Anti-Human HER2 (Trastuzumab) [Clone 4D5-8]

Anti-Human HER2 (Trastuzumab) [Clone 4D5-8]

Product No.: LT1500

- -
- -
Product No.LT1500
Clone
4D5-8
Target
HER-2/neu
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
ErbB-2, NEU, NGL, HER2, TKR1, CD340, MLN 19, HER-2/neu
Isotype
Human IgG1κ
Applications
CyTOF®
,
ELISA
,
FC
,
IHC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
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.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
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.
Additional Applications Reported In Literature ?
ELISA,
WB,
IP,
FA,
FC
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.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars

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 Trastuzumab Biosimilars as Calibration Standards in PK Bridging ELISAs

Research-grade trastuzumab biosimilars are employed as calibration standards and reference controls in pharmacokinetic (PK) bridging enzyme-linked immunosorbent assays (ELISAs) to quantify drug concentrations in serum samples. Here’s how this process works:

Step-by-Step Application

  • Standard Curve Construction: A range of known concentrations of the trastuzumab biosimilar is used to generate a calibration (standard) curve in the ELISA. This curve establishes the relationship between the signal generated (e.g., optical density) and the actual concentration of the drug.
  • Sample Quantification: Serum samples from patients or study subjects containing unknown concentrations of trastuzumab or its biosimilars are assayed alongside these standards. The signal from each sample is compared to the standard curve, allowing back-calculation of the drug concentration in the sample.
  • Quality Control: Reference controls (often at high, mid, and low concentrations) are included in each assay run to monitor assay performance, ensuring accuracy and reproducibility over time.
  • Specificity and Accuracy: These assays are designed to be highly specific for free (unbound) trastuzumab, avoiding cross-reactivity with other serum proteins or therapeutic antibodies. The use of monoclonal anti-idiotype antibodies further enhances specificity for the target molecule.

Technical Details and Validation

  • Format: The most common format is a sandwich ELISA, where one antibody captures trastuzumab and a second, labeled antibody detects it, producing a measurable signal.
  • Detection Range: Commercial kits may offer a detection range from 0 to 300 ng/mL, with a lower limit of quantitation as low as 75 ng/mL in validated assays.
  • Validation: Assays are validated for parameters such as precision, accuracy, linearity, and specificity before clinical use. This ensures that the standard curve is reliable for interpolating unknown sample concentrations.
  • Bridging Studies: Trastuzumab biosimilars provide reference points for bridging pharmacokinetic studies between originator and biosimilar products, allowing direct comparison of drug exposure in different populations or with different formulations.

Summary Table: Key Roles of Biosimilar Standards

FunctionDescription
Calibration StandardsUsed to construct a standard curve for converting ELISA signals into drug concentrations.
Reference ControlsEnsure assay validity and monitor performance across runs.
SpecificityDemonstrate assay selectivity for free trastuzumab, not cross-reacting with other proteins.
PK BridgingEnable direct comparison of drug exposure between originator and biosimilar products.

Conclusion

Research-grade trastuzumab biosimilars are essential tools in PK bridging ELISAs, serving as calibration standards and reference controls to ensure accurate, specific, and reproducible measurement of drug concentrations in serum. Their use underpins the reliability of pharmacokinetic studies critical for regulatory approval and therapeutic monitoring of trastuzumab and its biosimilars.

The primary in vivo models for studying the effects of research-grade anti-HER-2/neu antibody on tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models—especially those engineered to express human HER2/neu in a fully immunocompetent background.

Key details:

  • Syngeneic mouse models (mouse HER2/neu): Traditional models, like the TUBO mammary carcinoma model in BALB/c mice, overexpress rat or mouse neu (the HER2 ortholog). The therapeutic effect of anti-HER2/neu monoclonal antibodies such as 7.16.4 (which recognizes the rat neu antigen) can be robustly studied in these mice. These models allow for analysis of immune cell infiltration after antibody treatment, since the immune system is intact.

  • Syngeneic models with human HER2: To evaluate human anti-HER2 therapeutics (e.g., trastuzumab), models use murine tumor cells engineered to stably express either full-length or truncated human HER2 (e.g., 4T1.2-HER2T cells in BALB/c mice). These models are critical since human HER2-targeted antibodies do not cross-react efficiently with rodent HER2/neu, and immune-competent backgrounds are necessary for assessing the immune response, including TIL composition.

  • Mechanisms assessed: These models enable investigation of tumor growth inhibition, antibody-dependent cellular cytotoxicity (ADCC), and changes in TILs—including CD4+ and CD8+ T cells, NK cells, and other immune subsets—in response to anti-HER2/neu antibody administration.

