MedPath

Denosumab Advanced Drug Monograph

Published:Jul 14, 2025

Generic Name

Denosumab

Brand Names

Prolia, Xgeva, Wyost, Jubbonti, Stoboclo, Obodence, Osenvelt, Xbryk, Jubereq, Osvyrti, Evfraxy

Drug Type

Biotech

CAS Number

615258-40-7

Associated Conditions

Bone Fractures, Bone Giant Cell Tumor, Bone Loss, Bone pain, Spinal Cord Compression, Bone destruction, High risk of fracture Osteoporosis, Refractory Hypercalcemia of malignancy

Denosumab (DB06643): A Comprehensive Monograph on a First-in-Class RANKL Inhibitor

1.0 Executive Summary

Denosumab is a landmark therapeutic agent, representing a first-in-class, fully human immunoglobulin G2 (IgG2) monoclonal antibody that functions as a potent and specific inhibitor of the Receptor Activator of Nuclear Factor Kappa-B Ligand (RANKL) [1, 2, 3, 4]. Developed by Amgen, it is marketed under two distinct brand names, Prolia® and Xgeva®, which correspond to different dosages, administration schedules, and clinical indications; these formulations are not interchangeable [5, 6, 7]. Prolia® is primarily indicated for the treatment of osteoporosis and related conditions of bone loss, while Xgeva® is used in oncologic settings to prevent skeletal-related events (SREs) and manage other cancer-related bone complications [3, 5].

The mechanism of action of denosumab is a feat of biomimicry, targeting the fundamental signaling pathway that governs bone remodeling. By binding to and neutralizing RANKL, denosumab mimics the function of the body's natural decoy receptor, osteoprotegerin (OPG), thereby preventing the formation, function, and survival of osteoclasts—the cells responsible for bone resorption [1, 8]. This targeted intervention leads to a rapid and profound reduction in bone turnover, resulting in increased bone mineral density (BMD), improved bone strength, and a significant reduction in fracture risk [5, 9].

Clinical evidence supporting denosumab's efficacy is robust. In postmenopausal osteoporosis, the pivotal FREEDOM trial demonstrated significant reductions in vertebral (68%), hip (40%), and non-vertebral (20%) fractures over three years [10, 11]. Long-term data extending to 10 years show continuous, non-plateauing gains in BMD, a feature that distinguishes it from bisphosphonates, and sustained low fracture rates [12, 13]. In oncology, Xgeva® has proven superior to the previous standard of care, zoledronic acid, in preventing SREs in patients with bone metastases from solid tumors like breast and prostate cancer, and non-inferior in patients with multiple myeloma [14, 15, 16].

The therapeutic benefits of denosumab are balanced by a unique and complex safety profile that is intrinsically linked to its potent mechanism. A critical risk is severe, sometimes fatal, hypocalcemia, which prompted the U.S. Food and Drug Administration (FDA) to issue a Boxed Warning, particularly for patients with advanced chronic kidney disease [17, 18]. Other significant risks include osteonecrosis of the jaw (ONJ) and atypical femoral fractures, which are class effects for potent antiresorptives [7]. Uniquely, the reversibility of denosumab's effect leads to a well-documented rebound in bone turnover upon discontinuation, creating a substantial risk of multiple vertebral fractures if not managed with a transition to an alternative antiresorptive therapy [10, 19]. This report provides an exhaustive analysis of denosumab, covering its molecular biology, pharmacology, extensive clinical trial data, comprehensive safety considerations, and its established position in the therapeutic landscape of bone diseases.

2.0 Molecular Profile and Manufacturing

[2.1 Chemical and Structural Identity]

Denosumab (CAS Number: 615258-40-7) is a fully human monoclonal antibody of the immunoglobulin G2 (IgG2) isotype [5, 8, 20]. The selection of the IgG2 subclass is a critical aspect of its molecular design. Among human IgG subclasses, IgG2 exhibits the weakest effector functions; it binds poorly to the C1q component of the complement system and has very low affinity for activating Fcγ receptors, resulting in minimal antibody-dependent cellular cytotoxicity (ADCC) and complement-dependent cytotoxicity (CDC) [21, 22]. This characteristic is highly desirable for a therapeutic antibody whose primary purpose is to neutralize a target ligand (RANKL) rather than to eliminate target cells. By engineering denosumab as an IgG2, its pharmacologic activity is almost exclusively focused on antigen binding and neutralization, minimizing the potential for off-target inflammatory or cytotoxic effects mediated by its Fc region. This design contributes to a more targeted therapeutic action, which is particularly important for a drug intended for long-term use in chronic conditions [22].

The antibody has an approximate molecular weight of 147 kDa [23, 24]. Its chemical formula is cited as C6404​H9912​N1724​O2004​S50​, with a calculated molar mass of approximately 144,720 g/mol [1, 25, 26]. The structure is typical of an immunoglobulin, consisting of two identical heavy chains and two identical light chains [3]. Specifically, each heavy chain is composed of 448 amino acids, and each light chain contains 215 amino acids. The integrity of the structure is maintained by four intramolecular disulfide bridges [3]. The precise primary amino acid sequences of these chains determine the antibody's high specificity and affinity for its target [23, 27]. Advanced techniques such as random peptide phage display library screenings have been used to identify the specific epitope on RANKL that denosumab recognizes. These studies have pinpointed a linear epitope on the DE loop of human RANKL, located between amino acid residues Threonine-233 (T233) and Tyrosine-241 (Y241), which overlaps with the binding sites of both RANK and OPG [25, 28].

[2.2 Recombinant Production and Formulation]

Denosumab is a biotech drug produced via recombinant DNA technology [29]. The antibody is expressed in a genetically engineered mammalian cell line, specifically Chinese Hamster Ovary (CHO) cells, which have been transfected with the DNA encoding the human heavy and light chains of denosumab [23, 30, 31, 32]. This production method was developed using XenoMouse transgenic mouse technology, a platform for generating fully human antibodies [33].

The manufacturing process begins with large-scale cell culture in bioreactors [30]. Following the culture period, the denosumab antibody is harvested from the surrounding culture medium. An extensive, multi-step purification process is employed to isolate the drug substance and ensure its purity, potency, and safety. This process includes three critical chromatography steps: Protein A affinity chromatography, cation exchange chromatography, and hydrophobic interaction chromatography. These steps are designed to separate the denosumab antibody from host cell proteins and other impurities. The process also incorporates robust viral inactivation and removal procedures to address any potential viral or prion safety concerns, including those related to animal-derived components used in the cell culture medium [30].

The final drug product is formulated as a sterile, preservative-free, clear, and colorless to pale yellow injectable solution [23, 24, 29]. For the Prolia® and Xgeva® pre-filled syringe presentations, the purified drug substance is diluted to its final concentration (60 mg/mL for Prolia®, 70 mg/mL for Xgeva®) in a formulation buffer. This buffer consists of 10 mM acetate at a pH of 5.2, with 4.7% (w/v) sorbitol as a stabilizer [29, 30, 32]. Polysorbate 20 (0.01%) is added as a surfactant to prevent protein aggregation [24, 30]. The pre-filled syringe is constructed from Type 1 glass and features a plunger stopper made of bromobutyl rubber laminated with a fluoropolymer film. The materials, including the needle shield, are not made with natural rubber latex, minimizing the risk of latex allergies [19, 30].

