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 Labetuzumab. hMN-14 (Labetuzumab) activity is directed against
human CEACAM5.
Background
CEACAM5 is a member of the carcinoembryonic antigen-related cellular adhesion molecules
(CEACAM) family1. In normal tissues, CEACAM5 functions as an adhesion molecule and has
roles in differentiation, immune modulation, and anoikis inhibition. Additionally, CEACAM5
likely inhibits terminal differentiation and is also involved in activation of CD8+ suppressor T
cells. Dysregulation of CEACAM5 has been implicated in tumor initiation, progression,
invasion, and metastasis1,2 as well as in immune-related disease, particularly ulcerative colitis1.
CEACAM5 is a clinical biomarker for colon cancer1 and is also a promising target of anti-cancer
immunotherapy2. CEACAM5 is a mediator of CEACAM1-induced apoptosis, and preventing
this interaction may prove beneficial to cancer treatment by blocking immune evasion by cancer
cells.
hMN-14 (Labetuzumab; IMMU-130) is a CEACAM5 antibody-drug conjugate composed of a
humanized CEACAM5 monoclonal antibody conjugated via the hydrolysable linker CL2A to 7-
ethyl-10-hydroxycamptothecin (SN-38), a potent topoisomerase I inhibitor3,4. The antitumor
activity of labetuzumab has been tested for colorectal4,7 and prostate cancers2.
This non-therapeutic biosimilar is not a drug conjugate and thus does not contain the drug SN-
38.
Antigen Distribution
In normal tissues, CEACAM5 is expressed on gastric and intestinal
mucous cells, particularly esophageal squamous epithelia, as well as in the pancreas. In cancers,
CEACAM5 is expressed on the majority of solid tumors and is upregulated in a variety of human
epithelial malignancies (gastric, colorectal, pancreatic cancers) as well as in non-small cell lung
cancer and melanoma. Tumors release CEACAM5 in a soluble form that can be used as a
biomarker for cancer.
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 Labetuzumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by serving as the single analytical standard against which both the biosimilar and the reference (originator) drugs are quantitatively measured in serum samples. This practice ensures consistency, minimizes variability, and enables direct bioanalytical comparison in PK similarity studies.
Context and Supporting Details:
In PK bridging ELISA development, a single PK assay is usually established with a single analytical standard—most commonly the biosimilar itself—used to generate the calibration curve for quantifying drug concentrations in all serum samples (test, reference, and control).
The biosimilar standard is prepared at known concentrations in human serum and serially diluted to cover the assay’s full dynamic range (e.g., 50–12800 ng/mL in one study).
All sample types (e.g., patient samples containing either the reference Labetuzumab or biosimilar) are measured against the same calibration curve, ensuring bioanalytical equivalence and robust cross-comparability.
Procedure and Rationale:
The biosimilar standard acts as a reference calibration material for quantifying both the test biosimilar and the originator drug in serum, which is critical to minimize confounding variability and ensure the accuracy and precision required for PK bioequivalence assessment.
This methodology undergoes rigorous validation for parameters such as specificity, accuracy, precision, linearity, and range in accordance with regulatory guidelines. Performance is evaluated with multiple sets of biosimilar and reference product spiked QC samples, all measured against the biosimilar standard curve.
The use of a single standard (typically the biosimilar) in bridging ELISA is supported by industry consensus: it avoids the potential analytical bias that could result from the use of separate standards (i.e., reference and biosimilar), thus supporting regulatory requirements for robust comparability.
Additional Information:
This PK bridging strategy is not unique to Labetuzumab; it is also used for other monoclonal antibodies and their biosimilars (e.g., Trastuzumab), as shown in multiple published studies and validated ELISA kits.
Calibration standards are critical for generating accurate serum concentration measurements, which then support calculation of key PK endpoints such as Cmax (maximum concentration) and AUC (area under the concentration-time curve).
Summary Table: Usage of Biosimilar as PK ELISA Standard
Component
Role in PK Bridging ELISA
Rationale
Biosimilar STD
Calibration curve for all sample quantification
Minimizes analytical variability
Reference Drug
Assayed against biosimilar standard curve
Ensures. bioanalytical comparability
QC Samples
Biosimilar and reference products spiked in serum
Validates assay performance
Key Point: Using research-grade Labetuzumab biosimilars as calibration standards enables unbiased, robust measurement of serum drug concentrations for both the biosimilar and the originator, fulfilling the methodological requirements for PK comparability studies in biosimilar development.
