Download as pdf or txt
Download as pdf or txt
You are on page 1of 16

PRIMER

Postmenopausal osteoporosis
Richard Eastell1, Terence W. O’Neill2, Lorenz C. Hofbauer3, Bente Langdahl4, Ian R. Reid5,
Deborah T. Gold6 and Steven R. Cummings7
Abstract | Osteoporosis is a metabolic bone disorder that is characterized by low bone mass and
micro-architectural deterioration of bone tissue. Fractures of the proximal femur, the vertebrae
and the distal radius are the most frequent osteoporotic fractures, although most fractures in the
elderly are probably at least partly related to bone fragility. The incidence of fractures varies greatly
by country, but on average up to 50% of women >50 years of age are at risk of fractures. Fractures
severely affect the quality of life of an individual and are becoming a major public health problem
owing to the ageing population. Postmenopausal osteoporosis, resulting from oestrogen deficiency,
is the most common type of osteoporosis. Oestrogen deficiency results in an increase in bone turnover
owing to effects on all types of bone cells. The imbalance in bone formation and resorption has effects
on trabecular bone (loss of connectivity) and cortical bone (cortical thinning and porosity).
Osteoporosis is diagnosed using bone density measurements of the lumbar spine and proximal femur.
Preventive strategies to improve bone health include diet, exercise and abstaining from smoking.
Fractures may be prevented by reducing falls in high-risk populations. Several drugs are licensed to
reduce fracture risk by slowing down bone resorption (such as bisphosphonates and denosumab) or
by stimulating bone formation (such as teriparatide). Improved understanding of the cellular basis
for osteoporosis has resulted in new drugs targeted to key pathways, which are under development.

Osteoporosis has been defined as a systemic ­skeletal 10 years; the most widely used tool is the WHO Fracture
disease that is characterized by low bone mass and Risk Assessment Tool (FRAX) score3.
micro-architectural deterioration of bone tissue, with These approaches of characterizing osteoporosis
a consequent increase in bone fragility and susceptibil­ prove helpful in explaining individual risk and can help
ity to fracture1. The most frequent osteoporotic frac- with deciding whom to treat. When we make a diagno-
tures are fractures of the hip, wrist and spine (FIG. 1), sis of osteoporosis in our clinical practice, we take into
although most fractures in the elderly are probably at account the level of BMD, the presence of a fracture
least partly related to osteoporosis. Osteoporosis is more strongly linked to osteoporosis (such as proximal femur
common in women and, in this Primer, we focus on or spine) or a high 10‑year fracture risk4. Osteoporosis
postmenopausal osteoporosis. is a treatable condition and yet many people with this
Bone mineral density (BMD) is a key tool in identify­ condition remain undiagnosed and untreated5. This
ing osteoporosis and its introduction into clinical prac- Primer examines the epidemiology and pathophysiology
tice 30 years ago was a major step forward. A working of postmenopausal osteoporosis and considers its diag-
group of the WHO has defined osteoporosis oper­ nosis, prevention and treatment and its effect on quality
ationally as a BMD that is 2.5 standard deviations below of life, as well as looks to the future.
the peak bone mass for adult women, or lower 2. The
­number of standard deviations from average is defined Epidemiology
by the T-score, and a T-score of ≤–2.5 is thus defined Incidence and prevalence
Correspondence to R.E. as osteoporosis. The limitation of this approach is that Osteoporosis is a considerable clinical and public health
Metabolic Bone Centre, it increases a risk factor to the status of a diagnostic burden because of its association with age-related
Northern General Hospital, ­criterion and so ignores the other determinants of bone fractures. In the United States, 10 million individuals
Herries Road, Sheffield,
strength and fracture propensity. In the same way that >50 years of age are estimated to have osteoporosis
S5 7AU, UK.
r.eastell@sheffield.ac.uk we estimate the risk of cardiovascular disease from key of the hip (based on a T-score of ≤–2.5), with about
risk factors, there has been a move to use BMD along 1.5 million osteoporotic fractures each year. In Europe,
Article number: 16069
doi:10.1038/nrdp.2016.69 with key risk factors for fracture (particularly age, 27.6 million people are estimated to have age-related
Published online 29 Sep 2016 weight and prior fracture) to predict fracture in the next osteoporosis and >3.5 million fragility fractures are

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 1


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Author addresses data from non-western populations, although there is


evidence that hip fracture rates may be continuing to
1
Academic Unit of Bone Metabolism, and Mellanby Centre rise in some Asian countries. For example, the age-­
for Bone Research, University of Sheffield, Sheffield, UK. specific rate of hip fractures rose threefold to fourfold in
2
Arthritis Research UK Centre for Epidemiology, Centre for Beijing, China, from 1990–1992 to 2002–2006 (REF. 16).
Musculoskeletal Research, Manchester Academic Health
The risk might be due to westernization of lifestyle that
Science Centre (MAHSC), The University of Manchester,
Manchester, UK. includes using cars instead of bikes and chairs instead of
3
Division of Endocrinology, Diabetes, and Bone Diseases ­squatting. The number of fractures globally is expected
and Centre for Healthy Aging, Technische Universität to rise considerably over the next 30 years because of
Dresden Medical Center, Dresden, Germany. ageing; the number of people in the world ≥65 years
4
Department of Endocrinology and Internal Medicine, of age is expected to double between 2010 and 2040
Aarhus University Hospital, Aarhus, Denmark. (REF. 17). The increase in the number of old people will
5
Department of Medicine, University of Auckland, be most marked in Asia where it is estimated that half of
Auckland, New Zealand. all hip fractures will occur by 2050 (REF. 18).
6
Departments of Psychiatry & Behavioral Sciences,
Sociology, and Psychology & Neuroscience, Duke
Genetics factors
University Medical Center, Durham, North Carolina, USA.
7
San Francisco Coordinating Center, California Pacific Genetic factors have an important role in the develop­
Medical Center Research Institute and the University of ment of osteoporosis. Twin and family studies have
California, San Francisco, San Francisco, California, USA. shown that the heritability of bone mass is 50–70%19,20.
The heritability estimates for fracture strongly depend
on age. The heritability of, for example, hip fractures
observed each year 6,7. The economic cost of these frac- in individ­uals <69 years of age was 70%, whereas it
tures is high with an annual cost of US$17 billion in the was almost negligible in individuals >79 years of age21.
United States in 2005 and €37 billion in Europe based on Variants in the genes encoding factors known to regu-
data from 2010 (REFS 7,8). late bone mass (such as oestrogen receptor-α (ERα)22,
From an individual perspective, the lifetime risk for vitamin D receptor 23, transforming growth factor-β
sustaining an osteoporotic fracture from 50 years of (TGFβ)24, low-density lipoprotein receptor-related pro-
age is estimated to be one in two for women and one in tein 5 (LRP5) and LRP6 (both involved in WNT sig-
five for men based on data from the United Kingdom nalling))25 and bone matrix components (for example,
General Practice Research Database9. The incidence collagen type I α1 (REF. 26)) were found to be associated
of osteoporotic fractures increases with age and is with bone mass and fracture risk, but these variants could
approximately twice as high in women as in men at all be used to explain only very little of the inter-­individual
ages (FIG. 2). The preferential location of fractures also variation. A study in 2015 suggested that at least 500
varies with age. In women, the incidence of wrist and different genetic variants influence bone mass and/or
spine fractures begins to rise considerably from around fracture risk27. After the discovery of the importance of
50 years of age, whereas hip fractures occur later in life. the WNT pathway in bone bio­logy, genetic vari­ants in
In general, the pattern of fracture incidence with age some of the genes related to this pathway have also been
is similar in men, although, in contrast to women, the shown to affect bone mass and fracture risk. In addition
incidence of wrist fracture in men remains low until to LRP5 and LRP6, variants in WNT1 (REF. 28), WNT16
later life9. (REF. 29) and SOST (encoding ­s clerostin)30 have been
Globally, the frequency of the major osteoporotic found to affect bone mass. Large international collabor­
fractures (including fractures of the spine, hip, wrist ative efforts are ongoing to unravel more of the under-
and shoulder) varies substantially, being highest in lying genetic variance. Genome-wide association studies
Scandinavia and lowest in Africa10,11. The variation is including >200,000 individuals have identified 56 loci
most marked for hip fracture, with rates in women vary­ associated with BMD and 14 of these were also associ­
ing by >200‑fold. This may partly reflect ethnic influ- ated with fracture risk31. However, before these vari-
ences on fracture: higher fracture risk is seen in white ants can be included in risk assessment of i­ ndividuals,
women than in black women. However, there is variation a ­functional effect will have to be established.
also within geographical regions. For example, hip frac-
ture rates vary in Europe by a factor of approximately Mechanisms/pathophysiology
11‑fold: the highest rates are in northern Europe and the Bone remodelling — the constant and coordinated
lowest rates are in the Mediterranean area12. turnover of mineralized bone matrix in response to
Since the early-to-mid-twentieth century, the mechanical and endocrine stimuli — is accomplished
age-specific incidence of hip fractures has increased by two specialized cells: bone-resorbing osteoclasts
in many western countries. In most countries, rates and bone-forming osteoblasts (FIG. 3). Osteocytes sense
stabil­ized towards the end of the twentieth century mechanical strain and regulate bone remodelling. About
and in some areas, particularly northern Europe and 95% of the body’s calcium is incorporated into the bone
North America, the incidence has begun to decline13–15. matrix by the action of osteoblasts, which produce
This decline in hip fractures has been associated with type I collagen as the main bone matrix protein. The
higher BMD in the elderly, which might be attributed absorption and distribution of calcium to the skeleton
to an increased prevalence of obesity. There are fewer is under endocrine control of, for example, vitamin D,

