Anti-Human HER2 (Trastuzumab) CHO [Clone 4D5-8] — Purified No Carrier Protein

Anti-Human HER2 (Trastuzumab) CHO [Clone 4D5-8] — Purified No Carrier Protein

Product No.: LT1508

- -
- -
Product No.LT1508
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
CHO 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.

Research-grade Trastuzumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as known-concentration reference materials to construct standard curves and ensure assay specificity and accuracy when quantifying drug concentrations in serum samples.

Essential context and application details:

  • In a PK bridging ELISA, a series of known concentrations of research-grade Trastuzumab biosimilars (or originator) are prepared to generate a standard curve. This curve allows the conversion of optical density (OD) or other signal outputs from the assay into the actual drug concentrations present in unknown serum samples.

  • Calibration standards: These are serial dilutions of the biosimilar or reference Trastuzumab, typically spanning the expected therapeutic and subtherapeutic range (e.g., 0–300 ng/mL in some commercial kits). The assay measures the ELISA signal from each calibration standard, producing a standard (calibration) curve that is then used to interpolate the concentrations of Trastuzumab in test samples.

  • Reference controls: These are quality control samples formulated at specific known concentrations of Trastuzumab biosimilar. They are analyzed alongside unknown samples to assess and monitor the accuracy, precision, and reproducibility of the assay. Concordance between the measured and expected concentrations confirms proper assay performance.

  • Biosimilar suitability: Research-grade Trastuzumab biosimilars are selected for calibration and control purposes because they are highly similar in structure, binding activity, and immunoreactivity to the originator drug. Demonstrated comparability in physicochemical and functional properties (such as HER2 binding, glycosylation, and FcRn affinity) ensures that the ELISA will recognize both biosimilar and reference molecules equivalently.

  • Assay specificity: The use of Trastuzumab-specific antibodies in the ELISA ensures that readings correspond specifically to Trastuzumab (and its biosimilars), without cross-reactivity to endogenous IgG or other therapeutic antibodies present in the serum.

  • Bridging concept: In PK bridging ELISAs, biosimilars can be used in parallel with the originator to verify that the assay's sensitivity, specificity, and quantitation are consistent, further establishing biosimilar comparability and facilitating regulatory acceptance.

Summary of use steps in a PK bridging ELISA:

  • Spike blank serum with known concentrations of research-grade Trastuzumab biosimilar to prepare calibration standards.
  • Run standards and unknown samples together in the assay.
  • Generate a calibration curve from the standards' signal outputs.
  • Use this curve to interpolate Trastuzumab (or biosimilar) concentrations in serum samples, ensuring accurate PK analysis.

This strategy is central to both method validation and routine quantitative drug monitoring for originator and biosimilar Trastuzumab in clinical and research settings.

The primary models used to study in vivo administration of research-grade anti-HER-2/neu antibodies for tumor growth inhibition and analysis of tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models engineered to express human or mouse HER2/neu, and, less commonly, humanized mouse models. Syngeneic models are the most widely utilized in immunocompetent settings for TIL characterization.

Essential context and supporting details:

  • Syngeneic Mouse Models:
    These involve transplanting HER2/neu-expressing murine tumor cells (such as the TUBO or 4T1.2-HER2T lines) into immunocompetent mice of the same genetic background (e.g., BALB/c mice).

    • Anti-HER2/neu antibodies (e.g., 7.16.4) can be administered to these mice to study in vivo tumor regression, immune responses, and TIL composition.
    • These models allow for detailed assessment of both innate (NK cells, dendritic cells) and adaptive (CD4^+^ and CD8^+^ T cells, B cells) immune infiltration into the tumor following antibody therapy.
    • Many studies explicitly demonstrate T cell and FcR-dependent mechanisms for antibody-induced tumor clearance and enhanced TIL abundance.
    • Syngeneic models can be further engineered to express truncated human HER2 (HER2T) to enable testing of clinical-grade anti-human HER2 therapeutics such as trastuzumab or T-DM1.
  • Humanized Mouse Models:
    Less commonly, studies employ humanized mice (immunodeficient mice reconstituted with human immune cells) and then xenograft human HER2^+^ tumor cell lines and treat with human-specific antibodies.

    • These models are valuable for evaluation of human-targeted therapeutics but have limitations in native immune cell interactions due to incomplete immune system reconstitution.
  • TIL Characterization:
    In both syngeneic and humanized models, flow cytometry, immunohistochemistry, and transcriptomic profiling are used post-treatment to quantify and characterize TIL populations (e.g., activated CD8^+^ T cells, CD4^+^ T cells, NK cells).

    • Studies often link increased infiltration and activation of TILs with successful tumor regression and immune memory.
  • Other Experimental Models:
    Additional syngeneic tumor types (colorectal, ovarian) can be engineered for HER2T expression to allow anti-HER2 activity assessment in varied tissue contexts.

    • Novel approaches (e.g., CAR-M macrophage therapy targeting HER2) further expand the use of such models to dissect mechanisms of TME remodeling and antigen spreading.

