75
Views
3
CrossRef citations to date
0
Altmetric
Review

Impact of denosumab on bone mass in cancer patients

&
Pages 117-129 | Published online: 04 Jul 2013

Abstract

Cancer therapy-induced bone loss (CTIBL) is a form of secondary osteoporosis associated with systemic chemotherapy and hormonal ablation therapy. The monitoring and treatment of CTIBL is an important component of comprehensive cancer care, especially for patients with curable disease and long life expectancies. Whereas oral bisphosphonates remain the most commonly used therapeutic option for CTIBL, additional treatment options may be required for patients who do not respond adequately or are intolerant to bisphosphonates, have renal insufficiency, or are receiving treatment with nephrotoxic medications. For these patients, denosumab, a monoclonal antibody targeting the receptor activator of nuclear factor-κB ligand (RANKL), offers an effective and well-tolerated alternative. Several recent randomized trials have examined the use of denosumab as treatment for CTIBL associated with hormone ablation therapy for breast and prostate cancer. Recent data suggest a possible role for RANKL inhibitors in both chemoprevention and the prevention of cancer recurrence through direct effects on breast tissue and breast cancer stem cells. The outcomes of several international Phase III clinical trials currently underway will help clarify the role of denosumab in patients undergoing cancer therapy.

Introduction

Osteoporosis is widely recognized as a major health issue in the United States, and tremendous resources are spent screening, diagnosing, and treating this disease. According to the National Institutes of Health Consensus Development Panel on Osteoporosis, 10 million people in the United States have osteoporosis, and almost twice as many have decreased bone mass.Citation1 Osteoporosis is defined as a decline in bone density and bone quality resulting in an increased risk for fragility fractures.Citation1 Primary osteoporosis results from gradual bone loss with aging, whereas secondary osteoporosis is due to a variety of medical disorders, including chronic corticosteroid use, hypogonadism, alcoholism, and hyperthyroidism. Cancer therapy-induced bone loss (CTIBL) is a form of secondary osteoporosis associated with systemic chemotherapy used in the treatment of both hematologic malignancies and solid tumors and with hormone ablation therapy used in the treatment of breast and prostate cancer. The final common pathway for both primary and secondary osteoporosis is increased bone resorption in relation to new bone formation.

Diagnosis and screening for osteoporosis in men and women rely on the estimation of bone mineral density (BMD), most commonly achieved through the use of dual-energy X-ray absorptiometry (DXA).Citation1,Citation2 The World Health Organization (WHO) defines osteoporosis and osteopenia on the basis of the T-score (the number of standard deviations above or below the average BMD value for young healthy white women) and the Z-score (the number of standard deviations above or below the average BMD for age-and sex-matched controls). The WHO defines osteoporosis as a T-score at least 2.5 standard deviations below normal and osteopenia as a T-score between 1.0 and 2.5 standard deviations below normal.Citation3 These definitions were developed for estimating fracture risk in postmenopausal women and have since been extrapolated to the population at large.

The WHO-developed Fracture Risk Assessment Tool (FRAX) predicts the 10-year risk of a major fracture (defined as a fracture in the hip, clinical spine, proximal humerus, or forearm).Citation4 The FRAX tool is customizable for use in various countries and ethnic populations and is available online at http://www.shef.ac.uk/FRAX. The FRAX score is derived from the patient’s BMD or T-score at the femoral neck after adjusting for other fracture-related risk factors, including age, gender, body mass index, history of previous fracture in the patient or his/her parents, smoking status, alcohol consumption, and the presence of other secondary risks for osteoporosis. Although useful as a standardized estimate of risk, FRAX may underestimate the risk of fracture in the setting of CTIBL, particularly in breast cancer patients with bone loss associated with aromatase inhibitor (AI) therapy.Citation5

The National Osteoporosis Foundation recommends that all men and postmenopausal women >50 years of age, regardless of BMD, take supplemental calcium (1200 mg daily) and vitamin D3 (800–1000 international units daily).Citation6 This recommendation is somewhat controversial, however, as several retrospective studies have identified a potential increased risk for cardiovascular events in patients receiving calcium supplements.Citation7,Citation8

Lifestyle modifications, including increased physical activity and smoking and alcohol cessation, are also encouraged in the National Osteoporosis Foundation guidelines. Additional pharmacotherapeutic intervention is considered for patients with osteoporosis or osteopenia at the femoral neck or spine and a 10-year hip fracture probability ≥3% or a 10-year major osteoporosis-related fracture probability ≥20% based on the US-adapted FRAX evaluation.Citation6 Pharmacologic treatment options for osteoporosis in the general population include bisphosphonates, hormone replacement therapy, selective estrogen-receptor modulators (SERMs), strontium ranelate (not currently available in the United States), teriparatide, and denosumab. However, only bisphosphonates and denosumab are generally used in the treatment of CTIBL. Other pharmacologic treatment options for primary osteoporosis, including teriparatide and exogenous estrogens, are infrequently used in cancer patients because of perceived risks or associated toxicities. For example, teriparatide has been associated with an increased risk for osteosarcoma, particularly in patients who have received prior radiation therapy. Estrogen therapy is contraindicated in patients with hormone receptor-positive breast cancer and is associated with gynecomastia and thrombotic complications in men.

This manuscript focuses on the treatment of CTIBL, particularly that associated with hormone ablation therapy for the treatment of breast and prostate cancer, with denosumab, a monoclonal antibody targeting the receptor activator of nuclear factor-κB (RANK) ligand (RANKL).

Role of estrogen and testosterone in bone turnover

Normal bone metabolism is characterized by continual osteoclast-mediated bone resorption balanced by osteoblast-mediated new-bone formation, a process predominantly regulated by the RANK–RANKL system.Citation9 RANKL is produced by osteoblasts, bone marrow stromal cells, and other cells under the control of various proresorptive growth factors, hormones, and cytokines. Osteoblasts and stromal cells also produce osteoprotegerin (OPG), a decoy receptor, which binds to and thereby prevents RANKL from activating its receptor, RANK. In the absence of OPG, RANKL activates RANK expressed on osteoclasts and preosteoclast precursors, promoting preosteoclast recruitment, fusion into multinucleated osteoclasts, osteoclast activation, and osteoclast survival. Activated osteoclasts secrete acids and other factors that break down the bone matrix, releasing calcium and phosphate into circulation. Thus, the bone serves as the body’s reservoir for calcium homeostasis.

