Human epidermoid carcinoma cells (A431) over-expressing EGFR.
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.
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.
Applications and Recommended Usage? Quality Tested by Leinco
FC The suggested concentration for Trastuzumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
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 Trastuzumab. Clone 4D5-8 recognizes human erbB-2. This product is for research use only.
Background
Trastuzumab is a monoclonal antibody targeting HER2, a 185 kDa transmembrane glycoprotein that contains an extracellular domain and intracellular tyrosine kinase activity. When it is functioning normally, the HER2 pathway supports cell growth and division. On the other hand, the over expression of HER2 propels cell growth beyond its typical range. This overexpression is associated with some cancers, namely breast and stomach, in which the HER2 protein can be expressed up to 100 times more than in typical cells. Trastuzumab induces an immune-mediated response that triggers the internalization and downregulation of HER2 making it an excellent target for immunotherapy. Several clinical studies are under way which show that anti-HER-2/neu antibodies inhibit the growth and proliferation of these tumor cells In vitro as well as In vivo.
Antigen Distribution
Ubiquitous expression with highest expression levels found in the kidney, skin, esophagus, and small intestine.
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 Trastuzumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as reference materials of known concentration to generate standard curves, enabling accurate quantification of Trastuzumab levels in serum samples.
Context and Supporting Details:
Calibration Standards: In a bridging ELISA for PK studies, vials of research-grade Trastuzumab biosimilars, precisely quantified, are serially diluted to create a standard curve. Known concentrations—often ranging from low (e.g., 10 ng/mL) to high (e.g., 300 ng/mL)—are included with every ELISA plate to correlate optical density (OD) readings to actual drug concentration.
Reference Controls: These biosimilars are also used as quality controls to ensure assay consistency and validity across runs and between laboratories. Using biosimilars with verified equivalence to the reference product ensures that antibody binding in the assay matches the characteristics of the therapeutic drug found in patient serum.
Assay Specificity and Accuracy: The availability of a biosimilar that closely matches the structure and HER2-binding capacity of reference Trastuzumab ensures that the ELISA specifically detects free Trastuzumab (not other immunoglobulins or unrelated antibodies in serum), maximizing both specificity and sensitivity.
ELISA Format: Most PK bridging ELISAs are quantitative (often sandwich-type) and may use Trastuzumab-specific capture and detection antibodies, with colorimetric or enzymatic readouts fitted to a standard curve generated from biosimilar reference standards.
Additional Relevant Information:
The use of biosimilar standards is particularly important in biosimilar development, comparability, and regulatory submission because it ensures that the measured pharmacokinetic parameters reflect actual exposure to therapeutically equivalent molecules.
Regulatory agencies expect the use of well-characterized, research-grade standards and controls for assay validation, reproducibility, and cross-study comparability during both clinical trials and routine therapeutic drug monitoring.
Spiked and unspiked serum samples (using these standards) verify recovery and linearity, further confirming assay robustness.
In summary, Trastuzumab biosimilars serve a dual role as both calibration standards and reference controls in PK ELISAs, supporting the accurate measurement of serum drug concentrations and the functional comparability required for biosimilar development and therapeutic monitoring.
The primary models used to administer research-grade anti-HER-2/neu antibody in vivo for tumor growth inhibition studies and TIL characterization are immunocompetent syngeneic mouse models expressing murine or human HER-2/neu, with humanized mouse models reserved for human-specific translational research.
Syngeneic Mouse Models:
Most commonly, mouse strains (e.g., BALB/c or C57BL/6) are implanted with murine tumor cell lines engineered to express either mouse neu or human HER2 proteins.
Examples include models using TUBO (mouse mammary carcinoma line expressing rat neu) and 4T1.2-HER2T (4T1.2 cells engineered with a human HER2 transgene).
These models allow the use of anti-HER-2/neu antibodies, such as 7.16.4 (targets mouse neu) or Trastuzumab/variants (targets human HER2), to assess tumor growth inhibition and analyze TILs through flow cytometry and immune phenotyping of tumor and draining lymph nodes.
Studies show these models can recapitulate the adaptive and innate immune involvement in tumor clearance post-antibody therapy, making them ideal for TIL characterization.