The primary in vivo models where a research-grade anti-CEACAM5 antibody is administered to study tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) include syngeneic mouse models, xenograft/PDX models in immunocompromised mice, and humanized mouse models.
Key Models and Their Context:
Syngeneic Models:
Use mouse tumor cells implanted into immunocompetent mice of the same strain, which allows evaluation of immune responses, including characterization of TILs after antibody administration.
Example: One study used a syngeneic mouse model engineered to express human CD47 and SIRPα to test a bispecific antibody targeting CEACAM5, allowing analysis of both tumor growth inhibition and immune cell composition in the tumor microenvironment.
Xenograft/PDX Models:
Involve transplantation of human CEACAM5-positive tumor cells (or patient-derived xenografts) into immunocompromised mice (such as NOD-SCID or SCID mice), with antibody therapies assessed for growth inhibition.
These models focus on tumor-targeting efficacy, but TIL characterization is limited unless the mice are "humanized."
Humanized Mouse Models:
Immunocompromised mice engrafted with human hematopoietic stem cells or peripheral blood mononuclear cells (PBMCs), allowing establishment of a human-like immune system.
Anti-CEACAM5 antibody or ADC administration can be used to monitor both tumor growth and interaction with human TIL populations.
Studies such as those using anti-CEACAM5 CAR-T cells often use xenograft tumors in mice reconstituted with aspects of the human immune system to allow TIL characterization.
Summary Table: Models in Anti-CEACAM5 Antibody Research
Model Type
Host Immune System
Example Use
TILs Characterization
Syngeneic
Murine
Human CEACAM5-expressing cells in engineered mice
Yes, mouse TILs (may require knock-in for human receptors)
Xenograft/PDX
Immunocompromised
Human tumor cells in SCID/NOD-SCID mice
Limited, unless humanized
Humanized
Human (in mouse)
Human tumors in mice with human immune system
Yes, human TILs
Additional Notes:
Syngeneic and humanized models are especially used when the goal includes detailed characterization of TILs following antibody treatment, as their immune systems allow for immune cell infiltration and analysis.
Xenograft/PDX models are dominant for measuring pure tumor growth inhibition, with antibody-drug conjugates (such as SGN-CEACAM5C/SAR445953) showing efficacy in these settings across colorectal, gastric, PDAC, and lung cancer.
Characterization of TILs typically involves flow cytometry or immunohistochemistry on excised tumors post-treatment in these models.
For specific study designs or published examples, search results indicate that anti-CEACAM5 therapeutics have been tested in:
NOD-SCID xenograft models (colon cancer)
Syngeneic mouse models engineered for human targets
Humanized mouse models for CAR-T therapy and TIL analysis
In summary, researchers primarily use syngeneic models (with engineered expression of human antigens), humanized mouse models, and xenograft/PDX models in immunocompromised mice to study anti-CEACAM5 antibody-mediated tumor growth inhibition and to characterize TILs. The specific choice depends on whether immune response characterization (syngeneic/humanized) or tumor growth inhibition (xenograft/PDX) is the primary endpoint.
Researchers use Labetuzumab biosimilars—which target the cancer-associated protein CEACAM5—in combination with other checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 biosimilars) to investigate potential synergistic immune-oncology effects, particularly aiming to overcome tumor resistance and enhance anti-tumor immunity.
In these studies:
Labetuzumab biosimilars serve as reliable, cost-effective agents for preclinical models, reproducing the original drug's specificity for CEACAM5-positive tumors, including colorectal, lung, and pancreatic cancers.
Researchers combine Labetuzumab with checkpoint inhibitors to test whether the dual targeting of tumor antigens (e.g., CEACAM5) and immune regulatory pathways (e.g., CTLA-4 or LAG-3) produces greater inhibition of tumor growth than either agent alone.
Experiments commonly use animal models or advanced cell-based systems to simulate complex tumor microenvironments. These models allow evaluation of immune cell activation patterns and tumor regression, helping elucidate mechanisms behind the combined actions.
Synergies and Mechanistic Insights:
The goal of combining a CEACAM5-targeted antibody (like Labetuzumab biosimilar) with immune checkpoint inhibitors is to simultaneously eliminate tumor cells and release immune suppression, which can result in more robust and durable anti-tumor responses.
Mechanistic studies show that different checkpoint combinations (such as PD-1/CTLA-4 vs. PD-1/LAG-3) trigger distinct immune cell activation profiles. For instance, anti-PD-1/LAG-3 combination therapy increases CD4+ T-cell activity and decreases regulatory T-cell (Treg) function, while anti-PD-1/CTLA-4 tends to directly activate cytotoxic CD8+ T cells. Although these specific pathways are described for checkpoint inhibitor combinations, similar immune profiling approaches are applied in studies where Labetuzumab biosimilars are added to checkpoint regimens.