2 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

In addition, bone geometry of long bones differs sub-


a b c
stantially between women and men. This partly explains
why women are more prone to developing osteoporotic
bone fractures39 (FIG. 4). In the process of bone model-
ling during puberty, boys experience a greater periosteal
apposition rate at the outer cortical surface under the
influence of testosterone, leading to wider bones and
a higher cortical thickness. During puberty, girls have
enhanced periosteal apposition, but mainly endocortical
thickening of their cortical bone. Starting from young
d e f g adulthood and extending just before perimenopause,
cortical remodelling in women involves endocortical
resorption, which is almost compensated for by peri-
osteal apposition40. When oestrogen levels decline after
menopause in women, endocortical resorption is further
enhanced, whereas periosteal apposition is decreased,
translating into a thinner cortex. Oestrogen replacement
Figure 1 | Osteoporotic bone. a–c | Radiographs showing fractures
Nature (arrows)
Reviews of thePrimers
| Disease
therapy can prevent, but not reverse, cortical bone loss40.
proximal femur (part a), distal radius (part b) and vertebra (part c). d–g | High resolution Apart from thinning of the cortex, an additional struc-
quantitative CT allows study of the trabecular (the sponge-like interior compartment tural basis for impaired bone quality has been identified:
of bone) and cortical (the compact, outer layer of bone) components of the radius (part d increased cortical porosity 41. Swiss cheese-like pores
and part f) and the tibia (part e and part g). Compared with controls (part d and part e), (FIG. 4) have been observed in the cortex of women with
osteoporosis (part f and part g) is characterized by thinning of the cortex and reduced postmenopausal osteoporosis41, aged men and patients
trabecular bone mass with a loss of trabecular connectivity.
with type 2 diabetes mellitus42, and have been linked to
increased fragility.
parathyroid hormone (PTH) and sex steroids, including
oestrogens. The levels of PTH increase with age, which Oestrogens and osteoporosis
might partly be because of vitamin D insufficiency, and Reduced ovarian production of oestrogens after meno­
contribute to the increase in bone turnover with ageing. pause is the cause for an initial phase of rapid bone loss
in women, with an annual bone loss rate of 3–5% for
Bone structure and remodelling in osteoporosis 5–10 years, which mainly affects trabecular bone. This
A consistent feature of postmenopausal women is phenomenon, sex-specific differences in bone geo­
the concurrent increase of bone resorption and bone metry (FIG. 4) and the lack of a bona fide male equiva-
formation at the tissue level, termed high bone turn­ lent of meno­pause contribute to a higher incidence of
over. However, at the cellular level — the multicellular osteoporotic fractures in postmenopausal women than
bone-remodelling unit — there is a negative imbalance in age-matched men34,43. The onset and the severity of
in bone remodelling, with bone resorption outpacing postmenopausal osteoporosis are modulated by genetic
bone formation. Bone resorption on the basis of bio- factors, the degree of weight-bearing physical activity,
chemical markers of bone turnover is increased by 90% nutrition, smoking, body mass index (BMI), concurrent
after menopause, but bone formation is only increased diseases and medications, and ageing.
by 45%, thus resulting in net bone loss with each The effect of fat mass and oestrogen on BMD is
remodel­ling cycle32. Mechanisms that contribute to this explained by the fact that residual oestrogens are prod­
negative remodelling imbalance in osteoporosis include uced by conversion of adrenal androgens (for example,
the increased number of multicellular bone-remodelling androstenedione and dehydroepiandrosterone sulfate) by
units, when bone is remodelled with a prolongation of the enzyme aromatase (also known as CYP19A1), which
the resorption period and a concurrent shortening of the is highly expressed in white adipose tissue44. Thus, obese
formation period33–36, thus favouring sustained loss. women tend to have higher circulating levels of oestro-
Bone loss in postmenopausal women occurs in two gen throughout their lifetime but especially after meno-
phases: an initial shorter phase lasting 3–5 years with pause than age-matched individuals with a normal BMI,
rapid loss of mainly trabecular bone (menopause-­ which confer partial protection against osteoporosis45.
related bone loss) and a second longer phase of slower In circumstances in which this peripheral androgen-to‑­
loss lasting 10–20 years that occurs in women and men oestrogen conversion is blocked, for example, by the use
(age-related bone loss) and involves both cortical (the of aromatase inhibitors as in hormone receptor-positive
outer shell) and trabecular compartments37,38 (FIG. 4). breast cancer, bone loss is further enhanced45. The pro-
This explains why fracture incidence in men increases tective effect of oestrogens on the female skeleton also
about a decade later than in women and why fragility becomes apparent in transient periods of oestrogen
fractures of the spine occur earlier than fractures at deficiency, such as post-partum and during lactation46.
other skeletal sites (FIG. 2). Vertebral bodies are mainly In these circumstances, increased bone resorption is
(approximately 80%) composed of trabecular bone, thought to mobilize calcium from the maternal skele-
whereas long bones and hip consist mainly of cortical ton to ensure calcium transfer to the offspring via breast
bone (80% and 40%, respectively). milk, underscoring the evolutionary importance of

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 3


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

350
Women
350
Men increasing the secretion of IL‑1, IL‑6 and tumour necro-
sis factor (TNF) (FIG. 5). The net result is enhanced bone
300 300 turnover with increased bone resorption and subse-
Incidence per 10,000 per year

Incidence per 10,000 per year


quently increased bone formation, with a net effect of
250 250
resorption exceeding formation. The increase in bone
200 200
turnover is augmented by a pro-inflammatory cytokine
milieu and immune cell activation induced by oestrogen
150 150 deficiency 49. In principle, these alterations are reversed by
oestrogen replacement. Oestrogens and oestrogen defi-
100 100 ciency affect all three bone cell types and immune cells
50 50
in a distinct manner, which is highlighted in the sections
below (FIG. 5).
0 0
Osteoblasts and bone formation. Osteoblast function
55 4
60 9
65 4
70 9
75 4
80 9
85 4
90 9
4

55 4
60 9
65 4
70 9
75 4
80 9
85 4
90 9
4
–5
–5
–6
–6
–7
–7
–8
–8
–9

–5
–5
–6
–6
–7
–7
–8
–8
–9
50

50 and bone formation require the activation of bone


Age groups (years) Age groups (years)
morpho­genetic protein (BMP), TGFβ and particularly
Vertebral fracture Forearm fracture Femur and hip fractures the canonical WNT signalling pathway. WNT signal-
ling is under negative control by WNT inhibitors such
Figure 2 | Fracture incidence with increasing age. Incidence of radiographic vertebral,
Nature Reviews | Disease Primers as sclerostin, Dickkopf 1 and secreted Frizzled-related
forearm, and femur and hip fractures in women and men are shown. Data from
REFS 197,198.
protein 1, all of which are secreted by osteoblasts and
osteocytes52,53 (FIG. 3).
Oestrogens are crucial regulators of osteoblast differ-
a dynamic bone metabolism. Of note, oestrogen is also entiation and activity. Oestrogens enhance osteogenic
crucial for bone health in men, as men with inactive ERα differentiation of cultured mesenchymal stem cells
have severe osteoporosis47, and bone loss in men is more (MSCs), promote differentiation from pre-osteoblasts to
closely corre­lated to serum o ­ estradiol levels rather than osteoblasts and prolong osteoblast and osteocyte lifespan
to serum testosterone levels48. by suppressing apoptosis54. Moreover, oestrogens stim-
ulate the production of IGF1 and TGFβ by osteoblasts
Altered oestrogen signalling. ERs with a high affinity as well as procollagen synthesis55 (FIG. 5). These marked
for oestrogens have been detected in osteoblasts, osteo­ in vitro effects have been confirmed in humans follow-
clasts and osteocytes; these receptors include ERα and ing dermal oestrogen application to postmenopausal
ERβ isoforms, which bind to different ligands, mediate women for 6 years, which leads to increased trabecular
different effects and have different spatial distributions37. thickness of 12% and increased trabecular bone volume
ERα mediates most effects of natural oestrogen ligands of 61%, as a result of enhanced bone formation56. The
and is mainly expressed in bone cells that reside in cor- bone-forming effects of oestrogens in humans depend
tical bone, whereas ERβ mediates the effects of phyto-­ on the p ­ reparation, dose and mode of administration.
oestrogens (for example, found in soy products) on bone
and is predominantly expressed in bone cells present in Osteoclasts and bone resorption. Oestrogens function
trabecular bone38,49. ERs are nuclear receptors and ligand as anti-resorptive hormones that limit bone resorption
binding induces conformational changes that result in ER and bone loss (FIG. 5). Oestrogens regulate the receptor
dimerization and subsequent interactions with coactiv­ activator of NF‑κB (RANK; also known as TNFRSF11A)
ator or co-repressor proteins to either stimulate or inhibit ligand (RANKL; also known as TNFRSF11), RANK and
the expression of oestrogen target genes encoding pro- osteoprotegerin (OPG; also known as TNFRSF11B)
teins such as IL‑1, insulin-like growth factor 1 (IGF1) and pathway (FIG. 3) at several levels. Oestrogens upregulate
TGFβ. These effects are referred to as classical or genomic the expression of RANKL in the osteoblastic cell lineage
ER effects of ER activation. However, ERs can also bind to and induce the expression of OPG, a naturally occur-
transcription factors such as nuclear factor-κβ (NF‑κβ), ring antagonist of RANKL, in vitro57. Moreover, oestro-
which prevents their action, for example, the inhibition gens interfere with osteoclast signalling downstream of
of IL6 expression49. Moreover, membrane receptor-­ RANK, thereby preventing osteoclast differentiation and
mediated effects have been reported for certain types of promoting osteoclast apoptosis via increased produc-
oestrogens in which these compounds induce rapid non- tion of TGFβ by osteoblasts. Under conditions of acute
genomic effects, for example, phosphorylation of extra- oestro­gen deficiency, osteoclast differentiation and activ-
cellular signal-regulated kinase (ERK) kinases and other ity are increased by a higher expression of RANKL49,58
enzymes50. The relative contribution of genomic versus and other pro-osteoclastic cytokines, including TNF,
non-genomic effects on the skeleton in health and disease IL‑1 and IL‑6, by MSCs and lymphocytes. These effects
is unclear and requires further investigation51. are partially suppressed by oestrogen administration in
Lack of oestrogen affects all bone cells either directly ­postmenopausal women58.
by regulating cellular differentiation, activity and apop-
tosis or indirectly via altered expression of oestrogen-­ Osteocytes and mechanosensing. Osteocytes control
responsive target genes; proteins encoded by these genes bone remodelling and mineralization and function as
might function in an autocrine or paracrine manner by endocrine cells. Among the key molecules produced by