Both estrogen receptors and androgen receptors are present in chondrocytes, bone marrow stromal cells, osteoblasts, osteoclasts, and osteoclast progenitors.Citation10 Estrogen deficiency is a major cause of postmenopausal bone loss in women and of age-related bone loss in both sexes. Estrogen deficiency results primarily in the loss of cortical bone, which comprises more than 80% of the skeleton, and is the major contributor to overall fracture risk.Citation11 Estrogen, acting via the estrogen receptor, stimulates osteoclast apoptosis and, conversely, suppresses osteoblast and osteocyte apoptosis.Citation12 Cell-culture studies demonstrate that estrogen can also increase OPG production and in some cases suppress RANKL expression.Citation9 Estrogen deficiency is also associated with an increase in the levels of circulating inflammatory cytokines, including tumor necrosis factor-α and interleukin-1α, which are potent stimulators of RANKL expression.Citation12 Thus, the cumulative effect of a low-estrogen state is to increase the RANKL:OPG ratio, which results in increased osteoclast activation and survival and unbalanced bone resorption.

The role of androgens in bone homeostasis is less well defined. Testosterone is peripherally converted via the aromatase enzyme into estrogens essential for bone health in both men and postmenopausal women.Citation12,Citation13 Thus, decreased BMD in men undergoing androgen-deprivation therapy (ADT) is largely secondary to the resulting estrogen deficiency. Testosterone may also have direct antiresorptive effects and is important for the maintenance of bone formation.Citation13 By increasing cortical bone thickness and preserving muscle strength, testosterone may also decrease the risk of falls and the likelihood of posttrauma fracture.Citation13,Citation14

Bone loss with hormone ablation in cancer treatment

Breast cancer

AIs are currently the most commonly prescribed adjuvant endocrine therapy for postmenopausal women with estrogen receptor-positive breast cancer. AIs inhibit aromatase, the cytochrome P450 CYP-19 enzyme responsible for the peripheral conversion of androgens to estrogens. Commercially available options include the nonsteroidal AIs letrozole and anastrozole and the steroidal AI exemestane. Other options for adjuvant endocrine therapy in both premenopausal and postmenopausal women with breast cancer are SERMs, including tamoxifen, and in premenopausal women are oophorectomy and lutenizing hormone-releasing hormone (LHRH) agonists, including goserelin and leuprolide. AIs, LHRH agonists, and oophorectomy are associated with bone density loss, whereas SERMs increase BMD in postmenopausal women due to their proestrogenic effects on bone. Several clinical trials evaluating the use of AIs in the adjuvant setting have demonstrated a decrease in BMD with an associated increase in fractures. As an example, a subset analysis was performed in the 5-year Arimidex, Tamoxifen, Alone or in Combination (ATAC) trial in 108 evaluable postmenopausal women with breast cancer randomly assigned to anastrozole versus tamoxifen.Citation15 Lumbar spine and total hip BMD were assessed at baseline and after 1 year, 2 years, and 5 years. Women assigned to anastrozole had a greater decrease in median BMD from baseline to year 5 at the lumbar spine (6% loss) and total hip (7.2% loss), compared with women assigned to tamoxifen treatment. In this trial, women treated with tamoxifen demonstrated an increase in BMD at the lumbar spine (2.8% gain) and total hip (0.74% gain).Citation15 When the entire trial population of 6241 women was considered, significantly more fractures were observed in patients randomized to anastrozole than in patients randomized to tamoxifen (451 fractures with anastrozole versus 351 with tamoxifen; odds ratio [OR] = 1.33; 95% confidence interval [CI] = 1.15–1.55; P < 0.0001). However, after treatment completion, the incidence of fractures equalized between the two groups (110 anastrozole versus 112 tamoxifen; OR = 0.98; 95% CI = 0.74–1.30; P = 0.9).Citation16

Many other trials have confirmed these results. A meta-analysis of seven randomized trials comprising approximately 30,000 postmenopausal women with early-stage breast cancer found that the use of AIs significantly increased the risk of fractures (OR = 1.47; 95% CI = 1.34–1.61) compared with tamoxifen.Citation17 Guidelines from the American Society of Clinical Oncology, the National Comprehensive Cancer Network (NCCN), and the European Society for Medical Oncology recommend BMD testing by DXA for all postmenopausal women taking AIs.Citation18Citation20

Prostate cancer

The prolonged use of LHRH agonists in both men and women is associated with a decline in BMD. Loss of BMD can be detected in the first year of ADT in men with prostate cancer, with longer durations of therapy conferring higher risk.Citation21,Citation22 In a retrospective analysis of more than 50,000 men with prostate cancer in the Surveillance, Epidemiology, and End Results database, 19.4% of men treated with ADT and surviving at least 5 years after diagnosis had a documented fracture, compared with only 12.6% of those not receiving ADT (P < 0.001).Citation23 A statistically significant association was found between the number of doses of gonadotropin-releasing hormone received during the 12 months after diagnosis and the subsequent risk of fracture.Citation23 NCCN guidelines currently recommend obtaining a baseline DXA scan prior to initiating ADT in men at increased risk of fracture on the basis of their FRAX score.Citation24 A follow-up DXA scan after 1 year of therapy is suggested in higher-risk patients; however, there is currently no consensus on the optimal monitoring schedule.

CTIBL in other settings

While men and women undergoing hormone ablation therapy are the most studied group of patients with CTIBL, other cancer patients are also at risk. For example, a population-based cohort study of older adult patients with non-Hodgkin lymphoma treated with chemotherapy demonstrated a significantly higher risk of osteoporosis and fractures.Citation25 The observed decrease in BMD in patients undergoing treatment with cytotoxic chemotherapy is multifactorial. Hypogonadism resulting from exposure to chemotherapeutic agents is the dominant mechanism in younger patients.Citation26 In a small study, 35 breast cancer patients with ovarian failure after adjuvant chemotherapy had significant decreases in BMD, most pronounced in the lumbar spine, at 6 months and 12 months, compared with women with preserved ovarian function.Citation27

Cytotoxic chemotherapy agents, including methotrexate, cyclophosphamide, and doxorubicin, also have been shown to have direct inhibitory effects on bone formation in animal models.Citation26,Citation28,Citation29 Ifosfamide and platinum compounds are thought to impact BMD at least in part by inducing hypophosphatemia and hypomagnesemia, respectively.Citation26,Citation30,Citation31 Much of this data come from studies in children treated for acute lymphoblastic leukemia; however, there are multiple other factors contributing to osteoporosis in these patients, including failure to reach a normal peak bone mass in early adulthood, prolonged chronic illness, prolonged corticosteroid use, cranial irradiation with damage to pituitary function, and vitamin D deficiency.Citation32

Glucocorticoids are used commonly in the treatment of hematologic malignancies and in the prevention of nausea and hypersensitivity reactions associated with chemotherapy for solid tumors. Glucocorticoids contribute to bone loss via several mechanisms, including increasing the RANKL:OPG ratio, impacting calcium balance, directly decreasing bone formation via glucocorticoid-induced apoptosis of both osteoblasts and osteocytes, and prolonging the life span of osteoclasts.Citation26,Citation33,Citation34

