Skip to main content
Erschienen in: Osteoporosis International 1/2012

Open Access 01.01.2012 | Original Article

Infections in postmenopausal women with osteoporosis treated with denosumab or placebo: coincidence or causal association?

verfasst von: N. B. Watts, C. Roux, J. F. Modlin, J. P. Brown, A. Daniels, S. Jackson, S. Smith, D. J. Zack, L. Zhou, A. Grauer, S. Ferrari

Erschienen in: Osteoporosis International | Ausgabe 1/2012

Abstract

Summary

Serious adverse events of infections that occurred in subjects receiving denosumab or placebo in the Fracture Reduction Evaluation of Denosumab in Osteoporosis every 6 Months (FREEDOM) study were examined in detail. Serious adverse events of infections in denosumab subjects had heterogeneous etiology, with no clear clinical pattern to suggest a relationship to time or duration of exposure to denosumab.

Introduction

Denosumab reduces the risk for new vertebral, hip, and nonvertebral fractures compared with placebo. In the pivotal phase 3 fracture trial (FREEDOM), the overall safety profile and incidence of adverse events including adverse events of infections were similar between groups. Serious adverse events of erysipelas and cellulitis were more frequent in denosumab-treated subjects. In this report, we further evaluate the details of infectious events in FREEDOM to better understand if RANKL inhibition with denosumab influences infection risk.

Methods

FREEDOM was an international multicenter, randomized, double-blind, placebo-controlled study in postmenopausal women with osteoporosis randomly assigned to receive placebo (n = 3,906) or denosumab 60 mg every 6 months (n = 3,902). The incidence of adverse events and serious adverse events categorized within the Medical Dictionary for Regulatory Activities system organ class, “Infections and Infestations,” was compared between the placebo and denosumab groups by body systems and preferred terms. The temporal relationship between occurrence of serious adverse events of infections of interest and administration of denosumab was explored.

Results

Serious adverse events of infections involving the gastrointestinal system, renal and urinary system, ear, and endocarditis were numerically higher in the denosumab group compared with placebo, but the number of events was small. No relationship was observed between serious adverse events of infections and timing of administration or duration of exposure to denosumab.

Conclusions

Serious adverse events of infections that occurred with denosumab treatment had heterogeneous etiology, with no clear clinical pattern to suggest a relationship to time or duration of exposure to denosumab.
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00198-011-1755-2) contains supplementary material, which is available to authorized users.

Introduction

RANKL is recognized as an essential factor in the regulation of bone resorption. By signaling through its receptor RANK, RANKL increases osteoclast formation, differentiation, and activity and prolongs osteoclast survival [16]. In clinical trials, denosumab, a RANKL inhibitor, has demonstrated efficacy to reduce bone resorption, increase bone mineral density (BMD) and strength in both cortical and trabecular bone, and reduce the risk of vertebral, hip, and nonvertebral fractures [711].
In addition to expression in bone, RANKL and RANK are expressed by cells of the immune system including activated T lymphocytes, B cells, and dendritic cells [3, 12, 13], suggesting that immune cells might affect bone homeostasis or that RANKL inhibition might alter immune function. Gene deletion studies in rodents show that complete absence of RANKL or its receptor RANK during embryogenesis leads to absence of lymph nodes and changes in thymus architecture [3, 14]. However, in both RANKL and RANK deletion, dendritic cell and macrophage components were normal. In humans with osteoclast-poor osteopetrosis due to absence of RANKL and complete loss of function, there appears to be minimal, if any, effect on immune system development and function [15].
In studies of genetically modified rodents and in pharmacologic experiments in cynomolgus monkeys, inhibition of RANKL, rather than complete RANKL or RANK ablation, increased BMD but did not appear to have significant consequences on basal immune parameters, generation of T or B cell immune responses, or responses to immunization or other immune challenges [1618]. In five distinct preclinical models of inflammatory arthritis and in a T cell-driven model of inflammatory bowel disease, RANKL inhibition decreased bone resorption while having no effect on parameters of inflammation including local edema, pannus formation, and cytokine and chemokine profiles or histopathologically evaluated gut inflammation [1928].
Some in vitro and in vivo effects of RANKL inhibition on immune function have been noted in rodents with major defects in immune system stimulation mechanisms, such as IL-2 or CD40 depletion [12, 29, 30], which suggests that there may be redundancy within the immune system, with the RANKL pathway having a secondary role [29]. Additionally, it has been postulated that the RANKL–RANK interaction may modify immune responses in specific tissues such as the skin, potentially through an effect on the intensity of the inflammatory response, rather than through an immunosuppressive effect [31, 32].
In a dose-ranging study of denosumab in healthy postmenopausal women, no clinically meaningful differences in overall lymphocyte counts, T cells, or B cells were observed in subjects treated with denosumab [33]. In the phase 3 international, double-blind pivotal trial demonstrating fracture reduction efficacy of denosumab in postmenopausal women with osteoporosis (Fracture Reduction Evaluation of Denosumab in Osteoporosis every 6 Months (FREEDOM)], the overall incidence of adverse events and serious adverse events was similar between denosumab- and placebo-treated subjects; however, some numeric imbalances in specific events were reported, including serious adverse events of infections involving the skin [8]. To better understand the potential influence of RANKL inhibition on infections, we examined the incidence and types of infections as well as details of individual cases among participants in the pivotal phase 3 denosumab fracture trial, which represents 10,826 patient-years of exposure to denosumab.

Materials and methods

Subjects and database

Adverse events and serious adverse events of infections as reported in the denosumab pivotal phase 3 fracture trial were examined. The study design and primary results of the study have been previously reported [8]. Briefly, it was a 3-year multicenter, international, randomized, double-blind, placebo-controlled study in 7,808 postmenopausal women with osteoporosis. Subjects received placebo or denosumab subcutaneously 60 mg every 6 months (Q6M). The study was conducted in accordance with the Declaration of Helsinki, and the protocol was approved by an institutional review board or ethics committee for each study site. All subjects provided written informed consent.
Safety was assessed through adverse event reporting for all women who received at least one dose of investigational product (3,876 placebo and 3,886 denosumab). Information about adverse events was collected by investigators at each study visit. The investigator’s verbatim description of an adverse event was converted into standardized terminology based on the Medical Dictionary for Regulatory Activities (MedDRA) version 11 and entered in the safety database as preferred terms. Adverse events and serious adverse events were defined according to regulatory criteria: an adverse event was defined as any untoward medical occurrence in a clinical investigation subject administered a pharmaceutical product and which does not necessarily have a causal relationship with this treatment. A serious adverse event was defined as any adverse event that resulted in any of the following outcomes: death, life-threatening adverse drug experience, inpatient hospitalization or prolongation of existing hospitalization, persistent or significant disability/incapacity, or a congenital anomaly/birth defect. In addition, any event that did not meet the regulatory definition of a serious adverse event, but in the opinion of the investigator or sponsor represented a significant medical hazard, was also considered a serious adverse event.
Adverse events and serious adverse events of infections—those adverse events categorized in the MedDRA system organ class “Infections and Infestations”—were evaluated for this report. This category is broad and includes contagious as well as noncontagious (e.g., appendicitis, cholecystitis, diverticulitis) events.
Information about antibiotic treatments was obtained from case narratives and/or concomitant medication listings. Microbial classification (bacterial, viral, or fungal) could only be determined if cultures were collected at the time of event and culture results were reported by the investigators. Microbial classification was listed as unknown if cultures were not collected at the time of event, no organisms were isolated upon culture, or no culture results were reported.
Serious adverse events of opportunistic infections were identified by a search of the clinical trial safety database using predefined MedDRA terms that included fungal and mycobacterial infections. The presence of an organism by itself was not sufficient to qualify an adverse event as a serious opportunistic infection; events needed to meet the regulatory definition of serious (described above) and were verified by medical review. Colonization or localized infections were distinguished from invasive or disseminated infections. For example, shingles confined to a single dermatome would not be considered opportunistic, but herpes zoster infection that was disseminated or involved multiple dermatomes would be included. Queries were generated by the sponsor to obtain additional information from investigators if important case-level detail was missing.

