Abstract
Denosumab is a fully human monoclonal antibody to receptor activator of nuclear factor kappa-B ligand (RANKL), a cytokine member of the tumor necrosis factor family that is the principal regulator of osteoclastic bone resorption. Postmenopausal osteoporosis (PMO) is a systemic skeletal disease associated with high levels of RANKL, resulting in a high rate of bone remodeling and an imbalance of bone resorption over bone formation. By inhibiting RANKL in women with PMO, denosumab reduces the rate of bone remodeling, thereby increasing bone mineral density, improving bone strength, and reducing the risk of fractures. In clinical trials of women with osteoporosis and low bone mineral density, denosumab has been well tolerated, with overall rates of adverse events and serious adverse events in women treated with denosumab similar to those receiving placebo. In the largest clinical trial of denosumab for the treatment of women with PMO, there was a significantly greater incidence of cellulitis reported as a serious adverse event, with no difference in the overall incidence of cellulitis, and a significantly lower incidence of the serious adverse event of concussions with denosumab compared with placebo. The evidence supports a favorable balance of benefits versus risks of denosumab for the treatment of PMO. Assessments of the long-term safety of denosumab are ongoing. Denosumab 60 mg subcutaneously every 6 months is an approved treatment for women with PMO who are at high risk for fracture.
Keywords: denosumab, osteoporosis, safety, risk, benefit, FDA
Introduction
Osteoporosis is a skeletal disease characterized by low bone mineral density (BMD) and poor bone quality that reduces bone strength and increases the risk of fractures.1 It is a major public health concern, with more than 200 million men and women reported to have osteoporosis worldwide,2 including about 75 million individuals in Europe, Japan, and the USA.3 Approximately one-third of postmenopausal women in Europe and the USA have osteoporosis, with about 40% of them expected to have at least one fragility fracture during their remaining lifetimes.4 Any fracture may be painful and disabling. Hip fractures and vertebral fractures are associated with an increased risk of death.5,6
Osteoporosis is diagnosed by measuring BMD with dual-energy X-ray absorptiometry. In patients with osteoporosis, many pharmacological agents, including the bisphosphonates (eg, alendronate, risedronate, ibandronate, zoledronic acid), raloxifene, salmon calcitonin, strontium ranelate, teriparatide, parathyroid hormone (PTH) 1–84, and denosumab have been shown to reduce fracture risk with generally favorable safety profiles, with other compounds for the treatment of osteoporosis being investigated.7 There is accumulating evidence that by preventing fractures, particularly fractures of the hip, drug therapy can lower mortality rates.8–11 However, despite these advances, the care of osteoporosis in clinical practice has been disappointing. Osteoporosis is underdiagnosed12 and undertreated;13 when treatment is prescribed, compliance and persistence (collectively called adherence) is often poor,14 resulting in higher fracture risk15 and greater health care costs than in patients with good adherence.14 While many factors have been associated with poor adherence to osteoporosis therapy, side-effects, fear of side-effects, and poor understanding of the balance between the expected benefits and potential risks of therapy are important considerations.16 More effective risk communication and shared decision making offer the potential of improving clinical outcomes by enhancing patient understanding of the balance of benefits and risks and improving adherence to therapy.17,18
Denosumab (Prolia®, Amgen Inc, Thousand Oaks, CA) is a compound with a novel mechanism of action that is approved for the treatment of postmenopausal women with osteoporosis at high risk for fracture, treatment to increase bone mass in men at high risk for fracture receiving androgen deprivation therapy for nonmetastatic prostate cancer, and treatment to increase bone mass in women at high risk for fracture receiving adjuvant aromatase inhibitor therapy for breast cancer. It is administered by a health care professional as a 60 mg subcutaneous (SC) injection every 6 months (Q6M). The same drug, used with a higher dose (120 mg) and shorter interval between doses (every 4 weeks [Q4W]), is approved as XgevaTM (Amgen Inc) for the prevention of skeletal-related events in patients with bone metastases from solid tumors.
This review focuses on the safety and tolerability of denosumab in treating women with postmenopausal osteoporosis (PMO).
Methodology
Randomized controlled clinical trials of denosumab were selected from a PubMed search for matches with “denosumab.” US regulations on safety reporting in clinical trials and safety reports for denosumab were obtained from the website of the US Food and Drug Administration (FDA). Additional information on denosumab safety was selected from abstracts and oral presentations at recent scientific congresses.
Bone remodeling
The pathophysiology of PMO and the mechanism of action of the drugs used to treat osteoporosis, including denosumab, involve alterations of bone remodeling. The adult skeleton undergoes a lifelong process of resorption and formation in numerous localized areas called “bone multicellular units” (BMUs), with each BMU having an estimated volume of about 0.025 mm3 and a lifespan of about 6–9 months, resulting in a turnover of approximately 10% of the entire skeleton each year.19 BMUs can be identified microscopically as tiny pits on the surface of trabecular bone (the type of bone in the interior of vertebral bodies and inside the ends of long bones) and tunnels (Haversian systems) in cortical bone, the exterior envelope around all bones. Bone remodeling may be “targeted” at repairing areas of microdamage that occur from normal physical activities as well as with severe repetitive microtrauma, or may be “nontargeted” when it occurs at apparently random locations on bone to maintain mineral homeostasis, keeping extracellular calcium levels within a very narrow range.20 In healthy premenopausal women, there is a low rate of remodeling and an even balance between bone resorption and formation. With declining estrogen levels in perimenopausal and postmenopausal women, the remodeling rate is accelerated and unbalanced, with bone resorption being greater than formation. As a consequence, there is a degradation of microarchitectural elements that may eventually result in measurable bone loss, increased skeletal fragility, and high fracture risk.
The cells involved in the highly coordinated process of bone remodeling are osteoclasts (bone resorbing cells), osteoblasts (bone forming cells), and osteocytes (mechanosensory cells). Receptor activator of nuclear factor kappa-B ligand (RANKL), a member of the tumor necrosis family of proteins that is expressed by cells of the osteoblast lineage, has been identified as the principal regulator of osteoclastic bone resorption.21 When RANKL binds to its receptor, RANK, on the cell membrane of osteoclasts and preosteoclasts,22 it stimulates their formation, activity, and survival, thereby increasing the rate of bone resorption.23 Osteoprotegerin (OPG), also expressed by osteoblasts, is a counter-regulatory nonsignaling “decoy receptor” for RANKL. The binding of RANKL to OPG reduces the amount of RANKL available for binding to RANK, resulting in a decrease in osteoclast formation, activity, and survival. The balance between RANKL and OPG is thought to be a major determinant of the rate of bone resorption, with an abundance of RANKL favoring more bone resorption and, conversely, an abundance of OPG favoring less bone resorption. The decline of estrogen in postmenopausal women leads to an excess of RANKL over OPG24 and an increase in bone resorption. The discovery that high levels of RANKL were associated with increased bone resorption and bone loss suggested that inhibition of RANKL might be an effective treatment for osteoporosis and other skeletal diseases characterized by high bone turnover.
Early studies of RANKL inhibition
In vitro studies have shown that recombinant OPG inhibits osteoclast differentiation in a dose-dependent manner.25 Recombinant OPG prevents bone loss in ovariectomized rats25 and causes nonlethal osteopetrosis in normal mice.25 In young male rats, RANKL inhibition increases bone mineralization and improves mechanical strength in the femur.26 These preclinical studies and others with similar findings paved the way for the study of RANKL inhibition in humans.
The first clinical trial of RANKL inhibition was a Phase I randomized, double-blind, placebo-controlled, sequential dose-escalation study of OPG-Fc, a fusion molecule of recombinant OPG with the Fc fragment of immunoglobulin G, in 52 healthy postmenopausal women.27 Administration of a single SC dose of OPG-Fc resulted in a rapid, reversible, and dose-dependent suppression of bone resorption, and was not associated with serious adverse events or identification of neutralizing antibodies. The results of this study led to further clinical investigation of RANKL inhibition with denosumab, a fully human monoclonal antibody to RANKL that acts similarly to OPG-Fc, with the additional advantages of having greater antiresorptive potency, a longer duration of action, and avoidance of the potential risk of an OPG molecule generating anti-OPG antibodies that might crossreact with endogenous OPG.
