Anti-Human TNF-α Adalimumab [Clone D2E7] — Fc Muted™ Biotin

Anti-Human TNF-α Adalimumab [Clone D2E7] — Fc Muted™ Biotin

Product No.: LT106

- -
- -
Product No.LT106
Clone
D2E7
Target
TNF-α
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
DIF; TNFA; TNFSF2; TNLG1F; TNF-alpha
Isotype
Human IgG1κ
Applications
ELISA
,
FA
,
FC

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Immunogen
Human TNF alpha
Product Concentration
≥ 2.0 mg/ml
Formulation
This Biotinylated antibody is formulated in 0.01 M phosphate buffered saline (150 mM NaCl) PBS pH 7.4, 1% BSA and 0.09% sodium azide as a preservative.
Storage and Handling
This biotinylated antibody is stable when stored at 2-8°C. Do not freeze.
Regulatory Status
Research Use Only (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
Next Day 2-8°C
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Adalimumab biosimilar antibody for staining cells in flow cytometry is ≤ 1.0 μg per 106 cells in a volume of 100 μl. Titration of the reagent is recommended for optimal performance for each application.
Additional Applications Reported In Literature ?
ELISA
FA
Additional Reported Applications For Relevant Conjugates ?
B
N
IP
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 Adalimumab. Clone D2E7 binds to soluble TNF- α, but not to TNF- β (lymphotoxin). This product is for research use only.
Background
Adalimumab is a research-grade monoclonal antibody that works by inactivating tumor necrosis factor-alpha (TNF-α). TNF-α is a 17.5 kD protein that mediates inflammation and immunity caused by the invasion of viruses, bacteria, and parasites by initiating a cascade of cytokines that increase vascular permeability, thus bringing macrophages and neutrophils to the site of infection. TNF-α secreted by the macrophage causes the blood to clot which provides containment of the infection. TNF-α inactivation has proven to be important in downregulating the inflammatory reactions associated with autoimmune diseases such as rheumatoid arthritis, psoriatic arthritis, ankylosing spondylitis, Crohn's disease, moderate to severe chronic psoriasis, and juvenile idiopathic arthritis. Adalimumab blocks the interaction with the p55 and p75 cell surface TNF receptors thus, neutralizing the biological function of TNF. Anti-Human TNF alpha (Adalimumab) utilizes the same variable regions from the therapeutic antibody Adalimumab making it ideal for research projects.
Antigen Distribution
TNF-α is secreted by macrophages, monocytes, neutrophils, T cells, B cells, NK cells, LAK cells.
PubMed
NCBI Gene Bank ID
Research Area
Biosimilars

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 adalimumab biosimilars play a critical role as calibration standards and reference controls in pharmacokinetic bridging ELISA assays for measuring drug concentrations in serum samples. These applications are essential for ensuring accurate, reproducible measurements across different laboratories and assay platforms.

Role as Calibration Standards

Research-grade adalimumab biosimilars serve as primary calibration standards in ELISA-based therapeutic drug monitoring assays. The World Health Organization has established an international standard (IS) for adalimumab that functions as a universal reference point for calibrating bioassays and clinical monitoring assays. This standardized approach helps harmonize measurements across different laboratories and assay platforms, ensuring consistent and reliable results regardless of the specific method employed.

When used as calibration standards, these biosimilars are prepared in serial dilutions to create standard curves that typically span the therapeutic range of adalimumab concentrations. For example, validated ELISA assays often use standard curves ranging from 1.54 ng/ml to 300 ng/ml, with the linear quantification range typically falling between 5.2 ng/ml to 200 ng/ml. The biosimilar standards are reconstituted fresh for each assay run to maintain consistency and accuracy.

Quality Control Applications

Research-grade biosimilars function as quality control samples at multiple concentration levels within the assay range. These quality control points are strategically placed throughout the standard curve to monitor assay performance. Typical quality control concentrations include low, medium, and high levels such as 5.2, 16, 27, 150, and 200 ng/ml. These controls help identify potential issues with assay drift, matrix effects, or systematic errors during the analytical run.

Bridging Study Applications

In pharmacokinetic bridging studies, research-grade adalimumab biosimilars serve as common reference standards to compare different adalimumab formulations or biosimilar products. Multi-center collaborative studies have demonstrated that when laboratories use the same adalimumab international standard alongside their in-house standards, excellent agreement is observed in adalimumab content estimates regardless of the specific assay method or platform used. This standardization is crucial for establishing bioequivalence between originator products and biosimilars.

