Humanized antibody derived from mouse clone targeting CD25
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 Camidanlumab but is not linked to the toxin warhead SG3199. Camidanlumab antibody activity is directed against Human CD25.This product is for research use only.
Background
CD25 is the alpha-chain of the interleukin-2 receptor1, which regulates normal immune
function2, and is involved in signal transduction, cell growth and survival1. CD25 is
considered an attractive target for conjugated antibody chemotherapeutic development
because it is overexpressed in various hematologic malignancies and is associated with
poor prognosis.
Antibody drug conjugates (ADC) use tumor-associated surface antigens to specifically
target cancer cells with cytotoxic agents. Human therapeutic grade Camidanlumab is an
ADC composed of a human CD25-directed antibody (HuMax-TAC) that is maleimide-
conjugated at reduced interchain cysteines via a cathepsin-cleavable valine-alanine
linker to a PBD toxin warhead (SG3199)3.
Camidanlumab has a strong binding affinity to CD25-positive human anaplastic large
cell lymphoma derived cell lines. This research grade biosimilar has the same specificity
as the original therapeutic antibody but lacks the conjugated PBD toxin warhead.
Antigen Distribution
CD25 is expressed by approximately 30% of human peripheral
blood B cells, particularly those belonging to the memory B cell population. Additionally,
CD25 is expressed on the cell surface of many lymphomas, including classical Hodgkin
lymphoma and non-Hodgkin lymphoma.
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 Camidanlumab biosimilars serve as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by establishing the standard curve against which serum drug concentrations are quantified, ensuring assay comparability between biosimilar and reference products.
In a PK bridging ELISA:
Calibration standards are prepared by serially diluting research-grade biosimilar Camidanlumab into matrix-matched serum samples, generating a range of known concentrations (e.g., 50–12,800 ng/mL).
These standards are run alongside patient serum samples, and the assay quantifies the drug concentration by comparing the sample’s signal to the calibration curve generated from the standards.
Reference controls (i.e., known concentrations of biosimilar and/or reference drug) are included as quality controls (QCs) in assay runs to monitor performance, accuracy, and precision.
Why biosimilars are used:
Industry consensus recommends using a single analytical standard—typically the biosimilar—as the calibrator for both biosimilar and reference products in PK bridging ELISAs, to minimize variability and facilitate cross-comparison.
Before adopting this approach, a method qualification study establishes bioanalytical comparability (precision, accuracy, equivalence) between the biosimilar and reference, confirming that the biosimilar is suitable as a calibration standard.
This single-standard approach reduces crossover test requirements, streamlines data comparison, and supports regulatory expectations for PK similarity studies.
Assay procedure:
Coated wells with capture antibody specific for Camidanlumab bind drug in samples and standards.
A labeled detection antibody binds captured Camidanlumab, and substrate is added to produce a measurable signal proportional to drug concentration.
Sample concentrations are interpolated from the calibration curve established with biosimilar standards.
Summary of roles:
Calibration standards: Establish the quantification range and standard curve using research-grade biosimilar Camidanlumab.
Reference controls/QCs: Verify assay performance and comparability across all test samples with known controls of biosimilar and/or reference product.
This methodological rigor ensures that drug concentrations measured in PK studies are accurate, comparable, and compliant with regulatory guidance for biosimilar development.
The primary in vivo models where research-grade anti-CD25 antibodies are administered to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization are syngeneic mouse models and, increasingly, humanized mouse models.
Key model types:
Syngeneic mouse models
Murine tumors (e.g., MC38, CT26, B16) are implanted into immunocompetent mice of the same genetic background.
Anti-mouse CD25 antibodies (such as clone PC61) are administered, systemically or intratumorally, to deplete regulatory T cells (Tregs) within the tumor microenvironment.
These models allow preservation and monitoring of the native immune system, enabling the study of TIL expansion, phenotype, and functional changes following Treg depletion.
Tumor growth is measured, and TILs (often including CD8+ T cells and Tregs) are analyzed by flow cytometry and immunohistochemistry.
Humanized mouse models
Immunodeficient mice are engrafted with human hematopoietic stem cells or peripheral blood mononuclear cells, and then engrafted with human tumor cells.
Human anti-CD25 antibodies are used to target human Tregs in these models.
These systems better approximate human immune-tumor interactions and permit the study of human TILs but are more complex and expensive than syngeneic systems.
Supporting details:
Direct intratumoral delivery of anti-CD25 antibody or antibody-based immunotoxins can selectively deplete Tregs in the tumor while minimizing systemic side effects.
Tumor growth inhibition is typically accompanied by increased infiltration and activation of effector lymphocytes (notably CD8+ T cells) and a decrease in intratumoral Tregs.
Studies frequently use FoxP3 immunostaining or flow cytometry panels to characterize Tregs and other TIL subsets after anti-CD25 treatment.
Additional notes:
Anti-CD25 therapy is most established and reproducible in syngeneic murine models due to the ready availability of reagents and the intact mouse immune system.
PDX (patient-derived xenograft) models are sometimes used but are less suited for functional TIL studies since they generally lack a complete immune compartment unless humanized.
In summary, syngeneic mouse models are the standard for anti-CD25 antibody in vivo studies of tumor growth and TILs, while humanized mice are employed for translational and human-specific TIL investigations.
Researchers use Camidanlumab—an anti-CD25 antibody-drug conjugate—either as a monotherapy or in combination with other checkpoint inhibitors like pembrolizumab (anti-PD-1), to explore potential synergistic effects in complex immuno-oncology models, especially in solid tumors and lymphomas. There is currently no published evidence directly describing combinations with anti-CTLA-4 or anti-LAG-3 biosimilars, but the methodology for studying such synergy parallels other checkpoint inhibitor strategies.