Pharmacodynamics and pharmacokinetics of denosumab
Bone turnover markers (BTMs) such as N-telopeptide (NTX) and C-telopeptide, markers of bone resorption, and bone-specific alkaline phosphatase (BSAP), a marker of bone formation, have been used to assess the effect of denosumab on the rate of bone turnover. Changes in NTX and BSAP were measured in a Phase I randomized, placebo-controlled, double-blind, single-dose, dose-escalation study of SC denosumab (0.01 to 3.0 mg/kg) in 49 healthy postmenopausal women followed for up to 9 months. Treatment with denosumab was followed by a rapid (within 12 hours of dosing) dose-dependent decrease in urinary NTX, with a maximum decrease of 84% at 3 months and a rise in NTX levels at the end of the observational period.28 Decreases in BSAP occurred later and were less pronounced than NTX.
In the same study, the pharmacokinetics of denosumab was nonlinear with dose, as reported with other fully human monoclonal antibodies. Serum profiles were characterized by three phases: (1) a prolonged absorption phase with maximum serum concentration (Cmax) observed 5–21 days postdose, with the Cmax increasing as dose increased; (2) a prolonged β-phase, with serum half-life as long as 32 days with the maximum dose; and (3) a rapid terminal phase with serum concentration dropping below 1000 ng/ml.
Efficacy of denosumab
The rapid, profound, sustained, and reversible decrease of NTX in the Phase I study of denosumab28 supported further investigation of the efficacy and safety of this antiresorptive compound as a potential treatment for osteoporosis. A Phase II randomized, placebo-controlled, dose-ranging study evaluated the effects of denosumab in postmenopausal women with low BMD, defined as a lumbar spine T-score of −1.8 to −4.0 or total hip or femoral neck T-score of −1.8 to −3.5, with an initial enrollment of 412 subjects. The findings of this study have been reported for the time points of 1 year,29 2 years,30 4 years,31 6 years,32 and 8 years.33 Subjects received either denosumab 6, 14, or 30 mg every 3 months; 14, 60, 100, or 210 mg Q6M, open-label oral alendronate 70 mg once weekly; or placebo for the first 24 months of study. The primary endpoint was change in lumbar spine BMD compared with baseline at 12 months. It was found that lumbar spine BMD increased significantly at 12 months by 3.0%–6.7% with SC denosumab doses of 6, 14, or 30 mg every 3 months or 14, 60, 100, or 210 mg Q6M, with smaller but significant increases at other measured skeletal sites, including total hip and one-third radius (P < 0.001 for all).29 BTMs decreased in a dose-dependent manner. Continuous treatment in 80 subjects who received denosumab for 8 years, with a dose of 60 Q6M (the dose that was later approved for osteoporosis treatment) after 24 months, was associated with progressive gains in BMD: a mean increase of 16.8% at the lumbar spine and 6.9% at the total hip compared with baseline.33 Reductions in C-telopeptide and BSAP levels were sustained for the entire time of treatment.
The Fracture Reduction Evaluation of Denosumab in Osteoporosis every 6 Months (FREEDOM) trial was an international 3-year, randomized, double-blind, placebo-controlled Phase III study in 7868 women with PMO. This was the pivotal fracture trial to determine the efficacy of denosumab in reducing fracture risk. Subjects were randomized to receive SC denosumab 60 mg (n = 3902) or placebo (n = 3906) Q6M.34 The primary efficacy endpoint was new vertebral fractures at 36 months, with secondary endpoints that included time to first hip fracture and non-vertebral fractures. Baseline T-score at the lumbar spine or total hip ranged from less than −2.5 to greater than or equal to −4.0. Approximately 23% of subjects had at least one baseline prevalent vertebral fracture. Subjects were excluded from participation for any severe prevalent vertebral fracture or more than two moderate prevalent vertebral fractures. Treatment with denosumab was associated with a statistically significant 68% decrease in the risk of new vertebral fractures compared with placebo (2.3% denosumab versus 7.2% placebo, P < 0.0001), a 40% decrease in the risk of hip fractures (0.7% denosumab versus 1.2% placebo, P = 0.036), and a 20% decrease in the risk of nonvertebral fractures (6.5% denosumab versus 8.0% placebo, P = 0.011).34
Other Phase III studies have been conducted to evaluate the efficacy and safety of denosumab in a variety of circumstances that could provide guidance to physicians considering the use of this agent in clinical practice. Denosumab Fortifies Bone Density (DEFEND) was a 2-year randomized, double-blind, placebo-controlled Phase III study of denosumab in 332 postmenopausal women with low BMD, defined as lumbar spine T-score between −1.0 and −2.5.35 This study evaluated the efficacy of denosumab to stabilize or increase BMD in postmenopausal women with osteopenia. Subjects were randomized to receive SC denosumab 60 mg Q6M or placebo, with a primary efficacy endpoint of percentage change of lumbar spine BMD at 24 months compared with placebo. Denosumab significantly increased BMD at the lumbar spine compared with placebo (denosumab 6.5% versus placebo −0.6%, P < 0.0001), with significant BMD increases at other measured skeletal sites as well (P < 0.0001). Denosumab significantly reduced levels of BTMs compared with placebo.
Determining Efficacy: Comparison of Initiating Denosumab versus Alendronate (DECIDE) was a 1-year randomized, double-blind, double-dummy Phase III noninferiority study in 1189 postmenopausal women with lumbar spine or total hip T-score of −2.0 or less.36 DECIDE was a head-to-head comparison of the effects of denosumab and alendronate, the most commonly prescribed bisphosphonate for the treatment of osteoporosis. Subjects were randomized to receive SC denosumab SC 60 mg Q6M plus weekly oral placebo or oral alendronate 70 mg weekly plus placebo SC injections Q6M. The primary efficacy endpoint was the percentage change from baseline of total hip BMD after 12 months of treatment with denosumab compared with alendronate. It was found that subjects treated with denosumab had a significantly greater BMD increase at the total hip (denosumab 3.5% versus alendronate 2.6%, P < 0.0001) and at all other measured skeletal sites at 12 months compared with alendronate. Reductions in BTM levels were significantly greater with denosumab than alendronate.
The Study of Transitioning from Alendronate to Denosumab (STAND) addressed the response to treatment with denosumab in women previously treated with alendronate. This was a 1-year double-blind, active-controlled, double-dummy Phase III study in 504 postmenopausal women treated with alendronate for at least the past 6 months (median 36 months, range 6–192 months) and having a baseline lumbar spine or total hip T-score of −2.0 to −4.0.37 Subjects were randomized to either switching from alendronate to SC denosumab 60 mg Q6M or continuing oral alendronate 70 mg weekly. The primary efficacy endpoint was the percentage change in total hip BMD at 12 months in subjects switched to denosumab compared with those who continued alendronate. A significantly greater total hip BMD increase was reported in subjects switched to denosumab (denosumab 1.90%, alendronate 1.05%, P < 0.0001). BMD increases with denosumab were also greater at the lumbar spine, and distal one-third radius (P < 0.0125 for all).
These clinical trials show that denosumab is associated with a reduction in BTM levels, increase in BMD, and, in women with postmenopausal osteoporosis, a decrease in the risk of vertebral fractures, nonvertebral fractures, and hip fractures.
Denosumab safety reporting in clinical trials and postmarketing surveillance
Safety has been evaluated in all of the denosumab clinical trials and their extensions. Postmarketing safety data are collected through reports submitted to Amgen Medical Information,38 the Amgen Post-marketing Active Safety Surveillance Program39 (available to enrolled physicians), and the MedWatch Program of the FDA.40 The denosumab Risk Evaluation and Mitigation Strategy (REMS)41 is a strategy to manage known or potential risks associated with denosumab to ensure that the benefits of treatment outweigh the risks. The REMS program includes methods for communicating risks to health care professionals and patients in the form of “Prescribing Information,”42 a “Medication Guide,”43 and “Dear Healthcare Professional” letters,44 all of which are accessible through the Prolia product website45 and the REMS website.41
The required terminology for premarketing safety reporting of investigational new drugs is defined by the US Code of Federal Regulations (CFR), recently updated in the Federal Food, Drug and Cosmetic Act (21 CFR Parts 312 and 320) (Table 1).46 A reported untoward (unfavorable, negative, or harmful) medical occurrence is designated as an adverse “event” or “reaction” that may be modified by the terms “serious,” “life-threatening,” “suspected,” or “unexpected.” When an untoward occurrence is reported as an adverse “event,” there may or may not be a causal relationship with the drug. When reported as an adverse “reaction” without a modifier, there is reason to conclude that the drug caused the event. The modifier “suspected” with “reaction” is intended to mean that there is a reasonable possibility that the drug caused the event – that is, a lesser degree of certainty than without the modifier.
Table 1.
