Humanized antibody bispecifically binds to human CD20 and CD3ε to engage T cells
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.
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 Mosunetuzumab. This product is for research use only. Mosunetuzumab simultaneously binds human CD3 and CD20.
Background
Mosunetuzumab is a CD20xCD3 Bispecific T cell Engager (BiTE) antibody developed as a
cancer immunotherapeutic drug1 using knobs-in-holes engineering2. Simultaneous targeting of
CD20 B cell on lymphomas and CD3 on T cells, leads to T cell activation, the release of perforin
and granzymes, and ultimately the depletion of malignant B cells through lysis and cell death1.
Mosunetuzumab benefits from CH mutations that limit its effector functions3, having a
modified, aglycosylated2 Fc with no Fcγ receptor or complement binding and only one binding
site to CD203.
Mosunetuzumab is subject to splicing-mediated mechanisms of epitope loss4. CD20 is a
nonglycosylated 33-37 kDa phosphoprotein member of the MS4A family5,6 that encodes four
variants (V1-4)4, with V1 and V3 being the most abundant. Mosunetuzumab is only effective
against V3-expressing B cells, and this likely plays a role in resistance to Mosunetuzumab in
some patients4. Mosunetuzumab is approved to treat follicular lymphoma, the second most
common subtype of non-Hodgkin’s lymphoma1.
The biological role of CD20 remains poorly understood; however, it is thought to be involved in
calcium ion influx5,6. 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. CD3 is an invariant antigen of the T cell TCR (T cell receptor), which is responsible
for recognizing peptides bound to major histocompatibility complex molecules.
Antigen Distribution
CD20 is widely expressed on normal B cells during all stages of
development, as well as by most B cell malignancies. CD3 is a T cell surface glycoprotein.
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 Mosunetuzumab biosimilars are typically used as calibration standards or reference controls in PK bridging ELISA to ensure accurate quantification of drug concentration in serum. The general approach is to use a single analytical standard, often the biosimilar, as the assay calibrator for both the biosimilar and innovator products during serum sample testing.
Essential context and supporting details:
The biosimilar is first characterized for analytical comparability to the innovator (reference) Mosunetuzumab through robust method qualification studies, including precision and accuracy metrics. This involves running comparative analytical procedures—such as immunochemical or electrophoretic assays—using both the innovator and biosimilar under identical conditions to ensure that their quantitation profiles are sufficiently similar.
Once bioanalytical equivalence is established (i.e., both reference and biosimilar yield comparable assay results with low variability), the biosimilar is selected as the calibration standard for the ELISA method. This standard is used to generate the standard curve: known concentrations of the biosimilar are prepared in serum matrix, enabling quantification of Mosunetuzumab concentrations in unknown test samples through interpolation against this curve.
Quality control (QC) samples, formulated with both biosimilar and innovator molecules, are quantified against the biosimilar standard curve to demonstrate the assay's accuracy, precision, and lack of bias for both products.
The ELISA method itself is often a sandwich or bridging format, wherein both the drug and the standard are bound and detected via dual specificity for the analyte. This format ensures that the assay equally recognizes Mosunetuzumab, regardless of whether it derives from the innovator or the biosimilar source.
The use of a single reference calibration standard minimizes assay variability and streamlines method validation, particularly during blinded PK or clinical studies where interchangeability and accuracy are paramount.
Regulatory guidance recommends this approach as best practice for biosimilar PK study assays, provided there is strong evidence for analytical equivalence.
Additional relevant information:
Preparation and analysis of both standard and QC samples occur in human serum matrix to best mimic clinical testing conditions and ensure matrix effects are accounted for.
Validation studies typically include a range of concentrations (e.g., 50 ng/mL to 12,800 ng/mL) and are run repeatedly by multiple analysts to confirm reproducibility and robustness.
This strategy can be adapted for other biologics beyond Mosunetuzumab and is part of harmonized bioanalytical practices in biosimilar development.
In summary, research-grade Mosunetuzumab biosimilars are used as calibration standards/reference controls in PK bridging ELISA after confirmation of analytical equivalence with the innovator product, ensuring precise and unbiased quantification of serum drug concentrations during biosimilar development and clinical studies.
The primary in vivo models for administering research-grade anti-CD20/CD3 bispecific antibodies to study tumor growth inhibition and to characterize resulting tumor-infiltrating lymphocytes (TILs) are:
Syngeneic mouse models genetically humanized for both CD3 and CD20 (double-humanized models).
Syngeneic tumors expressing human CD20 in immunocompetent mice with humanized CD3.
