Anti-Human CD276 (B7-H3) (Mirzotamab) [Clone ABBV-155]

Anti-Human CD276 (B7-H3) (Mirzotamab) [Clone ABBV-155]

Product No.: LT640

- -
- -
Product No.LT640
Clone
ABBV-155
Target
CD276
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
ABBV-155, anti-CD276, B7-H3, B7H3
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
Active
Immunogen
Human CD276/B7-H3
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 ?
FC,
FA,
ELISA,
WB,
IP
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 Mirzotamab. This product is for research use only. Mirzotamab activity is directed against Human CD276 (B7-H3).
Background
CD276, also known as B7 homolog 3 protein (B7-H3), is a member of the B7 superfamily and acts as an immune checkpoint molecule and a costimulatory/coinhibitory immunoregulatory protein1. CD276 influences innate and adaptive immunity, regulates the aggressiveness of cancer cells, and is thought to play an important role in tumor development and cancer immunity. CD276 has been studied in many cancers, including breast, lung, ovarian, brain, gastric, and squamous cell carcinoma.

Human CD276 exists as either a soluble isoform or as a ~45–66 kDa type I transmembrane protein that is composed of an extracellular domain, a transmembrane domain, and a short intracellular domain1. Soluble CD276 is produced by cleavage from the cell surface or via alternative intron splicing and has been found in the secretomes of exosomes and other extracellular vesicles.

In normal human tissues, CD276 mRNA is widely and abundantly expressed but protein abundance is low1. miR-124 is thought to cause translational repression of CD276 by targeting its 3’-UTR, while other miRNAs are known to affect CD276 expression. In tumor cells, CD276 mRNA and protein are abundant, and its presence is correlated with worsened prognosis, poor survival, recurrence rate, and enhanced invasive and migratory properties1, 2. CD276 is known to act as a T cell inhibitor that promotes tumor proliferation and invasion and is an immune checkpoint molecule in the epithelial mesenchymal transition pathway2.

Blocking CD276 with monoclonal antibodies reduces tumor growth and prolongs survival in mouse models of various cancers 1, 2. Additionally, a first-in-human study shows that monotherapy with mirzotamab clezutoclax, a first-in class antibody drug conjugate composed of mirzotamab conjugated via a solubilizing linker to a B cell lymphoma – extra long (BCL-XL) inhibitor, has potential anti-tumor activity3, 4.
Antigen Distribution
CD276 is weakly expressed on activated lymphocytes, macrophages, dendritic cells, nasal and airway epithelial cells, and osteoblasts. A soluble form is secreted by monocytes, dendritic cells, and activated T cells. CD276 can be abundant in tumor cells.
Ligand/Receptor
CD276/B7-H3
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
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 Mirzotamab biosimilars play a crucial role as analytical standards in pharmacokinetic bridging ELISA assays for measuring drug concentrations in serum samples, following established bioanalytical strategies for biosimilar development.

Single PK Assay Approach

The current industry best practice involves establishing a single PK assay that uses a single analytical standard for quantification of both the biosimilar and reference product(s) within test samples. In the case of Mirzotamab biosimilars, the biosimilar product itself serves as the assay calibrator for quantifying both the biosimilar and reference products in serum matrix. This approach offers significant advantages by decreasing the inherent variability that would be associated with running multiple methods and eliminating the need for crossover analysis when conducting blinded clinical studies.

Bioanalytical Comparability Assessment

Before implementing the single assay approach, a comprehensive testing paradigm must be followed. The process begins with a robust method qualification study that generates precision and accuracy data sets of both the biosimilar and reference products. Statistical analysis is then applied to determine if the test products are bioanalytically equivalent within the method.

The analytical equivalence is evaluated by comparing the 90% confidence interval to pre-defined equivalence interval [0.8, 1.25] and concluding bioanalytical equivalence by combining the totality of evidence. This stringent criteria around the measurement of test products within the assay is necessary to minimize confounding variability, particularly given that the method supports PK similarity studies.

Calibration Standards and Quality Controls

Once bioanalytical comparability is established across test products, the biosimilar is selected as the analytical standard for the single method. The validation process involves preparing independent sets of biosimilar standards in human serum at multiple concentration levels. For example, validation studies typically use nine independent sets of standards at nominal concentrations ranging from 50 to 12,800 ng/mL.

Quality Control (QC) samples are prepared with both the biosimilar and reference products at various concentrations and quantified against the biosimilar standard curve. This ensures that the method can accurately measure both products using the same calibration standards.

Method Validation Requirements

The PK assay undergoes full validation for performance parameters consistent with quantitative pharmacokinetic methods as described in FDA bioanalytical guidance documents. The validation study is conducted across multiple assays performed over several days by different analysts to ensure robustness and reproducibility.

This scientifically rigorous approach ensures that research-grade Mirzotamab biosimilars can serve as reliable calibration standards, providing accurate and precise measurements of drug concentrations in serum samples while supporting the overall biosimilarity assessment required for regulatory approval.

The primary in vivo models used to study the effects of anti-CD276 (B7-H3) antibodies on tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are syngeneic mouse tumor models and human xenograft (including PDX and CDX) models.

