Glucocorticoid-Induced Osteoporosis: Pathophysiology and Therapy

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 10

Osteoporos Int (2007) 18:1319–1328

DOI 10.1007/s00198-007-0394-0

REVIEW

Glucocorticoid-induced osteoporosis: pathophysiology


and therapy
E. Canalis & G. Mazziotti & A. Giustina & J. P. Bilezikian

Received: 11 February 2007 / Accepted: 30 April 2007 / Published online: 14 June 2007
# International Osteoporosis Foundation and National Osteoporosis Foundation 2007

Abstract Glucocorticoid-induced osteoporosis (GIO) is the induce the apoptosis of mature osteoblasts and osteocytes.
most common form of secondary osteoporosis. Fractures, These effects lead to a suppression of bone formation, a
which are often asymptomatic, may occur in as many as 30– central feature in the pathogenesis of GIO. Glucocorticoids
50% of patients receiving chronic glucocorticoid therapy. also favor osteoclastogenesis and as a consequence increase
Vertebral fractures occur early after exposure to glucocorti- bone resorption. Bisphosphonates are effective in the
coids, at a time when bone mineral density (BMD) declines prevention and treatment of GIO. Anabolic therapeutic
rapidly. Fractures tend to occur at higher BMD levels than in strategies are under investigation.
women with postmenopausal osteoporosis. In human sub-
jects, the early rapid decline in BMD is followed by a slower Keywords Bone mineral density . Fractures .
progressive decline in BMD. Glucocorticoids have direct Glucocorticoid-induced osteoporosis
and indirect effects on the skeleton. The primary effects are
on osteoblasts and osteocytes. Glucocorticoids impair the
replication, differentiation and function of osteoblasts and Introduction

Synthetic glucocorticoids are used in a wide variety of


disorders, including autoimmune, pulmonary, and gastroin-
This work was supported by Grant AR21707 (E. Canalis) from the testinal diseases, as well as in patients following organ
National Institute of Arthritis and Musculoskeletal and Skin Diseases, transplantation and with malignancies. Although the indi-
Grants DK42424 and DK45227 (E. Canalis), and DK32333 (J.P.
Bilezikian) from the National Institute of Diabetes & Digestive & cations for glucocorticoids in these various conditions are
Kidney Diseases and by MIUR and Centro di Ricerca clear, their use is fraught with a host of potential side
sull’Osteoporosi-University of Brescia/EULO (A. Giustina). effects. One organ system that has the potential to be
E. Canalis profoundly affected by glucocorticoids is the skeleton, and
Saint Francis Hospital and Medical Center, glucocorticoid-induced osteoporosis (GIO) is the most
Hartford, CT 060105, USA common form of secondary osteoporosis [1]. Despite the
E. Canalis
fact that glucocorticoids can cause bone loss and fractures,
University of Connecticut School of Medicine, many patients receiving or initiating long-term glucocorti-
Farmington, CT 06030, USA coid therapy are not evaluated for their skeletal health.
Furthermore, patients often do not receive specific preven-
G. Mazziotti : A. Giustina (*)
Department of Internal Medicine, University of Brescia,
tive or therapeutic agents when indicated [2–4]. New
Brescia, Italy knowledge of the pathophysiological mechanisms underly-
e-mail: a.giustina@libero.it ing GIO has been accompanied by the availability of
effective strategies to prevent and treat GIO. This article
J. P. Bilezikian
Department of Medicine, College of Physicians and Surgeons,
focuses on the cellular aspects of glucocorticoid action in
Columbia University, bone, highlighting the mechanisms that are responsible for
New York, NY 10032, USA bone loss (Fig. 1) [5]. We also review current guidelines
1320 Osteoporos Int (2007) 18:1319–1328

Fig. 1 Diagram showing the


direct and indirect effects of Glucocorticoids
glucocorticoids on bone leading
to glucocorticoid-induced osteo-
porosis and fractures

BONE Neuroendocrine system Calcium Metabolism Muscle

RANKL
⇓ intestinal absorption Proteolysis
OSTEOCYTES OSTEOBLASTS
CSF
OSTEOCLASTS ⇑ Renal excretion of myofibrils
⇓ Function ⇓ Differentiation ⇑ Genesis
⇑ Apoptosis ⇓ Function ⇓ Apoptosis
⇓ GH/IGF-I ⇓ sex steroids
⇑ Apoptosis ⇓ Fibrils

Negative calcium
⇓ bone formation ⇑ bone resorption
balance

⇓ Bone Quality ⇓ Bone Mass

Myopathy

Increased risk of fracture ⇑ risk of falls Muscle weakness

and therapeutic approaches for the prevention and treatment proliferator-activated receptor γ 2 (PPARγ 2), both of which
of GIO. play essential roles in adipogenesis [5, 13, 14]. In
accordance with these observations, thiazolidinediones,
which are known to activate PPARγ 2, inhibit osteoblastic
Direct effects of glucocorticoids on bone cells cell differentiation in murine models. Recent observations
in human subjects indicate that diabetic patients receiving
Osteoblasts thiazolidinediones have a higher incidence of fractures [15].
An additional mechanism by which glucocorticoids
Glucocorticoids decrease the number and the function of inhibit osteoblast cell differentiation is by opposing Wnt/
osteoblasts. These effects lead to a suppression of bone β-catenin signaling [5, 16, 17]. Wnt signaling has emerged
formation, a central feature in the pathogenesis of GIO. as a key regulator of osteoblastogenesis. Wnt uses four
Glucocorticoids decrease the replication of cells of the known signaling pathways, but in skeletal cells the
osteoblastic lineage, reducing the pool of cells that may canonical Wnt/β-catenin signaling pathway operates [18].
differentiate into mature osteoblasts [6, 7]. In addition, In this pathway, when Wnt is absent, β-catenin is
glucocorticoids impair osteoblastic differentiation and mat- phosphorylated by glycogen-synthase kinase-3β (GSK-3β),
uration [5]. Under certain experimental conditions, on the and then degraded by ubiquitination. When Wnt is present,
other hand, glucocorticoids have been reported to favor it binds to specific receptors, called frizzled, and to co-
osteoblastic differentiation [8]. In murine models, basal receptors, low density lipoprotein receptor related proteins
levels of glucocorticoids seem to be required for cortical (LRP)-5 and -6, leading to inhibition of GSK-3β activity.
bone acquisition and osteoblast differentiation [9]. Howev- When GSK-3β is not active, stabilized β-catenin trans-
er, the effects of glucocorticoids to favor osteoblast locates to the nucleus, where it associates with transcription
differentiation seem to be highly dependent on experimen- factors to regulate gene expression [19]. Deletions of either
tal conditions, and do not reflect the loss of cells of the Wnt or β-catenin result in the absence of osteoblastogenesis,
osteoblastic lineage regularly seen following glucocorticoid and increased osteoclastogenesis [19, 20]. The Wnt pathway
exposure [5]. can be inactivated by Dickkopf, an antagonist that prevents
In the presence of glucocorticoids, bone marrow stromal Wnt binding to its receptor complex. Glucocorticoids
cells, the precursors of osteoblasts, do not differentiate or are enhance Dickkopf expression and maintain GSK 3-β in an
directed, instead, toward cells of the adipocytic lineage [10– active state, leading ultimately to the inactivation of β-catenin
12]. Mechanisms involved in this redirection of stromal cells [16, 17, 21].
include induction of nuclear factors of the CCAAT enhancer In addition to inhibiting the differentiation of osteoblasts,
binding protein family and the induction of peroxisome glucocorticoids inhibit the function of the differentiated
Osteoporos Int (2007) 18:1319–1328 1321

