Anti-Human Amyloid-β (Donanemab)

Anti-Human Amyloid-β (Donanemab)

Product No.: A530

- -
- -
Product No.A530
Clone
LY3002813
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
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Hamster
Expression Host
CHO Cells
FC Effector Activity
Active
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,
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 Donanemab. Donanemab binds to the insoluble, modified, N- terminal truncated form of the β-amyloid.
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.

LY3002813, known as Donanemab, is a human IgG1, kappa monoclonal antibody designed to target amyloid-beta plaques, which are linked to Alzheimer's disease. By binding to these plaques, Donanemab aims to reduce their accumulation and may slow disease progression. It is primarily used in clinical research and trials, with studies demonstrating its potential to lower amyloid plaque levels and improve cognitive function in early-stage Alzheimer's patient3-7.
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 Donanemab biosimilars are typically used as calibration standards (analytical standards) or reference controls in pharmacokinetic (PK) bridging ELISAs to ensure accurate measurement and comparability of Donanemab drug concentrations in human serum samples during PK studies.

Key Points and Methodology:

  • Biosimilar as Analytical Standard:
    In a single PK bridging ELISA used for biosimilar development, the biosimilar Donanemab (instead of the originator/reference product) is often selected as the analytical standard (calibrator). The biosimilar is used to generate the standard curve, against which the concentrations in serum samples—including those containing either biosimilar or reference Donanemab—are quantified.

  • Validation and Equivalence:
    The strategy involves thorough assay qualification to demonstrate that the biosimilar and the reference Donanemab are bioanalytically equivalent in the ELISA platform. This is achieved by parallel testing of both the biosimilar and the reference product in the assay to ensure similar detection and quantification characteristics. Statistical equivalence (e.g., 90% CI within 0.8–1.25) must be met to confirm that both forms behave similarly within the assay context.

  • Calibration Curve Preparation:
    The Donanemab biosimilar, usually of verified research grade and purity, is serially diluted in matrix-matched serum to prepare the calibration standards at defined concentrations (e.g., 50–12,800 ng/mL as cited in validation studies). The calibration curve is then constructed from these standards and enables quantitation of Donanemab in patient serum samples.

  • Reference Controls/QC Samples:
    Quality control (QC) samples may be prepared using both the biosimilar and the reference product at several concentrations. These controls are analyzed together with study samples to ensure assay accuracy, precision, and reliable performance throughout sample analysis.

  • Assay Execution:
    The PK ELISA typically uses a sandwich format: anti-Donanemab antibodies coat the wells to capture Donanemab from standards/QCs/samples, followed by detection using a labeled (e.g., HRP-conjugated) anti-Donanemab antibody. The readout (colorimetric or chemiluminescent) correlates with the amount of Donanemab present, enabling quantification based on the standard curve prepared from the biosimilar.

  • Source and Quality of Biosimilars:
    Research-grade Donanemab biosimilars used for calibration must closely match the reference in terms of sequence, binding, and functional attributes to ensure reliable cross-quantification. Suppliers provide these as recombinant monoclonal antibodies expressed in mammalian systems, with high purity and characterized for research or analytical use.

Summary Table: Biosimilar Donanemab in PK ELISA

Role in AssayPurposeImportance
Calibration StandardConstruct standard curve for quantifying Donanemab in samplesEnsures accurate, reproducible data
Reference Control/QCValidate assay precision, accuracy, comparabilityConfirms assay reliability
Analytical EquivalenceDemonstrate comparable assay response between biosimilar and referenceSupports regulatory PK bridging

Additional Context:

  • Using a single standard (the biosimilar) across samples minimizes variability and eliminates the need for assay bridging or cross-referencing between separate calibrators.
  • This approach is recommended by regulatory agencies and industry due to robustness, regulatory acceptance, and ease of method transfer.

In summary, research-grade Donanemab biosimilars serve as essential calibration and control materials in PK ELISAs for measuring Donanemab concentrations in serum, provided their analytical equivalence to the reference product is rigorously demonstrated and validated within the assay system.

The primary models used to study the impact of anti-Amyloid-β (Aβ) antibodies on tumor growth and to characterize tumor-infiltrating lymphocytes (TILs) are murine syngeneic tumor models. These models are widely adopted because they possess a fully functional immune system and allow for characterization of TILs in response to immunotherapy, including antibodies not originally designed for oncology.