Patients undergoing hematopoietic stem cell transplantation (SCT) are uniquely prone to osteopenia or osteoporosis. Most bone loss in SCT patients occurs during the first 3–6 months after transplant and is most severe at the femoral neck.Citation35 These patients are typically treated with highdose cytotoxic chemotherapy, resulting in hypogonadism. They also receive prolonged systemic corticosteroids and other immunosuppressive agents, have lengthy periods of inactivity, and are frequently vitamin-D deficient; all of which contribute to excessive bone resorption.Citation26 SCT may also directly damage the marrow, resulting in an inability to regenerate normal numbers of osteoblastic precursors in the stromal stem cell compartment.Citation36

Treatment of CTIBL

Bisphosphonates

In addition to adequate calcium and vitamin D intake, bisphosphonates have been the mainstay of treatment for CTIBL. Although frequently prescribed, none of the bisphosphonates are specifically approved by the US Food and Drug Administration (FDA) for the treatment of CTIBL. However, data from several randomized trials have proven their effectiveness in the treatment of CTIBL in patients with breast and prostate cancer, and several bisphosphonates are Medicare compendia-approved for this indication.Citation37 Bisphosphonates are rapidly incorporated into bone and are released during osteoclast-mediated bone resorption, during which they impair the ability of the osteoclasts to adhere to the bony surface and inhibit further bone resorption. Bisphosphonates also decrease osteoclast progenitor development and recruitment and induce osteoclast apoptosis. Oral (PO) bisphosphonates available for the treatment of osteoporosis in the United States include alendronate, risedronate, and ibandronate, all of which have been shown in clinical trials to be effective at preventing bone loss, improving BMD, and reducing fracture risk. Intravenous (IV) bisphosphonates, including zoledronic acid, pamidronate, and ibandronate (not available in the United States), have also been shown to be effective for this indication.

The most common side effects associated with PO bisphosphonates are gastrointestinal and include reflux, esophagitis, and esophageal ulcers. Common side effects reported with IV bisphosphonates include flu-like symptoms, bone pains, and hypocalcemia. Bisphosphonates are also renally cleared and thus can cause nephrotoxicity. Consequently, they are contraindicated in patients with severe or moderate renal insufficiency (creatinine [Cr] clearance <30–35 mL/minute) and should be used with caution in patients receiving other nephrotoxic medications. Rare, but serious, side effects associated with both PO and IV bisphosphonates include osteonecrosis of the jaw (ONJ) and atypical femur fractures. The full side-effect profile associated with bisphosphonate therapy was recently reviewed by McClung et al.Citation38

Several trials have established a role for both PO and IV bisphosphonates in the treatment of bone loss associated with AI therapy in breast cancer.Citation39Citation43 Similarly, bisphosphonates have been shown to increase BMD in men receiving ADT for prostate cancer.Citation44Citation47 Of note, none of the breast and prostate cancer trials evaluating the use of bisphosphonates for the treatment and prevention of CTIBL have been powered to evaluate fracture rates as a primary outcome.

Denosumab

The pivotal role of the RANK–RANKL pathway in bone physiology renders it a prime target for the treatment of bone disease. The first commercially available drug to specifically target the RANK–RANKL pathway is denosumab, a fully human monoclonal antibody that binds and neutralizes RANKL, thereby inhibiting osteoclast function. The initial Phase I trials demonstrated that osteoclastic activity was almost completely abolished by denosumab therapy.Citation48 However, the effect is reversible, as indicated by a rise in markers of bone turnover when the drug is cleared by the reticuloendothelial system.Citation48 Denosumab has FDA approval under two brand names. Under the brand name Prolia® (Amgen Inc, Thousand Oaks, CA, USA), denosumab is indicated for the treatment of postmenopausal osteoporosis and CTIBL, at a dose of 60 mg subcutaneously (SC) every 6 months. Under the brand name XGEVA® (Amgen Inc), denosumab is FDA approved for the prevention of skeletal-related events (SREs) in patients with bone metastases from solid tumors, at a dose of 120 mg SC every 4 weeks.

Initial trials established the efficacy of denosumab in the treatment of postmenopausal women with primary osteoporosis. In the Phase III Fracture Reduction Evaluation of Denosumab in Osteoporosis Every 6 Months (FREEDOM) trial, 7868 postmenopausal women (60–90 years of age) with osteoporosis were randomly assigned to denosumab (60 mg SC every 6 months) or placebo.Citation49 At 3 years, the incidence of new vertebral fractures (the primary endpoint) was 2.3% in the denosumab arm versus 7.2% in the control arm, a relative-risk reduction of 68% (P < 0.001). The risk of nonvertebral and hip fractures was also significantly reduced in the treatment arm.

Denosumab was also evaluated in the setting of primary osteoporosis in a Phase III, double-blind, active-controlled trial comparing the efficacy and safety of denosumab (60 mg SC every 6 months) with alendronate (70 mg PO weekly) in 1189 postmenopausal women with a BMD T-score <−2.0.Citation50 At 12 months, the denosumab-treated patients had a statistically significant increase in BMD at the total hip (the primary endpoint) compared with the alendronate-treated patients (3.5% versus 2.6%, respectively; P < 0.0001).Citation50 Although this study was not powered to compare fracture rates, they were similar between the groups, with 18 fractures (3.0%) in the denosumab group compared with 13 (2.2%) in the alendronate group (P = 0.37).Citation50

summarizes the outcomes and fracture rates from the Phase III clinical trials evaluating denosumab in the treatment of osteoporosis, osteopenia, and CTIBL.

Table 1 Phase III trials of denosumab for low bone mineral density and cancer therapy-induced bone loss: outcomes

Denosumab and CTIBL in breast cancer

Denosumab has also been evaluated specifically in the treatment and prevention of CTIBL associated with adjuvant AI therapy in breast cancer. In a randomized, double-blind, placebo-controlled trial, 252 women with hormone receptor-positive early-stage breast cancer treated with adjuvant AI therapy were randomly assigned to receive placebo or denosumab (60 mg SC every 6 months).Citation51 At enrollment, all patients were required to have evidence of low bone mass, excluding osteoporosis (lumbar spine, total hip, or femoral neck BMD corresponding to a T-score of −1.0 to −2.5). Patients were also required to have 25-hydroxyvitamin D levels ≥12 ng/mL. Key exclusion criteria included current use of bisphosphonates and the use of antineoplastic therapy aside from AI. The primary endpoint was percentage change in the lumbar spine BMD at months 12 and 24, compared with baseline. At 12 and 24 months, lumbar spine BMD increased by 5.5% and 7.6%, respectively, in the denosumab group compared with the placebo group (P = 0.0001). This increase was observed irrespective of the duration of prior AI or tamoxifen therapy. As in previous trials, markers of bone metabolism were rapidly suppressed. At only 252 patients, the study was not powered to see differences in fracture rates; however, the incidence of nonvertebral fractures was 6% in both the denosumab and placebo arms (). A randomized Phase III clinical trial is currently underway to determine whether, compared with placebo, denosumab will reduce the rate of first clinical fracture in women with nonmetastatic breast cancer receiving AI therapy.Citation52

