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 |
Antibody DetailsProduct DetailsReactive 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. DescriptionDescriptionSpecificity 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 AdvisorPowered 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
2. Assay Protocol
3. Calibration and Measurement
4. Validation and Controls
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:
Summary Table: Model Comparison
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 ApproachesResearchers 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 EffectsThe 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 ConsiderationsResearch 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 ValidationThe 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 OutcomesResearchers 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 DirectionsThe 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 TestingImmunogenicity 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 WorksIn 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 BiosimilarsWhen 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.
This approach ensures that the assay specifically detects antibodies against the biosimilar, not the originator or other irrelevant molecules. Key Considerations
Alternative MethodsSome 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
ConclusionIn 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 & Citations1. 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. Technical ProtocolsCertificate of Analysis |
Related Products
Prod No. | Description |
---|---|
D230 | |
LT2800 | |
LT9010 |
Formats Available
Prod No. | Description |
---|---|
LT2800 | |
LT2805 |