3.0 Mechanism of Action: Targeted Inhibition of the RANK-RANKL Pathway

[3.1 The Physiology of Bone Remodeling]

Bone is a dynamic tissue that undergoes a continuous process of remodeling throughout life, ensuring the maintenance of skeletal integrity and mineral homeostasis. This process is tightly regulated by a delicate balance between bone resorption, mediated by osteoclasts, and bone formation, mediated by osteoblasts [1, 34]. At the core of this regulation lies a critical signaling axis composed of three key proteins: Receptor Activator of Nuclear Factor Kappa-B (RANK), its ligand (RANKL), and a natural inhibitor, osteoprotegerin (OPG) [35].

RANKL, a member of the tumor necrosis factor (TNF) cytokine superfamily, is the principal mediator of osteoclast activity. It is expressed as both a membrane-bound and a soluble protein by osteoblasts, osteocytes, and other cells within the bone marrow microenvironment [1, 8, 36]. RANKL exerts its effects by binding to its cognate receptor, RANK, which is expressed on the surface of osteoclast precursors and mature osteoclasts [1, 34]. This binding event triggers a downstream signaling cascade that is essential for the differentiation of hematopoietic precursors into mature, multinucleated osteoclasts, as well as for the activation and survival of these mature cells [8, 37].

To prevent excessive bone resorption, the body produces OPG, a soluble decoy receptor also secreted by osteoblasts [1, 35]. OPG functions as a physiological antagonist by binding to RANKL with high affinity, thereby preventing it from interacting with RANK. The relative ratio of RANKL to OPG is a critical determinant of bone mass. In pathological states such as postmenopausal osteoporosis, the decline in estrogen levels leads to an upregulation of RANKL expression, shifting the RANKL/OPG ratio in favor of RANKL. This overwhelms the protective capacity of OPG, resulting in increased osteoclast formation and activity, excessive bone resorption, and a net loss of bone mass [20].

[3.2 Pharmacologic Intervention with Denosumab]

Denosumab is a RANKL inhibitor that functions as a therapeutic mimic of endogenous OPG [1, 2, 5]. It is a fully human monoclonal antibody that binds with very high affinity (dissociation equilibrium constant, Kd​, of 3 pM) and specificity to both soluble and membrane-bound forms of human RANKL [25, 38]. This binding is highly specific; denosumab does not bind to other members of the TNF family, such as TNFα or TRAIL [25].

By sequestering RANKL, denosumab effectively blocks its ability to bind to and activate the RANK receptor on osteoclasts and their precursors [4, 8, 39, 40]. This targeted interruption of the RANKL/RANK signaling pathway has two profound consequences: it inhibits the differentiation and maturation of pre-osteoclasts into functional, bone-resorbing osteoclasts, and it induces apoptosis (programmed cell death) in existing mature osteoclasts, thereby reducing their lifespan and overall number [1, 8].

The ultimate pharmacodynamic effect is a rapid, deep, and sustained suppression of bone resorption [41]. This allows the balance of bone remodeling to shift away from resorption and toward a state of relative bone conservation or formation. The result is a measurable increase in bone mass and strength throughout the skeleton, in both the dense cortical bone that forms the outer shell of bones and the spongy trabecular bone found within [1, 9].

This "signal-blocking" mechanism of action represents a fundamental departure from that of bisphosphonates, the other major class of antiresorptive agents. Bisphosphonates must first bind to hydroxyapatite in the bone matrix and are then internalized by actively resorbing osteoclasts, where they induce apoptosis through enzymatic inhibition [41]. In contrast, denosumab acts extracellularly within the bone microenvironment to neutralize a key signaling molecule. This distinction explains several key clinical differences between the two drug classes. Denosumab's mechanism allows for a more rapid onset of action, with bone resorption markers decreasing by over 80% within days of administration [3]. It also accounts for its potent effects on cortical bone, a site where bisphosphonates are considered less effective, by reducing intracortical remodeling and porosity [42]. Most critically, because denosumab is a circulating antibody with a defined half-life and is not incorporated into the bone matrix, its effects are fully reversible upon drug clearance—a property that is both a potential therapeutic advantage and the source of its most significant clinical challenge: the rebound effect upon discontinuation [10, 41].

4.0 Pharmacokinetics and Pharmacodynamics (ADME)

[4.1 Absorption and Distribution]

Denosumab is administered exclusively via subcutaneous (SC) injection, typically into the upper arm, upper thigh, or abdomen [8, 43]. Intravenous or intramuscular administration is not appropriate [8].

Following a single 60 mg SC dose (the Prolia® dose), denosumab is absorbed into the systemic circulation, reaching a mean maximum serum concentration (Cmax​) of 6.75 mcg/mL. The time to reach this maximum concentration (Tmax​) is a median of 10 days, with a range of 3 to 21 days [3, 8]. The absolute bioavailability of a 60 mg SC dose is estimated to be 61% [8]. The pharmacokinetics of denosumab are non-linear at doses below 60 mg but exhibit approximately dose-proportional increases in exposure at higher doses, such as the 120 mg Xgeva® dose [8, 44]. Upon repeated dosing every six months (Prolia®), no drug accumulation or time-dependent changes in pharmacokinetics are observed [19]. For the more frequent Xgeva® regimen (120 mg every 4 weeks), steady-state serum concentrations are typically achieved by the sixth month of treatment [3].

Regarding distribution, studies in male volunteers have shown that denosumab is distributed into seminal fluid, though at very low concentrations. The maximum seminal fluid concentration is approximately 2% of the corresponding serum level, suggesting minimal paternal exposure [3, 8].

[4.2 Metabolism and Elimination]

As a large protein, denosumab follows the metabolic and elimination pathways typical of endogenous immunoglobulins. It is not metabolized by hepatic cytochrome P450 enzymes, nor is it eliminated via renal excretion [8, 45]. Instead, denosumab is cleared from the circulation primarily through breakdown into small peptides and individual amino acids by the reticuloendothelial system, a process known as proteolysis [1, 8].

This non-renal, non-hepatic clearance pathway is a clinically significant feature. It means that no dose adjustment is necessary for patients with either renal or hepatic impairment [8, 45]. This contrasts sharply with many bisphosphonates, which are cleared by the kidneys and are often contraindicated in patients with severe chronic kidney disease (CKD) [46, 47].

Following peak concentrations, serum denosumab levels decline over a period of 4 to 5 months. The elimination half-life is approximately 25.4 to 32 days, which supports the long dosing intervals of every 6 months for Prolia® and every 4 weeks for Xgeva® [8, 19].

The pharmacokinetic and pharmacodynamic profile of denosumab gives rise to a significant clinical paradox. Its elimination pathway makes it a suitable and often preferred antiresorptive agent for patients with severe CKD, in whom bisphosphonates may be contraindicated [8, 48]. However, this is precisely the patient population at the highest risk for the drug's most severe adverse effect: life-threatening hypocalcemia [5, 18]. Patients with advanced CKD frequently suffer from CKD-Mineral and Bone Disorder (CKD-MBD), a systemic condition characterized by impaired calcium homeostasis, abnormal vitamin D metabolism, and secondary hyperparathyroidism [19]. In these patients, the kidneys' ability to conserve calcium and produce active vitamin D is compromised. When denosumab is administered, it potently and rapidly shuts down bone resorption, which is the body's primary mechanism for mobilizing stored calcium into the bloodstream [3]. In a patient with healthy kidneys, the endocrine system would compensate for this. In a patient with advanced CKD, these compensatory mechanisms are defunct. The result is an unbuffered drop in serum calcium that can be rapid, profound, and difficult to manage. Thus, the very pharmacokinetic property that makes denosumab an option in CKD interacts with the underlying pathophysiology of the disease to create its most dangerous on-target pharmacodynamic risk.

