Anti-Human CD20 (Obinutuzumab) [Clone GA101] — APC

Anti-Human CD20 (Obinutuzumab) [Clone GA101] — APC

Product No.: LT907

- -
- -
Product No.LT907
Clone
GA101
Target
CD20
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Obinutuzumab, CD20, MS4A1
Isotype
Human IgG1κ
Applications
ELISA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
Immunogen
Human lymphoblastoid cell line SB.
Product Concentration
0.2 mg/ml
Formulation
This Allophycocyanin (APC) conjugate is formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.4, 1% BSA and 0.09% sodium azide as a preservative.
State of Matter
Liquid
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
Excitation Laser
Red Laser (650 nm)
Applications and Recommended Usage?
Quality Tested by Leinco
FC,
ELISA
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.

Role of Obinutuzumab Biosimilars in PK Bridging ELISA

Research-grade Obinutuzumab biosimilars—molecules highly similar but not identical to the reference drug—can be used as calibration standards (reference controls) in pharmacokinetic (PK) bridging ELISA assays designed to measure Obinutuzumab concentration in serum samples. Here’s how this process typically works, based on current bioanalytical practice:

Preparation and Characterization

  • Calibration Standards: Biosimilars are dissolved at known concentrations in the same matrix as the unknown samples (typically human serum). These standards create a calibration curve against which unknown samples are measured.
  • Reference Controls: Alongside standards, additional control samples (quality controls, QCs) are prepared at specified concentrations to monitor assay performance across runs.
  • Method Qualification and Validation: Before routine use, the assay must be qualified and validated to ensure that the biosimilar standards behave comparably to the reference product in terms of sensitivity, accuracy, precision, and dynamic range.
  • Single Standard Approach: For bridging assays supporting biosimilar product development, the consensus is to use a single analytical standard (often the biosimilar itself) for both calibration and quality control, provided that bioanalytical comparability has been demonstrated.

Assay Execution

  • Plate Setup: Each microplate includes both calibration standards (prepared from the biosimilar) and QC samples, run alongside the clinical samples.
  • Calibration Curve: Absorbance data from the standards are used to generate a calibration curve, typically using a 4-parameter logistic function. Unknown sample concentrations are interpolated from this curve.
  • Cross-Validation: To ensure reliability, the assay should be validated using both the biosimilar and the reference product at several concentrations, confirming that both generate equivalent results within the assay’s dynamic range.

Quality Assurance

  • Bioanalytical Comparability: The biosimilar’s performance as a standard should be statistically equivalent to the reference product within pre-specified margins (e.g., 90% confidence interval within [0.8, 1.25]).
  • Robustness: The assay must be robust, reproducible, and minimally affected by inter-operator or inter-laboratory variation. Strict adherence to protocols is essential for meaningful results.

Summary Table

ComponentRole in PK Bridging ELISAExample References
Biosimilar StandardGenerates calibration curve for concentration interpolation
Reference ControlsMonitors assay precision and accuracy each run
Validation SamplesConfirms biosimilar and reference comparability in the assay matrix
Quality ControlsAssess run-to-run variability and assay validity

Practical Considerations

  • Specificity: The assay must specifically detect Obinutuzumab and not cross-react with other serum proteins or ADA (anti-drug antibodies).
  • Sample Preparation: Serum samples may need dilution to fall within the assay’s linear range.
  • Assay Type: Most commercial ELISA kits are colorimetric and quantitative, designed for research use only, not clinical diagnostics.
  • Storage and Stability: Standards and kits must be stored under appropriate conditions to maintain activity.

Conclusion

Research-grade Obinutuzumab biosimilars serve as robust calibration standards and reference controls in bridging ELISA for PK studies, provided their analytical comparability to the reference product is thoroughly validated. This approach minimizes variability, supports regulatory requirements for biosimilarity assessment, and enables reliable measurement of drug concentrations in clinical serum samples.

Flow cytometry protocols using conjugated obinutuzumab biosimilars to validate CD20 expression and binding capacity typically follow standardized procedures that have been validated in research settings. Based on established methodologies, here are the key protocols:

Cell Preparation and Staining Protocol

Sample PreparationThe standard protocol begins with harvesting target cells (such as B-cell lymphoma lines or primary B cells) and washing them in phosphate-buffered saline (PBS) containing 2% fetal bovine serum. Cells should be counted and adjusted to approximately 1×10^6 cells per test to ensure consistent results.

Antibody Conjugation and StainingFor PE or APC-labeled obinutuzumab biosimilars, cells are typically incubated with the conjugated antibody at 4°C for 30 minutes in the dark. The standard concentration ranges from 1-10 μg/ml, though titration experiments should be performed to determine optimal concentrations for each specific cell line. After incubation, cells are washed twice with cold PBS to remove unbound antibody.

Validation Controls and Analysis

Essential ControlsThe protocol requires several critical controls: unstained cells (autofluorescence control), isotype control antibody (typically mouse IgG1-PE or IgG1-APC), and cells stained with unconjugated obinutuzumab followed by secondary detection antibody. These controls help distinguish specific binding from non-specific interactions.