The overall incidence of grade 3 or higher adverse events (AEs) was similar between treatment groups and included AEs commonly associated with AI therapy, including arthralgia, pain in the extremities, back pain, and fatigue ().Citation51 Significant hypocalcemia was not reported. Two deaths (one in each group) were reported, both attributed to breast cancer progression. In a subgroup analysis, an increase in BMD from baseline was seen with denosumab compared with placebo across all subgroups and skeletal sites. This increase was statistically significant, with two exceptions: (1) radial BMD in patients who received prior steroidal AI therapy; and (2) femoral-neck BMD in patients aged ≥65 years at baseline.Citation53

Table 2 Adverse events from denosumab trials

Denosumab at 60 mg SC every 6 months is currently the only FDA-approved antiresorptive therapy for the treatment of AI-induced bone loss in women with early-stage breast cancer. However, several guidelines, including those from the American Society of Clinical Oncology and the NCCN, recommend either a bisphosphonate or denosumab as appropriate therapy for those with documented osteoporosis (T-score ≤−2.5) or significant osteopenia (T-score ≤−2.0) and an increased fracture risk.Citation18,Citation19 The European Society for Medical Oncology recognizes that bisphosphonates prevent bone loss in patients with iatrogenic premature menopause and in postmenopausal patients treated with AIs, but the organization does not make specific recommendations about treatment.Citation20

Denosumab and CTIBL in prostate cancer

In the denosumab Hormone Ablation Bone Loss Trial (HALT), 1468 men receiving ADT (bilateral orchiectomy or gonadotropin-releasing hormone agonist) for nonmetastatic prostate cancer were randomly assigned to denosumab (60 mg SC every 6 months) or placebo.Citation54 The primary endpoint was change in BMD at the lumbar spine at 24 months. Eligibility included men ≥70 years of age, or men <70 years of age with baseline osteopenia (T-score at the lumbar spine, total hip, or femoral neck of <−1.0). Key exclusion criteria included concurrent chemotherapy or radiation, prostate-specific antigen (PSA) >5 ng/mL after 1 month of ADT, current or long-term prior bisphosphonate use, and a BMD T-score <−4.0. At 24 months, denosumab was associated with increased BMD at all measured sites, including the lumbar spine, total hip, femoral neck, distal third of the radius, and whole body (absolute difference versus placebo, 6.7%, 4.8%, 3.9%, 5.5%, and 4.0%, respectively; P < 0.001 for all comparisons). A significant increase in BMD was observed in all patient subgroups. With a sample size of almost 1500 patients, this study was powered to capture a difference in fracture rate, and a statistically significant decrease in new vertebral fractures was observed at 36 months in the denosumab arm (1.5% versus 3.9% with placebo, relative risk = 0.38; 95% CI = 0.19–0.78; P = 0.006). Fractures at any site also occurred in fewer denosumab-treated patients compared with the placebo group (38 patients [5.2%] versus 53 patients [7.2%]); however, this did not reach statistical significance (P = 0.10). In addition, denosumab significantly decreased the number of patients developing more than one fracture (denosumab, five patients [0.7%] versus placebo, 18 patients [2.5%], P = 0.006). AE rates were similar between the groups, with no cases of ONJ reported in either arm ().

The NCCN recommends that men undergoing ADT be treated according to the National Osteoporosis Foundation guidelines as detailed above, with treatment recommended in patients with osteoporosis or osteopenia at high risk for fracture as evaluated by their FRAX score.Citation6,Citation24 Recommended treatment options include denosumab (60 mg SC every 6 months), zoledronic acid (5 mg IV annually), or alendronate (70 mg PO weekly).Citation24

Denosumab dosing

Denosumab absorption is rapid and sustained, with a bioavailability of 62%–64%, a steady-state mean serum concentration of 20.5 μg/mL, and an elimination half-life of 28 days.Citation55,Citation56 A decrease in bone-resorption markers is observed within 24 hours after denosumab administration, and steady-state levels are achieved after 6 months of multiple doses at 120 mg per month.Citation48,Citation55 The initial Phase I trial in healthy postmenopausal women demonstrated that a single denosumab dose of 3.0 mg/kg could suppress markers of bone turnover, including urinary collagen type 1 cross-linked N-telopeptide (uNTx), up to 80% for several months.Citation48 A subsequent Phase II trial in patients with bone metastases from solid tumors with elevated uNTx:Cr levels despite IV bisphosphonate therapy compared denosumab (30–180 mg administered every 4 or 12 weeks) with continued IV bisphosphonate therapy.Citation57 In this study, 71% of patients who switched to denosumab treatment had a decline in uNTx:Cr ratio compared with 29% of patients who continued therapy with IV bisphosphonates (P < 0.001), providing initial evidence that denosumab may be superior to IV bisphosphonates at suppressing bone resorption.Citation57 A second Phase II trial in patients with metastatic breast cancer treated with multiple doses and schedules of denosumab confirmed a greater reduction in uNTx:Cr levels in denosumab-treated compared to IV bisphosphonate-treated patients (74% versus 63%).Citation58 In a recently published meta-analysis of multiple Phase I, Phase II, and Phase III trials evaluating the pharmacokinetics of denosumab in more than 1500 healthy subjects and postmenopausal women with low BMD, a fixed dose of denosumab at 60 mg (the recommended dose for CTIBL) provided similar RANKL inhibition to that achieved with weight-based dosing.Citation56 Importantly, patient age and race had little impact on denosumab’s pharmacokinetics. Denosumab has not been studied in the pediatric population.

Denosumab elimination occurs through the immunoglobulin clearance pathway via the reticuloendothelial system, similar to that of other monoclonal antibodies, and is thus thought to be independent of renal or hepatic function.Citation59

Dose reductions and renal monitoring are not required with denosumab therapy; however, there is a lack of safety data in patients with severe renal dysfunction. In the only published trial of denosumab use in renal impairment, a single 60 mg dose was given to 55 patients, including 17 patients having severe chronic renal disease (Cr clearance ≤30 mL/minute) or requiring hemodialysis. Pharmacokinetics and changes in biomarkers of bone resorption were unaffected by renal function.Citation60 Hypocalcemia was observed more frequently in patients with renal dysfunction with hypocalcemia of any grade in eight (15%) patients and severe hypocalcemia requiring hospitalization in two (4%) patients.Citation60 The two patients with severe hypocalcemia had advanced renal disease and were enrolled before the protocol required baseline assessment of parathyroid hormone, calcium, and vitamin D. Thus, more frequent monitoring of serum calcium levels and the ensuring of adequate vitamin D levels prior to initiating denosumab therapy is recommended in this population.

