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 Ublituximab. TG-1101 targets a unique epitope on the CD20
antigen.
Background
CD20 is a transmembrane protein that is prominently present on the surface of B-cells from
the early to mature stages, but notably absent on hematopoietic stem cells, pro-B cells, or
plasma cells. Its significance lies in its role in B-cell functions such as activation and
differentiation. It is a key target for monoclonal antibodies used in the treatment of B-cell-
related diseases and autoimmune conditions. Monoclonal antibodies targeting CD20 have
been widely used to treat B-cell lymphomas, leukemias, and autoimmune diseases like
rheumatoid arthritis and systemic lupus erythematosus. These antibodies work by selectively
targeting and depleting B-cells that express CD20, thereby modulating the immune response
and reducing inflammation. This targeted approach has shown promising results in managing
various B-cell disorders and has significantly improved the prognosis for patients with these
conditions1,2.
TG-1101, also known as Ublituximab, is a monoclonal antibody that targets the CD20
antigen. This helps enhance the immune responses that can destroy B-cells expressing CD20.
Ublituximab is currently being studied for its potential in treating B-cell disorders and
autoimmune diseases. It has received its first global approval in the USA for the treatment of
relapsing forms of multiple sclerosis (MS), including clinically isolated syndrome, relapsing-
remitting disease, and active secondary progressive disease3-5.
Antigen Distribution
CD20 is primarily expressed on the surface of B lymphocytes,
including both normal and malignant B cells.
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 Ublituximab biosimilars are used as analytical standards or reference controls in PK bridging ELISA assays by serving as the calibration standard curve against which unknown serum sample concentrations are quantified. This approach allows for precise and accurate measurement of drug levels in clinical pharmacokinetic (PK) studies.
Essential details and methodology:
Calibration Standard Selection: In PK assays for biosimilars, industry best practice is to use a single analytical standard—either the biosimilar or reference product—to generate the standard curve for quantitation of both biosimilar and reference drug in serum samples.
Reference Control Role: The research-grade Ublituximab biosimilar, once shown to be bioanalytically equivalent to the innovator/reference product, can be used as the calibration standard. Quality control (QC) samples prepared from both biosimilar and reference products are quantified against this standard curve.
Assay Configuration (PK Bridging ELISA):
The assay uses a sandwich ELISA format with anti-idiotypic antibodies specific to Ublituximab’s unique variable regions, giving specificity for the drug even in the presence of other human IgGs or serum proteins.
The standard curve is created by serially diluting the biosimilar Ublituximab in drug-naïve human serum to span the expected clinical PK range (e.g., 50–12,800 ng/mL).
Unknown serum samples from treated subjects are analyzed alongside this standard curve, and their concentrations are interpolated.
The integrity and comparability of the biosimilar as a standard are validated, ensuring its assay behavior matches that of the reference product (for example, by demonstrating parallel response and similar binding in the ELISA).
Validation and Controls:
Analytical equivalence between biosimilar and innovator standards is established by comparing signal responses over the calibration range, often by statistical analysis of the curve data.
QC samples at multiple concentrations with both the biosimilar and reference protein are included to demonstrate consistent quantification regardless of standard origin, further supporting the biosimilar’s suitability as a reference standard.
Resulting Data Usage: With this approach, the PK bridging ELISA can accurately measure the concentration of Ublituximab (whether from biosimilar or reference source) in serum, supporting both PK similarity and bioequivalence evaluations in biosimilar development programs.
Key technical highlights:
Using research-grade Ublituximab biosimilar as the calibration standard is only appropriate after rigorous bioanalytical comparability has been established with the reference product through well-designed qualification and validation studies.
The PK bridging ELISA is highly specific for Ublituximab thanks to anti-idiotypic antibodies, minimizing interference from endogenous IgG or other drugs.
This unified standard approach minimizes assay variability and eliminates the need for multiple, separate standard curves for biosimilar and reference measurements, thus supporting regulatory and scientific requirements for biosimilar comparability.
In summary: Research-grade Ublituximab biosimilars serve as calibration standards or reference controls in PK bridging ELISA by providing the benchmark standard curve for quantitative drug measurement in serum, but only after comparability with the reference product is analytically validated to ensure accuracy and regulatory acceptability.
The primary models where a research-grade anti-CD20 antibody is administered in vivo to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are mouse syngeneic tumor models, especially those engineered to express human CD20, as well as specialized humanized models in select cases.
Key models and approaches:
Syngeneic mouse tumor models:
These models (such as A20-human CD20, TC-1, MC38, RENCA, and CT26) involve transplanting murine tumor cells (sometimes engineered to express human CD20) into immunocompetent mice.
This allows the evaluation of anti-CD20 antibodies’ effects on tumor growth and immune cell (notably TIL) composition within a functioning mouse immune system.
Studies have shown that anti-CD20 treatment results in B cell depletion and can inhibit tumor growth or augment immunotherapy efficacy. For example, injecting an anti-mouse CD20 antibody in syngeneic models retards tumor growth and increases CD8+ T cell infiltration and activity within tumors.
Human CD20 transgenic mice with syngeneic tumors:
Some models use murine lymphoma lines (like A20) engineered to express human CD20, implanted into mice that are also transgenic for human CD20 and/or other relevant immune molecules.
These models are suitable for preclinical testing of research-grade anti-human CD20 antibodies and enable detailed analysis of TILs.
Characterization of TILs:
Most studies using these models include profiling of TILs via flow cytometry or immunohistochemistry to observe shifts in populations, such as increases in CD8+ T cells or other immune compartments following antibody therapy.
