Anti-Human Trophoblast Cell Surface Antigen 2 (TROP-2) (Sacituzumab) – Fc Muted™

Anti-Human Trophoblast Cell Surface Antigen 2 (TROP-2) (Sacituzumab) – Fc Muted™

Product No.: T2035

- -
- -
Product No.T2035
Clone
IMMU-132
Target
TROP-2
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
TACSTD2; GA733-1; M1S1; Tumor-associated calcium signal transducer 2
Isotype
Human IgG1κ
Applications
FA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
Immunogen
Parental murine clone RS7 developed by immunizing mice with cell membranes isolated from squamous non-small cell lung cancer.
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
Country of Origin
USA
Shipping
2 – 8° C Wet Ice
Additional Applications Reported In Literature ?
FA
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 Sacituzumab. IMMU-132 (Sacituzumab) activity is directed against human TROP-2.
Background
Trophoblast cell surface antigen 2 (TROP-2), also known as tumor-associated calcium signal transducer 2 (TACSTD2), is a cell surface glycoprotein that plays roles in cell-cell adhesion1, cell proliferation, invasion, migration, and apoptosis in healthy tissues as well as tumorigenesis in cancers2. TROP-2 acts as a transmembrane transducer of intracellular calcium signals. In cancer, TROP-2 is necessary and sufficient to drive cancer growth1 and promotes migration and invasion of cancer cells by regulating the calcium ion signaling pathway and cyclin expression, stimulating ERK signaling, and reducing fibronectin adhesion2. TROP-2 stimulates proliferation in some cancers (cervical, bladder) but suppresses proliferation in others (cholangiocarcinoma, MCF7 breast cancer cell lines). TROP-2 is a target for the treatment of advanced metastatic cancers.

IMMU-132 (Sacituzumab) is an antibody-drug conjugate developed as an immunotherapy against cancer3. Sacituzumab was developed by site-specific conjugation of SN-38, an irinotecan active metabolite that acts as a DNA topoisomerase I inhibitor, to hRS7, a humanized monoclonal antibody against TROP-2. The original RS7 is a murine monoclonal antibody developed by immunizing mice with cell membranes isolated from squamous non-small cell lung cancer4. SN-38 is covalently linked to hRS7 via a hydrolyzable CL2A linker3. SN-38 is moderately toxic and is conjugated to hRS7 at a high mean drug-to-antibody ratio. Sacituzumab activity is performed.

This non-therapeutic biosimilar antibody is not conjugated to SN-38.

Antigen Distribution
TROP-2 is expressed in many normal tissues and is overexpressed in a variety of tumors, including pancreatic, ovarian, prostate, and breast cancers. TROP-2 was first identified in human placental trophoblasts.
Ligand/Receptor
N/A
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Immuno-Oncology
.
Immunology
.
Stem Cell

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 Sacituzumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the reference analyte to generate a standard curve, allowing accurate quantification of drug concentrations in serum samples from both reference and biosimilar products. This single-assay approach is central to PK bridging studies for biosimilars.

Essential context and supporting details:

  • In typical PK bridging ELISA for biosimilars:

    • A single research-grade Sacituzumab biosimilar is selected as the analytical standard (calibrator).
    • Serial dilutions of this biosimilar are prepared in a matrix (usually human serum) to create a standard curve covering the required concentration range.
    • Serum test samples containing either the biosimilar or the reference product (e.g., Sacituzumab govitecan) are analyzed in parallel with the standards.
    • The ELISA plate is coated with capture reagents (usually anti-Sacituzumab antibodies or the relevant antigen), which bind Sacituzumab from both samples and standards.
    • Detection antibodies (often HRP-conjugated anti-human IgG) generate a colorimetric signal proportional to Sacituzumab concentration.
    • Resulting absorbance values from test samples are interpolated against the standard curve generated from the biosimilar, providing quantitative measurement.
  • Why research-grade biosimilar as calibrator?