  • Humanized models: While not emphasized in the retrieved sources, humanized mouse models (immunodeficient mice engrafted with a human immune system and human tumor xenografts) are sometimes used to study human antibody therapies. However, they are less commonly employed for detailed TIL characterization due to limitations in immune system reconstitution.

Summary Table: Models for in vivo anti-HER-2/neu antibody studies

Model TypeSpecies/HostTumor Cell LineHER2/neu ExpressedAntibody ExampleImmune CompetenceTIL Assessment
Syngeneic (mouse HER2/neu)MouseTUBO, MMC, othersRat/mouse neu7.16.4 (anti-neu)YesYes
Syngeneic (human HER2)Mouse4T1.2-HER2T, others engineeredHuman HER2 (full/T)Trastuzumab, T-DM1YesYes
Humanized (xenograft, less common)MouseHuman HER2+ tumorsHuman HER2Trastuzumab, pertuzumabPartialLimited

Supporting insights:

  • 7.16.4 antibody is frequently used in mouse models with rat/mouse neu, showing that tumor inhibition and TIL modulation are T cell-dependent and require intact Fc receptor (FcγR) signaling.
  • 4T1.2-HER2T model permits immune profiling of TILs after huHER2-targeted therapy, allowing for detailed immune mechanistic studies.
  • Fully immunocompetent models provide the best platform to characterize how anti-HER2/neu therapy modulates the tumor immune microenvironment.

These syngeneic mouse models are widely regarded as gold-standard preclinical systems for concurrently evaluating both tumor growth inhibition and immune cell infiltration after anti-HER2/neu antibody administration.

Based on the available research, the combination of trastuzumab biosimilars with checkpoint inhibitors represents an emerging area of investigation in immune-oncology, though specific studies directly combining these agents are still limited in the current literature.

Mechanistic Rationale for Combination Approaches

The theoretical basis for combining trastuzumab biosimilars with checkpoint inhibitors stems from their complementary mechanisms of action. Trastuzumab biosimilars, such as MYL-1401O and CT-P6, have demonstrated bioequivalent immunomodulation profiles to the reference trastuzumab, including the ability to induce transient IL-6 peaks and modulate mononuclear cell subset profiles, particularly affecting CD16+ cells. This immunomodulatory activity provides a foundation for potential synergistic interactions with checkpoint inhibitors.

Current Checkpoint Inhibitor Combination Strategies

Researchers are actively exploring multi-checkpoint targeting approaches based on the principle that different checkpoint inhibitors have distinct mechanisms of action. The combination of CTLA-4 and PD-1/PD-L1 blockade has shown particular promise, with anti-CTLA-4 agents primarily acting in lymph node compartments to restore T-cell induction and proliferation, while anti-PD-1 agents work at the tumor periphery to prevent cytotoxic T-cell neutralization.

The landmark CheckMate 067 trial demonstrated that in patients with PD-L1-negative tumors, the combination of ipilimumab plus nivolumab achieved longer progression-free survival (11.2 months) compared to nivolumab alone (5.3 months). At five-year follow-up, combination therapy achieved complete response rates of 22%, with 74% of patients remaining alive and treatment-free.

Emerging Combination Approaches

Recent advances include the exploration of LAG-3 inhibitors in combination therapy. The RELATIVITY-047 study demonstrated that combining nivolumab with relatlimab (a LAG-3 inhibitor) improved progression-free survival in advanced melanoma patients, leading to FDA approval for first-line treatment. This success has prompted interest in combining LAG-3 and TIM-3 inhibitors with PD-1/PD-L1 inhibitors based on preclinical data showing their coordinated function.

Research Methodology and Considerations

When studying these combinations, researchers typically employ comprehensive immunomodulation profiling approaches. For trastuzumab biosimilars, this includes assessment of sixty parameters encompassing serum cytokines, peripheral mononuclear cell subsets, cell activation responses, and cytokine release assays. The demonstrated bioequivalence of biosimilars like CT-P6, which showed comparable pharmacokinetic ratios (AUC∞ 99.05, Cmax 96.58) and similar safety profiles to reference trastuzumab, provides confidence in their use for combination studies.

Clinical Translation and Challenges

While the theoretical framework supports combining trastuzumab biosimilars with checkpoint inhibitors, researchers must address several challenges. The increased toxicity risk associated with combination checkpoint inhibitor therapy, as demonstrated by higher grade 3-4 toxicities in multi-agent regimens, necessitates careful dose optimization and patient selection. Additionally, the development of subcutaneous formulations and cost-effectiveness considerations play important roles in clinical implementation.