Reporting term | Definition |
---|---|
Adverse event | Any untoward medical occurrence associated with the use of a drug in humans, whether or not considered drug related |
Adverse reaction | Any adverse event caused by a drug |
Life-threatening adverse event or life-threatening suspected adverse reaction | An adverse event or suspected adverse reaction is considered “life-threatening” if, in the view of either the investigator or sponsor, its occurrence places the patient or subject at immediate risk of death |
Serious adverse event or serious suspected adverse reaction | An adverse event or suspected adverse reaction is considered “serious” if, in the view of either the investigator or sponsor, it results in any of the following outcomes: death, a life-threatening adverse event, inpatient hospitalization or prolongation of existing hospitalization, a persistent or significant incapacity or substantial disruption of the ability to conduct normal life functions, or a congenital anomaly/birth defect |
Suspected adverse reaction | Any adverse event for which there is a reasonable possibility that the drug caused the adverse event |
Unexpected adverse event or unexpected suspected adverse reaction | An adverse event or suspected adverse reaction is considered “unexpected” if it is not listed in the investigator brochure or is not listed at the specificity or severity that has been observed; or, if an investigator brochure is not required or available, is not consistent with the risk information described in the general investigational plan or elsewhere in the current application, as amended |
Notes: The US Code of Federal Regulations defines the terms used for identifying unfavorable, negative, or harmful medical occurrences in clinical trials of INDs and bioavailability and bioequivalence studies. An “adverse event” is an untoward medical occurrence in any clinical trial subject (eg, receiving study drug or placebo) regardless of causality, while an “adverse reaction” is an untoward medical occurrence that is caused by a drug. All definitions are from: US Food and Drug Administration. Investigational new drug safety reporting requirements for human drug and biological products and safety reporting requirements for bioavailability and bioequivalence studies in humans. Federal Register. 21 CFR Parts 312 and 320. 2010;75(188):59935–59963. Available from: http://frwebgate.access.gpo.gov/cgi-bin/getdoc.cgi?dbname=2010_register&docid=fr29se10-3.pdf. Accessed September 2, 2011.
The implementation of this terminology may help to resolve some of the uncertainty and confusion generated from earlier definitions that might mislead patients and physicians regarding safety and causality with denosumab. For example, the FDA stated47 that “An adverse drug reaction, also called a side effect, is any undesirable experience associated with the use of a medicine in a patient.” There is no mention of causality, despite designating “side effect” as synonymous with “adverse drug reaction.” The use of “side effect” in this fashion is contrary to common usage and quite different from the dictionary definition of “side effect,” which clearly recognizes a causal relationship between the drug and the untoward occurrence.48 The distinction between an untoward occurrence that is caused by a drug and one that is likely to be a consequence of the disease state being studied (eg, a fracture with osteoporosis) or one that commonly occurs in the population being studied (eg, back pain in the elderly) is an important one.
A “Medication Guide” is a paper handout or pamphlet for distribution of FDA-approved information to patients for medications that the FDA determines pose a serious and significant public health concern. US federal regulations (21 CFR 208.20) state that it “shall be scientifically accurate and shall be based on, and shall not conflict with, the approved professional labeling for the drug product under 201.57;”49 21 CFR 201.57 defines adverse reactions as “only those adverse events for which there is some basis to believe there is a causal relationship between the drug and the occurrence of the adverse event.”50 However, the Medication Guide for Prolia® in use at the time of writing lists the most common “side effects” as back pain, arm and leg pains, high cholesterol, muscle pain, and bladder infections.43 While these untoward medical occurrences were commonly reported in FREEDOM trial subjects, the differences in incidence between those receiving denosumab and placebo were trivial (Table 2). The Medication Guide for Prolia® does not provide information on the frequency of these occurrences with treatment compared with placebo and does not state whether there is a clinically or statistically significant difference in these events in subjects taking denosumab and those on placebo; there is no mention of severity, causality, or biological plausibility.
Table 2.
Adverse reaction | Denosumab (n = 3886) | Placebo (n = 3876) |
---|---|---|
Back pain | 1347 (34.7%) | 1340 (34.6%) |
Pain in extremity | 453 (11.7%) | 430 (11.1%) |
Musculoskeletal pain | 297 (7.65) | 291 (7.5%) |
Hypercholesterolemia | 280 (7.2%) | 236 (6.1%) |
Cystitis | 228 (5.9%) | 225 (5.8%) |
Notes: These are listed as “side effects” of denosumab in the Prolia® medication guide43 without explanation regarding statistical significance, causality, or clinical relevance. These untoward medical occurrences are those that are common in elderly populations, with the differences in incidence between the two groups being very small.
Safety concerns
At a meeting of the Advisory Committee for Reproductive Health Drugs held on August 13, 2009, the FDA safety analysis identified the following as “adverse events of special interest”: infection, new malignancy, tumor progression, dermatologic events, hypocalcemia, osteonecrosis of the jaw (ONJ), bone histomorphometry findings, hypersensitivity/immunogenicity, cardiovascular adverse events, pancreatitis, and ocular adverse events.51 For some of these categories, biological plausibility was clearly present, while others appeared to be listed due to unexpected clinical trial findings. After consideration of the efficacy and safety data available at that time, the committee voted unanimously that the benefit of treatment with denosumab is likely to outweigh the risks in women with PMO at high risk for fracture.52 This ultimately led to FDA approval for this indication.
The biological plausibility for possible increased risk of infections, new malignancies, and tumor progression with denosumab is derived from what is known about the role of the RANKL/RANK/OPG signaling pathway with the immune system. RANKL plays an essential role in the development and maturation of the immune system in rodents,53 is expressed by activated T cells,54 and is an activator of dendritic cells, monocytes, and macrophages.55,56 OPG is expressed by mature B cells.57 However, a small permissive level of RANKL and RANK in transgenic rats is associated with normal lymph node development58,59 and inhibition of RANKL in adult humans has not been shown to adversely affect measured parameters of immune function.27,28 Carcinogenicity studies were not performed with denosumab, since it is an antibody specific to human and nonhuman primate RANKL and is not active in the rodent. Potential adverse effects of denosumab on immunity continue to be assessed in clinical trials and postmarketing surveillance.
RANK and RANKL are expressed in normal human skin. Preclinical studies have shown that RANKL is a regulator of epidermal dendritic cell function and T cell activity in the skin.60 The finding that cutaneous RANKL expression suppresses inflammation of the skin in mice61 suggests the possibility that RANKL inhibition in humans might lead to an enhanced inflammatory response, providing a rationale for the observation of some of the dermatological untoward occurrences reported with denosumab.34
Hypocalcemia, usually subclinical but sometimes symptomatic, with associated increases in serum PTH levels, has been observed with the use of potent antiresorptive agents, including bisphosphonates62 and denosumab.29
Denosumab is a robust antiresorptive agent, as demonstrated by the magnitude of reduction in BTMs28–34 and the results of histomorphometry in bone biopsy substudies in clinical trials.63 There has been concern that prolonged treatment with drugs that inhibit bone remodeling might cause oversuppression of bone turnover64 that could impair the healing of microcracks, possibly leading to reduced bone strength with unusual fractures,65 impairment of fracture healing,66 or ONJ.67
There is a potential for any therapeutic protein, including denosumab, to elicit an immune response. Most clinical studies with denosumab included evaluations of immunogenicity with a screening immunoassay to detect binding antibodies, a second immunoassay to confirm binding antibodies, and a cell-based bioassay to evaluate for neutralizing antibodies.28–34
Cardiovascular concerns with denosumab have been raised since RANKL, RANK, and OPG are expressed by vascular endothelial cells, and studies in humans have shown an association of elevated OPG levels with cardiovascular endpoints, such as coronary artery disease,68 stroke,69 progression of atherosclerosis,70 vascular calcification,71 and mortality. 72 However, it now appears that increased OPG levels are a compensatory response to vascular disease rather than a causal factor, and that OPG may inhibit vascular calcification by reducing bone turnover.73 An increased risk of atrial fibrillation as a serious adverse event has been reported in a single randomized, placebo-controlled trial of zoledronic acid,74 another potent antiresorptive agent, suggesting the possibility that this might be a class effect of bisphosphonates or all antiresorptive drugs. After a review of all available data, the FDA found that there was insufficient evidence to conclude that bisphosphonates caused atrial fibrillation and that physicians should not change prescribing habits due to this concern.75
FDA concerns regarding pancreatitis focused on a numerical imbalance in reported events in denosumab-treated subjects compared with those receiving placebo in pooled PMO studies. There was also a numerical imbalance of an ocular adverse event (cataracts) in a trial of denosumab in men with prostate cancer but not in PMO studies.76 There is no clear rationale for a causal relationship between denosumab and either of these adverse events.
