Anti-Human CD20 (Obinutuzumab) [Clone GA101] — Fc Muted™

Anti-Human CD20 (Obinutuzumab) [Clone GA101] — Fc Muted™

Product No.: LT912

- -
- -
Product No.LT912
Clone
GA101
Target
CD20
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Obinutuzumab, CD20, MS4A1, 949142-50-1
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 lymphoblastoid cell line SB.
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.
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 Obinutuzumab. This product is for research use only. Obinutuzumab (GA101) activity is directed against human CD20.
Background
CD20 is a nonglycosylated 33-37 kDa phosphoprotein 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. Anti-CD20 mAbs bind to the 44 amino acid extracellular portion.

Obinutuzumab (GA101) is a new generation, type II, anti-CD20 antibody2. Obinutuzumab was humanized by grafting the complementarity-determining sequences of murine IgG1-κ antibody B-Ly1 onto human VH and VL acceptor frameworks3. The Fc segment was glycoengineered to attach bisected, complex, nonfucosylated oligosaccharides to asparagine 297, leading to increased affinity to FcgRIII.

Obinutuzumab causes homotypic adhesion4,5,6, induces direct cell death via largely caspase-independent mechanisms4,6,7,8,9, does not localize into lipid rafts4,10,11, displays half-maximal CD20 binding at saturating conditions7, and displays minimal complement dependent cytotoxicity7.

Compared to rituximab, obinutuzumab recognizes a distinct but overlapping CD20 epitope, in a different orientation that results in increased pro-apoptotic potential12,13,14. A modified elbow-hinge residue, characterized by a leucine to valine mutation at Kabat position 11, is key to superior phosphatidylserine exposure and cell death relative to rituximab3.
Antigen Distribution
CD20 is a general B cell marker expressed by the majority of normal B cells in all stages of their development as well as by most B cell malignancies.
Ligand/Receptor
Src family tyrosine kinases, MHC class I, II, CD53, CD81, CD82
PubMed
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immunology
.
Oncology

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.

Research-grade Obinutuzumab biosimilars are commonly used as calibration standards and reference controls in pharmacokinetic (PK) bridging ELISAs to accurately measure drug concentrations in serum samples in biosimilar development.

In a PK bridging ELISA, the analytical procedure typically involves:

  • Calibration standard preparation: Quantitative ELISA kits for Obinutuzumab biosimilars contain lyophilized biosimilar protein which is reconstituted to prepare a series of known concentrations, forming a standard curve across the assay’s validated range (for example, 0.31–5 μg/mL or 93.75 ng/mL–6000 ng/mL).
  • Reference controls: Quality control (QC) samples, prepared using both the biosimilar and reference product (originator monoclonal antibody), are included to ensure comparability. In validated methods, these QC samples are assessed for precision and accuracy by being measured across the assay and compared to the calibration curve established with the biosimilar standard.

Key rationale and usage:

  • According to industry consensus and regulatory guidance, best practice is to use a single PK ELISA method employing a single analytical standard (usually the biosimilar) as the calibrator for quantifying both the biosimilar and reference product in serum samples. This approach reduces analytical variability and eliminates the need for separate assay methods for each product.
  • The method is rigorously validated, including statistical comparison of biosimilar and reference products within the assay, ensuring bioanalytical equivalence. Analytical equivalence is typically defined by comparing measured values to predefined equivalence intervals (such as 0.8 to 1.25 for the ratio of biosimilar to reference product concentrations).
  • Calibration standards (biosimilar) and QC samples (both biosimilar and reference) are prepared in human serum at multiple concentrations spanning the assay range, enabling accurate quantification of drug concentrations in actual study samples.
  • Research-grade biosimilars (for example, lyophilized Obinutuzumab in ELISA kits) are supplied for research use only, not for therapeutic or diagnostic use.

Summary table – Uses in PK bridging ELISA:

MaterialRole in PK ELISA
Biosimilar (Obinutuzumab) standardCalibration curve, quantification of drug levels
Biosimilar QC sampleAccuracy/precision validation, reference control
Reference (originator) QC sampleAssay comparability, equivalence validation
Serum samples (test)Measurement of drug concentration

This approach ensures robust, comparable, and regulatory-compliant measurement of Obinutuzumab in clinical PK studies with biosimilar candidates.

The primary preclinical models used to study in vivo administration of research-grade anti-CD20 antibodies for tumor growth inhibition and analysis of tumor-infiltrating lymphocytes (TILs) are syngeneic mouse tumor models engineered to express human CD20 and models utilizing murine-specific anti-CD20 antibodies in fully immunocompetent mice.

