only.
Background
CD20 is a nonglycosylated 33-37 kDa transmembrane-spanning phosphoprotein that is a member of the MS4A family which is widely expressed on normal B cell surfaces during all stages of development as well as by most B cell malignancies1, 2. The biological role of CD20 remains poorly understood; however, it is thought to be involved in calcium ion influx. CD20 has no natural ligand and is not immediately internalized upon antibody binding. Thus, mAbs directed against CD20 depend on the recruitment of a host response. CD20 is a popular target for mAb therapy because depleting developing B-cells generally does not cause permanent side effects (due to the fact that mature plasma cells and B-cell progenitors do not express CD20 and that there is limited expression of CD20 among other cell lineages).
Rituximab is a chimeric monoclonal antibody that binds to CD20. Rituximab is used to treat some autoimmune diseases and types of cancer such as non-Hodgkin lymphoma, chronic lymphocytic leukemia, and rheumatoid arthritis among others. The Fc portion of Rituximab mediates antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC). Rituximab increases MHC II and adhesion molecules LFA-1 and LFA-3 (lymphocyte function-associated antigen) and also induces apoptosis of CD20+ cells. This ultimately results in the elimination of B cells (including the cancerous ones) from the body, and thus allows a new population of healthy B cells to develop from lymphoid stem cells. Anti-Human CD20 (Rituximab) utilizes the same variable regions from the therapeutic antibody Rituximab making it ideal for research projects.
Antigen Distribution
CD20 is primarily found on the surface of immune system B cells. CD20 is highly expressed in the lymph node, and to a lesser extent, the spleen and appendix.
Ligand/Receptor
Src family tyrosine kinases, MHC class I, II, CD53, CD81, CD82
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 Rituximab biosimilars are used as calibration standards or reference controls in PK bridging ELISA assays by providing known, quantifiable concentrations for constructing standard curves against which unknown serum samples can be measured and compared.
In a pharmacokinetic (PK) bridging ELISA designed to measure drug concentration in serum samples, the use of Rituximab biosimilars as standards involves several key steps:
Calibration Standard Preparation: Biosimilar Rituximab is formulated at multiple known concentrations, often spanning a relevant range (e.g., 0, 30, 100, 300, 1000 ng/mL), and is included in the ELISA kit as standards for the assay. These standards are sometimes specifically calibrated against international standards (such as those from the National Institute for Biological Standards and Control, NIBSC) and reference products (original Rituxan™).
Assay Principle: The ELISA plate is coated with a capture antibody specific for Rituximab. The biosimilar standard and the patient serum samples (which may contain either reference or biosimilar Rituximab) are added to the plate, enabling binding to the capture antibody.
Detection and Quantification: After washing, a detection antibody binds specifically to the drug molecule, commonly targeting the human IgG Fc region. The colorimetric or chemiluminescent readout is proportional to the Rituximab concentration present in the wells.
Standard Curve Generation: The optical density (or signal) measured for each biosimilar standard concentration establishes a standard curve. The drug concentrations in serum samples are interpolated from this curve, providing quantitative PK data.
Lot-specific and Matrix Matching: Calibration is lot-specific because different lots of biosimilar Rituximab may have slight variations. Standards are often formulated in a matrix that matches human serum to mimic sample behavior.
Bridging Across Reference and Biosimilar: These biosimilar standards demonstrate high comparability and can be used to measure concentrations of both originator and biosimilar Rituximab in clinical samples due to equipotency and similar detection in ELISA formats. This ensures consistency and reliability in PK assessments during biosimilar development or interchangeability studies.
Key advantages:
Use of biosimilar standards ensures assay calibration is representative for both reference and biosimilar products, supporting regulatory bridging and interchangeability studies.
Accurate and reproducible quantification supports reliable pharmacokinetic profiling in clinical and preclinical studies.
Summary Table: | Purpose | Role of Biosimilar Standard | Example Concentrations | Calibration/Reference ||-----------------------|-------------------------------------------|----------------------------|---------------------------------|| Standard curve | Quantifies Rituximab in samples | 0, 30, 100, 300, 1000 ng/mL| Calibrated vs reference/biosimilar || PK bridging | Compares biosimilar and reference product | Same assay, same detection | Equipotent, comparable || Control/Validation | Ensures accuracy and reproducibility | Lot-specific, spiked serum | International standards (NIBSC)|
This process is essential in pharmacokinetic studies for biosimilar development, interchangeability assessment, and clinical monitoring of Rituximab therapy.