As with the study of any investigational new drug, adverse effects that are very uncommon or ones that do not emerge until treatment has been continued for many years may not be recognized in clinical trials. The reasons that clinical trials fail to identify all adverse drug effects include the relatively small number of subjects compared with the total number of patients eventually exposed to the drug, the homogeneity of the study population compared with patients in clinical practice, and the limited duration of observation during studies compared with a much longer time of treatment for many clinical practice patients. For these reasons, postmarketing surveillance for the safety of approved medications is appropriate and necessary.
Safety data
In the report of the Phase I study of denosumab in healthy postmenopausal women, there were no related serious adverse events and no discontinuations from the study due to adverse events.28 Mild transient dose-dependent decreases in albumin-adjusted serum calcium and corresponding increases in serum intact PTH levels were observed. Infectious adverse events were similar with the combined denosumab dosing groups (38%) and placebo (33%). Two subjects treated with denosumab had mild injection-site reactions. There were no changes in white blood cell counts, T-, B-, or NK-cell counts, immunoglobulins, or coagulation parameters associated with denosumab treatment.
Safety data for subjects in the Phase II study of denosumab in postmenopausal women with low BMD have been incrementally reported for time points beginning at 1 year and progressing most recently to 8 years.29–33 At 4 years, all adverse events, including infections, and all serious adverse events, including malignancies, were similar in groups receiving denosumab, alendronate, and placebo.31 Most patients reported an adverse event at some stage during the 4 years, the most common events being an upper respiratory tract infection, arthralgia, and back pain. Infections requiring hospitalization were reported in 3.2% of subjects treated with denosumab and none of those receiving placebo or alendronate. The infections were typical community-acquired infections that responded appropriately to conventional antibiotic therapy. No opportunistic infections suggestive of impaired immune function were reported. Serum calcium levels remained stable and within the normal range with denosumab, with no patient having symptomatic hypocalcemia. No patient developed neutralizing antibodies to denosumab. Of the 262 subjects who completed the 4-year parent study, 200 enrolled in the 4-year single-arm study extension, with all receiving open-label SC denosumab 60 mg Q6M; 116 completed the extension and 80 of these received 8 years of continuous denosumab treatment. The adverse event profile at 6 years32 and 8 years33 was similar to what was reported after 1–4 years of drug exposure, with no report of increased frequency of any specific event over time.
FREEDOM, the largest of the denosumab clinical trials, provides the single most robust database for evaluation of safety endpoints, with 3-year data published in a peer-reviewed journal34 and more detailed information presented to the FDA as a report to the Advisory Committee for Reproductive Health Drugs.77 After 3 years of observation, there were no significant differences in the total incidence of adverse events or serious adverse events in subjects receiving denosumab or placebo. There was no evidence of hypersensitivity or allergic drug reactions. Neutralizing antibodies to denosumab were not detected in any of the clinical trial subjects. The overall risk of infections, opportunistic infections, cardiovascular events, atrial fibrillation, stroke, cataracts, hypocalcemia, malignancies, and death was similar in subjects receiving denosumab or placebo. In pooled data from FREEDOM and other studies in women with PMO (n = 4050 for denosumab-treated subjects, n = 4041 for placebo-treated subjects), pancreatitis was reported in eight subjects on denosumab and four subjects on placebo; cataracts were reported for 232 subjects on denosumab and 253 subjects on placebo.51
In a prespecified analysis, complications in the healing of nonvertebral fractures were evaluated.78 Complications associated with the fracture or its surgical repair occurred in 1.7% of denosumab subjects and 5.7% of placebo subjects (P < 0.01). There were two reports of delayed union with denosumab and four with placebo, and one nonunion with placebo. There was no signal for impaired fracture healing with denosumab. There were no reports of atypical femur fractures or ONJ in denosumab-treated subjects.
Assessment of adverse events showed that eczema and flatulence were significantly more common with denosumab, while falling was less common with denosumab; with serious adverse events, cellulitis was significantly more common with denosumab, although there was no significant difference in the incidence of cellulitis overall, and concussions were less common with denosumab (Table 3). Skin infections were not related to the injection site or to the timing of the injection. Denosumab was well tolerated with no reported injection-site reactions. Local reactions after injection occurred in 0.8% of subjects in the denosumab group and 0.7% in the placebo group.
Table 3.
Type of event | Denosumab (n = 3886) | Placebo (n = 3876) | P value |
---|---|---|---|
Adverse events occurring in at least 2% of subjects in either group with P ≤ 0.05 | |||
Eczema | 118 (3.0%) | 65 (1.7%) | <0.001 |
Falling | 175 (4.5%) | 219 (5.7%) | 0.02 |
Flatulence | 84 (2.2%) | 53 (1.4%) | 0.008 |
Serious adverse events occurring in at least 0.1% of subjects in either group with P ≤ 0.01 | |||
Cellulitis (including erysipelas) | 12 (0.3%) | 1 (<0.1%) | 0.002 |
Concussion | 1 (<0.1%) | 11 (0.3%) | 0.004 |
All adverse events and serious adverse events | 3605 (92.8%) | 3607 (93.1%) | 0.91 |
All serious adverse events | 1004 (25.8%) | 972 (25.1%) | 0.61 |
Notes: Five events were identified as being statistically significantly different between subjects treated with denosumab and placebo. Three of these (eczema, flatulence, cellulitis) favored placebo while two (falling, concussions) favored denosumab. There was no difference in total adverse events or serious adverse events.
The quality of bone with denosumab treatment and the mechanism of action of denosumab have been evaluated in bone biopsy substudies of FREEDOM and STAND. Transiliac bone biopsies with a standardized double-tetracycline labeling procedure were obtained at 24 and/or 36 months in FREEDOM (47 women on denosumab, 46 on placebo) and at 12 months in STAND (15 women on denosumab after alendronate, 21 continuing alendronate).63 Qualitative bone histology was unremarkable in all women treated with denosumab, showing normal lamellar bone, normal mineralization, and the absence of marrow fibrosis. Data from FREEDOM biopsies at 24 and 36 months were not significantly different and were therefore combined to provide greater statistical power. In the FREEDOM substudy, double tetracycline labeling was observed in trabecular or cortical bone in all biopsies of subjects on placebo, while for subjects treated with denosumab, 40% had double label, 25% had single label only, and 36% had no label. There was no correlation between BTM levels and the presence or absence of labeling. Nine subjects in the substudy had fractures: six in the placebo group (all with double labels) and three in the denosumab group (one with single labels and two with no labels), with no reports of impaired fracture healing. Structural indices measured by histomorphometry and microcomputed tomography were consistent with reduced bone turnover in the denosumab group. Reduced cortical porosity and increased cortical volumetric BMD were observed in the denosumab group with microcomputed tomography at 24 months. In the STAND substudy, indices of bone turnover tended to be lower in the denosumab group than the alendronate group; double labeling was observed in trabecular or cortical bone in all biopsies of subjects in the alendronate group, while for subjects switched to denosumab, 60% had double label, 20% had single label only, and 20% had no label. These bone biopsy substudies confirmed that denosumab is a robust inhibitor of bone remodeling. The clinical relevance and long-term safety of such a low level of remodeling with a reduction or absence of detectable tetracycline labeling in a substantial number of subjects is unknown. Reversibility of the antiresorptive effects of denosumab has been demonstrated by changes in BTMs and BMD in subjects discontinuing denosumab in the Phase II study.31 Reversibility was confirmed in a bone biopsy study in 15 women who had been treated with denosumab and discontinued treatment an average of 2 years before a double tetracycline-labeled bone biopsy was done.79 These biopsies showed normal histology and bone remodeling similar to untreated women with postmenopausal osteoporosis.