4 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

osteocytes are RANKL59, inhibitors of the WNT signalling been observed in women with osteoporosis, which
pathway (including sclerostin and Dickkopf 1) and fibro- suggests that there is decreased c­ onnectivity between
blast growth factor 23 (FGF23), a regulator of phosphate ­adjacent osteocytes52.
homeostasis (FIG. 3). Oestrogen deficiency is associated
with impaired mechanosensing by osteocytes; oestro- Immune response and microenvironment. Several
gens were shown to inhibit the production of sclerostin inflammatory pathways are negatively controlled by
by osteoblastic lineage cells, thus providing a rationale oestrogens and contribute to a pro-inflammatory milieu
for the bone-forming ability of oestrogens60,61 (FIG. 5). in the acute phase of oestrogen deficiency 49. Extensive
In mice that systemically lack ERα, mechanical load- characterization of the RANKL–RANK pathway 57 and
ing did not evoke an adequate bone-anabolic response, identification of the role of activated T cells and their
indicating that functioning ER signalling is required for prominent cytokine TNF on osteoclast biology 49 have
mechanosensing and mechano­transduction62. Of note, led to the emerging concept of osteoimmunology — the
structural abnormalities in the osteocytic network have mutual interactions between the immune system and
bone metabolism. Rodents without appropriate T cell
activation, or defective RANKL or TNF production or
action fail to enhance osteoclastic bone resorption and
MSC HSC are protected against bone loss after ovariectomy 38,49,57.
Most of these findings are based on ovariectomy models
of inbred mouse strains and thus need to be cautiously
RANK interpreted. In humans, RANKL levels are upregulated in
Pre-osteoblast Pre-osteoclast
MSCs and T cells as well as in B cells immediately after
menopause and these levels return to ­premenopausal
­levels with oestrogen replacement therapy 49,58.

Alternative mechanisms. The oestrogen-centric para­


RANKL digm of bone loss in postmenopausal osteoporosis
M-CSF has recently been challenged and ageing and oxidative
stress have been proposed as alternative mechanisms
Osteoclast based on experiments in animal models35. These con-
Osteoblast
cepts are not mutually exclusive. In fact, some of the
OPG non-genomic effects of the ER on osteoblast apoptosis
have been linked to the attenuation of reactive oxygen
Cathepsin K species (ROS) production in mice63. In line with this,
inhib­ition of NADPH oxidase 4 (NOX4), an enzyme
Cl– that generates ROS, protected mice from ovariectomy-­
H+ induced trabecu­lar bone loss64. In summary, oestro­gen
Cl–
H+ deficiency contributes to a pro-inflammatory milieu,
Sclerostin with enhanced activities of osteoclastogenic cytokines,
Bone formation Bone resorption ­activated T cells and higher levels of ROS.
Osteocyte
Bone Diagnosis, screening and prevention
Diagnosis
Figure 3 | Bone remodelling. In the adult skeleton, bone is constantly
Nature Reviewsremodelled at
| Disease Primers Postmenopausal women or men >50 years of age who
multiple sites by the coordinated and sequential actions of bone-resorbing osteoclasts are at risk of having osteoporosis should be assessed.
and bone-forming osteoblasts to repair microdamage and to adapt bone structure to The risk is evaluated in the individual based on the
the mechanical and metabolic needs. Osteocytes represent >95% of all bone cells, are
presence of clinical risk factors, which include previ-
embedded in the mineral, function as mechanosensors and fine-tune bone remodelling
by osteoblasts and osteoclasts. Osteoblasts are derived from mesenchymal stem cells ous fragility fracture, genetic predisposition, smoking,
(MSCs) and are specialized to produce an extracellular bone matrix, including type I >14 units of alcohol per week, early-onset menopause
collagen, and non-collagenous proteins, including osteocalcin, osteonectin and (<45 years of age), low BMI (<19 kg per m2) or other fac-
osteopontin. The bone matrix is subsequently mineralized and stiffened by deposition tors involved in secondary osteoporosis (BOX 1). General
of calcium hydroxyapatite. WNT signalling, and its inhibitors, has an important role in screening of populations for osteoporosis is not recom-
osteoblast differentiation. Osteoclasts are derived from haematopoietic stem cells mended because the risk of fractures in postmeno­pausal
(HSCs) of the macrophagic and monocytic lineage. Differentiation from precursor cells women without clinical risk factors with a T-score of
towards activated multinucleated cells is crucially dependent on receptor activator of ≤–2.5 is lower than in women with the same T-score
nuclear factor‑κB (RANK) ligand (RANKL) that acts on osteoclastic RANK, and who have risk factors65–67.
permissive levels of macrophage colony-stimulating factor (M‑CSF). Osteoprotegerin
Osteoporosis is defined by BMD at the lumbar spine,
(OPG; also known as TNFRSF11B) functions as a decoy to block this interaction. RANKL
is mainly produced by osteoblastic lineage cells (MSCs, osteoblasts and osteocytes) and total hip or femoral neck of 2.5 standard deviations or
lymphocytes57. Mature bone-resorbing osteoclasts are large multinucleated cells. Using more below the population mean for young healthy
a sealing zone to attach at the bone surface and augmenting their surface with a ruffled adults (peak bone mass); the difference in standard
border, mature osteoclasts secrete HCl to create an acidic microenvironment where deviations is defined as the T-score68. BMD is measured
enzymes such as cathepsin K, which degrades type I collagen, are most active. by dual-energy X‑ray absorptiometry (DXA). DXA is