Flow Cytometric AnalysisData acquisition should collect at least 10,000 events per sample. The analysis focuses on forward scatter (FSC) and side scatter (SSC) to gate viable cells, followed by fluorescence intensity measurement in the appropriate channel (PE: ~575 nm, APC: ~660 nm). Results typically show a distinct population shift in CD20-positive cells compared to negative controls.

Binding Affinity Validation

Quantitative AssessmentTo validate binding capacity, saturation binding experiments can be performed using serial dilutions of the conjugated obinutuzumab biosimilar. This allows calculation of the dissociation constant (K_D) and maximum binding capacity (B_max). Research has demonstrated that obinutuzumab maintains high binding affinity with CD20-expressing cells, with K_D values typically in the low nanomolar range.

Specificity ConfirmationBinding specificity is confirmed through competitive inhibition assays, where cells are pre-incubated with excess unconjugated obinutuzumab before adding the fluorescent conjugate. True specific binding should be significantly reduced under these conditions.

Quality Control Parameters

Antibody ValidationEach new lot of conjugated obinutuzumab biosimilar should be validated against reference standards. The protocol includes testing antibody stability, confirming appropriate fluorophore conjugation ratios, and verifying consistent binding patterns across known CD20-positive and negative cell lines.

Data InterpretationResults are typically expressed as mean fluorescence intensity (MFI) ratios comparing specific to non-specific binding, or as percentage of positive cells above the isotype control threshold. Strong CD20 expression typically shows MFI ratios >10-fold above background, while weak expression may show 2-5 fold increases.

These protocols provide robust validation of both CD20 expression levels and the binding capacity of obinutuzumab biosimilars, making them valuable tools for both research applications and clinical diagnostic development.

Biopharma companies typically employ a comprehensive suite of analytical assays to confirm the structural and functional similarity of a proposed biosimilar to its originator (reference) drug. These assays span physicochemical, structural, and functional testing, and are backed by regulatory expectations such as those outlined in ICH Q6B.

Analytical Assays for Biosimilar Characterization

  • Structural Characterization:

    • Primary structure: Peptide mapping, mass spectrometry, and amino acid analysis to verify the exact amino acid sequence.
    • Higher-order structure: Circular dichroism, NMR spectroscopy, and X-ray crystallography to assess secondary, tertiary, and quaternary structures.
    • Post-translational modifications: Glycosylation, phosphorylation, oxidation, and deamidation profiles using advanced mass spectrometry and chromatographic methods.
    • Impurity and variant profiling: Size-exclusion chromatography, capillary electrophoresis, and SDS-PAGE to detect aggregates, fragments, host cell proteins, and process-related impurities.
  • Physicochemical Analysis:

    • Techniques like isoelectric focusing and differential scanning calorimetry for profiling charge variants and thermal stability.
    • Chromatographic methods including HPLC and UPLC for purity and impurity assessment.
  • Functional and Biological Assays:

    • Potency assays (often cell-based), assessing the biosimilar’s ability to elicit expected biological activities.
    • Binding assays (e.g., ELISA, surface plasmon resonance) measure binding affinity/specificity to targets (such as antigens, receptors, or immune components like Fc receptors).
    • Enzyme kinetics and other mechanism-specific assays.
    • Orthogonal methods—multiple, complementary techniques are often used for higher confidence.
  • Comparability Approach:

    • Head-to-head comparative studies are essential: the biosimilar is directly compared with the reference product across all critical attributes.
    • Analytical similarity is judged by whether the biosimilar falls within the defined range for each critical quality attribute (CQA) set by the originator.

Role of Leinco Biosimilars in Analytical Studies

In the context of biosimilar testing, Leinco develops and provides biosimilar reference materials, which can be used as comparators or controls in analytical assays. While the provided search results do not specify the precise use of "Leinco biosimilar" in these studies, standard industry practice is as follows:

  • A commercially available biosimilar, like those provided by Leinco, can serve as an analytical benchmark for developing new biosimilars or validating assays.
  • Such materials might be used in side-by-side comparisons to demonstrate assay performance or to provide alternative reference points if the originator product is scarce or unavailable.

However, the primary comparator in regulatory biosimilarity studies must always be the original reference biological product, not another biosimilar. Leinco's products are more typically used for research, assay development, or as secondary standards—not as a direct substitute for the originator in regulatory submissions.

Summary Table: Typical Assays in Biosimilar Comparison

AttributeExample AssaysUse
Primary structurePeptide mapping, Mass spectrometrySequence confirmation
Glycosylation/post-trans.Mass spec, HILIC, N-glycan profilingGlycan structure/heterogeneity
Higher-order structureCD, NMR, DSCFolding and conformational check
Purity, AggregatesSEC, Capillary electrophoresis, SDS-PAGEDetect impurities/aggregates
Biological ActivityCell-based potency assay, Enzyme kineticsConfirm functional equivalence
Binding characteristicsELISA, SPR, Fc receptor assaysAffinity/specificity to target

According to the available information, Leinco biosimilars can be used as research tools, for assay validation, or for developing analytical methods, but the main regulatory comparability studies always require the originator product as the benchmark.

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
Flow Cytometry

Certificate of Analysis

Formats Available

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