Anti-Human CD19 (Loncastuximab) [Clone ADCT-402] – Fc Muted™

Anti-Human CD19 (Loncastuximab) [Clone ADCT-402] – Fc Muted™

Product No.: C3165

- -
- -
Product No.C3165
Clone
ADCT-402
Target
CD19
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
B-lymphocyte antigen CD19, B-lymphocyte surface antigen B4, Differentiation antigen CD19, T-cell surface antigen Leu-12
Isotype
Human IgG1κ
Applications
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
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 ?
WB
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 Loncastuximab. ADCT-402 specifically targets the CD19 antigen, which is expressed on the surface of B cells.
Background
CD19 is a 95 kDa type I transmembrane glycoprotein found on the surface of B cells at all stages of their growth into plasma cells. It works as a co-receptor alongside the B cell receptor (BCR) and is essential in activating, growing, and transforming B cells. CD19 assists in lowering the threshold for BCR signaling, making B cells more sensitive to antigens. Because of its crucial involvement in the life of B cells, CD19 is an important marker for identifying B cell lineage and is a target for treatments focusing on B cell-related cancers1-7.

ADCT-402, also known as loncastuximab tesirine, is an antibody-drug conjugate (ADC) that targets CD19-expressing cells. It is made up of a humanized monoclonal antibody that targets CD19 and is linked to a pyrrolobenzodiazepine (PBD) dimer cytotoxin. When ADCT-402 binds to CD19- expressing cells, it is taken inside the cell, and the cytotoxin is released, leading to DNA crosslinking and cell death. This targeted approach allows for strong and selective anti-tumor activity against CD19-expressing blood cancers, such as B-cell lymphomas and leukemias8,9.

This non-therapeutic biosimilar is not a drug conjugate and thus does not contain the cytotoxin PDB.

Antigen Distribution
CD19 is expressed on all B lineage cells, from early pre-B cells to mature B cells.
Ligand/Receptor
B-cell antigen receptor complex (BCR); CR2/CD21, CD81, IFITM1/CD225, VAV, GRB2, SOS, PLCG2, LYN
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Cancer
.
Costimulatory Molecules
.
Immuno-Oncology
.
Immunology
.
Tumor Suppressors

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 Loncastuximab biosimilars are commonly used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISA assays to accurately measure drug concentration in serum samples. In these assays, the biosimilar is used to generate a standard curve, enabling quantification of both the biosimilar and its reference originator in test samples.

Key roles and procedures:

  • Calibration Standard Function: The research-grade biosimilar is prepared at known concentrations in serum to create a calibration (standard) curve. This curve is essential for converting ELISA signal intensity into absolute drug concentrations in patient or research samples.

  • Bioanalytical Comparability: Before routine use, a method qualification study is performed. This study directly compares the biosimilar and reference product in the ELISA, ensuring they are analytically equivalent within the method. Both products are tested across multiple concentration ranges, analysts, and days to verify similar assay response (signal, accuracy, and precision).

  • Assay Validation: Once equivalency is confirmed, the biosimilar is typically designated as the single analytical standard for the assay. This minimizes variability, simplifies quantitative analysis, and eliminates the need for multiple reference curves or crossover corrections.

  • Reference Controls: Quality control (QC) samples are prepared using both the biosimilar and reference product at various concentrations to monitor assay performance, confirm consistent quantitation, and provide ongoing verification during assay runs. These QC samples help ensure the assay's reliability across different batches and over time.

  • Bridging ELISA Specifics: In a bridging ELISA, the assay design detects free Loncastuximab via its binding to specific capture and detection reagents (such as anti-idiotype antibodies or target protein). The biosimilar (or reference) is used both to establish the calibration curve and to serve as a control to verify bioanalytical equivalency.

  • Regulatory and Quality Context: International and in-house reference standards, when used correctly, improve the harmonization and reproducibility of PK assays, allowing robust data generation for regulatory submissions and biosimilar development.

Summary Table: Use of Research-Grade Biosimilar in PK Bridging ELISA

RoleFunction
Calibration StandardForms standard curve for quantitation of drug concentration in samples
Reference ControlVerifies ongoing assay performance and precision
ComparabilityConfirms similar assay response vs. reference biologic prior to use

In practice:

  • Research-grade Loncastuximab biosimilar is serially diluted in serum to generate the standard curve.
  • Study serum samples are run in parallel with standards in the bridging ELISA.
  • QC samples with known concentrations of biosimilar and/or reference drug are included in every assay run for method control.

Note: The use of these biosimilars as standards is predicated on rigorous analytical demonstration of equivalency to the reference product within the specific ELISA method. If a formally recognized international reference standard exists, this may be preferred to harmonize results between laboratories.

