Humanized antibody derived from mouse clone targeting CD79b
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 Polatuzumab but is not linked to MMAE. This product is for research use only. Polatuzumab antibody activity is directed against CD79b.
Background
CD79 is a covalent heterodimer, composed of CD79a and CD79b, that acts as the signaling component of the B cell receptor (BCR) 1 and is also a tumor associated antigen 2. CD79 together with surface Ig forms the BCR complex, and cross-linking of BCR triggers downstream signaling that can lead either to apoptosis or, when rescue signals from T cells are present, cell activation 1. Cross-linked BCR is internalized and targeted by a lysosome-like compartment, the major histocompatibility complex class II positive compartment, making CD79b a target antigen for antibody drug conjugates (ADC) against cancerous B cells 1.
Polatuzumab is an ADC composed of an antibody directed against CD79b on B cells covalently bound via a cleavable linker to Microtubule-disrupting anti-mitotic agent monomethyl auristatin (MMAE) 2, an apoptosis stimulant that inhibits mitosis, tubulin, and tubulin polymerization 2.
This research grade biosimilar has the same specificity as the original therapeutic antibody but lacks the conjugated MMAE drug.
Polatuzumab was generated by immunizing mice with the extracellular domain of CD79b 1, 4. Recombinant technology was then used to humanize the anti-CD79b antibody and sequence optimize it.
Polatuzumab has been approved for the treatment of some adults with relapsed/refractory diffuse large B cell lymphoma (DLBCL) and various clinical trials are in progress 1.
Antigen Distribution
CD79b is expressed on the majority of B cells and is moderately to
strongly expressed in a majority of malignant lymphomas, including almost all non-Hodgkin
lymphoma.
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 Polatuzumab biosimilars are commonly used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays to determine the concentration of Polatuzumab (or its biosimilar) in serum samples from preclinical or clinical studies.
Essential context and use in PK bridging ELISA:
ELISA Format: In a typical PK bridging ELISA, anti-idiotypic antibodies (specific for Polatuzumab's unique variable region) are used as both capture and detection reagents to ensure high specificity for the drug molecule. The capture antibody is coated on the plate, and after sample/drug binding, an HRP-labeled anti-idiotypic detection antibody is added to generate a measurable signal.
Calibration Standards: Research-grade biosimilar Polatuzumab preparations serve as the calibrator—serially diluted to generate a standard curve spanning the expected drug concentration range in study samples. This standard curve allows for quantification of unknown serum concentrations by comparison of their signal to the curve.
Reference Controls: Parallel to standards and patient samples, reference controls (low, medium, high concentration samples, often spiked with known amounts of Polatuzumab biosimilar) are run on each plate to ensure assay accuracy, precision, and reproducibility across batches and over time.
Why Use Biosimilars: Research-grade biosimilar versions of Polatuzumab are structurally and functionally comparable to the clinical drug, and thus their use avoids the need to use the commercial clinical product (which may be limited or regulated), while providing a reliable, consistent material for assay standardization.
Summary Table: Use of Research-Grade Polatuzumab Biosimilars in PK Bridging ELISA
Role
Function in ELISA
Rationale
Calibration Standard
Generate standard curve for quantification
Ensures accurate, traceable measurement
Reference Control
Monitor run/plate performance and assay consistency
Confirms assay reliability and precision
Substitute for Clinical Drug
Used in place of commercial/clinical material
Readily available, consistent, non-regulated
Additional Details:
Assay Validation: The ELISA must be validated for parallelism (the biosimilar standard behaves identically to native drug in matrix) and cross-reactivity (the assay does not detect endogenous or unrelated antibodies).
Critical Considerations: While minor differences in post-translational modifications between biosimilar and reference product may exist, a properly validated assay confirms that these do not impact assay performance in the PK/bridging format.
Common Practice: The use of biosimilars for assay development and PK assessment is well established in industry, especially for drugs where clinical supply is restricted or costly.
Illustrative Example:
Bio-Rad describes this process generically: “Biosimilars of monoclonal antibody drugs [are used] for research use in bridging ELISAs as reference standard or capture and detection reagents”.
In conclusion, research-grade Polatuzumab biosimilars are essential standard materials for PK bridging ELISAs, acting both as calibration standards for quantitation and as reference controls for quality assurance, ensuring accurate measurement of drug concentrations in serum samples throughout preclinical and clinical studies.
The primary in vivo models for administering research-grade anti-CD79b antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are xenograft models reconstituted with human immune cells (humanized models) and, less commonly, syngeneic mouse models.