A detailed analysis of adverse events and serious adverse events of specific types of infections reported in the 3-year FREEDOM dataset has recently been released.80 The incidence of serious adverse events of infections did not change with increasing duration of denosumab exposure. The incidence of serious adverse events of opportunistic infections was low and similar in both groups, with no temporal relationship between administration of drug and the onset of infection. Serious adverse events of skin infections in denosumab-treated subjects were mostly cellulitis and clinically diagnosed erysipelas in the lower extremities (unrelated to the injection site) that resolved with the use of common antibiotics. Preexisting risk factors of venous ulcers and skin wounds were reported in five of the twelve denosumab-treated subjects with these infections. There was no temporal relationship between administration of drug and the onset of skin infections. Serious adverse events of urinary tract infections were reported in 29 (0.7%) of denosumab-treated subjects and 20 (0.5%) of those receiving placebo; these were typically caused by Escherichia coli or other common Gram-negative bacteria. Endocarditis, without identification of a causative organism, was reported in three subjects in the denosumab group and none in the placebo group. Two of the subjects with reported endocarditis underwent echocardiography and were treated, while the third subject had suspected acute bacterial endocarditis associated with fatal multiorgan failure, but no further details were available and no autopsy was performed. Neutrophil, lymphocyte, and monocyte counts were similar in both groups with no change in cell counts with increased duration of exposure to denosumab.
Of the 6468 women who completed the 3-year study, 4550 enrolled in the 7-year extension (total of 10 years) to receive open-label SC denosumab 60 mg Q6M. Preliminary data are available for the first 2 years of the extension, with 2343 women in the group with 5 years of continuous exposure to denosumab (“long-term group”) and 2207 in the group receiving placebo for 3 years followed by denosumab for 2 years (“cross-over group”).81,82 The yearly incidence rates for serious adverse events of infections, including cellulitis and erysipelas, and adverse events of malignancies for the long-term and cross-over groups in the first 2 years of the extension were similar to or lower than the observed yearly rates in the FREEDOM placebo group. The imbalances in serious adverse events of skin infections in the FREEDOM trial were not observed in the extension study. One reported case of oral osteomyelitis and one case of oral bone necrosis in the cross-over group were adjudicated as consistent with ONJ.
In DEFEND,35 DECIDE,36 and STAND,37 total adverse events and serious adverse events were similar among groups. Differences reported for some categories of adverse events and serious adverse events. In DEFEND, there were more reports of rashes, sore throats, and infections treated in hospital in the denosumab group (8.5%, 9.1%, and 4.9%, respectively) than placebo (3.0%, P = 0.035; 3.0%, P = 0.022; and 0.6%, P = 0.020, respectively). Adverse events of infections in the denosumab (60%) and placebo groups (61%) were similar, and there was no significant difference among groups in the incidence of neoplasms. In DECIDE and STAND, there were no significant differences among groups in the incidence of adverse events or serious adverse events of infections or neoplasms.
A meta-analysis83 evaluated the efficacy and safety of denosumab in 8864 subjects with low bone mass or osteoporosis in four randomized placebo-controlled trials.30,34,35,84 The analysis was dominated by the large number of subjects (7868) in FREEDOM and included one study of 252 women with nonmetastatic breast cancer on aromatase inhibitor therapy. The risk ratio for all serious adverse events, serious adverse events of infections, serious adverse events of neoplasms, and adverse events leading to study discontinuation was not statistically significantly different for women receiving denosumab and placebo. A sensitivity analysis excluding the study in cancer patients made little difference in the findings.
In three large clinical trials in cancer patients, SC denosumab 120 mg every Q4W was compared with intravenous zoledronic acid 4 mg adjusted for creatinine clearance Q4W.85–87 The primary endpoint was time to first on-study skeletal-related event, defined as radiation therapy to alleviate pain or prevent fracture, surgery to bone to treat or prevent fractures, pathologic fracture, and spinal cord compression that can result in paresthesias, incontinence, and paralysis. While the safety data from these cancer studies are not directly applicable to women treated with denosumab for osteoporosis, it may be noteworthy that no increase in the incidence of skin reactions (eg, eczema or cellulitis as a serious adverse event) was reported in denosumab-treated cancer subjects, despite the use of a higher dose and shorter dosing interval than in FREEDOM.
Patient-focused perspectives
The Denosumab Adherence, Preference, Satisfaction (DAPS) study evaluated patient perspectives with SC denosumab 60 mg Q6M compared with oral alendronate 70 mg weekly in 250 women with PMO.88 In this randomized, open-label, 2-year cross-over study, the primary study endpoint was adherence at 1 year. It was reported that adherence, defined as meeting study criteria for both compliance and persistence, was significantly greater with denosumab (87.3%) than alendronate (76.6%). Subject ratings for treatment preference and satisfaction were also significantly greater for denosumab than alendronate. Adverse events were similar with both drugs. In the second 12 months of the DAPS study, with each treatment group crossing over to the other, serious adverse events were reported in 3.5% of subjects receiving denosumab and 3.9% of those receiving alendronate.89
Role of denosumab in clinical practice
Denosumab is indicated for the treatment of women with PMO who are at high risk for fracture, defined by the FDA as a history of osteoporotic fracture, multiple risk factors for fracture, or patients who have failed or are intolerant to other available osteoporosis therapy. The focus on treating women “at high risk for fracture” is generally consistent with current clinical practice guidelines, although different definitions of “high risk” have been used. The National Osteoporosis Foundation in the USA, for example, considers a postmenopausal woman to be at sufficiently high risk for fracture to initiate pharmacological therapy, including denosumab, under any of the following conditions:90 (1) T-score of −2.5 or below at the femoral neck or lumbar spine (without the need for additional risk factors); (2) T-score between −1.0 and −2.5 at the femoral neck or lumbar spine with a FRAX (World Health Organization fracture risk assessment tool) 10-year probability of major osteoporotic fracture ≥20% or 10-year probability of hip fracture ≥3% (without the need for a densitometric classification of osteoporosis); or (3) previous hip or vertebral fracture. Denosumab, as well as alendronate, risedronate, and zoledronic acid, is recommended by the American Association of Clinical Endocrinologists as a first-line agent for the treatment of PMO.91 Denosumab is distinguished by: its infrequent injectable administration, which avoids issues of malabsorption and gastrointestinal intolerance, and may improve long-term adherence to therapy compared with oral bisphosphonates; its SC method of injection, which is less disruptive to the office routine of most physicians than intravenous bisphosphonates and expands the potential number of facilities willing to administer the drug compared with intravenous bisphosphonates; the absence of restrictions on its usage in patients with severe chronic kidney disease; and by reversibility of antiresorptive effect soon after the 6-month dosing period has passed, which may be reassuring to patients who are concerned with the long skeletal half-life of bisphosphonates. Treatment decisions should be made after an evaluation for secondary causes of osteoporosis has been conducted. The expected benefit and potential risks with each medication under consideration should be discussed with the patient prior to starting treatment.92
Conclusion
Denosumab is a fully human monoclonal antibody to RANKL associated with a rapid, sustained, and reversible reduction in BTM levels, an increase in BMD, and decrease in the risk of vertebral fractures, hip fractures, and nonvertebral fractures in women with PMO. Compared with alendronate, denosumab is associated with a greater increase in BMD and, in women previously treated with alendronate, switching to denosumab increases BMD more than continuing alendronate. It is well tolerated with total adverse events and serious adverse events generally similar to placebo. Differences between denosumab and placebo have been reported for some individual safety endpoints. The evidence supports a favorable balance between the expected benefit and potential risks with the use of denosumab for the treatment of PMO.
Acknowledgment
The author thanks Shirley Murphy, MD, for review of sections of the manuscript and helpful comments.
Footnotes
Disclosure
The author reports no conflicts of interest in this work.