Denosumab side effects

Much of what is known about the side effects of denosumab comes from its use in the setting of primary osteoporosis and in patients with bone metastasis.

The FREEDOM trial is the largest single trial comparing denosumab to placebo for the prevention of fractures in primary osteoporosis.Citation49 In this study, there were no significant differences between the 3900 subjects who received denosumab and those who received placebo in the total incidence of AEs, serious adverse events (SAEs), or discontinuation of study treatment because of AEs.Citation49 In addition, there was no increase in the risk of cancer, cardiovascular disease, delayed fracture healing, or hypocalcemia, and there were no cases of osteonecrosis of the jaw. Cellulitis was more frequently observed in the denosumab group, although there was no difference in the overall rate of infections. Neutralizing antibodies against denosumab were not identified. As summarized in , a similar safety profile was observed in the trials using denosumab to treat CTIBL in the settings of breast and prostate cancer.

Three international Phase III randomized, doubleblind, double-dummy, active-controlled studies comparing denosumab (120 mg SC every 4 weeks) with zoledronic acid (4 mg IV every 4 weeks) for the prevention of SREs in more than 5700 patients with bone metastases have been completed.Citation61Citation63 These three registration trials were of identical design and focused on the prevention of SREs in patients with breast cancer, prostate cancer, and other solid tumors or multiple myeloma and lead to the FDA approval of denosumab for this indication. In a meta-analysis of these trials, the incidence of all AEs and SAEs was similar between treatment groups.Citation64 However, as in the individual trials, there was an increased incidence of hypocalcemia (9.6% versus 5.0%) and acute phase reactions (20.2% versus 8.7%) in the denosumab group compared with the zoledronic acid group, respectively. Incidence of ONJ was infrequent and similar between treatment groups, with a cumulative incidence of 1.3% (37 events) in the zoledronic acid group compared with 1.8% (52 events) in the denosumab group (P = 0.13).

Recently, there have been reports of atypical femur fractures in patients treated with denosumab.Citation55,Citation65 Atypical subtrochanteric and diaphyseal femur fractures, initially reported in patients using bisphosphonates long term (>5 years), have a characteristic prodrome of mid-thigh pain, which may be accompanied by cortical thickening on plain radiographs obtained prior to fracture. The rate of atypical femur fractures in patients treated with both bisphosphonates and denosumab is much lower than the fracture risk associated with untreated osteoporosis.

As RANKL is expressed on subsets of T and B cells, there is a theoretical possibility that denosumab may be immunosuppressive. RANKL-deficient mice lack normal lymph node development and have inhibition of early T- and B-lymphocyte development.Citation66 However, in clinical trials, a statistically significant or clinically meaningful effect on the immune system has not been observed to date. In one early trial of denosumab therapy, there was no significant effect on mean white blood cell counts, absolute lymphocyte counts, T-cell or B-cell counts, or immunoglobulins, nor was there a meaningful difference in incidence of infection.Citation48 As noted above, trials of denosumab for the treatment and prevention of osteoporosis suggested a slight increase in the rate of certain infectious complications, including cellulitis.Citation49,Citation67 However, the overall infection rate did not differ significantly from placebo, and an association between denosumab and infectious SAEs was not observed in the three large Phase III registrational trials comprising over 5700 patients with metastatic cancer.Citation64

Sequential use

The safety of switching from bisphosphonates to denosumab therapy has been studied in the treatment of patients with primary osteoporosis as well as in the setting of metastatic disease to prevent SREs, but not specifically in the setting of CTIBL.Citation68Citation70 The Phase III Study of Transitioning from Alendronate to Denosumab trial evaluated the sequential use of alendronate followed by denosumab in postmenopausal women with primary osteoporosis.Citation68 Eligible patients were postmenopausal women at least 55 years of age who had a lumbar spine or total hip BMD T-score of −2.0 to −4.0 and who had been receiving alendronate at 70 mg/week for at least 6 months. Subjects were randomized to denosumab (60 mg SC every 6 months) versus continued oral alendronate. The primary endpoint was change in total hip BMD after 12 months of therapy. BMD at the total hip increased significantly in patients transitioned to denosumab (1.90%; 95% CI = 1.61%–2.18%) compared with patients continuing on alendronate (1.05%; 95% CI = 0.76%–1.34%) (P < 0.0001). These trials suggest a role for switching to denosumab in patients who are currently receiving bisphosphonate therapy and who continue to have a decline in BMD on DXA or have a persistently elevated uNTx level. Other indications for switching to denosumab might include progressive renal insufficiency, intolerance to bisphosphonate side effects, or the necessity for concomitant use of nephrotoxic medications. No data support the combined use of a bisphosphonate plus denosumab to further reduce osteoclast activity. As both agents are extremely potent osteoclast inhibitors, the risk of increased toxicity with combination therapy, especially with regard to ONJ development and hypocalcemia, would be particularly concerning.

Future uses

The use of bone-modifying agents in the prevention of cancer, cancer recurrence, and/or cancer progression is currently an area of active research. Several trials in early-stage breast cancer patients suggest a role for bone-modifying agents in improving disease-free survival. The Austrian Breast and Colorectal Cancer Study Group Trial-12 in premenopausal women with early-stage breast cancer is the most quoted of the recent trials in support of a role for bone-targeted therapy in the prevention of breast cancer recurrence.Citation71 In this trial, patients treated with three years of 4 mg IV zoledronic acid twice yearly had a 36% lower incidence of breast cancer recurrence compared with patients treated with hormonal therapy alone. Unfortunately, the much larger Adjuvant Zoledronic Acid to Reduce Recurrence trial performed in higher-risk early-stage breast cancer patients with stage 2 and 3 disease treated predominantly with chemotherapy alone versus chemotherapy plus higher-intensity zoledronic acid failed to show a similar improvement in breast cancer recurrence.Citation72 However, in a subgroup analysis there was an improvement in both disease-free and overall survival in older women (>5 years postmenopausal or >60 years of age) treated with zoledronic acid. Several other trials have also recently been presented that offer additional conflicting results about the effectiveness of intravenous bisphosphonates in preventing breast cancer recurrence in patients with early-stage disease (reviewed in Wong et al).Citation73 One hypothesis for the results of the Austrian Breast and Colorectal Cancer Study Group Trial-12 and Adjuvant Zoledronic Acid to Reduce Recurrence trials is that bisphosphonates play a role in the prevention of breast cancer recurrence, but only in very-low-estrogen states.Citation71,Citation72 However, the exact mechanism or effect bisphosphonates play in this setting is still controversial.