Key Models:

  • Syngeneic Models:
    These involve transplanting mouse tumor cell lines into immunocompetent mice of the same genetic background (e.g., MC38 colon cancer in C57BL/6 mice). These models are crucial for studying antitumor immune responses, including TILs, because they maintain an intact, functional mouse immune system and allow for interrogation of immune checkpoint blockade, such as CD276 inhibition.

    • Example: MC38 (colon), TC-1 (lung), and other commonly used syngeneic models. Anti-CD276 antibodies or antibody-drug conjugates are administered, and changes in tumor growth and immune cell composition—especially TILs—are evaluated.
  • Humanized or Xenograft Models:
    These rely on the implantation of human tumor cells or patient-derived xenografts (PDX) into immunodeficient mice (such as NOD/SCID or athymic nude mice). For detailed immune cell analysis (including human TILs), "humanized" mice (engrafted with human hematopoiesis) may be used.

    • Example: Studies have shown that anti-CD276 antibody-drug conjugates (such as enoblituzumab, m276-SL-PBD, or MGC018) inhibit tumor growth in human xenograft or PDX models of various cancers, including Ewing sarcoma, neuroblastoma, and non-small cell lung cancer. TIL analysis in these settings typically requires humanized immune systems, or focuses on stromal/tumor-infiltrating mouse immune cells in non-humanized hosts.

TIL Characterization:

  • In syngeneic models, the immune TILs (e.g., CD8+ and CD4+ T cells, NK cells, myeloid cells) are murine, and their abundance and activation state following treatment can be directly measured.
  • In xenograft or humanized models, TILs are either murine (in standard xenografts) or human/mixed (in humanized mouse models). TIL characterization in true humanized models allows for direct assessment of how anti-CD276 affects human immune infiltration.

Summary Table:

Model TypeImmune SystemTumor SourceAntibody CompatibilityTIL Characterization
Syngeneic mouse tumor modelMouse (intact)Mouse cell linesMurine/humanized AbMurine TILs, robust analysis
Human xenograft (standard)Mouse (deficient)Human cell/PDXHuman AbLimited (murine TILs mainly)
Humanized mouse (immune system)Human (reconstituted)Human cell/PDXHuman AbHuman TILs, best fidelity

In summary:

  • Syngeneic mouse tumor models are the most routinely used for in vivo immune profiling of TILs and anti-tumor efficacy of anti-CD276 agents.
  • Human xenograft and especially humanized mouse models are used for direct testing of human-specific antibodies or antibody-drug conjugates, and can support TIL studies involving human immune cells in reconstituted hosts.

For comparative TIL analysis and immune characterization, syngeneic models are generally preferred, unless the antibody is strictly human-specific, in which case humanized or certain xenograft models may be required.

Researchers are increasingly exploring combination strategies using Mirzotamab biosimilars alongside other checkpoint inhibitors to understand synergistic effects in complex immune-oncology models, although specific protocols for Mirzotamab combinations require careful experimental design.

Understanding Mirzotamab Biosimilar as a Research Tool

Mirzotamab biosimilar targets CD276 (B7-H3), a checkpoint protein that represents a distinct pathway from the more commonly studied PD-1/PD-L1 and CTLA-4 checkpoints. This research-grade biosimilar uses the same variable region sequence as the therapeutic antibody, making it an valuable tool for investigating B7-H3 pathway interactions in preclinical models.

Rationale for Combination Checkpoint Inhibitor Studies

The scientific foundation for combining multiple checkpoint inhibitors stems from their different mechanisms of action and sites of activity. Researchers have established that targeting multiple checkpoints can increase the activity of each other and help overcome the limitations of monotherapy approaches. For instance, anti-CTLA-4 primarily acts in the lymph node compartment to restore induction and proliferation of activated T cells, while anti-PD-1 mainly functions at the tumor periphery, preventing neutralization of cytotoxic T cells by PD-L1 expressing tumor cells.

Established Combination Strategies in Research

Dual Checkpoint Blockade Models

Current research demonstrates successful combination approaches, particularly with PD-1 and LAG-3 pathway targeting. The combination of relatlimab (anti-LAG-3) with nivolumab (anti-PD-1) has shown that dual checkpoint blockade can double median progression-free survival compared to single-agent therapy. This success provides a framework for researchers studying Mirzotamab combinations.

Multi-Pathway Targeting Approaches

Researchers are focusing on combination immunotherapy agents that target multiple pathways simultaneously. The logic extends beyond traditional PD-1/CTLA-4 combinations to include novel targets like LAG-3, which represents the first new checkpoint pathway approved in eight years.

Experimental Design Considerations for Mirzotamab Combinations

Sequential vs. Concurrent Administration

When designing studies with Mirzotamab biosimilars and other checkpoint inhibitors, researchers must consider timing strategies. The different sites of action suggest that concurrent administration may provide optimal synergy, though sequential protocols may help identify individual pathway contributions.

