Anti-Human RANKL (Denosumab) – Fc Muted™

Anti-Human RANKL (Denosumab) – Fc Muted™

Product No.: LT2805

- -
- -
Product No.LT2805
Clone
AMG-162
Target
RANKL
Product Type
Biosimilar Recombinant Human Monoclonal Antibody
Alternate Names
osteoprotegerin ligand (OPGL), osteoclast differentiation factor (ODF), TNF related activation-induced cytokine (TRANCE), tumor necrosis factor ligand superfamily member 11 (TNFSF11)
Isotype
Human IgG2κ
Applications
ELISA
,
FA
,
IHC
,
WB

- -
- -
Select Product Size
- -
- -

Antibody Details

Product Details

Reactive Species
Human
Host Species
Human
Expression Host
HEK-293 Cells
FC Effector Activity
Muted
Recommended Isotype Controls
Immunogen
Purified Recombinant Human RANKL
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.
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
Additional Applications Reported In Literature ?
ELISA
WB
IHC
FA
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 Denosumab. This product is for research use only. Denosumab activity is directed against human RANKL (receptor activator of NFκB ligand).
Background
Osteoporosis is a disease of bone microarchitecture deterioration commonly seen in postmenopausal women1. Estrogen deficiency leads to low bone mass and increased bone fragility due to bone resorption increasing more than formation. Those affected have an increased risk of fracture. RANKL (receptor activator of NFκB ligand) is a TNF family member that acts as a key bone resorption protein by mediating osteoclast formation, activation, and survival via activating its receptor RANK1,2.

Denosumab, a fully human monoclonal antibody originally generated using transgenic Xenomouse technology, selectively and with high affinity binds to and inhibits human RANKL, thus preventing interaction with and activation of its receptor RANK on the surface of osteoclasts and their precursors2. This blocking activity inhibits the formation, function, and survival of osteoclasts, resulting in reduced bone resorption and consequently reduces the risk of vertebral, nonvertebral and hip fractures. Denosumab increases bone mineral density (BMD) and trabecular and cortical bone strength, with continued antifracture and BMD benefits over 10 years of therapy. Bone resorption is inhibited in cynomolgus monkeys and humans, but not normal mice or rats.

Unlike bisphosphonates, denosumab is not incorporated into bone and its effects on bone turnover markers, BMD and histomorphometric measures are generally reversed upon its discontinuation1.
Antigen Distribution
RANKL binds to its receptor RANK, which is located on osteoclasts and osteoclast precursors.
Ligand/Receptor
RANK/RANKL
NCBI Gene Bank ID
UniProt.org
Research Area
Biosimilars
.
Immunology
.
Osteoporosis

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 Denosumab biosimilars are used as calibration standards or reference controls in pharmacokinetic (PK) bridging ELISAs to measure drug concentration in serum samples through a structured process:

1. Preparation of Standard Curve

  • Calibration Standards: Known concentrations of research-grade Denosumab biosimilars are prepared to create a standard curve. This curve allows for the accurate quantification of Denosumab in unknown samples by comparing the optical density (OD) values to the known concentrations.
  • Range of Concentration: The standard curve typically covers a range of concentrations, for example, from 0.125 ng/mL to 8,000 ng/mL, to ensure that the assay can detect a wide range of drug concentrations in serum samples.

2. Assay Protocol

  • Capture and Detection Antibodies: The PK bridging ELISA uses specific anti-Denosumab antibodies (e.g., HCA280 or HCA282 as capture antibodies and HCA283P as the detection antibody) to bind Denosumab. This sandwich ELISA format ensures that the assay specifically detects Denosumab in the presence of other serum components.
  • Sample Preparation: Serum samples are prepared by diluting them appropriately to fit within the range of the standard curve. This ensures that the assay can accurately measure the concentration of Denosumab in the sample.

3. Calibration and Measurement

  • Calibration: The known concentrations of the Denosumab biosimilars are used to calibrate the assay, ensuring that the response (typically measured as OD) is linear across the concentration range.
  • Measurement: The concentration of Denosumab in serum samples is determined by comparing their OD values to the standard curve. This allows for the accurate measurement of drug concentration.

4. Validation and Controls

  • Validation: The PK ELISA must be validated to ensure accuracy, precision, and specificity. This involves demonstrating that the assay can reliably measure Denosumab concentrations across the expected range without interference from other components in the serum.
  • Reference Controls: Reference controls, prepared from the same biosimilars, are used to verify the accuracy of each assay run. These controls help ensure that the assay is functioning correctly and provide a quality control measure.

By using research-grade Denosumab biosimilars as calibration standards, PK bridging ELISAs can reliably measure the concentration of Denosumab in serum samples, which is crucial for pharmacokinetic studies and biosimilar development.

The primary in vivo models used to study tumor growth inhibition and tumor-infiltrating lymphocyte (TIL) characterization following administration of research-grade anti-RANKL antibodies are syngeneic mouse tumor models with fully functional immune systems. There is limited direct evidence in the literature for the routine use of humanized models (i.e., immunodeficient mice engrafted with a human immune system) for anti-RANKL antibody studies focused on immune characterization.