Key models include:
Humanized Xenograft Models:
These models involve immunodeficient mice (such as NOG or NSG strains) engrafted with human tumor cells (e.g., human B-cell non-Hodgkin lymphoma lines) and reconstituted with human immune cells, typically human peripheral blood mononuclear cells (PBMCs) or CD34+ hematopoietic stem cells.
Example: An anti-CD79b/CD3 bispecific antibody (IBI38D9-L) was studied in NOG mice engrafted with both human PBMCs and human B-NHL tumor cells. This approach allowed assessment of both tumor inhibition and TIL phenotype, showing eradication of B-NHL tumors and robust infiltration of activated human T cells into the tumor tissue. Similar approaches are used for bispecific and antibody-drug conjugate (ADC) anti-CD79b therapies.
Syngeneic Mouse Models (limited use for anti-CD79b):
Traditional syngeneic models involve transplanting murine lymphoma cells (expressing endogenous CD79b) into immunocompetent mice with the same genetic background. These provide a fully functional mouse immune system and can support TIL analysis. However, unless a mouse-reactive anti-CD79b antibody is used, these models are less common for human antibody evaluation due to species specificity.
Syngeneic models are most widely used to test general immunotherapeutic mechanisms but are less directly applicable for research-grade human anti-CD79b antibodies (which typically do not cross-react with mouse CD79b).
Traditional Human Xenograft Models:
These models implant human lymphoma cells into immunodeficient mice without human immune reconstitution. Tumor growth inhibition can be assessed, but analysis of TILs is not feasible because the host is immunodeficient.
Summary Table of Models:
Model Type
Antibody Tested
Immune Context
TIL Study Feasible?
References
Humanized xenograft (NOG/NSG + PBMC)
Human anti-CD79b
Human immune cells + tumor
Yes
Syngeneic (mouse tumor in mouse)
Mouse anti-CD79b
Mouse immune system + tumor
Yes
Standard human xenograft (no reconst.)
Human anti-CD79b
No functional immune system
No
Suitability for TIL Analysis:
Humanized xenograft models are the gold standard for characterizing TILs in response to human anti-CD79b antibody intervention, allowing study of both tumor inhibition and immune landscape changes at the tumor site.
Syngeneic tumor models are suitable for murine immunotherapy studies but generally not used for characterizing human anti-CD79b antibodies, unless mouse cross-reactivity is engineered.
Additional Details:
In preclinical studies, both unconjugated and ADC-conjugated anti-CD79b antibodies have shown efficacy in xenograft models, including triggering immune-mediated tumor cell death.
The infiltration of activated T cells within tumors following anti-CD79b therapy can be robustly characterized in humanized models.
For research specifically requiring analysis of TIL phenotypes after anti-CD79b treatment, humanized xenograft models provide the most relevant and widely used preclinical platform.
Researchers use Polatuzumab biosimilars in combination with checkpoint inhibitors—such as anti-CTLA-4 or anti-LAG-3 biosimilars—to investigate whether these agents produce synergistic antitumor effects in complex immune-oncology models. While clinical studies have established Polatuzumab vedotin's efficacy primarily in combination with chemotherapy agents like bendamustine and rituximab in B-cell lymphomas, the mechanistic rationale for combining multiple immunotherapy agents, including checkpoint inhibitors, is grounded in their complementary immune-activating properties.
Context and Experimental Approach:
Polatuzumab vedotin is an antibody-drug conjugate targeting CD79b, a protein expressed on B-cell lymphomas, leading to direct cytotoxicity against malignant B cells.
Anti-CTLA-4 acts in lymph nodes to enhance activation and proliferation of T cells.
Anti-LAG-3 and Anti-PD-1 act at the tumor periphery, supporting sustained cytotoxic T-cell activity and reducing regulatory T-cell suppression.
Synergistic Investigation in Immune-Oncology Models:
Researchers typically use preclinical mouse models or ex vivo human tumor cultures to combine Polatuzumab biosimilars with checkpoint inhibitors. The goal is to determine whether dual targeting of B-cell malignancy (via Polatuzumab) and T-cell modulation (via checkpoint inhibitors) results in more robust tumor regression than either agent alone.
Combinations like anti-PD-1/CTLA-4 and anti-PD-1/LAG-3 have exhibited distinct immune cell activation mechanisms; for instance, anti-PD-1/LAG-3 requires CD4 T-cell presence to reduce regulatory T-cell (Treg) activity and indirectly boost cytotoxic CD8 T-cell responses, while anti-PD-1/CTLA-4 leads to direct activation of more CD8 T cells.