mature cells. Glucocorticoids inhibit osteoblast-driven an osteoclastogenic cytokine, and suppress the expression
synthesis of type I collagen, the major component of the of interferon-beta, an inhibitor of osteoclastogenesis [32,
bone extracellular matrix, with a consequent decrease in 33]. Glucocorticoids decrease the apoptosis of mature
bone matrix available for mineralization [5]. The decrease osteoclasts [34]. Consequently, there is increased formation
in type I collagen synthesis occurs by transcriptional and of osteoclasts with a prolonged life span explaining, at the
post-transcriptional mechanisms [22]. cellular level, the enhanced and prolonged bone resorption
Glucocorticoids have pro-apoptotic effects on osteoblasts observed in GIO. The direct effects of glucocorticoids on
and osteocytes due to activation of caspase 3, a common osteoclasts also may contribute to an operational decline in
downstream effector of several apoptotic signaling path- osteoblast function during glucocorticoid exposure [35].
ways [23, 24]. Caspases are synthesized as proenzymes and Although the net effect of glucocorticoids is to enhance
are activated through autocatalysis or a caspase cascade. osteoclast number, osteoclast function may be tempered
Active caspases contribute to apoptosis by cleaving target with cells that no longer spread and resorb mineralized
cellular proteins. Caspase 3 is a key mediator of apoptosis matrix normally. Osteoblast signals that depend upon
and is a common downstream effector of multiple apoptotic normal osteoclast function could, thus, be impaired [35].
signaling pathways [24]. The inhibitory effects of gluco- However, these novel findings have been challenged by
corticoids on osteoblastic cell replication and differentiation studies demonstrating a primary effect of glucocorticoids
and the increased apoptosis of mature osteoblasts, all on cells of the osteoblastic lineage [34]. In accordance with
contribute to the depletion of the osteoblastic cellular pool their effects on bone resorption, glucocorticoids enhance
and decreased bone formation. the expression of selected matrix metalloproteinases
(MMP). Osteoblasts secrete MMP1 or collagenase 1 and
Osteocytes MMP13 or collagenase 3, and both cleave type I collagen
fibrils at neutral pH [36, 37]. Cortisol increases collagenase
Osteocytes serve as mechanosensors, and play a role in the 3 synthesis by post-transcriptional mechanisms, by regulat-
repair of bone microdamage [25]. Loss of osteocytes ing specific cytosolic RNA binding proteins, and their
disrupts the osteocyte-canalicular network resulting in a binding to specific RNA sequences [38]. Glucocorticoids
failure to detect signals that normally stimulate processes may also have effects on bone remodelling at the basic
associated with the replacement of damaged bone [26]. multicellular unit (BMU) level, mainly manifested as a
Disruption of the osteocyte-canalicular network can disrupt reduction in wall width (reduced amount of bone formed
fluid flow within the network adversely affecting the per BMU) [39, 40]. In addition, there is some evidence that
material properties of the surrounding bone, independent increased resorption depth (increased amount of bone
of changes in bone remodeling or architecture [26]. resorbed per BMU) may occur in the early stages of
Glucocorticoids affect the function of osteocytes, by therapy, particularly at high doses of glucocorticoids.
modifying the elastic modulus surrounding osteocytic
lacunae [27]. Glucocorticoids induce the apoptosis of Effects of glucocorticoids on bone cells mediated
osteocytes [23]. As a result, the normal maintenance of by growth factors
bone through this mechanism is impaired and the biome-
chanical properties of bone are compromised [27]. In addition to the direct actions of glucocorticoids on bone
target cells, other effects are mediated by changes in the
Osteoclasts synthesis, receptor binding or binding proteins of growth
factors present in the bone microenvironment. Glucocorticoids
In human subjects, GIO occurs in two phases: a rapid, early influence the expression of insulin-like growth factor
phase in which bone mineral density (BMD) is reduced, (IGF) I. IGF-I increases bone formation and the synthesis
presumably due to excessive bone resorption, and a slower, of type I collagen, and decreases bone collagen degrada-
progressive phase in which BMD declines due to impaired tion and osteoblast apoptosis [41]. Glucocorticoids suppress
bone formation [28]. Osteoclasts are members of the IGF I gene transcription, but do not alter IGF I receptor
monocyte/macrophage family of cells that differentiate number or affinity in osteoblasts [5]. Glucocorticoids
under the influence of two requisite cytokines, namely decrease IGF II receptor number, but the skeletal functions
macrophage colony stimulating factor (M-CSF) and receptor of the IGF II receptor have remained elusive [42]. The
activator of NF-κB ligand (RANK-L) [29]. Glucocorticoids activities of IGFs are regulated by six IGF binding proteins
increase the expression of M-CSF and RANK-L, and (IGFBP), all of which are expressed by the osteoblast [43].
decrease the expression of its soluble decoy receptor, Of these, IGFBP-5 was reported to have anabolic effects for
osteoprotegerin, in stromal and osteoblastic cells [30, 31]. skeletal cells, and its transcription is suppressed by
Glucocorticoids also enhance the expression of Interleukin-6, glucocorticoids [44]. The inhibition of IGFBP-5 synthesis
1322 Osteoporos Int (2007) 18:1319–1328

by glucocorticoids is probably not key to the ultimate effect inhaled corticosteroids, suggesting that inhaled steroids
of glucocorticoids on osteoblastic function, because trans- may alter the synthesis or release of GH [59]. However,
genic mice overexpressing IGFBP-5 exhibit decreased, and the cause or consequence of this effect is not clear, since
not increased bone formation [45]. The effects of gluco- serum levels of cortisol and of IGF-I are not suppressed [59].
corticoids on IGF-I expression by the osteoblast are Glucocorticoids inhibit the release of gonadotropins, and as a
reversed by parathyroid hormone (PTH), an observation result estrogen and testosterone production. This effect of
that may help explain why PTH may be effective in the glucocorticoids on the gonadal axis may be an additional
treatment of GIO [46]. factor playing a role in the pathogenesis of GIO [60].