    • Using a single analytical standard (either biosimilar or reference) for both test and reference products reduces assay variability, facilitates comparison, and is considered best practice in regulatory bioanalytical guidance.
    • Before adoption, the analytical comparability must be established: both biosimilar and reference drug should behave equivalently in the assay (e.g., parallelism of standard curves, equivalent recovery across concentration range).
    • This approach is validated via head-to-head precision and accuracy experiments using quality control (QC) samples prepared with both biosimilar and reference material.
  • Controls in PK ELISA:

    • Reference controls (often both low and high concentration QCs) are made with either the biosimilar or reference drug, and included in each assay run to ensure performance.
    • Acceptance criteria (precision and accuracy) must be met across all QCs for the assay to be valid.
  • Example from industry:

    • One published PK assay validation (using monoclonal antibody biosimilars as the calibrator for both the biosimilar and reference products) prepared independent sets of standards in human serum for a range of 50–12800 ng/mL. QC samples were derived from both the biosimilar and reference sources, and all were quantified against the biosimilar standard curve. The validation included multiple analysts, assay runs, and cross-comparability assessments.

Summary of application:

  • Research-grade Sacituzumab biosimilar is diluted to generate calibrators for a standard curve in a bridging ELISA.
  • Test and reference serum samples are quantified relative to this curve.
  • QC samples made from both sources ensure system suitability.
  • Parallelism and bioanalytical equivalence between biosimilar and reference are confirmed during validation before using the assay in PK studies.

This methodology enables unbiased, standardized comparison of pharmacokinetics for both the reference and biosimilar drugs, supporting regulatory bioequivalence assessments.

The primary in vivo models used to study the effects of research-grade anti-TROP-2 antibodies on tumor growth inhibition and characterization of tumor-infiltrating lymphocytes (TILs) are:

  • Human tumor xenograft models in immunodeficient mice (often with human tumors or engineered tumors expressing human TROP-2)
  • Syngeneic mouse tumor models (using mouse tumors with murine TROP-2, or genetically engineered mouse strains expressing human TROP-2 for compatibility with humanized antibodies)
  • Humanized mouse models (immunodeficient mice engrafted with human immune cells and human tumors, used to study interactions between antibody therapy, tumor, and human immune system)

Key details on these models:

  • Syngeneic models provide fully intact immune systems and are commonly used for immunotherapy studies to evaluate TIL populations and mechanisms of immune modulation. These models require antibodies or reagents cross-reactive with the mouse version of TROP-2, or use genetically engineered mice to express human TROP-2. Syngeneic models such as MC38 (colon), 4T1 (breast), and others are widely used to examine both tumor growth inhibition and changes in TILs.

  • Humanized models (e.g., NSG or NOG mice engrafted with human hematopoietic cells) enable studies with human-specific anti-TROP-2 antibodies and allow for in vivo TIL characterization in a human immune context. These are especially valuable when the antibody does not cross-react with murine TROP-2 or when human immune responses are required.

  • Xenograft models in immunodeficient mice allow for testing of antibody efficacy on human tumors but lack a fully functional immune system, so TIL studies are limited unless supplemented with human immune cells.

Anti-TROP-2 antibody examples:

  • Research-grade antibodies such as AR47A6.4.2 and 7E6 have been studied in xenograft models for their ability to inhibit tumor growth, elucidating mechanisms including complement-dependent cytotoxicity and signaling disruption.
  • The antibody-drug conjugate IMMU-132 (sacituzumab govitecan), built on the humanized RS7 antibody backbone, has also been evaluated in human tumor xenograft models, demonstrating potent tumor growth inhibition.

Model selection depends on the specific goals:

  • If the focus is on immune cell dynamics and TILs, syngeneic and humanized models are essential due to their functional immune systems.
  • If only direct antitumor effects are needed, traditional xenograft models may suffice, but immune mechanisms and TIL characterization will be limited.

In summary, syngeneic models are the most broadly used for TIL characterization in the context of research-grade anti-TROP-2 antibodies in vivo, with humanized models or engineered syngeneic models used where species cross-reactivity is an issue or full human immune-tumor interactions are desired.

Researchers use Sacituzumab biosimilars—antibodies targeting Trop-2—in combination with other checkpoint inhibitor biosimilars (such as anti-CTLA-4 or anti-LAG-3) to study potential synergistic effects in immune-oncology models, particularly by evaluating enhanced tumor cell killing and immune activation.

Essential context and details:

  • Sacituzumab biosimilars act as antibody-drug conjugates (ADCs), binding specifically to the Trop-2 antigen highly expressed on many tumor cells. Upon internalization, they release cytotoxic agents like SN-38 directly into tumor cells, causing targeted cell death and minimizing systemic toxicity.