Key model types:

  • Syngeneic Mouse Models Expressing Human CD20

    • Example: A20 or EL4 murine lymphoma cell lines genetically engineered to express human CD20, used in immunocompetent syngeneic mice. This allows for testing anti-CD20 antibodies (including clinically relevant or research-grade variants) and for collecting tumors to study both tumor growth inhibition and immune infiltrates (TILs) using mouse-specific reagents.
    • These models can be used for both solid and hematological malignancies by stably transducing a mouse tumor cell line with human CD20, e.g., EL4-CD20 in C57BL/6 mice.
    • The approach allows for assessment of both antibody anti-tumor efficacy and changes in lymphocyte subsets within tumors.
  • Syngeneic Solid Tumor Models with Murine-specific Anti-CD20 Antibodies

    • Some studies utilize anti-mouse CD20 antibodies (that target endogenous mouse B cells but not tumor cells directly) to deplete B cells in syngeneic tumor models such as TC1 (mouse lung cancer), RENCA (renal carcinoma), CT26 (colon carcinoma) in immunocompetent mice. This enables investigation into how B-cell depletion affects tumor growth, immune microenvironment, and specifically the composition and activity of TILs.
    • Example: A newly developed anti-mouse CD20 antibody used in mice bearing TC1 tumors significantly altered the CD8+ TIL compartment and enhanced immunotherapy responses.
    • These models inform on immunomodulatory roles of B cells and their interaction with T cell immunity in the tumor.
  • Humanized Mouse Models

    • These are more rarely used for anti-CD20 due to complexities in establishing a functional human immune system within mice, and the need for cross-reactivity of antibody and human immune components. Most published work relevant to your question focuses on syngeneic models for mechanistic studies. When humanized models are used, they're primarily in the context of hematologic malignancies or for pharmacokinetic/toxicity studies.
Model TypeAntibody TargetTumor CellsHost ImmunityTypical Use
Syngeneic (huCD20+)Human CD20Mouse tumor cells (e.g., A20, EL4) expressing huCD20Fully immunocompetent mouseStudy antibody efficacy & TIL profiling
Syngeneic (muCD20)Mouse CD20Mouse tumor cells (natural or engineered)Fully immunocompetent mouseStudy B cell depletion & TIL impact
Humanized mouseHuman CD20Human lymphoma/leukemia cellsPartially reconstituted mouseMechanism/pharmacology, limited for TIL studies
  • Syngeneic models with engineered CD20 expression are the gold standard for mechanistic and TIL-compartment studies due to their fully functional immune systems, which is critical for accurate TIL characterization and tumor-immune interaction assessment.

  • Common readouts include tumor growth delay/regression and flow cytometric or immunohistochemical quantification of TIL subsets (CD8+, CD4+, myeloid cells, etc.) in the tumor microenvironment.

References:Describes use of A20-huCD20 in syngeneic models for tumor inhibition and TIL assessment.Uses EL4-huCD20 cells in a syngeneic mouse model to study anti-CD20 mechanisms, tumor cell killing, and effector functions involving TILs.Describes anti-mouse CD20 in syngeneic solid tumor models for studying B-cell depletion effects on TILs.Reviews immune profiling across multiple syngeneic models, including effects of immunotherapies on TILs.

Researchers use Obinutuzumab biosimilars in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to evaluate synergistic antitumor effects within complex immune-oncology models, primarily through both preclinical (in vitro and animal) studies and early-phase clinical trials.

Key approaches in synergy studies:

  • Preclinical Models: Researchers employ animal models (e.g., human lymphoma xenografts, mouse colon adenocarcinoma) to test the ability of obinutuzumab biosimilars to enhance immune-mediated tumor killing. These models often include dual or multi-agent regimens—for instance, obinutuzumab combined with chemotherapy or other monoclonal antibodies (mAbs) targeting immune checkpoints like CTLA-4 or PD-1.

  • Mechanistic Rationale: Combining agents that target distinct immune regulatory pathways (e.g., CD20 on B cells [obinutuzumab] and CTLA-4 or LAG-3 on T cells) is hypothesized to act on different stages or locations in the immune response. For example:

    • Obinutuzumab biosimilars deplete B cells by binding CD20, enhancing antibody-dependent cellular cytotoxicity (ADCC) and direct cell death.
    • Checkpoint inhibitors (e.g., anti-CTLA-4, anti-LAG-3) release the "brakes" on T cells by blocking inhibitory signals, facilitating both T cell priming (lymph nodes) and effector function (tumor microenvironment).
    • Combined, these can foster both improved tumor antigen presentation (via B cell depletion) and more potent T cell-mediated cytotoxicity, extending the anti-cancer immune response.
  • Evaluation Metrics: In these studies, researchers measure:

    • Tumor growth inhibition (primary endpoint in animal models)
    • Immune cell infiltration and activation, such as increased cytotoxic T cells and reduced regulatory B/T cells in tumor tissue
    • Survival rates in treated animals or patient-derived xenografts
    • Toxicity profiles, since combination immunotherapy often increases adverse event incidence
  • Clinical Translation: Findings from preclinical models inform the design of early-phase clinical trials that assess combined efficacy and safety in patients with refractory malignancies. For example, in advanced melanoma, dual checkpoint blockade (CTLA-4 plus PD-1/PD-L1) yields superior survival but heightened toxicity—the same synergy logic underpins newer combinations, including those integrating anti-CD20 antibodies like obinutuzumab.