Humanized mouse models (with a human immune system) are less commonly used specifically for anti-CD20 tumor/TIL studies due to complexity and expense, but can be considered for translational research if human immune interaction is essential. More frequently, the models above are preferred due to reliable immune context and existing tools for characterization.
Summary Table
Model Type
Antibody Target
Immunological Context
Application
Mouse syngeneic (e.g., TC-1, MC38, RENCA)
Anti-mouse CD20
Native mouse immune system
Tumor growth inhibition, TIL characterization
Human CD20 transgenic mice + human CD20 tumor
Anti-human CD20
Mouse, engineered human CD20
Efficacy, TIL composition, mechanistic studies
Humanized mouse models
Anti-human CD20
Human immune system
Specialized, limited use for TIL studies
These models are widely used as standard preclinical tools to evaluate anti-CD20 mechanism of action, anti-tumor activity, and immune microenvironment remodeling, particularly regarding TIL alterations.
Researchers use the Ublituximab biosimilar, an anti-CD20 monoclonal antibody, in combination with other checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—in complex immune-oncology models to examine potential synergistic effects on tumor control and immune modulation. While public clinical data on specific use of ublituximab with checkpoint inhibitors are still emerging, the combination strategies align with broader approaches demonstrated with checkpoint inhibitor pairings.
Key experimental strategies and rationale:
Immune Pathway Targeting: Ublituximab targets CD20 on B cells, mediating their depletion and modifying the immune landscape—reducing immunosuppression and facilitating greater T-cell activation. Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) release the brakes on T cells, allowing enhanced cytotoxic responses against tumor cells.
Synergistic Mechanisms: By combining a B-cell depleting agent (ublituximab) with T cell–activating agents (checkpoint inhibitors), researchers seek to augment antitumor immunity compared to either agent alone. B cell depletion may reduce regulatory B cells and dampen immune escape mechanisms, while checkpoint inhibitors counteract T cell exhaustion.
Model Systems: These combinations are tested in murine xenograft models (mouse tumor models with humanized immune components), which allow assessment of tumor growth, immune cell infiltration, and cytokine milieu following treatment.
Experimental readouts and endpoints include:
Tumor volume and growth kinetics.
T cell infiltration, activation status, and cytotoxicity markers within the tumor microenvironment.
Biomarkers of B cell depletion and changes in cytokine or chemokine profiles.
Toxicity and overall survival in preclinical subjects.
Comparison and design insights:
Component
Mechanism
Expected Outcome When Combined
Ublituximab
CD20-mediated B cell depletion
Reduces immune suppression, primes TME for T cells
The approach mirrors that of other antibody combinations (e.g., anti-PD-1 plus anti-CTLA-4), where synergy is shown in preclinical and some clinical models. The diversity of immune compartment targeting is critical: CTLA-4 blockade enhances T cell priming/activation, and ublituximab shifts the tumor milieu by eliminating B cell–driven immunosuppression.
Limitations and future directions:
Data on co-administration of ublituximab specifically with checkpoint inhibitors is still mostly preclinical; combinations are under study for optimizing efficacy and reducing overlapping toxicities.
Translational research seeks biomarkers to define which tumors and patient populations will benefit most from such regimens and to monitor immune activation and toxicity.
In summary, ublituximab biosimilar is combined with checkpoint inhibitors in preclinical models to investigate whether B cell depletion can amplify the immune-activating effects of checkpoint blockade, potentially leading to improved antitumor effects through complementary mechanisms.
A Ublituximab biosimilar can serve as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA, enabling the monitoring of a patient’s immune response against ublituximab by detecting antibodies that a patient develops against the therapeutic drug.
How this works in bridging ADA ELISA:
Bridging ADA ELISA principle: The bridging format is specifically designed to detect antibodies against therapeutic monoclonal antibodies in patient samples. The assay uses the therapeutic antibody (or its biosimilar) labeled in two different ways: one form is immobilized on the plate (capture reagent), and another form is labeled for detection (detection reagent).
Step-by-step context:
The ELISA plate is coated with biotinylated Ublituximab biosimilar, which captures any anti-ublituximab antibodies (ADAs) present in the patient’s serum.
The patient’s serum is added, allowing any ADAs specific to ublituximab to bind to the immobilized drug.
After washing to remove unbound components, HRP-labeled Ublituximab biosimilar (or another appropriately labeled form) is added. This binds to the unoccupied ADA paratope, thus "bridging" between two drug molecules (immobilized and labeled) via the ADA.
After additional washes, the extent of ADA binding is detected using a chromogenic substrate (e.g., TMB with HRP), resulting in a measurable color change proportional to the ADA concentration.
Key details and benefits:
Specificity: Using a biosimilar ensures that the detected anti-drug antibodies target epitopes present in both the biosimilar and the reference therapeutic, reflecting true immunogenicity to the therapeutic drug.
Interchangeability: It is common to use either the original biologic or a biosimilar in such assays, as long as the critical binding epitopes are preserved between the two, allowing reliable monitoring of immunogenic responses.
Summary Table: Ublituximab Biosimilar in Bridging ADA ELISA
Role of Ublituximab Biosimilar
Description
Capture Reagent
Immobilized (e.g., biotinylated) on ELISA plate to bind ADAs in the patient’s serum
Detection Reagent
Labeled (e.g., HRP-labeled) to detect ADA by bridging across captured antibodies
Purpose
Quantifies ADAs specific to ublituximab, indicating patient immune response against the drug
Additional context:
Using a biosimilar is validated by regulatory and clinical studies that show immunogenicity profiles of biosimilars are closely comparable to their reference biologics, making them suitable surrogates for assay reagents in immunogenicity monitoring.