The most widely used in vivo models for evaluating research-grade anti-CD19 antibodies in studies of tumor growth inhibition and tumor-infiltrating lymphocytes (TILs) are human tumor xenograft models (in immunodeficient mice), syngeneic mouse models, and humanized mouse models.

Key Model Types:

  • Human Tumor Xenografts (Immunodeficient Mice):

    • Human B cell tumor lines (e.g., BJAB, BJAB/ADR) are implanted in immunodeficient mice, such as BALB/c nude or SCID mice.
    • Anti-CD19 antibodies (including research-grade formats and engineered variants like anti-CD19(Fab)-LDM or MEDI-551) are administered to assess the inhibition of human tumor growth.
    • These models are commonly used for tumor growth inhibition studies but typically lack a fully functional immune system for TIL analysis.
  • Syngeneic Models (Immunocompetent Mice):

    • Murine B cell tumor lines expressing mouse or engineered human CD19 are implanted in immunocompetent, genetically identical mice.
    • These models allow the study of murine anti-CD19 antibodies and robust characterization of the immune microenvironment and endogenous TILs.
    • Administration of research-grade murine anti-CD19 antibodies can be used to assess efficacy and TIL responses.
  • Humanized Mouse Models:

    • These mice are engrafted with human hematopoietic stem cells or a human immune system and then implanted with human tumors that express CD19.
    • Anti-CD19 antibodies (human-specific) are administered, enabling the assessment of both tumor inhibition and the impact on human TILs within the mouse tumor microenvironment.
    • These models are often considered the gold standard for preclinical immunotherapy studies because they allow TIL characterization in the context of a human immune system.
Model TypeTumor CellsImmune SystemAntibody UtilityTIL Characterization
Xenograft (nude/SCID)HumanDeficient (mouse)Human-specificLimited
SyngeneicMouse (± human antigen)ImmunocompetentMouse-specificRobust (murine TILs)
HumanizedHumanHumanizedHuman-specificRobust (human TILs)

Summary:

  • Xenograft models are most frequently used for tumor growth inhibition by anti-CD19 antibodies, but have limited TIL analysis due to immune deficiency.
  • Syngeneic models allow detailed TIL analysis but require murine-format antibodies or murine tumors expressing human CD19.
  • Humanized mouse models enable both robust tumor inhibition and thorough characterization of human TILs with research-grade human anti-CD19 antibodies.

The choice of model depends on whether the primary goal is to examine antibody efficacy on tumor growth, characterize immune/TIL responses, or both. For TIL studies with human antibodies, humanized models are the most appropriate. For mechanistic studies of immune interactions, syngeneic mouse models are preferred.

Researchers use Loncastuximab tesirine (a CD19-targeted antibody-drug conjugate, or ADC) in combination with other immunotherapy agents—including checkpoint inhibitors like anti-CTLA-4 or anti-LAG-3—in preclinical and computational models to study possible synergistic anti-tumor effects and mechanisms of resistance in complex immune-oncology settings.

Key methods and findings include:

  • Quantitative Systems Pharmacology (QSP) Modeling: Researchers construct in silico models combining loncastuximab with T-cell engagers and checkpoint inhibitors to predict how these combinations affect tumor volume and response in virtual patient populations. The latest QSP models allow simulation of various dosing regimens and heterogeneity in immune cell composition, providing predictions of additive or synergistic tumor responses (measured by "combination index") and resistance patterns as functions of tumor biology and immune context.

  • Mechanistic Rationale for Combination: The primary rationale for combining ADCs like Loncastuximab with checkpoint inhibitors is rooted in their distinct mechanisms:

    • Loncastuximab directly kills CD19+ tumor cells by delivering a cytotoxic payload.
    • Checkpoint inhibitors (such as anti-CTLA-4, anti-LAG-3, and anti-PD-1/PD-L1) aim to restore exhausted T cell function and enhance anti-tumor immunity.

    Combining these products is hypothesized to increase overall anti-tumor efficacy by both increasing tumor cytolysis and alleviating immune exhaustion, potentially yielding synergy beyond either agent alone.

  • Synergy Measurement and Predictive Indices:

    • Researchers calculate a combination index (CI) to quantify synergy (with CI > 1 indicating more-than-additive effects). Modeling work has shown, for example, that combining Loncastuximab with T-cell-engaging bispecific antibodies yields synergistic or additive effects on tumor reduction, suggesting that combining Loncastuximab with checkpoint inhibition may behave similarly.
  • Preclinical & Clinical Context:

    • In preclinical models, combination immunotherapies have shown improved tumor clearance compared to monotherapy—particularly if targeting non-overlapping aspects of T-cell regulation (e.g., CTLA-4, PD-1, LAG-3).
    • Clinical trials have established that combined checkpoint blockade (e.g., anti-PD-1 plus anti-CTLA-4 or anti-LAG-3) can lead to improved response rates at the cost of increased adverse events.
  • Factors Studied in Models:

    • Effects of immune cell ratios (T:B cell ratio), tumor size, antigen expression density, and mechanisms of resistance (such as emergence of antigen-negative clones or immune suppression) are systematically varied to predict which patients might benefit most from combination regimens and under what circumstances resistance might occur.
  • Experimental Extensibility:

    • While most direct data combine Loncastuximab with T-cell bispecifics (rather than checkpoint inhibitors), the modeling and mechanistic insights are generalizable to other checkpoint inhibitors such as anti-CTLA-4 and anti-LAG-3 biosimilars, which are under active translational investigation.

