Anti-Human CD20 (Rituximab) [Clone 10F381] — Fc Muted™

Anti-Human CD20 (Rituximab) [Clone 10F381] — Fc Muted™

Product No.: LT905

- -
- -
Product No.LT905
Clone
10F381
Target
CD20
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Rituximab, CD20, MS4A1,
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC
,
IP
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human CD20
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.
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Additional Applications Reported In Literature ?
ELISA,
FA,
FC,
IP,
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 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
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology

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.

Use of Research-Grade Rituximab Biosimilars as Calibration Standards in PK Bridging ELISA

Pharmacokinetic (PK) bridging ELISA is a standard method for quantifying therapeutic monoclonal antibodies like rituximab in serum, supporting both original and biosimilar development. Here’s how research-grade biosimilars function as calibration standards or reference controls in these assays:

Calibration and Reference Role of Biosimilars

  • Calibration Curve Construction: In PK bridging ELISA, a series of known concentrations of rituximab (original or biosimilar) are used to generate a standard curve. The optical density (OD) values from these standards are plotted against their concentrations to establish a relationship used to quantify unknown samples. Biosimilars, when characterized and validated for analytical similarity, can serve as reference materials alongside the originator product.
  • Batch-Specific Calibration: The calibration is typically lot-specific, meaning each batch of reference standard (originator or biosimilar) is independently qualified. Standards provided in commercial ELISA kits may be calibrated against both the originator (Rituxan™) and alternate biosimilar recombinant rituximab injections to ensure broad applicability.
  • International Standards: Some kits are calibrated against international reference standards (e.g., from NIBSC), with specified conversion factors (e.g., 100 μg/ml = 1,000 IU of rituximab). Biosimilars can be cross-referenced to such standards to ensure traceability and comparability across assays.

Assay Validation and Biosimilar Equivalence

  • Validation of Biosimilar Standards: For a biosimilar to be used as a reference, it must demonstrate analytical and functional equivalence to the originator in terms of binding characteristics, linearity, precision, and recovery in the ELISA system. Published studies confirm that biosimilar rituximab can be “comparable and equipotent” in cell-based and immunoassays when compared to the reference standard.
  • Assay Performance: Validation parameters such as linearity (regression coefficient >0.9), precision (CV <10–25%), accuracy (91–125%), and spike recovery (within ±15%) are critical. These metrics ensure that the biosimilar-derived standard curve reliably reflects the concentration of rituximab in patient samples. Dilutional linearity and parallelism are also confirmed to handle the expected range of serum concentrations.
  • Specificity and Cross-Reactivity: The assay must be specific for rituximab and show minimal cross-reactivity with endogenous immunoglobulins. This is typically confirmed during validation, regardless of whether originator or biosimilar standards are used.

Practical Protocol for PK Bridging ELISA

A typical PK bridging ELISA protocol involves:

  • Coating plates with an anti-idiotypic antibody specific to rituximab.
  • Blocking to minimize nonspecific binding.
  • Adding standards (originator or biosimilar rituximab at known concentrations) and serum samples.
  • Detection with an enzyme-conjugated anti-human IgG antibody, producing a signal proportional to rituximab concentration.
  • Quantification by interpolating sample OD values on the standard curve.

The detection range is broad (e.g., 0.1–1,000 ng/ml) to cover expected serum levels post-administration. Standards, whether originator or biosimilar, should cover this full range, including a zero concentration for background subtraction.

Summary Table: Key Aspects of Using Biosimilars as Standards

AspectDescriptionReference
Calibration CurveBiosimilars used alongside originator to generate standard curve for quantification
ValidationMust demonstrate equivalence in linearity, precision, accuracy, and recovery
SpecificityAssay must be specific for rituximab, with minimal cross-reactivity
TraceabilityStandards may be referenced to international biological standards
Practical UseUsed in the same way as originator in ELISA protocols for serum PK analysis

Conclusion

Research-grade rituximab biosimilars, when analytically validated, are routinely used as calibration standards and reference controls in PK bridging ELISAs to measure drug concentration in serum samples. They must demonstrate equivalence to the originator in critical assay performance parameters and can be cross-referenced to international standards for traceability. This approach supports robust, comparable pharmacokinetic assessments for both originator and biosimilar rituximab products in clinical and research settings.