The current research landscape suggests that while direct studies combining trastuzumab biosimilars with checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3 agents are still emerging, the established immunomodulatory properties of trastuzumab biosimilars and the proven efficacy of multi-checkpoint inhibitor approaches provide a strong foundation for future combination studies in complex immune-oncology models.

In immunogenicity testing for trastuzumab and its biosimilars, the bridging ELISA format utilizes the drug itself as both capture and detection reagent to monitor anti-drug antibodies (ADAs) in patient samples. This approach is particularly valuable for assessing immune responses against therapeutic monoclonal antibodies like trastuzumab, which is used to treat HER2-positive breast cancers.

Bridging ELISA Principle

The bridging ELISA technique exploits the bivalent nature of anti-drug antibodies to create a "bridge" between two drug molecules. In this format, trastuzumab (or its biosimilar) serves dual roles: one molecule acts as the capture reagent immobilized on the plate surface, while a second labeled molecule functions as the detection reagent. When ADAs are present in patient serum, they bind to both the capture and detection drug molecules simultaneously, forming a detectable complex.

Specific Implementation for Trastuzumab ADA Testing

Capture Phase SetupThe assay begins with trastuzumab being immobilized onto microplate wells, either through direct coating or via biotin-streptavidin capture systems. Quality control samples and patient serum samples are then added to wells, allowing any anti-trastuzumab antibodies present to bind to the immobilized drug.

Detection PhaseAfter washing to remove unbound materials, a labeled version of trastuzumab is introduced as the detection reagent. This can be biotinylated trastuzumab followed by streptavidin-HRP, or directly HRP-labeled trastuzumab. The detection reagent binds to the captured ADAs, completing the "bridge" formation.

Alternative ACE Methodology

Some laboratories employ an Affinity Capture Elution (ACE) approach, where trastuzumab captures ADAs from serum, followed by acid dissociation and transfer to a second plate for detection with biotinylated trastuzumab and streptavidin-HRP. This method can help overcome matrix interferences that may affect standard bridging formats.

Clinical Significance and Limitations

The formation of anti-trastuzumab antibodies has been associated with reduced therapeutic efficacy, making ADA monitoring clinically relevant. However, bridging ELISA assays face specificity challenges in complex biological matrices due to matrix components, soluble target molecules, or residual drug that can interfere with detection. High-quality reagents and appropriate blocking solutions are essential for obtaining meaningful results in patient monitoring applications.

The bridging ELISA format provides high sensitivity and enables high-throughput screening of patient samples, making it particularly suitable for clinical immunogenicity studies and routine therapeutic drug monitoring programs.

References & Citations

1. Jayaswamy PK, Vijaykrishnaraj M, Patil P, et al. Ageing Res Rev. 83:101791. 2023.
2. Romano R, Bucci C. Cells. 9(8):1887. 2020.
3. Sigismund S, Avanzato D, Lanzetti L. Mol Oncol. 12(1):3-20. 2018.
4. Iwamoto M, Saso W, Sugiyama R, et al. Proc Natl Acad Sci U S A. 116(17):8487-8492. 2019.
5. Lupberger J, Zeisel MB, Xiao F, et al. Nat Med. 17(5):589-595. 2011.
6. Hu W, Zhang S, Shen Y, et al. Virology. 521:33-43. 2018.
7. Klann K, Bojkova D, Tascher G, et al. Mol Cell. 80(1):164-174.e4. 2020.
8. Xu G, Li Y, Zhang S, et al. Cell Res. 31(12):1230-1243. 2021.
9. Wang S, Qiu Z, Hou Y, et al. Cell Res. 31(2):126-140. 2021.
10. Sigismund S, Avanzato D, Lanzetti L. Mol Oncol. 12(1):3-20. 2018.
11. Garnock-Jones KP. Drugs. 76(2):283-289. 2016.
12. Yang XD, Jia XC, Corvalan JR, et al. Crit Rev Oncol Hematol. Apr;38(1):17-23. 2001.
13. https://www.accessdata.fda.gov/drugsatfda_docs/label/2009/125147s080lbl.pdf
14. Dubois EA, Cohen AF. Br J Clin Pharmacol. 68(4):482-483. 2009.
15. Saltz L, Easley C, Kirkpatrick P. Nat Rev Drug Discov. 5(12):987-988. 2006.
16. Giusti RM, Shastri KA, Cohen MH, et al. Oncologist. 12(5):577-583. 2007.
CyTOF®
Indirect Elisa Protocol
Flow Cytometry
IHC

Certificate of Analysis

Formats Available

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