Anti-Human CD79b (Polatuzumab) [Clone RG7596]

Anti-Human CD79b (Polatuzumab) [Clone RG7596]

Product No.: C1090

- -
- -
Product No.C1090
Clone
RG7596
Target
CD79b
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
Ig B chain
Isotype
Human IgG1κ
Applications
ELISA
,
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
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.
Regulatory Status
Research Use Only
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
FA,
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 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.
Ligand/Receptor
heterodimer CD79a/CD79b
NCBI Gene Bank ID
UniProt.org
Research Area
Agammaglobulinemia
.
Biosimilars
.
Immunology

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 Polatuzumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as a quantifiable benchmark to determine the concentration of Polatuzumab in serum samples. These biosimilars are structurally and functionally similar to the clinical drug, ensuring relevant and accurate quantitation within the assay.

How biosimilars are used in PK bridging ELISA:

  • Calibration Curve Creation: Known concentrations of the research-grade Polatuzumab biosimilar are spiked into serum or buffer to generate serial dilution standards. These standards are run alongside unknown samples to construct a calibration (standard) curve. The measured optical density (OD) signal from the standards is plotted against their known concentrations, allowing interpolation of drug concentrations in test samples.

  • Reference Control: Along with calibration standards, biosimilars may be used as quality control (QC) samples at different concentration levels (e.g., low, medium, high). QC controls ensure assay performance and validity for each batch.

Assay format—Bridging ELISA:

  1. Capture: An anti-idiotypic (anti-drug) antibody coated on the plate captures the Polatuzumab molecule from standards or serum samples.

  2. Detection: A second, labeled anti-idiotypic antibody (often HRP-conjugated), specific for a different epitope of Polatuzumab, binds to the captured antibody-drug.

  3. Quantification: The detected signal (typically absorbance) is proportional to the Polatuzumab concentration, and unknown serum concentrations are extrapolated from the calibration curve established using the research-grade biosimilar.

Key Parameters for Calibration Standards:

  • Standards should cover the assay’s dynamic range (e.g., 0.31–5 μg/mL for some commercial Polatuzumab kits).
  • Standards and controls are included in every plate/run to ensure accuracy, reproducibility, and quality.

Why research-grade biosimilars are suitable:

  • They mimic the structure and immunoactivity of the clinical product, allowing them to serve reliably for both calibration and control, provided they are well-characterized and validated for the assay.
  • The similarity in pharmacokinetic and immunological properties, as shown in published biosimilar ELISA methods, supports the use of research biosimilars for quantitative serum assays.

Summary Table:

RoleResearch-Grade Polatuzumab Biosimilar Use
Calibration StandardSerial dilutions for standard curve in ELISA
Reference Control (QC)Ensures assay precision and accuracy between runs

Essential references for further methodological detail:

  • See schematic and explanations in Bio-Rad documents for bridging ELISA use of research biosimilars.
  • See example ELISA kit ranges and formats in commercial Polatuzumab ELISA kits.
  • See detailed PK ELISA methodology using biosimilar antibodies as standards/QC in literature examples.

The primary in vivo models used to study the effects of research-grade anti-CD79b antibodies on tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are humanized mouse models, specifically:

  • Xenograft models (NOG or NPG mice) reconstituted with human PBMCs (peripheral blood mononuclear cells)
  • HSC-NPG mice (NOD-Prkdc<scid> IL2rg<tm1Sug>/ShiJicTac mice transplanted with human hematopoietic stem cells)

Key model characteristics:

  • Humanized PBMC xenograft model: Human B-cell non-Hodgkin lymphoma (B-NHL) cell lines are inoculated into severely immunocompromised NOG (NOD/Shi-scid/IL-2Rγnull) or NPG mice, followed by engraftment with human PBMCs to partially reconstitute a human-like immune system. Anti-CD79b antibodies, including bispecific variants (CD79b/CD3), are administered in vivo to assess both tumor growth inhibition and immune cell dynamics.

  • Evaluation of TILs: These models allow for robust analysis of human T-cell infiltration within the tumor microenvironment following anti-CD79b treatment. Studies using this platform have demonstrated strong infiltration of activated human T cells and depletion of malignant B cells within tumors after antibody administration.

  • HSC-NPG mice: These mice are transplanted with human hematopoietic stem cells, enabling longer-term and more complete immune system reconstitution compared to PBMC models. In these models, anti-CD79b antibodies can be evaluated for both tumor regression and their effects on circulating and intratumoral immune cells.

  • Syngeneic models (e.g., mouse B-cell lymphoma in immunocompetent mice) are rarely used for anti-human CD79b antibody testing, because human antibodies generally do not cross-react with mouse CD79b, and the readout of human TILs would not be possible in mouse-only systems.

Supporting data:

  • Anti-CD79b (and anti-CD79b/CD3) antibodies resulted in tumor eradication in NOG mice engrafted with human PBMCs, with accompanying increases in activated human TILs in the tumor tissue.
  • Related studies using anti-CD79b antibody-drug conjugates (ADCs) reported tumor growth inhibition in xenograft models (e.g., BJAB or DoHH2) in immunodeficient mice, but these models, unless reconstituted with human immune cells, do not permit detailed TIL characterization.

Summary Table:

Model TypeImmune SystemTumor LineTIL Analysis Possible?References
NOG/NPG mice + human PBMCs (xenograft)Human PBMC-reconstitutedHuman B-NHLYes (human TILs)
HSC-NPG mice (xenograft)Human HSC-reconstitutedHuman B-NHLYes (human immune)
Immunodeficient mice, no humanization (xenograft)Mouse only (deficient)Human B-NHLNo (non-human TILs)
Syngeneic mouse (e.g., A20 in BALB/c)Full mouseMouse B-lymphomaOnly mouse immuneNot reported

Conclusion:The main platforms for in vivo characterization of anti-CD79b on tumor growth and TILs are humanized xenograft models with human PBMC or HSC engraftment. Fully immunocompetent murine syngeneic models are not suitable for research-grade anti-human CD79b antibody studies requiring human TIL characterization.

