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.
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.
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 commonly used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to quantitatively measure Sacituzumab concentration in serum samples. Their use is essential for ensuring comparability between test (biosimilar) and reference (originator) products in PK studies supporting biosimilar development.
Key roles and approach:
Calibration Standard: The biosimilar is typically selected as the analytical standard for the assay. This standard is used to generate a calibration (standard) curve by spiking known concentrations of the biosimilar into matrix-matched samples (e.g., human serum). These calibration points define the ELISA’s quantitative range, enabling the quantification of unknown sample concentrations by interpolating their signals against this curve.
Reference Controls: Both the biosimilar and the reference product (originator Sacituzumab govitecan) are used to prepare quality control (QC) samples at various concentrations across the assay’s range. These controls are run in parallel with test samples to assess assay accuracy, precision, and equivalence between the products.
PK Bridging ELISA Design:
The assay is validated using both biosimilar and reference products spiked into serum to demonstrate bioanalytical equivalence—that the assay measures both products with equivalent sensitivity and accuracy.
The method validation involves comparing assay performance for both products (standard curves, QC recoveries, etc.). If they meet pre-defined equivalence criteria (often confidence intervals between 0.8–1.25 of the ratio of means), a single method using the biosimilar as the standard can be justified for routine measurements.
Conducting all quantification with a single standard avoids inter-assay variability and is now regulatory best practice for biosimilar PK studies.
Quality Controls: QC samples using both test and reference materials ensure that the assay’s performance is monitored continuously for both biosimilar and originator antibodies, confirming c accuracy throughout the analysis.
Assay Principle Example:
Plates may be coated with anti-Sacituzumab antibodies or the relevant antigen (e.g., Trop-2 for Sacituzumab govitecan), which captures Sacituzumab in the sample. Detection is typically via an anti-human IgG conjugated to an enzyme, generating a colorimetric (or other) readout proportional to drug concentration.
Summary Table: Role of Biosimilars as Standards in PK ELISA
Assay Component
How Biosimilar is Used
Purpose
Calibration Standard
Prepared at defined concentrations in serum
Generates quantitation curve
Reference Controls
Spiked at QC levels using both biosimilar and originator
Validate comparability and accuracy
PK Bridging Design
Direct comparison of ELISA performance for both agents
Ensures assay works equally for both
References:
Regulatory and industry consensus recommend a single PK ELISA using the biosimilar as standard when analytical equivalence is demonstrated.
Typical ELISA kits for Sacituzumab are designed for native antibody in body fluids and can be adapted for serum PK if validated appropriately.
If more technical detail is needed (e.g., validation parameters such as accuracy, precision, recovery, or specific statistical approaches), or if your question concerns regulatory submission requirements, please clarify.
The primary models used to study in vivo tumor growth inhibition and analyze tumor-infiltrating lymphocytes (TILs) with research-grade anti-TROP-2 antibodies include both mouse syngeneic models and human tumor xenograft models (PDX or CDX), often in humanized immune system mice.
Key model types and examples:
Syngeneic mouse models: These use immunocompetent mice implanted with murine tumor cell lines naturally expressing Trop2, allowing for intact mouse immune system responses and robust analysis of TILs. Commonly used lines are CT26 (colon), EMT6 (breast), RENCA (renal), MC38 (colon), and B16F10 (melanoma). Syngeneic models are considered the gold standard for TIL characterization after immunotherapy because the host immune system is fully functional.
Human xenograft models (PDX/cell line-derived): These employ immunodeficient mice (e.g., NSG or NOD/SCID) implanted with either human cancer cell lines or patient-derived tumors that are TROP-2 positive. For mechanistic TIL studies, these may be augmented with human immune cell engraftment ("humanized mice"). Example models showing anti-TROP2 efficacy include Calu-3, NCI-H2170, HCC827, and EBC-1 for lung cancer.
Applications and supporting details:
Syngeneic models (e.g., CT26, EMT6, RENCA, MC38) enable investigation of how anti-TROP-2 antibodies affect tumor growth and alter endogenous mouse TIL populations, including profiling of CD8+ T cells, regulatory T cells, and myeloid cells before and after treatment. Their immune context makes them especially valuable for dissecting antitumor immune mechanisms and response stratification. These models have supported studies of immune checkpoint inhibitors and antibody therapies, with detailed TIL analysis.
Humanized xenograft models allow human anti-TROP-2 antibodies (or ADCs) to be tested against human tumors, with some platforms providing a partial or fully human immune context for TIL analysis. These are essential for evaluating direct effects on human TROP-2 and downstream immune infiltration, especially when using human-specific antibodies. Various antibodies, such as AR47A6.4.2 or 7E6, and ADCs like datopotamab deruxtecan, have been used in these models to demonstrate tumor inhibition.
Limitations: Syngeneic models are limited to murine TROP-2, so some human antibodies do not cross-react, necessitating surrogate antibodies or humanized models. Humanized mice are costly and have partial immune reconstitution, affecting depth of TIL analysis.
Summary Table: Models for In Vivo Anti-TROP-2 Antibody Testing and TIL Characterization
Model Type
Example Tumors
Immune Context
Antibody Source
Typical Use Case
TIL Characterization
Syngeneic (mouse-mouse)
CT26, EMT6, RENCA
Fully murine
Mouse or cross-reactive
Immune mechanism, responder vs non-responder analysis
High
Human xenograft (PDX/CDX)
Calu-3, HCC827
Immunodeficient
Human-specific
Tumor efficacy, growth inhibition
Limited (unless humanized)
Humanized immune PDX/CDX
Human tumors/cell lines
Human (reconstituted) + mouse
Human-specific
Efficacy + human TIL response
Moderate
Syngeneic mouse tumor models (e.g., CT26, MC38, EMT6, RENCA, B16F10) are most commonly used for TIL profiling after antibody administration.
