Denosumab is a fully human IgG2 monoclonal antibody that specifically binds to receptor activator of nuclear factor kappa-B ligand (RANKL), a key mediator of osteoclastdifferentiation, activation, and survival.[1] By inhibiting the RANKL-RANK interaction, denosumab potently suppresses osteoclast-mediated bone resorption, thereby increasing bone mineral density and reducing fracture risk.[2] Developed by Amgen Inc., it was first approved by the U.S. Food and Drug Administration (FDA) in 2010 and is administered via subcutaneous injection.[1]Denosumab is marketed under two brand names with distinct indications and dosing regimens. As Prolia (60 mg every 6 months), it is indicated for the treatment of postmenopausal osteoporosis in women at high risk for fracture, osteoporosis in men at high risk for fracture, glucocorticoid-induced osteoporosis in men and women at high risk for fracture, treatment to increase bone mass in men receiving androgen deprivation therapy for nonmetastatic prostate cancer, and treatment to increase bone mass in women receiving adjuvant aromatase inhibitor therapy for breast cancer.[1] As Xgeva (120 mg every 4 weeks), it is approved for the prevention of skeletal-related events in patients with multiple myeloma and bone metastases from solid tumors, treatment of adults and skeletally mature adolescents with unresectable giant cell tumor of bone or those for whom surgical resection would cause severe morbidity, and treatment of hypercalcemia of malignancy refractory to bisphosphonate therapy.[3] These formulations share the same active ingredient but differ in concentration and scheduling to address osteoporosis versus oncology-related bone pathology.[2]Clinical use of denosumab requires careful monitoring due to potential adverse effects, including severe hypocalcemia (particularly in patients with chronic kidney disease), osteonecrosis of the jaw, atypical subtrochanteric and diaphyseal femoral fractures, and increased risk of multiple vertebral fractures upon discontinuation.[1] Patients must receive adequate calcium and vitamin D supplementation, and dental evaluations are recommended prior to initiation to mitigate jaw-related risks.[3] With a bioavailability of approximately 61% and a half-life of about 32 days, denosumab provides sustained inhibition of bone turnover.[2] Biosimilars, such as Jubbonti and Wyost (denosumab-bbdz) approved by the FDA in 2024, and additional ones including Enoby/Xtrenbo (denosumab-qbde), Osvyrti/Jubereq (denosumab-desu), Conexxence/Bomyntra, and Stoboclo/Osenvelt (denosumab-bmwo) approved in 2025, are interchangeable alternatives for both Prolia and Xgeva indications.[4][5]
Medical uses
Osteoporosis
Denosumab, marketed as Prolia, is approved for the treatment of osteoporosis in postmenopausal women at high risk for fracture, defined as those with a history of osteoporotic fracture or multiple risk factors for fracture.[1] It is also indicated to increase bone mass in men with osteoporosis at high risk for fracture.[1] Additionally, denosumab is approved for the treatment of glucocorticoid-induced osteoporosis in men and women at high risk for fracture receiving systemic glucocorticoid therapy (daily dosage equivalent to 7.5 mg or greater of prednisone) for chronic conditions expected to last at least 6 months.[1] It is further indicated to increase bone mass in men at high risk for fracture receiving androgen deprivation therapy for nonmetastatic prostate cancer, and to increase bone mass in women at high risk for fracture receiving adjuvant aromatase inhibitor therapy for breast cancer.[1]The recommended dosing regimen for denosumab in osteoporosis is 60 mg administered as a subcutaneous injection every 6 months.[1] This formulation is specifically designed for osteoporosis management and differs from the higher-dose regimen used in other indications. Denosumab is indicated for individuals at high risk for fracture and may be particularly useful in those intolerant to other osteoporosis therapies or with contraindications, such as renal impairment precluding bisphosphonate use.[2]Efficacy in reducing fracture risk was demonstrated in the pivotal FREEDOM trial, a 3-year randomized, placebo-controlled study involving postmenopausal women with osteoporosis, where denosumab reduced the risk of new vertebral fractures by 68% (2.3% vs. 7.2% incidence; relative risk, 0.32; 95% CI, 0.26-0.41), hip fractures by 40% (0.7% vs. 1.2% incidence; hazard ratio, 0.60; 95% CI, 0.37-0.97), and nonvertebral fractures by 20% (6.5% vs. 8.0% incidence; hazard ratio, 0.80; 95% CI, 0.67-0.95).[6] Long-term data from the 10-year FREEDOM extension trial showed sustained efficacy, with continued increases in bone mineral density (BMD) reaching 21.7% at the lumbar spine and 9.2% at the total hip compared to baseline, alongside persistently low fracture rates.[7] By inhibiting RANKL, denosumab reduces osteoclast activity and bone resorption, thereby enhancing BMD and fracture protection in these populations.[1]