The primary in vivo models for administering a research-grade anti-CD20 antibody to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models, often genetically engineered to express human CD20, and occasionally humanized mouse models bearing human immune cells and/or hematologic tumors.
Key Model Types:
Syngeneic Mouse Models
These models use murine tumor cell lines (e.g., EL4, A20, TC1) implanted into immunocompetent mice of the same genetic background. For anti-CD20 studies, tumor cells are frequently transduced with human CD20 to allow recognition by therapeutic antibodies that target human CD20.
Example: The EL4-CD20 model involves EL4 murine lymphoma cells transduced to express human CD20 and implanted into syngeneic C57BL/6 mice, which are then treated with anti-CD20 antibodies to assess tumor inhibition and local immune responses.
The A20-human CD20 model utilizes A20 B-cell lymphoma cells engineered to express human CD20 in BALB/c mice, enabling evaluation of antibody efficacy and detailed immune profiling, including TIL analysis.
In vivo anti-CD20 treatment with these models readily allows for analysis of the TIL composition via flow cytometry or immunohistochemistry.
Syngeneic Models with Native Murine CD20
Some studies use anti-mouse-CD20 antibodies (for example, the 18B12 clone) that target native murine CD20, enabling B-cell depletion and assessment of the immunologic consequences for tumor control in solid or hematologic malignancies.
Example: The TC1 model (murine lung cancer expressing HPV-E7 antigen) in C57BL/6 mice has been used to examine how anti-mouse-CD20 therapy impacts both tumor growth and CD8+ T-cell infiltration into tumors.
Humanized Mouse Models (less common in preclinical anti-CD20 TIL studies)
These are immunodeficient mice engrafted with human hematopoietic stem cells to reconstitute a human immune system, sometimes bearing human CD20+ tumor xenografts.
This approach allows testing of human anti-CD20 agents on both human tumors and immune effectors but is more complex, costly, and less frequently employed for systematic TIL analyses compared to syngeneic models (inferred from general literature and model use patterns).
Summary Table — Commonly Used In Vivo Models:
Model Type
Tumor Type
CD20 Expression
Immune System
Use Cases
Syngeneic (e.g., EL4-CD20)
Mouse lymphoma or solid tumor
Human CD20 (transduced)
Mouse
Antibody efficacy, TIL characterization
Syngeneic (native CD20)
Mouse lymphoma/solid tumor
Murine CD20
Mouse
B-cell depletion, TIL consequences
Humanized mouse
Human CD20+ lymphoma/xenograft
Human CD20
Human (reconstituted)
Translational bridging, immune profiling
Key Insights:
Syngeneic models are the mainstay for in vivo studies of anti-CD20 antibodies, due to their reproducible tumor growth, manipulable immune environment, and suitability for detailed TIL profiling.
For evaluation of human anti-CD20 antibodies, syngeneic models expressing human CD20 are preferred, permitting use of research-grade or clinical antibodies like rituximab analogues.
Humanized models offer higher translational value but are less commonly used in the published TIL studies, possibly due to their complexity and variation in immune reconstitution.
References:
Murine syngeneic tumor models (including those with B-cell depletion using an anti-CD20 antibody) are used to study tumor inhibition and TIL characteristics.
The EL4-human CD20 and A20-human CD20 syngeneic models are specifically designed to examine anti-CD20 efficacy and immune cell infiltration.
General immunoprofiling methods for TILs in murine models are widely established and applicable to these systems.
Researchers use rituximab biosimilars in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to explore potential synergistic effects in immune-oncology models by targeting complementary mechanisms in cancer immunotherapy. This strategy aims to enhance anti-tumor responses by simultaneously modulating B cell populations and T cell regulatory pathways.
Context and Supporting Details:
Rituximab biosimilars target CD20 on B cells, leading to B cell depletion via mechanisms like complement-mediated cytotoxicity (CMC), antibody-dependent cellular cytotoxicity (ADCC), and induction of apoptosis. Their main function in oncology models is to eliminate cancerous or dysfunctional B cells and modulate immune regulation.
Checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) act primarily on T cells, either restoring activation and proliferation (CTLA-4 blockade—mostly in lymph nodes) or enhancing cytotoxicity at the tumor site (PD-1/PD-L1 blockade). Dual checkpoint blockade, such as CTLA-4 and PD-1/PD-L1, has demonstrated enhanced antitumor efficacy in several preclinical and clinical models due to their non-overlapping mechanisms.