The RANK–RANKL system is also implicated in mammary epithelial proliferation and carcinogenesis. In murine models, overexpression of RANK promotes hormone- and carcinogen-induced murine mammary tumor formation, whereas pharmacologic inhibition of RANKL attenuates tumor development.Citation74 This and other animal data suggest that there may be a role for RANKL inhibitors like denosumab in both chemoprevention and the prevention of cancer recurrence through direct effects on breast tissue and breast cancer stem cells.Citation74Citation76 Similarly, there is data supporting an integral role for RANK–RANKL in the spread and propagation of cancer cells in bone, helping to explain the osteotropism of tumors, including prostate, breast, and lung cancer, that express RANK.Citation77 Denosumab may have the potential to interrupt the “vicious cycle” of cancer-induced bone destruction and tumor cell expansion and to inhibit breast cancer stem cell survival.Citation78 The international Phase III Denosumab as Adjuvant Treatment for Women With High Risk Early Breast Cancer Receiving Neoadjuvant or Adjuvant Therapy (D-CARE) trial has recently completed accrual and is powered to assess the effect of denosumab on disease recurrence in patients with stage 2 and 3 high-risk early stage breast cancer.Citation79 The Austrian Breast and Colorectal Cancer Study Group Study 18 trial is currently enrolling patients and will similarly evaluate the effect of denosumab on the time to first clinical fracture and bone metastasis, disease-free survival, and overall survival.Citation52

The role of denosumab in delaying the development of bone metastasis has been demonstrated in the prostate cancer 147 trial.Citation80 In this Phase III double blind, placebo-controlled trial, 716 men with castrate-resistant prostate cancer at high risk for the development of bone metastases (PSA > 8.0 ug/L or PSA doubling time <10.0 months) were randomized to denosumab 120 mg SC or SC placebo every 4 weeks.81 The primary end point of bone metastasis-free survival time was 25.2 months in the placebo group compared with 29.5 months in the denosumab group (hazard ratio = 0.85; 95% CI = 0.73–0.98; P = 0.028), with those at greatest risk of metastases (PSA doubling time ≤6 months) having the greatest benefit. As overall survival was similar between the two groups, it is currently unclear if denosumab merely delayed rather than prevented bone metastases in this patient population. The FDA declined to approve denosumab for this indication.

The role of denosumab in preventing or delaying cancer development and progression awaits the results of the D-CARE and Austrian Breast and Colorectal Cancer Study Group Study 18 trials as well as further follow-up of the 147 prostate cancer trial.

Conclusion

Attention to bone health is an essential component of cancer care, particularly in women and men undergoing hormone ablation therapy for the treatment of breast and prostate cancer. Oral bisphosphonates remain the most commonly used therapeutic option in this setting, but additional treatment options are required for patients who do not respond adequately or are intolerant to bisphosphonates, have renal insufficiency, or require treatment with nephrotoxic medications. For these patients, denosumab offers an effective and well-tolerated alternative. In addition, bisphosphonates are retained in the bone for decades and the long-term implications, especially in younger patients, are unknown. As with bisphosphonates, data regarding the long-term toxicities of denosumab are lacking. However denosumab’s shorter half-life and reversibility are attractive when considering long-term use or therapy for younger patients. The choice between bisphosphonates and denosumab will become better informed as the role these agents play in cancer progression, recurrence, and prevention is established. The added cost of denosumab as a novel and newly approved biologic agent, particularly in comparison with the notably lower costs of generic bisphosphonates, must also be considered when recommending a therapy. The data presented highlight the essential role that bone-modifying agents play in maintaining bone health in patients undergoing cancer care and the emerging role of denosumab for this indication.

Disclosure

A Stopeck has consulted, received honoraria, and received research and clinical trial support from Amgen, received honoraria from GlaxoSmithKline, and received clinical trial support from Novartis. U Brown-Glaberman reports no conflict of interest in this work. The authors report no other conflicts of interest in this work.