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 5


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

an X‑ray-based technique that quantifies bone mineral history into account, including clinical and laboratory
content (BMC) by using two X‑ray sources of different investigations. For example, in patients with back pain,
energies allowing subtraction of the attenuation of radi- loss of height and a difference in BMD between adjacent
ation by soft tissues from the total attenuation to deter- vertebrae of >1 standard deviation, the presence of a
mine the attenuation caused by bone mineral. By adding verte­bral fracture should be assessed. Prevalent vertebral
information about the area of the bone investig­ated, areal fractures can be investigated either by a radiograph of the
BMD can be calculated as BMC divided by bone area. spine or by assessment of vertebral fractures using DXA
The technique is very precise, fairly fast and exposes the with similar diagnostic accuracy.
patient to very low doses of radiation. These character­ The level of bone turnover can be determined by
istics in combination with the fact that BMD is the single measuring bone turnover markers. The recommended
best predictor of fracture risk69,70 are the main reasons markers are serum procollagen type I amino‑terminal
for choosing BMD as the diagnostic criterion for osteo­ propeptide, which is a marker of bone formation, and
porosis. There have been discussions whether the diag- serum carboxy‑terminal collagen crosslinks, which
nostic criteria should be expanded to reflect treatment are markers of bone resorption71. The bone turnover
indications and reimbursements4, but as these vary markers are not used for diagnosis but are helpful in the
worldwide, the above-mentioned definition is still valid. monitor­ing of treatment response and the identifi­cation
A prerequisite for the definition of osteoporosis on the of failure to respond72. The greater the reduction in bone
basis of BMD is that other modifiable causes of low BMD turnover markers with anti-resorptive treatment, the
are excluded. Secondary osteoporosis (BOX 1) should be greater the fracture risk reduction71. The characterization
investigated and any underlying disorder treated. Renal of the disease will enable more relevant treatment deci-
osteodystrophy can be difficult to exclude in patients sions. If bone turnover is high, a greater BMD response
with chronic kidney disease (stage 4 and stage 5), as bio­ can be expected to both anti-resorptive and anabolic
chemical markers of bone turnover are more difficult to treatments than if bone turnover is low (in which case
interpret in these patients. A bone biopsy might therefore observation without treatment might be appropriate).
be needed to diagnose the patient correctly. As part of the
diagnostic work‑up, it is important to take the full patient Prevention
Prevention of osteoporosis comprises primary and
Men Phase 2
secondary prevention. Primary prevention is directed
Periosteal surface towards the general population, whereas secondary
prevention focuses on individuals who are at increased
risk of developing osteoporosis because of the presence
of clinical risk factors. Treatment of patients with estab-
lished osteoporosis is sometimes referred to as tertiary
prevention (discussed below).
Prevention of osteoporosis can generally be achieved
through distinct mechanisms. One mechanism to pre-
Endocortical surface vent osteoporosis is to augment bone gain during child-
hood and adolescence and the peak bone mass that the
Age individual is genetically determined to achieve. This
18 50 60 75 (years)
is important as modelling suggests that the onset of
Menopause osteo­porosis will be delayed by 13 years by increasing
(only in women) peak bone mass by 10%73. The other mechanism to
prevent osteoporosis is by minimizing the menopause-­
related and age-related bone loss. In addition to meas-
ures that affect BMD, osteoporotic fractures can be
prevented by reducing the risk of falls. Nutritional
­interventions, maintenance of a BMI of >20 kg per m2,
regular physical activity, moderation of alcohol intake,
Women Phase 1 Phase 2
falls prevention and smoking cessation are all impor-
tant in the p
­ revention of osteoporosis and osteoporotic
Figure 4 | Sex-specific differences in bone modelling (and remodelling).
Nature Reviews | Disease Primers fractures (BOX 2).
a | Bone modelling (and remodelling) in men. During puberty, androgens promote a
marked periosteal apposition, leading to an outwards displacement with wider bone and Nutrition and/or supplementation. Nutrition includes
thicker cortices. Despite some endocortical resorption during adulthood, bone strength an endless number of components of which many are
is mostly maintained during ageing, until cortical thinning and cortical porosity develop. likely to be important for bone mass and quality. Calcium
b | Bone modelling (and remodelling) in women. During puberty, women have more
and vitamin D supplementation have been researched
endocortical rather than periosteal bone apposition, yielding more gracile bones, than
men do. In the first phase of bone loss after menopause, oestrogen deficiency markedly the most. The suggestion that calcium supplementation
stimulates endocortical resorption and mitigates periosteal apposition, thus causing may prevent osteoporosis is based on the underlying
rapid cortical thinning. The effects on trabecular bone loss during this phase are even physiology: calcium insufficiency leads to secondary
greater. In a second phase, endocortical resorption continues at a slower rate and hyperparathyroidism, which again leads to increased
cortical porosity develops, rendering the bone more prone to fragility fractures. bone resorption. However, evidence for a preventive

6 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

a With oestrogen b Oestrogen deficiency


MSC HSC ↑ Activated
Oestrogen T cells

Active
Pre-osteoblast T cell Pre-osteoclast

Oestrogen
Oestrogen
WNT WNT
Osteoblast Sclerostin RANKL ↑ Sclerostin ↑ RANKL
Apoptosis

Bone ↑ Bone formation

Osteoclast ↑↑ Bone resorption

Osteocyte ↓ Mechanosensing

Oestrogen Oestrogen Bone loss

Figure 5 | Role of oestrogens in bone remodelling. a | Physiological bone remodelling. Oestrogens enhance bone
formation by stimulating osteoblast differentiation and function and suppress osteoclast differentiation
Nature Reviewsand activityPrimers
| Disease
through direct cellular effects and reduced production of receptor activator of nuclear factor‑κB ligand (RANKL) by
activated T cells, mesenchymal stem cells (MSCs), osteoblasts and osteocytes. In addition, oestrogens are required for
adequate mechanosensing and mechanotransduction by the osteocytes. b | Oestrogen deficiency. RANKL production is
upregulated translating into a higher osteoclast number and activity. Concurrently, the function of individual osteoblasts
is suppressed, partly by upregulation of the WNT inhibitor sclerostin, whereas the total number of osteoblasts increases.
The role of osteocytes in mechanosensing is also impaired. HSC, haematopoietic stem cell.

effect of calcium supplementation on the development Vitamin D deficiency (serum 25‑OH vitamin D lev-
of osteoporosis is weak. Calcium intake either as dairy els of <30 nmol per l) leads to rickets in children and
products or supplements has in some studies been osteomalacia in adults and should therefore be avoided.
found to be positively associated with bone gain dur- In population-based studies, vitamin D insufficiency
ing childhood and adolescence74,75; however, this has (serum 25‑OH vitamin D levels of <50 nmol per l) has
not been confirmed in a meta-analysis of randomized been shown to lead to increases in PTH secretion and
clinical trials in children76. A population-based study in it is therefore recommended to ensure sufficient intake
the United States showed an association between cal- of vitamin D to reach a serum level of vitamin D that is
cium intake and bone mass in adults with vitamin D >50 nmol per l. However, it is probably not a vitamin D
insufficiency (serum 25‑hydroxy vitamin D (25‑OH level of <50 nmol per l that has a risk for osteoporosis
vitamin D) levels of <50 nmol per l), but not in those and fracture, but the secondary increase in the levels of
with normal vitamin D levels (>50 nmol per l)77, but PTH. A meta-analysis showed an approximately 20%
a meta-analysis of randomized clinical trials in adults reduction in non-vertebral and hip fractures in individ­
found that calcium supplementation was associated with uals and patients who were supplemented with vit­
increases in BMD78. Increased calcium intake or sup- amin D alone or in combination with calcium86. A dose
plementation cannot prevent menopause-related bone dependency of the effects was observed; only studies
loss but has, in some studies, been shown to reduce age-­ providing a daily dose of >20 μg (800 IU) showed preven-
related bone loss79. Finally, meta-analyses of the effect tion of fractures and a serum level of 25‑OH vitamin D
of calcium supplementation in adults have not shown of >75 nmol per l was needed to prevent hip fractures.
any effect on fracture prevention78,80. In addition, cal- Another meta-analysis, which included postmenopausal
cium supplementation might be associated with adverse women as well as older men (mean or median popula-
effects. Calcium supplementation might increase the risk tion of >65 years of age) concluded that supplementation
of myocardial infarction81, but meta-analyses82,83 produce with vitamin D alone is unlikely to prevent fractures, but
inconsistent results depending on how the data from supplementation with vitamin D in combination with
the Women’s Health Initiative (WHI) study are treated. calcium is likely to do so87. The underlying mechanism
In the WHI study, calcium and vitamin D supplemen­ is probably a combination of effects of vitamin D on
tation were associated with a 17% increased risk of bone metabolism and muscle function. Vitamin D defi-
nephro­lithiasis84; the same is not seen in ­individuals with ciency leads to reduced muscle strength as a result of a
a high-calcium diet 85. ­reduction in type IIa muscle fibres88.

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 7


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Box 1 | Drugs and diseases associated with increased fracture risk


• Drugs • Rheumatological diseases
-- Glucocorticoids -- Rheumatoid arthritis
-- Aromatase inhibitors -- Ankylosing spondylitis
-- Proton pump inhibitors • Haematological diseases
-- Glitazones (also known as thiazolidinediones) -- Multiple myeloma
-- Anti-epileptics -- Systemic mastocytosis
-- Loop-acting diuretics -- Leukaemia
-- Heparin and warfarin
• Neurological diseases
-- Antidepressants
-- Stroke
• Endocrine diseases -- Immobilization
-- Anorexia nervosa -- Parkinson disease
-- Hypogonadism
• Other diseases
-- Cushing syndrome
-- Congestive cardiac failure
-- Vitamin D deficiency
-- Chronic obstructive lung disease
-- Hyperparathyrodism
-- Chronic renal insufficiency
-- Hyperthyroidism
-- Osteogenesis imperfecta
-- Diabetes mellitus (type 1 and type 2)
-- Marfan syndrome
• Gastrointestinal diseases -- Ehlers–Danlos syndrome
-- Malabsorption -- Human immunodeficiency virus infection
-- Inflammatory bowel diseases (and its treatment)