[4.3 Pharmacodynamic Effects]

The pharmacodynamic response to denosumab is characterized by a rapid, profound, and sustained suppression of bone turnover markers (BTMs), reflecting its potent inhibition of osteoclast activity [34].

For the 60 mg Prolia® dose used in osteoporosis, levels of serum type 1 C-telopeptide (CTX), a marker of bone resorption, are reduced by approximately 85% within just 3 days of injection. Maximal suppression is observed by one month, and this effect is sustained with continued dosing every six months [3]. For the 120 mg Xgeva® dose used in oncology, levels of urinary N-terminal telopeptide corrected for creatinine (uNTx/Cr) show a median reduction of 82% within one week of the first dose [3, 44].

The pharmacodynamic effects of denosumab are also fully reversible. Following discontinuation of therapy, as the antibody is cleared from the system, the inhibitory effect on RANKL ceases. This leads to a marked rebound in bone resorption. BTMs not only return to baseline but overshoot, increasing to levels 40% to 60% above pre-treatment values within the first year after the last dose, before eventually returning to baseline [3, 10]. This rebound phenomenon is the direct physiological cause of the increased risk of vertebral fractures observed upon treatment cessation.

5.0 Clinical Efficacy in Osteoporosis and Related Bone Loss Conditions (Prolia®)

[5.1 Postmenopausal Osteoporosis: The FREEDOM Trial and Long-Term Extension]

The cornerstone of evidence for denosumab's efficacy in postmenopausal osteoporosis is the FREEDOM (Fracture REduction Evaluation of Denosumab in Osteoporosis Every 6 Months) trial [10]. This pivotal, three-year, international, randomized, double-blind, placebo-controlled Phase 3 study enrolled 7,868 women aged 60 to 90 with osteoporosis, defined by a bone mineral density (BMD) T-score below -2.5 at the lumbar spine or total hip [11, 49, 50]. Participants were randomized to receive either denosumab 60 mg subcutaneously every 6 months or a matching placebo, with all participants receiving daily calcium and vitamin D supplementation [49].

The primary endpoint was the incidence of new radiographic vertebral fractures at 36 months. The results were unequivocally positive and clinically significant. As detailed in Table 1, denosumab treatment led to a 68% relative risk reduction in new vertebral fractures compared to placebo. The trial also met its key secondary endpoints, demonstrating a 40% relative risk reduction in hip fractures and a 20% relative risk reduction in all non-vertebral fractures [10, 11, 51].

Table 1: Key Efficacy Outcomes of the Pivotal 3-Year FREEDOM Trial in Postmenopausal Osteoporosis
Fracture TypeOutcomeDenosumab 60 mg Q6M (n=3902)Placebo (n=3906)Cumulative Incidence (%)Relative Risk Reduction (%)95% Confidence Interval
New Vertebral FractureIncidence892762.3% vs. 7.2%68%0.26 to 0.41
Hip FractureIncidence27460.7% vs. 1.2%40%0.37 to 0.97
Non-Vertebral FractureIncidence2553136.5% vs. 8.0%20%0.67 to 0.98
Data synthesized from sources.10

Following the completion of the 3-year FREEDOM trial, eligible participants were invited to enroll in a 7-year open-label extension study, in which all patients received denosumab. This provided data for up to 10 years of continuous therapy for the original denosumab group (long-term group) and 7 years of therapy for those who crossed over from placebo (crossover group) [12, 52, 53].

The long-term extension study yielded two critical findings. First, the fracture protection was sustained. The yearly incidence of new vertebral and non-vertebral fractures remained low throughout the 10-year period, with rates for non-vertebral fractures being even lower in years 4 through 10 than in the initial 3-year trial [12, 54]. A sophisticated "virtual twin" analysis, which modeled a hypothetical placebo group over 10 years, estimated that continuous denosumab treatment resulted in a 51% relative risk reduction for major osteoporotic fractures [52].

Second, and perhaps most remarkably, denosumab treatment resulted in continuous, non-plateauing gains in BMD over the entire 10-year period. This stands in stark contrast to bisphosphonates, for which BMD gains typically reach a plateau after 3 to 4 years of treatment [12, 47]. In the long-term denosumab group, BMD from the original baseline increased by an impressive 21.7% at the lumbar spine and 9.2% at the total hip after 10 years [10, 12, 13, 53]. This unique BMD trajectory suggests that denosumab's profound and consistent suppression of bone resorption allows for a prolonged period during which bone formation can continuously outpace resorption, leading to a net accrual of bone mineral over a much longer timeframe than seen with other antiresorptives. This finding has significant implications for long-term management, challenging the applicability of "drug holidays" (common with bisphosphonates) and positioning denosumab as a therapy for which long-term continuous use is intended.

[5.2 Efficacy in Other Osteoporosis Populations]

The efficacy of Prolia® has been established in other important patient populations at high risk for fracture:

  • Male Osteoporosis: In a one-year, randomized, placebo-controlled trial involving 242 men with low BMD, treatment with Prolia® resulted in significantly greater increases in BMD compared to placebo. At 12 months, the mean increase in lumbar spine BMD was 5.7% in the denosumab group versus 0.9% in the placebo group [5, 19, 55]. Significant gains were also seen at the total hip, femoral neck, and trochanter [42].
  • Glucocorticoid-Induced Osteoporosis (GIOP): GIOP is the most common form of secondary osteoporosis. A pivotal Phase 3 study evaluated denosumab in 795 patients receiving long-term systemic glucocorticoid therapy (equivalent to ≥7.5 mg/day of prednisone) [5, 56]. The study compared denosumab 60 mg every 6 months to the oral bisphosphonate risedronate 5 mg daily. After 12 months, denosumab demonstrated superior increases in BMD at both the lumbar spine and total hip compared to the active comparator, risedronate [55, 56]. These results were consistent in both patients who were initiating glucocorticoids and those who were continuing long-term therapy, leading to the FDA's approval of this indication in May 2018 [56, 57].

[5.3 Management of Cancer Treatment-Induced Bone Loss]

Certain cancer treatments can accelerate bone loss and increase fracture risk. Prolia® is approved to mitigate this in two key settings:

  • Androgen Deprivation Therapy (ADT) for Prostate Cancer: ADT is a cornerstone of treatment for nonmetastatic prostate cancer but induces rapid bone loss. In a three-year study of 1,468 men on ADT, Prolia® significantly increased BMD at all measured sites compared to placebo. Critically, it also reduced the incidence of new vertebral fractures by 62% over 36 months [10, 19, 58].
  • Aromatase Inhibitor (AI) Therapy for Breast Cancer: Adjuvant AI therapy is standard for hormone receptor-positive breast cancer but also causes bone loss. A study in 252 postmenopausal women with early-stage breast cancer receiving AI therapy showed that Prolia® led to significant increases in lumbar spine BMD (7.6% vs -1.4% for placebo) and total hip BMD (4.7% vs -0.8% for placebo) over 24 months [10, 19, 58].