Study design considerations:

  • Researchers select biosimilar antibodies for cost-effectiveness and reproducibility across experimental conditions.
  • The complexity of immune-oncology models—especially humanized mouse models—allows simultaneous assessment of B cell and T cell-directed therapeutics under controlled microenvironment settings.

Limitations and ongoing research:

  • Most published data involve combinations of checkpoint inhibitors with chemotherapy or with PD-1/CTLA-4 blockade; studies directly pairing obinutuzumab biosimilars with anti-CTLA-4 or anti-LAG-3 biosimilars are emerging, but peer-reviewed results remain limited.
  • Toxicity remains a major concern, making translational studies crucial for optimizing dose and schedule.

In sum, researchers leverage obinutuzumab biosimilars together with checkpoint inhibitors in controlled models to investigate multi-faceted immune activation, aiming to potentiate tumor clearance while monitoring safety—laying the groundwork for rational clinical combination strategies in immune-oncology.

In the context of immunogenicity testing, a biosimilar of obinutuzumab, like other therapeutic antibodies, can be used as part of a bridging ELISA to monitor a patient's immune response against therapeutic drugs. Here's how it might be applied:

Bridging ELISA Overview

A bridging ELISA is a diagnostic tool used to detect anti-drug antibodies (ADAs), which are antibodies that a patient's immune system produces in response to therapeutic drugs, including monoclonal antibodies like obinutuzumab.

Use of Obinutuzumab Biosimilar in Bridging ELISA

  1. Preparation of the Assay:

    • Capture Reagent: One form of obinutuzumab biosimilar is biotinylated and used to coat streptavidin plates. This acts as the capture reagent.
    • Detection Reagent: A different form of the obinutuzumab biosimilar, typically labeled with a dye or enzyme like horseradish peroxidase (HRP), is used as the detection reagent.
  2. Sample Preparation and Assay Process:

    • Sample Addition: Serum samples from patients are added to the wells containing the biotinylated obinutuzumab biosimilar. If the patient has developed ADAs against obinutuzumab, these antibodies will bind to the captured drug.
    • Detection: The labeled obinutuzumab biosimilar is then added to detect the bound ADAs. If ADAs are present, they will bind both the captured and labeled drug molecules, forming a "bridge" that is essential for detection.
  3. Detection and Analysis:

    • The presence of ADAs is detected through a colorimetric reaction using a chromogenic substrate like TMB. The intensity of the color produced is proportional to the amount of ADAs present in the sample.
  4. Advantages and Considerations:

    • Advantages: Bridging ELISA is highly sensitive and allows for high-throughput screening of samples.
    • Considerations: The specificity of the assay can be affected by matrix components in serum or by the presence of high drug concentrations, emphasizing the need for high-quality reagents and careful assay optimization.

Conclusion

Using a biosimilar of obinutuzumab in a bridging ELISA provides a precise method to monitor patient immune responses by detecting ADAs, which is crucial for assessing the efficacy and safety of therapeutic antibodies. However, specific technical details about using obinutuzumab or its biosimilar in this context would typically be established based on the specific requirements and protocols of the implementing laboratory.

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. Mössner E, Brünker P, Moser S, et al. Blood. 115(22):4393-4402. 2010.
4. Chan HT, Hughes D, French RR, et al. Cancer Res. 63(17):5480-5489. 2003.
5. Ivanov A, Beers SA, Walshe CA, et al. J Clin Invest. 119(8):2143-2159. 2009.
6. Alduaij W, Ivanov A, Honeychurch J, et al. Blood. 117(17):4519-4529. 2011.
7. Herter S, Herting F, Mundigl O, et al. Mol Cancer Ther. 12(10):2031-2042. 2013.
8. Honeychurch J, Alduaij W, Azizyan M, et al. Blood. 119(15):3523-3533. 2012.
9. Golay J, Zaffaroni L, Vaccari T, et al. Blood. 95(12):3900-3908. 2000.
10. Cragg MS, Morgan SM, Chan HT, et al. Blood. 101(3):1045-1052. 2003.
11. Cragg MS, Glennie MJ. Blood. 103(7):2738-2743. 2004.
12. Niederfellner G, Lammens A, Mundigl O, et al. Blood. 118(2):358-367. 2011.
13. Klein C, Lammens A, Schäfer W, et al. MAbs. 5(1):22-33. 2013.
14. Könitzer JD, Sieron A, Wacker A, Enenkel B. PLoS One. 10(12):e0145633. 2015.
15. Terszowski G, Klein C, Stern M. J Immunol. 192(12):5618-5624. 2014.
16. Bologna L, Gotti E, Manganini M, et al. J Immunol. 186(6):3762-3769. 2011.
17. Ysebaert L, Laprévotte E, Klein C, Quillet-Mary A. Blood Cancer J. 5(11):e367. 2015.
18. Cartron G, Hourcade-Potelleret F, Morschhauser F, et al. Haematologica. 101(2):226-234. 2016.
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.