Statistical analysis

Demographic data for all randomized subjects were summarized by treatment group. Safety data were summarized by actual treatment received. Thus, seven subjects assigned to placebo who received a single dose of denosumab at some point during the study were included in the denosumab group for purposes of safety assessments. Yearly incidence rates of serious adverse events of infection were calculated. The temporal relationship between occurrence and resolution of serious adverse events of infections of interest and administration of investigational product was explored. P values were based on the log-rank test. The analyses did not include any adjustments for multiplicity and should be considered exploratory.

Results

Baseline characteristics of subjects enrolled in the pivotal phase 3 fracture trial have been previously reported [8]. Subjects were primarily Caucasian (93%); the mean (SD) age was 72.3 (5.2) years and 74% were 70 years of age or older. As previously reported, the overall incidence of adverse events of infections was similar between the placebo and denosumab groups (54.4% vs 52.9%, respectively; P = 0.17), and serious adverse events of infections were reported in 3.4% of placebo subjects and 4.1% of denosumab subjects (P = 0.14) [8].
About 40% of the serious adverse events of infection (41.3% with placebo and 44.7% with denosumab) were of mild or moderate severity, although they met the regulatory definition of “serious adverse events.” Usually, the “serious” definition was applied due to hospitalization of the subject. The number of subjects discontinuing the study as a result of adverse events of infection was low and similar between treatment groups (four placebo, three denosumab; Table 1). No increased risk for fatal infections was observed with denosumab (six placebo, six denosumab; Table 1).
Table 1
Summary of adverse events and serious adverse events of infection
 
Placebo (N = 3,876), n (%)
Denosumab (N = 3,886), n (%)
P value
Adverse events of infections
2,108 (54.4)
2,055 (52.9)
0.1721
Serious adverse events of infection
133 (3.4)
159 (4.1)
0.1399
Serious opportunistic infection
3 (<0.1)
4 (0.1)
0.7130
AEs of infection leading to study discontinuation
4 (0.1)
3 (<0.1)
0.6979
Fatal infections
6 (0.2)
6 (0.2)
0.9787

Serious adverse events of infections over time

The incidence of serious adverse events of infection across the 3 years of study was examined. The rate of infection did not change with increasing duration of denosumab exposure (Table 2). The rates of known bacterial, viral, and fungal infections also did not increase with duration of denosumab exposure (Table 2).
Table 2
Incidence of serious adverse events of infections by year of study and microbial classification
 
Year 1
Year 2
Year 3
Incidence of serious adverse events of infection by year
   
 Placebo
42 (1.1%)
49 (1.3%)
47 (1.4%)
 Denosumab
55 (1.4%)
58 (1.6%)
54 (1.5%)
Positively identified bacterial infections
   
 Placebo
10 (0.3%)
12 (0.3%)
10 (0.3%)
 Denosumab
13 (0.3%)
15 (0.4%)
19 (0.5%)
Positively identified viral infections
   
 Placebo
0 (0.0%)
1 (<0.1%)
5 (0.1%)
 Denosumab
2 (0.1%)
4 (0.1%)
2 (0.1%)
Positively identified fungal infections
   
 Placebo
1 (<0.1%)
0 (0.0%)
0 (0.0%)
 Denosumab
1 (<0.1%)
0 (0.0%)
1 (<0.1%)

Opportunistic infections

Serious adverse events of opportunistic infections were prospectively identified as events of interest. The incidence of serious adverse events of opportunistic infections was low and similar in the placebo (three [<0.1%]) and denosumab (four [0.1%]) groups [8]. No clear pattern in the type of infections was observed. In the placebo group, all three subjects had tuberculosis (preferred terms of tuberculosis or pulmonary tuberculosis) and one event was fatal. In the denosumab group, the opportunistic infections were tuberculosis (two subjects), aspergillosis of the face, and disseminated herpes zoster. No risk factors for the opportunistic infections were identified in these subjects. No temporal relationship was observed between the occurrence of these opportunistic infections and administration of the investigational product (Fig. 1a). Nonserious adverse events of opportunistic infections were not specifically identified and categorized as such, but individual terms included tuberculosis, which was reported as a nonserious adverse event in four subjects receiving placebo and no subjects receiving denosumab.

Skin infections

Serious adverse events of infections involving the skin occurred in 3 (<0.1%) placebo subjects and 15 (0.4%) denosumab subjects (P < 0.05; Table 3). These were not injection-site reactions. In the denosumab group, most of these skin infections were cellulitis or clinically diagnosed erysipelas involving the lower extremities that resolved with administration of common antibiotics. The overall incidence of adverse events of cellulitis and erysipelas (i.e., both serious and nonserious adverse events) was not significantly different between treatment groups (0.9% placebo, 1.2% denosumab) [8]. There was no temporal association between the onset of serious adverse events of cellulitis and erysipelas and duration of treatment or time since last dose of investigational product (Fig. 1b).
Table 3
Incidence of serious adverse events of skin infection
 
Placebo (N = 3,876)a, n (%)
Denosumab (N = 3,886)a, n (%)
Serious adverse events of infection involving the skin
3 (<0.1)
15 (0.4)*
Cellulitis and erysipelas
1 (<0.1)
12 (0.3)b
Skin bacterial infection
0 (0)
2 (<0.1)
Staphylococcal infection
1 (<0.1)
1 (<0.1)
Infected skin ulcer
0 (0)
1 (<0.1)b
Subcutaneous abscess
1 (<0.1)
0 (0)
*P < 0.05 vs placebo
aNumber of subjects who received ≥1 dose of investigational product
bOne subject in the denosumab group experienced events of cellulitis and erysipelas and infected skin ulcer
Cellulitis and erysipelas are usually caused by Streptococcus pyogenes, Staphylococcus aureus, and other gram-positive bacterial infections. In this study, serious adverse events of cellulitis and erysipelas were diagnosed clinically and not usually confirmed by culture. A positive S. pyogenes culture was obtained for 1 of the 12 subjects experiencing a serious adverse event of cellulitis or erysipelas in the denosumab group.
A detailed description of the cases of serious adverse events of cellulitis and erysipelas is provided in Table 4. The median duration of hospitalization for denosumab subjects was 5.5 days (range, 1–17 days), and most subjects responded well to treatment with common antibiotics (Table 4). Preexisting risk factors including venous ulcers and skin wounds were reported in 5 of 12 denosumab subjects reporting serious adverse events of cellulitis and erysipelas.
Table 4
Case descriptions for subjects with serious adverse events of cellulitis and erysipelas
Subject
Age (years)
Study day
Number of days hospitalized
Culture
Event description
Treatment
Denosumab subject 1
82
113
15
No cultures
Erysipelas of the left leg
IV benzylpenicillin, IM amikacin, oral roxithromycin
Denosumab subject 2
84
750
15
No cultures
Bilateral leg cellulitis
IV and oral antibiotics (unspecified)
History of atrial fibrillation, asthma
Denosumab subject 3
79
204
17
No cultures
Severe erysipelas of the left lower extremity
IV cefotaxime
History of venous ulcer, chronic cardiac failure
Denosumab subject 4
71
177
5
No cultures
Cellulitis.
Oral flucloxacillin, metronidazole, and phenoxymethylpenicillin
Subject cut her left foot on thorns while gardening.
Denosumab subject 5
65
∼180 days
<1 day as subject died on day of hospitalization
Streptococcus pyogenes
The subject had a confirmed neuroendocrine carcinoma of pancreas with penetration to spleen and ventricle and presented with a painful, bluish and swollen right leg. Cellulitis was confirmed by culture. At admission, the subject experienced atrial fibrillation and low blood pressure with same-day deterioration to multiorgan failure, severe shock, and death.
IV oxacillin
Denosumab subject 6
66
939
2
No cultures
Cellulitis of the face following possible insect bite
IV vancomycin and cefazolin, oral cephalexin
History of scleroderma and hypothyroidism
Denosumab subject 7
73
39
6
No cultures
Moderate erysipelas of left lower limb. Bilateral phlebitis in both legs, and edema
IV cefalotin, IM penicillin
75
861
6
No cultures
Worsening of infection of right lower limb varicose ulcer.
IV penicillin
76
1,030
3
No cultures
Lower extremity cellulitis
Oral cephalexin
History of bilateral varicose ulceration of lower extremity, stasis dermatitis, thrombophlebitis of the leg, and obesity.
Denosumab subject 8
81
889
10
Klebsiella pneumonia
Erysipelas of right leg
Unspecified antibiotics
Denosumab subject 9
87
952
7
No cultures
Right leg erysipelas
Oral penicillin
Denosumab subject 10
85
369
5
Negative blood cultures
Right lower limb erysipelas
IV cefalotin and oral amoxicillin
History of lower extremity varicose veins and left first toe cellulitis
Denosumab subject 11
87
922
2
Culture results not reported
Erysipelas. Ulcer on the left leg with formation of a progressive swollen red patch associated with local edema and heat although the subject was afebrile.
IV cefalotin and oral cefaclor
Denosumab subject 12
73
1,059
4
Negative cultures
Cellulitis of the left foot
IV cefazolin and ceftriaxone
Placebo subject 1
80
1,072
2
 