Essential Context and Supporting Details
1. Syngeneic Double-Humanized Mouse Model
These mice are engineered to express human forms of both CD3 (on T cells) and CD20 (on B cells or tumor cells), allowing for specific engagement by anti-CD20/CD3 bispecific antibodies (T-BsAbs) that require human antigen co-recognition.
In these models, mice can be engrafted with human CD20-expressing leukemia or lymphoma cell lines (such as E2A leukemia, A20 lymphoma), which grow robustly due to immune tolerance to human CD20.
Treatment with CD20/CD3 T-BsAbs results in dynamic changes in TIL profiles, including rapid and extensive T cell activation, redistribution of T cells to lymphoid organs and tumor tissues, and induction of anti-tumor immune responses.
These models accurately recapitulate human pharmacodynamics, including cytokine release syndrome and T cell kinetics seen in patients receiving clinical T-BsAbs such as blinatumomab.
2. A20-Human CD20 Syngeneic Mouse Model
This model utilizes the A20 murine B lymphoma line engineered to express human CD20, in syngeneic immunocompetent mice (sometimes also expressing humanized CD3) for direct evaluation of anti-CD20/CD3 efficacy in tumor growth inhibition.
The model enables detailed characterization of the tumor-immune microenvironment, including TIL activation, phenotyping, and functional analysis in response to treatment.
The anti-tumor effects—including combination therapy with immune checkpoint inhibitors—have been systematically evaluated in this model, making it a standard for preclinical TIL assessment and mechanism studies.
Additional Notes
Conventional immunodeficient xenograft models are generally unsuitable for T-BsAbs targeting human CD3, as human T cells are needed for full activity and the TIL landscape does not recapitulate an intact immune system.
Humanized immune system mice (reconstituted with human hematopoietic stem cells) have limited application compared to the double-humanized syngeneic models, especially for mechanistic TIL studies, due to remaining murine components and inconsistent human immune cell reconstitution.
Summary Table: Principal Models Used
Model Type
Mouse Immune System
Human CD20/CD3 Required?
Major Use
Key Strengths
Double-humanized syngeneic mouse (CD20/CD3)
Intact/syngeneic
Yes
Tumor growth inhibition, TIL profiling
Physiologic, predictive
Syngeneic tumor (A20-huCD20)
Intact/syngeneic
CD20 required
Efficacy, mechanism, TIL characterization
Standardized, mechanistic
Humanized immune system (HIS) mouse
Partially human
Yes
Limited for TIL due to incomplete system
Not gold-standard for TILs
These humanized syngeneic models are currently considered the most physiologically relevant and widely used for preclinical in vivo studies of anti-CD20/CD3 bispecific antibody function, tumor inhibition, and detailed TIL analysis.
Researchers explore the use of mosunetuzumab biosimilars—bispecific antibodies targeting CD20 and CD3—in combination with other immune checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3 biosimilars) to investigate possible synergistic effects in complex immune-oncology models, but most current clinical trials and preclinical models primarily report combinations with PD-1/PD-L1 inhibitors rather than CTLA-4 or LAG-3 specifically.
Context and Supporting Details:
Mosunetuzumab is a bispecific antibody that engages T cells to target malignant B cells and is approved for relapsed/refractory B-cell lymphomas. It is generally tested in combination with other agents to enhance antitumor responses or overcome limitations of monotherapies.
In early-phase clinical trials, mosunetuzumab has been combined with checkpoint inhibitors like atezolizumab (a PD-L1 inhibitor). These studies aim to harness complementary immune mechanisms: mosunetuzumab redirects T cells to tumors, while checkpoint inhibitors block immune-suppressive pathways in the tumor microenvironment. This strategy reflects a broader approach in immuno-oncology where multiple checkpoint pathways are targeted in tandem to augment T-cell activation, proliferation, and effector function.
Although direct clinical evidence for combinations with anti-CTLA-4 or anti-LAG-3 biosimilars is presently limited, the rationale for such combinations is well-established. Preclinical and clinical studies with other checkpoint combinations (such as CTLA-4 plus PD-1 inhibitors) consistently show enhanced antitumor activity compared to monotherapy, albeit with increased toxicity.
Experimental Use in Complex Models:
In complex immune-oncology models (such as mouse xenografts with human immune cells or patient-derived tumor samples), researchers simulate combinations of T-cell engagers (e.g., mosunetuzumab) and checkpoint inhibitors to study:
Synergistic antitumor activity, such as increased T-cell infiltration, cytokine production, and tumor cell killing.