Indirect effects of glucocorticoids on bone metabolism Pathogenesis of fractures in GIO

Glucocorticoids inhibit calcium absorption from the gastro- Fractures may occur in as many as 30–50% of patients
intestinal tract, by opposing vitamin D actions, and by receiving chronic glucocorticoid therapy [61–63]. They
decreasing the expression of specific calcium channels in the occur more frequently in postmenopausal women and men
duodenum [47]. Renal tubular calcium reabsorption also is at sites enriched in cancellous bone, such as the vertebrae
inhibited by glucocorticoids [1]. As a consequence of these and femoral neck [64, 65]. As with vertebral fractures
effects, secondary hyperparathyroidism could exist in the occurring in post-menopausal osteoporosis, vertebral frac-
context of glucocorticoid use. But a hyperparathyroid state tures associated with glucocorticoid therapy often are
does not explain the bone disorder observed in GIO. Most asymptomatic [63]. When assessed by X-ray-based mor-
patients with GIO do not exhibit serum levels of PTH that phometric measurements of vertebral bodies, 37% of
are frankly elevated. Although vertebral and non-vertebral postmenopausal women on chronic (> 6 months) oral
fractures occur in GIO, this condition is associated with a glucocorticoid therapy sustain one or more vertebral
preferential loss of cancellous bone, whereas hyperparathy- fractures [63]. Vertebral fractures occur early after exposure
roidism, is associated with a preferential loss of cortical to glucocorticoids, at a time when BMD declines rapidly
bone [48, 49]. Moreover, bone histomorphometric analysis [66]. The early rapid loss of bone predisposes to fracture,
demonstrates reduced bone turnover in GIO, in contrast to even in individuals whose T-scores are only in the
the increased bone turnover that characterizes hyperpara- osteopenic range.
thyroidism [1, 28]. These observations indicate that Although fractures can occur early in the course of
hyperparathyroidism does not play a central role in the glucocorticoid therapy, their incidence is also related to the
development of the skeletal manifestations of GIO. Never- dose and duration of glucocorticoid exposure. Doses as low
theless, there may be subtle, but important effects of as 2.5 to 7.5 mg of prednisolone equivalents per day can be
glucocorticoids on the secretory dynamics of PTH, with a associated with a 2.5-fold increase in vertebral fractures,
decrease in the tonic release of PTH and an increase in but the risk is greater at higher doses for prolonged periods
pulsatile bursts of the hormone [50]. In healthy subjects, of time [1, 67]. Following the exposure to prednisone
PTH is secreted by low amplitude and high frequency equivalents of 10 mg daily for longer than 90 days, the risk
pulses superimposed upon tonic secretion. Pulsatile PTH of fractures of the hip and spine is increased by 7- and 17-
secretion may be important for the regulation of the actions fold, respectively [67]. The risk of fracture declines after
of the hormone on bone [51]. Abnormal PTH pulsatility is discontinuation of glucocorticoid therapy [1].
found not only following glucocorticoid exposure, but also The reason for the individual heterogeneity in the
in post-menopausal women and in acromegaly [52, 53]. response to glucocorticoids is not known, but differential
Additionally, glucocorticoids may enhance the sensitivity of responses may be associated with polymorphisms of the
skeletal cells to PTH, by increasing the number and affinity glucocorticoid receptor gene. Glucocorticoid receptor poly-
of PTH receptors [54]. morphisms are associated with differences in BMD and
In addition to the direct effects of glucorticoids on body composition [68–70]. Indeed, body composition and
skeletal IGF-I, glucocorticoids decrease the secretion of risk of fracture during glucocorticoid treatment seem to be
growth hormone (GH) and may alter the systemic GH/IGF- closely related [71]. Another explanation for individual
I axis [55]. However, serum levels of IGF-I are normal in variability in the response of patients exposed to glucocor-
GIO. GH secretion is blunted by glucocorticoids by an ticoids is related to peripheral enzymes that interconvert
increase in hypothalamic somatostatin tone, and GH active and inactive glucocorticoid molecules. 11β-hydrox-
administration could reverse some of the negative effects ysteroid dehydrogenases regulate the interconversion be-
of chronic glucocorticoid treatment on bone [56–58]. tween cortisone and hormonally active cortisol, and play a
Secretion of GH is blunted in asthmatic patients receiving role in the regulation of glucocorticoid activity [72]. Two
Osteoporos Int (2007) 18:1319–1328 1323