In summary: Researchers use complex computational models, alongside preclinical animal studies, to evaluate Loncastuximab combinations with other checkpoint inhibitors. These approaches help to identify synergistic effects, optimize dosing regimens, and select rational combinations for clinical trials to enhance the efficacy of immune-oncology therapies in lymphoma and other cancers.

A Loncastuximab biosimilar can be used as either the capture or detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response to the therapeutic drug by leveraging the bivalent nature of patient-derived ADAs, which can simultaneously bind two drug molecules.

Bridging ADA ELISA Context and Principle:

  • In a typical bridging ELISA format for ADA detection, the assay uses the drug or its biosimilar labeled in two different ways (e.g., one biotinylated, one HRP-conjugated, or similar) to "bridge" ADAs present in patient samples.
  • The ADA in a patient sample binds to the capture reagent (e.g., plate-coated Loncastuximab biosimilar) with one antigen-binding site, while its other binding site captures the detection reagent (e.g., labeled Loncastuximab biosimilar), completing the bridge.
  • Only samples containing ADA that recognize Loncastuximab will generate a detectable signal, ensuring specificity for anti-drug immune responses.

Use of the Biosimilar Reagent:

  • A biosimilar of Loncastuximab provides an analytically equivalent surrogate of the innovator drug for use in the assay.
  • The biosimilar can be:
    • Coated on the ELISA plate (as capture reagent), where it immobilizes any ADA present in the sample.
    • Labeled (e.g., with HRP or biotin, as detection reagent), which binds the other ADA paratope.
  • Because patient-derived ADA typically recognizes the therapeutic drug regardless of originator or biosimilar formulation (assuming structural similarity), the biosimilar is effective in the assay.

Advantages and Considerations:

  • Bridging ELISAs are the gold standard for ADA screening in clinical immunogenicity programs due to their sensitivity and ability to detect bivalent, specific ADAs.
  • Using a biosimilar in place of the originator can conserve expensive clinical material, simplify regulatory concerns, and validate biosimilar comparability.
  • It's essential to confirm analytical equivalence (matching glycosylation, epitope recognition) to avoid missing ADA populations specific to unique attributes of the originator.

Summary Table: Loncastuximab Biosimilar Use in ADA Bridging ELISA

RoleFunctionNotes
CapturePlate-bound to immobilize ADA from patientMust preserve epitope for ADA recognition
DetectionLabeled form for detection of ADA bridgeShould not interfere with ADA binding
BothConfirm specificity and cross-reactivityValidates biosimilar as functionally equivalent

In sum, a Loncastuximab biosimilar enables the ADA bridging ELISA by acting as both the target and detection agent for anti-drug antibodies, thus allowing clinicians to monitor immunogenicity by detecting ADAs generated by the patient's immune system in response to the therapeutic drug.

References & Citations

1. Morbach H, Schickel JN, Cunningham-Rundles C, et al. J Allergy Clin Immunol. 2016;137(3):889-898.e6.
2. Wang K, Wei G, Liu D. Experimental Hematology & Oncology. 2012;1(1):36.
3. Ishiura N, Nakashima H, Watanabe R, et al. Eur J Immunol. 2010;40(4):1192-1204.
4. Krop I, Shaffer AL, Fearon DT, Schlissel MS. J Immunol. 1996;157(1):48-56.
5. Depoil D, Weber M, Treanor B, et al. Sci Signal. 2009;2(63):pt1.
6. Otero DC, Anzelon AN, Rickert RC. J Immunol. 2003;170(1):73-83.
7. Li X, Sandoval D, Freeberg L, Carter RH. J Immunol. 1997;158(12):5649-5657.
8. Breton CS, Nahimana A, Aubry D, et al. J Hematol Oncol. 2014;7:33.
9. Zammarchi F, Havenith KE, Sachini N, et al. Mol Cancer Ther. 2024;23(4):520-531.
10. Tarantelli C, Wald D, Munz N, et al. Published online August 18, 2023:2023.08.17.553668.
General Western Blot Protocol

Certificate of Analysis

Formats Available

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