Observational studies have shown that other nutri- still lacking. Whole-body vibration has in some stud-
ents might influence calcium metabolism and therefore ies been shown to reduce falls and improve BMD, but
potentially the risk of osteoporosis. Intake of vegetables not ­consistently 107,108, and no effect on fracture risk has
and fruits leads to alkalization of the urine owing to the been shown.
high content of potassium and thereby decreases cal-
cium excretion, whereas intake of meat and fish acid- Smoking. Smoking, current as well as past, has in a
ifies the urine via the sulfur-rich amino acids and can meta-analysis been shown to be associated with increased
cause hypercalcuria89,90. However, the effects might only risk of fractures, including hip fractures109. Smoking is
be minimal as it has been shown that these effects on associated with impaired intestinal calcium absorption
urinary calcium excretion are balanced by changes in and increased age-related bone loss110. Smoking impairs
calcium absorption91. High protein intake accompanied the effects of postmenopausal hormone replacement on
by low calcium intake is associated with an increased risk bone mass, probably through liver induction of metab-
of fractures, whereas a high protein intake accompanied olizing enzymes and thereby increased metabolism
by a high calcium intake is associated with a reduced risk of oestrogen111. Smoking cessation has been found to
of fracture92,93. improve BMD in postmenopausal women, probably
through changes in sex hormone levels and thereby
BMI. Body weight and body composition is also of rel- bone turnover 112.
evance in the prevention of osteoporosis. Weight loss
in older women is associated with a general increased Fall prevention. The main complication of osteoporo-
fracture risk94, whereas weight gain is associated with sis is fractures. The risk of hip fracture and other non-­
reduced risk of hip fractures but increased risk of other vertebral fractures is closely associated with the risk
peripheral fractures95,96. Sarcopaenia is associated with of falling 95,113. It is therefore relevant to investigate the
increased risk of fractures97, although sarcopaenia is risk of falling in patients with osteoporosis. Often, an
very poorly defined. According to some definitions, it is increased risk of falling is part of more general frailty.
defined as poor muscle function, either because of low However, sometimes the risk of falling is associated with
muscle mass or impaired function of the muscles. specific pathologies, such as reduced vision, cardiac
arrhythmias or medical treatments (for example, anti-
Physical activity. The bone cells, especially osteocytes, hypertensives and sedatives). The risk of falling may be
respond to mechanical strains or absence of such98,99 improved considerably by addressing these conditions.
(FIG. 3). Clinical studies investigating the effect of physical Even in individuals without these conditions, recurrent
training have shown that it is possible to increase BMD, falls may be prevented by multifactorial approaches114.
especially in younger individuals (<25 years of age)100–102. Although, a considerable reduction in falls would be
Although some evidence suggests that the skeleton expected to reduce fractures, this association remains to
responds less well to physical activity with increasing be proven for the majority of fall prevention strat­egies.
age103, some studies show an effect of exercise on BMD in Two strategies have been shown to prevent both falls and
postmenopausal women104–106. However, positive effects fractures. Cataract surgery has been shown to reduce
on BMD are lost if the exercise programme is stopped. falls by 34% and fractures by 46% in elderly, mainly
Evidence that physical training prevents fractures is white, women115. Randomized clinical trials investigating

8 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

the effects of vitamin D supplementation on the risk denosumab have been shown to have broad anti-fracture
of falls and fractures and meta-analyses of these have efficacy (for vertebral, non-vertebral and hip fractures)
shown that vitamin D supplementation decreases the and are approved for use. Generally, bisphosphonates are
risk of falling and non-vertebral fractures, including used as a first-line treatment, because they are less expen-
hip fractures, by approximately 20%86,116; these effects sive than denosumab and their efficacy is comparable.
were only seen with doses of vitamin D of >17.5–20 μg Some bisphosphonates are available as generics, which
(700–800 IU) per day. Later analyses of the same clinical increases ease of access. Thus far, the only approved
studies have come to different conclusions regarding the agent that stimulates bone formation is teriparatide and
consistency of the above-mentioned findings; the dif- intact PTH; the others are anti-resorptive agents.
ferences are mainly caused by a difference in the criteria
for inclusion or exclusions of studies, as some analyses Bisphosphonates. Bisphosphonates (including oral alen-
included all studies, whereas others only included studies dronate, oral risedronate, oral or intravenous ibandro-
using medium-dose or high-dose supplementation, good nate, and intravenous zoledronate) have been the most
adherence and high-quality assessment of falls117–120. widely used agents for managing osteoporosis over the
past two decades. They bind avidly to bone mineral
Management and are subsequently ingested by osteoclasts, the bone-­
Patient selection resorbing activity of which is suppressed through the
Following the practice established in cardiovascular inhibition of a crucial enzyme in the mevalonate path-
disease, it is now common to make treatment decisions way 125. Their efficacy in fracture prevention is shown
for osteoporosis on the basis of estimated event risk, in in FIG. 6, along with that of other agents. Although the
this case, the risk of a fracture. This strategy acknow­ reduction in non-vertebral fractures is only small, they
ledges that several factors, including age, determine typically reduce hip fracture risk by about 50%126 and
fracture risk, not just BMD or previous fracture history. vertebral fracture risk by 40–70%127. Cost, convenience,
Two widely used fracture risk calculators are WHO compliance and adverse-effect profiles usually guide the
FRAX3 and that from the Garvan Institute121 (TABLE 1). choice between intravenous and oral bisphosphonates.
As they use different clusters of risk factors, they dif- Oral bisphosphonates cause gastrointestinal disturb­
fer in treatment recommendation in approximately ance in about 20% of users, which probably contributes
25% of patients122. In patients with borderline fracture to poor long-term compliance with these drugs in some
risk, the use of more than one calculator may permit a reports. About one-third of patients having their first infu-
more comprehensive appreciation of their real risk. The sion of an intravenous bisphosphonate will experience a
choice of threshold for intervention should be based on transient, flu-like illness. In addition, intravenous bis-
a cost-effectiveness analysis, which is influenced not phosphonates carry a risk of acute renal failure in patients
only by fracture risk but also by the cost and the safety with severe pre-existing kidney disease (glomer­ular fil-
of each intervention. The use of a single intervention tration rate of <35 ml/min). Adequate renal ­function is
criterion is not appropriate for agents of widely different necessary for use of intravenous bisphosphonates.
cost. A 10‑year hip fracture risk of 3% has been advo- The occurrence of osteonecrosis of the jaw in patients
cated as a criterion for treatment123, although it should be with cancer receiving high doses of intravenous bisphos­
noted that this will result in the treatment of individuals phonates has caused concern that this may also be a
whose BMD and fracture history would not have quali- consequence of the lower doses used in osteoporosis
fied them for entry to most of the pivotal phase III trials treatment with these agents. Although osteo­necrosis has
of the agents we are using, so this practice is not strictly been described in patients with osteoporosis, its inci-
evidence-based. dence is very low 128 and in some studies does not seem
to be greater in those who receive bisphosphonates
Management than in patients with osteoporosis who are not taking
When considering pharmaceutical intervention, gen- these drugs129–131.
eral preventive measures (as discussed above) should
be implemented. Vitamin D (800 IU daily) should be
prov­ided for those at risk of osteomalacia (for example, Box 2 | Prevention strategies for at‑risk women
the frail elderly who are housebound or institutional-
Recommendations for postmenopausal women with
ized). The issue of whether to give calcium supplements
documented osteoporosis or increased risk of
to patients along with an anti-resorptive drug remains developing osteoporosis:
controversial and may be more suitable for those with
• Sufficient daily intake of calcium (800–1,000 mg per day),
low habitual dietary calcium intake. Calcium supple- preferentially through diet
ments have traditionally been provided when anti-­
• Vitamin D supplementation in those who are likely
resorptive drugs are prescribed, but fracture prevention to be deficient in vitamin D; dosage to achieve serum
has been shown in the absence of calcium and calcium 25‑hydroxy vitamin D3 levels of >50 nmol per l
does not seem to influence the effects of these agents • Avoid weight loss and low body mass index
on BMD124.
• Daily physical activity (weight-bearing, such as
Although there are a range of agents available for skipping, steps and walking)
treating osteoporosis, only the potent bisphospho-
• Cessation of smoking
nates (alendronate, risedronate and zoledronate) and