Key details and context:

  • Syngeneic models involve transplanting tumor cell lines derived from the same genetic background as the host mouse strain, preserving intact mouse immune responses. These models are crucial for studying how immune-targeting therapies such as anti-RANKL manipulate the tumor immune microenvironment, including TILs.

    • Common syngeneic models used for anti-RANKL studies include:
      • 4T1, 67NR, and E0771 (murine triple-negative breast cancer)
      • Other frequently used syngeneic tumor models in immuno-oncology research are EMT6, CT26, RENCA, and B16F10.
  • Study design for anti-RANKL antibody administration:

    • These studies examine effects on tumor growth inhibition after antibody treatment and comprehensively characterize changes in TILs including functional status, differentiation state, and antigen-presenting cell activation.
    • Immune profiling typically involves analysis of various lymphoid and myeloid cell populations within the tumor microenvironment, often via flow cytometry and sometimes transcriptomic profiling.
    • The dependency of efficacy on adaptive immunity is demonstrated by the absence of anti-tumor effects in immunodeficient hosts.
  • Rationale for model preference:

    • Syngeneic models are preferred because they allow mechanistic interrogation of immune cell–tumor interactions, which cannot be modeled in immunodeficient or xenograft settings.
    • Humanized models are occasionally used for translational studies but are less common for early discovery-phase immune modulation with anti-RANKL agents due to complexity, cost, and variable immune reconstitution.

Summary Table: Model Comparison

Model TypeExamplesImmune SystemTIL Analysis Feasible?Cited in Context of Anti-RANKL?
Syngeneic (mouse)4T1, 67NR, E0771Intact (mouse)YesYes
Humanized (immunodeficient with human immune system)VariableHuman (partial)Yes, but uncommon for anti-RANKLNot directly cited
XenograftHuman tumorNone or partialNoNot preferred

Conclusion:Syngeneic mouse tumor models (e.g., 4T1, 67NR, E0771) are the primary and best-established preclinical systems in which research-grade anti-RANKL antibodies are administered in vivo for studying both tumor growth and detailed TIL characterization. Use of humanized models for this purpose has not been substantiated in the cited studies and remains uncommon.

Research into combining denosumab biosimilars with immune checkpoint inhibitors represents a promising frontier in cancer immunotherapy, particularly for patients with bone metastases. Current studies demonstrate that this combination may produce synergistic effects that enhance antitumor activity through multiple mechanisms.

Current Research Approaches

Researchers are primarily conducting retrospective analyses and clinical trials to evaluate the efficacy of denosumab biosimilar combinations with immune checkpoint inhibitors (ICIs). The most comprehensive research has focused on non-small cell lung cancer (NSCLC) patients with bone metastases, where studies have enrolled patients receiving ICI treatment and stratified them into denosumab combination groups versus non-combination groups. These studies evaluate key outcomes including response rates for bone metastases, disease control rates, overall survival, real-world progression-free survival, and the incidence of immune-related adverse events.

Mechanistic Understanding of Synergistic Effects

The theoretical basis for combining denosumab with checkpoint inhibitors centers on the RANK/RANKL axis, which plays crucial roles in both bone metabolism and immune regulation. Denosumab, a monoclonal antibody targeting RANKL, may enhance ICI efficacy through several mechanisms:

Immune Microenvironment Modulation: ICI treatment can cause upregulation of RANKL expression in T cells, which promotes interaction with immunosuppressive RANK-expressing cells in the tumor microenvironment. Denosumab blocks this process, potentially relieving immunosuppression and allowing for enhanced immune responses.

Dendritic Cell Effects: RANK is expressed on dendritic cells that can mediate immunosuppression by blocking T cell activation. Denosumab treatment may relieve this suppression, resulting in increased numbers of active T cells and enhanced immune responses.

Timing and Sequencing Considerations

Research has revealed that the sequence of administration is critically important for achieving optimal synergistic effects. Preclinical studies have shown dramatic advantages when anti-PD-1 and anti-CTLA-4 treatments are used before denosumab, whereas initiating denosumab first followed by checkpoint inhibitors produced results similar to placebo.

Clinical data supports this sequencing approach. Patients who received ICI followed by denosumab showed significant advantages compared to those who received no denosumab or those who received denosumab before ICI initiation. This finding suggests that researchers should prioritize ICI-first protocols when designing combination studies.

Biosimilar Development and Validation

The development of denosumab biosimilars has enabled broader research applications while maintaining therapeutic equivalence. Studies of biosimilars like QL1206 and LY01011 have demonstrated equivalent efficacy, safety, and pharmacokinetics compared to reference denosumab. These biosimilars showed median percentage changes in bone turnover markers that were statistically equivalent to the original denosumab, with similar adverse event profiles and immunogenicity.

Current Study Designs and Outcomes

Researchers are employing both retrospective database analyses and prospective clinical trials to evaluate combination therapies. The IMMUCARE database analysis, which included 268 patients with bone metastases, stratified patients into groups without denosumab, ICI followed by denosumab, and denosumab followed by ICI. While overall survival and progression-free survival showed no significant differences between ICI monotherapy and ICI with denosumab combinations, the sequential ICI-then-denosumab approach showed promising numerical improvements.