References

  • NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and TherapyOsteoporosis prevention, diagnosis and therapyJAMA2001285678579511176917
  • LuiHPaigeNMGoldzweigCLScreening for osteoporosis in men: a systematic review for an American College of Physicians guidelineAnn Intern Med2008148968570118458282
  • Prevention and management of osteoporosisWorld Health Organ Tech Rep Ser2003921116415293701
  • World Health OrganizationFRAX® WHO fracture risk assessment tool: welcome to FRAX® [homepage on the Internet]Sheffield, United KingdomWorld Health Organization Collaborating Centre for Metabolic Bone Diseases2013 Available from: http://www.sheffield.ac.uk/FRAX/Accessed February 5, 2013
  • HadjiPAaproMSBodyJJManagement of aromatase inhibitor-associated bone loss in postmenopausal women with breast cancer: practical guidance for prevention and treatmentAnn Oncol201122122546255521415233
  • National Osteoporosis Foundation2013 clinician’s guide to the prevention and treatment of osteoporosis [webpage on the Internet]Washington, DCNational Osteoporosis Foundation2013 Available from: http://nof.org/hcp/resources/913Accessed February 10, 2013
  • BollandMJAvenellABaronJAEffect of calcium supplements on risk of myocardial infarction and cardiovascular events: meta-analysisBMJ2010341c369120671013
  • BollandMJGreyAAvenellAGambleGDReidIRCalcium supplements with or without vitamin D and risk of cardiovascular events: reanalysis of the Women’s Health Initiative limited access dataset and meta-analysisBMJ2011342d204021505219
  • KearnsAEKhoslaSKostenuikPJReceptor activator of nuclear factor kappaB ligand and osteoprotegerin regulation of bone remodeling in health and diseaseEndocr Rev200829215519218057140
  • ManolagasSCKousteniSJilkaRLSex steroids and boneRecent Prog Horm Res20025738540912017554
  • KhoslaSMeltonLJ3rdRiggsBLThe unitary model for estrogen deficiency and the pathogenesis of osteoporosis: is a revision needed?J Bone Miner Res201126344145120928874
  • KhoslaSUpdate on estrogens and the skeletonJ Clin Endocrinol Metab20109583569357720685883
  • KhoslaSAminSOrwollEOsteoporosis in menEndocr Rev200829444146418451258
  • LorentzonMSwansonCAnderssonNMellströmDOhlssonCFree testosterone is a positive, whereas free estradiol is a negative, predictor of cortical bone size in young Swedish men: the GOOD studyJ Bone Miner Res20052081334134116007330
  • EastellRAdamsJEColemanREEffect of anastrozole on bone mineral density: 5-year results from the anastrozole, tamoxifen, alone or in combination trial 18233230J Clin Oncol20082671051105718309940
  • ForbesJFCuzickJBuzdarAHowellATobiasJSBaumMArimidex, Tamoxifen, Alone or in Combination (ATAC) Trialists’ GroupEffect of anastrozole and tamoxifen as adjuvant treatment for early-stage breast cancer: 100-month analysis of the ATAC trialLancet Oncol200891455318083636
  • AmirESerugaBNiraulaSCarlssonLOcañaAToxicity of adjuvant endocrine therapy in postmenopausal breast cancer patients: a systematic review and meta-analysisJ Natl Cancer Inst2011103171299130921743022
  • HillnerBEIngleJNChlebowskiRTAmerican Society of Clinical OncologyAmerican Society of Clinical Oncology 2003 update on the role of bisphosphonates and bone health issues in women with breast cancerJ Clin Oncol200321214042405712963702
  • GralowJRBiermannJSFarookiANCCN Task Force report: bone health in cancer careJ Natl Compr Canc Netw20097Suppl 3S1S32 quiz S33–S3519555589
  • AebiSDavidsonTGruberGCardosoFEMSO Guidelines Working GroupPrimary breast cancer: ESMO Clinical Practice Guidelines for diagnosis, treatment and follow-upAnn Oncol201122Suppl 6vi12vi2421908498
  • SaadFAdachiJDBrownJPCancer treatment-induced bone loss in breast and prostate cancerJ Clin Oncol200826335465547618955443
  • KrupskiTLSmithMRLeeWCNatural history of bone complications in men with prostate carcinoma initiating androgen deprivation therapyCancer2004101354154915274067
  • ShahinianVBKuoYFFreemanJLGoodwinJSRisk of fracture after androgen deprivation for prostate cancerN Engl J Med2005352215416415647578
  • National Comprehensive Cancer NetworkNCCN Clinical Practice Guidelines in Oncology (NCCN Guideline®): Prostate Cancer. V.2.2013Fort Washington, PANational Comprehensive Cancer Network2013 Available from: http://www.nccn.org/professionals/physician_gls/pdf/prostate.pdfAccessed February 8, 2013
  • CabanillasMELuHFangSDuXLElderly patients with non-Hodgkin lymphoma who receive chemotherapy are at higher risk for osteoporosis and fracturesLeuk Lymphoma20074881514152117701582
  • HuMILuHGagelRFCancer therapies and bone healthCurr Rheumatol Rep201012317718520437119
  • ShapiroCLManolaJLeboffMOvarian failure after adjuvant chemotherapy is associated with rapid bone loss in women with early-stage breast cancerJ Clin Oncol200119143306331111454877
  • NilssonOSBauerHCBrostromLAComparison of the effects of adriamycin and methotrexate on orthotopic and induced heterotopic bone in ratsJ Orthop Res1990821992042303952
  • WangTMShihCStudy of histomorphometric changes of the mandibular condyles in neonatal and juvenile rats after administration of cyclophosphamideActa Anat (Basel)1986127293993788461
  • KötherMSchindlerJOetteKBertholdFAbnormalities in serum osteocalcin values in children receiving chemotherapy including ifosfamideIn Vivo1992622192211525342
  • Von HoffDDSchilskyRReichertCMToxic effects of cis-dichlorodiammineplatinum(II) in manCancer Treat Rep1979639–1015271531387223
  • WarnerJTEvansWDWebbDKBellWGregoryJWRelative osteopenia after treatment for acute lymphoblastic leukemiaPediatr Res1999454 Pt 154455110203147
  • JiaDO’BrienCAStewartSAManolagasSCWeinsteinRSGlucocorticoids act directly on osteoclasts to increase their life span and reduce bone densityEndocrinology2006147125592559916935844
  • HofbauerLCGoriFRiggsBLStimulation of osteoprotegerin ligand and inhibition of osteoprotegerin production by glucocorticoids in human osteoblastic lineage cells: potential paracrine mechanisms of glucocorticoid-induced osteoporosisEndocrinology1999140104382438910499489
  • GandhiMKLekamwasamSInmanISignificant and persistent loss of bone mineral density in the femoral neck after haematopoietic stem cell transplantation: long-term follow-up of a prospective studyBr J Haematol2003121346246812716369
  • TauchmanovàLSerioBDel PuenteALong-lasting bone damage detected by dual-energy x-ray absorptiometry, phalangeal osteosonogrammetry, and in vitro growth of marrow stromal cells after allogeneic stem cell transplantationJ Clin Endocrinol Metab200287115058506512414872
  • Truven Health AnalyticsMicromedex gateway [webpage on the Internet]Greenwood Village, COTruven Health Analytics Inc20122013 Available from: http://www.micromedexsolutions.com/micromedex2/librarian/Accessed May 15, 2013
  • McClungMHarrisSTMillerPDBisphosphonate therapy for osteoporosis: benefits, risks, and drug holidayAm J Med20131261132023177553
  • BrufskyAHarkerWGBeckJTZoledronic acid inhibits adjuvant letrozole-induced bone loss in postmenopausal women with early breast cancerJ Clin Oncol200725782983617159193
  • BundredNJCampbellIDDavidsonNEffective inhibition of aromatase inhibitor-associated bone loss by zoledronic acid in postmenopausal women with early breast cancer receiving adjuvant letrozole: ZO-FAST Study resultsCancer200811251001101018205185
  • LlombartAFrassoldatiAPaijaOImmediate administration of zoledronic acid reduces aromatase inhibitor-associated bone loss in postmenopausal women with early breast cancer: 12-month analysis of the E-ZO-FAST trialClin Breast Cancer2012121404822014381
