Anti-Human CD276 (B7-H3) (Mirzotamab) [Clone ABBV-155] — Fc Muted™

Anti-Human CD276 (B7-H3) (Mirzotamab) [Clone ABBV-155] — Fc Muted™

Product No.: LT645

- -
- -
Product No.LT645
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
Muted
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.

Use of Research-Grade Mirzotamab Biosimilars as Calibration Standards and Reference Controls in PK Bridging ELISA

Background
In pharmacokinetic (PK) studies for biosimilar development, it is essential to accurately measure the serum concentration of both the biosimilar and its reference product to establish bioequivalence. Enzyme-linked immunosorbent assays (ELISAs) are commonly used for this purpose due to their sensitivity and specificity for detecting therapeutic proteins in complex biological matrices like serum.

Calibration Standards and Reference Controls

Role of Biosimilars as Calibration Standards
For PK bridging studies, it has become a best practice in the industry to use the biosimilar candidate itself as the calibration standard in the bioanalytical assay, including the reference and biosimilar products in the measurement. This approach reduces variability that could arise from using different standards for the biosimilar and the reference product, and simplifies the statistical analysis required for demonstrating bioequivalence. The biosimilar standard curve is generated by spiking known concentrations of the biosimilar into the biological matrix (e.g., serum), and this curve is then used to quantify both the biosimilar and reference product in test samples.

Method Validation and Comparability
Before deploying the biosimilar as a single calibration standard, rigorous comparability testing is performed. The precision, accuracy, and robustness of the assay are evaluated using both the biosimilar and reference product over a range of concentrations. Statistical analysis determines if both products are bioanalytically equivalent within the assay, ensuring that the biosimilar standard accurately reflects the immuno-reactivity of the reference product. Only after establishing this comparability is the single-standard approach adopted for formal PK studies.

Reference Controls
In addition to calibration standards, quality control (QC) samples are prepared using both the biosimilar and the reference product at various concentrations. These QC samples are included in every assay run to monitor the performance and reliability of the ELISA over time and across different operators and days. This dual use of both biosimilar and reference product as QCs further confirms the suitability of the biosimilar standard for quantifying the reference product.

Technical and Regulatory Considerations

Assay Development
The ELISA must be fully validated according to regulatory guidelines (e.g., FDA, EMA). This includes assessing parameters such as sensitivity, specificity, linearity, precision, and accuracy, as well as demonstrating that the biosimilar and reference product yield comparable results within the assay.

Statistical Analysis
Bioanalytical equivalence is often determined by comparing the 90% confidence interval of the measured concentrations to a pre-defined equivalence interval (typically [0.8, 1.25]), using the totality of evidence to conclude similarity. This stringent criteria help ensure that any observed PK differences are due to true biological differences rather than assay variability.

Summary Table

ComponentRole in PK Bridging ELISAPreparation and Use
Biosimilar StandardCalibration curve for quantifying both biosimilar and reference product in serumSpiked into matrix at known concentrations
Reference ProductTested as samples and as QC to confirm method suitabilityUsed to validate the biosimilar standard
QC SamplesMonitor assay performance, precision, and accuracyPrepared from both biosimilar and reference at multiple levels

Conclusion

Research-grade biosimilars like Mirzotamab (theoretical, as no specific Mirzotamab data is cited) are used as calibration standards in PK bridging ELISAs only after demonstrating analytical comparability with the reference product. This approach harmonizes quantification, reduces variability, and supports the robust demonstration of PK similarity required for regulatory approval. QC samples from both products are included to ensure ongoing assay reliability and accuracy.

Primary Syngeneic and Humanized Mouse Models for Anti-CD276 Antibody Studies

Syngeneic models are commonly employed to evaluate the effects of anti-CD276 (B7-H3) antibodies on tumor growth and the tumor immune microenvironment in vivo. These models allow for tracking tumor growth dynamics and detailed characterization of tumor-infiltrating lymphocytes (TILs) in the presence of an intact immune system.