Cancer-related bone conditions
Denosumab, marketed as Xgeva, is approved for the prevention of skeletal-related events (SREs) in patients with bone metastases from solid tumors and in those with multiple myeloma.[3] It is also indicated for the treatment of hypercalcemia of malignancy that is refractory to bisphosphonate therapy.[8] SREs include pathologic fractures, radiation or surgery to bone, and spinal cord compression, which significantly impact quality of life in advanced cancer patients. In clinical practice, denosumab targets the RANKL pathway to inhibit osteoclast activity, thereby reducing bone resorption and the incidence of these events in oncology settings.Phase III randomized controlled trials have demonstrated denosumab's superiority over zoledronic acid in preventing SREs across various solid tumors. In a pooled analysis of three trials involving patients with bone metastases from breast cancer, prostate cancer, and other solid tumors, denosumab reduced the risk of first on-study SRE by 17% (hazard ratio 0.83; 95% CI 0.76-0.90) and delayed the median time to first SRE by 8.21 months compared to zoledronic acid.[9] For instance, in breast cancer patients, the median time to first SRE was 32.4 months with denosumab versus 27.4 months with zoledronic acid.[10] Similar benefits were observed in prostate cancer, where denosumab delayed time to first SRE (median 20.7 months versus 17.1 months) and reduced multiple SREs by 18%. These outcomes highlight denosumab's role in extending the interval between SREs, particularly in patients undergoing androgen deprivation therapy for prostate cancer, where bone loss is exacerbated.[11]The recommended dosing for SRE prevention is 120 mg administered as a subcutaneous injection every 4 weeks, typically in the upper arm, upper thigh, or abdomen.[3] For hypercalcemia of malignancy, the regimen includes 120 mg every 4 weeks, with additional 120 mg doses on days 8 and 15 of the first month.[12] All patients should receive calcium and vitamin D supplementation as necessary to mitigate hypocalcemia risk, alongside monitoring of serum calcium levels.[13] Denosumab's subcutaneous administration offers convenience over intravenous bisphosphonates, with demonstrated efficacy in breast, prostate, and other solid tumors, including a 23% reduction in subsequent SREs in breast cancer cohorts.[9]
Giant cell tumor of bone
Denosumab is approved for the treatment of adults and skeletally mature adolescents with unresectable giant cell tumor of bone (GCTB) or when surgical resection would likely result in severe morbidity.[14] This indication targets the locally aggressive, benign nature of GCTB, where denosumab inhibits RANKL to suppress osteoclast-like giant cell activity essential for tumor-driven bone destruction.[15]In a pivotal open-label phase 2 study involving 282 patients with advanced GCTB, denosumab demonstrated efficacy with 74% of patients achieving stable disease or better after 6 months of treatment, and an objective response rate of 25% (all partial responses) by modified RECIST criteria.[16][14] Histopathologic analyses from biopsies showed near-complete elimination of osteoclast-like giant cells in 100% of evaluable patients, accompanied by reduced proliferative stromal cells and increased fibro-osseous matrix formation, confirming the drug's impact on tumor biology.[15] These responses often allowed deferral of morbid surgery, with only 30% of patients undergoing procedures during the study period.[16]The recommended dosing regimen is 120 mg administered subcutaneously every 4 weeks, with additional loading doses of 120 mg on days 8 and 15 during the first month of therapy.[14] Patients require radiographic evaluations, such as CT or MRI, every 3 months to assess tumor response and determine if surgical intervention has become feasible.[14] Treatment continuation is advised as long as clinical benefit persists without disease progression.[14]