6.0 Clinical Efficacy in Oncologic Indications (Xgeva®)

[6.1 Prevention of Skeletal-Related Events (SREs)]

Xgeva®, administered as a 120 mg subcutaneous injection every 4 weeks, is a cornerstone therapy for preventing SREs in patients whose cancer has metastasized to the bone [5, 7]. SREs are defined as pathologic fractures, the need for radiation or surgery to bone for pain or stabilization, and spinal cord compression. These events cause significant morbidity, pain, and disability [59, 60]. The clinical development program for Xgeva® was notable for its large, head-to-head trials comparing its efficacy directly against the then-standard of care, the intravenous bisphosphonate zoledronic acid (Zometa®) [14, 60].

  • Solid Tumors (Breast and Prostate Cancer): Three pivotal Phase 3 trials established Xgeva's role in patients with bone metastases from solid tumors.
  • In a trial of 2,046 patients with advanced breast cancer, Xgeva® was found to be superior to zoledronic acid in delaying the time to the first on-study SRE, with a hazard ratio (HR) of 0.82 (95% CI, 0.71 to 0.95; p=0.01). It was also superior in delaying the time to first and subsequent SREs [15, 61, 62].
  • In a trial of 1,901 men with castration-resistant prostate cancer, Xgeva® was again superior to zoledronic acid, significantly delaying the time to first SRE with an HR of 0.82 (95% CI, 0.71 to 0.95; p=0.008) [14, 16].
  • A third trial in 1,776 patients with other solid tumors or multiple myeloma also showed Xgeva® reduced the risk of a first SRE by 16% compared to zoledronic acid [14].
  • Multiple Myeloma: In this disease, which is characterized by osteolytic bone lesions, the efficacy of Xgeva® was compared to zoledronic acid in the large, international '482' clinical trial involving 1,718 patients with newly diagnosed multiple myeloma [48]. The study demonstrated that Xgeva® was non-inferior to zoledronic acid in preventing SREs [14, 48]. This finding, coupled with Xgeva's favorable profile in patients with renal impairment (a common comorbidity in myeloma), led to its FDA approval for this indication in January 2018 [63, 64].

The differential outcomes—superiority in solid tumors versus non-inferiority in multiple myeloma—suggest a nuanced interaction with tumor-specific biology. The "vicious cycle" in bone metastases from breast and prostate cancer involves tumor cells secreting factors that stimulate osteoclasts, which in turn release growth factors from the bone matrix that fuel tumor growth [35]. Denosumab's potent and specific blockade of the RANKL pathway may interrupt this cycle more effectively than the less specific mechanism of zoledronic acid. In multiple myeloma, while RANKL is a key driver of osteolysis, other signaling pathways may also play a prominent role. Furthermore, some research has suggested that bisphosphonates may have direct anti-myeloma properties that could counterbalance denosumab's superior antiresorptive effect, leading to an overall non-inferior outcome. This distinction means that while Xgeva® is often a preferred first-line agent for SRE prevention in solid tumor metastases, the choice in multiple myeloma may be more influenced by factors such as a patient's renal function, risk of acute-phase reactions, and administration preference [48].

Table 2: Comparative Efficacy of Xgeva® vs. Zoledronic Acid for SRE Prevention in Bone Metastases
Trial PopulationPrimary EndpointHazard Ratio (HR) or Rate Ratio (RR) (Xgeva vs. Zoledronic Acid)95% Confidence Intervalp-valueKey Finding
Advanced Breast CancerTime to first on-study SREHR = 0.820.71 to 0.950.01Superiority
Castration-Resistant Prostate CancerTime to first on-study SREHR = 0.820.71 to 0.950.008Superiority
Multiple MyelomaTime to first on-study SREN/AN/AN/ANon-inferiority
Other Solid TumorsTime to first on-study SREHR = 0.84N/AN/ASuperiority
Data synthesized from sources.14 "N/A" indicates data not specified in the provided sources for that specific endpoint.

[6.2 Treatment of Giant Cell Tumor of Bone (GCTB)]

Xgeva® is approved for the treatment of GCTB in adults and skeletally mature adolescents where the tumor is unresectable or where surgical resection would result in severe morbidity, such as limb amputation or joint removal [5, 65, 66]. GCTB is a benign but locally aggressive tumor characterized by the presence of numerous osteoclast-like giant cells whose proliferation is driven by RANKL [14].

Denosumab's mechanism directly targets the underlying pathology of GCTB. Clinical trials have demonstrated its effectiveness in controlling the disease. In one key study involving 37 patients, 86% of participants achieved a treatment response, defined as the elimination of at least 90% of the giant cells or no progression of the tumor after 25 weeks [14]. A larger study of 507 patients showed that Xgeva® treatment prevented the need for planned morbid surgery in approximately half of the eligible patients. For others, it allowed for less extensive, function-sparing surgery than was initially planned [14]. This indication was approved by the FDA in June 2013 [63, 66].

[6.3 Management of Hypercalcemia of Malignancy (HCM)]

Xgeva® is also indicated for the treatment of HCM that is refractory to bisphosphonate therapy [5, 63, 67]. HCM is a serious metabolic complication of advanced cancer that can lead to renal failure, coma, and death if untreated [68].

The approval for this indication, granted in December 2014, was based on results from an open-label, single-arm study in patients with advanced cancer and persistent HCM despite recent intravenous bisphosphonate treatment [68]. The primary endpoint was the proportion of patients who achieved a response, defined as an albumin-corrected serum calcium level of 11.5 mg/dL or less, within 10 days of the first Xgeva® dose. The study met its primary endpoint, with a response rate of 63.6%. The median time to response was 9 days, highlighting denosumab's rapid action in this critical setting [46, 68].

7.0 Comprehensive Safety Profile and Risk Management

The safety profile of denosumab is well-characterized but complex, with risks that are direct, predictable consequences of its potent and specific inhibition of the RANKL pathway. Managing these risks is paramount to its safe and effective use.

[7.1 Adverse Reactions]

The profile of common adverse reactions differs slightly between the lower-dose, less frequent Prolia® regimen and the higher-dose, more frequent Xgeva® regimen.

  • Prolia® (Osteoporosis Indications): The most frequently reported adverse reactions in clinical trials (occurring in >5% of patients and more commonly than placebo) include back pain, pain in extremity, musculoskeletal pain, hypercholesterolemia, and cystitis. Pancreatitis has also been reported [19]. In men with osteoporosis, common reactions were back pain, arthralgia, and nasopharyngitis [19].
  • Xgeva® (Oncologic Indications): In patients with bone metastases from solid tumors, the most common adverse reactions were fatigue/asthenia, hypophosphatemia, and nausea. For patients with multiple myeloma, common reactions included diarrhea, nausea, anemia, back pain, thrombocytopenia, peripheral edema, and hypocalcemia [5, 7].

Across both formulations, other notable adverse events include an increased risk of infections, particularly serious skin infections like cellulitis, as well as abdominal, urinary tract, and ear infections [1, 5]. This is thought to be an on-target effect, as the RANKL pathway plays a role in immune function, including T-cell differentiation and dendritic cell maturation [1, 21]. Dermatologic reactions such as eczema, dermatitis, and rashes are also common [1, 19]. Hypersensitivity reactions, ranging from rash and urticaria to severe anaphylaxis, have been reported [5].

[7.2 Significant Risks and Mitigation Strategies]

Four major safety concerns require specific attention and proactive management strategies. These are summarized in Table 3.