Preclinical Research Approach:
Biosimilars offer scalable access and reproducibility, making them ideal for hypothesis-driven studies on efficacy and immune modulation.
Flow cytometry and immunohistochemistry are used to evaluate how combination therapy changes tumor immune infiltrate composition, the expression of immune exhaustion markers, and rates of tumor progression or regression.
Research context and limitations:
Labetuzumab biosimilars are strictly for research use; no clinical protocols using these combinations are currently approved. All results are preclinical and exploratory, designed to inform possible future therapy developments.
In summary, combining Labetuzumab biosimilars with other checkpoint inhibitors is a strategic method to explore and optimize immune-oncology therapies, with a focus on synergy, immune escape inhibition, and modeling complex interactions in experimental cancer systems.
A Labetuzumab biosimilar is typically used in bridging anti-drug antibody (ADA) ELISA assays as both the capture and detection reagent to monitor a patient’s immune response against the therapeutic drug. In this format, the biosimilar is immobilized on the microtiter plate and also labeled (e.g., with enzyme or biotin) for detection, enabling sensitive measurement of ADAs specific to labetuzumab in patient serum.
How the bridging ADA ELISA works with Labetuzumab biosimilar:
Plate coating: The Labetuzumab biosimilar is coated directly on the ELISA plate (or, alternately, a biotinylated version can be captured on a streptavidin-coated plate).
Sample incubation: Patient serum samples—potentially containing anti-Labetuzumab antibodies—are added to the wells.
Detection reagent: A labeled (e.g., HRP-conjugated or biotinylated) Labetuzumab biosimilar is then added. If ADAs are present, one arm binds to the plate-coated Labetuzumab while the other arm binds to the labeled detection Labetuzumab, forming a “bridge”.
Signal generation: After washing, a substrate for the detection enzyme is added. Signal development indicates ADA presence and can be quantified.
Key details and context:
This “bridging” design specifically relies on the bivalent nature of human ADAs, which must simultaneously bind both the immobilized and the labeled Labetuzumab, ensuring high specificity for antibody-mediated reactions.
Using the biosimilar version as both capture and detection reagent is valid due to demonstrated equivalence in structure and immunoreactivity with the originator, so immune responses detected are relevant for both reference and biosimilar products.
Interference from endogenous serum components (other antibodies, target antigen) may affect assay specificity; optimization is essential for clinical application.
The method is widely used for various monoclonal antibody therapies and their biosimilars, following the same principles as described with other mAbs like Metuzumab and Adalimumab.
Summary Table: Labetuzumab Biosimilar in Bridging ADA ELISA
Step
Role of Labetuzumab Biosimilar
Purpose
Plate coating
Immobilized on ELISA plate
Captures ADAs from patient serum
Detection
Labeled (e.g., biotin, HRP) version added after serum incubation
Detects bridged ADAs by signal report
Readout
Substrate added for enzyme reaction (HRP/TMB)
Quantifies ADA levels
This approach provides a high-sensitivity, high-specificity platform for ADA monitoring against both Labetuzumab reference and biosimilar products.
References & Citations
1. Thomas J, Klebanov A, John S, et al. Genes Cancer. 14:12-29. 2023.
2. DeLucia DC, Cardillo TM, Ang L, et al. Clin Cancer Res. 27(3):759-774. 2021.
3. Sharkey RM, Juweid M, Shevitz J, et al. Cancer Res. 55(23 Suppl):5935s-5945s. 1995.
4. Govindan SV, Cardillo TM, Moon SJ, et al. Clin Cancer Res. 15(19):6052-6061. 2009.
5. Shinmi D, Nakano R, Mitamura K, et al. Cancer Med. 6(4):798-808. 2017.
6. Iwano J, Shinmi D, Masuda K, et al. Drug Metab Dispos. 47(11):1240-1246. 2019.
7. Govindan SV, Cardillo TM, Rossi EA, et al. Mol Pharm. 12(6):1836-47. 2015.
8. Sharkey RM, Govindan SV, Cardillo TM, et al. Mol Cancer Ther. 17(1):196-203. 2018.
9. de Gooyer JM, Elekonawo FMK, Bremers AJA, et al. Nat Commun. 13(1):2621. 2022.
10. Imberti C, De Gregorio R, Korsen JA, et al. J Nucl Med. 65(7):1043-1050. 2024.