Pharmacology
Mechanism of action
Denosumab is a fully human monoclonal immunoglobulin G2 (IgG2) antibody that specifically binds to receptor activator of nuclear factor kappa-B ligand (RANKL) with high affinity, characterized by a dissociation constant (Kd) of approximately 3 × 10^{-12} M.[17] This binding prevents RANKL from interacting with its receptor RANK on the surface of osteoclast precursors and mature osteoclasts.[18] By blocking this key signaling pathway, denosumab inhibits the differentiation, maturation, activation, and survival of osteoclasts, the primary cells responsible for bone resorption.[2]The suppression of osteoclast activity leads to a rapid and profound decrease in bone resorption, as evidenced by reductions in serum markers such as C-terminal telopeptide of type I collagen (CTX) by approximately 85% within three days and up to 88% by one month, and urinary N-terminal telopeptide (NTX) by over 80% within one week.[18][19] These effects manifest within weeks of administration, reflecting potent target engagement and downstream inhibition of bone breakdown processes.[1]Physiologically, denosumab reduces overall bone turnover by markedly suppressing resorption while having minimal direct impact on bone formation, resulting in a net increase in bonemass through secondary mineralization and coupling effects.[20] Unlike anabolic agents, it exerts no direct stimulatory effect on osteoblasts or bone-forming pathways.[18]In contrast to bisphosphonates, which inhibit osteoclast function intracellularly by targeting farnesyl pyrophosphate synthase in the mevalonate pathway, denosumab acts extracellularly by neutralizing soluble and membrane-bound RANKL, offering a distinct mechanism that avoids intracellular accumulation.[21][22]
Pharmacokinetics
Denosumab is administered subcutaneously and exhibits a bioavailability of approximately 62% following this route. After a single 60 mg dose, the median time to maximum serum concentration (Tmax) is 10 days, with a range of 3 to 21 days, and the mean maximum concentration (Cmax) is 6.75 mcg/mL.[1][23]The volume of distribution for denosumab is approximately 5.2 L, reflecting its distribution primarily within the vascular compartment as a monoclonal antibody, with binding to receptor activator of nuclear factor kappa-B ligand (RANKL) occurring in both serum and bonetissue.[1][24]Denosumab undergoes catabolism via proteolytic degradation, similar to other immunoglobulin G antibodies, without involvement of hepatic cytochrome P450 enzymes. Its terminal half-life is 25 to 30 days, with serum concentrations declining over 4 to 5 months post-dose. Clearance is nonlinear and decreases at higher doses due to target-mediated drug disposition, resulting in approximately dose-proportional exposure at therapeutic doses above 60 mg, while pharmacokinetics are nonlinear at lower doses.[1][23]At the standard osteoporosis treatment regimen of 60 mg every 6 months, denosumab does not accumulate, and pharmacokinetics remain consistent with repeated dosing.[1]Pharmacokinetics of denosumab are similar across men, postmenopausal women, and patients with bone metastases from solid tumors, with no notable differences based on age (28 to 87 years), race, or body weight (36 to 140 kg). No dose adjustment is required for renal impairment, as the degree of impairment does not affect pharmacokinetics; hepatic impairment studies have not been conducted, but adjustment is not anticipated given the drug's elimination pathway.[1][24]
Contraindications and interactions
Contraindications
Denosumab is contraindicated in patients with pre-existing hypocalcemia that has not been corrected prior to initiating therapy, as the drug can exacerbate this condition and lead to severe symptomatic hypocalcemia, including a boxed warning for potentially fatal cases in patients with advanced chronic kidney disease (eGFR <30 mL/min/1.73 m²), particularly those on dialysis.[1][3] It is also contraindicated in individuals with a history of systemic hypersensitivity reactions to denosumab or any of its excipients, including cases of anaphylaxis, hypotension, dyspnea, throat tightness, facial and upper airway edema, lip swelling, rash, pruritus, and urticaria.