Cellulitis of left foot
Oral ciprofloxacin
No subjects in either group discontinued treatment due to skin infection, and in only one subject was a recurrent serious adverse event of skin infection observed (denosumab subject 7; Table 4). This subject had a history of varicose ulceration of a lower extremity before starting the study and experienced serious adverse events of lower left limb erysipelas, lower right limb skin ulcer, and lower right limb cellulitis over the course of the study, with the first event occurring on study day 39. One subject with a confirmed neuroendocrine carcinoma of pancreas experienced a fatal event associated with cellulitis of the right leg; the case was complicated by sepsis, shock, and multiple organ failure (denosumab subject 5; Table 4).

Gastrointestinal infections

Serious adverse events of infections were also examined in more detail according to body system. Serious adverse events of infections involving the gastrointestinal system occurred in 28 (0.7%) placebo subjects and 36 (0.9%) denosumab subjects (Table 5). The preferred terms categorized under the gastrointestinal body system correspond to infections with heterogeneous etiology, and no consistent pattern was observed in the type of infections. For individual preferred terms, the difference between treatment groups was 0.1% or less. The most common events were gastroenteritis, diverticulitis, and appendicitis.
Table 5
Incidence of serious adverse events of infections related to the gastrointestinal, renal and urinary, and ear and labyrinth body systems
 
Placebo (N = 3,876)a, n (%)
Denosumab (N = 3,886)a, n (%)
P value
Serious adverse events of infections related to the gastrointestinal system
28 (0.7)
36 (0.9)
0.3322
 Gastroenteritis
7 (0.2)
9 (0.2)
 Diverticulitis
6 (0.2)
8 (0.2)
 Appendicitis
7 (0.2)
7 (0.2)
 Abdominal abscess
0 (0)
2 (0.1)
 Helicobacter infection
0 (0)
2 (0.1)
 Clostridium difficile colitis
2 (0.1)
1 (<0.1)
 Anal abscess
0 (0)
1 (<0.1)
 Biliary tract infection fungal
0 (0)
1 (<0.1)
 Gastric infection
0 (0)
1 (<0.1)
 Gastroenteritis Escherichia coli
0 (0)
1 (<0.1)
 Gastroenteritis bacterial
0 (0)
1 (<0.1)
 Gastroenteritis rotavirus
0 (0)
1 (<0.1)
 Gastroenteritis viral
0 (0)
1 (<0.1)
 Post procedural infection
0 (0)
1 (<0.1)
 Salmonellosis
2 (0.1)
0 (0)
 Abscess intestinal
1 (<0.1)
0 (0)
 Gastrointestinal infection
1 (<0.1)
0 (0)
 Infected cyst
1 (<0.1)
0 (0)
 Peridiverticular abscess
1 (<0.1)
0 (0)
 Peritoneal abscess
1 (<0.1)
0 (0)
 Typhus
1 (<0.1)
0 (0)
Serious adverse events of infections related to the renal and urinary systems
20 (0.5)
29 (0.7)
0.2105
 Urinary tract infection
10 (0.3)
16 (0.4)
 Cystitis
2 (0.1)
6 (0.2)
 Pyelonephritis
2 (0.1)
5 (0.1)
 Urosepsis
2 (0.1)
1 (<0.1)
 Pyelonephritis acute
1 (<0.1)
1 (<0.1)
 Pyelonephritis chronic
0 (0)
1 (<0.1)
 Escherichia infection
2 (0.1)
0 (0)
 Bacterial pyelonephritis
1 (<0.1)
0 (0)
 Kidney infection
1 (<0.1)
0 (0)
 Renal abscess
1 (<0.1)
0 (0)
Serious adverse events of infections related to the ear and labyrinth systems
0 (0)
5 (0.1)
0.0260
 Labyrinthitis
0 (0)
4 (0.1)
 Otitis media
0 (0)
1 (<0.1)
aNumber of subjects who received ≥1 dose of investigational product
For subjects with serious adverse events of diverticulitis (six placebo, eight denosumab), the median hospital stay was similar between groups, 6 days (range, 1–8 days) for placebo subjects and 4 days (range, 1–15 days) for denosumab subjects. No subject in the placebo group and three subjects in the denosumab group had a history of diverticulitis before entering the study. One denosumab subject experienced two serious adverse events of diverticulitis on study.

Renal and urinary infections

Serious adverse events of infections involving the urinary tract were experienced by 20 (0.5%) placebo subjects and 29 (0.7%) denosumab subjects (Table 5). The most common serious adverse events included urinary tract infection, cystitis, and pyelonephritis. Culture results indicated these were typically due to Escherichia coli and other common gram-negative bacteria. The difference in incidence between treatment groups for individual preferred terms was 0.1% or less.

Ear infections

Serious adverse events of infections involving the ear occurred in no placebo subjects and five denosumab subjects (Table 5). These infections were primarily labyrinthitis (four cases), of which two cases were moderate and two were severe; the other serious adverse event was otitis media. Resolution of labyrinthitis occurred within 2 and 13 days in cases of moderate severity and in 6 weeks in a severe case. In one subject with a history of recurrent labyrinthitis, the event was ongoing. No apparent relationship was observed between onset of the events and time since initiation of denosumab (range, 6–31 months).
Most subjects with serious adverse events of ear infections had preexisting complicating factors. For example, three of the four subjects with labyrinthitis had a prior history of labyrinthitis. The subject with otitis media had a previous stapedectomy and tympanoplasty in the same ear approximately 3 years prior. She was hospitalized for an exploratory tympanoplasty.

Endocarditis

Three events of endocarditis (one adverse event and two serious adverse events) were reported in the denosumab group and none in the placebo group. No relationship was observed between the onset of endocarditis and the duration of treatment or time since last dose of denosumab (Fig. 1c), and a causative pathogen was not identified in any case. Two of the subjects underwent echocardiography and the diagnosis was reported to be confirmed. One of these subjects was hospitalized for treatment with antibiotics and the other was treated as an outpatient. In the third subject, acute bacterial endocarditis was suspected as a probable contributor to a fatal event of multiorgan failure; no treatment details from the case were available, including echocardiography, and an autopsy was not performed.

