Anti-Human Amyloid-β (Lecanemab) – Fc Muted™

Anti-Human Amyloid-β (Lecanemab) – Fc Muted™

Product No.: A495

- -
- -
Product No.A495
Clone
BAN-2401
Target
Amyloid-β
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
ABPP, APPI, Alzheimer disease amyloid A4 protein homolog, Alzheimer disease amyloid protein, Amyloid precursor protein
Isotype
Human IgG1κ
Applications
ELISA

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Muted
Immunogen
Unknown
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 biosimilar 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 -80°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 ?
ELISA
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 Lecanemab. BAN-2401 selectively binds to large, soluble Aβ protofibrils with high affinity.
Background
Amyloid-β (Aβ) is a peptide that accumulates in the brains of individuals with Alzheimer’s disease, forming plaques that are a hallmark of the condition. These plaques are believed to contribute to the neurodegenerative processes seen in Alzheimer’s by disrupting cell function and triggering inflammatory responses. Amyloid-β is derived from the amyloid precursor protein (APP) through enzymatic cleavage. The aggregation of Aβ into oligomers and fibrils is a key pathological feature of Alzheimer’s disease, making it a significant target for therapeutic interventions aimed at reducing or preventing plaque formation1,2.

Known as clone BAN-2401, Lecanemab is the humanized IgG1 version of the mouse monoclonal antibody mAb158, specifically engineered to selectively bind to large, soluble Aβ protofibrils. BAN-2401 has shown efficacy in reducing Aβ protofibrils in the brain and cerebrospinal fluid (CSF) of animal models. In clinical trials, it has exhibited potential as a disease-modifying treatment for Alzheimer's disease. Additionally, BAN-2401 has been utilized in preclinical research to study its binding to Aβ deposits in postmortem brain sections from individuals with Down's syndrome3,4.

Antigen Distribution
Amyloid-beta (Aβ) circulates in plasma, cerebrospinal fluid (CSF), and brain interstitial fluid (ISF), primarily as soluble Aβ40.
Ligand/Receptor
APBB1-KAT5, TNFRSF21, binds transient metals such as copper, zinc, and iron
NCBI Gene Bank ID
UniProt.org
Research Area
Neuroscience
.
Alzheimer's Disease
.
Neuroimmunology

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 Lecanemab biosimilars are employed as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISA assays by serving as precisely quantified model antibodies that mimic the drug’s biochemical activity and antigen binding profile. This allows for accurate measurement and quantification of Lecanemab concentrations in serum samples during PK evaluations.

To elaborate:

  • Calibration Standards: Lecanemab biosimilars are created using publicly available sequences to closely replicate the properties of the therapeutic antibody. In ELISA assays, these biosimilars are serially diluted and used to construct a standard curve, which relates known concentrations of the biosimilar to the assay’s signal output. The unknown Lecanemab concentrations in patient serum samples are then interpolated from this curve.
  • Reference Controls: Biosimilars also act as positive controls to validate assay performance and ensure specificity of detection for the target antibody. Their use guarantees that the assay reliably detects Lecanemab's pharmacologically relevant epitopes and conforms to established analytical standards.

Technical context:

  • The immunoreactivity of research-grade Lecanemab biosimilars is confirmed using ELISA and western blotting to verify their capacity to bind amyloid beta and interact with serum proteins similarly to the clinical-grade antibody.
  • These biosimilars are critical especially when the clinical-grade drug is limited or unavailable for assay validation, bridging experimental gaps during method development or regulatory submission.
  • Using biosimilar antibodies as standards also facilitates comparison across batches, studies, or analytical platforms by providing a consistent calibrator for quantification.

Other relevant points:

  • PK bridging ELISA assays may sometimes also use anti-human IgG capture/secondary reagents to detect Lecanemab or its biosimilar, further ensuring specificity and sensitivity.
  • The integrity and purity of research-grade biosimilars are generally verified by orthogonal methods such as size exclusion chromatography, western blot, and surface plasmon resonance before use in ELISA standardization.

In summary, research-grade Lecanemab biosimilars serve as both calibration standards for quantitation and as reference controls for assay validation in PK bridging ELISA, directly supporting reliable drug concentration measurement in serum during clinical or preclinical studies.

The primary in vivo models for administering research-grade anti-Amyloid-β (Aβ) antibodies to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are murine (mouse) syngeneic tumor models and, less frequently, humanized mouse models.