[1][3] For the osteoporosis indication (Prolia formulation), denosumab is contraindicated during pregnancy due to the risk of fetal harm; women of reproductive potential should undergo pregnancy testing prior to initiation and use effective contraception during treatment and for at least 5 months after the last dose.[1]Relative contraindications include unhealed lesions in the mouth or planned invasive dental procedures, particularly in patients receiving the oncology formulation (Xgeva), due to the elevated risk of osteonecrosis of the jaw; a complete dental examination and appropriate preventive dentistry are recommended before starting treatment in such cases.[3] Severe renal impairment (creatinine clearance <30 mL/min or receiving dialysis) is a relative contraindication without close monitoring, as these patients face a higher risk of severe hypocalcemia following administration, though no dose adjustment is required.[1][3] A history of atypical femoral fractures may warrant caution, as denosumab has been associated with such events, and therapy should be evaluated carefully in at-risk individuals.[1]Prior to initiating denosumab, hypocalcemia must be corrected to levels above the lower limit of normal, with supplementation of calcium, vitamin D, and magnesium as needed; serum calcium should be monitored closely, especially in the first weeks of therapy.[1][3] For oncology patients, a dental evaluation is essential to assess and mitigate risks of osteonecrosis of the jaw.[3]Denosumab is not recommended in pediatric patients with open growth plates due to potential impairment of bone growth and abnormal growth plate histology observed in animal studies; it is not approved for pediatric use in the osteoporosis indication and is limited to skeletally mature adolescents (aged 12 years and older) for giant cell tumor of bone.[1][3]
Drug interactions
Denosumab exhibits minimal pharmacokinetic interactions with other medications due to its lack of involvement with cytochrome P450 (CYP450) enzymes. Clinical studies have shown no significant alterations in the pharmacokinetics of midazolam, a CYP3A4 substrate, when co-administered with denosumab in postmenopausal women with osteoporosis, indicating that denosumab does not affect the metabolism of drugs primarily cleared via CYP3A4 pathways.[1] Similarly, no clinically meaningful changes in etanercept pharmacokinetics were observed following a single subcutaneous dose of denosumab in postmenopausal women with rheumatoid arthritis and low bone mineral density, suggesting limited impact on other biologic agents.[1] In patients receiving anticancer therapies, such as chemotherapy or hormoneablation, or those with prior intravenous bisphosphonate exposure, denosumab systemic exposure and pharmacodynamic effects (e.g., reductions in urinary N-telopeptide/creatinine) remained consistent, with no evidence of interference.[3]Pharmacodynamic interactions with denosumab primarily involve additive effects on calcium homeostasis. Concomitant use with agents that lower serum calcium levels or other calcium-lowering drugs can exacerbate hypocalcemia risk due to enhanced suppression of bone resorption and increased urinary calcium excretion.[1][3]Loop diuretics, in particular, have been associated with a higher incidence of denosumab-induced hypocalcemia in patients with osteoporosis, as demonstrated in retrospective analyses.[25] Additionally, calcimimetics like cinacalcet, which lower parathyroid hormone and serum calcium, may worsen hypocalcemia when combined with denosumab, especially in patients with advanced chronic kidney disease (eGFR <30 mL/min/1.73 m²).[1][3]To mitigate hypocalcemia risks, co-administration of calcium (at least 1000 mg daily) and vitamin D (at least 400 IU daily) is mandatory for all patients receiving denosumab, with supplementation assessed and optimized prior to initiation.[1] Concomitant use with calcimimetics should be avoided if possible, or closely monitored in high-risk populations. In oncology settings, denosumab increases the risk of osteonecrosis of the jaw (ONJ) when used alongside bisphosphonates or antiangiogenic agents like bevacizumab, as these therapies compound vascular and bone remodeling disruptions; prior intravenous bisphosphonate use is a noted risk factor for ONJ development.[3][26]Monitoring recommendations emphasize serum calcium assessment before initiating denosumab and approximately two weeks after each dose in at-risk patients, such as those with renal impairment, receiving concomitant hypocalcemia-inducing therapies, or inadequate supplementation; more frequent monitoring (e.g., weekly initially) is advised for advanced chronic kidney disease patients.[1][3] Pre-existing hypocalcemia must be corrected prior to starting therapy to prevent severe complications.[1]