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 9


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

However, atypical femoral fractures are a concern. is combined with teriparatide140, although the effect of
These seem to develop initially as stress fractures in the the combination on fractures is not known.
lateral cortex of the femoral shaft, often spontaneously
progressing to transverse fractures132,133. Bisphosphonate Teriparatide. Teriparatide, the N‑terminal 34 amino
use is more common in patients with these fractures acids of PTH, stimulates both bone formation and
(78%) than in those with ‘typical’ fractures of the fem- bone resorption. Daily or weekly subcutaneous injec-
oral shaft (10%), and incidence increases with time tions result in progressive increases in spine BMD, with
on these drugs132. Atypical femoral fractures might be smaller increases in the hip141. Teriparatide at a dose of
more common with alendronate than with risedron­ 20 μg per day reduced the risk of vertebral fractures by
ate or zoledronate134 and Asian patients may be more 70% and non-vertebral fractures by 50% over a median
susceptible135. Incidence seems to drop dramatically in treatment period of 21 months142, and weekly dosing with
the 1–2 years after drug discontinuation132. However, 56.5 μg over 72 weeks reduced vertebral but not non-­
atypical femoral fractures represent a small minority vertebral fractures (relative non-vertebral fracture risk:
of total femoral fractures132, so bisphosphonates are 0.98 (95% CI: 0.47–2.07))143. Teriparatide and intact PTH
believed to prevent many more fractures than they have not been shown to reduce hip fracture risk. Nausea,
might cause in those with increased fracture risk. headache and hypercalcaemia have caused withdrawals
It is important to periodically review the need for from clinical trials of teriparatide and intact PTH, and
continued bisphosphonate therapy, to consider drug there are post-marketing anecdotes of musculoskeletal
discontinuation in patients whose T‑score has risen pain. Despite concern from rat studies that parathyroid
to >–2.5 and to consider temporary drug h ­ olidays peptides increase the incidence of osteosarcoma, there
in those who have been on treatment for >5 years. is no evidence of this from 10 years of human use144.
However, effective interventions must continue to be Combinations of teriparatide with bisphosphonates or
provided for patients with osteoporosis who have a high with denosumab are additive in their BMD effects145,146.
fracture risk.
Strontium. Strontium is a divalent cation with chemical
Denosumab. Denosumab is a monoclonal antibody and biological properties similar to those of calcium.
directed against RANKL, administered as 6‑monthly, Its ranelate salt is used to treat osteoporosis, adminis-
subcutaneous injections for osteoporosis treatment. tered daily in powder form. It produces large increases
It markedly reduces bone turnover, results in sustained in BMD, which are at least in part an artefact of imag-
and substantial increases in BMD and achieves a similar ing owing to heavier strontium ions replacing calcium
fracture reduction as seen with zoledronate136,137 (FIG. 6). in hydroxyapatite. It has a weak anti-resorptive effect
Osteonecrosis of the jaw and atypical femoral fractures based on results of bone turnover markers147. A study
have been reported in patients treated for osteoporosis of 170 paired biopsy samples before and after stron-
with denosumab and the low incidences are compar­ tium treatment showed reductions in several ­formation
able to those seen with bisphosphonates137. There is ­indices at 6 and 12 months (including bone forma-
a rebound in bone turnover and a rapid loss of BMD tion rate), no consistent changes in resorption and
within months of a 6‑monthly injection being missed138 reduced bone volume at 12 months148. Vertebral fracture
and there have been case reports of increased vertebral ­numbers were decreased by 40%149 and non-vertebral
fracture risk following denosumab cessation139. BMD fractures by 16%150, but hip fracture was only decreased
studies have shown additive effects when denosumab in a post-hoc subgroup analysis150.

Table 1 | Comparison of FRAX and Garvan tools


Parameters FRAX tool* Garvan tool‡
Clinical risk • Age (40–90 years) • Age (60–96 years)§
factors • Sex • Sex
• Weight • Fractures since 50 years of age (up to 3)
• Previous fracture in adulthood (yes or no) • Falls over the past 12 months (up to 3)
• Family history of hip fractures
• Current smoking
• Glucocorticoid use
• Rheumatoid arthritis
• Secondary osteoporosis
• Alcohol
Other • Incorporates competing risk of mortality • No competing risk of mortality
considerations • Different algorithms can be used for different • No different algorithms for different
ethnicities and countries ethnicities
• Probably involves interactions between variables • No interactions between individual variables
Types of fracture • Hip fracture at 10 years • Hip fracture at 5 and 10 years
predicted • Major osteoporotic fracture at 10 years • Any fragility fracture at 5 and 10 years
FRAX, Fracture Risk Assessment Tool. *See REF. 3. ‡See REF. 121. §Since the time of data collection and analyses, the Garvan
calculator has expanded its eligible age range to include individuals 50–59 years of age. Reproduced with permission from
REF. 122, Maturitas–Elsevier.

10 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

Non-vertebral fracture Hip fracture Vertebral fracture


Calcium
Vitamin D
Calcium and vitamin D
Alendronate
Risedronate
Bisphosphonates
Ibandronate
Zoledronate
Denosumab
Teriparatide
PTH1–84
Strontium
Raloxifene
Odanacatib

0.2 0.5 1 2 0.6 0.2 0.5 1 2 4 8 0.1 0.2 0.5 1 2


Favours Favours Favours Favours Favours Favours
treatment control treatment control treatment control
Figure 6 | Fracture prevention by osteoporotic agents. Summary of trial data documenting Naturethe efficacy| Disease
Reviews of treatments
Primers
for the prevention of non-vertebral hip fractures and vertebral fractures. The results for strontium150, odanacatib199
and parathyroid hormone (PTH) 1–84 (REF. 200) are relative risks. Other data are odds ratios and from REF. 126. Values are
shown with 95% confidence intervals201.

Considerable safety concerns are associated with Selective oestrogen receptor modulators. Selective ER
strontium treatment. It causes rare severe dermato­ modulators (SERMs; for example, raloxifene, bazedox­
logical reactions 151, but more concern relates to a ifene and lasofoxifene) are compounds that inter-
60% increase in the risk of myocardial infarction in act with ERs and that show a range of tissue-specific
the original pivotal trials152. This finding caused the agonist and antagonist effects. Raloxifene has been
Pharmaco­v igilance Risk Assessment Committee of widely used in osteoporosis, but its effects on BMD are
the European Medicines Agency to recommend suspen- consider­ably less than those of oestrogen158. Although
sion of the medicine because of its cardiovascular risk, raloxifene reduces vertebral fracture numbers, it has
but the Agency subsequently continued availability of no effect on hip and other non-vertebral fractures159.
the med­icine, restricting its use to patients who ­cannot It is well tolerated but has been shown to increase the
be treated with other medicines that are approved for risk of thromboembolic disease in two different studies,
osteoporosis, subject to their regular screening and with a relative risk of 3.1 (95% CI: 1.5–6.2) found in
monitoring to exclude cardiovascular disease153. The one159 and a hazard ratio of 1.44 (95% CI: 1.06–1.95)
combination of limited efficacy, low compliance and found in another 160. In addition, the risk of fatal
major safety concerns results in low enthusiasm for its stroke was increased in one study (hazard ratio: 1.49
use in osteoporosis treatment. (95% CI: 1–2.24))160.
Bazedoxifene is marketed in combination with con-
Oestrogen replacement therapy. Oestrogen replace- jugated oestrogens. This preparation has been shown to
ment has been shown to prevent postmenopausal bone improve vasomotor symptoms, vulvovaginal ­atrophy
loss and to reduce fracture risk154–156. However, oestro­ and BMD, without adverse effects on the breast and
gen has effects on many tissues, some positive and endo­metrium161,162. It has positive effects on BMD,
some negative, creating a complex risk–benefit situ­ which are less than those of potent anti-resorptives163,
ation. In the WHI study, beneficial effects on fractures but its anti-fracture efficacy is unknown. Bazedoxifene
and colon cancer incidence were counterbalanced by alone has been shown to reduce vertebral fracture risk
changes in cardiovascular, cerebrovascular and venous by about 40%164. Lasofoxifene is the only SERM that
thromboembolic events155,156. This balance is likely to has been shown to reduce the risk of non-vertebral
be different for oestrogen alone and for oestrogen plus ­fractures165, but it is not yet marketed.
progestins and may also vary between specific oestro-
gens and progestins. This complexity has caused many Quality of life
patients and physicians to lose enthusiasm for oestrogen Osteoporosis is often a silent disease; loss of BMD by
as a bone-protective therapy, even though its efficacy itself rarely affects quality of life. However, the effect of
is beyond question. A reappraisal of the literature on fractures associated with osteoporosis on health-­related
this topic suggests that, in the small number of women quality of life (HRQOL) is typically profound166. HRQOL
who have increased fracture risk in the decade follow- reflects the following dimensions of well-­being: physi-
ing menopause, the balance of benefit versus risk is cal, psychological, social and financial167. Research has
­probably positive157. shown that low-trauma fractures initiate the downward

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 11


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

a 3,500 b 20
18
3,000
Incidence rate per 100,000

16
2,500 14

Prevalence (%)
2,000 12
10
1,500 8

1,000 6
4
500
2
0 0
86
87
88

19 9
90

19 1
92

19 3
94

19 5
96

19 7

20 9
98

00

20 1
02

20 3
04
05

96
97
98
99
00
01
02
03
04
05
06
07
08
09
10
11
12
8

0
19
19
19
19

19

19

19

19

19

20

20

20

19
19
19
19
20
20
20
20
20
20
20
20
20
20
20
20
20
Year Year
65–74 years 75–84 years >84 years Women Men

Figure 7 | Evolution of fracture incidence and bisphosphonate use. a | Evolution of hipNature


fractureReviews | Disease
incidence by agePrimers
for men and women in the United States. b | Prevalence of bisphosphonate use among men and women ≥55 years of age.
Values are shown with 95% confidence intervals. Part a is reproduced with permission from JAMA, 2009, 302 (14):
1573–1579. Copyright © (2009) American Medical Association. All rights reserved. Part b is reproduced with permission
from REF. 181, American Society for Bone and Mineral Research.