Immune exhaustion reversal, where additional checkpoint blockade (CTLA-4, LAG-3) may prevent or reverse adaptive resistance that develops after initial T-cell activation.
Adverse event profiles, evaluating cytokine release syndrome, autoimmunity, or other immune-mediated toxicities.
Key Insights and Limitations:
The combination of immune checkpoint inhibitors with bispecifics is promising but can result in increased adverse events, requiring careful dose optimization and patient monitoring.
Most published data focus on mosunetuzumab with PD-1/PD-L1 blockade in clinical or experimental settings; studies specifically using anti-CTLA-4 or anti-LAG-3 biosimilars are in earlier stages or preclinical phases.
Researchers continue to develop improved mouse models, ex vivo systems, and early-phase clinical trials addressing these combinations to clarify mechanisms and clinical benefit.
In summary, while direct published results on mosunetuzumab plus anti-CTLA-4 or anti-LAG-3 biosimilars are limited, the overall strategy follows the immuno-oncology paradigm of combining bispecific T-cell engagers with diverse checkpoint blockade to probe and potentially maximize synergistic immune effects in advanced cancer models.
A Mosunetuzumab biosimilar can be used as either a capture or detection reagent within a bridging anti-drug antibody (ADA) ELISA to detect patient antibodies that may develop against the therapeutic, serving to monitor the immunogenic response to Mosunetuzumab therapy.
Context and Principle:
A bridging ADA ELISA is designed to detect antibodies in patient serum (ADAs) that specifically bind to the therapeutic drug, in this case, Mosunetuzumab.
Biosimilar Mosunetuzumab is structurally and functionally close to the original drug and is used as the antigen/reagent in these immunogenicity assays.
How the Bridging ADA ELISA Works:
The assay typically follows these steps:
Coating/Capture: The biosimilar Mosunetuzumab is immobilized on the surface of a microplate well.
Sample Incubation: Patient serum samples are added. If anti-Mosunetuzumab ADAs are present, they will bind to the immobilized Mosunetuzumab on the plate.
Detection: After washing away unbound serum proteins, a labeled (e.g., biotinylated or enzyme-conjugated) Mosunetuzumab biosimilar is added. This detection reagent binds the other arm of any ADA that is already bound to the capture Mosunetuzumab, forming a "bridge".
Signal Generation: Following binding, the presence of the detection reagent is visualized using a substrate that yields a measurable signal (such as colorimetric change in the case of HRP/TMB), indicating the presence of anti-Mosunetuzumab ADA in the patient’s serum.
Why Use a Biosimilar?
The biosimilar must have identical immunoreactive epitopes to the reference product to ensure all clinically relevant ADAs are captured and detected.
These reagents are validated for in vitro testing and not for clinical diagnostic or therapeutic use.
Key Details:
Both the capture and detection Mosunetuzumab in this format must be non-overlapping in tags/labels to avoid artifact bridging.
The accuracy of ADA measurement depends on the quality, batch consistency, and purity of the biosimilar used.
Summary Table: Key Roles of Mosunetuzumab Biosimilar in Bridging ADA ELISA
Reagent Role
Function
Requirement
Capture
Coated on plate, binds patient ADA if present
Must represent reference drug epitope
Detection
Labeled (e.g., biotin/HRP), binds other arm of ADA, enabling “bridging” and signal readout
Must not cross-react with label or substance
This approach allows sensitive and specific monitoring of patients' immune responses against Mosunetuzumab by identifying anti-drug antibodies, critical for assessing the immunogenic risk of the therapeutic.
References & Citations
1 Kang C. Drugs. 82(11):1229-1234. 2022.
2 Kaplon H, Crescioli S, Chenoweth A, et al. MAbs. 15(1):2153410. 2023.
3 Tavarozzi R, Manzato E. Antibodies (Basel). 11(1):16. 2022 Feb 21;11(1):16.
4 Ang Z, Paruzzo L, Hayer KE, et al. Blood. Sep 8:blood.2023020400. 2023.
5 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.
6 Freeman CL, Sehn LH. Br J Haematol. 182(1):29-45. 2018.
7 Hernandez G, Huw LY, Belousov A, et al. Blood. 134(Suppl 1):1585. 2019.
8 Budde LE, Assouline S, Sehn LH, et al. J Clin Oncol. 40(5):481-491. 2022.
9 Bartlett NL, Assouline S, Giri P, et al. Blood Adv. 7(17):4926-4935. 2023.