PDX/Xenograft models are used for human-specific anti-TROP-2 reagents, especially when combined with humanized mouse strains for TIL studies.
In summary, syngeneic mouse models are broadly used for TIL characterization after anti-TROP-2 antibody treatment, while humanized xenograft models are necessary for human antibody and human TIL analyses in vivo.
Researchers use the Sacituzumab biosimilar in combination with other checkpoint inhibitor biosimilars (such as anti-CTLA-4 or anti-LAG-3) to investigate potential synergistic anti-tumor effects within complex immune-oncology models, particularly in preclinical and translational studies.
Essential context and supporting details:
Sacituzumab Function: Sacituzumab is a monoclonal antibody that targets the TROP-2 antigen, which is highly expressed on many epithelial tumors but minimally on normal cells, making it a selective and valuable tool for cancer immunotherapy research. The biosimilar form provides an affordable and widely available option for laboratory research.
Combination Rationale: The underlying logic of combining multiple checkpoint inhibitors (such as anti-CTLA-4, anti-PD-1, or anti-LAG-3 biosimilars) with an agent like Sacituzumab is to target different components of the immune response. For example, anti-CTLA-4 mainly enhances T cell activation in lymph nodes, while anti-PD-1/PD-L1 acts to prevent tumor-mediated suppression of cytotoxic T cells at the tumor site. Combining these with Sacituzumab, which delivers targeted cytotoxicity via TROP-2, may lead to enhanced immune activation and tumor cell killing.
Research Strategies:
In Vitro and In Vivo Studies: Researchers conduct laboratory (in vitro) and animal (in vivo) studies to explore whether combining these agents results in increased T cell activation, improved tumor cell lysis, and better anti-tumor response compared to monotherapy.
Synergy Assessment: These studies often measure synergistic effects by observing tumor regression rates, immune cell infiltration, cytokine release, and overall survival in model systems.
Use of Biosimilars: Biosimilars, including those for Sacituzumab and checkpoint inhibitors, reduce costs and make such combination studies more feasible on a large scale. This allows researchers to perform comparative studies to optimize dosing strategies and explore efficacy across different cancer types.
Translational and Clinical Context: While clinical data are still emerging, preclinical evidence strongly suggests that dual or triple combinations can result in superior anti-tumor activity, especially in tumors with poor baseline responsiveness to checkpoint blockade alone.
Additional relevant information:
Ongoing clinical trials are actively exploring these combinations in numerous cancer indications, expanding the scope of Sacituzumab-based regimens beyond established uses.
The combination approach also highlights the importance of biomarker-driven research (e.g., TROP-2 expression, tumor immune microenvironment profiling) to select patients most likely to benefit.
In summary, researchers utilize the Sacituzumab biosimilar alongside checkpoint inhibitor biosimilars to exploit complementary mechanisms of action, aiming to maximize anti-tumor effects in sophisticated preclinical immune-oncology models and inform future clinical application.
A Sacituzumab biosimilar can serve as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against Sacituzumab by detecting antibodies the patient generates against the therapeutic drug.
How this works in a bridging ADA ELISA:
Core principle: The bridging ELISA is designed to detect bivalent ADAs—antibodies produced by the patient's immune system against the therapeutic antibody (in this case, Sacituzumab). The approach uses the drug (here, the Sacituzumab biosimilar) in two forms: one immobilized on the ELISA plate ("capture") and the other labeled for detection ("detection").
Assay setup:
The ELISA plate is first coated with the Sacituzumab biosimilar (capture reagent) to immobilize it on the surface.
The patient's serum sample is added. If the patient has developed anti-Sacituzumab antibodies, these bivalent ADAs will bind to the immobilized Sacituzumab.
A reporter-labeled Sacituzumab biosimilar (e.g., biotin- or HRP-conjugated; detection reagent) is then added. The ADA, if present, will bridge between the immobilized Sacituzumab and the labeled Sacituzumab, forming a "bridge" complex.
After washing steps, a substrate (such as TMB when using HRP) is added and the resulting color change is measured, reflecting the amount of ADA present in the patient sample.
Key points about use of a Sacituzumab biosimilar:
The biosimilar is used in place of the innovator (reference) drug to ensure the assay detects ADAs that react to the same epitope structure as the therapeutic product.
The biosimilar can be effectively labeled for detection (HRP/biotin) due to its high purity (typically >95%) and its fully human IgG1 structure, similar to the marketed Sacituzumab.
This design allows for sensitive, high-throughput ADA detection, with specificity determined by the quality of the biosimilar and labeling.
Summary Table: Use of Sacituzumab Biosimilar in Bridging ADA ELISA
Component
Role of Sacituzumab Biosimilar
Capture reagent
Coated on ELISA plate to capture ADAs
Detection reagent
Labeled (e.g., HRP, biotin) to detect bridging ADAs
Assay readout
Signal proportional to ADA amount in sample
Relevant context:
ADA bridging ELISAs are a gold standard for immunogenicity testing in both biosimilar and reference biotherapeutics.
The choice of biosimilar in these assays ensures the immunogenicity profile measured closely matches that expected for the therapeutic or its biosimilar.
This approach is adaptable—either the biosimilar or the originator drug may be used as capture/detection reagent, provided they are functionally and structurally equivalent.
In summary: A Sacituzumab biosimilar is immobilized on the plate and/or labeled as a detection reagent in a bridging ADA ELISA, enabling the quantification of patient anti-drug antibodies specific for Sacituzumab, thus monitoring the patient’s immune response to the therapeutic 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.