Respiratory, mediastinal, and other thoracic infections

Serious adverse events of infections involving the respiratory tract occurred in 68 (1.8%) placebo subjects and 69 (1.8%) denosumab subjects (Supplementary Table 1). Incidence of individual preferred terms was similar between groups.

Osteomyelitis

One subject in each treatment group experienced a nonserious adverse event of osteomyelitis of the jaw. Both cases were adjudicated negative for osteonecrosis of the jaw. The denosumab subject received only one dose of denosumab on study; the event occurred 2 years after denosumab administration.

Peripheral white blood cell counts

Neutrophil, lymphocyte, and monocyte counts were similar between the placebo and denosumab groups throughout the study (Supplementary Fig. 1). Cell counts did not change with increased duration of denosumab exposure.

Discussion

This study examined the incidence, types, and details in individual subjects of adverse events of infections observed in postmenopausal women treated with the RANKL inhibitor denosumab or placebo in the phase 3 pivotal fracture trial, which represents more than 10,000 patient-years of denosumab exposure. The overall incidence of infections was similar between treatment groups. No increased risk of opportunistic infection was seen with denosumab.
Serious adverse events of cellulitis and erysipelas resulting in hospitalization occurred more frequently with denosumab, although the number of events was low. Hospitalized subjects responded to treatment with common antibiotics. No significant increase in overall incidence (serious and nonserious adverse events) of cellulitis and erysipelas was observed with denosumab. With the small numbers of subjects, the finding of more hospitalizations in the denosumab group might be due to chance or could indicate that skin infections were more severe with denosumab treatment. Preclinical data suggest another possibility: inhibition of RANKL in keratinocytes may decrease the number of regulatory T cells (cells that suppress immune responses), leading to an increased inflammatory response in the skin [31, 32]. Thus, it may be that the appearance of the skin lesions was suggestive of greater severity of the inflammatory process in subjects receiving denosumab, resulting in more frequent hospitalization.
When serious adverse events of infections were reviewed according to body systems, events involving the abdomen, urinary tract, and ear, as well as endocarditis, were numerically more frequent in denosumab than placebo subjects, while serious adverse events of infections of the respiratory tract were balanced between treatment groups. The body system groupings were broad and included contagious as well as noncontagious events. In general, when numerical imbalances were reported—for example, ear and labyrinthitis events—subjects had preexisting risk factors for the condition. Given the age of the study population, many had multiple comorbidities including preexisting conditions that may have predisposed to the type to infections observed. Although general medical histories were collected from all subjects at study start, information relating to such potentially predisposing comorbid conditions was not collected systematically. Therefore, we were unable to determine from the available data if the overall baseline level for certain risk factors was similar between groups. Similarly, we could not conclusively investigate whether patients with particular baseline characteristics might be at increased risk to develop certain infections with denosumab.
In denosumab-treated subjects, white blood cell counts remained stable over time and similar to placebo. Serious adverse events of infections that occurred with denosumab had heterogeneous etiology, with no clear clinical pattern to suggest a relationship to time or duration of exposure to denosumab. In aggregate, these findings are consistent with the evidence that suggests there is a redundancy of function in the adult immune system, with RANKL playing a minimal role [34] and inhibition of RANKL having little or no adverse effect in this regard.
Denosumab safety has been evaluated across the clinical development program. In a small phase 3 trial comparing denosumab and placebo in a younger population (mean age, 59 years) of 332 postmenopausal women with low bone mass, subjects treated with denosumab had significantly more serious adverse events of infections that were associated with hospitalization [7]. The serious adverse events of infections were common infections for the population studied and were treated successfully with standard antibiotics; no pattern was observed in the type of body systems affected. No significantly increased risk of serious adverse events of infections was observed in any other phase 2 and phase 3 clinical trials of denosumab compared with placebo or alendronate in postmenopausal women with low bone mass [7, 3537].
Denosumab has also been studied in other disease populations. No increased risk of infection with denosumab (60 or 180 mg Q6M) was noted in clinical trials of patients with rheumatoid arthritis receiving methotrexate or in patients receiving hormone ablation therapy for breast or prostate cancer (denosumab 60 mg Q6M) [3840]. Similarly, no increased risk of infection was observed for a higher dose of denosumab (120 mg every 4 weeks) compared with zoledronic acid in several large trials in patients with advanced cancer or multiple myeloma and bone metastases [4143].
In this analysis, we endeavored to develop a better understanding of the effects of RANKL inhibition with denosumab by evaluating infectious events in postmenopausal women with osteoporosis participating in the phase 3 pivotal fracture trial. Although this study represents the largest placebo-controlled trial with denosumab, it was not designed for evaluating statistical differences between groups of subjects who experienced adverse events where the number of events within a preferred term was low. The analysis of adverse events reported in a clinical trial relies on the mapping of investigator-provided terms for diagnoses to standardized terminology using a coding dictionary (MedDRA). This process can introduce a categorization bias when verbatim terms are grouped together into preferred terms based upon the judgment of the coding personnel. When these data are evaluated in aggregate, diagnostic subtlety may be lost, thus, apparent differences in outcome may reflect the lumping of verbatim terms into MedDRA categories as well as actual differences in the data.
The benefit/risk profile of denosumab continues to be evaluated in ongoing clinical trials, including an open-label extension of the phase 3 pivotal fracture trial that is planned to follow up subjects for up to 10 years. Over the first 3 years (reported here), there is no indication that inhibition of RANKL has any effect on defense mechanisms against infection. A preliminary report indicates that the safety profile of denosumab remains consistent over 5 years of treatment, with no evidence of an increase in the rate of infectious events over time [44].

Acknowledgements

Funding for this study was provided by Amgen. Holly Brenza Zoog, Ph.D., of Amgen provided medical writing support.

Conflicts of interest

N.B. Watts is a co-founder, stockholder, and director of OsteoDynamics, OSMB member for an NIH-sponsored study, and consultant for Amgen, Baxter, Bristol-Myers Squibb, Imagepace, Lilly, Medpace, Merck, Orexigen, and Pfizer/Wyeth. He also received grants (money to institution) from Amgen, Merck, and NPS, speaker fees from Amgen, Lilly, Novartis, and Warner Chilcott and payment for development of educational programs from Amgen.
C. Roux is a member of advisory boards and a consultant for Amgen, MSD, and Novartis. He also received grants (money to institution) from Amgen, MSD, and Novartis, speaker fees from Amgen and MSD, and travel support and review activity fees from Amgen.
J.F. Modlin is a consultant for and has received travel support from Amgen.
J.P. Brown is a member of the advisory board for Amgen, Eli Lilly, Novartis, and Warner-Chilcott and a consultant for Amgen, Eli Lilly, and Merck. He provided expert witness testimony for Merck. He also received grants (money to institution) from Abbott, Amgen, BMS, Eli Lilly, Merck, Novartis, Pfizer, Roche, Sanofi-Aventis, Servier, and Warner-Chilcott and speaker fees from Amgen, Eli Lilly, Merck, and Novartis.
A. Daniels, S. Jackson, S. Smith, D.J. Zack, L. Zhou, and A. Grauer are employees and shareholders of Amgen.
S. Ferrari is an advisory board member and consultant for Amgen. He also received grants (money to institution), lecture fees, payment for development of educational presentation, and travel support from Amgen.