spiral of quality of life that leads to worsen­ing prob- increase steadily 8,173. However, osteoporosis also has
lems in all four domains. All fractures have undesir­ indirect costs that are borne by individuals with frac-
able physical consequences; however, the physical and tures. These costs include lost wages, the need to hire
quality-­of-life consequences associated with spine people to complete home chores and other expenditures
and hip fractures are marked. Vertebral fracture out- that are not directly related to medical care. Estimates
comes include deformity, fear of falling, chronic pain suggest that indirect costs account for half of the total
and mortality, whereas hip fractures often cause activ- costs of osteoporosis174.
ities of daily living limitations, dysmobility, excessive Overall, osteoporotic fractures compromise HRQOL
morbidity and excess mortality, especially in the first in all domains. As the number of fractures increase,
year post-fracture168. HRQOL decreases, with hip and spine fractures having
The physical and functional outcomes of osteoporo- the biggest negative effect on HRQOL. Prevention of
tic fractures have been well studied, especially those fractures through screening and appropriate and rapid
associated with a hip fracture169 and/or two or more treatment of fractures when they occur might preserve
spinal fractures170. However, psychosocial and financial HRQOL and slow the downward spiral that can begin
consequences of these fractures are less well identified. with a single atraumatic fracture175.
In short, HRQOL is compromised in the psychological
realm by feelings of anxiety (for example, fear of falling Outlook
and fear of fractures) and depression. In fact, comorbid Trends in incidence
anxiety and depression occurs in patients with osteo- Since about 1976, the rates of hip fracture have been
porosis171. Reduced self-esteem and self-image can also declining by about 20% per decade in the United States
occur with increasing numbers of fractures167. (FIG. 7a), Canada and in most countries in Europe176,177.
The social consequences of osteoporosis are even Effective treatment can only account for a small fraction
less well understood and studied than the psycho­logical of this trend owing to the low prevalence of use of drugs
consequences. Perhaps the most serious is the loss of for osteoporosis178. Femoral neck BMD has also been
social roles; that is, the inability to continue to enact increasing in the United States during that period such
roles associated with employment, family leisure and that the prevalence of osteoporosis has decreased by
self-care activities, including hobbies, athletics, sexual more than half between 2002 and 2010 (REF. 179). Some
intimacy, bathing and dressing. Such limitations can take of the effect might be attributable to the rise in BMI,
away independence and feelings of self-worth and can but the reasons are not clear 180. Despite these trends, the
breed hopelessness. The interaction between these social global number of fractures will continue to rise rapidly
factors (such as loneliness and isolation) and psycho­ owing to the ageing population.
logical factors (such as anxiety and depression) leads to
an ever-deepening downward spiral172. Trends in management
Finally, the financial cost of osteoporotic fractures Since about 2005, prescriptions of drug treatments in the
adds yet another way in which HRQOL is impaired. United States have declined by 25–50% even in patients
Although there are few in‑depth studies of individual with hip fracture and particularly among women with
costs, evaluations of societal cost suggest that the eco- lower levels of education (FIG.  7b) . The decline is
nomic burden of osteoporosis is substantial8. Current probably mostly due to publicity about the occur-
and projected direct costs of fractures of which the rence of atypical femoral fractures and ­osteonecrosis
majority is covered by health care systems worldwide of the jaw with bisphosphonates and denosumab,

12 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

although the absolute risk in osteoporosis treatment Odanacatib, a cathepsin K inhibitor, suppresses
remains low 181. All bisphosphonates have become resorption of collagen matrix while allowing the forma-
generic, which is associated with reductions in price tion of bone. It might permit a more favourable balance
that means that treatment to prevent fractures is now of a small decrease in resorption with less inhibition of
more cost-effective and, therefore, should be more formation187. The LOFT trial found a 47% decrease in the
widely used. Efforts to better target the prescription of risk of hip fracture and a 54% decrease in the risk of verte­
treatments have included institution of ‘fracture liaison’ bral fractures188. The safety profile has resulted in halting
services in some hospitals to initiate drug treatments, or of drug development as there was an excess incidence of
to measure BMD and risk, in patients who are admit- stroke189; there were also rare instances of ­atypical femur
ted for fracture182. This promising approach has had fractures and morphoea-like skin lesions188.
mixed results because of the expense of employing a
clinical specialist to identify patients and because most Guidelines for treatment and screening
patients who start treatments after fracture discontinue The National Osteoporosis Foundation (United States)
them within a year. This is a particular concern with guidelines have been updated with a new emphasis on
denosumab, as there is an accelerated bone loss138 and screening for vertebral fractures using vertebral imag-
probable increase of multiple vertebral fractures after ing by DXA or radiograph in patients who are at risk
stopping therapy 139. of vertebral fractures as a result of older age, low BMD,
≥2 cm height loss or history of a fracture along with the
New treatments measurement of BMD190; treatment is recom­mended
The ultimate aim in osteoporosis management would with T‑scores of ≤–2.5 at any skeletal site independent
be to develop a treatment that reduces fracture risk, of age, a vertebral fracture, or a BMD T‑score between
­especially non-vertebral and hip fractures, by improving –1 and –2.5 along with an increased risk of fracture
bone mass, muscle strength and endurance. This would by the FRAX algorithm. By contrast, the National
reduce the risk of injurious falls, shorten rehabilitation Osteoporosis Guideline Group (United Kingdom) guide-
time and reduce disability due to fractures. Although lines e­ mphasize the assessment of risk using the FRAX
we are not there yet, several treatments with novel algor­ithm and treating on the basis of risks that are spe-
­mechanisms of action are due to make their debut soon. cific to age groups191. Besides these, there are guidelines
The anti-sclerostin antibody romosozumab has been of the International Osteoporosis Foundation192 and
shown to markedly increase bone formation for up to >60 available national guidelines193 that take account of
6 months after the initial dose and the highest dose local practices.
increases spine and hip BMD by about 11% and 4%,
respectively, over just 12 months of monthly sub­cutaneous Goal-directed treatment
administration183. Preliminary reports from the FRAME It has been proposed to treat patients with the aim to
study indicate that 1 year of treatment reduces the risk reach target levels of BMD, for example, a T‑score of
of vertebral fracture by 75% but with no significant ≥–2.5 at any skeletal site or a fracture risk that is lower
reduction in risk of non-vertebral fractures184. Complete than the level that would indicate starting treatment 194.
results from ongoing trials will determine whether In this scheme, the choice of drug would be based on
short-term treatment with an anti-­sclerostin antibody the likelihood of a patient reaching this goal using the
offers greater anti-fracture efficacy than ­denosumab treatment options. Achievement of BMD goals dur-
or zoledronate. ing treatment could guide when to pause or switch treat-
Abaloparatide, an analogue of PTH-related pro- ment. For denosumab and zoledronate, improvement in
tein, functions similarly to teriparatide. Treatment for BMD is accompanied by a decrease in fracture risk195,196;
18 months with the doses intended for use in practice this would support using changes in BMD as a goal to
tended to increase BMD of the spine and hip to a greater advise management choices. However, the association
degree than teriparatide185. The ACTIVE trial showed between BMD and fracture risk is not strong for other
an 86% reduction in the risk of vertebral fractures and treatments and the change in BMD will not be so pre-
a 43% decrease in the risk of non-vertebral fractures186. cise as to predict changes in the risk of fracture. Thus,
Full details about its safety have not been published, but, goal-directed treatment would be best if both improve-
if approved for use, abaloparatide might be a new option ments in BMD and data about how treatments reduce
for treatment of patients at high risk of fracture. risk were considered.

1. [No authors listed.] Consensus development 4. Siris, E. S. et al. The clinical diagnosis of osteoporosis: and the European Federation of Pharmaceutical
conference on osteoporosis. Hong Kong, April 1–2, a position statement from the National Bone Health Industry Associations (EFPIA). Arch. Osteoporos 8,
1993. Am. J. Med. 95, 1S–78S (1993). Alliance Working Group. Osteoporos Int. 25, 136–136 (2013).
2. Kanis, J. A. Assessment of fracture risk and 1439–1443 (2014). 8. Burge, R. et al. Incidence and economic burden of
its application to screening for postmenopausal 5. Eastell, R. Treatment of postmenopausal osteoporosis. osteoporosis-related fractures in the United States,
osteoporosis: synopsis of a WHO report. N. Engl. J. Med. 338, 736–746 (1998). 2005–2025. J. Bone Miner. Res. 22, 465–475
Osteoporos Int. 4, 368–381 (1994). 6. Office of the Surgeon General (US). Bone Health (2007).
This paper summarizes the key report from the and Osteoporosis: A Report of the Surgeon General 9. van Staa, T. P., Dennison, E. M., Leufkens, H. G.
WHO that defined osteoporosis on the basis of (Office of the Surgeon General, 2004). & Cooper, C. Epidemiology of fractures in England
a bone density T‑score. 7. Hernlund, E. et al. Osteoporosis in the European and Wales. Bone 29, 517–522 (2001).
3. Kanis, J. A. FRAX: WHO Fracture Risk Assessment Union: medical management, epidemiology and 10. Cheng, S. Y. et al. Geographic trends in incidence
Tool. QHO https://www.shef.ac.uk/FRAX/tool.jsp economic burden. A report prepared in collaboration of hip fractures: a comprehensive literature review.
(2016). with the International Osteoporosis Foundation (IOF) Osteoporos Int. 22, 2575–2586 (2011).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 13