Adverse effects
Common adverse effects
Denosumab is generally well-tolerated, with common adverse effects primarily involving the musculoskeletal system. In clinical trials for osteoporosis treatment, such as the FREEDOM trial involving postmenopausal women, back pain was reported in approximately 35% of patients receiving denosumab, compared to a similar rate in the placebo group. Arthralgia occurred in about 20% of patients, and pain in the extremities in 13%. These events were typically mild to moderate and did not differ significantly from placebo in terms of overall incidence or severity.[1][6]General adverse effects are also frequent but comparable to placebo. Nasopharyngitis affected around 10% of patients, headache 8%, and fatigue 5% in osteoporosis trials. Dermatologic reactions, including eczema and rash, each occurred in approximately 3% of patients, with eczema showing a slight increase over placebo (3.0% vs. 1.7%).[1][6]In patients with cancer-related bone conditions treated with denosumab (Xgeva formulation), musculoskeletal events are more prevalent, affecting about 50% of individuals, often due to underlying disease progression alongside treatment. Common events include arthralgia (13%), back pain (11%), and pain in extremity (11%), which are similar to comparator therapies like zoledronic acid.[27][1]These common adverse effects are managed symptomatically with analgesics, rest, or topical treatments as appropriate, and rarely require discontinuation of denosumab. Hypocalcemia may present as a common laboratory abnormality, particularly in patients with risk factors, and is typically addressed with calcium and vitamin D supplementation.[1]
Serious adverse effects
Denosumab, a monoclonal antibody that inhibits RANKL, carries several serious adverse effects, primarily related to its potent suppression of bone resorption and immune modulation. These risks, though infrequent, can lead to significant morbidity and require careful monitoring and preventive measures.Hypocalcemia is a notable risk, occurring in approximately 2-10% of patients, with severe cases (serum calcium <1.7 mmol/L) reported in up to 1% of treated individuals, particularly those with renal impairment. The FDA issued a black box warning in January 2024 highlighting the increased risk of severe, symptomatic, or fatal hypocalcemia in patients with advanced chronic kidney disease (stage 4 or 5), where incidence can reach 41% compared to 2% with oral bisphosphonates. Premedication with calcium and vitamin D is essential, and serum calcium levels should be monitored closely, especially within two weeks of administration and in patients with creatinine clearance <30 mL/min.[28][29][30]Osteonecrosis of the jaw (ONJ) develops in 0.04-2% of patients treated for osteoporosis and up to 2% in those with cancer, with cumulative incidence rising to 11.6% after prolonged denosumab exposure in oncology settings. Risk factors include invasive dental procedures, poor oral hygiene, smoking, and concomitant corticosteroid use, which can exacerbate the condition by impairing vascular supply and bone healing. Preventive strategies involve a baseline dental evaluation prior to initiation and avoidance of elective oral surgery during therapy; if ONJ occurs, denosumab should be discontinued and conservative management pursued.[29][31][32]Atypical femoral fractures, typically subtrochanteric or diaphyseal, occur in less than 1% of patients, with an estimated incidence of 0.8 per 10,000 patient-years, and are linked to prolonged use exceeding three years due to oversuppression of bone remodeling. These fractures often present with prodromal thigh or groin pain, and bilateral involvement is common; patients should be evaluated promptly for such symptoms, and therapy reassessed if confirmed.[29][33][34]Serious infections arise in 4-8% of patients, potentially stemming from RANKL's role in immune cell function, including T-cell activation and dendritic cell survival. Common manifestations include cellulitis, urinary tract infections, and pneumonia, with serious infections, including skin infections, reported more frequently than in placebo groups (4.0% vs. 3.3%). Patients with compromised immunity should be monitored for signs of infection, and prompt treatment initiated to mitigate complications.