Open Access

This article is distributed under the terms of the Creative Commons Attribution Noncommercial License which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.
Open AccessThis is an open access article distributed under the terms of the Creative Commons Attribution Noncommercial License (https://​creativecommons.​org/​licenses/​by-nc/​2.​0), which permits any noncommercial use, distribution, and reproduction in any medium, provided the original author(s) and source are credited.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

Literatur
1.
Zurück zum Zitat Dougall WC, Glaccum M, Charrier K, Rohrbach K, Brasel K, De Smedt T, Daro E, Smith J, Tometsko ME, Maliszewski CR, Armstrong A, Shen V, Bain S, Cosman D, Anderson D, Morrissey PJ, Peschon JJ, Schuh J (1999) RANK is essential for osteoclast and lymph node development. Genes Dev 13:2412–2424PubMedCrossRef Dougall WC, Glaccum M, Charrier K, Rohrbach K, Brasel K, De Smedt T, Daro E, Smith J, Tometsko ME, Maliszewski CR, Armstrong A, Shen V, Bain S, Cosman D, Anderson D, Morrissey PJ, Peschon JJ, Schuh J (1999) RANK is essential for osteoclast and lymph node development. Genes Dev 13:2412–2424PubMedCrossRef
2.
Zurück zum Zitat Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero A, Timms E, Tan HL, Elliott G, Kelley MJ, Sarosi I, Wang L, Xia XZ, Elliott R, Chiu L, Black T, Scully S, Capparelli C, Morony S, Shimamoto G, Bass MB, Boyle WJ (1999) Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc Natl Acad Sci U S A 96:3540–3545PubMedCrossRef Hsu H, Lacey DL, Dunstan CR, Solovyev I, Colombero A, Timms E, Tan HL, Elliott G, Kelley MJ, Sarosi I, Wang L, Xia XZ, Elliott R, Chiu L, Black T, Scully S, Capparelli C, Morony S, Shimamoto G, Bass MB, Boyle WJ (1999) Tumor necrosis factor receptor family member RANK mediates osteoclast differentiation and activation induced by osteoprotegerin ligand. Proc Natl Acad Sci U S A 96:3540–3545PubMedCrossRef
3.
Zurück zum Zitat Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Morony S, Oliveira-dos-Santos AJ, Van G, Itie A, Khoo W, Wakeham A, Dunstan CR, Lacey DL, Mak TW, Boyle WJ, Penninger JM (1999) OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397:315–323PubMedCrossRef Kong YY, Yoshida H, Sarosi I, Tan HL, Timms E, Capparelli C, Morony S, Oliveira-dos-Santos AJ, Van G, Itie A, Khoo W, Wakeham A, Dunstan CR, Lacey DL, Mak TW, Boyle WJ, Penninger JM (1999) OPGL is a key regulator of osteoclastogenesis, lymphocyte development and lymph-node organogenesis. Nature 397:315–323PubMedCrossRef
4.
Zurück zum Zitat Lacey DL, Tan HL, Lu J, Kaufman S, Van G, Qiu W, Rattan A, Scully S, Fletcher F, Juan T, Kelley M, Burgess TL, Boyle WJ, Polverino AJ (2000) Osteoprotegerin ligand modulates murine osteoclast survival in vitro and in vivo. Am J Pathol 157:435–448PubMedCrossRef Lacey DL, Tan HL, Lu J, Kaufman S, Van G, Qiu W, Rattan A, Scully S, Fletcher F, Juan T, Kelley M, Burgess TL, Boyle WJ, Polverino AJ (2000) Osteoprotegerin ligand modulates murine osteoclast survival in vitro and in vivo. Am J Pathol 157:435–448PubMedCrossRef
5.
Zurück zum Zitat Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ (1998) Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93:165–176PubMedCrossRef Lacey DL, Timms E, Tan HL, Kelley MJ, Dunstan CR, Burgess T, Elliott R, Colombero A, Elliott G, Scully S, Hsu H, Sullivan J, Hawkins N, Davy E, Capparelli C, Eli A, Qian YX, Kaufman S, Sarosi I, Shalhoub V, Senaldi G, Guo J, Delaney J, Boyle WJ (1998) Osteoprotegerin ligand is a cytokine that regulates osteoclast differentiation and activation. Cell 93:165–176PubMedCrossRef
6.
Zurück zum Zitat Nakagawa N, Kinosaki M, Yamaguchi K, Shima N, Yasuda H, Yano K, Morinaga T, Higashio K (1998) RANK is the essential signaling receptor for osteoclast differentiation factor in osteoclastogenesis. Biochem Biophys Res Commun 253:395–400PubMedCrossRef Nakagawa N, Kinosaki M, Yamaguchi K, Shima N, Yasuda H, Yano K, Morinaga T, Higashio K (1998) RANK is the essential signaling receptor for osteoclast differentiation factor in osteoclastogenesis. Biochem Biophys Res Commun 253:395–400PubMedCrossRef
7.
Zurück zum Zitat Bone HG, Bolognese MA, Yuen CK, Kendler DL, Wang H, Liu Y, San Martin J (2008) Effects of denosumab on bone mineral density and bone turnover in postmenopausal women. J Clin Endocrinol Metab 93:2149–2157PubMedCrossRef Bone HG, Bolognese MA, Yuen CK, Kendler DL, Wang H, Liu Y, San Martin J (2008) Effects of denosumab on bone mineral density and bone turnover in postmenopausal women. J Clin Endocrinol Metab 93:2149–2157PubMedCrossRef
8.
Zurück zum Zitat Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, Christiansen C (2009) Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 361:756–765PubMedCrossRef Cummings SR, San Martin J, McClung MR, Siris ES, Eastell R, Reid IR, Delmas P, Zoog HB, Austin M, Wang A, Kutilek S, Adami S, Zanchetta J, Libanati C, Siddhanti S, Christiansen C (2009) Denosumab for prevention of fractures in postmenopausal women with osteoporosis. N Engl J Med 361:756–765PubMedCrossRef
9.
Zurück zum Zitat Genant HK, Engelke K, Hanley DA, Brown JP, Omizo M, Bone HG, Kivitz AJ, Fuerst T, Wang H, Austin M, Libanati C (2010) Denosumab improves density and strength parameters as measured by QCT of the radius in postmenopausal women with low bone mineral density. Bone 47:131–139PubMedCrossRef Genant HK, Engelke K, Hanley DA, Brown JP, Omizo M, Bone HG, Kivitz AJ, Fuerst T, Wang H, Austin M, Libanati C (2010) Denosumab improves density and strength parameters as measured by QCT of the radius in postmenopausal women with low bone mineral density. Bone 47:131–139PubMedCrossRef
10.
Zurück zum Zitat McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, Peacock M, Miller PD, Lederman SN, Chesnut CH, Lain D, Kivitz AJ, Holloway DL, Zhang C, Peterson MC, Bekker PJ (2006) Denosumab in postmenopausal women with low bone mineral density. N Engl J Med 354:821–831PubMedCrossRef McClung MR, Lewiecki EM, Cohen SB, Bolognese MA, Woodson GC, Moffett AH, Peacock M, Miller PD, Lederman SN, Chesnut CH, Lain D, Kivitz AJ, Holloway DL, Zhang C, Peterson MC, Bekker PJ (2006) Denosumab in postmenopausal women with low bone mineral density. N Engl J Med 354:821–831PubMedCrossRef
11.