Context and Supporting Details:

  • Syngeneic Tumor Models:
    These involve implanting tumor cells derived from the same genetic background as the host mouse (e.g., CT26 in BALB/c, RENCA in BALB/c, MC38 or B16F10 in C57BL/6) into immunocompetent mice, preserving an intact immune system.
    These models are widely used to assess antitumor effects and immune cell infiltration (such as TILs) following immunotherapy administration, as they allow for detailed analysis of immune responses—including T cell and myeloid populations—within the tumor microenvironment.
    Syngeneic models are preferred for immunological studies because:

    • They retain a fully functional mouse immune system.
    • Tumor-immune interactions are preserved.
    • TIL analysis can be performed through flow cytometry and immunohistochemistry.
  • Characterization of TILs:
    Studies frequently use these models to evaluate how immunotherapies, such as anti-Aβ antibodies (or more commonly immune checkpoint inhibitors), modulate TIL populations (e.g., CD8+ cytotoxic T cells, CD4+ T cells, myeloid-derived suppressor cells).
    These models enable stratification of tumors by immune profiles and comparison of responders versus non-responders to immunotherapy based on TIL composition.

  • Humanized Models:
    These mice are engineered to possess human immune components (via transgenic expression or engraftment of human hematopoietic cells).
    While potentially useful for studying human-specific immune responses and antibody activity, they are less common for initial tumor immunology studies with anti-Aβ antibodies due to cost, complexity, and lower throughput compared to syngeneic models.
    Humanized models become relevant for translational research, especially if the antibody is specific for human Aβ and requires a human immune context.

  • Anti-Amyloid-β Antibodies in Oncology:
    Most published studies focus on anti-Aβ antibodies in Alzheimer’s disease models, but there is emerging evidence that Aβ can inhibit tumor cell growth, suggesting a rationale for testing such antibodies in syngeneic cancer models to assess tumor growth inhibition and immune effects.

Summary Table: Syngeneic vs Humanized Tumor Models

Model TypeSpeciesImmune SystemTumor SourceTypical Use
SyngeneicMouseMouse (intact)Mouse tumor linesStandard for immunotherapy/immune profiling
HumanizedMouse (eng.)Humanized (HSCs, etc.)Human tumor/xenograftAdvanced/translational, human immune interactions

Key Murine Syngeneic Models Used:

  • MC38 (colon adenocarcinoma, C57BL/6)
  • CT26 (colon carcinoma, BALB/c)
  • B16F10 (melanoma, C57BL/6)
  • RENCA (renal cell carcinoma, BALB/c)

In conclusion, syngeneic mouse models are the primary and most widely used platform for in vivo studies involving research-grade anti-Aβ antibody administration to interrogate tumor inhibition and TIL composition, with humanized models used for more advanced or translational studies.

Current literature does not show direct use of a Lecanemab biosimilar in combination with checkpoint inhibitors such as anti-CTLA-4 or anti-LAG-3 biosimilars in immune-oncology models, as Lecanemab is primarily an anti-amyloid beta antibody developed for Alzheimer’s disease, not cancer immunotherapy. However, the methodologies and preclinical strategies for combining monoclonal antibodies—including biosimilars—are well documented for checkpoint blockade therapies in immune-oncology.

Context and Supporting Details:

  • Lecanemab’s Indication and Research Use: Lecanemab, and its biosimilars, are used to target amyloid beta in neurodegenerative disease models. Research using Lecanemab biosimilar has focused on plasma protein binding and pharmacokinetics, specifically interactions with proteins such as fibrinogen. There is no documented preclinical or clinical use pairing Lecanemab biosimilar with immune checkpoint inhibitors for cancer immunotherapy.

  • Checkpoint Inhibitor Combinations in Oncology: In immune-oncology, combination therapy with checkpoint inhibitors (e.g., anti-PD-1, anti-CTLA-4, anti-LAG-3) is designed to target distinct immune regulatory pathways within the tumor microenvironment, often producing synergistic effects in suppressing tumor growth. These combinations are commonly modeled in mouse studies to examine:

    • Mechanistic synergy between checkpoint axes (e.g., CTLA-4 blockade primes T cell responses in lymph nodes, PD-1 blockade releases suppression at tumor sites).
    • Effects on specific T cell subsets (e.g., CD4+ T cell dependency for anti-PD-1/LAG-3; direct cytotoxic CD8+ T cell activation with anti-PD-1/CTLA-4).
    • Immunophenotyping, cytokine profiling, and tumor growth curve analysis to measure combined efficacy and immune modulation.
  • Biosimilar Use in Immunotherapy Research: Anti-CD39 and anti-CD73 biosimilars have been tested in immune checkpoint combination strategies in preclinical tumor models—suggesting a general research approach of using recombinant or biosimilar antibodies to interrogate synergistic inhibition of immunosuppressive pathways.