Key Syngeneic Models Used

  • MC38 Colon Carcinoma: This is a well-characterized, immunocompetent murine model frequently used to study the role of CD276 in tumor stroma (endothelial cells) and tumor cells. Researchers have generated CD276-knockout (KO) mice on a C57BL/6 background and transplanted MC38 colon cancer cells (with or without CD276 expression) into these mice. Anti-CD276 antibody-drug conjugates (ADCs), such as m276-PBD, showed significant tumor growth delay when CD276 was expressed on tumor cells, and even more potent effects when both tumor cells and tumor vasculature expressed CD276.
  • Chemically-Induced Head and Neck Squamous Cell Carcinoma (HNSCC): A recent study utilized a chemically induced murine HNSCC model to assess the efficacy of anti-CD276 antibodies. The model allowed researchers to evaluate tumor growth inhibition and the impact of combining CD276 targeting with integrin β6 (ITGB6) knockout, revealing synergistic therapeutic effects.
  • Panel of Syngeneic Models: While not specifically focused on CD276, the broader literature highlights the use of models such as RENCA (renal cell carcinoma), CT26 (colon carcinoma), EMT6 (breast carcinoma), and B16F10 (melanoma) for immunotherapy studies, each with distinct tumor-immune infiltrate profiles. These models are valuable for understanding how different immune microenvironments respond to targeted therapies, though direct evidence of anti-CD276 antibody use in these specific models is not detailed in the provided results.

Humanized Models

Humanized mouse models (where human tumors are grown in immunodeficient mice with a humanized immune system) are less commonly described in the provided literature for anti-CD276 studies. Most of the cited research focuses on syngeneic models with murine tumors or human xenografts in immunodeficient mice (e.g., nude mice) to isolate the role of host (stromal) versus tumor cell CD276. The latter approach helps determine if therapeutic effects are due to targeting the tumor microenvironment rather than the tumor cells themselves.

Experimental Strategies and Outcomes

  • Stromal vs. Tumor Cell Targeting: In MC38 models, researchers compared tumors grown in CD276 WT vs. KO mice to distinguish the contribution of stromal (host) CD276 from tumor cell CD276. Anti-CD276 ADCs were more effective when both tumor cells and stroma expressed CD276, leading to tumor regression and long-term survival in a subset of animals.
  • Combination Therapies: In HNSCC models, combining anti-CD276 antibody with genetic ablation of ITGB6 showed enhanced tumor growth inhibition compared to either treatment alone, suggesting potential for combination strategies in the clinic.
  • TIL Characterization: While the provided results emphasize tumor growth inhibition, they imply that syngeneic models are suitable for profiling TILs in response to anti-CD276 therapy, given their intact immune systems and the ability to perform flow cytometry and immunohistochemistry on excised tumors. However, detailed TIL phenotyping (e.g., CD8+ T cell, regulatory T cell, myeloid cell subsets) in the context of anti-CD276 therapy is not explicitly described in the cited studies.

Summary Table: Models and Applications

Model TypeTumor TypeKey FindingsReference
Syngeneic (C57BL/6)MC38 colon carcinomaAnti-CD276 ADC delays growth, especially when both tumor and stroma express CD276; some tumors regress
Chemically-induced syngeneicHNSCCAnti-CD276 + ITGB6 knockout synergistically inhibits growth
Syngeneic panel (CT26, RENCA, etc.)Various carcinomasModels differ in immune infiltrate; useful for immunotherapy mechanism studies (general, not CD276-specific)
Human xenograft (immunodeficient)Human colon cancers (DLD-1, etc.)Modest growth delay in CD276 KO hosts; used to isolate stromal CD276 effect

Conclusions

Syngeneic mouse models—particularly MC38 colon carcinoma and chemically induced HNSCC—are the primary platforms where research-grade anti-CD276 antibodies have been administered in vivo to study tumor growth inhibition and (implicitly) to characterize TIL responses. These models enable dissection of stromal versus tumor cell CD276 contributions and evaluation of combination therapies. Humanized models are less prominently featured in this context, with most work relying on immunocompetent syngeneic or immunodeficient xenograft systems. Detailed immune profiling of TILs in response to anti-CD276 therapy is an area ripe for further investigation, leveraging the strengths of these syngeneic systems.

Researchers use Mirzotamab biosimilar—a monoclonal antibody targeted against specific tumor antigens—primarily as a tool in preclinical and translational immune-oncology models to study synergistic effects when combined with other checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 biosimilars.

In laboratory and animal studies, these combinations are designed to:

  • Target multiple immune suppression pathways: Anti-CTLA-4 works mainly in the lymph nodes to boost early T cell activation, while anti-PD-1/PD-L1 or anti-LAG-3 agents restore exhausted T cell function at the tumor site. Using Mirzotamab (anti-ABBV-155 biosimilar, for research use only) with checkpoint inhibitors allows researchers to explore whether blocking both tumor-intrinsic targets and immune checkpoints can result in greater T cell activation, tumor infiltration, and tumor cell elimination.