References
- 1.NIH Consensus Development Panel on Osteoporosis Prevention, Diagnosis, and Therapy. Osteoporosis prevention, diagnosis, and therapy. JAMA. 2001;285(6):785–795. doi: 10.1001/jama.285.6.785. [DOI] [PubMed] [Google Scholar]
- 2.Cooper C, Campion G, Melton LJ., III Hip fractures in the elderly: a world-wide projection. Osteoporos Int. 1992;2(6):285–289. doi: 10.1007/BF01623184. [DOI] [PubMed] [Google Scholar]
- 3.European Foundation for Osteoporosis and Bone Disease, National Osteoporosis Foundation. Who are candidates for prevention and treatment for osteoporosis? Osteoporos Int. 1997;7(1):1–6. doi: 10.1007/BF01623453. [DOI] [PubMed] [Google Scholar]
- 4.Melton LJ, III, Chrischilles EA, Cooper C, Lane AW, Riggs BL. Perspective. How many women have osteoporosis? J Bone Miner Res. 1992;7(9):1005–1010. doi: 10.1002/jbmr.5650070902. [DOI] [PubMed] [Google Scholar]
- 5.Center JR, Nguyen TV, Schneider D, Sambrook PN, Eisman JA. Mortality after all major types of osteoporotic fracture in men and women: an observational study. Lancet. 1999;353(9156):878–882. doi: 10.1016/S0140-6736(98)09075-8. [DOI] [PubMed] [Google Scholar]
- 6.Cooper C. The crippling consequences of fractures and their impact on quality of life. Am J Med. 1997;103(2A):12S–19S. doi: 10.1016/s0002-9343(97)90022-x. [DOI] [PubMed] [Google Scholar]
- 7.Lewiecki EM, Bilezikian JP, Khosla S, et al. Osteoporosis update from the 2010 Santa Fe bone symposium. J Clin Densitom. 2011;14(1):1–21. doi: 10.1016/j.jocd.2010.12.001. [DOI] [PubMed] [Google Scholar]
- 8.Beaupre LA, Morrish DW, Hanley DA, et al. Oral bisphosphonates are associated with reduced mortality after hip fracture. Osteoporos Int. 2011;22(3):983–991. doi: 10.1007/s00198-010-1411-2. [DOI] [PubMed] [Google Scholar]
- 9.Center JR, Bliuc D, Nguyen ND, Nguyen TV, Eisman JA. Osteoporosis medication and reduced mortality risk in elderly women and men. J Clin Endocrinol Metab. 2011;96(4):1006–1014. doi: 10.1210/jc.2010-2730. [DOI] [PubMed] [Google Scholar]
- 10.Nurmi-Luthje I, Sund R, Juntunen M, Luthje P. Post-hip fracture use of prescribed calcium plus vitamin D or vitamin D supplements and antiosteoporotic drugs is associated with lower mortality: A nationwide study in Finland. J Bone Miner Res. 2011;26(8):1845–1853. doi: 10.1002/jbmr.375. [DOI] [PubMed] [Google Scholar]
- 11.Bolland MJ, Grey AB, Gamble GD, Reid IR. Effect of osteoporosis treatment on mortality: a meta-analysis. J Clin Endocrinol Metab. 2010;95(3):1174–1181. doi: 10.1210/jc.2009-0852. [DOI] [PubMed] [Google Scholar]
- 12.Curtis JR, Carbone L, Cheng H, et al. Longitudinal trends in use of bone mass measurement among older Americans, 1999–2005. J Bone Miner Res. 2008;23(7):1061–1067. doi: 10.1359/JBMR.080232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Foley KA, Foster SA, Meadows ES, Baser O, Long SR. Assessment of the clinical management of fragility fractures and implications for the new HEDIS osteoporosis measure. Med Care. 2007;45(9):902–906. doi: 10.1097/MLR.0b013e3180536764. [DOI] [PubMed] [Google Scholar]
- 14.McCombs JS, Thiebaud P, Laughlin-Miley C, Shi J. Compliance with drug therapies for the treatment and prevention of osteoporosis. Maturitas. 2004;48(3):271–287. doi: 10.1016/j.maturitas.2004.02.005. [DOI] [PubMed] [Google Scholar]
- 15.Caro JJ, Ishak KJ, Huybrechts KF, Raggio G, Naujoks C. The impact of compliance with osteoporosis therapy on fracture rates in actual practice. Osteoporos Int. 2004;15(12):1003–1008. doi: 10.1007/s00198-004-1652-z. [DOI] [PubMed] [Google Scholar]
- 16.McHorney CA, Schousboe JT, Cline RR, Weiss TW. The impact of osteoporosis medication beliefs and side-effect experiences on non-adherence to oral bisphosphonates. Curr Med Res Opin. 2007;23(12):3137–3152. doi: 10.1185/030079907X242890. [DOI] [PubMed] [Google Scholar]
- 17.Lewiecki EM. Risk communication and shared decision making in the care of patients with osteoporosis. J Clin Densitom. 2010;13(4):335–345. doi: 10.1016/j.jocd.2010.06.005. [DOI] [PubMed] [Google Scholar]
- 18.Montori VM, Shah ND, Pencille LJ, et al. Use of a decision aid to improve treatment decisions in osteoporosis: the osteoporosis choice randomized trial. Am J Med. 2011;124(6):549–556. doi: 10.1016/j.amjmed.2011.01.013. [DOI] [PubMed] [Google Scholar]
- 19.Parfitt AM. Osteonal and hemi-osteonal remodeling: the spatial and temporal framework for signal traffic in adult human bone. J Cell Biochem. 1994;55(3):273–286. doi: 10.1002/jcb.240550303. [DOI] [PubMed] [Google Scholar]
- 20.Burr DB. Targeted and nontargeted remodeling. Bone. 2002;30(1):2–4. doi: 10.1016/s8756-3282(01)00619-6. [DOI] [PubMed] [Google Scholar]
- 21.Collin-Osdoby P. Regulation of vascular calcification by osteoclast regulatory factors RANKL and osteoprotegerin. Circ Res. 2004;95(11):1046–1057. doi: 10.1161/01.RES.0000149165.99974.12. [DOI] [PubMed] [Google Scholar]
- 22.Hsu H, Lacey DL, Dunstan CR, et al. Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc Natl Acad Sci U S A. 1999;96(7):3540–3545. doi: 10.1073/pnas.96.7.3540. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Lacey DL, Timms E, Tan HL, et al. Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell. 1998;93(2):165–176. doi: 10.1016/s0092-8674(00)81569-x. [DOI] [PubMed] [Google Scholar]
- 24.Eghbali-Fatourechi G, Khosla S, Sanyal A, Boyle WJ, Lacey DL, Riggs BL. Role of RANK ligand in mediating increased bone resorption in early postmenopausal women. J Clin Invest. 2003;111(8):1221–1230. doi: 10.1172/JCI17215. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Simonet WS, Lacey DL, Dunstan CR, et al. Osteoprotegerin: a novel secreted protein involved in the regulation of bone density. Cell. 1997;89(2):309–319. doi: 10.1016/s0092-8674(00)80209-3. [DOI] [PubMed] [Google Scholar]
- 26.Ross AB, Bateman TA, Kostenuik PJ, et al. The effects of osteoprotegerin on the mechanical properties of rat bone. J Mater Sci Mater Med. 2001;12(7):583–588. doi: 10.1023/a:1011229324412. [DOI] [PubMed] [Google Scholar]
- 27.Bekker PJ, Holloway D, Nakanishi A, Arrighi M, Leese PT, Dunstan CR. The effect of a single dose of osteoprotegerin in postmenopausal women. J Bone Miner Res. 2001;16(2):348–360. doi: 10.1359/jbmr.2001.16.2.348. [DOI] [PubMed] [Google Scholar]
- 28.Bekker PJ, Holloway DL, Rasmussen AS, et al. A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res. 2004;19(7):1059–1066. doi: 10.1359/JBMR.040305. [DOI] [PubMed] [Google Scholar]
- 29.McClung MR, Lewiecki EM, Cohen SB, et al. Denosumab in post-menopausal women with low bone mineral density. N Engl J Med. 2006;354(8):821–831. doi: 10.1056/NEJMoa044459. [DOI] [PubMed] [Google Scholar]
- 30.Lewiecki EM, Miller PD, McClung MR, et al. Two-year treatment with denosumab (AMG 162) in a randomized phase 2 study of post-menopausal women with low bone mineral density. J Bone Miner Res. 2007;22(12):1832–1841. doi: 10.1359/jbmr.070809. [DOI] [PubMed] [Google Scholar]
- 31.Miller PD, Bolognese MA, Lewiecki EM, et al. Effect of denosumab on bone density and turnover in postmenopausal women with low bone mass after long-term continued, discontinued, and restarting of therapy: A randomized blinded phase 2 clinical trial. Bone. 2008;43(2):222–229. doi: 10.1016/j.bone.2008.04.007. [DOI] [PubMed] [Google Scholar]
- 32.Miller PD, Wagman RB, Peacock M, et al. Effect of denosumab on bone mineral density and biochemical markers of bone turnover: six-year results of a phase 2 clinical trial. J Clin Endocrinol Metab. 2011;96(2):394–402. doi: 10.1210/jc.2010-1805. [DOI] [PubMed] [Google Scholar]