11β-hydroxysteroid dehydrogenase enzymes have been may all contribute to bone loss, independent of the use of
described: 11β-hydroxysteroid dehydrogenase type-1 is glucocorticoids [60, 78, 79].
primarily a glucocorticoid activator, converting cortisone A direct relationship between BMD and fracture risk in
to cortisol, and 11β-hydroxysteroid dehydrogenase type II GIO has not been established [80–82]. It is likely to be
is an inhibitor enzyme expressed in mineralocorticoid target different from that established in postmenopausal osteopo-
tissues. The type I enzyme is widely expressed in rosis because fractures in GIO occur at higher BMDs [83]
glucocorticoid target tissues, including bone, and its activity (Fig. 2). This point has to be considered when making
and the potential to generate cortisol from cortisone in treatment decisions in GIO. The Royal College of Physi-
human osteoblasts is increased by glucocorticoids [72–75]. cians recommends a vertebral T-score of −1.5 as the
There seems to be an inverse relationship between 11β- intervention threshold. The American College of Rheuma-
hydroxysteriod dehydrogenase type I activity and osteoblast tology (ACR) recommends a more stringent therapeutic
differentiation [75]. An increase of 11β-hydroxysteriod intervention at a T-score of ≤ −1. These scores are higher
dehydrogenase type I activity occurs with aging, possibly than the treatment threshold T-Scores of −2.0 to −2.5, often
providing an explanation for the enhanced sensitivity of the used in the management guidelines for post-menopausal
elderly to the effects of glucocorticoids on the skeleton [75]. osteoporosis [84]. The reasons for the altered relationship
An important point that is often minimized in discus- between BMD and risk of fracture are complex [85, 86]. In
sions about GIO is that many disorders for which the addition to the rapid decline in BMD that occurs following
glucocorticoids are prescribed are themselves causes of glucocorticoid exposure (the faster the bone loss, the
osteoporosis. One has to take into account, therefore, the greater the risk), other factors influence bone strength and
underlying disease itself along with the use of glucocorticoids fracture risk in GIO. These include the underlying disease
when considering the management of GIO. Inflammatory for which patients receive glucocorticoids, and multiple
bowel disease, rheumatoid arthritis and chronic obstructive cellular events that lead to structural changes in bone [87].
pulmonary disease (COPD), for example, all are associated In GIO, the negative effects of glucocorticoids on osteo-
with bone loss, independent of glucocorticoid treatment blasts and osteocytes affect adversely the architecture of
[76, 77]. The systemic release of inflammatory cytokines, cancellous bone. However, these changes often are not
which affect bone formation and bone resorption seem to translated into a decrease in BMD. Reductions in trabecular
underlie the pathophysiology of the bone loss in these thickness, number, and connectivity cannot be determined
settings [76, 77]. However, there are additional factors that by currently available non-invasive imaging modalities.
may play a role in the bone loss. In inflammatory bowel Newer technologies, such as high resolution peripheral
disease, bone loss may be due, in part, to malabsorption of quantitative computed tomography or micro magnetic
vitamin D, calcium and other nutrients [76]. In COPD, resonance imaging may be helpful in identifying individual
hypoxia, acidosis, reduced physical activity, and smoking fracture risk of patients on glucocorticoids [88].

Fig. 2 Relationship between


bone mineral density (BMD)
at lumbar spine and femoral
neck and incidence of radio-
logical spinal deformities in
post-menopausal women with
glucocorticoid-induced osteopo-
rosis as compared with post-
menopausal osteoporosis (from
Van Staa TP, et al. Arthritis
Rheum. 2003 [ref. 83], with
permission)
1324 Osteoporos Int (2007) 18:1319–1328

Although biochemical markers of bone turnover can be Table 1 Guidelines for prevention and treatment of glucocorticoid-
induced osteoporosis
useful measures of bone remodeling activity and can
predict fracture risk, their value in GIO has not been American College Royal College
established and their levels vary with the stage of the of Rheumatology of Physicians
disease [89]. Following the initial exposure to glucocorti-
Calcium All In vitamin D
coids, there is an increase in biochemical markers of bone
and vitamin D insufficiency
resorption, which is followed by a suppression of markers Bisphosphonates If glucocorticoids If older than
of bone formation and bone resorption [89]. for primary (≥ 5 mg) are 65 yrs and/
In addition to the direct effects of glucocorticoids on prevention planned for > or with history
bone cells, the catabolic effects of glucocorticoids on 3 months* of fragility fracture
muscle may contribute to fracture risk since these steroids Bisphosphonates BMD T-score BMD T-score
cause muscular weakness, which can increase the incidence in secondary < −1.0 <−1.5 or with
prevention a reduction in BMD
of falls. Glucocorticoid-induced myopathy may occur
of > 4% after 1 year
following early exposure to glucocorticoids [90]. Chronic
Sex hormones Hypogonadism Hypogonadism
glucocorticoid-induced myopathy is generally manifested
by weakness particularly of the pelvic girdle musculature *with caution in premenopusal women
and may affect up to 60% of patients treated with
glucocorticoids [60]. The myopathy involves muscle loss
due to glucocorticoid induced proteolysis of myofibrils daily), including lifestyle changes, such as tobacco cessa-
[91]. This is mediated by activation of lysosomal and tion and reduction of alcohol consumption, an exercise
ubiquitin-proteasome enzymes. Recent studies have demon- program, restriction of sodium intake in the presence of
strated that glucocorticoids induce myostatin, a negative hypercalciuria, sufficient calcium intake and adequate
regulator of muscle mass. Deletion of the myostatin gene vitamin D supplementation. ACR guidelines recommend
prevents glucocorticoid induced myofibril proteolysis and that treatment with bisphosphonates should be started in
muscle loss in murine models [91]. This would suggest that these subjects if their T-score is ≤ −1.0, as assessed by
myostatin plays a role in the mechanism of muscular vertebral densitometry [84, 92]. Table 1 shows similarities
atrophy in GIO. and differences between ACR and UK guidelines [84].

Specific therapeutic approaches


Management of GIO
Vitamin D plays an important adjuvant role in the
Guidelines management of GIO [93–97]. Vitamin D increases the
intestinal absorption of calcium and reabsorption of
ACR and the Royal College of Physicians have proposed calcium in the distal renal tubules. In addition to its role
primary prevention and treatment guidelines. Primary in calcium homeostasis, vitamin D plays a function in the
prevention is intervention with a bone-sparing agent at the maintenance of muscular strength and balance. A practical
initiation of (or very soon after) commencement of consideration regarding vitamin D therapy in subjects
glucocorticoid therapy. Secondary prevention is initiation receiving glucocorticoids, relates to vitamin D resistance
of bone protective medication in patients already estab- that occurs in this setting. Rather than maintaining 25-
lished on glucocorticoid therapy (Table 1). The guidelines hydroxyvitamin D levels at an adequate level of 30 ng/ml
advocate the following measures for the prevention and (82 nmoles/L), some experts recommend that the goal be
treatment of GIO: general health awareness, administration set at higher serum levels of 25 hydroxyvitamin D of ≥
of sufficient calcium and vitamin D, reduction of the dose 80 nmol/L. In order to maintain these levels, patients often
of glucocorticoids to a minimum and, when indicated, require amounts of 1,000–2,000 International Units of
therapeutic intervention with bisphosphonates and possibly vitamin D daily [98].
other agents [92]. UK guidelines recommend primary Bisphosphonates are considered to be the pharmacologic
prevention in all men and women over the age of 65 years, gold standard for the prevention and treatment of GIO [84,
in individuals with a history of previous fractures and in 99]. The benefits of bisphosphonates in GIO have been
younger people with BMD T scores of ≤ − 1.5, who are ascribed to their anti-resorptive effect. Bisphosphonates are
committed to at least 3 months of oral glucocorticoid more effective than vitamin D in the prevention of bone
therapy at any dose (Table 1). ACR recommends preventive loss and fractures in GIO, but should be given with
measures in patients exposed to glucocorticoids for longer supplemental calcium and vitamin D [100]. The primary
than 3 months (5 mg prednisone equivalent, or higher, end-point of bisphosphonate trials has been the stabilization
Osteoporos Int (2007) 18:1319–1328 1325