Future Research Directions

The field is moving toward more sophisticated study designs that focus on first-line immunotherapy combinations and prospective trial methodologies. Researchers are particularly interested in exploring optimal timing windows for denosumab initiation, duration of concomitant treatment, and identification of biomarkers that predict response to combination therapy.

The expanding availability of multiple denosumab biosimilars, including denosumab-bmwo, denosumab-bnht, and denosumab-dssb formulations, provides researchers with additional tools for conducting larger-scale combination studies while managing cost considerations. This increased accessibility may facilitate more comprehensive investigations into synergistic mechanisms and optimal dosing strategies in complex immune-oncology models.

Role of Denosumab Biosimilar in Bridging ADA ELISA for Immunogenicity Testing

Immunogenicity testing—the assessment of a patient’s immune response to a therapeutic drug—is critical for biologics like monoclonal antibodies, including Denosumab and its biosimilars. A bridging anti-drug antibody (ADA) ELISA is a widely used assay format for detecting and quantifying ADAs elicited by these drugs.

How the Bridging ADA ELISA Works

In a standard bridging ELISA for ADA detection, the therapeutic drug (here, Denosumab or its biosimilar) is biotinylated and immobilized on a streptavidin-coated plate. When patient serum containing ADAs is added, these antibodies bind simultaneously to captured drug and to a labeled (typically horseradish peroxidase, HRP) version of the drug, forming an immune complex that can be detected enzymatically. This “bridging” occurs because the ADA recognizes two drug molecules—one immobilized, one labeled—thus allowing specific detection of bivalent ADAs (usually IgG).

Application to Denosumab Biosimilars

When monitoring immunogenicity against Denosumab biosimilars, the biosimilar itself must be used as both capture and detection reagent in the bridging ELISA. This is essential because ADAs can recognize subtle differences between originator and biosimilar, and only the biosimilar can faithfully represent the immunogenic epitopes present in the administered drug.

  • Capture: The denosumab biosimilar is biotinylated and immobilized on the plate.
  • Detection: A second preparation of the denosumab biosimilar is conjugated to a detectable label (e.g., HRP).
  • Patient Sample: Serum is added; if anti-biosimilar ADAs are present, they bridge the immobilized and labeled drug, resulting in a signal proportional to ADA concentration.

This approach ensures that the assay specifically detects antibodies against the biosimilar, not the originator or other irrelevant molecules.

Key Considerations

  • Specificity: The bridging ELISA is highly specific for ADAs that recognize both drug molecules—minimizing false positives but potentially missing certain ADA types (e.g., some IgM or low-affinity IgG).
  • Reagent Quality: Both the biotinylated and labeled drug must be of high quality and purity to avoid assay interference.
  • Matrix Effects: Human serum contains many proteins and other substances that can interfere with assay performance; robust blocking and wash steps are essential.
  • Drug Tolerance: Patient samples often contain free drug, which can interfere with ADA detection; specialized sample pre-treatment (such as acid dissociation) may be needed to improve assay sensitivity.
  • Regulatory Acceptance: While ELISA is commonly used, newer formats (e.g., electrochemiluminescence) may offer improved sensitivity and drug tolerance.

Alternative Methods

Some specialized ADA assays for Denosumab (and biosimilars) use the drug’s target (RANKL) as capture, with anti-Denosumab for detection, but this format is used for pharmacokinetic (PK) assays, not ADA detection. For ADA, the bridging ELISA using the drug itself as both capture and detection is standard.

Summary Table

Assay ComponentRole in Bridging ADA ELISA (Denosumab Biosimilar)
Capture ReagentBiotinylated Denosumab biosimilar
Detection ReagentLabeled (e.g., HRP) Denosumab biosimilar
Patient SampleSerum (potential source of ADAs)
SignalGenerated only if ADAs bridge capture and detection drug

Conclusion

In immunogenicity testing for Denosumab biosimilars, the biosimilar is used as both capture and detection reagent in a bridging ADA ELISA to specifically monitor patient immune responses against the administered drug. This approach is highly specific and clinically relevant, provided that assay conditions are carefully controlled to manage matrix effects and drug interference.

References & Citations

1. Deeks ED. Drugs Aging. 35(2):163-173. 2018.
2. Kostenuik PJ, Nguyen HQ, McCabe J, et al. J Bone Miner Res. 24(2):182-195. 2009.
3. Cummings SR, San Martin J, McClung MR, et al. N Engl J Med. 361(8):756-765. 2009.
4. Eastell R, Christiansen C, Grauer A, et al. J Bone Miner Res. 26(3):530-537. 2011.
5. Simon JA, Recknor C, Moffett AH Jr, et al. Menopause. 20(2):130-137. 2013.
6. Bone HG, Wagman RB, Brandi ML, et al. Lancet Diabetes Endocrinol. 5(7):513-523. 2017.
Indirect Elisa Protocol
FA
IHC
General Western Blot Protocol

Certificate of Analysis

- -
- -

Formats Available

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