Table 3: Summary of Key Safety Risks and Recommended Management for Denosumab
RiskRisk Profile & Key FeaturesHigh-Risk Patient PopulationsRecommended Monitoring
Severe Hypocalcemia (FDA Boxed Warning)Potentially life-threatening drop in serum calcium. Can cause muscle spasms, seizures, cardiac arrhythmias, and death. Risk is highest in the first few weeks after dosing [5, 18, 19].Patients with advanced Chronic Kidney Disease (CKD), especially eGFR < 30 mL/min or on dialysis. Patients with CKD-Mineral and Bone Disorder (CKD-MBD) [18, 69].Correct pre-existing hypocalcemia before first dose. Monitor serum calcium, PTH, and vitamin D levels at baseline. In CKD patients, monitor serum calcium weekly for the first month, then monthly [19].
Osteonecrosis of the Jaw (ONJ)Exposed, necrotic bone in the jaw, often following a dental procedure. Can cause pain, infection, and tooth loss. Incidence is higher with the Xgeva® dose/schedule [1, 7].History of invasive dental procedures (e.g., tooth extraction), poor oral hygiene, cancer, use of dental appliances, concomitant corticosteroids or anti-angiogenic therapy [7].A routine oral exam by a dentist is recommended before starting therapy. Periodic monitoring during therapy [7].
Atypical Femoral Fracture (AFF)Transverse or short oblique fractures of the subtrochanteric or diaphyseal femoral shaft with minimal or no trauma. May be bilateral. Often preceded by prodromal thigh or groin pain [1, 7, 8].Patients on long-term antiresorptive therapy. Concomitant glucocorticoid use may be a risk factor [7].Evaluate any patient presenting with new or unusual thigh, hip, or groin pain to rule out an incomplete fracture. Assess contralateral limb if an AFF is identified [7].
Multiple Vertebral Fractures (MVF) Upon DiscontinuationRapid reversal of antiresorptive effect upon drug clearance leads to a rebound in bone turnover above baseline, causing rapid bone loss and a high risk of multiple, often spontaneous, vertebral fractures [10, 19].All patients discontinuing denosumab. Higher risk in those with a history of osteoporosis or prior fractures [43].No specific monitoring test. The risk is predictable based on the drug's mechanism and half-life.

[7.3 Contraindications, Warnings, and Drug Interactions]

  • Contraindications: Denosumab is strictly contraindicated in patients with pre-existing, uncorrected hypocalcemia and in patients with a known history of systemic hypersensitivity to denosumab or any of its components [29, 58]. It is also contraindicated during pregnancy due to the potential for fetal harm, as RANKL is crucial for normal development [8, 58].
  • Warnings and Precautions: In addition to the major risks detailed above, the labeling includes warnings for:
  • Serious Infections: Including skin, abdominal, urinary tract, and ear infections that may require hospitalization, and endocarditis [5, 19, 58].
  • Dermatologic Adverse Reactions: Including dermatitis, eczema, rashes, and rare cases of Drug Reaction with Eosinophilia and Systemic Symptoms (DRESS) [5, 19].
  • Severe Musculoskeletal Pain: Severe and occasionally incapacitating bone, joint, and/or muscle pain has been reported, which may require discontinuation [58].
  • Suppression of Bone Turnover: The significance of long-term, profound suppression of bone turnover is not fully known, but it is the likely underlying cause of ONJ and AFF [58].
  • Drug Interactions: As a monoclonal antibody cleared by proteolysis, denosumab has a low potential for pharmacokinetic drug-drug interactions, and no formal interaction studies have been conducted [29, 45]. However, pharmacodynamic interactions are a concern. Co-administration with immunosuppressive medications or systemic corticosteroids may increase the risk of serious infections [8, 70]. Concomitant use with calcimimetic agents like etelcalcetide, which also lower serum calcium, can severely exacerbate the risk of hypocalcemia and should be avoided or managed with extreme caution [8].

8.0 Regulatory and Commercial Landscape

[8.1 Global Approval History]

The clinical development of denosumab by Amgen was a highly strategic and successful program, leveraging a single molecule to address distinct but mechanistically related diseases in both metabolic bone disorders and oncology. This dual-pronged approach led to a rapid succession of approvals for its two brand formulations, Prolia® and Xgeva®, from major regulatory agencies worldwide.

  • U.S. Food and Drug Administration (FDA):
  • Prolia®: The Biologics License Application (BLA) was submitted in December 2008 [57]. Following a review, Prolia® received its first FDA approval on June 1, 2010, for the treatment of postmenopausal women with osteoporosis at high risk for fracture [1, 3, 51, 57]. The approval was subsequently expanded to include: treatment of bone loss in patients undergoing hormone ablation therapy for breast or prostate cancer (September 2011), treatment to increase bone mass in men with osteoporosis (September 2012), and treatment of glucocorticoid-induced osteoporosis (May 2018) [56, 57].
  • Xgeva®: The BLA for Xgeva® was granted a priority review. It was first approved on November 18, 2010, for the prevention of SREs in patients with bone metastases from solid tumors [1, 60, 63, 71]. This was followed by a series of indication expansions: treatment of giant cell tumor of bone (GCTB) in June 2013, treatment of hypercalcemia of malignancy (HCM) refractory to bisphosphonates in December 2014, and prevention of SREs in patients with multiple myeloma in January 2018 [63, 66, 68].
  • European Medicines Agency (EMA):
  • Prolia®: Following a positive opinion from the Committee for Medicinal Products for Human Use (CHMP) in December 2009, the European Commission granted a marketing authorisation for Prolia® on May 26, 2010 [1, 55].
  • Xgeva®: The marketing authorisation for Xgeva® was granted in July 2011 [1].

This timeline illustrates a remarkably efficient translation of a foundational scientific discovery—the RANKL pathway—into two blockbuster drugs addressing large unmet medical needs.

[8.2 The Emergence of Biosimilars]

With the expiration of key patents for denosumab, the market has entered a new phase characterized by the introduction of biosimilar competitors. Biosimilars are biologic products that are demonstrated to be highly similar to an already-approved reference product, with no clinically meaningful differences in terms of safety, purity, and potency [9, 36].

  • In the United States: The first denosumab biosimilars received FDA approval in March 2024. Sandoz's products, Jubbonti™ (denosumab-bbdz), a biosimilar to Prolia®, and Wyost™ (denosumab-bbdz), a biosimilar to Xgeva®, were approved with an interchangeability designation, meaning a pharmacist can substitute them for the reference product without consulting the prescriber (subject to state laws) [1, 17, 72]. Subsequently, other biosimilars have been approved, including Bomyntra/Conexxence (denosumab-bnht) and Ospomyv/Xbryk (denosumab-dssb), signaling a significant shift in the market landscape [1, 46, 72].
  • In the European Union: The CHMP issued positive opinions recommending marketing authorization for the first denosumab biosimilars, including Wyost and Jubbonti, in March 2024 [1]. These were subsequently authorized for medical use.

The arrival of biosimilars is expected to increase market competition, potentially lower healthcare costs, and improve patient access to this important class of therapy [36, 73]. This transition marks the end of Amgen's market exclusivity and will require ongoing pharmacovigilance to ensure that the safety and efficacy profiles of the biosimilars continue to match that of the reference products in real-world clinical practice.

9.0 Synthesis and Future Perspectives

[9.1 Denosumab in Clinical Context: A Comparative Analysis vs. Bisphosphonates]

Denosumab and bisphosphonates are the two main classes of antiresorptive therapies used to treat bone loss. While both aim to reduce bone resorption, their distinct mechanisms of action lead to important differences in their clinical profiles.