ELISA Design and Validation

The most common ELISA format for adalimumab measurement uses recombinant human TNF-α (rhTNF-α) as the capture antibody, taking advantage of adalimumab's mechanism of action as a TNF-α antagonist. In this design, the research-grade biosimilar standards are captured by the immobilized rhTNF-α and detected using anti-human IgG antibodies conjugated to detection enzymes.

The validation process requires that biosimilar standards demonstrate acceptable binding kinetics, with dissociation constants (KD) typically in the range of 2.38×10⁻⁷ nM. The assays must meet stringent precision and accuracy criteria, with coefficient of variation (CV) values less than 20% for inter- and intra-assay measurements, and accuracy within 25% at the upper and lower limits of quantification.

Matrix Effects and Dilution Linearity

Research-grade biosimilars are used to validate dilution linearity in serum matrices, ensuring that samples with concentrations above the assay range can be accurately diluted and measured. This validation typically involves preparing spiked serum samples using pooled normal human sera and demonstrating that diluted samples maintain accuracy within ±20% of expected values across the entire dilution range.

Inter-Laboratory Standardization

One of the most significant advantages of using research-grade adalimumab biosimilar standards is their ability to harmonize results across different laboratories and platforms. Studies involving multiple laboratories, including contract research organizations, national control laboratories, commercial kit manufacturers, hospital laboratories, and clinical diagnostic centers, have shown that using common adalimumab international standards reduces inter-laboratory variability and improves result comparability.

This standardization is particularly important for therapeutic drug monitoring in clinical settings, where accurate adalimumab level measurements are essential for optimizing dosing regimens and improving patient outcomes. The use of validated research-grade biosimilar standards ensures that clinicians can make informed treatment decisions based on reliable pharmacokinetic data, regardless of which laboratory performs the analysis.

Standard flow cytometry protocols for evaluating expression levels or binding capacity of TNF-α using conjugated Adalimumab biosimilars (such as PE or APC-labeled reagents) primarily involve the following approaches:

1. Direct Staining of Cells Expressing TNF-α:

  • Cells (e.g., human peripheral blood lymphocytes or engineered cell lines like HEK293 overexpressing membrane-bound TNF-α) are incubated with a fluorescently labeled Adalimumab biosimilar, such as PE, APC, or BV480-labeled biosimilar anti-TNF.
  • After washing, cells are analyzed by flow cytometry. The fluorescent signal intensity correlates with the expression level or binding capacity of TNF-α on the cell surface.
  • Specificity is typically controlled using IgG1 isotype controls.

2. Functional Assay in Whole Blood or Cell Cultures:

  • Human whole blood or cell lines are stimulated (e.g., using LPS) to induce TNF-α expression.
  • Cells are incubated with the labeled Adalimumab biosimilar, potentially in the presence of other immune markers (for multi-color panels).
  • Changes in TNF-α surface expression, as well as related activation markers (CD69, TM-TNF, etc.), are measured.
  • This setup may also allow for analysis of Adalimumab-induced signaling events (e.g., changes in CD69, CD16), which indirectly reflect Adalimumab binding and modulation of TNF-α signaling.

3. Dose-Response Binding Curves:

  • Cells expressing membrane TNF-α are incubated with serial dilutions of the fluorescent Adalimumab biosimilar.
  • Following incubation and washing, flow cytometry is performed.
  • Mean fluorescence intensity (MFI) is plotted against biosimilar concentration to deduce binding affinity, EC₅₀, or relative potency using non-linear curve fitting (e.g., 4PL regression).
  • Controls: Background autofluorescence is subtracted, and negative controls are included.

Protocol Example Steps (from published biosimilar validation studies):

  1. Culture cells expressing membrane TNF-α (engineered lines or activated immune cells).
  2. Incubate cells (e.g., 5x10⁵ per sample) with various concentrations of a fluorescent-labeled Adalimumab biosimilar in FACS buffer (PBS + 0.5% BSA) for 30 min at 4°C in the dark.
  3. Include isotype control and unstained control samples.
  4. Wash cells with buffer, centrifuge, and resuspend.
  5. (If using unconjugated Adalimumab) Stain with fluorescent anti-human IgG1 Fc-specific antibody, wash, and analyze.
  6. Acquire data on flow cytometer (e.g., BD FACSCalibur or similar).
  7. Analyze MFI; plot dose-response curves if quantifying binding affinity.