Humanized Mouse Models:
Used when studying the function of human HER2/neu antibodies in vivo, especially to assess human-specific immune responses and antibody effects on human TIL populations.
These are often immune-deficient mice reconstituted with human immune cells and implanted with human HER2-expressing tumor xenografts.
While more translational for clinical relevance, these models are less standardized due to variability in immune reconstitution.
Model Selection Rationale:
Syngeneic models are preferred for immune mechanism studies because they maintain a fully functional mouse immune system, enabling robust analysis of TIL populations (CD4+ T cells, B cells, NK cells, dendritic cells, etc.) following antibody treatment.
Humanized models are best for investigating mechanisms specific to human immune cell interactions or antibody Fc domains that specifically engage human Fc receptors, but offer reduced reproducibility compared to syngeneic setups.
Key Features for TIL Characterization:
Tumors and draining lymph nodes are harvested after antibody administration.
Immune infiltrate composition (CD4+, CD8+ T cells, NK, dendritic cells) is characterized by flow cytometry and immunostaining.
In summary, syngeneic mouse models expressing murine or human HER2/neu are standard for mechanistic studies of antibody efficacy and TIL phenotyping, while humanized models provide translational relevance for human antibody and TIL studies.
Trastuzumab Biosimilars in Immune Oncology Research
Trastuzumab—and its biosimilars—are monoclonal antibodies targeting the HER2 receptor, primarily used in HER2-positive breast cancer. While the original use of trastuzumab is to block HER2 signaling and recruit immune cells (via antibody-dependent cellular cytotoxicity, ADCC), its potential synergy with immune checkpoint inhibitors (ICIs) is a growing area of translational and clinical research.
Rationale for Combination Studies
Trastuzumab's Immunomodulatory Effects
Direct Antitumor Action: Trastuzumab inhibits HER2 signaling, induces cell cycle arrest, and promotes tumor cell death via ADCC, primarily mediated by natural killer (NK) cells.
Immune Modulation: Trastuzumab can alter the tumor microenvironment (TME) by increasing production of cytokines like TGFβ and interferon-γ (IFN-γ), and by modulating NK cell activity. However, the TME can also counteract these effects through immunosuppressive molecules (e.g., HLA-G, PD-L1).
Resistance Mechanisms: Tumor expression of inhibitory ligands (e.g., HLA-G, which engages NK cell inhibitory receptors) or upregulation of PD-L1 can render tumors resistant to trastuzumab.
Checkpoint Inhibitors
Targeted Pathways: ICIs such as anti-CTLA-4, anti-PD-1/PD-L1, and anti-LAG-3 act at different nodes of immune regulation. Anti-CTLA-4 primarily enhances T-cell priming in lymphoid organs, while anti-PD-1/PD-L1 and anti-LAG-3 act at the tumor site to prevent T-cell exhaustion.
Synergistic Potential: Combining trastuzumab with ICIs aims to overcome resistance by simultaneously enhancing NK and T-cell activation while blocking inhibitory signals in the TME.
Experimental Approaches
Preclinical and Early Clinical Models
Immune Profiling: Studies often compare the immunomodulation profiles of trastuzumab biosimilars (e.g., MYL-1401O, CT-P6) with the originator, demonstrating bioequivalence in immune parameters such as cytokine release and mononuclear cell subset modulation. This ensures that biosimilars can be used interchangeably in combination studies.
Combination with ICIs: Researchers expose HER2-positive tumor models (cell lines, patient-derived xenografts, or organoids) to trastuzumab biosimilars together with anti-CTLA-4, anti-PD-1/PD-L1, or anti-LAG-3 agents. Outcomes measured include tumor growth inhibition, immune cell infiltration, cytokine profiles, and expression of immune checkpoint molecules.
Mechanistic Studies: For example, blocking HLA-G (which desensitizes tumors to trastuzumab) in combination with PD-1/PD-L1 inhibition has been shown to restore NK cell cytotoxicity and enhance trastuzumab efficacy in resistant models. Similarly, combining trastuzumab with anti-CTLA-4 may amplify T-cell priming while ADCC is ongoing.
Toxicity and Efficacy: Researchers assess not only antitumor efficacy but also immune-related adverse events, as combination therapies often increase toxicity.