Researchers study the synergistic effects of Polatuzumab biosimilars in combination with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 agents by designing preclinical or early-phase clinical experiments that integrate these agents in complex immune-oncology models, often using mouse models or humanized systems.

Checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 act on different stages and locations of T cell activation—CTLA-4 mainly in lymphoid organs during the priming phase, and LAG-3 (often partnered with PD-1 blockade) within the tumor microenvironment to reverse T cell exhaustion. The rationale for combining these with agents like Polatuzumab, an antibody-drug conjugate targeting B cells, is to harness complementary mechanisms to boost anti-tumor immunity beyond monotherapy limitations.

Key elements of such studies include:

  • In Vitro and In Vivo Models: Researchers use immune-competent mouse models or patient-derived xenografts to recreate complex tumor-immune environments, enabling them to track the interactions between tumor, immune cells, and multiple therapeutic agents.
  • Immune Profiling: Experiments map the specific types of immune cells (e.g., CD4+ helper T cells, CD8+ cytotoxic T cells, regulatory T cells) activated by each drug or combination. For instance, anti-PD-1/LAG-3 efficacy depends more on CD4+ T cells, while anti-PD-1/CTLA-4 mainly boosts CD8+ T cells, as observed in melanoma models.
  • Combination Strategies: Agents like Polatuzumab (biosimilar or originator) are combined with checkpoint inhibitors to test whether the cytotoxic effects of B cell depletion (from Polatuzumab) can be amplified by concurrent T-cell checkpoint blockade, leading to more robust and durable anti-tumor responses.
  • Endpoints: Synergy is assessed by measuring tumor regression, progression-free survival, and detailed immune cell activation and cytokine profiles. Researchers look for improved outcomes (e.g., higher complete remission rates, extended survival), as seen in the clinical setting combining novel agents with standard regimens in B-cell malignancies.
  • Mechanistic Studies: Detailed immunophenotyping is used to distinguish whether improved responses are due to enhanced T-cell priming and infiltration, reduced immune suppression in the tumor microenvironment, or other immune modulation.

Published reviews and studies highlight that combining agents with distinct mechanisms—such as B cell targeting (Polatuzumab) and checkpoint blockade (CTLA-4, LAG-3)—is a promising way to overcome tumor resistance and enhance anti-tumor efficacy, but these combinations must be carefully evaluated for both efficacy and additive toxicity in robust models before translation to larger clinical trials.

Currently, there are limited published clinical data specifically combining Polatuzumab with checkpoint inhibitors, but the rationale and early preclinical findings set a strong foundation for ongoing and future research into these multi-agent setups. If you need specific protocols or detailed immune-oncology model descriptions, searching recent immunotherapy literature and registered early-phase combination trials may provide the most updated methodologies.

A Polatuzumab biosimilar can be used as a capture or detection reagent in a bridging ADA (anti-drug antibody) ELISA by exploiting the bivalent binding nature of patient-generated ADAs to form a bridge between two labeled forms of the biosimilar drug—one for capture and one for detection.

How it works:

  • Assay setup: Typically, you label the biosimilar with two different tags (for example, biotin on one and horseradish peroxidase [HRP] or digoxigenin on the other). Patient serum is incubated with both, allowing any ADAs to bridge the two labeled drug molecules, forming an immune complex.
  • Capture phase: The complex is captured onto a streptavidin-coated plate (if biotin is used) via the biotinylated drug. The bound complex consists of the biosimilar-ADA-biosimilar "sandwich."
  • Detection phase: The detection reagent (HRP- or digoxigenin-labeled biosimilar) is recognized by a secondary detection antibody (such as anti-digoxin-HRP), producing a quantifiable signal proportional to ADA levels.

Rationale for using a biosimilar as reagent:

  • Using a labeled Polatuzumab biosimilar as the capture or detection agent ensures the ADA response being measured is directed against the drug administered or its biosimilar variant, providing relevant clinical immunogenicity data.
  • Regulatory guidelines generally require immunogenicity monitoring with an assay matching the drug product received by the patient. For biosimilars, you can use either a "same product" approach (biosimilar with biosimilar, reference with reference) or a "single-antigen" approach (biosimilar for all samples). The latter is often acceptable and widely used, provided assay validation demonstrates comparable sensitivity and specificity.
  • This approach also allows detection of antibodies against any domain of the antibody-drug conjugate (ADC)—such as the antibody portion, the linker, or the payload—since the entire molecule is used as both capture and detection reagent.

Critical details:

  • Customization and validation: Each lab must optimize and validate the assay using well-characterized reagents and robust controls to ensure the assessment is sensitive, specific, and drug-tolerant.
  • Interpretation: The presence of bridging indicates the presence of ADA. Subsequent confirmatory and titer assays further characterize specificity and strength of the immune response, while domain mapping assays can identify the ADA target region.

Summary protocol (generalized):

  1. Label Polatuzumab biosimilar with biotin (capture) and HRP/digoxigenin (detection).
  2. Incubate patient serum with both reagents to form bridging complexes if ADA is present.
  3. Transfer to streptavidin-coated plate to immobilize complexes.
  4. Wash and add detection system (e.g., anti-digoxin-HRP).
  5. Add substrate and read the signal to quantify ADA levels.

This strategy is broadly applicable for monitoring immunogenicity of biosimilars, including Polatuzumab, in clinical and regulatory settings.

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.
Indirect Elisa Protocol
FA

Certificate of Analysis

Formats Available

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