or the increase of BMD, with fracture outcomes as 2. Feldstein AC, Elmer PJ, Nichols GA et al (2005) Practice patterns
in patients at risk for glucocorticoid-induced osteoporosis.
secondary end points. In selected trials, bisphosphonates
Osteoporos Int 16:2168–2174
were shown to decrease the incidence of vertebral fractures 3. Cruse LM, Valeriano J, Vasey FB et al (2006) Prevalence of
in GIO [61, 62, 101–103]. It is advisable to continue evaluation and treatment of glucocorticoid-induced osteoporosis
bisphosphonates for the duration of glucocorticoid treat- in men. J Clin Rheumatol 12:221–225
4. Saag KG, Gelbach SH, Curtis JR et al (2006) Trends in
ment. This recommendation is consistent with data demon-
prevention of glucocorticoid-induced osteoporosis. J Rheumatol
strating that fracture risk is highest during the time of 33:1651–1657
glucocorticoid exposure [104]. The use of bisphosphonates 5. Canalis E (2005) Mechanisms of glucocorticoid action in bone.
in women of childbearing age has to be carefully Curr Osteoporos Rep 3:98–102
6. Canalis E (1984) Effect of cortisol on periosteal and non-
considered, since bisphosphonates have a prolonged half periosteal collagen and DNA synthesis in cultured rat calvariae.
life, and they can cross the placenta with potential Calcif Tissue Int 36:158–166
unwanted effects on fetal skeletal development. 7. Canalis E (1983) Effect of glucocorticoids on type I collagen
In postmenopausal women, estrogens preserve bone mass synthesis, alkaline phosphatase activity, and deoxyribonucleic
acid content in cultured rat calvariae. Endocrinology 112:
whether in the context, or not, of glucocorticoid exposure,
931–939
and they can be a treatment option [105]. However, the use 8. Eijken M, Koedam M, van Driel M et al (2006) The essential
of estrogens has to be carefully considered because of role of glucocorticoids for proper human osteoblast differentia-
unwanted adverse effects. tion and matrix mineralization. Mol Cell Endocrinol 248:87–93
9. Sher LB, Harrison JR, Adams DJ et al (2006) Impaired cortical
An anabolic approach to the treatment of GIO might
bone acquisition and osteoblast differentiation in mice with
have promise since the disorder is primarily one of reduced osteoblast-targeted disruption of glucocorticoid signaling. Calcif
bone formation. PTH is an attractive candidate because it Tissue Int 79:118–125
induces skeletal IGF I and inhibits osteoblast apoptosis, 10. Ito S, Suzuki N, Kato S et al (2007) Glucocorticoids induce the
differentiation of a mesenchymal progenitor cell line, ROB-C26
increasing osteoblast cell number [36, 106–108]. The use of
into adipocytes and osteoblasts, but fail to induce terminal
PTH in GIO has been examined in postmenopausal women osteoblast differentiation. Bone 40:84–92
with rheumatoid arthritis treated with prednisone and 11. Pereira RC, Delany AM, Canalis E (2002) Effects of cortisol and
estrogen replacement [109, 110]. In this population, daily bone morphogenetic protein-2 on stromal cell differentiation:
correlation with CCAAT-enhancer binding protein expression.
treatment with hPTH (1–34) increased vertebral BMD.
Bone 30:685–691
Modest increases in bone mass were observed at the hip. 12. Pereira RC, Delany AM, Canalis E (2004) CCAAT/enhancer
PTH administration induces an initial uncoupling of bone binding protein homologous protein (DDIT3) induces osteoblas-
remodeling with an early increase in bone formation tic cell differentiation. Endocrinology 145:1952–1960
13. Wu Z, Bucher NLR, Farmer SR (1996) Induction of peroxisome
followed by a more gradual increase of bone resorption
proliferator-activated receptor g during the conversion of 3T3
[111]. According to the concept of the “anabolic window”, fibroblasts into adipocytes is mediated by C/EBPh, C/EBPy, and
PTH rapidly and powerfully stimulates osteoblast function glucocorticoids. Mol Cell Biol 16:4128–4136
[112]. PTH is currently being tested in clinical trials to 14. Wedel A, Ziegler-Heitbrock HW (1995) The C/EBP family of
transcription factors. Immunobiology 193:171–185
determine its efficacy in GIO.
15. Schwartz AV, Sellmeyer DE, Vittinghoff E et al (2006)
Thiazolidinedione use and bone loss in older diabetic adults. J
Clin Endocrinol Metab 91:3349–3354
Conclusions 16. Ohnaka K, Tanabe M, Kawate H et al (2005) Glucocorticoid
suppresses the canonical Wnt signal in cultured human osteo-
blasts. Biochem Biophys Res Commun 329:177–181
GIO is a serious skeletal disorder that is associated with 17. Smith E, Frenkel B (2005) Glucocorticoids inhibit the transcrip-
fractures. New knowledge of mechanisms of glucocorticoid tional activity of LEF/TCF in differentiating osteoblasts in a
action has demonstrated detrimental effects on osteoblast glycogen synthase kinase-3beta-dependent and -independent
manner. J Biol Chem 280:2388–2394
and osteocyte function, and prolonged osteoclast life span.
18. Westendorf JJ, Kahler RA, Schroeder TM (2004) Wnt signaling
The extent and rate of bone loss determine the risk of in osteoblasts and bone diseases. Gene 341:19–39
fractures. GIO can be prevented and treated with 19. Glass DA, Bialek P, Ahn JD et al (2005) Canonical Wnt
bisphosphonates. Future therapies may include the use of signaling in differentiated osteoblasts controls osteoclast differ-
PTH and other anabolic agents. entiation. Dev Cell 8:751–764
20. Holmen SL, Zylstra CR, Mukherjee A et al (2005) Essential role
of beta-catenin in postnatal bone acquisition. J Biol Chem
280:21162–21168
21. Kawano Y, Kypta R (2003) Secreted antagonists of the Wnt
References signalling pathway. J Cell Sci 116:2627–2634
22. Delany AM, Gabbitas BY, Canalis E (1995) Cortisol down
1. Mazziotti G, Angeli A, Bilezikian JP et al (2006) Glucocorticoid- regulates osteoblast 1(I) procollagen mRNA by transcriptional
induced osteoporosis: an update. Trends Endocrinol Metab and post-transcriptional mechanisms. J Cell Biochem 57:
7:144–149 488–494
1326 Osteoporos Int (2007) 18:1319–1328