Model System Selection

Complex immune-oncology models require careful selection of:

  • Tumor models that express relevant checkpoint proteins
  • Immune-competent systems that can demonstrate T cell activation and proliferation
  • Biomarker assessment capabilities to track multiple pathway interactions

Dosing Optimization

Combination studies must account for potential increased toxicity profiles when multiple checkpoint pathways are blocked simultaneously. Research protocols typically start with established monotherapy doses and adjust based on observed interactions.

Monitoring Synergistic Effects

Immune Response Measurements

Researchers evaluate synergistic effects through multiple parameters:

  • T cell activation markers across different compartments
  • Cytokine profiling to assess inflammatory responses
  • Tumor infiltration studies to measure immune cell recruitment
  • Long-term survival outcomes in preclinical models

Mechanistic Understanding

The combination of Mirzotamab with other checkpoint inhibitors allows researchers to investigate how B7-H3 pathway blockade interacts with established checkpoint mechanisms. This includes studying whether B7-H3 inhibition enhances the efficacy of PD-1/PD-L1 or CTLA-4 blockade through complementary mechanisms.

The research landscape for combination checkpoint inhibitor studies continues expanding, with Mirzotamab biosimilars providing researchers with tools to explore novel therapeutic combinations that may overcome resistance mechanisms and improve patient outcomes in complex immune-oncology models.

Immunogenicity Testing and ADA Assays

Immunogenicity testing is critical in clinical development to assess whether a patient’s immune system generates anti-drug antibodies (ADAs) against a therapeutic, such as monoclonal antibodies (mAbs) or their biosimilars like Mirzotamab. The presence of ADAs can alter drug pharmacokinetics, reduce efficacy, or induce adverse effects. A bridging ELISA is a widely used method to detect and quantify ADAs in patient samples.

Principle of Bridging ADA ELISA

A bridging ELISA for ADA detection works by capturing antibodies in patient serum that specifically bind to the drug of interest. Here’s the general workflow:

  • Plate Coating: The microtiter well is coated with the drug (e.g., Mirzotamab biosimilar).
  • Sample Incubation: Patient serum is added; any ADAs present will bind to the drug.
  • Detection Reagent: A labeled (e.g., biotinylated or enzyme-conjugated) form of the same drug is added. This will bind to the other arm of the ADA, “bridging” the ADA between the coated drug and the labeled drug.
  • Signal Detection: The presence of this “bridge” produces a detectable signal (e.g., colorimetric, fluorescence), proportional to the amount of ADAs present.

This format increases specificity, as only antibodies binding to both the capture and detection forms of the drug generate a signal.

Role of Mirzotamab Biosimilar

In the context of testing for ADAs against Mirzotamab (or its reference product):

  • Capture Reagent: The Mirzotamab biosimilar is immobilized on the plate to bind ADAs from patient serum.
  • Detection Reagent: The same Mirzotamab biosimilar, but labeled (e.g., with biotin or HRP), is used in solution to detect the ADAs that have been captured.
  • Bridging Mechanism: Only ADAs that can bind both the immobilized and the labeled drugs simultaneously will generate a signal, ensuring specificity for antibodies against Mirzotamab.

Key Considerations

  • Assay Validation: The assay must be validated for specificity, sensitivity, and robustness, especially to avoid interference from drug present in the sample (drug interference), which can mask ADA detection.
  • Matrix Effects: Serum or plasma components may interfere, so assay optimization (e.g., sample dilution, acid dissociation) is often required.
  • Neutralizing Capacity: If ADAs are detected, further testing (e.g., cell-based assays) may be needed to determine if they can neutralize the drug’s therapeutic effect.
  • Regulatory Standards: Regulatory guidelines emphasize using the same assay for both biosimilar and reference drug, and for biosimilars, that the immunogenicity profile is highly similar to the reference product.

Summary Table: Bridging ELISA Using Mirzotamab Biosimilar

StepReagent UsedPurpose
Plate CoatingMirzotamab biosimilarCapture ADAs from patient serum
Sample IncubationPatient serumAllow ADAs to bind to coated biosimilar
DetectionLabeled Mirzotamab biosimilar“Bridge” and detect bound ADAs
Signal ReadoutChromogenic/fluorogenic substrateQuantify ADA levels

Conclusion

A Mirzotamab biosimilar can be used as both the capture and detection reagent in a bridging ADA ELISA to sensitively and specifically monitor a patient’s immune response to the drug. The assay design ensures that only antibodies recognizing both forms of the biosimilar are detected, providing a reliable measure of immunogenicity in clinical samples. Careful validation and optimization are essential to ensure accurate and reproducible results, aligning with regulatory expectations for biosimilar development.

References & Citations

1. Zhou WT, Jin WL. Front Immunol. 12:701006. 2021.
2. Liu S, Liang J, Liu Z, et al. Front Oncol. 11:654684. 2021.
3. Tolcher AW, Carneiro BA, Dowlati A, et al. J. Clin. Oncol. 39(15):suppl. 2021.
4. https://ncit.nci.nih.gov/ncitbrowser/ConceptReport.jsp?dictionary=NCI%20Thesaurus&code=C157279
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.