  • BrufskyAMHarkerWGBeckJTFinal 5-year results of Z-FAST trial: adjuvant zoledronic acid maintains bone mass in postmenopausal breast cancer patients receiving letrozoleCancer201211851192120121987386
  • Van PoznakCHannonRAMackeyJRPrevention of aromatase inhibitor-induced bone loss using risedronate: the SABRE trialJ Clin Oncol201028696797520065185
  • SmithMRMcGovernFJZietmanALPamidronate to prevent bone loss during androgen-deprivation therapy for prostate cancerN Engl J Med20013451394895511575286
  • SmithMREasthamJGleasonDMShashaDTchekmedvianSZinnerNRandomized controlled trial of zoledronic acid to prevent bone loss in men receiving androgen deprivation therapy for nonmetastatic prostate cancerJ Urol200316962008201212771706
  • GreenspanSLNelsonJBTrumpDLResnickNMEffect of once-weekly oral alendronate on bone loss in men receiving androgen deprivation therapy for prostate cancer: a randomized trialAnn Intern Med2007146641642417371886
  • KlotzLHMcNeillIYKebabdjianMZhangLChinJCanadian Urology Research ConsortiumA phase 3, double-blind, randomised, parallel-group, placebo-controlled study of oral weekly alendronate for the prevention of androgen deprivation bone loss in nonmetastatic prostate cancer: the cancer and osteoporosis research with alendronate and leuprolide (CORAL) studyEur Urol201363592793523040208
  • BekkerPJHollowayDLRasmussenASA single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. 2004J Bone Miner Res200520122275228216432953
  • CummingsSRSan MartinJMcClungMRFREEDOM TrialDenosumab for prevention of fractures in postmenopausal women with osteoporosisN Engl J Med2009361875676519671655
  • BrownJPPrinceRLDealCComparison of the effect of denosumab and alendronate on BMD and biochemical markers of bone turnover in postmenopausal women with low bone mass: a randomized, blinded, phase 3 trialJ Bone Miner Res200924115316118767928
  • EllisGKBoneHGChlebowskiRRandomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancerJ Clin Oncol200826304875488218725648
  • AmgenStudy to determine treatment effects of denosumab in patients with breast cancer receiving aromatase inhibitor therapyClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2013 [updated March 15, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT00556374Accessed February 15, 2013
  • EllisGKBoneHGChlebowskiREffect of denosumab on bone mineral density in women receiving adjuvant aromatase inhibitors for non-metastatic breast cancer: subgroup analyses of a phase 3 studyBreast Cancer Res Treat20091181818719308727
  • SmithMREgerdieBHernándezToriz NDenosumab HALT Prostate Cancer Study GroupDenosumab in men receiving androgen-deprivation therapy for prostate cancerN Engl J Med2009361874575519671656
  • XGEVA® (denosumab) [package insert]Thousand Oaks, CAAmgen Inc2010
  • SutjandraLRodriguezRDDoshiSPopulation pharmacokinetic meta-analysis of denosumab in healthy subjects and postmenopausal women with osteopenia or osteoporosisClin Pharmacokinet2011501279380722087866
  • FizaziKLiptonAMarietteXRandomized phase II trial of denosumab in patients with bone metastases from prostate cancer, breast cancer, or other neoplasms after intravenous bisphosphonatesJ Clin Oncol200927101564157119237632
  • LiptonAStegerCGFigueroaJRandomized active-controlled phase II study of denosumab efficacy and safety in patients with breast cancer-related bone metastasesJ Clin Oncol200725284431443717785705
  • TabriziMATsengCMRoskosLKElimination mechanisms of therapeutic monoclonal antibodiesDrug Discov Today2006111–2818816478695
  • BlockGABoneHGFangLLeeEPadhiDA single-dose study of denosumab in patients with various degrees of renal impairmentJ Bone Miner Res20122771471147922461041
  • StopeckATLiptonABodyJJDenosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind studyJ Clin Oncol201028355132513921060033
  • FizaziKCarducciMSmithMDenosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind studyLancet2011377976881382221353695
  • HenryDHCostaLGoldwasserFRandomized, double-blind study of denosumab versus zoledronic acid in the treatment of bone metastases in patients with advanced cancer (excluding breast and prostate cancer) or multiple myelomaJ Clin Oncol20112991125113221343556
  • LiptonAFizaziKStopeckATSuperiority of denosumab to zoledronic acid for prevention of skeletal-related events: a combined analysis of 3 pivotal, randomised, phase 3 trialsEur J Cancer201248163082309222975218
  • PaparodisRBuehringBPelleyEBinkleyNA case of unusual subtrochanteric fracture in a patient receiving denosumabEndocr Pract2013117 [Epub ahead of print]
  • KongYYYoshidaHSarosiIOPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesisNature199939767173153239950424
  • MillerPDWagmanRBPeacockMEffect of denosumab on bone mineral density and biochemical markers of bone turnover: six year results of a phase 2 clinical trialJ Clin Endocrinol Metab201196239440221159841
  • KendlerDLRouxCBenhamouCLEffects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapyJ Bone Miner Res2010251728119594293
  • FizaziKBrownJECarducciMDenosumab in patients with metastatic prostate cancer previously treated with denosumab or zoledronic acid: 2-year open-label extension phase results from the pivotal phase 3 studyEuropean Society for Medical Oncology 2012 CongressSeptember 28–October 2, 2012Vienna, Austria Abstract 937P
  • StopeckATLiptonAMartínMDenosumab in patients with breast cancer and bone metastases previously treated with zoledronic acid or denosumab: results from the 2-year open-label extension treatment phase of a pivotal phase 3 study2011 CTRC-AACR San Antonio Breast Cancer SymposiumDecember 6–10, 2011San Antonio, TX Abstract P3-16-07
  • GnantMMineritschBSchippingerWABCSG-12 Trial InvestigatorsEndocrine therapy plus zoledronic acid in premenopausal breast cancerN Engl J Med2009360767969119213681
  • ColemanREMarshallHCameronDAZURE InvestigatorsBreast-cancer adjuvant therapy with zoledronic acidN Engl J Med2011365151396140521995387
  • WongMHStocklerMRPavlakisNBisphosphonates and other bone agents for breast cancerCochrane Database Syst Rev20122CD00347422336790
  • Gonzalez-SuarezEJacobAPJonesJRANK ligand mediates progestin-induced mammary epithelial proliferation and carcinogenesisNature2010468732010310720881963
  • SchramekDLeibbrandtASiglVOsteoclast differentiation factor RANKL controls development of progestin-driven mammary cancerNature201046873209810220881962
  • Gonzalez-SuarezEBranstetterDArmstrongADinhHBlumbergHDougallWCRANK overexpression in transgenic mice with mouse mammary tumor virus promoter-controlled RANK increases proliferation and impairs alveolar differentiation in the mammary epithelia and disrupts lumen formation in cultured epithelial aciniMol Cell Biol20072741442145417145767
  • JonesDHNakashimaTSanchezOHRegulation of cancer cell migration and bone metastasis by RANKLNature2006440708469269616572175
  • RoodmanGDMechanisms of bone metastasisN Engl J Med2004350161655166415084698
  • AmgenStudy of denosumab as adjuvant treatment for women with high risk early breast cancer receiving neoadjuvant or adjuvant therapy (D-CARE)ClinicalTrialsgov [website on the Internet]Bethesda, MDUS National Library of Medicine2013 [updated January 25, 2013]. Available from: http://clinicaltrials.gov/ct2/show/NCT01077154Accessed February 15, 2013
  • SmithMRSaadFColemanRDenosumab and bone-metastasis-free survival in men with castration-resistant prostate cancer: results of a phase 3, randomised, placebo-controlled trialLancet20123799810394622093187