Essential context and supporting details:

  • Syngeneic models use mouse tumor cell lines implanted in immunocompetent mice of the same genetic background, maintaining an intact mouse immune system. This enables robust evaluation of tumor growth, immune cell infiltration, and therapeutic response.

  • Commonly used mouse syngeneic tumor models include:

    • MC38 (colon carcinoma, C57BL/6 background)
    • CT26 (colon carcinoma, BALB/c background)
    • B16F10 (melanoma, C57BL/6 background)
    • RENCA (renal carcinoma, BALB/c background)
    • EMT6 (breast carcinoma, BALB/c background).
  • These models are well characterized for gene expression, baseline TIL populations (such as CD8+ T cells, regulatory T cells, and myeloid-derived suppressor cells), and their response to immune checkpoint inhibitors and other immunotherapies.

  • Immune profiling in these models enables detailed analysis of how therapeutic agents, including research-grade anti-Aβ antibodies, influence both tumor growth and the composition and activity of TILs.

  • Humanized models (mice engrafted with human immune cells and/or tissues) have been used for certain immunotherapy studies but are less commonly employed for anti-Aβ antibody testing in oncology, primarily due to their complexity, cost, and the potential for xenogeneic immune interactions. The provided search results do not document anti-Aβ antibody use in humanized tumor models.

Additional relevant information:

  • The typical workflow involves administration of the antibody (e.g., anti-Aβ) to tumor-bearing mice and subsequent assessment of:

    • Tumor volume and growth inhibition
    • Flow cytometric or immunohistochemical analysis of TILs, including the abundance and phenotype of CD8+, CD4+, regulatory T cells, and myeloid populations.
  • These models are used to explore mechanistic questions, including the effect of antibodies (even those with non-traditional or repurposed targets, like anti-Aβ) on the anti-tumor immune response.

  • No specific studies from the provided results directly indicate that anti-Aβ antibodies have been routinely tested in humanized models for this purpose; syngeneic models remain the dominant preclinical system for such investigations.

In summary: Research-grade anti-Amyloid-β antibodies are primarily tested for tumor growth effects and TIL characterization in well-defined syngeneic murine tumor models such as MC38, CT26, RENCA, and B16F10, taking advantage of their fully functional immune systems and established immune profiling protocols.

Researchers studying synergistic effects in complex immune-oncology models use combinations of biosimilars—such as anti-amyloid agents like the Donanemab biosimilar—alongside checkpoint inhibitors (e.g., anti-CTLA-4 or anti-LAG-3 biosimilars) to interrogate immune system interactions, though direct evidence for Donanemab’s use in cancer immunotherapy is lacking in available search results. Most frequently, multiple checkpoint inhibitors are paired to study synergy, leveraging their complementary mechanisms in immune modulation.

Essential context and supporting details:

  • Checkpoint inhibitors such as anti-CTLA-4 and anti-PD-1/PD-L1 are co-administered in preclinical models to analyze how blocking distinct inhibitory pathways enhances T cell activation and anti-tumor immune responses. Anti-CTLA-4 acts mainly in lymph nodes, boosting T cell priming, while anti-PD-1/PD-L1 acts at tumor sites, maintaining cytotoxic T cell activity.
  • Synergy is studied by observing changes in tumor growth, immune cell populations, cytokine profiles, and survival outcomes in animal models or in vitro co-culture systems when drugs are combined versus used singly.
  • Experimental combinations: The scientific logic behind combining agents is that targeting different immune checkpoints—often those that act in separate immune compartments—may overcome resistance or suboptimal neutralization that occurs with monotherapies.

Donanemab biosimilar context:

  • The Donanemab biosimilar is an antibody that targets amyloid-beta 42, used predominantly in the study of Alzheimer's disease and not traditionally applied in cancer models. Its mechanism involves amyloid plaque clearance via microglial activation. Existing clinical trials focus on amyloid reduction, not immune-oncology synergy.
  • No current published evidence directly supports Donanemab biosimilar being used as part of immune-oncology synergy experiments or in combination with checkpoint inhibitors (CTLA-4, LAG-3) in cancer settings per the search results.