Zurück zum Zitat Seeman E, Delmas PD, Hanley DA, Sellmeyer D, Cheung AM, Shane E, Kearns A, Thomas T, Boyd SK, Boutroy S, Bogado C, Majumdar S, Fan M, Libanati C, Zanchetta J (2010) Microarchitectural deterioration of cortical and trabecular bone: differing effects of denosumab and alendronate. J Bone Miner Res 25:1886–1894PubMedCrossRef Seeman E, Delmas PD, Hanley DA, Sellmeyer D, Cheung AM, Shane E, Kearns A, Thomas T, Boyd SK, Boutroy S, Bogado C, Majumdar S, Fan M, Libanati C, Zanchetta J (2010) Microarchitectural deterioration of cortical and trabecular bone: differing effects of denosumab and alendronate. J Bone Miner Res 25:1886–1894PubMedCrossRef
12.
Zurück zum Zitat Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, Tometsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, Galibert L (1997) A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 390:175–179PubMedCrossRef Anderson DM, Maraskovsky E, Billingsley WL, Dougall WC, Tometsko ME, Roux ER, Teepe MC, DuBose RF, Cosman D, Galibert L (1997) A homologue of the TNF receptor and its ligand enhance T-cell growth and dendritic-cell function. Nature 390:175–179PubMedCrossRef
13.
Zurück zum Zitat Bachmann MF, Wong BR, Josien R, Steinman RM, Oxenius A, Choi Y (1999) TRANCE, a tumor necrosis factor family member critical for CD40 ligand-independent T helper cell activation. J Exp Med 189:1025–1031PubMedCrossRef Bachmann MF, Wong BR, Josien R, Steinman RM, Oxenius A, Choi Y (1999) TRANCE, a tumor necrosis factor family member critical for CD40 ligand-independent T helper cell activation. J Exp Med 189:1025–1031PubMedCrossRef
14.
Zurück zum Zitat Li J, Sarosi I, Yan XQ, Morony S, Capparelli C, Tan HL, McCabe S, Elliott R, Scully S, Van G, Kaufman S, Juan SC, Sun Y, Tarpley J, Martin L, Christensen K, McCabe J, Kostenuik P, Hsu H, Fletcher F, Dunstan CR, Lacey DL, Boyle WJ (2000) RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci U S A 97:1566–1571PubMedCrossRef Li J, Sarosi I, Yan XQ, Morony S, Capparelli C, Tan HL, McCabe S, Elliott R, Scully S, Van G, Kaufman S, Juan SC, Sun Y, Tarpley J, Martin L, Christensen K, McCabe J, Kostenuik P, Hsu H, Fletcher F, Dunstan CR, Lacey DL, Boyle WJ (2000) RANK is the intrinsic hematopoietic cell surface receptor that controls osteoclastogenesis and regulation of bone mass and calcium metabolism. Proc Natl Acad Sci U S A 97:1566–1571PubMedCrossRef
15.
Zurück zum Zitat Sobacchi C, Frattini A, Guerrini MM, Abinun M, Pangrazio A, Susani L, Bredius R, Mancini G, Cant A, Bishop N, Grabowski P, Del Fattore A, Messina C, Errigo G, Coxon FP, Scott DI, Teti A, Rogers MJ, Vezzoni P, Villa A, Helfrich MH (2007) Osteoclast-poor human osteopetrosis due to mutations in the gene encoding RANKL. Nat Genet 39:960–962PubMedCrossRef Sobacchi C, Frattini A, Guerrini MM, Abinun M, Pangrazio A, Susani L, Bredius R, Mancini G, Cant A, Bishop N, Grabowski P, Del Fattore A, Messina C, Errigo G, Coxon FP, Scott DI, Teti A, Rogers MJ, Vezzoni P, Villa A, Helfrich MH (2007) Osteoclast-poor human osteopetrosis due to mutations in the gene encoding RANKL. Nat Genet 39:960–962PubMedCrossRef
16.
Zurück zum Zitat Stolina M, Dwyer D, Ominsky MS, Corbin T, Van G, Bolon B, Sarosi I, McCabe J, Zack DJ, Kostenuik P (2007) Continuous RANKL inhibition in osteoprotegerin transgenic mice and rats suppresses bone resorption without impairing lymphorganogenesis or functional immune responses. J Immunol 179:7497–7505PubMed Stolina M, Dwyer D, Ominsky MS, Corbin T, Van G, Bolon B, Sarosi I, McCabe J, Zack DJ, Kostenuik P (2007) Continuous RANKL inhibition in osteoprotegerin transgenic mice and rats suppresses bone resorption without impairing lymphorganogenesis or functional immune responses. J Immunol 179:7497–7505PubMed
17.
Zurück zum Zitat Miller RE, Branstetter D, Armstrong A, Kennedy B, Jones J, Cowan L, Bussiere J, Dougall WC (2007) Receptor activator of NF-kappa B ligand inhibition suppresses bone resorption and hypercalcemia but does not affect host immune responses to influenza infection. J Immunol 179:266–274PubMed Miller RE, Branstetter D, Armstrong A, Kennedy B, Jones J, Cowan L, Bussiere J, Dougall WC (2007) Receptor activator of NF-kappa B ligand inhibition suppresses bone resorption and hypercalcemia but does not affect host immune responses to influenza infection. J Immunol 179:266–274PubMed
18.
Zurück zum Zitat Stolina M, Kostenuik PJ, Dougall WC, Fitzpatrick LA, Zack DJ (2007) RANKL inhibition: from mice to men (and women). Adv Exp Med Biol 602:143–150PubMedCrossRef Stolina M, Kostenuik PJ, Dougall WC, Fitzpatrick LA, Zack DJ (2007) RANKL inhibition: from mice to men (and women). Adv Exp Med Biol 602:143–150PubMedCrossRef
19.
Zurück zum Zitat Stolina M, Guo J, Faggioni R, Brown H, Senaldi G (2003) Regulatory effects of osteoprotegerin on cellular and humoral immune responses. Clin Immunol 109:347–354PubMedCrossRef Stolina M, Guo J, Faggioni R, Brown H, Senaldi G (2003) Regulatory effects of osteoprotegerin on cellular and humoral immune responses. Clin Immunol 109:347–354PubMedCrossRef
20.
Zurück zum Zitat Stolina M, Schett G, Dwyer D, Vonderfecht S, Middleton S, Duryea D, Pacheco E, Van G, Bolon B, Feige U, Zack D, Kostenuik P (2009) RANKL inhibition by osteoprotegerin prevents bone loss without affecting local or systemic inflammation parameters in two rat arthritis models: comparison with anti-TNFalpha or anti-IL-1 therapies. Arthritis Res Ther 11:R187PubMedCrossRef Stolina M, Schett G, Dwyer D, Vonderfecht S, Middleton S, Duryea D, Pacheco E, Van G, Bolon B, Feige U, Zack D, Kostenuik P (2009) RANKL inhibition by osteoprotegerin prevents bone loss without affecting local or systemic inflammation parameters in two rat arthritis models: comparison with anti-TNFalpha or anti-IL-1 therapies. Arthritis Res Ther 11:R187PubMedCrossRef
21.
Zurück zum Zitat Stolina M, Ominsky MS, Smith SY (2008) Long-term denosumab administration had no observed effects on WBC counts, immune parameters, or T-cell-dependent immune response in non-human primates. In 35th European Symposium on Calcified Tissues. European Calcified Tissue Society, Barcelona Stolina M, Ominsky MS, Smith SY (2008) Long-term denosumab administration had no observed effects on WBC counts, immune parameters, or T-cell-dependent immune response in non-human primates. In 35th European Symposium on Calcified Tissues. European Calcified Tissue Society, Barcelona
22.
Zurück zum Zitat Byrne FR, Morony S, Warmington K, Geng Z, Brown HL, Flores SA, Fiorino M, Yin SL, Hill D, Porkess V, Duryea D, Pretorius JK, Adamu S, Manoukian R, Danilenko DM, Sarosi I, Lacey DL, Kostenuik PJ, Senaldi G (2005) CD4+CD45RBHi T cell transfer induced colitis in mice is accompanied by osteopenia which is treatable with recombinant human osteoprotegerin. Gut 54:78–86PubMedCrossRef Byrne FR, Morony S, Warmington K, Geng Z, Brown HL, Flores SA, Fiorino M, Yin SL, Hill D, Porkess V, Duryea D, Pretorius JK, Adamu S, Manoukian R, Danilenko DM, Sarosi I, Lacey DL, Kostenuik PJ, Senaldi G (2005) CD4+CD45RBHi T cell transfer induced colitis in mice is accompanied by osteopenia which is treatable with recombinant human osteoprotegerin. Gut 54:78–86PubMedCrossRef