The primary in vivo models for studying the effects of research-grade anti-CD20 antibody on tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are predominantly syngeneic mouse tumor models engineered to express human CD20, as well as fully murine models using species-specific anti-CD20 antibodies.

Key model types:

  • Syngeneic mouse tumor models:

    • Mouse tumor cell lines (e.g., EL4, A20, CT26, RENCA, B16F10) are genetically modified to express human CD20 so that human or research-grade anti-CD20 antibodies can target them. These models are immunocompetent, allowing for TIL characterization.
    • Example: EL4 murine lymphoma cells stably transduced with human CD20 (EL4-CD20) in C57BL/6 mice were used to study different anti-CD20 monoclonal antibody mechanisms under low and high tumor burden, enabling analysis of effector cell recruitment and TIL composition in situ.
    • Example: A20-human CD20 syngeneic mouse lymphoma model is used to assess anti-tumor activity and TIL profiles with CD20-TDB (T-cell dependent bispecific antibody) and anti-PD-L1, highlighting combinatorial therapeutic effects and immune infiltration.
    • Solid tumor models expressing CD20, such as mouse lung cancer TC1 expressing HPV-E7, have been used to study B-cell depletion by anti-mouse CD20 antibody, revealing changes in tumor growth and increased activation of intratumoral CD8+ T cells.
  • Humanized mouse models:

    • Less commonly referenced for anti-CD20 mechanistic studies linked to TIL analysis in the cited results, but these involve immunodeficient mice engrafted with a humanized immune system and human CD20+ tumor cells. These models allow testing clinical-grade antibodies and detailed phenotyping of human immune cell infiltrates.

Syngeneic vs. Humanized Model Comparison Table

Model TypeExample Cell Lines/FeaturesImmune SystemProsCons
Syngeneic-hCD20EL4, A20 (murine, hCD20+)Fully murineImmunocompetent; TIL profilingAntibody must be cross-reactive
Syngeneic-mCD20TC1, B16F10, CT26, RENCA (murine)Fully murineDirect B-cell depletionNot human-specific antibody
HumanizedHuman lymphoma or solid tumor cellsHumanized (mouse + human)Human antibody testing; human TILsSetup is costly and complex

Supporting Details:

  • Syngeneic models are widely used with anti-CD20 antibodies to study immunotherapeutic mechanisms, tumor growth inhibition, and TILs, since their intact immune system allows robust analysis of lymphocyte infiltration and function.
  • These models enable quantification and phenotyping of TILs under different conditions (e.g., tumor burden, single agent vs. combination therapy), and facilitate dissection of antibody effector mechanisms such as antibody-dependent cellular cytotoxicity (ADCC), complement-dependent cytotoxicity (CDC), and interplay with other immunotherapies.
  • B-cell depletion with anti-CD20 antibody not only retards tumor growth but also modulates the immune microenvironment, increasing activated CD8+ T cells in tumors.

In summary, syngeneic mouse models engineered to express human CD20 are the principal preclinical systems for in vivo administration of research-grade anti-CD20 antibodies to analyze tumor growth and TILs. Humanized models are occasionally used for higher translational relevance but are less frequently reported in these mechanistic studies.

Researchers use Rituximab biosimilars alongside other checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars to study potential synergistic effects in complex immune-oncology models by employing combination strategies that target both B cell-mediated immunity (via CD20) and T cell regulation (via immune checkpoints).

Key approaches and supporting details:

  • Model Selection: Combination studies are often performed in in vitro cellular systems or in vivo preclinical models (e.g., mouse tumor xenografts) that replicate aspects of the human immune system and tumor microenvironment.

  • Mechanistic Rationale:

    • Rituximab biosimilars deplete B cells by targeting CD20, through mechanisms such as complement-mediated cytotoxicity (CMC), antibody-dependent cellular cytotoxicity (ADCC), and apoptosis.
    • Immune checkpoint inhibitors (anti-CTLA-4, anti-LAG-3, or anti-PD-1/PD-L1 biosimilars) relieve inhibitory signals that normally restrain T cell activation, enhancing anti-tumor immunity.
    • By combining these agents, researchers evaluate whether B cell depletion can modulate the tumor immune microenvironment to make checkpoint blockade therapy more effective, or whether checkpoint inhibition enhances immune cell-mediated destruction of residual tumor B cells.
  • Experimental Workflow:

    • Researchers administer Rituximab biosimilars to selectively deplete B cells in the model system.
    • They then treat with a checkpoint inhibitor biosimilar (e.g., anti-CTLA-4, anti-LAG-3) to activate T cells by blocking suppressive pathways.
    • Functional endpoints measured include tumor growth, immune cell infiltration, cytokine profiles, and survival.
    • Assays such as flow cytometry (e.g., for immune cell profiling), cytotoxicity assays, and histopathology are used to assess effects.
  • Why Use Biosimilars?