23. Liu Y, Porta A, Peng X et al (2004) Prevention of glucocorticoid- 43. Okazaki R, Riggs BL, Conover CA (1994) Glucocorticoid
induced apoptosis in osteocytes and osteoblasts by calbindin-D28k. regulation of insulin-like growth factor-binding protein expres-
J Bone Miner Res 19:479–490 sion in normal human osteoblast-like cells. Endocrinology
24. O’Brien CA, Jia D, Plotkin LI et al (2004) Glucocorticoids act 134:126–132
directly on osteoblasts and osteocytes to induce their apoptosis 44. Gabbitas B, Pash JM, Delany AM et al (1996) Cortisol inhibits
and reduce bone formation and strength. Endocrinology the synthesis of insulin-like growth factor binding protein-5 in
145:1835–1841 bone cell cultures by transcriptional mechanisms. J Biol Chem
25. Thornberry NA, Lazebnik Y (1998) Caspases: enemies within. 271:9033–9038
Science 281:1312–1316 45. Devlin RD, Du Z, Buccilli V et al (2002) Transgenic mice
26. Baylink DJ, Wergedal JE (1971) Bone formation by osteocytes. overexpressing insulin-like growth factor binding protein-5 dis-
Am J Physiol 221:669–678 play transiently decreased osteoblastic function and osteopenia.
27. Lane NE, Yao W, Balooch M et al (2006) Glucocorticoid- Endocrinology 143:3955–3962
treated mice have localized changes in trabecular bone material 46. Lane NE, Sanchez S, Modin GW et al (1998) Parathyroid hormone
properties and osteocyte lacunar size that are not observed in treatment can reverse corticosteroid-induced osteoporosis. Results of
placebo-treated or estrogen-deficient mice. J Bone Miner Res a randomized controlled clinical trial. J Clin Invest 102:1627–1633
21:466–476 47. Huybers S, Naber TH, Bindels RJ et al (2006) Prednisolone-
28. Canalis E, Bilezikian JP, Angeli A et al (2004) Perspectives on induced Ca2+malabsorption is caused by diminished expression
glucocorticoid-induced osteoporosis. Bone 34:593–598 of the epithelial Ca2+channel TRPV6. Am J Physiol Gastrointest
29. Teitelbaum SL (2000) Bone resorption by osteoclasts. Science Liver Physiol Aug [Epub ahead of print]
289:1504–8. 48. Rubin MR, Bilezikian JP (2002) The role of parathyroid
30. Hofbauer LC, Gori F, Riggs BL et al (1999) Stimulation of hormone in the pathogenesis of glucocorticoid-induced osteopo-
osteoprotegerin ligand and inhibition of osteoprotegerin pro- rosis: a re-examination of the evidence. J Clin Endocrinol Metab
duction by glucocorticoids in human osteoblastic lineage cells: 87:4033–4041
potential paracrine mechanisms of glucocorticoid-induced 49. Laan RF, Buijs WC, van Erning LJ et al (1993) Differential
osteoporosis. Endocrinology 140:4382–4389 effects of glucocorticoids on cortical appendicular and cortical
31. Rubin J, Biskobing DM, Jadhav L et al (1998) Dexamethasone vertebral bone mineral content. Calcif Tissue Int 52:5–9
promotes expression of membrane-bound macrophage colony- 50. Bonadonna S, Burattin A, Nuzzo M et al (2005) Chronic
stimulating factor in murine osteoblast-like cells. Endocrinology glucocorticoid treatment alters spontaneous pulsatile parathyroid
139:1006–1012 hormone secretory dynamics in human subjects. Eur J Endo-
32. Dovio A, Perazzolo L, Saba L et al (2006) High-dose crinol 152:199–205
glucocorticoids increase serum levels of soluble IL-6 receptor 51. Samuels MH, Veldhuis J, Cawley C et al (1993) Pulsatile
alpha and its ratio to soluble gp130: an additional mechanism for secretion of parathyroid hormone in normal young subjects:
early increased bone resorption. Eur J Endocrinol 154:745–751 assessment by deconvolution analysis. J Clin Endocrinol Metab
33. Takuma A, Kaneda T, Sato T et al (2003) Dexamethasone 76:399–403
enhances osteoclast formation synergistically with transforming 52. Samuels MH, Veldhuis JD, Kramer P et al (1993) Episodic
growth factor-beta by stimulating the priming of osteoclast secretion of parathyroid hormone in post-menopausal women:
progenitors for differentiation into osteoclasts. J Biol Chem assessment by deconvolution analysis and approximate entropy.
278:44667–44674 J Bone Miner Res 12:616–623
34. Jia D, O’Brien CA, Stewart SA et al (2006) Glucocorticoids act 53. Mazziotti G, Cimino V, De Menis E et al (2006) Active
directly on osteoclasts to increase their life span and reduce bone acromegaly enhances spontaneous parathyroid hormone pulsa-
density. Endocrinology 147:5592–5599 tility. Metabolism 55:736–740
35. Kim HJ, Zhao H, Kitaura H et al (2006) Glucocorticoids 54. Urena P, Iida-Klein A, Kong XF et al (1994) Regulation of
suppress bone formation via the osteoclast. J Clin Invest 116: parathyroid hormone (PTH)/PTHrelated peptide receptor mes-
2152–2160 senger ribonucleic acid by glucocorticoids and PTH in ROS 17/
36. Knauper V, Will H, Lopez-Otin C et al (1996) Cellular 2.8 and OK cells. Endocrinology 134:451–456
mechanisms for human procollagenase-3 (MMP-13) activation. 55. Giustina A, Veldhuis JD (1998) Pathophysiology of the neuro-
J Biol Chem 271:17124–17131 regulation of growth hormone secretion in experimental animals
37. Freije JMP, Diez-Itza I, Balbin M et al (1994) Molecular cloning and the human. Endocr Rev 19:717–797
and expression of collagenase 3, a novel human matrix metal- 56. Giustina A, Bossoni S, Bodini C et al (1992) Arginine normalizes
loproteinase produced by breast carcinomas. J Biol Chem the growth hormone (GH) response to GH-releasing hormone in
269:16766–16773 adult patients receiving chronic daily immunosuppressive gluco-
38. Delany AM, Jeffrey JJ, Rydziel S et al (1995) Cortisol increases corticoid therapy. J Clin Endocrinol Metab 74:1301–1305
interstitial collagenase expression in osteoblasts by post-tran- 57. Giustina A, Bussi AR, Jacobello C et al (1995) Effects of
scriptional mechanisms. J Biol Chem 270:26607–26612 recombinant human growth hormone (GH) on bone and
39. Dempster DW, Arlot MA, Meunier PJ (1983) Mean wall thickness intermediary metabolism in patients receiving chronic glucocor-
and formation periods of trabecular bone packets in corticosteroid- ticoid treatment with suppressed endogenous GH response to
induced osteoporosis. Calcif Tissue Int 35: 410–417 GH-releasing hormone. J Clin Endocrinol Metab 80:122–129
40. Stellon AJ, Webb A, Compston JE (1988) Bone histomorphom- 58. Manelli F, Carpinteri R, Bossoni S et al (2002) Growth hormone in
etry and structure in corticosteroid treated chronic active glucocorticoid-induced osteoporosis. Front Horm Res 30:174–183
hepatitis. Gut 29:378–384 59. Malerba M, Bossoni S, Radaeli A et al (2005) Growth hormone
41. Canalis E, Centrella M, Burch J et al (1989) Insulin-like growth response to growth hormone-releasing hormone is reduced in
factor I mediates selected anabolic effects of parathyroid adult asthmatic patients receiving long-term inhaled corticoste-
hormone in bone cultures. J Clin Invest 83:60–65 roid treatment. Chest 127:515–521
42. Rydziel S, Canalis E (1995) Cortisol represses insulin-like 60. van Staa TP (2006) The pathogenesis, epidemiology and
growth factor II receptor transcription in skeletal cell cultures. management of glucocorticoid-induced osteoporosis. Calcif
Endocrinology 136:4254–4260 Tissue Int 79:129–137
Osteoporos Int (2007) 18:1319–1328 1327