23.
Zurück zum Zitat Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, Capparelli C, Li J, Elliott R, McCabe S, Wong T, Campagnuolo G, Moran E, Bogoch ER, Van G, Nguyen LT, Ohashi PS, Lacey DL, Fish E, Boyle WJ, Penninger JM (1999) Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature 402:304–309PubMedCrossRef Kong YY, Feige U, Sarosi I, Bolon B, Tafuri A, Morony S, Capparelli C, Li J, Elliott R, McCabe S, Wong T, Campagnuolo G, Moran E, Bogoch ER, Van G, Nguyen LT, Ohashi PS, Lacey DL, Fish E, Boyle WJ, Penninger JM (1999) Activated T cells regulate bone loss and joint destruction in adjuvant arthritis through osteoprotegerin ligand. Nature 402:304–309PubMedCrossRef
24.
Zurück zum Zitat Pettit AR, Ji H, von Stechow D, Muller R, Goldring SR, Choi Y, Benoist C, Gravallese EM (2001) TRANCE/RANKL knockout mice are protected from bone erosion in a serum transfer model of arthritis. Am J Pathol 159:1689–1699PubMedCrossRef Pettit AR, Ji H, von Stechow D, Muller R, Goldring SR, Choi Y, Benoist C, Gravallese EM (2001) TRANCE/RANKL knockout mice are protected from bone erosion in a serum transfer model of arthritis. Am J Pathol 159:1689–1699PubMedCrossRef
25.
Zurück zum Zitat Redlich K, Hayer S, Maier A, Dunstan CR, Tohidast-Akrad M, Lang S, Turk B, Pietschmann P, Woloszczuk W, Haralambous S, Kollias G, Steiner G, Smolen JS, Schett G (2002) Tumor necrosis factor alpha-mediated joint destruction is inhibited by targeting osteoclasts with osteoprotegerin. Arthritis Rheum 46:785–792PubMedCrossRef Redlich K, Hayer S, Maier A, Dunstan CR, Tohidast-Akrad M, Lang S, Turk B, Pietschmann P, Woloszczuk W, Haralambous S, Kollias G, Steiner G, Smolen JS, Schett G (2002) Tumor necrosis factor alpha-mediated joint destruction is inhibited by targeting osteoclasts with osteoprotegerin. Arthritis Rheum 46:785–792PubMedCrossRef
26.
Zurück zum Zitat Romas E, Sims NA, Hards DK, Lindsay M, Quinn JW, Ryan PF, Dunstan CR, Martin TJ, Gillespie MT (2002) Osteoprotegerin reduces osteoclast numbers and prevents bone erosion in collagen-induced arthritis. Am J Pathol 161:1419–1427PubMedCrossRef Romas E, Sims NA, Hards DK, Lindsay M, Quinn JW, Ryan PF, Dunstan CR, Martin TJ, Gillespie MT (2002) Osteoprotegerin reduces osteoclast numbers and prevents bone erosion in collagen-induced arthritis. Am J Pathol 161:1419–1427PubMedCrossRef
27.
Zurück zum Zitat Schett G, Redlich K, Hayer S, Zwerina J, Bolon B, Dunstan C, Gortz B, Schulz A, Bergmeister H, Kollias G, Steiner G, Smolen JS (2003) Osteoprotegerin protects against generalized bone loss in tumor necrosis factor-transgenic mice. Arthritis Rheum 48:2042–2051PubMedCrossRef Schett G, Redlich K, Hayer S, Zwerina J, Bolon B, Dunstan C, Gortz B, Schulz A, Bergmeister H, Kollias G, Steiner G, Smolen JS (2003) Osteoprotegerin protects against generalized bone loss in tumor necrosis factor-transgenic mice. Arthritis Rheum 48:2042–2051PubMedCrossRef
28.
Zurück zum Zitat Zwerina J, Hayer S, Tohidast-Akrad M, Bergmeister H, Redlich K, Feige U, Dunstan C, Kollias G, Steiner G, Smolen J, Schett G (2004) Single and combined inhibition of tumor necrosis factor, interleukin-1, and RANKL pathways in tumor necrosis factor-induced arthritis: effects on synovial inflammation, bone erosion, and cartilage destruction. Arthritis Rheum 50:277–290PubMedCrossRef Zwerina J, Hayer S, Tohidast-Akrad M, Bergmeister H, Redlich K, Feige U, Dunstan C, Kollias G, Steiner G, Smolen J, Schett G (2004) Single and combined inhibition of tumor necrosis factor, interleukin-1, and RANKL pathways in tumor necrosis factor-induced arthritis: effects on synovial inflammation, bone erosion, and cartilage destruction. Arthritis Rheum 50:277–290PubMedCrossRef
29.
Zurück zum Zitat Padigel UM, Kim N, Choi Y, Farrell JP (2003) TRANCE-RANK costimulation is required for IL-12 production and the initiation of a Th1-type response to Leishmania major infection in CD40L-deficient mice. J Immunol 171:5437–5441PubMed Padigel UM, Kim N, Choi Y, Farrell JP (2003) TRANCE-RANK costimulation is required for IL-12 production and the initiation of a Th1-type response to Leishmania major infection in CD40L-deficient mice. J Immunol 171:5437–5441PubMed
30.
Zurück zum Zitat Wong BR, Rho J, Arron J, Robinson E, Orlinick J, Chao M, Kalachikov S, Cayani E, Bartlett FS 3rd, Frankel WN, Lee SY, Choi Y (1997) TRANCE is a novel ligand of the tumor necrosis factor receptor family that activates c-Jun N-terminal kinase in T cells. J Biol Chem 272:25190–25194PubMedCrossRef Wong BR, Rho J, Arron J, Robinson E, Orlinick J, Chao M, Kalachikov S, Cayani E, Bartlett FS 3rd, Frankel WN, Lee SY, Choi Y (1997) TRANCE is a novel ligand of the tumor necrosis factor receptor family that activates c-Jun N-terminal kinase in T cells. J Biol Chem 272:25190–25194PubMedCrossRef
31.
Zurück zum Zitat Barbaroux JB, Beleut M, Brisken C, Mueller CG, Groves RW (2008) Epidermal receptor activator of NF-kappaB ligand controls Langerhans cells numbers and proliferation. J Immunol 181:1103–1108PubMed Barbaroux JB, Beleut M, Brisken C, Mueller CG, Groves RW (2008) Epidermal receptor activator of NF-kappaB ligand controls Langerhans cells numbers and proliferation. J Immunol 181:1103–1108PubMed
32.
Zurück zum Zitat Loser K, Mehling A, Loeser S, Apelt J, Kuhn A, Grabbe S, Schwarz T, Penninger JM, Beissert S (2006) Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat Med 12:1372–1379PubMedCrossRef Loser K, Mehling A, Loeser S, Apelt J, Kuhn A, Grabbe S, Schwarz T, Penninger JM, Beissert S (2006) Epidermal RANKL controls regulatory T-cell numbers via activation of dendritic cells. Nat Med 12:1372–1379PubMedCrossRef
33.
Zurück zum Zitat Bekker PJ, Holloway DL, Rasmussen AS, Murphy R, Martin SW, Leese PT, Holmes GB, Dunstan CR, DePaoli AM (2004) A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 19:1059–1066PubMedCrossRef Bekker PJ, Holloway DL, Rasmussen AS, Murphy R, Martin SW, Leese PT, Holmes GB, Dunstan CR, DePaoli AM (2004) A single-dose placebo-controlled study of AMG 162, a fully human monoclonal antibody to RANKL, in postmenopausal women. J Bone Miner Res 19:1059–1066PubMedCrossRef
34.