Essential Context and Methodology:
Model Systems: Preclinical synergy studies typically employ mouse models (including syngeneic tumor models and humanized mouse models), and occasionally organoids or ex vivo human tumor samples.
Combination Rationale: The rationale stems from the complementary or distinct mechanisms that each inhibitor has on the immune system—for instance, combining a CD25-targeting agent like Camidanlumab (which can deplete regulatory T cells/Tregs and enhance T-cell priming) with checkpoint inhibitors (which remove brakes from effector T cells) may unleash a more robust anti-tumor response.
Endpoints & Analyses: Researchers measure outcomes such as:
Enhanced tumor regression/growth delay compared to monotherapies.
Immune profiling (flow cytometry, single-cell RNA sequencing) to assess T cell activation, Treg depletion, and shifts in the tumor microenvironment.
Pharmacokinetics and safety profiling to establish optimal dosing and sequence.
Biomarker analysis, monitoring immune signatures, cytokine production, and the presence of effector vs. suppressive cell populations.
Illustrative Example (Anti-PD-1/CTLA-4 or LAG-3 Combinations):
A recent study in melanoma used anti-PD-1 + anti-CTLA-4 and anti-PD-1 + anti-LAG-3 combinations in mouse models to dissect mechanistic differences:
Anti-PD-1/LAG-3 effects relied on CD4 T cells to drive antitumor activity, suppress Tregs, and boost CD8 responses.
Anti-PD-1/CTLA-4 directly activated CD8 cells often independent of CD4 T cell help.
Similar readouts are expected when testing Camidanlumab biosimilars in analogous combination settings to decipher whether additional depletion of Tregs (CD25+) augments the activity of CTLA-4 or LAG-3 blockade.
Clinical and Translational Studies:
As of 2020, a Phase 1b clinical trial combined Camidanlumab tesirine with pembrolizumab in advanced solid tumors to determine dosing, pharmacodynamics, and biomarkers of immune activation.
These trials seek to identify regimens where immune activation (via Treg depletion + effector T cell checkpoint release) translates into higher objective response rates and durable tumor control compared to monotherapy.
Summary Table: Experimental Strategy for Investigating Synergy
Parameter
Use in Camidanlumab + Checkpoint Inhibitor Studies
Although clear data for Camidanlumab biosimilar combinations with anti-CTLA-4 or anti-LAG-3 biosimilars in published preclinical or clinical literature is lacking, researchers would logically follow the approaches proven with other checkpoint inhibitor combinations.
A Camidanlumab biosimilar can be used as either the capture reagent or the detection reagent in a bridging anti-drug antibody (ADA) ELISA to monitor a patient’s immune response against Camidanlumab or its biosimilar. In a bridging ELISA, the biosimilar (functionally identical to the therapeutic drug) is typically conjugated (e.g., biotinylated or HRP-labeled) and utilized in both the capture and the detection steps, allowing the detection of bivalent ADAs formed in response to treatment.
How the bridging ADA ELISA works with the Camidanlumab biosimilar:
Plate Coating/Capture: The Camidanlumab biosimilar is immobilized on the solid phase, such as a microtiter plate (commonly by direct adsorption, via biotin-streptavidin interaction, or other high-affinity binding). This immobilized drug serves to "capture" any ADA present in the patient sample by binding to one of the antigen-binding sites of bivalent ADA molecules.
Sample/Serum Addition: Patient serum samples are added. If anti-drug antibodies are present, one "arm" of the ADA binds to the plate-bound biosimilar.
Detection: The same Camidanlumab biosimilar, conjugated to an enzyme (e.g., HRP) or other detectable label, is added as the detection reagent. This labeled drug molecule binds to the other “arm” of the ADA, thus forming a "bridge" between the capture and detection reagents via the ADA's two binding sites.
Signal Generation: A substrate is provided for the enzyme, generating a measurable signal proportional to the amount of ADA present.
Why use the biosimilar as both capture and detection reagent?
The biosimilar is chosen when it is demonstrated to be structurally and functionally equivalent (including immunological epitopes) to the original drug, ensuring it will effectively bind to all relevant anti-Camidanlumab antibodies in the patient's serum.
Using the biosimilar (rather than the original) ensures the assay's applicability in settings where the biosimilar is the administered product, and it allows for direct measurement of immunogenicity toward the biosimilar itself.
Specific Considerations for Bridging ELISA:
The bridging ELISA is highly sensitive for detecting bivalent ADAs (IgG, IgM) but generally not monovalent antibodies.
The technique may be affected by interfering substances in patient serum (e.g., high residual drug, soluble target antigen, or heterophilic antibodies) and typically requires proper controls and, if needed, sample pretreatment.
High-quality, well-characterized, and stable biosimilar reagents are essential for assay consistency and reliability.
In summary: The Camidanlumab biosimilar is used as both the capture (plate-bound or biotinylated) and detection (enzyme-labeled) reagent in a bridging ELISA to measure the presence of ADAs in patient samples. The appearance and titer of ADAs detected by this method indicate the patient’s immunogenic response to the administered therapeutic.
References & Citations
1 Epperla N, Hamadani M. Curr Hematol Malig Rep. 16(1):19-24. 2021.
2 Zammarchi F, Havenith K, Bertelli F, et al. J Immunother Cancer. 8(2):e000860. 2020.
3 Flynn MJ, Zammarchi F, Tyrer PC, et al. Mol Cancer Ther. 15(11):2709-2721. 2016.