  • Simulate complex immune-tumor interactions: Researchers use complex systems such as co-culture assays (tumor cells + immune cells), 3D spheroid models, and humanized mouse models to test these combinations, analyzing outcomes like immune cell activation, cytokine secretion, tumor growth inhibition, and cell death. Mirzotamab biosimilar can serve either as a direct tumor-targeting antibody or, in ADC (antibody-drug conjugate) formats, to deliver cytotoxins, thereby increasing tumor immunogenicity—potentially making checkpoint blockade more effective.

  • Study mechanistic synergy: Combining Mirzotamab with anti-CTLA-4 or anti-LAG-3 biosimilars helps dissect synergy—e.g., whether Mirzotamab’s targeted tumor cell lysis releases antigens that prime T cells, which are then further activated by checkpoint inhibition, overcoming each agent’s monotherapy limitations.

Key techniques in these studies include:

  • In vitro assays measuring T cell proliferation, cytokine release, and tumor cell apoptosis after combination treatment.
  • Flow cytometry and immunohistochemistry to assess immune cell infiltration and phenotypes.
  • In vivo mouse models (e.g., syngeneic or humanized immune system mice), measuring tumor volume, survival, and immune landscape in response to combination therapy.

Research consistently shows that such combinations can induce stronger and more durable anti-tumor responses than either agent alone, especially in “cold” tumors (poorly infiltrated by T cells). However, increased efficacy is often accompanied by higher toxicity risk, necessitating careful dose and schedule optimization in translational studies before clinical application.

While Mirzotamab biosimilar is not as widely published as other checkpoint blockers, its use in combination research models follows the same fundamental scientific approach: rational multi-target intervention to maximize immune-mediated tumor destruction and to model combination immunotherapy strategies relevant to clinical oncology.

Mirzotamab Biosimilar and Bridging ADA ELISA: Technical Approach

In the context of immunogenicity testing for a biosimilar such as Mirzotamab (a hypothetical biosimilar mAb, as no real "Mirzotamab" appears in the literature), the bridging anti-drug antibody (ADA) ELISA is a standard method to monitor whether patients develop antibodies against the therapeutic drug, which can compromise efficacy or safety. Here’s how the biosimilar is integrated into the assay:

Bridging ADA ELISA: General Principle

A bridging ELISA captures ADAs using immobilized drug (the antigen) and then detects them using the same drug conjugated to a reporter enzyme (e.g., horseradish peroxidase, HRP). The steps typically include:

  1. Capture: Patient serum (potentially containing ADAs) is added to a microplate coated with the drug (in this case, the Mirzotamab biosimilar).
  2. Wash: Unbound components are washed away.
  3. Detection: The same drug, now conjugated to an enzyme (e.g., HRP), is added. If ADAs are present, they bridge the immobilized and enzyme-conjugated drug, forming an immune complex on the plate.
  4. Signal Development: Addition of a chromogenic substrate generates a measurable signal proportional to the amount of ADA present.

Biosimilar as Capture/Detection Reagent

  • Capture Reagent: The Mirzotamab biosimilar is immobilized on the plate to specifically capture any antibodies in the patient’s serum that recognize the drug.
  • Detection Reagent: The same Mirzomab biosimilar (or a batch with equivalent binding characteristics), conjugated to an enzyme (e.g., HRP), is used to detect the captured antibodies, relying on the “bridging” of the ADA between the two drug molecules.
  • Ensuring Equivalence: For biosimilars, it is critical that the assay can detect antibodies against both the biosimilar and the originator to ensure the test is relevant for all marketed versions of the drug. Regulatory guidance recommends rigorous cross-validation to confirm that the biosimilar-based assay performs equivalently to one based on the originator.

Considerations for Biosimilar Testing

  • Antigenic Equivalence: The biosimilar and originator must be compared to ensure they are recognized similarly by ADAs, a key regulatory requirement for biosimilar immunogenicity testing.
  • Assay Validation: The assay should be validated with both the biosimilar and originator to confirm similar sensitivity and specificity.
  • No Clinically Meaningful Differences: Minor differences in ADA rates between biosimilar and originator are generally acceptable if they do not translate into significant clinical effects.

Summary Table: Biosimilar Role in Bridging ADA ELISA

StepReagent UsedPurpose
Plate CoatingMirzotamab biosimilarCapture ADAs from patient serum
DetectionMirzotamab biosimilar-HRPDetect captured ADAs via enzyme signal
Assay ValidationBoth biosimilar & originatorEnsure equivalent ADA detection

Conclusion

A Mirzotamab biosimilar—like any biosimilar monoclonal antibody—can be used as both the capture and detection reagent in a bridging ADA ELISA to monitor patient immune responses, provided the assay is thoroughly validated for equivalence with the originator product. This approach ensures that the assay is fit-for-purpose to support biosimilar development and post-marketing pharmacovigilance.

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.