- 33.McClung M, Lewiecki EM, Bolognese MA, et al. Effects of denosumab on bone mineral density and biochemical markers of bone turnover: 8-Year results of a phase 2 clinical trial. J Bone Miner Res. 2011;24(Suppl 1):S20. doi: 10.1007/s00198-012-2052-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Cummings SR, San Martin J, McClung MR, et al. Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med. 2009;361(8):756–765. doi: 10.1056/NEJMoa0809493. [DOI] [PubMed] [Google Scholar]
- 35.Bone HG, Bolognese MA, Yuen CK, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women. J Clin Endocrinol Metab. 2008;93(6):2149–2157. doi: 10.1210/jc.2007-2814. [DOI] [PubMed] [Google Scholar]
- 36.Brown JP, Prince RL, Deal C, et al. Comparison 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 trial. J Bone Miner Res. 2009;24(1):153–161. doi: 10.1359/jbmr.0809010. [DOI] [PubMed] [Google Scholar]
- 37.Kendler DL, Roux C, Benhamou CL, et al. Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res. 2010;25(1):72–81. doi: 10.1359/jbmr.090716. [DOI] [PubMed] [Google Scholar]
- 38.Amgen. Amgen medical information [web page on the Internet] Thousand Oaks, CA: Amgen Inc; 2009, 2010. [Accessed September 15, 2011]. Available from: https://www.amgenmedinfo.com/Home. [Google Scholar]
- 39.Amgen. Prolia® Postmarketing Active Safety Surveillance Program [website on the Internet] Thousand Oaks, CA: Amgen Inc; 2011. [Accessed August 26, 2011]. Available from: https://proliasafety.secure.force.com/prolia. [Google Scholar]
- 40.US Food and Drug Administration. MedWatch: The FDA Safety Information and Adverse Event Reporting Program [website on the Internet] Silverspring, MD: FDA; 2011. [Accessed October 4, 2011]. [updated October 3]. Available from: http://www.fda.gov/Safety/MedWatch/default.htm. [Google Scholar]
- 41.Amgen. Prolia® (denosumab) risk evaluation and mitigation strategy [website on the Internet] Thousand Oaks, CA: Amgen Inc; 2011. [Accessed August 26, 2011]. Available from: http://www.proliahcp.com/risk-evaluation-mitigation-strategy/ [Google Scholar]
- 42.Amgen. Prolia® prescribing information. Thousand Oaks, CA: Amgen Inc; [Accessed August 28, 2011]. pp. 2010–2011. Available from: http://pi.amgen.com/united_states/prolia/prolia_pi.pdf. [Google Scholar]
- 43.Amgen. Medication Guide: Prolia®. Thousand Oaks, CA: Amgen Inc; 2011. [Accessed August 28, 2011]. Available from: http://pi.amgen.com/united_states/prolia/prolia_mg.pdf. [Google Scholar]
- 44.Amgen. Important Drug Warning Regarding Prolia® (densoumab). [Dear healthcare professional letter.] Thousand Oaks, CA: Amgen Inc; nd. [Accessed August 28, 2011]. Available from: http://www.proliahcp.com/pdf/dear_healthcare_professional_letter.pdf. [Google Scholar]
- 45.Amgen. Prolia® (denosumab) Injection [homepage on the Internet] Thousand Oaks, CA: Amgen Inc; 2011. [Accessed September 15, 2011]. Available from: http://www.prolia.com/ [Google Scholar]
- 46.US Food and Drug Administration. Investigational new drug safety reporting requirements for human drug and biological products and safety reporting requirements for bioavailability and bioequivalence studies in humans. [Accessed September 2, 2011];Federal Register. 21 CFR Parts 312 and 320. 2010 75(188):59935–59963. Available from: http://frwebgate.access.gpo.gov/cgi-bin/getdoc.cgi?dbname=2010_register&docid=fr29se10-3.pdf. [PubMed] [Google Scholar]
- 47.US Food and Drug Administration. An FDA guide to drug safety terms [web page on the Internet] Silverspring, MD: FDA; 2011. [Accessed October 4, 2011]. [updated September 9]. Available from: http://www.fda.gov/ForConsumers/ConsumerUpdates/ucm107970.htm. [Google Scholar]
- 48.Houghton Miflin Company. The American Heritage Dictionary. 4th ed. Boston, MA, and New York, NY: Houghton Miflin Company; 2000. [Google Scholar]
- 49.US Food and Drug Administration. CFR – Code of Federal Regulations Title 21. Medication Guides for Prescription Drug Products. Silverspring, MD: FDA; 2011. [Accessed October 12, 2011]. [updated April 1]. Available at: http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?CFRPart=208&showFR=1. [Google Scholar]
- 50.US Food an Drug Admistration. CFR – Code of Federal Regulations Title 21: Food and Drugs. Part 201 – Labeling. Subpart B – Labeling Requirements for Prescription Drugs and/or Insulin. Silverspring, MD: FDA; 2011. [Accessed August 30, 2011]. [updated April 1]. Available from: http://www.accessdata.fda.gov/scripts/cdrh/cfdocs/cfcfr/CFRSearch.cfm?fr=201.57. [Google Scholar]
- 51.Rothstein A. Denosumab Safety: FDA Analysis. Silverspring, MD: FDA; nd. [Accessed September 6, 2011]. Available from: http://courses.washington.edu/bonephys/denosumab/Rothstein%20FDA%20deno%20safety.pdf. [Google Scholar]
- 52.Advisory Committee for Reproductive Health Drugs of the Center for Drug Evaluation and Research. Summary Minutes of the Advisory Committee for Reproductive Health Drugs: August 13, 2009. Washington DC North, Githersburg, MD: [Accessed September 6, 2011]. Available from: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ReproductiveHealthDrugsAdvisoryCommittee/UCM249267.pdf. [Google Scholar]
- 53.Dougall WC, Glaccum M, Charrier K, et al. RANK is essential for osteoclast and lymph node development. Genes Dev. 1999;13(18):2412–2424. doi: 10.1101/gad.13.18.2412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Josien R, Wong BR, Li HL, Steinman RM, Choi Y. TRANCE, a TNF family member, is differentially expressed on T cell subsets and induces cytokine production in dendritic cells. J Immunol. 1999;162(5):2562–2568. [PubMed] [Google Scholar]
- 55.Anderson DM, Maraskovsky E, Billingsley WL, et al. A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature. 1997;390(6656):175–179. doi: 10.1038/36593. [DOI] [PubMed] [Google Scholar]
- 56.Seshasayee D, Wang H, Lee WP, et al. A novel in vivo role for osteoprotegerin ligand in activation of monocyte effector function and inflammatory response. J Biol Chem. 2004;279(29):30202–30209. doi: 10.1074/jbc.M403968200. [DOI] [PubMed] [Google Scholar]
- 57.Li Y, Toraldo G, Li A, et al. B cells and T cells are critical for the preservation of bone homeostasis and attainment of peak bone mass in vivo. Blood. 2007;109(9):3839–3848. doi: 10.1182/blood-2006-07-037994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Stolina M, Dwyer D, Ominsky MS, et al. Continuous RANKL inhibition in osteoprotegerin transgenic mice and rats suppresses bone resorption without impairing lymphorganogenesis or functional immune responses. J Immunol. 2007;179(11):7497–7505. doi: 10.4049/jimmunol.179.11.7497. [DOI] [PubMed] [Google Scholar]
- 59.Stolina M, Dwyer D, Morony S, et al. Rats and mice overexpressing soluble OPG have high bone mass but no alteration in immunological parameters or lymphocyte function. Arthritis Rheum. 2005;20:S708. [Google Scholar]
- 60.Barbaroux JB, Beleut M, Brisken C, Mueller CG, Groves RW. Epidermal receptor activator of NF-kappaB ligand controls Langerhans cells numbers and proliferation. J Immunol. 2008;181(2):1103–1108. doi: 10.4049/jimmunol.181.2.1103. [DOI] [PubMed] [Google Scholar]
- 61.Loser K, Mehling A, Loeser S, et al. Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat Med. 2006;12(12):1372–1379. doi: 10.1038/nm1518. [DOI] [PubMed] [Google Scholar]
- 62.Recker RR, Lewiecki EM, Miller PD, Reiffel J. Safety of bisphosphonates in the treatment of osteoporosis. Am J Med. 2009;122(2 Suppl):S22–S32. doi: 10.1016/j.amjmed.2008.12.004. [DOI] [PubMed] [Google Scholar]
- 63.Reid IR, Miller PD, Brown JP, et al. Effects of denosumab on bone histomorphometry: the FREEDOM and STAND studies. J Bone Miner Res. 2010;25(10):2256–2265. doi: 10.1002/jbmr.149. [DOI] [PubMed] [Google Scholar]