In summary, immunocompetent syngeneic mouse models (murine or human HER2/neu-expressing) are the standard in vivo system to study anti-HER2/neu antibody-induced tumor inhibition and TIL dynamics, with humanized models reserved for more translational human-specific studies.

Researchers use Trastuzumab biosimilars in combination with other checkpoint inhibitors, like anti-CTLA-4 or anti-LAG-3 biosimilars, to investigate whether these agents produce synergistic antitumor effects by modulating distinct, complementary pathways of the immune system in complex immune-oncology models.

Trastuzumab biosimilars, which target HER2-positive tumor cells, elicit immune responses primarily through mechanisms like antibody-dependent cell-mediated cytotoxicity (ADCC) and antibody-dependent cellular phagocytosis (ADCP), engaging NK cells and macrophages. The antitumor activity of Trastuzumab can be dampened by the tumor microenvironment (TME), such as increased expression of inhibitory ligands (e.g., HLA-G, PD-L1) that suppress immune cell activation. Researchers study combinations with checkpoint inhibitors because these agents block immune checkpoints (such as CTLA-4, PD-1/PD-L1, or LAG-3), which can restore or enhance the function of cytotoxic T cells and NK cells suppressed by the TME.

Experimental paradigms typically include:

  • Preclinical models (cell lines and animal models): Sequential or simultaneous administration of Trastuzumab biosimilars and checkpoint inhibitors to HER2-positive cancers, followed by monitoring tumor growth and detailed analysis of immune cell infiltration and activity.
  • Functional readouts: Researchers use assays to assess changes in cytokine levels, immune cell activation (e.g., CD16+ NK cells), tumor cell killing, and modulation of various PBMC (peripheral blood mononuclear cell) subsets before and after combination therapy.
  • Translational strategies: Integrating biomarker and immunophenotyping data to correlate immune modulation (e.g., increase in cytotoxicity, decrease in Treg suppressive function) with clinical benefit or tumor response.

The rationale for using multiple checkpoint inhibitors (for example, anti-CTLA-4 and anti-LAG-3) with Trastuzumab biosimilars is that each checkpoint acts at different stages or locations of the immune response. For instance:

  • CTLA-4 inhibitors mainly enhance T cell activation and proliferation in lymphoid tissue.
  • PD-1/PD-L1 inhibitors restore cytotoxic T cell function in the tumor microenvironment.
  • LAG-3 inhibitors can further relieve T cell exhaustion and enhance effector functions.

By blocking distinct inhibitory pathways, combinations can theoretically produce greater immune activation and tumor clearance than monotherapies, although there is also an increased risk of immune-related toxicities. Researchers systematically profile these effects in immuno-oncology models to optimize efficacy and safety for clinical translation.

In summary, scientists employ Trastuzumab biosimilars combined with multiple checkpoint inhibitors in complex preclinical and translational models to study the immunologic crosstalk, seeking to maximize synergistic antitumor immunity by unlocking multiple layers of immune activation—especially in tumors with immunosuppressive TMEs.

A Trastuzumab biosimilar can be used as both the capture and detection reagent in a bridging ADA ELISA to sensitively monitor anti-drug antibodies (ADA) generated in a patient's serum in response to Trastuzumab therapy. In this assay format, the biosimilar reproduces the relevant antigenic epitopes of the reference drug, so patient antibodies that recognize Trastuzumab will also bind its biosimilar form, enabling effective immunogenicity testing.

Essential context and supporting details:

  • In a bridging ELISA for ADA detection:

    • The biosimilar Trastuzumab is immobilized (directly or via biotin-streptavidin linkage) on the ELISA plate as the capture reagent.
    • Patient serum is added; if anti-Trastuzumab antibodies are present, they will bind to the drug on the plate (one antibody arm engages the immobilized biosimilar).
    • A labeled Trastuzumab biosimilar (e.g., conjugated with HRP or biotin) is next added as the detection reagent. The other arm of any bivalent ADA binds the labeled biosimilar, forming a "bridge" between the plate-bound and labeled drug.
    • Following washing, a substrate is added for color development, which is proportional to ADA levels in the serum sample.
  • The use of biosimilar Trastuzumab (instead of originator) in this role is accepted because both share high sequence and structural similarity, hence patient ADAs will generally recognize both forms equivalently, supporting assay reliability. Numerous immunogenicity studies confirm that Trastuzumab and biosimilars induce similar ADA responses when measured via these methods.

  • Bridging ADA ELISA is highly sensitive and suitable for high-throughput screening, but its specificity can be impacted by soluble drug, matrix effects, or presence of soluble antigen; appropriate controls and blocking reagents are recommended.

Additional relevant information:

  • Some protocols employ biotinylated biosimilar drug for capture and HRP-conjugated biosimilar drug for detection.
  • The bridging format detects a broad range of ADA isotypes, provided they are bivalent and can bridge between capture and detection reagents.
  • The technique is extensively validated for monoclonal antibody drugs including Trastuzumab and its biosimilars.

In summary, a Trastuzumab biosimilar enables robust ADA monitoring in bridging ELISA by serving as both the capture and detection reagent, accurately reflecting immunogenicity against the therapeutic agent in treated patients.

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.