Relevant Combination Study Methodology:

  1. Selection of appropriate mouse or humanized models for tumor growth and immune cell profiling.
  2. Coadministration of biosimilars: Each antibody is administered at optimal dose and timing, chosen to maximize potential synergy based on in vitro data or pilot studies.
  3. Assessment of antitumor response, immune cell infiltration (flow cytometry, IHC), T cell activation/phenotype, and survival metrics.
  4. Interpretation of mechanistic interactions: For instance, studies have documented that anti-PD-1/CTLA-4 and anti-PD-1/LAG-3 regimens operate through distinct immune pathways as determined by the requirement for specific T cell subpopulations.

Key Point: While the research paradigm for combining monoclonal and biosimilar antibodies to interrogate immune-oncology synergy is well characterized (especially with anti-PD-1, CTLA-4, LAG-3, CD39, and CD73), there is currently no evidence that lecanemab biosimilars have been applied in this setting, due both to its molecular target (amyloid beta) and current research indications.

If your question is speculative—about how a Lecanemab biosimilar might be used in such models—researchers would design studies as above but would need scientific rationale linking amyloid beta or the antibody’s immune effects with tumor immune modulation, a hypothesis not supported in current literature.

A Lecanemab biosimilar is used as both the capture and detection reagent in a bridging ADA ELISA by taking advantage of its ability to bind anti-drug antibodies (ADAs) with high specificity, enabling sensitive monitoring of a patient's immune response to Lecanemab therapy.

Context and Protocol Details:

  • Assay Principle:
    In a bridging ADA ELISA, the therapeutic drug (here, Lecanemab, or its biosimilar) is immobilized on the plate as the capture reagent, and a labeled version (often HRP- or biotin-conjugated) serves as the detection reagent. When patient serum containing ADAs is added, these bivalent antibodies bind to the immobilized drug and simultaneously to the labeled drug, forming a "bridge".

  • Biosimilar Advantages:
    Using a biosimilar (non-therapeutic research-grade antibody with identical binding domains as the original drug) ensures consistency and avoids interference from residual therapeutic drug in patient samples. The biosimilar's identical variable region allows detection of all antibody specificities against the original Lecanemab.

  • Steps Involved:

    1. Plate Coating: The Lecanemab biosimilar is immobilized onto microtiter plate wells (as the capture reagent).
    2. Patient Sample Incubation: Patient serum is added, allowing any ADAs present to bind to the biosimilar-coated plate.
    3. Detection: A labeled (e.g., HRP- or biotin-conjugated) Lecanemab biosimilar is added, which binds to the other arm of the ADA, completing the bridge.
    4. Readout: Signal from the labeled biosimilar (chromogenic, fluorescent, or luminescent substrate) is measured, quantifying ADA presence.
  • Specificity Notes:

    • This format is highly sensitive for detecting bivalent IgG-class ADAs.
    • Matrix components (e.g., soluble amyloid beta or other plasma proteins) may compete or interfere, so assay optimization and controls are necessary.
  • Related Studies:
    The cited studies demonstrate use of the biosimilar in binding assays and confirm its suitability for ELISA-based applications by showing it retains high affinity for amyloid-beta and relevant specificity.

Key Points to Monitor Patient Immunogenicity:

  • The bridging ADA ELISA using a Lecanemab biosimilar reliably detects anti-drug antibodies generated in response to Lecanemab therapy.
  • Data obtained informs clinicians about potential loss of efficacy or risk of hypersensitivity due to immune response.

This approach is standard for monoclonal antibody therapies, with adaptation for specific drugs like Lecanemab implemented through use of a biosimilar reagent.

References & Citations

1. Sevigny J, Chiao P, Bussière T, et al. Nature. 2016;537(7618):50-56.
2. Arndt JW, Qian F, Smith BA, et al. Sci Rep. 2018;8(1):6412.
3. Logovinsky V, Satlin A, Lai R, et al. Alzheimer’s Research & Therapy. 2016;8(1):14.
4. https://www.alzforum.org/therapeutics/leqembi
Indirect Elisa Protocol

Certificate of Analysis

Formats Available

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