- 64.Odvina CV, Zerwekh JE, Rao DS, Maalouf N, Gottschalk FA, Pak CY. Severely suppressed bone turnover: a potential complication of alendronate therapy. J Clin Endocrinol Metab. 2005;90(3):1294–1301. doi: 10.1210/jc.2004-0952. [DOI] [PubMed] [Google Scholar]
- 65.Visekruna M, Wilson D, McKiernan FE. Severely suppressed bone turnover and atypical skeletal fragility. J Clin Endocrinol Metab. 2008;93(8):2948–2952. doi: 10.1210/jc.2007-2803. [DOI] [PubMed] [Google Scholar]
- 66.Jorgensen NR, Schwarz P. Effects of anti-osteoporosis medications on fracture healing. Curr Osteoporos Rep. 2011;9(3):149–155. doi: 10.1007/s11914-011-0065-0. [DOI] [PubMed] [Google Scholar]
- 67.Marx RE, Cillo JE, Jr, Ulloa JJ. Oral bisphosphonate-induced osteonecrosis: risk factors, prediction of risk using serum CTX testing, prevention, and treatment. J Oral Maxillofac Surg. 2007;65(12):2397–2410. doi: 10.1016/j.joms.2007.08.003. [DOI] [PubMed] [Google Scholar]
- 68.Rasmussen LM, Tarnow L, Hansen TK, Parving HH, Flyvbjerg A. Plasma osteoprotegerin levels are associated with glycaemic status, systolic blood pressure, kidney function and cardiovascular morbidity in type 1 diabetic patients. Eur J Endocrinol. 2006;154(1):75–81. doi: 10.1530/eje.1.02049. [DOI] [PubMed] [Google Scholar]
- 69.Browner WS, Lui LY, Cummings SR. Associations of serum osteoprotegerin levels with diabetes, stroke, bone density, fractures, and mortality in elderly women. J Clin Endocrinol Metab. 2001;86(2):631–637. doi: 10.1210/jcem.86.2.7192. [DOI] [PubMed] [Google Scholar]
- 70.Kiechl S, Schett G, Wenning G, et al. Osteoprotegerin is a risk factor for progressive atherosclerosis and cardiovascular disease. Circulation. 2004;109(18):2175–2180. doi: 10.1161/01.CIR.0000127957.43874.BB. [DOI] [PubMed] [Google Scholar]
- 71.Nitta K, Akiba T, Uchida K, et al. Serum osteoprotegerin levels and the extent of vascular calcification in haemodialysis patients. Nephrol Dial Transplant. 2004;19(7):1886–1889. doi: 10.1093/ndt/gfh263. [DOI] [PubMed] [Google Scholar]
- 72.Morena M, Terrier N, Jaussent I, et al. Plasma osteoprotegerin is associated with mortality in hemodialysis patients. J Am Soc Nephrol. 2006;17(1):262–270. doi: 10.1681/ASN.2005030260. [DOI] [PubMed] [Google Scholar]
- 73.Kearns AE, Khosla S, Kostenuik PJ. Receptor activator of nuclear factor kappaB ligand and osteoprotegerin regulation of bone remodeling in health and disease. Endocr Rev. 2008;29(2):155–192. doi: 10.1210/er.2007-0014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Black DM, Delmas PD, Eastell R, et al. Once-yearly zoledronic acid for treatment of postmenopausal osteoporosis. N Engl J Med. 2007;356(18):1809–1822. doi: 10.1056/NEJMoa067312. [DOI] [PubMed] [Google Scholar]
- 75.US Food and Drug Administration. Early communication about the ongoing safety review of bisphosphonates [web page on the Internet] Silverspring, MD: FDA; 2010. [Accessed August 11, 2011]. Update of safety review follow-up to the October 1, 2007. [updated March 4]. Available from: http://www.fda.gov/Drugs/DrugSafety/PostmarketDrugSafetyInformationforPatientsandProviders/DrugSafetyInformationforHeathcareProfessionals/ucm136201.htm. [Google Scholar]
- 76.Smith MR, Egerdie B, Hernandez TN, et al. Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med. 2009;361(8):745–755. doi: 10.1056/NEJMoa0809003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Division of Reproductive and Urologic Products, Office of New Drugs, Center for Drug Evaluation and Research, Food and Drug Administration. Denosumab (Proposed trade name: PROLIA), Amgen, Inc. Silverspring, MD: FDA; 2009. [Accessed September 15, 2011]. Background Document for Meeting of Advisory Committee for Reproductive Health Drugs (August 13, 2009) Available from: http://www.fda.gov/downloads/AdvisoryCommittees/CommitteesMeetingMaterials/Drugs/ReproductiveHealthDrugsAdvisoryCommittee/UCM176605.pdf. [Google Scholar]
- 78.Adami S, Palacios S, Pavelka K, et al. FREEDOM trial: denosumab is not associated with fracture healing complications in postmenopausal women with osteoporosis. Osteoporos Int. 2011;22:S243. [Google Scholar]
- 79.Brown JP, Dempster DW, Ding B, et al. Bone remodeling in postmenopausal women who discontinued denosumab treatment: Off-treatment biopsy study. J Bone Miner Res. 2011 doi: 10.1002/jbmr.448. Epub 2011 Oct 20. [DOI] [PubMed] [Google Scholar]
- 80.Watts NB, Roux C, Modlin JF, et al. Infections in postmenopausal women with osteoporosis treated with denosumab or placebo: coincidence or causal association? Osteoporos Int. 2011 doi: 10.1007/s00198-011-1755-2. Epub 2011 Sep 3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Bone HG, Chapurlat R, Libanati C, et al. Safety observations from denosumab long-term extension and cross-over studies in postmenopausal women with osteoporosis. J Bone Miner Res. 2011;26:S22. [Google Scholar]
- 82.Bone HG, III, Chapurlat R, Brandi ML, et al. Denosumab treatment for 5 years of postmenopausal women with osteoporosis: results from the first two years of the FREEDOM trial extension. American Association of Clinical Endocrinologists 20th Annual Meeting and Clinical Congress; 2011. Abstract 503. [Google Scholar]
- 83.von Keyserlingk C, Hopkins R, Anastasilakis A, et al. Clinical efficacy and safety of denosumab in postmenopausal women with low bone mineral density and osteoporosis: a meta-analysis. Semin Arthritis Rheum. 2011;41:178–186. doi: 10.1016/j.semarthrit.2011.03.005. [DOI] [PubMed] [Google Scholar]
- 84.Ellis GK, Bone HG, Chlebowski R, et al. Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancer. J Clin Oncol. 2008;26(30):4875–4882. doi: 10.1200/JCO.2008.16.3832. [DOI] [PubMed] [Google Scholar]
- 85.Fizazi K, Carducci M, Smith M, et al. Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet. 2011;377(9768):813–822. doi: 10.1016/S0140-6736(10)62344-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Henry DH, Costa L, Goldwasser F, et al. Randomized, 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 myeloma. J Clin Oncol. 2011;29(9):1125–1132. doi: 10.1200/JCO.2010.31.3304. [DOI] [PubMed] [Google Scholar]
- 87.Stopeck AT, Lipton A, Body JJ, et al. Denosumab compared with zoledronic acid for the treatment of bone metastases in patients with advanced breast cancer: a randomized, double-blind study. J Clin Oncol. 2010;28(35):5132–5139. doi: 10.1200/JCO.2010.29.7101. [DOI] [PubMed] [Google Scholar]
- 88.Kendler DL, McClung MR, Freemantle N, et al. Adherence, preference, and satisfaction of postmenopausal women taking denosumab or alendronate. Osteoporos Int. 2011;22(6):1725–1735. doi: 10.1007/s00198-010-1378-z. [DOI] [PubMed] [Google Scholar]
- 89.McClung M, Freemantle N, Kaur P, et al. The denosumab adherence, preference, satisfaction (DAPS) study: bone mineral density results at 24 months. J Clin Densitom. 2011;14:159. [Google Scholar]
- 90.National Osteoporosis Foundation. Clinician’s Guide to Prevention and Treatment of Osteoporosis. Washington DC: National Osteoporosis Foundation; 2010. [Accessed October 10, 2011]. Available at: http://www.nof.org/sites/default/files/pdfs/NOF_ClinicianGuide2009_v7.pdf. [Google Scholar]
- 91.Watts NB, Bilezikian JP, Camacho PM, et al. American Association of Clinical Endocrinologists Medical Guidelines for Clinical Practice for the diagnosis and treatment of postmenopausal osteoporosis. Endocr Pract. 2010;16(Suppl 3):1–37. doi: 10.4158/ep.16.s3.1. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Lewiecki EM. The role of risk communication in the care of osteoporosis. Curr Osteoporos Rep. 2011;9(3):141–148. doi: 10.1007/s11914-011-0056-1. [DOI] [PubMed] [Google Scholar]