  • Efficacy: Denosumab consistently demonstrates superior efficacy in increasing BMD compared to oral bisphosphonates like alendronate, particularly at cortical bone sites [42, 47]. The long-term, non-plateauing BMD gains observed with denosumab over 10 years are unique [12]. In oncology, Xgeva® is superior to zoledronic acid for preventing SREs in patients with bone metastases from solid tumors, though it is non-inferior in multiple myeloma [14, 74]. Overall, for bone density improvement and SRE prevention in many settings, denosumab represents a more potent agent.
  • Administration and Adherence: The administration of denosumab as a subcutaneous injection every 6 months (Prolia®) or every 4 weeks (Xgeva®) is generally perceived as more convenient than the strict oral dosing requirements of oral bisphosphonates (e.g., fasting, remaining upright) or the need for intravenous infusions for drugs like zoledronic acid [42]. This convenience has been shown to lead to greater patient satisfaction and better medication adherence, which is critical for long-term efficacy in chronic diseases like osteoporosis [42, 75].
  • Safety and Tolerability: The safety profiles present a complex trade-off. Denosumab avoids the acute-phase reactions (fever, flu-like symptoms) and potential for renal toxicity associated with intravenous bisphosphonates, making it a valuable option for patients with kidney disease [47, 74, 76]. However, it introduces its own set of significant risks. The risk of severe hypocalcemia in patients with advanced CKD is a major concern that requires careful management [18]. The risk of rebound vertebral fractures upon discontinuation is a unique and critical liability of denosumab that is not seen with bisphosphonates, whose effects wane much more gradually [10]. The risks of ONJ and AFF are associated with both classes of potent antiresorptives, though the risk of ONJ with denosumab appears to be reversible, declining after the drug is cleared, which may not be the case for bisphosphonates that remain in the bone matrix for years [1].

[9.2 Unresolved Questions and Future Research]

Despite over a decade of clinical use and extensive study, several important questions regarding denosumab remain, pointing to key areas for future research.

  • Optimal Duration of Therapy: The FREEDOM extension trial provides robust data for up to 10 years of use, showing continued benefits [12, 13]. However, osteoporosis is a lifelong condition. The long-term effects of therapy beyond 10 years are unknown. It is unclear if BMD gains continue indefinitely, if fracture risk remains low, and whether the risk of rare adverse events like AFF increases with even longer duration of profound bone turnover suppression.
  • Optimal Discontinuation Management: The prevention of rebound vertebral fractures upon cessation is a critical unmet need. While transitioning to a bisphosphonate is the current recommendation, the optimal regimen is not established [10]. Research is ongoing to determine the ideal timing, dose, and duration of bisphosphonate therapy needed to effectively "lock in" the bone mass gains from denosumab and prevent the surge in bone resorption [77]. Studies like the ZOOM study are evaluating different zoledronic acid regimens following denosumab discontinuation [77].
  • Potential Direct Anti-Tumor Effects: The RANK/RANKL pathway is implicated not only in bone metabolism but also in immune regulation and tumorigenesis [21, 78]. There is emerging evidence from preclinical models and early clinical trials that denosumab may have direct or indirect anti-tumor effects beyond its action on osteoclasts. For instance, a recent trial suggested that denosumab could boost the anti-tumor immune response in early-stage breast cancer by increasing the infiltration of immune cells into the tumor [79]. Further investigation into this potential immune-modulatory role could open new therapeutic avenues for denosumab in oncology.
  • Optimal Sequencing with Anabolic Therapies: The ideal way to sequence denosumab with anabolic (bone-building) agents like teriparatide is an area of active study. Clinical trials are exploring whether starting with an anabolic agent to build new bone, followed by denosumab to consolidate and maintain those gains, provides a greater overall benefit than either approach alone [77, 80]. Understanding this interplay is key to optimizing lifelong management strategies for severe osteoporosis.

10.0 Conclusion

Denosumab has fundamentally altered the therapeutic landscape for a wide spectrum of bone disorders, from common postmenopausal osteoporosis to life-threatening oncologic complications. As the first and only approved inhibitor of the RANKL pathway, it represents a triumph of translational medicine, turning a basic science discovery into a potent and widely used therapeutic agent. Its efficacy in increasing bone mineral density and reducing fracture risk is well-established and, in many cases, superior to previous standards of care. The convenience of its subcutaneous administration has further solidified its role as a preferred option for many patients and clinicians.

However, the power of denosumab is inextricably linked to a distinct and challenging safety profile. The major adverse events—severe hypocalcemia in renal impairment, osteonecrosis of the jaw, atypical femoral fractures, and rebound vertebral fractures upon cessation—are not idiosyncratic toxicities but are logical, on-target consequences of its potent and reversible mechanism. This reality demands a sophisticated and proactive approach to clinical management. The decision to initiate denosumab therapy is not a short-term choice but a commitment to a long-term strategy that requires careful patient selection, diligent monitoring, and, most critically, a well-defined plan for either continuous lifelong therapy or a carefully managed transition to an alternative agent. As the era of biosimilars begins, increasing access to this molecule, the imperative for clinicians to master the nuances of its use will only grow. Denosumab remains a powerful and indispensable tool, but one that commands respect for its unique biological effects.