    • Cost and accessibility: Biosimilars are less expensive than original biologics, making complex combination and mechanistic studies more feasible.
    • Reproducibility: Research-grade biosimilars are engineered to closely mimic therapeutic antibodies, ensuring consistent results for preclinical studies.
  • Emergent Research Questions:

    • Does B cell depletion sensitize tumors to T cell checkpoint blockade?
    • Are specific B cell subpopulations immunosuppressive and does their removal via Rituximab enhance T cell responses?
    • What is the safety and efficacy profile of these combinations compared to monotherapy?

Limitations and considerations:

  • While biosimilars allow robust preclinical modeling, translation to human efficacy requires clinical validation.
  • Most evidence for combinatorial mechanistic synergy remains preclinical; published clinical data combining Rituximab with checkpoint inhibitors is limited as of now.

Summary Table: Rituximab Biosimilar + Checkpoint Inhibitor Combination

Agent TypeImmune TargetMechanismModel UseResearch Benefit
Rituximab biosimilarCD20 on B cellsB cell depletion (CDC, ADCC, apoptosis)Cancer/autoimmune modelsRemoves immunosuppressive B cells, mimics clinical therapy
Anti-CTLA-4/LAG-3/etc. biosimilarsCTLA-4, LAG-3, etc. on T cellsImmune checkpoint blockade, enhances T cell activityOncology, immune modelsProbes synergy with B cell depletion, improves adaptive immune response

This combinatorial approach allows researchers to test hypotheses about synergistic anti-tumor immunity arising from simultaneous targeting of both arms of the immune system—B cells and T cells—using cost-effective, research-grade biosimilars as surrogates for clinical antibodies.

A Rituximab biosimilar is commonly used in a bridging anti-drug antibody (ADA) ELISA as both the capture and detection reagent to specifically identify patient-generated antibodies against the therapeutic drug (Rituximab or its biosimilar).

In a typical bridging ADA ELISA for Rituximab:

  • Coating/Capture Step: The biosimilar Rituximab is immobilized on the ELISA plate’s surface.
  • Sample Incubation: Patient samples (typically serum) are added. If the patient has generated anti-Rituximab antibodies (ADAs), these will bind to the immobilized Rituximab via one arm of the antibody.
  • Detection/Bridging Step: A labeled (often biotin- or HRP-conjugated) version of Rituximab (the same biosimilar) is added. If ADAs are present, they “bridge” between the immobilized drug and the labeled drug, binding both via their two antigen-binding sites.
  • Signal Development: The presence of the ADA–drug–labeled-drug complex is detected using a reporter system (e.g., HRP and TMB substrate for color development). The resulting signal is directly proportional to the concentration of ADA in the patient sample.

Why a Rituximab biosimilar is used:

  • Immunochemical identity: Biosimilars are structurally and functionally similar to the originator drug, making them equally effective in binding to anti-Rituximab antibodies generated in response to therapy.
  • Specificity: Using the biosimilar (rather than irrelevant antibodies) ensures that the assay specifically detects antibodies produced against Rituximab and not against unrelated proteins.
  • Practicality: Biotinylated or HRP-labeled versions of the biosimilar can be easily produced, facilitating high-throughput and sensitive detection in clinical monitoring.

This format is especially sensitive because ADAs are typically bivalent, permitting the “bridging” formation necessary for detection. Importantly, this method can distinguish between true anti-drug antibodies and other serum components, provided appropriate controls and specificity reagents are included.

In summary, a Rituximab biosimilar serves as the key antigen in the bridging ADA ELISA: immobilized on the plate for capture and labeled for detection, enabling identification and quantitation of patient antibodies against the therapeutic Rituximab.

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.
Indirect Elisa Protocol
FA
Flow Cytometry
Immunoprecipitation Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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