61. Cohen S, Levy RM, Keller M et al (1999) Risedronate therapy 81. Bonadonna S, Mazziotti G, Nuzzo M et al (2005) Increased
prevents corticosteroid-induced bone loss: a twelve-month, prevalence of radiological spinal deformities in active acromegaly:
multicenter, randomized, double-blind, placebo-controlled, par- a cross-sectional study in postmenopausal women. J Bone Miner
allel-group study. Arthritis Rheum 42:2309–2318 Res 20:1837–1844
62. Wallach S, Cohen S, Reid DM et al (2000) Effects of risedronate 82. Mazziotti G, Bianchi A, Bonadonna S et al (2006) Increased
treatment on bone density and vertebral fracture in patients on prevalence of radiological spinal deformities in adult patients
corticosteroid therapy. Calcif Tissue Int 67:277–285 with GH deficiency: influence of GH replacement therapy. J
63. Angeli A, Guglielmi G, Dovio A et al (2006) High prevalence of Bone Miner Res 21:520–528
asymptomatic vertebral fractures in post-menopausal women 83. Van Staa TP, Laan RF, Barton IP et al (2003) Bone density threshold
receiving chronic glucocorticoid therapy: a cross-sectional and other predictors of vertebral fracture in patients receiving oral
outpatient study. Bone 39:253–259 glucocorticoid therapy. Arthritis Rheum 48: 3224–3229
64. Shaker JL, Lukert BP (2005) Osteoporosis associated with excess 84. Compston J (2004) US and UK guidelines for glucocorticoid-
glucocorticoids. Endocrinol Metab Clin North Am 34:341–356 induced osteoporosis: similarities and differences. Curr Rheu-
65. van Staa TP, Leufkens HGM, Cooper C (2002) The epidemiology matol Rep 6:66–69
of corticosteroid-induced osteoporosis: a metaanalysis. Osteoporos 85. Cefalu CA (2004) Is bone mineral density predictive of fracture
Int 13:777–787 risk reduction? Curr Med Res Opin 20:341–349
66. Cohen A, Shane E (2003) Osteoporosis after solid organ and 86. Lunt M, Felsenberg D, Reeve J et al (1997) Bone density
bone marrow transplantation. Osteoporos Int 14:617–630 variation and its effects on risk of vertebral deformity in men and
67. Steinbuch M, Youket TE, Cohen S (2004) Oral glucocorticoid women studied in thirteen European centers: the EVOS Study. J
use is associated with an increased risk of fracture. Osteoporos Bone Min Res 12:1883–1894
Int 15:323–328 87. Olesik A, Ott SM, Vedi S et al (2000) Bone structure in patients
68. van Rossum EF, Koper JW, van den Beld AW et al (2003) with low bone mineral density with or without vertebral
Identification of the BclI polymorphism in the glucocorticoid fractures. J Bone Min Res 15:1368–1375
receptor gene: association with sensitivity to glucocorticoids in 88. Carballido-Gamio J, Majumdar S (2006) Clinical utility of
vivo and body mass index. Clin Endocrinol 59:585–592 microarchitecture measurements of trabecular bone. Curr Osteo-
69. Russcher H, Smit P, van den Akker EL et al (2005) Two poros Rep 4:64–70
polymorphisms in the glucocorticoid receptor gene directly affect 89. Masaki H, Miki T (2006) The biochemical markers of bone in
glucocorticoid-regulated gene expression. J Clin Endocrinol steroid (glucocorticoid)-induced osteoporosis (GIOP). Clin Cal-
Metab 90:804–810 cium 16:51–60
70. van Rossum EF, Voorhoeve PG, te Velde SJ et al (2004) The 90. Natsui K, Tanaka K, Suda M et al (2006) High-dose glucocor-
ER22/23EK polymorphism in the glucocorticoid receptor gene is ticoid treatment induces rapid loss of trabecular bone mineral
associated with a beneficial body composition and muscle density and lean body mass. Osteoporos Int 17:105–108
strength in young adults. J Clin Endocrinol Metab 89:4004–4009 91. Gilson H, Schakman O, Combaret L et al (2007) Myostatin gene
71. Kaji H, Tobimatsu T, Naito J et al (2006) Body composition and deletion prevents glucocorticoid-induced muscle atrophy. Endo-
vertebral fracture risk in female patients treated with glucocor- crinology 148:452–460
ticoid. Osteoporos Int 17:627–633 92. American College of Rheumatology Ad Hoc Committee on
72. Tomlinson JW, Walker EA, Bujalska IJ et al (2004) 11beta- Glucocorticoid-Induced Osteoporosis (2001) Recommendations
hydroxysteroid dehydrogenase type 1: a tissue-specific regulator for the prevention and treatment of glucocorticoid-induced
of glucocorticoid response. Endocr Rev 25:31–66 osteoporosis: 2001 update. Arthritis Rheum 44:1496–1503
73. Cooper MS, Bujalska I, Rabbitt E et al (2001) Modulation of 11â- 93. Boonen S, Vanderschueren D, Haentjens P et al (2006) Calcium
hydroxysteroid dehydrogenase isozymes by proinflammatory and vitamin D in the prevention and treatment of osteoporosis - a
cytokines in osteoblasts: an autocrine switch from glucocorticoid clinical update. J Intern Med 259:539–552
inactivation to activation. J Bone Miner Res 16:1037–1044 94. Shiraishi A, Takeda S, Masaki T et al (2000) Alfacalcidol
74. Williams LJ, Lyons V, MacLeod I et al (2000) C/EBP regulates inhibits bone resorption and stimulates formation in an ovariec-
hepatic transcription of 11beta-hydroxysteroid dehydrogenase tomized rat model of osteoporosis: distinct actions from
type1. A novel mechanism for cross-talk between the C/EBP estrogen. J Bone Miner Res 15:770–779
and glucocorticoid signaling pathways. J Biol Chem 275: 95. de Nijs RN, Jacobs JW, Algra A et al (2004) Prevention and
30232–30239 treatment of glucocorticoid-induced osteoporosis with active
75. Cooper MS, Rabbitt EH, Goddard PE et al (2002) Osteoblastic vitamin D(3) analogues: a review with meta-analysis of
11â-hydroxysteroid dehydrogenase type 1 activity increases with randomized controlled trials including organ transplantation
age and glucocorticoid exposure. J Bone Miner Res 17:979–986 studies. Osteoporos Int 15:589–602
76. van Hogezand RA, Hamdy NA (2006) Skeletal morbidity in 96. Schacht E (1999) Rationale for treatment of involutional
inflammatory bowel disease. Scand J Gastroenterol 243:S59–S64 osteoporosis in women and for prevention and treatment of
77. Romas E (2005) Bone loss in inflammatory arthritis: mecha- corticosteroid-induced osteoporosis with alfacalcidol. Calcif
nisms and therapeutic approaches with bisphosphonates. Best Tissue Int 65:317–327
Pract Res Clin Rheumatol 19:1065–1079 97. Buckley LM, Leib ES, Cartularo KS et al (1996) Calcium and
78. Lekamwasam S, Trivedi DP, Khaw KT (2002) An association vitamin D3 supplementation prevents bone loss in the spine
between respiratory function and bone mineral density in women secondary to low-dose corticosteroids in patients with rheuma-
from the general community: a cross sectional study. Osteoporos toid arthritis. A randomized, double-blind, placebo-controlled
Int 13:710–715 trial. Ann Intern Med 125:961–968
79. Sin DD, Man JP, Man SF (2003) The risk of osteoporosis in 98. Heaney RP (2005) The Vitamin D requirement in health and
Caucasian men and women with obstructive airways disease. disease. J Steroid Biochem Mol Biol 97:13–19
Am J Med 114:10–14 99. Amin S, Lavalley MP, Simms RW et al (2002) The comparative
80. Khan AA, Hanley DA, Bilezikian JP et al (2006) Standards for efficacy of drug therapies used for the management of
performing DXA in individuals with secondary causes of corticosteroid-induced osteoporosis: a meta-regression. J Bone
osteoporosis. J Clin Densitom 9:47–57 Miner Res 17:1512–1526
1328 Osteoporos Int (2007) 18:1319–1328

