Anti-Human RANKL (Denosumab)

Anti-Human RANKL (Denosumab)

Product No.: LT2800

- -
- -
Product No.LT2800
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
Active
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 concentrations in serum samples by employing a standardized and validated protocol. Here's a detailed explanation of how this is typically done:

Principle of PK Bridging ELISA

  1. Calibration Curve: The PK bridging ELISA involves creating a calibration curve using known concentrations of Denosumab. This curve allows for the quantification of Denosumab in serum samples by comparing the optical density (OD) of the sample to the OD values of the calibration standards.

  2. Standard Curve Range: The calibration standards typically cover a wide range of concentrations, such as from 0.125 ng/mL to 8,000 ng/mL, ensuring that the assay can detect Denosumab across the entire therapeutic range.

  3. Zero Concentration Control: A zero Denosumab concentration is included as a background value to account for any non-specific binding or background noise in the assay.

Reagents and Assay Setup

  • Capture Antibodies: Anti-Denosumab antibodies, such as HCA280 or HCA282, are used as capture antibodies to bind Denosumab in the sample.
  • Detection Antibody: A detection antibody like HCA283P is used to detect the captured Denosumab.
  • Sample Preparation: Serum samples are prepared according to the protocol, which may include dilution if necessary.

Assay Performance

  • Sensitivity and Specificity: The assay is designed to be sensitive and specific for Denosumab. Sensitivity levels can vary, but the assay should be able to detect concentrations as low as 25 ng/mL.
  • Inter- and Intra-Assay Variability: The assay should be validated to ensure low variability between and within assays to ensure reliable results.

Validation Considerations

  • Regulatory Compliance: The bioanalytical method must be validated according to regulatory guidelines, such as those from the EMA or FDA.
  • Precision and Accuracy: The assay should demonstrate high precision and accuracy across the calibration range.

By using research-grade Denosumab biosimilars as calibration standards in PK bridging ELISAs, researchers can ensure accurate and reliable measurement of Denosumab concentrations in serum samples, which is crucial for pharmacokinetic studies and biosimilar development.

Primary Syngeneic and Humanized Models for Anti-RANKL Antibody Studies

The main research models in which anti-RANKL antibodies have been administered in vivo to study tumor growth inhibition and to characterize tumor-infiltrating lymphocytes (TILs) are syngeneic murine tumor models, particularly in the context of breast cancer.

Syngeneic Mouse Models

Key Models Used:
Triple-negative breast cancer (TNBC) syngeneic models are prominently referenced, specifically the 4T1, 67NR, and E0771 cell lines orthotopically implanted in immune-competent mice. These models are widely employed because they retain a functional immune system, allowing researchers to investigate the interplay between immunotherapy (such as anti-RANKL) and the host immune response, including TIL dynamics.

Experimental Approach:
Researchers administer research-grade anti-RANKL antibodies to these syngeneic hosts, then track tumor growth and perform detailed immunophenotyping of the tumor microenvironment. This includes the quantification and characterization of 14 immune cell (sub)populations and assessment of antigen-presenting cell activation status within the tumor. These studies have demonstrated that RANKL blockade inhibits tumor growth in an immune-dependent manner, as the effect is lost in immunocompromised mice. The tumor growth inhibition correlates with a remodeling of the tumor immune microenvironment, particularly affecting dendritic cells (DCs) and plasmacytoid DCs (pDCs), and indirectly promoting regulatory T cell differentiation via mTOR pathway modulation in antigen-presenting cells.

Relevance of Syngeneic Models:
Syngeneic models like 4T1, 67NR, and E0771 are chosen for their ability to mimic human tumor-immune interactions and to evaluate the efficacy of immunotherapies in a setting that preserves adaptive immunity. They are instrumental in elucidating how anti-RANKL treatment alters the composition and function of TILs, including T cells, DCs, and myeloid-derived suppressor cells.

Humanized Mouse Models

Current Evidence:
There is no explicit mention in the provided search results of humanized mouse models (e.g., mice engrafted with human immune systems and human tumors) being used to study the effects of anti-RANKL antibodies on tumor growth and TIL characterization. The focus of the cited research is squarely on syngeneic, immune-competent murine systems.

Combination and Mechanistic Studies

Combination Therapies:
The efficacy of anti-RANKL can be enhanced when combined with neoadjuvant chemotherapy, which primes effector T cells and increases their infiltration into the tumor. However, combination with PD-1 checkpoint inhibition did not show a synergistic effect in these models. There is also evidence for the development of bispecific antibodies targeting both RANKL and PD-1, which have shown potent tumor growth inhibition, including in settings of immune checkpoint inhibitor resistance.

Summary Table: Primary Models for Anti-RANKL Antibody Studies

Model TypeExample Cell LinesKey FindingsTIL CharacterizationReferences
Syngeneic murine4T1, 67NR, E0771RANKL blockade inhibits tumor growth via immune remodeling; enhanced by chemo14 immune subsets, DC/pDC, Tregs
Syngeneic murineEMT6, CT26, RENCA(General use in immuno-oncology, not specifically with anti-RANKL in cited studies)Various T cell, myeloid populations
HumanizedNot specifiedNot described in provided literatureNot applicable

Conclusion

Syngeneic mouse models—especially 4T1, 67NR, and E0771 orthotopic TNBC models—are the primary platforms where research-grade anti-RANKL antibodies have been administered in vivo to study tumor growth inhibition and to characterize resulting TILs. These models enable detailed analysis of immune cell infiltration, activation, and functional changes in response to RANKL blockade, underscoring the importance of the adaptive immune system in mediating anti-tumor effects. Humanized models are not highlighted in the current literature for this specific application. Combination strategies, particularly with chemotherapy, further enhance the immunotherapeutic potential of anti-RANKL in these syngeneic systems.