Zurück zum Zitat Ferrari-Lacraz S, Ferrari S (2010) Do RANKL inhibitors (denosumab) affect inflammation and immunity? Osteoporos Int 22:435–446PubMedCrossRef Ferrari-Lacraz S, Ferrari S (2010) Do RANKL inhibitors (denosumab) affect inflammation and immunity? Osteoporos Int 22:435–446PubMedCrossRef
35.
Zurück zum Zitat Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, de Gregorio LH, Hadji P, Hofbauer LC, Alvaro-Gracia JM, Wang H, Austin M, Wagman RB, Newmark R, Libanati C, San Martin J, Bone HG (2009) 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 24:153–161PubMedCrossRef Brown JP, Prince RL, Deal C, Recker RR, Kiel DP, de Gregorio LH, Hadji P, Hofbauer LC, Alvaro-Gracia JM, Wang H, Austin M, Wagman RB, Newmark R, Libanati C, San Martin J, Bone HG (2009) 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 24:153–161PubMedCrossRef
36.
Zurück zum Zitat Kendler DL, Roux C, Benhamou CL, Brown JP, Lillestol M, Siddhanti S, Man HS, San Martin J, Bone HG (2010) Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res 25:72–81PubMedCrossRef Kendler DL, Roux C, Benhamou CL, Brown JP, Lillestol M, Siddhanti S, Man HS, San Martin J, Bone HG (2010) Effects of denosumab on bone mineral density and bone turnover in postmenopausal women transitioning from alendronate therapy. J Bone Miner Res 25:72–81PubMedCrossRef
37.
Zurück zum Zitat Miller PD, Bolognese MA, Lewiecki EM, McClung MR, Ding B, Austin M, Liu Y, San Martin J (2008) 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 43:222–229PubMedCrossRef Miller PD, Bolognese MA, Lewiecki EM, McClung MR, Ding B, Austin M, Liu Y, San Martin J (2008) 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 43:222–229PubMedCrossRef
38.
Zurück zum Zitat Cohen SB, Dore RK, Lane NE, Ory PA, Peterfy CG, Sharp JT, van der Heijde D, Zhou L, Tsuji W, Newmark R (2008) Denosumab treatment effects on structural damage, bone mineral density, and bone turnover in rheumatoid arthritis: a twelve-month, multicenter, randomized, double-blind, placebo-controlled, phase II clinical trial. Arthritis Rheum 58:1299–1309PubMedCrossRef Cohen SB, Dore RK, Lane NE, Ory PA, Peterfy CG, Sharp JT, van der Heijde D, Zhou L, Tsuji W, Newmark R (2008) Denosumab treatment effects on structural damage, bone mineral density, and bone turnover in rheumatoid arthritis: a twelve-month, multicenter, randomized, double-blind, placebo-controlled, phase II clinical trial. Arthritis Rheum 58:1299–1309PubMedCrossRef
39.
Zurück zum Zitat Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Smith J, Fan M, Jun S (2008) Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancer. J Clin Oncol 26:4875–4882PubMedCrossRef Ellis GK, Bone HG, Chlebowski R, Paul D, Spadafora S, Smith J, Fan M, Jun S (2008) Randomized trial of denosumab in patients receiving adjuvant aromatase inhibitors for nonmetastatic breast cancer. J Clin Oncol 26:4875–4882PubMedCrossRef
40.
Zurück zum Zitat Smith MR, Egerdie B, Hernandez Toriz N, Feldman R, Tammela TL, Saad F, Heracek J, Szwedowski M, Ke C, Kupic A, Leder BZ, Goessl C (2009) Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 361:745–755PubMedCrossRef Smith MR, Egerdie B, Hernandez Toriz N, Feldman R, Tammela TL, Saad F, Heracek J, Szwedowski M, Ke C, Kupic A, Leder BZ, Goessl C (2009) Denosumab in men receiving androgen-deprivation therapy for prostate cancer. N Engl J Med 361:745–755PubMedCrossRef
41.
Zurück zum Zitat Stopeck AT, Lipton A, Body JJ, Steger GG, Tonkin K, de Boer RH, Lichinitser M, Fujiwara Y, Yardley DA, Viniegra M, Fan M, Jiang Q, Dansey R, Jun S, Braun A (2010) 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 28:5132–5139PubMedCrossRef Stopeck AT, Lipton A, Body JJ, Steger GG, Tonkin K, de Boer RH, Lichinitser M, Fujiwara Y, Yardley DA, Viniegra M, Fan M, Jiang Q, Dansey R, Jun S, Braun A (2010) 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 28:5132–5139PubMedCrossRef
42.
Zurück zum Zitat Henry DH, Costa L, Goldwasser F, Hirsch V, Hungria V, Prausova J, Scagliotti GV, Sleeboom H, Spencer A, Vadhan-Raj S, von Moos R, Willenbacher W, Woll PJ, Wang J, Jang Q, Jun S, Dansey R, Yeh H (2011) 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 29:1125–1132PubMedCrossRef Henry DH, Costa L, Goldwasser F, Hirsch V, Hungria V, Prausova J, Scagliotti GV, Sleeboom H, Spencer A, Vadhan-Raj S, von Moos R, Willenbacher W, Woll PJ, Wang J, Jang Q, Jun S, Dansey R, Yeh H (2011) 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 29:1125–1132PubMedCrossRef
43.
Zurück zum Zitat Fizazi K, Carducci M, Smith M, Damiao R, Brown J, Karsh L, Milecki P, Shore N, Rader M, Wang H, Jiang Q, Tadros S, Dansey R, Goessl C (2011) Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet 377:813–822PubMedCrossRef Fizazi K, Carducci M, Smith M, Damiao R, Brown J, Karsh L, Milecki P, Shore N, Rader M, Wang H, Jiang Q, Tadros S, Dansey R, Goessl C (2011) Denosumab versus zoledronic acid for treatment of bone metastases in men with castration-resistant prostate cancer: a randomised, double-blind study. Lancet 377:813–822PubMedCrossRef
44.
Zurück zum Zitat Papapoulos S, Chapurlat R, Brandi ML, Brown JP, Czerwinski E, Daizadeh NS, Grauer A, Krieg M-A, Libanati C, Man Z, Mellstrom D, Radominski S, Reginster J-Y, Resch H, Roman JA, Roux C, Cummings SR, Bone HG (2011) Five-year denosumab treatment of postmenopausal women with osteoporosis: results from the first two years of the FREEDOM trial extension. Osteoporos Int 22(Suppl 1):S107 Papapoulos S, Chapurlat R, Brandi ML, Brown JP, Czerwinski E, Daizadeh NS, Grauer A, Krieg M-A, Libanati C, Man Z, Mellstrom D, Radominski S, Reginster J-Y, Resch H, Roman JA, Roux C, Cummings SR, Bone HG (2011) Five-year denosumab treatment of postmenopausal women with osteoporosis: results from the first two years of the FREEDOM trial extension. Osteoporos Int 22(Suppl 1):S107
Metadaten
Titel
Infections in postmenopausal women with osteoporosis treated with denosumab or placebo: coincidence or causal association?
verfasst von
N. B. Watts
C. Roux
J. F. Modlin
J. P. Brown
A. Daniels
S. Jackson
S. Smith
D. J. Zack
L. Zhou
A. Grauer
S. Ferrari
Publikationsdatum
01.01.2012
Verlag
Springer-Verlag
Erschienen in
Osteoporosis International / Ausgabe 1/2012
Print ISSN: 0937-941X
Elektronische ISSN: 1433-2965
DOI
https://doi.org/10.1007/s00198-011-1755-2

Weitere Artikel der Ausgabe 1/2012

Osteoporosis International 1/2012 Zur Ausgabe

Arthropedia

Grundlagenwissen der Arthroskopie und Gelenkchirurgie. Erweitert durch Fallbeispiele, Videos und Abbildungen. 
» Jetzt entdecken

Update Orthopädie und Unfallchirurgie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.