[29][35][36]Discontinuation of denosumab leads to a rebound effect characterized by rapid bone turnover resurgence, resulting in accelerated bone loss and an elevated risk of multiple vertebral fractures (up to 15% within 18 months). This phenomenon, observed in 3-10% of cases post-cessation, underscores the need for sequential antiresorptive therapy, such as bisphosphonates, to bridge the gap and prevent fractures.[29][37][38]Other rare serious effects include dermatologic reactions such as Stevens-Johnson syndrome, with incidence below 0.1% based on postmarketing surveillance and case reports, often requiring immediate discontinuation. Denosumab is contraindicated in pregnancy due to teratogenic effects observed in animal studies, including increased fetal loss, stillbirths, and developmental abnormalities; women of reproductive potential must use effective contraception during treatment and for at least five months thereafter.[29][39][40]
Society and culture
Development and history
Denosumab, developed by Amgen as the first biologic targeting receptor activator of nuclear factor kappa-B ligand (RANKL), originated from research into the osteoprotegerin (OPG)-RANK-RANKL pathway in the early 1990s. Amgen scientists identified OPG through DNA sequencing of a fetal rat intestine cDNA library, revealing its role as a decoy receptor that inhibits osteoclast formation and activity.[41][42] Preclinical studies during the 1990s demonstrated that RANKL inhibition reduced osteoclast numbers and increased bone density in animal models, including transgenic mice overexpressing OPG, paving the way for antibody-based therapies.[41][43]The first human dose of denosumab (then AMG 162), a fully humanmonoclonal antibody generated using XenoMouse technology, was administered in 2001. Pivotal clinical development advanced through phase III trials, including the FREEDOM study published in 2009, which evaluated denosumab's efficacy in postmenopausal women with osteoporosis over three years.[6] In parallel, phase III oncology trials reported in 2010 demonstrated denosumab's superiority to zoledronic acid in delaying skeletal-related events (SREs) in patients with bone metastases from solid tumors, including breast cancer.[44]Regulatory milestones began with U.S. Food and Drug Administration (FDA) approval of denosumab as Prolia (60 mg every six months) on June 1, 2010, for postmenopausal osteoporosis at high fracture risk, followed by approval as Xgeva (120 mg every four weeks) on November 18, 2010, for SRE prevention in bone metastases.[45] In the European Union, the European Medicines Agency authorized Prolia on May 26, 2010, and Xgeva on July 13, 2011.[46] Expanded indications included FDA approval for giant cell tumor of bone in June 2013 and European approval for glucocorticoid-induced osteoporosis in June 2018.[47]Post-approval research included long-term extension studies of the FREEDOM trial, with data up to 10 years of continuous denosumab treatment presented in 2016, confirming sustained bone mineral density gains and low fracture rates.[48]Biosimilar development has been accelerated by patent expirations, including key U.S. protections ending February 19, 2025, and earlier European expirations in 2022 for most markets.[49]Amgen entered a collaboration with GlaxoSmithKline in July 2009 to co-commercialize denosumab for postmenopausal osteoporosis in Europe and select other regions, while retaining full rights in the U.S., Canada, and for oncology indications.[50] The partnership was terminated in 2014, with Amgen regaining full commercialization rights by 2015.[51]
Brand names and formulations
Denosumab is commercially available under the primary brand names Prolia and Xgeva, both developed by Amgen. Prolia is supplied as a sterile, preservative-free, clear, colorless to slightly yellow solution containing 60 mg of denosumab in 1 mL volume, packaged in a single-use prefilled syringe for subcutaneous administration, and is indicated for osteoporosis treatment. Xgeva is formulated as a similar sterile, preservative-free solution with 120 mg of denosumab in 1.7 mL (70 mg/mL), provided in a single-use vial for subcutaneous injection, targeting oncology-related bone conditions.