100. de Nijs RN, Jacobs JW, Lems WF et al (2006) Alendronate or 107. Iu MF, Kaji H, Naito J et al (2005) Low-dose parathyroid
alfacalcidol in glucocorticoid-induced osteoporosis. N Engl J hormone and estrogen reverse alkaline phosphatase activity
Med 355:675–684 suppressed by dexamethasone in mouse osteoblastic cells. J
101. Saag KG, Emkey R, Schnitzer TJ et al (1998) Alendronate for Bone Miner Metab 23:450–455
the prevention and treatment of glucocorticoid-induced osteopo- 108. Calvi LM, Adams GB, Weibrecht KW et al (2003) Osteoblastic
rosis. Glucocorticoid-Induced Osteoporosis Intervention Study cells regulate the haematopoietic stem cell niche. Nature
Group. N Engl J Med 339:292–299 425:841–846
102. Reid DM, Hughes RA, Laan RF et al (2000) Efficacy and safety 109. Lane NE, Sanchez S, Modin GW et al (1998) Parathyroid
of daily risedronate in the treatment of corticosteroid-induced hormone treatment can reverse corticosteroid-induced osteopo-
osteoporosis in men and women: a randomized trial. European rosis. Results of a randomized controlled clinical trial. J Clin
Corticosteroid-Induced Osteoporosis Treatment Study. J Bone Invest 102:1627–1633
Miner Res 15:1006–1013 110. Lane NE, Sanchez S, Genant HK et al (2000) Short-term
103. Adachi JD, Saag KG, Delmas PD et al (2001) Two-year effects of increases in bone turnover markers predict parathyroid hor-
alendronate on bone mineral density and vertebral fracture in mone-induced spinal bone mineral density gains in postmeno-
patients receiving glucocorticoids: a randomized, double-blind, pausal women with glucocorticoid-induced osteoporosis.
placebo-controlled extension trial. Arthritis Rheum 44:202–211 Osteoporos Int 11:434–442
104. van Staa TP, Geusens P, Bijlsma JW et al (2006) Clinical 111. Rehman Q, Lang TF, Arnaud CD et al (2003) Daily treatment
assessment of the long-term risk of fracture in patients with with parathyroid hormone is associated with an increase in
rheumatoid arthritis. Arthritis Rheum 54:3104–3112 vertebral cross-sectional area in postmenopausal women with
105. Franchimont N, Canalis E (2003) Management of glucocorticoid glucocorticoid-induced osteoporosis. Osteoporos Int 14:77–81
induced osteoporosis in premenopausal women with autoimmune 112. Buxton EC, Yao W, Lane NE (2004) Changes in serum receptor
disease. Autoimmun Rev 4:224–228 activator of nuclear factor-kappaB ligand, osteoprotegerin, and
106. Jilka RL, Weinstein RS, Bellido T et al (1999) Increased bone interleukin-6 levels in patients with glucocorticoid-induced
formation by prevention of osteoblast apoptosis with parathyroid osteoporosis treated with human parathyroid hormone (1–34). J
hormone. J Clin Invest 104:439–446 Clin Endocrinol Metab 89:3332–3336

You might also like