Summary of Mechanistic Studies:
Combining multiple agents often exploits non-overlapping mechanisms of action. Polatuzumab induces B-cell lymphoma cell death, potentially increasing antigen availability for uptake by dendritic cells and subsequent presentation to T cells.
Checkpoint inhibitors then reinvigorate these T cells, enhancing their cytotoxic function, sometimes requiring specific subsets (CD4 or CD8) depending on the combination chosen.
Key Experimental Outcomes Researchers Measure:
Tumor regression and response rates
Progression-free survival (PFS) and overall survival (OS) in animal models
Activation status of CD4/CD8 T cells and reduction of Treg activity
Immunophenotyping of tumor-infiltrating lymphocytes via flow cytometry and histology
Clinical translation is in early stages, but the available preclinical evidence suggests that combining Polatuzumab biosimilars with checkpoint inhibitors may overcome monotherapy limitations and drive more potent antitumor immune responses. There are not yet published studies explicitly combining Polatuzumab with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3, but the mechanistic rationale, based on their distinct and complementary immune effects, supports ongoing and future investigations in this direction.
A Polatuzumab biosimilar can be used as the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor immune responses to Polatuzumab by exploiting the bivalent nature of ADAs, which typically bind to two identical epitopes on the therapeutic antibody.
In a bridging ADA ELISA designed for a biosimilar such as Polatuzumab:
Both the capture and detection reagents are the Polatuzumab biosimilar (the actual biosimilar protein, not a different anti-idiotype antibody). One preparation is typically biotinylated for immobilization (capture) on a streptavidin-coated plate; the second is usually labeled (e.g., with HRP or digoxigenin) for detection.
Patient serum is incubated with both the biotinylated and labeled biosimilar drug. If ADAs are present, their two binding arms allow them to form a "bridge" between the capture drug and the detection drug, creating a drug–ADA–drug sandwich complex.
The biosimilar serves as a close structural match to the reference drug, which is essential for sensitive, specific, and unbiased ADA detection in the context of biosimilar immunogenicity studies.
Protocol Details:
Biotinylated Polatuzumab biosimilar is first added to a streptavidin-coated well to capture it on the solid phase.
Patient sample (serum or plasma) is then added. If anti-Polatuzumab ADAs are present, they bind to the immobilized drug.
Detection reagent (Polatuzumab biosimilar conjugated with a reporter such as HRP or digoxigenin) is added, binding the other arm of ADA, forming the “bridge”.
Detection is via chromogenic substrate (e.g., TMB for HRP), generating a signal proportional to the amount of ADA present.
Why use the biosimilar as both capture and detection reagent?
In biosimilar immunogenicity studies, regulatory guidelines permit use of the biosimilar as the reagent to ensure the assay is sensitive to any ADA response specific to the biosimilar.
If the reference product and biosimilar are highly similar, a single validated assay using the biosimilar is acceptable, simplifying assay development and validation.
This approach is especially relevant for demonstrating that the biosimilar does not provoke immunogenicity different from the reference product.
Special notes:
The design must minimize drug interference (the presence of drug masking ADA).
Some protocols may also use a two-antigen strategy (reference and biosimilar, tested separately) in comparative trials, but the single-antigen (biosimilar-based) format is common and accepted for routine monitoring.
References to other formats:
Alternative ADA detection (e.g., using immunocapture LC/MS) can also utilize the biosimilar drug as the immunocapture agent on beads, but the principle remains that the therapeutic molecule captures antibodies directed against itself.
In summary: The Polatuzumab biosimilar is used both as capture and detection reagent in a bridging ELISA to specifically and sensitively detect anti-Polatuzumab ADAs, enabling immunogenicity monitoring in patients treated with the biosimilar or in comparative studies with the originator.
References & Citations
1 Polson AG, Yu SF, Elkins K, et al. Blood. 110(2):616-623. 2007.
2 Deeks ED. Drugs. 79(13):1467-1475. 2019.
3 Pfeifer M, Zheng B, Erdmann T, et al. Leukemia. 29(7):1578-1586. 2015.
4 Dornan D, Bennett F, Chen Y, et al. Blood. 114(13):2721-2729. 2009.
5 Polson AG, Williams M, Gray AM, et al. Leukemia. 24(9):1566-1573. 2010.