[1][3]The formulations of both Prolia and Xgeva include the active ingredient denosumab, a human IgG2 monoclonal antibody, along with excipients such as 4.7% sorbitol (Prolia) or 4.6% sorbitol (Xgeva), acetate buffer (17 mM for Prolia, 18 mM for Xgeva), 0.01% polysorbate 20, water for injection, and sodium hydroxide to adjust pH to 5.2. These single-use presentations are designed for direct subcutaneous delivery without dilution, ensuring stability and ease of administration by healthcare providers.[1][3]Biosimilars to denosumab have entered the market, offering equivalent formulations to the reference products. In 2024, Sandoz received approval for Jubbonti (interchangeable with Prolia) and Wyost (interchangeable with Xgeva) in both the US and EU, featuring identical solution compositions, strengths, and delivery devices as the originators. Additional approvals followed in 2025, including Ospomyv and Stoboclo in the US for osteoporosis and oncology uses, respectively, and Obodence and Xbryk in the EU, all maintaining the same excipient profiles, pH, and single-use syringe or vial formats without differences from the reference formulations. By late 2025, over 10 biosimilars were approved in the US.[4][52][53][54][55][5]All denosumab products, including originators and biosimilars, require storage under refrigeration at 2°C to 8°C (36°F to 46°F) in their original carton to protect from light, with instructions to avoid freezing and vigorous shaking. Once removed from refrigeration, they can be kept at controlled room temperature up to 25°C (77°F) for a maximum of 14 days prior to use, after which any unused portion must be discarded.[1][3][52]Denosumab is accessible globally through these originator and biosimilar options, though regional availability varies depending on local regulatory authorizations and market entry timelines.[56][57]
Legal status
Denosumab, marketed as Prolia and Xgeva, received initial approval from the U.S. Food and Drug Administration (FDA) in 2010, with Prolia authorized on June 1 for osteoporosis treatment and Xgeva on November 18 for bone metastasis-related events.[58] In January 2024, the FDA added a black box warning to Prolia's labeling regarding the increased risk of severe hypocalcemia in patients with advanced chronic kidney disease.[59] The first denosumab biosimilars, Jubbonti and Wyost, were approved by the FDA in March 2024 as interchangeable products for all reference indications.[4] Additional biosimilars followed in 2025, including Stoboclo and Osenvelt in February, Enoby in September, and others such as Bildyos and Bosaya later that year, expanding options for osteoporosis and cancer-related bone conditions. By late 2025, over 10 were approved in the US.[56][5] In the United States, denosumab requires a prescription and is covered under Medicare Part B for physician-administered doses or Part D for self-administration, subject to medical necessity criteria.[60]In the European Union, the European Medicines Agency (EMA) authorized Prolia in May 2010 for postmenopausal osteoporosis and Xgeva on July 13, 2011, for skeletal-related events in cancer patients, with centralized marketing authorization valid across member states.[61] Biosimilars Jubbonti and Wyost received EMA approval in May 2024, followed by approvals in 2025 for products including Obodence, Xbryk, Jubereq, and Osvyrti, all referencing the originator for similar indications. By September 2025, the EMA had recommended over 20 denosumab biosimilars.[62][63] Reimbursement varies by national health systems, with centralized EMA approval facilitating but not guaranteeing uniform access.[49]Health Canada approved denosumab in 2011 for osteoporosis and bone metastasis indications, with biosimilars Jubbonti and Wyost authorized in February 2024.[64] Coverage is available under provincial public drug plans, typically requiring prior authorization for eligible patients.[65]Denosumab was approved in Japan in March 2013 for osteoporosis under the brand Praria and in Australia in 2011 by the Therapeutic Goods Administration for similar uses.[66] Biosimilars are emerging in Asia, including Henlius's HLX14, approved in the United States in September 2025 and advancing in regional markets.[67] In Australia, it is classified as a Schedule 4 prescription-only medicine.The originator's patent exclusivity for denosumab expired in major markets between 2024 and 2025, enabling over 10 biosimilars globally by late 2025, primarily targeting high-burden indications like osteoporosis and oncology.[49][68]