Notes:

  • Conjugated Adalimumab directly detects TNF-α, as the drug specifically binds its epitope.
  • This method provides a direct measure of target availability, relative affinity, and biosimilar comparability.
  • Such protocols are widely used in both research reagent validation (demonstrating specific binding of Adalimumab biosimilars) and biosimilar functional characterization.

References for protocol details:

  • BD provides a conjugated BV480 biosimilar anti-TNF (Adalimumab) reagent validated for multiparameter flow cytometry of TNF-α on peripheral blood lymphocytes.
  • Published biosimilar studies use flow cytometry to compare binding of Adalimumab or its biosimilars to TNF-α on engineered HEK293 cells, with MFI serving as the quantitative readout, often following the approach outlined above.

Summary Table: Typical Flow Cytometry Protocol Steps

StepDetails
Cell PreparationEngineered/TNF-expressing or activated immune cells
Reagent IncubationPE/APC/BV480-labeled Adalimumab biosimilar (conjugated)
ControlsIsotype control, unstained, and specificity controls
WashPBS or FACS buffer, centrifugation
Data AcquisitionFlow cytometer (e.g., BD FACSCanto, FACSCalibur)
Data AnalysisMFI vs. controls; dose-response curve (if appropriate)

These approaches are standard for initial validation and for detailed characterization of binding interactions between Adalimumab biosimilars and TNF-α using flow cytometry.

Biosimilar analytical testing involves a comprehensive battery of assays designed to demonstrate structural and functional similarity between a proposed biosimilar and the reference originator drug. This analytical similarity assessment forms the foundation for biosimilar approval and relies on multiple complementary analytical techniques.

Structural Characterization Assays

The structural analysis encompasses multiple levels of molecular architecture. Primary structure analysis involves peptide mapping to examine the amino acid sequence and identify any post-translational modifications. Secondary and higher order structure assessment uses techniques like circular dichroism spectroscopy and nuclear magnetic resonance to evaluate protein folding patterns.

Mass spectrometry and chromatographic techniques are extensively deployed to characterize the molecular properties and assess process impurities and product-related impurity profiles. These physicochemical studies provide detailed information about molecular weight, charge variants, and other critical structural attributes that could impact biological function.

Functional Assessment

Functional assays serve as the crucial link between structural data and clinical expectations, answering whether any observed structural differences translate into meaningful biological differences. The functional characterization program typically includes:

Binding Assays evaluate the biosimilar's ability to bind to its target receptor or antigen with equivalent affinity to the reference product. For antibody-based biosimilars, Fc receptor binding assays are particularly important to demonstrate that any glycosylation differences don't impact immune system interactions.

Biological Potency Assays use cell-based systems to measure the biosimilar's ability to elicit the same biological response as the originator drug. These assays evaluate enzyme kinetics and other functional parameters that directly relate to therapeutic efficacy.

Orthogonal Methods are employed to provide multiple complementary approaches for characterizing product properties and more sensitively assess for potential differences between the biosimilar and reference product.

Impurity and Aggregate Analysis

The analytical similarity assessment must include rigorous comparison of purity and impurity profiles, serving as a sensitive fingerprint of the entire manufacturing process. This analysis evaluates:

  • Product-related variants including aggregates, precursors, fragments, and other modified forms
  • Process-related impurities that could arise from the manufacturing process
  • Degradation products that might form during storage or handling

Risk-Based Approach

Molecular properties are ranked by their risk of impact on the product's activity, pharmacokinetics, pharmacodynamics, safety, efficacy, or immunogenicity. This risk assessment determines which attributes are most critical for demonstrating biosimilarity, with particular focus placed on properties based on the protein's nature and mechanism of action.

Manufacturers conduct head-to-head comparisons where results must fall within appropriate limits, ranges, or distributions established through extensive characterization of the reference product. These highly sensitive analytical methods allow measurement of molecular properties across multiple lots of both the proposed biosimilar and reference product.

Regarding the Leinco biosimilar specifically, the search results do not contain information about this particular biosimilar or its use in analytical studies. The analytical approaches described above represent the standard methodologies used across the biosimilar industry for demonstrating similarity to originator biologics, but specific details about Leinco's biosimilar products are not available in the provided sources.

References & Citations

1. Omidinia, E. et al. (2019) Protein Expr Purif. 155:59-65.
Indirect Elisa Protocol
FA
Flow Cytometry

Certificate of Analysis

Formats Available

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