Researchers investigate synergistic effects between the denosumab biosimilar and immune checkpoint inhibitors (ICIs)—including anti-CTLA-4 and potentially anti-LAG-3 biosimilars—by co-administering these agents in preclinical models and clinical studies, especially for cancer patients with bone metastases.

The core methodology involves:

  • Combination Therapy Studies: Patients or animal models receive both a denosumab biosimilar (targeting RANKL) and one or more ICIs (e.g., anti-PD-1, anti-CTLA-4), seeking enhanced antitumor responses beyond each agent alone.
  • Sequence Investigation: Evidence suggests that sequencing is crucial—administering ICIs before denosumab may provide more pronounced antitumor effects, possibly due to initial ICI-induced RANKL upregulation on T cells, which denosumab then targets, reducing immune suppression in the tumor microenvironment. For example, preclinical studies show that giving anti-PD-1 and anti-CTLA-4 before denosumab in animal models results in stronger tumor control than the reverse order or simultaneous use.
  • Outcome Measurement: Researchers evaluate objective measures such as overall survival, progression-free survival, immune cell activation, and tumor growth in both clinical and preclinical models. Specifically, studies in patients with bone metastases compare endpoints like bone response rate, disease control, and immune-related adverse events when combining denosumab biosimilars with ICIs.
  • Mechanistic Analysis: Molecular and cellular mechanisms are studied, focusing on how the RANK/RANKL pathway interacts with immune modulation. Denosumab can relieve RANK-mediated immunosuppression (e.g., in dendritic cells and T cells), potentially enhancing ICI efficacy.

For checkpoint inhibitors outside anti-PD-1 and anti-CTLA-4 (such as anti-LAG-3 biosimilars), direct published clinical synergy with denosumab biosimilars is not explicitly documented in these results, but similar principles would theoretically apply, and preclinical research may be ongoing.

Key insights:

  • Denosumab biosimilars are shown to be clinically equivalent to reference denosumab in safety, efficacy, and immunogenicity, making them suitable for such combination studies.
  • Future research is encouraged to focus on prospective trials and complex immune-oncology models to clarify optimal combination strategies, including dosing, sequencing, and selection of checkpoint inhibitors.

In summary, researchers use denosumab biosimilars with ICIs by administering each agent in controlled sequence and combination trials, measuring immune and cancer outcomes, and exploring mechanistic interactions, particularly the modulation of RANK/RANKL signaling and antitumor immunity.

A Denosumab biosimilar can be used as both the capture and detection reagent in a bridging ADA ELISA to monitor a patient’s immune response (i.e., development of anti-drug antibodies, or ADAs) against the therapeutic drug.

In the bridging ADA ELISA format:

  • The Denosumab biosimilar is labeled in two different ways: typically, one aliquot is biotinylated (for capture), and another is enzyme-conjugated (e.g., HRP-labeled, for detection).
  • Patient serum is incubated with both the biotinylated and labeled versions of the biosimilar. If anti-drug antibodies (ADAs) are present in the serum, their bivalent structure allows them to bind both versions of the drug: one arm binds the immobilized (biotinylated) biosimilar on the plate, and the other binds the detection-labeled biosimilar, forming a "bridge".

Typical workflow:

  1. Coating/Immobilization: Streptavidin-coated plates capture the biotinylated Denosumab biosimilar.
  2. Sample Addition: Patient serum (potentially containing ADAs) is added, allowing any ADAs to bind the immobilized drug.
  3. Detection: The HRP-labeled (or otherwise conjugated) Denosumab biosimilar is added. If ADAs are present, the detection reagent binds to the ADA, which is already attached to the capture drug—as a result, a "bridge" is formed.
  4. Readout: A substrate for the enzyme (such as TMB for HRP) is added; a quantifiable colorimetric or chemiluminescent signal proportional to the amount of ADA present is generated.

Rationale for using the biosimilar in this way:

  • Regulatory guidelines and best practices for biosimilar immunogenicity assessment recommend using the biosimilar itself as the reagent (both for capture and detection), subject to cross-validation with the reference product, to ensure epitopes and detection profiles are similar. This approach ensures the assay is sensitive to antibodies generated specifically against the biosimilar and maintains consistency when comparing immunogenicity between biosimilar and reference in clinical studies.
  • It is also crucial to demonstrate during validation that the biosimilar and the reference product are equivalent as assay reagents, especially in how they present potential epitopes to ADAs.

Additional Notes:

  • While general details about Denosumab-specific ADA protocols are not provided, the described method is standard for therapeutic antibodies, and biosimilar-specific guidelines align with this approach.
  • Assay sensitivity, specificity, and drug tolerance must be validated for the biosimilar-based bridging ADA ELISA to ensure robust monitoring in clinical practice.

Summary Table: Use of Denosumab Biosimilar in Bridging ADA ELISA

StepReagentRole
Plate CoatingBiotinylated Denosumab biosimilarCapture ADA from patient serum
Sample IncubationPatient serumMay contain ADA
DetectionHRP-labeled Denosumab biosimilarBinds second site on ADA
SignalSubstrate for HRP (e.g., TMB)Quantifies ADA bound

This method is widely recommended for biosimilar ADA assays to ensure accurate and equivalent detection of immune responses to both biosimilar and originator drugs.

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.