Anti-Human TNF-α Adalimumab [Clone D2E7]

Anti-Human TNF-α Adalimumab [Clone D2E7]

Product No.: LT100

- -
- -
Product No.LT100
Clone
D2E7
Target
TNF-α
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
DIF; TNFA; TNFSF2; TNLG1F; TNF-alpha
Isotype
Human IgG1κ
Applications
B
,
ELISA
,
FA
,
FC
,
IF
,
IHC
,
IP
,
N

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Active
Immunogen
Human TNF alpha
Product Concentration
≥ 5.0 mg/ml
Endotoxin Level
< 1.0 EU/mg as determined by the LAL method
Purity
≥98% monomer by analytical SEC
>95% by SDS Page
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.
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 (RUO). Non-Therapeutic.
Country of Origin
USA
Shipping
2-8°C Wet Ice
Applications and Recommended Usage?
Quality Tested by Leinco
FC The suggested concentration for Adalimumab biosimilar antibody for staining cells in flow cytometry is ≤ 0.25 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 ?
N
B
ELISA
FA
IF
IP
IHC
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 are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs by constructing a standard curve against which the unknown drug concentrations in serum samples are measured. This allows the precise quantification of adalimumab (originator or biosimilar) in patient samples and supports cross-comparisons between biosimilar and reference products.

Key aspects:

  • Role in Calibration/Standard Curves: Research-grade adalimumab biosimilars are serially diluted in serum or assay buffer to generate a standard curve that spans the expected drug concentration range in clinical samples. This curve forms the basis for quantifying unknowns by interpolating their ELISA signals.

  • Reference or Control: The biosimilar can be used as a reference calibrator or quality control sample to ensure assay consistency. PK bridging studies often use both the originator (e.g., Humira®) and biosimilar (e.g., ABP501) as standards to confirm assay equivalence and enable bridging of PK data between the two products.

  • Demonstrating Interchangeability and Assay Suitability: Studies validate that both biosimilar and reference standards yield overlapping standard curves and comparable results, ensuring the assay is sensitive and specific for both molecules. For example, the LISA-TRACKER Adalimumab assay demonstrated high correlation (R² ~0.94-0.98) and similar slopes when measuring both ABP501 biosimilar and Humira® in human serum, confirming suitability for quantifying either in clinical samples.

  • Assay Harmonization: International standards (e.g., the WHO International Standard for Adalimumab) or well-characterized biosimilar reference preparations are employed to enable assay harmonization across labs. Concordance in assay readouts using different standards (either in-house, commercial kit, or international) demonstrates interchangeability and supports reproducible PK comparisons in bridging studies.

  • Serum Matrix Consideration: Calibration standards must be prepared in matrices matched to patient samples—typically pooled human serum spiked with known concentrations of the biosimilar—to account for serum-specific effects on assay signal.

  • Assay Validation and Controls: In addition to calibration curves, biosimilars may also serve as positive controls (QCs) in each run. Assay accuracy, precision, specificity, and parallelism between biosimilar and originator standards are key validation criteria.

Summary Table: Use of Adalimumab Biosimilars as Calibration Standards in PK Bridging ELISA

PurposeImplementationSupporting Detail
Calibration StandardSerially dilute biosimilar in serum to generate standard curveInterpolate unknowns from curve
Reference/ControlCompare biosimilar and originator curves in same assay for PK bridgingValidate interchangeability
Assay HarmonizationUse of WHO or unified standard to enable cross-lab comparabilityStandardize reporting
Positive Controls (QC)Use biosimilar at defined concentration(s) as run controlsMonitor assay performance
Matrix MatchingPrepare standards in pooled human serum to reflect clinical sample matrixMinimize matrix effects

In summary, PK ELISA bridging studies rely on well-characterized biosimilar and/or originator adalimumab standards for accurate, reproducible drug quantification, enabling regulatory and clinical cross-comparison of biosimilar and reference products.