Combination Rationale: By combining a rituximab biosimilar with checkpoint inhibitors, researchers aim to:
Remove immunosuppressive B cells from the tumor microenvironment (TME).
Simultaneously activate cytotoxic T cells and reduce regulatory T cell inhibition.
Overcome limitations observed in monotherapy, like incomplete tumor response or resistance.
Experimental Application: In complex immune-oncology models:
Mice or humanized models are treated with rituximab biosimilars to deplete B cells, followed by (or in combination with) checkpoint inhibitors to maximize T cell activation.
Researchers then assess tumor growth rates, immune cell infiltration, cytokine production, and survival rates to detect synergistic (more-than-additive) effects.
Such combinations have shown promise in augmenting immune responses and can inform new therapeutic regimens for cancers, especially when monotherapies are insufficient.
Benefits of Biosimilars: Research-grade biosimilars enable cost-effective and reproducible studies, closely simulating clinical biologics while allowing for large-scale experimental testing without prohibitive expenses.
Clinical Correlates: Although most combination therapies have focused on PD-1/PD-L1 with CTLA-4, the same approach can be extended to novel checkpoints like LAG-3, and with anti-CD20 biosimilars, to address distinct components of the immune system. Combination therapy improves outcomes in some patient subpopulations, with drawbacks including higher toxicity rates.
Summary of Use in Research:In summary, rituximab biosimilars, when combined with checkpoint inhibitors (including anti-CTLA-4 or anti-LAG-3 biosimilars), are used in research to probe synergistic immune effects by:
Depleting B cells,
Enhancing T cell anti-tumor functions,
Mapping immune interactions in complex tumor models,
Reducing costs for translational and preclinical studies.
This combined approach is central to developing more effective cancer immunotherapies and better understanding the tumor immune microenvironment.
A Rituximab biosimilar is commonly used as both the capture and detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against the therapeutic drug by detecting anti-Rituximab antibodies (ADAs) in the patient's serum.
Mechanism in Bridging ELISA:
Capture Phase: The microplate wells are coated with Rituximab (the biosimilar). When patient serum is added, any anti-Rituximab antibodies present (from the patient’s immune response) bind to the immobilized Rituximab on the plate.
Detection Phase: After washing, biotinylated Rituximab (the same biosimilar, labeled for detection) is added. If a patient's anti-Rituximab antibody is present, it will bind to both the immobilized Rituximab (via one of its antigen-binding sites) and the biotinylated Rituximab (via its other antigen-binding site), effectively bridging between the two drug molecules.
Signal Development: Streptavidin-conjugated HRP (or another suitable reporter) is added to bind the biotin on the detection Rituximab, followed by substrate addition for color development. The intensity of the resulting signal correlates with the amount of ADA in the sample.
Key Points:
The assay detects bivalent ADAs (antibodies using both arms to bind drug molecules).
Using the same drug (here, a Rituximab biosimilar) as both the capture and detection reagent ensures specificity to anti-drug antibodies generated in response to therapy.
The bridging format is highly sensitive and widely used for ADA immunogenicity testing but typically detects only bivalent antibodies, possibly missing low-affinity or monovalent antibodies.
Rationale for Using a Biosimilar:
Biosimilars are structurally and functionally equivalent to the original (originator) therapeutic antibody, so they can be used interchangeably in immunogenicity assays to detect patient-generated antibodies against either the biosimilar or the reference product.
Protocols and Examples:
Actual kit protocols use this format: plates are coated with rituximab (biosimilar or originator); after incubation with serum, labeled rituximab is added; binding indicates presence of ADAs.
Summary Table:
Step
Reagent
Function
Capture
Rituximab biosimilar (coated)
Captures ADAs in patient's serum
Detection
Biotinylated Rituximab
Bridges bound ADAs for detection
Signal
Streptavidin-HRP + substrate
Measures color development (quantifies ADA)
This approach is standard in immunogenicity testing for monoclonal antibody therapies, allowing clinicians to monitor the emergence of anti-drug antibodies in treated patients.
References & Citations
1. Middleton O, Wheadon H, Michie AM. Classical Complement Pathway. In MJH Ratcliffe (Ed.), Reference Module in Biomedical Sciences Encyclopedia of Immunobiology Volume 2 (pp. 318-324). Elsevier. 2016.
2. Freeman CL, Sehn LH. Br J Haematol. 182(1):29-45. 2018.
3. Mato, A. et al. (2018) Oncologist. 23(3):288-296.
4. Richards, K. et al. (2018) Front Oncol. 8: 163.