  • Checkpoint inhibitors (including anti-CTLA-4, anti-LAG-3) block negative immune regulators to enhance T-cell activation and anti-tumor immunity. When used in combination, these inhibitors trigger distinct immune pathways—for instance, CTLA-4 blockade enhances T-cell priming and proliferation, while PD-1/PD-L1 blockade boosts effector T-cell function at the tumor site.

  • Synergistic studies in complex models:

    • Researchers combine Sacituzumab biosimilars with checkpoint inhibitors to examine if dual targeting results in greater anti-tumor activity than either agent alone.
    • The ADC kills tumor cells and may release antigens, enhancing immune recognition. Concurrently, checkpoint inhibitors relieve immune suppression, potentially increasing the effectiveness of both therapies.
    • Such combinations are tested in animal models and cellular systems to assess effects on tumor regression, immune cell infiltration, and cytokine profiles.
    • Combination strategies help model potential clinical outcomes, such as improved progression-free survival and increased rates of complete response, but may also be associated with higher incidences of immune-related toxicities.
  • Translational and mechanistic research:

    • Preclinical studies utilize biosimilars that replicate the variable regions of clinical antibodies, allowing researchers to map interactions, dose responses, and biomarkers under standardized conditions.
    • Mechanistic approaches include flow cytometry to monitor immune populations, cytokine assays, and histological examination of tumor tissue to evaluate synergistic effects.

Additional relevant information:

  • Combination therapies are part of precision oncology, maximizing efficacy while reducing harm to non-tumor tissue.
  • The use of biosimilars allows broader access to research-grade agents, facilitating innovative study designs and reproducibility.

In summary, Sacituzumab biosimilars in combination with checkpoint inhibitor biosimilars are leveraged in immune-oncology research to dissect how targeted tumor cell killing and immune activation can be optimized for synergistic anti-cancer effects, using advanced preclinical models to inform future clinical strategies.

A Sacituzumab biosimilar can be used as both the capture reagent and the detection reagent in a bridging ADA ELISA to monitor a patient’s immune response against the therapeutic drug by detecting anti-drug antibodies (ADAs) specific to Sacituzumab in patient serum.

In a typical bridging ADA ELISA protocol:

  • The capture reagent (either the biosimilar or reference Sacituzumab) is first immobilized onto the ELISA plate, frequently via biotin-streptavidin interaction for high sensitivity.
  • Patient serum is added. If the sample contains ADAs against Sacituzumab (the biosimilar or original drug), these antibodies will bind to the immobilized drug.
  • The detection reagent is Sacituzumab (biosimilar or reference), conjugated to an enzyme such as horseradish peroxidase (HRP) or a dye. This labeled drug binds to the other “arm” of any bivalent ADA that has already bound to the capture reagent, thus forming a “bridge.”
  • Upon addition of the substrate, a measurable signal is produced, proportional to the amount of ADA present.

Key points:

  • Biosimilar or reference drug can be used as capture/detection reagents, provided they exhibit similar epitope and immunoreactivity profiles; using the biosimilar is justified if it has been thoroughly characterized to match the reference product.
  • The bridging format is highly sensitive for detecting bivalent (i.e., IgG class) ADAs because both antigen-binding sites of the ADA participate in the bridging reaction.
  • Specificity controls and use of high-purity biosimilar reagents are crucial to avoid false positives due to matrix effects or non-specific binding.

In summary, the Sacituzumab biosimilar serves as both “bait” and “hook” in the ELISA: immobilized to capture circulating ADAs, and labeled to detect those ADAs capable of binding two Sacituzumab molecules, thus enabling monitoring of patient immune response to either the reference or biosimilar drug.

References & Citations

1 Guerra E, Trerotola M, Aloisi AL, et al. Oncogene. 32(12):1594-1600. 2013.
2 Wen Y, Ouyang D, Zou Q, et al. Ann Transl Med. 10(24):1403. 2022.
3 Syed YY. Drugs. 80(10):1019-1025. 2020.
4 Goldenberg DM, Stein R, Sharkey RM. Oncotarget. 9(48):28989-29006. 2018.
5 Cardillo TM, Govindan SV, Sharkey RM, et al. Clin Cancer Res. 17(10):3157-3169. 2011.
6 Goldenberg DM, Cardillo TM, Govindan SV, et al. Oncotarget. 6(26):22496-22512. 2015.
7 Chang CH, Wang Y, Zalath M, et al. Mol Cancer Ther. 15(8):1910-1919. 2016.

Formats Available

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