The primary in vivo models where a research-grade anti-TNF-α antibody is administered to study tumor growth inhibition and characterize tumor-infiltrating lymphocytes (TILs) are syngeneic mouse models and, to a lesser extent, humanized mouse models.

Key details:

  • Syngeneic models use mouse tumor cell lines (e.g., CT26 colorectal carcinoma) implanted into immunocompetent mice (such as BALB/c or C57BL/6).

    • Anti-TNF-α mAbs are administered to these mice to study effects on tumor growth, immune pathways, stromal response, angiogenesis, and TIL populations such as lymphocytes and macrophages.
    • TILs can be characterized by flow cytometry and immunohistochemistry for specific immune markers after antibody treatment.
  • Humanized mouse models involve engrafting immunodeficient mice with human immune cells and sometimes human tumor cells.

    • While less common for anti-TNF-α studies (since most anti-TNF-α mAbs are mouse- or rat-specific in standard preclinical work), these models enable assessment of human TIL responses and cytokine interactions in an in vivo context relevant to human immunotherapies.
Model TypeDescriptionKey Features for Anti-TNF-α Study
SyngeneicMouse tumor cells in immunocompetent mice (e.g., CT26 in BALB/c)Permits robust TIL analysis, preserves full mouse immune system
HumanizedHuman tumor and immune cells in immunodeficient miceEnables study of human TILs; may be limited by mAb species selectivity
  • Experimental endpoints commonly include tumor volume/weight, flow cytometry for TIL subsets (e.g., CD8+, CD4+, macrophages), and evaluation of immune gene expression or protein markers.
  • These models are considered most relevant for assessing both tumor growth inhibition and TIL phenotype/function in response to anti-TNF-α intervention.

Patient-derived xenograft (PDX) models are rarely used for direct anti-TNF-α mAb studies targeting the immune compartment, as these mice lack fully functional immune systems unless humanized.

Summary:
Syngeneic mouse models are the gold standard for in vivo anti-TNF-α administration to characterize TILs and tumor growth inhibition. Humanized mice are also employed for translational studies involving human immune-tumor interactions, though less frequently for anti-TNF-α mAbs unless human-specific reagents are used.

Current State of Adalimumab Biosimilar Use in Complex Immune-Oncology

Adalimumab is a monoclonal antibody targeting tumor necrosis factor-alpha (TNF-α) and is widely used in autoimmune diseases. Its biosimilars—near-identical copies with similar efficacy and safety profiles—are increasingly available and studied in various contexts. However, there is no published evidence in the provided search results or in the literature that adalimumab or its biosimilars are being combined with checkpoint inhibitors (such as anti-CTLA-4 or anti-LAG-3 antibodies, or their biosimilars) in complex immune-oncology models to study synergistic effects.

Synergistic Studies in Immune-Oncology: Focus on Checkpoint Inhibitor Combinations

Current research on immune-oncology combination therapies primarily explores combinations of immune checkpoint inhibitors (ICIs) themselves (e.g., anti-CTLA-4 plus anti-PD-1/PD-L1), or ICIs with chemotherapy, targeted therapies, radiotherapy, and other agents that modulate the tumor microenvironment. These combinations aim to overcome resistance, enhance T-cell infiltration, and improve clinical outcomes in various cancers.

  • Double checkpoint blockade (e.g., ipilimumab + nivolumab) is well-studied, demonstrating synergy in some cancers, but with increased toxicity.
  • Novel checkpoint targets like LAG-3, TIGIT, and others are under investigation, often in combination with PD-1/PD-L1 or CTLA-4 inhibitors.
  • Combining ICIs with other pathways—such as PI3K/AKT/mTOR inhibitors, BRAF/MEK inhibitors, or drugs targeting myeloid-derived suppressor cells—is a growing field, with preclinical and early-phase clinical data suggesting potential benefits.