Translational and Clinical Implications
Personalized Combinations: The choice of ICI (anti-CTLA-4, anti-PD-1/PD-L1, anti-LAG-3) may depend on the dominant resistance mechanism in the tumor (e.g., high HLA-G, PD-L1, or LAG-3 expression).
Biosimilar Utility: Trastuzumab biosimilars offer a cost-effective, clinically equivalent alternative for these combination studies, enabling broader access and larger-scale trials.
Future Directions: Ongoing research is exploring optimal sequencing, dosing, and patient selection for these combinations, with a focus on biomarkers predictive of response and resistance.
Summary Table: Key Synergistic Mechanisms
Component
Mechanism in TME
Synergy with Trastuzumab
Research Focus
Trastuzumab (biosimilar)
HER2 blockade, ADCC, cytokine modulation
Enhances NK and macrophage activity
Bioequivalence, immune profiles
Anti-CTLA-4
T-cell priming in lymph nodes
Boosts T-cell response to HER2+ cells
Toxicity, efficacy in PD-L1 low tumors
Anti-PD-1/PD-L1
Prevents T-cell exhaustion at tumor site
Overcomes PD-L1-mediated resistance
Biomarker-driven combinations
Anti-LAG-3
Blocks inhibitory signals on T cells
Potentiates T-cell cytotoxicity
Emerging, preclinical models
Conclusion
Researchers use trastuzumab biosimilars in combination with checkpoint inhibitors to model and overcome complex resistance mechanisms in HER2-positive cancers. By leveraging the complementary actions of ADCC-inducing antibodies and ICIs that modulate T-cell and NK cell activity, these studies aim to identify synergistic regimens that improve outcomes for patients with refractory disease. The use of biosimilars ensures that such research is both scientifically rigorous and economically feasible.
In the context of immunogenicity testing, a Trastuzumab biosimilar can be used as both the capture and detection reagent in a bridging Anti-Drug Antibody (ADA) ELISA to monitor a patient's immune response against Trastuzumab. Here's how this process generally works:
Bridging ADA ELISA Protocol
Principle:
The bridging ELISA is a technique used for detecting ADAs in patient sera. It involves capturing ADAs using one form of the drug tagged with an enzyme or biotin, and then detecting them with another form of the drug, often labeled.
Application with Trastuzumab Biosimilar:
Capture Reagent:
Biotinylation: The Trastuzumab biosimilar is biotinylated to act as the capture reagent. This biotinylated form is immobilized on streptavidin-coated plates.
Function: The biotinylated Trastuzumab captures ADAs present in the patient's serum sample.
Detection Reagent:
Horseradish Peroxidase (HRP) Labeling: Another form of the Trastuzumab biosimilar is labeled with HRP. This labeled form serves as the detection reagent.
Function: The HRP-labeled Trastuzumab binds to the ADAs that have been captured by the biotinylated form, creating a bridging effect.
Detection:
After washing away unbound reagents, a chromogenic substrate like 3,3’,5,5’-tetramethylbenzidine (TMB) is added to the wells. The TMB reacts with HRP to produce a color change proportional to the amount of ADAs present.
Interpretation:
The intensity of the color development indicates the level of ADAs in the patient's serum. Higher color intensity corresponds to a higher concentration of ADAs.
Advantages and Considerations
Sensitivity and Specificity: The bridging ELISA is highly sensitive and can detect bivalent ADAs, which are crucial for assessing immunogenicity. However, specificity can be challenged by matrix components or drug components in serum.
Customization: Each laboratory should customize the protocol to suit specific requirements, ensuring optimal performance and reliability of the assay.
By using the Trastuzumab biosimilar in this manner, researchers can effectively monitor a patient's immune response to Trastuzumab treatment, which is critical for understanding treatment efficacy and potential side effects related to immunogenicity.
References & Citations
1. Fendly, B. et al. (1990) Cancer Research 50: 1550-1558.
2. McBride, H. et al. (2019) Pharm Res. 36(12): 177.
3. Zielinski, C. et al. (1997) Int. J. Cancer 73: 875–879
4. Valone, FH. et al. (1995) J. Clin. Oncology 13 (9): 2281-92.
5. Hynes, NE. et al. (1993) Br J Cancer. 68(6): 1140–1145.