Works cited

  1. Denosumab - Wikipedia, accessed July 14, 2025, https://en.wikipedia.org/wiki/Denosumab
  2. medlineplus.gov, accessed July 14, 2025, [https://medlineplus.gov/druginfo/meds/a610023.html#:~:text=Denosumab%20injection%20products%20(Xgeva%C2%AE,body%20to%20decrease%20bone%20breakdown.](https://www.google.com/url?q=https://medlineplus.gov/druginfo/meds/a610023.html%23:~:text%3DDenosumab%2520injection%2520products%2520(Xgeva%25C2%25AE,body%2520to%2520decrease%2520bone%2520breakdown.&sa=D&source=editors&ust=1752468164447763&usg=AOvVaw1h-kuSYQm7ak54uyMI1TqQ)
  3. Denosumab: Uses, Interactions, Mechanism of Action | DrugBank Online, accessed July 14, 2025, https://go.drugbank.com/drugs/DB06643
  4. What are RANKL inhibitors and how do they work? - Patsnap Synapse, accessed July 14, 2025, https://synapse.patsnap.com/article/what-are-rankl-inhibitors-and-how-do-they-work
  5. Denosumab: Bone Loss Uses, Side Effects, Warnings, Dosage - MedicineNet, accessed July 14, 2025, https://www.medicinenet.com/denosumab/article.htm
  6. Prolia® (denosumab) for Postmenopausal Osteoporosis, accessed July 14, 2025, https://www.prolia.com/
  7. Bone Metastases Treatment | XGEVA® (denosumab) for HCPs, accessed July 14, 2025, https://www.xgevahcp.com/
  8. Denosumab - StatPearls - NCBI Bookshelf, accessed July 14, 2025, https://www.ncbi.nlm.nih.gov/books/NBK535388/
  9. Prolia injection (denosumab): Side effects, cost, and more - Medical News Today, accessed July 14, 2025, https://www.medicalnewstoday.com/articles/prolia
  10. Denosumab - International Osteoporosis Foundation, accessed July 14, 2025, https://www.osteoporosis.foundation/health-professionals/treatment/denosumab
  11. Denosumab for prevention of fractures in postmenopausal women with osteoporosis - PubMed, accessed July 14, 2025, https://pubmed.ncbi.nlm.nih.gov/19671655/
  12. Denosumab in the Treatment of Osteoporosis: 10 Years Later: A Narrative Review - PMC, accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC8799550/
  13. Long-Term Efficacy and Safety of Denosumab: Insights beyond 10 Years of Use - Endocrinology and Metabolism, accessed July 14, 2025, https://www.e-enm.org/journal/view.php?doi=10.3803/EnM.2024.2125
  14. Xgeva | European Medicines Agency (EMA), accessed July 14, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/xgeva
  15. Denosumab Demonstrates Superiority Over Zometa(R) in Pivotal Phase 3 Head-to-Head Trial in Breast Cancer Patients With Bone Metastases - Amgen's investor, accessed July 14, 2025, https://investors.amgen.com/news-releases/news-release-details/denosumab-demonstrates-superiority-over-zometar-pivotal-phase-3/
  16. Denosumab Demonstrated Superiority Over Zometa(R) in Pivotal Phase 3 Head-to-Head Trial in Prostate Cancer Patients With Bone Metastases - Amgen's investor, accessed July 14, 2025, https://investors.amgen.com/news-releases/news-release-details/denosumab-demonstrated-superiority-over-zometar-pivotal-phase-3/
  17. Sandoz receives FDA approval for first and only denosumab biosimilars, accessed July 14, 2025, https://www.sandoz.com/sandoz-receives-fda-approval-first-and-only-denosumab-biosimilars/
  18. FDA Adds Boxed Warning to Denosumab (Prolia) for Severe Hypocalcemia - HCPLive, accessed July 14, 2025, https://www.hcplive.com/view/fda-adds-boxed-warning-to-denosumab-prolia-for-severe-hypocalcemia
  19. Treatment for Postmenopausal Osteoporosis | Prolia® (denosumab ..., accessed July 14, 2025, https://www.proliahcp.com/
  20. prolia-epar-public-assessment-report_en.pdf - EMA, accessed July 14, 2025, https://www.ema.europa.eu/en/documents/assessment-report/prolia-epar-public-assessment-report_en.pdf
  21. Anti-human RANKL Denosumab biosimilar | CAS # 615258-40-7 - InvivoGen, accessed July 14, 2025, https://www.invivogen.com/anti-human-rankl-denosumab
  22. The solution structure of the human IgG2 subclass is distinct from those for human IgG1 and IgG4 providing an explanation for their discrete functions - PMC, accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6635440/
  23. US20150246974A1 - Methods related to denosumab - Google Patents, accessed July 14, 2025, https://patents.google.com/patent/US20150246974A1/en
  24. Prolia® (denosumab) - GlobalRPH, accessed July 14, 2025, https://globalrph.com/drugs/prolia-denosumab/
  25. Denosumab | RANKL Inhibitor | CAS NO.:615258-40-7 - GlpBio, accessed July 14, 2025, https://www.glpbio.com/denosumab.html
  26. Denosumab - KEGG DRUG, accessed July 14, 2025, https://www.kegg.jp/entry/D03684
  27. Prolia comparability and biosimilarity studies - BioPharmaSpec, accessed July 14, 2025, https://biopharmaspec.com/biosimilar-testing/prolia-denosumab/
  28. Denosumab mimics the natural decoy receptor osteoprotegerin by interacting with its major binding site on RANKL | Oncotarget, accessed July 14, 2025, https://www.oncotarget.com/article/2160/text/
  29. Prolia, INN-denosumab - EMA, accessed July 14, 2025, https://www.ema.europa.eu/en/documents/product-information/prolia-epar-product-information_en.pdf
  30. Australian Public Assessment Report for Denosumab - Therapeutic ..., accessed July 14, 2025, https://www.tga.gov.au/sites/default/files/auspar-prolia.pdf
  31. Denosumab Biosimilar Antibody - BiCell Scientific®, accessed July 14, 2025, https://bicellscientific.com/product/denosumab-biosimilar-antibody/
  32. Denosumab 1080147A - Evidentic GmbH, accessed July 14, 2025, https://evidentic.com/products/denosumab-1080147/
  33. Denosumab - Blog - GlpBio, accessed July 14, 2025, https://www.glpbio.com/de/blog/post/denosumab.html
  34. Denosumab for the treatment of osteoporosis: A systematic literature review, accessed July 14, 2025, https://www.reumatologiaclinica.org/en-denosumab-for-treatment-osteoporosis-a-articulo-S1699258X12001787
  35. Denosumab in the treatment of bone metastases - Open Access Journals, accessed July 14, 2025, https://www.openaccessjournals.com/articles/denosumab-in-the-treatment-of-bone-metastases.pdf
  36. Full article: Pharmacokinetics and pharmacodynamics of the proposed biosimilar denosumab GP2411 and reference denosumab in healthy males - Taylor & Francis Online, accessed July 14, 2025, https://www.tandfonline.com/doi/full/10.1080/14712598.2024.2308645
  37. The mechanism of action of anti-RANK ligand (RANK-L) antibody on bone... - ResearchGate, accessed July 14, 2025, https://www.researchgate.net/figure/The-mechanism-of-action-of-anti-RANK-ligand-RANK-L-antibody-on-bone-turnover-Denosumab_fig1_230624748
  38. Denosumab (AMG-162) | RANKL Inhibitor - MedchemExpress.com, accessed July 14, 2025, https://www.medchemexpress.com/denosumab.html
  39. The RANKL pathway and denosumab - PubMed, accessed July 14, 2025, https://pubmed.ncbi.nlm.nih.gov/22023901/
  40. Pros and Cons of Denosumab Treatment for Osteoporosis and Implication for RANKL Aptamer Therapy - Frontiers, accessed July 14, 2025, https://www.frontiersin.org/journals/cell-and-developmental-biology/articles/10.3389/fcell.2020.00325/full
  41. Denosumab: mechanism of action and clinical outcomes - PubMed, accessed July 14, 2025, https://pubmed.ncbi.nlm.nih.gov/22967310/
  42. Efficacy and safety of denosumab compared to bisphosphonates in ..., accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC6658653/
  43. Prolia® - Denosumab | OncoLink, accessed July 14, 2025, https://www.oncolink.org/cancer-treatment/oncolink-rx/prolia-R-denosumab