Scientific Rationale for TNF-α Inhibition in Immuno-Oncology

While TNF-α inhibitors like adalimumab are not standard in cancer immunotherapy, there is theoretical interest in their potential to modulate the tumor microenvironment and inflammation. However, clinical and preclinical evidence supporting the combination of TNF-α inhibitors with checkpoint inhibitors is lacking. In fact, TNF-α inhibitors are primarily used to manage immune-related adverse events (irAEs) from checkpoint inhibitors rather than as direct anti-cancer agents.

Challenges and Considerations

  • Lack of Precedent: No published studies describe the combination of adalimumab (or its biosimilars) with checkpoint inhibitors (or their biosimilars) for synergistic anti-tumor effects.
  • Mechanistic Concerns: TNF-α blockade may suppress certain inflammatory processes necessary for effective anti-tumor immunity.
  • Safety: Combining biologic agents increases the risk of immune suppression, infections, and other adverse events.
  • Regulatory and Clinical Hurdles: Given the absence of preclinical rationale and safety data, such combinations are not being actively pursued in clinical trials.

Conclusion

Current research in immune-oncology focuses on synergistic combinations of checkpoint inhibitors with each other or with other classes of anti-cancer agents, not with TNF-α inhibitors like adalimumab. There is no evidence from the literature that researchers are using adalimumab biosimilars in conjunction with checkpoint inhibitor biosimilars to study synergistic effects in complex immune-oncology models. Any future exploration of such combinations would require strong preclinical rationale, careful safety assessment, and rigorous clinical evaluation.

In a bridging ADA ELISA for immunogenicity testing, a biosimilar Adalimumab can serve as both the capture and detection reagent to specifically measure anti-adalimumab antibodies (ADAs) generated in patients undergoing therapy with Adalimumab or its biosimilars.

Key methodology details:

  • Capture reagent: The biosimilar Adalimumab is immobilized on the microtiter plate. When patient serum is added, any ADA present will bind to this immobilized biosimilar Adalimumab.
  • Detection reagent: After washing away unbound substances, a second molecule of Adalimumab (labeled, e.g., with HRP or biotin) is added as the detection reagent. Because ADAs are typically bivalent, they can simultaneously bind both the immobilized and the labeled Adalimumab, forming a “bridge”.
  • Result interpretation: The formation of this bridged complex allows for enzyme-mediated colorimetric detection, where the signal intensity is proportional to the amount of ADA present in the sample.

Why use a biosimilar as both capture and detection reagent?

  • Biosimilar Adalimumab is structurally and functionally nearly identical to the originator, making it appropriate for ADA assays against both forms.
  • It ensures reagent availability and cost-effectiveness, particularly for commercial ADA test kits.
  • The use of biosimilar mitigates issues of supply restrictions or high cost associated with the originator biologic, while maintaining assay specificity.

Bridging ELISA advantages:

  • It reliably detects bivalent ADAs, the most immunogenic and clinically-relevant species.
  • Assay specificity comes from the use of authentic drug (here, a biosimilar) for both capture and detection, reducing cross-reactivity.

Context of patient monitoring:

  • By quantitatively measuring ADA levels, clinicians can evaluate the immunogenicity of Adalimumab therapy.
  • The technique allows monitoring for loss of drug efficacy, hypersensitivity reactions, and other immune-mediated adverse effects.

Summary table: Biosimilar Adalimumab in bridging ADA ELISA

Reagent RoleAction in Bridging ELISA
Capture (plate)Immobilized biosimilar Adalimumab binds ADA
Detection (solution)Labeled biosimilar Adalimumab completes bridge through ADA's second Fab arm
Patient sampleADA in serum bridges immobilized and labeled drug
ReadoutColorimetric via enzyme label, proportional to ADA

This approach represents a standardized and sensitive method for monitoring a patient’s immune response against therapeutic Adalimumab or its biosimilars during treatment.

References & Citations

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

Certificate of Analysis

Formats Available

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