Relevant methodologies in immune-oncology synergistic studies:

  • Preclinical models involve administering checkpoint inhibitors like anti-CTLA-4, anti-PD-1, or anti-LAG-3, singly or in combination, and monitoring the immune response and cancer progression.
  • Synergy assessment methodologies include comparing tumor rejection rates, immune cell infiltration (CD8+, CD4+), cytokine analysis, and transcriptomic profiling between groups treated with single agents or combinations.
  • Sequencing and timing: Some studies highlight that the sequence of drug administration may greatly affect outcomes; for example, giving checkpoint inhibitors before other agents (like denosumab, similar in investigative principle) can amplify response.

Limitations:

  • There is no direct evidence in the provided sources that the Donanemab biosimilar is used with immune checkpoint inhibitors for cancer synergy studies. Most use cases relate to Alzheimer’s and Down syndrome amyloid pathology. Synergy research primarily involves combinations of known checkpoint inhibitors.
  • Any references to anti-LAG-3, anti-CTLA-4, or similar biosimilars combined with non-oncologic antibodies (like anti-amyloids) in immune-oncology are presently speculative, not empirical per available data.

In summary, synergy research in immune-oncology models is focused on combinations of checkpoint inhibitors where distinct immune pathways are co-targeted, and effects are measured via established preclinical and clinical models. Use of Donanemab biosimilar in such models has not been documented in existing literature, as it is tailored to neurodegenerative disease research rather than cancer immunotherapy.

A Donanemab 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 (i.e., development of ADAs) against therapeutic Donanemab.

Context and Mechanism:

  • Bridging ADA ELISA principle: The assay detects ADAs (e.g., human IgG generated in response to Donanemab) by exploiting their ability to bind two identical drug molecules: one presented on the plate (capture) and another as a labeled detection reagent.
  • Donanemab biosimilar use: Because a biosimilar has the same variable regions as therapeutic Donanemab, it can bind patient-generated ADAs equivalently without risking the use of clinical material, making it suitable for both capture and/or detection.

Typical Bridging ADA ELISA Workflow:

  1. Coating (Capture): The plate is coated with Donanemab biosimilar, presenting the same epitope as the therapeutic.
  2. Sample Incubation: Patient serum is added. If ADAs are present, they bind to the immobilized Donanemab biosimilar.
  3. Detection: A labeled Donanemab biosimilar (commonly biotinylated or conjugated to HRP/dye) is added. If ADAs are bound, this detection reagent can attach via its antigen-binding sites to a different epitope on the ADA, creating a "bridge."
  4. Signal Development: After washing, substrate is added for signal detection, proportional to the ADA amount.

Why Use a Biosimilar?

  • Functional equivalence: The biosimilar mimics the therapeutic’s binding characteristics with ADAs.
  • Supply & safety: Biosimilars are suitable for large-scale research and do not require use of costly or scarce therapeutic product for assay setup.
  • Assay specificity: Quality and fidelity of the biosimilar ensure specificity of ADA detection in the patient’s sample.

Summary:
In a bridging ADA ELISA, a Donanemab biosimilar is immobilized on the assay plate (capture) and/or used as a labeled detection reagent to "bridge" ADAs from patient samples, measuring the immunogenic response to Donanemab therapy by exploiting the bivalent nature of ADAs and the biosimilar’s identical epitope presentation.

References & Citations

1. Arndt JW, Qian F, Smith BA, et al. Sci Rep. 2018;8(1):6412.
2. Sevigny J, Chiao P, Bussière T, et al. Nature. 2016;537(7618):50-56.
3. Song T, Wang Y, Silverglate BD, Grossberg GT. Expert Opin Drug Metab Toxicol. 2024;20(6):411-417.
4. Shukla AK, Misra S. J Basic Clin Physiol Pharmacol. 2024;35(1-2):25-29.
5. Sims JR, Zimmer JA, Evans CD, et al. JAMA. 2023;330(6):512-527.
6. Sato S, Hatakeyama N, Fujikoshi S, Katayama S, Katagiri H, Sims JR. Neurol Ther. 2024;13(3):677-695.
7. Mintun MA, Lo AC, Duggan Evans C, et al. N Engl J Med. 2021;384(18):1691-1704.
Indirect Elisa Protocol
General Western Blot Protocol

Certificate of Analysis

Formats Available

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