  44. The pharmacokinetics and pharmacodynamics of denosumab in patients with advanced solid tumours and bone metastases: a systematic review - PMC - PubMed Central, accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC4243899/
  45. Denosumab: Uses, Dosage, Side Effects, Warnings - Drugs.com, accessed July 14, 2025, https://www.drugs.com/denosumab.html
  46. Denosumab Products : Denosumab (Xgeva®); Denosumab-bnht (Bomyntra®); Denosumab-bmwo (Osenvelt®); Denosumab-bbdz (Wyost®); Denosumab-dssb (Xbryk™), accessed July 14, 2025, https://www.bcbst.com/mpmanual/Denosumab_Xgeva.htm
  47. Comparison of denosumab and oral bisphosphonates for the treatment of glucocorticoid-induced osteoporosis: a systematic review and meta-analysis - PMC, accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC9706902/
  48. Xgeva (denosumab) - International Myeloma Foundation, accessed July 14, 2025, https://www.myeloma.org/xgeva-denosumab
  49. Study Summary: FREEDOM - American Journal of Managed Care, accessed July 14, 2025, https://www.ajmc.com/view/study-summary-freedom
  50. A Study to Evaluate Denosumab in the Treatment of Postmenopausal Osteoporosis, accessed July 14, 2025, https://clinicaltrials.gov/study/NCT00089791
  51. FDA Approves Amgen's Prolia(TM) (Denosumab) for Treatment of, accessed July 14, 2025, https://www.amgen.com/newsroom/press-releases/2010/06/fda-approves-amgens-proliatm-denosumab-for-treatment-of-postmenopausal-women-with-osteoporosis-at-high-risk-for-fracture
  52. Estimation of Long Term Efficacy of Denosumab Treatment in Postmenopausal Women With Osteoporosis: A FRAX and Virtual Twin Based Post Hoc Analysis From the FREEDOM and FREEDOM Extension Trials | JBMR Plus | Oxford Academic, accessed July 14, 2025, https://academic.oup.com/jbmrplus/article/4/4/e10348/7479028
  53. 10 years of denosumab treatment in postmenopausal women with osteoporosis: results from the phase 3 randomised FREEDOM trial and open-label extension - PubMed, accessed July 14, 2025, https://pubmed.ncbi.nlm.nih.gov/28546097/
  54. Denosumab Shows Favorable Results in FREEDOM Extension | MDedge - The Hospitalist, accessed July 14, 2025, https://community.the-hospitalist.org/content/denosumab-shows-favorable-results-freedom-extension
  55. Prolia | European Medicines Agency (EMA), accessed July 14, 2025, https://www.ema.europa.eu/en/medicines/human/EPAR/prolia
  56. FDA Approves Prolia® (Denosumab) For Glucocorticoid-Induced Osteoporosis - Amgen, accessed July 14, 2025, https://www.amgen.com/newsroom/press-releases/2018/05/fda-approves-prolia-denosumab-for-glucocorticoidinduced-osteoporosis
  57. Prolia (denosumab) FDA Approval History - Drugs.com, accessed July 14, 2025, https://www.drugs.com/history/prolia.html
  58. Prolia Finder™, accessed July 14, 2025, https://www.proliafinder.com/
  59. Serious Bone Problem Prevention | XGEVA® (denosumab) Injection, accessed July 14, 2025, https://www.xgeva.com/
  60. FDA Approves Amgen's XGEVA(TM) (Denosumab) for the Prevention of Skeletal-Related Events in Patients with Bone Metastases from Solid Tumors, accessed July 14, 2025, https://investors.amgen.com/news-releases/news-release-details/fda-approves-amgens-xgevatm-denosumab-prevention-skeletal/
  61. Bone-Related Complications and Quality of Life in Advanced Breast Cancer: Results from a Randomized Phase III Trial of Denosumab versus Zoledronic Acid - AACR Journals, accessed July 14, 2025, https://aacrjournals.org/clincancerres/article/18/17/4841/283648/Bone-Related-Complications-and-Quality-of-Life-in
  62. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study - PubMed, accessed July 14, 2025, https://pubmed.ncbi.nlm.nih.gov/21060033/
  63. Xgeva (denosumab) FDA Approval History - Drugs.com, accessed July 14, 2025, https://www.drugs.com/history/xgeva.html
  64. FDA Approves XGEVA® (denosumab) For The Prevention Of Skeletal-Related Events In Patients With Multiple Myeloma - Amgen, accessed July 14, 2025, https://www.amgen.com/newsroom/press-releases/2018/01/fda-approves-xgeva-denosumab-for-the-prevention-of-skeletalrelated-events-in-patients-with-multiple-myeloma
  65. Denosumab - NCI, accessed July 14, 2025, https://www.cancer.gov/about-cancer/treatment/drugs/denosumab
  66. FDA Approves Denosumab to Treat Giant Cell Tumor of the Bone - The ASCO Post, accessed July 14, 2025, https://ascopost.com/News/5233
  67. Prolia Injection (Denosumab): Uses & Side Effects - Cleveland Clinic, accessed July 14, 2025, https://my.clevelandclinic.org/health/drugs/18334-denosumab-injection
  68. FDA Approves New Indication for Denosumab (Xgeva) - Medscape, accessed July 14, 2025, https://www.medscape.com/viewarticle/836252
  69. Prolia - This label may not be the latest approved by FDA. For current labeling information, please visit https://www.fda.gov/drugsatfda, accessed July 14, 2025, https://www.accessdata.fda.gov/drugsatfda_docs/label/2024/125320s216lbl.pdf
  70. Prolia, Xgeva (denosumab) dosing, indications, interactions, adverse effects, and more, accessed July 14, 2025, https://reference.medscape.com/drug/xgeva-prolia-denosumab-999566
  71. FDA Approves Amgen's XGEVA™ (Denosumab) for the Prevention of Skeletal-Related Events in Patients with Bone Metastases from Solid Tumors - PR Newswire, accessed July 14, 2025, https://www.prnewswire.com/news-releases/fda-approves-amgens-xgeva-denosumab-for-the-prevention-of-skeletal-related-events-in-patients-with-bone-metastases-from-solid-tumors-109065144.html
  72. FDA Approves First Denosumab Biosimilars, accessed July 14, 2025, https://www.centerforbiosimilars.com/view/fda-approves-first-denosumab-biosimilar
  73. Pharmacokinetics, Pharmacodynamics, Safety and Immunogenicity of CMAB807, a New Denosumab Biosimilar, in Healthy Chinese Subjects - Frontiers, accessed July 14, 2025, https://www.frontiersin.org/journals/pharmacology/articles/10.3389/fphar.2022.821944/full
  74. Comparison of the efficacy and safety of denosumab versus ..., accessed July 14, 2025, https://www.spandidos-publications.com/10.3892/ol.2014.1982
  75. Osteoporosis treatment preferences and satisfaction in postmenopausal women: Denosumab compared with oral bisphosphonates - Australian Journal of Advanced Nursing, accessed July 14, 2025, https://www.ajan.com.au/archive/Vol35/Issue3/5Bajger.pdf
  76. Comparative effectiveness and safety outcomes between denosumab and bisphosphonate in South Korea | Journal of Bone and Mineral Research | Oxford Academic, accessed July 14, 2025, https://academic.oup.com/jbmr/article/39/7/835/7667804
  77. Denosumab Completed Phase 4 Trials for Osteoporosis Treatment | DrugBank Online, accessed July 14, 2025, https://go.drugbank.com/drugs/DB06643/clinical_trials?conditions=DBCOND0015928&phase=4&purpose=treatment&status=completed
  78. Denosumab for the treatment of bone metastases in breast cancer: evidence and opinion, accessed July 14, 2025, https://pmc.ncbi.nlm.nih.gov/articles/PMC3169928/
  79. Rediscovering denosumab: from osteoporosis treatment to a possible role against breast cancer - Centro Nacional de Investigaciones Oncológicas - CNIO, accessed July 14, 2025, https://www.cnio.es/en/news/rediscovering-denosumab-from-osteoporosis-treatment-to-a-possible-role-against-breast-cancer/
  80. Denosumab - Drug Targets, Indications, Patents - Patsnap Synapse, accessed July 14, 2025, https://synapse.patsnap.com/drug/2dde5d0ddf4d49a2b0df4c4050b91d95

Published at: July 14, 2025

This report is continuously updated as new research emerges.

MedPath

Empowering clinical research with data-driven insights and AI-powered tools.

© 2025 MedPath, Inc. All rights reserved.