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

11. Cauley, J. A., Chalhoub, D., Kassem, A. M. pathogenesis of osteoporosis. Endocr. Rev. 31, 61. Fujita, K. et al. Effects of estrogen on bone mRNA
& Fuleihan, G. E.‑H. Geographic and ethnic disparities 266–300 (2010). levels of sclerostin and other genes relevant to bone
in osteoporotic fractures. Nat. Rev. Endocrinol. 10, 36. Seeman, E. & Delmas, P. D. Bone quality — the metabolism in postmenopausal women. J. Clin.
338–351 (2014). material and structural basis of bone strength and Endocrinol. Metab. 99, E81–E88 (2014).
12. Johnell, O., Gullberg, B., Allander, E. & Kanis, J. A. fragility. N. Engl. J. Med. 354, 2250–2261 (2006). 62. Lee, K., Jessop, H., Suswillo, R., Zaman, G.
The apparent incidence of hip fracture in Europe: 37. Manolagas, S. C., O’Brien, C. A. & Almeida, M. & Lanyon, L. Endocrinology: bone adaptation requires
a study of national register sources. Osteoporos Int. The role of estrogen and androgen receptors in bone oestrogen receptor-α. Nature 424, 389–389 (2003).
2, 298–302 (1992). health and disease. Nat. Rev. Endocrinol. 9, 699–712 63. Almeida, M. et al. Estrogens attenuate oxidative
13. Cooper, C. et al. Secular trends in the incidence of hip (2013). stress and the differentiation and apoptosis of
and other osteoporotic fractures. Osteoporos Int. 22, 38. Clowes, J. A., Riggs, B. L. & Khosla, S. The role osteoblasts by DNA-binding-independent actions
1277–1288 (2011). of the immune system in the pathophysiology of of the ERα. J. Bone Miner. Res. 25, 769–781 (2010).
14. Morin, S. N., Lix, L. M., Majumdar, S. R. osteoporosis. Immunol. Rev. 208, 207–227 (2005). 64. Goettsch, C. et al. NADPH oxidase 4 limits bone mass
& Leslie, W. D. Temporal trends in the incidence 39. Bonjour, J.‑P. & Chevalley, T. Pubertal timing, bone by promoting osteoclastogenesis. J. Clin. Invest. 123,
of osteoporotic fractures. Curr. Osteoporos Rep. 11, acquisition, and risk of fracture throughout life. 4731–4738 (2013).
263–269 (2013). Endocr. Rev. 35, 820–847 (2014). 65. Kanis, J. A., Johnell, O., Oden, A., Johansson, H.
15. Leslie, W. D. et al. Trends in hip fracture rates in 40. Szulc, P., Seeman, E., Duboeuf, F., Sornay‑Rendu, E. & McCloskey, E. FRAX™ and the assessment of fracture
Canada. JAMA 302, 883–889 (2009). & Delmas, P. D. Bone fragility: failure of periosteal probability in men and women from the UK.
16. Xia, W. B. et al. Rapidly increasing rates of hip fracture apposition to compensate for increased endocortical Osteoporos Int. 19, 385–397 (2008).
in Beijing, China. J. Bone Miner. Res. 27, 125–129 resorption in postmenopausal women. J. Bone Miner. 66. Nguyen, N. D., Frost, S. A., Center, J. R., Eisman, J. A.
(2012). Res. 21, 1856–1863 (2006). & Nguyen, T. V. Development of prognostic
17. Kinsella, K. & Wan, H. U.S. Census Bureau, 41. Nishiyama, K. K., Macdonald, H. M., Buie, H. R., nomograms for individualizing 5‑year and 10‑year
International Population Reports, P95/09‑1, An Aging Hanley, D. A. & Boyd, S. K. Postmenopausal women fracture risks. Osteoporos Int. 19, 1431–1444
World: 2008 (U.S. Government Printing Office, 2009). with osteopenia have higher cortical porosity and (2008).
18. Cooper, C., Campion, G. & Melton, L. J. Hip fractures thinner cortices at the distal radius and tibia than 67. Hippisley‑Cox, J. & Coupland, C. Predicting risk of
in the elderly: a world-wide projection. Osteoporos Int. women with normal aBMD: an in vivo HR‑pQCT study. osteoporotic fracture in men and women in England
2, 285–289 (1992). J. Bone Miner. Res. 25, 882–890 (2010). and Wales: prospective derivation and validation of
This paper highlights for the first time the epidemic 42. Patsch, J. M. et al. Increased cortical porosity in QFractureScores. BMJ 339, b4229 (2009).
in hip fractures. type 2 diabetic postmenopausal women with fragility 68. [No authors listed.] Assessment of fracture risk and
19. Pocock, N. A. et al. Genetic determinants of bone fractures. J. Bone Miner. Res. 28, 313–324 (2013). its application to screening for postmenopausal
mass in adults. A twin study. J. Clin. Invest. 80, 43. Gennari, L., Khosla, S. & Bilezikian, J. P. Estrogen osteoporosis. Report of a WHO Study Group. World
706–710 (1987). and fracture risk in men. J. Bone Miner. Res. 23, Health Organ. Techn. Rep. Ser. 843, 1–129 (1994).
20. Hernandez‑de Sosa, N. et al. Heritability of bone 1548–1551 (2008). 69. Marshall, D., Johnell, O. & Wedel, H. Meta-analysis
mineral density in a multivariate family-based study. 44. Santen, R. J., Brodie, H., Simpson, E. R., Siiteri, P. K. of how well measures of bone mineral density predict
Calcif. Tissue Int. 94, 590–596 (2014). & Brodie, A. History of aromatase: saga of an occurrence of osteoporotic fractures. BMJ 312,
21. Michaëlsson, K., Melhus, H., Ferm, H., Ahlbom, A. important biological mediator and therapeutic target. 1254–1259 (1996).
& Pedersen, N. L. Genetic liability to fractures in the Endocr. Rev. 30, 343–375 (2009). 70. Kanis, J. A. Diagnosis of osteoporosis and assessment
elderly. Arch. Intern. Med. 165, 1825–1830 (2005). 45. Khosla, S. Update on estrogens and the skeleton. of fracture risk. Lancet 359, 1929–1936 (2002).
22. Ioannidis, J. P. A. et al. Differential genetic effects of J. Clin. Endocrinol. Metab. 95, 3569–3577 (2010). 71. Vasikaran, S. et al. Markers of bone turnover for
ESR1 gene polymorphisms on osteoporosis outcomes. 46. Kovacs, C. S. Osteoporosis presenting in pregnancy, the prediction of fracture risk and monitoring of
JAMA 292, 2105–2114 (2004). puerperium, and lactation. Curr. Opin. Endocrinol. osteoporosis treatment: a need for international
23. Uitterlinden, A. G. et al. The association between Diabetes Obes. 21, 468–475 (2014). reference standards. Osteoporos Int. 22, 391–420
common vitamin D receptor gene variations and 47. Smith, E. P. et al. Estrogen resistance caused by a (2011).
osteoporosis: a participant-level meta-analysis. mutation in the estrogen-receptor gene in a man. 72. Diez‑Perez, A. et al. Treatment failure in osteoporosis.
Ann. Intern. Med. 145, 255–264 (2006). N. Engl. J. Med. 331, 1056–1061 (1994). Osteoporos Int. 23, 2769–2774 (2012).
24. Langdahl, B. L. et al. Large-scale analysis of 48. Falahati‑Nini, A. et al. Relative contributions of 73. Hernandez, C. J., Beaupré, G. S. & Carter, D. R.
association between polymorphisms in the testosterone and estrogen in regulating bone A theoretical analysis of the relative influences of peak
transforming growth factor β1 gene (TGFB1) resorption and formation in normal elderly men. BMD, age-related bone loss and menopause on the
and osteoporosis: the GENOMOS study. Bone 42, J. Clin. Invest. 106, 1553–1560 (2000). development of osteoporosis. Osteoporos Int. 14,
969–981 (2008). 49. Weitzmann, M. N. & Pacifici, R. Estrogen deficiency 843–847 (2003).
25. van Meurs, J. B. J. et al. Large-scale analysis of and bone loss: an inflammatory tale. J. Clin. Invest. 74. Cadogan, J., Eastell, R., Jones, N. & Barker, M. E.
association between LRP5 and LRP6 variants and 116, 1186–1194 (2006). Milk intake and bone mineral acquisition in adolescent
osteoporosis. JAMA 299, 1277–1290 (2008). 50. Kousteni, S. et al. Nongenotropic, sex-nonspecific girls: randomised, controlled intervention trial. BMJ
26. Ralston, S. H. et al. Large-scale evidence for the effect signaling through the estrogen or androgen receptors: 315, 1255–1260 (1997).
of the COLIA1 Sp1 polymorphism on osteoporosis dissociation from transcriptional activity. Cell 104, 75. Chevalley, T., Rizzoli, R., Hans, D., Ferrari, S.
outcomes: the GENOMOS study. PLoS Med. 3, e90 719–730 (2001). & Bonjour, J.‑P. Interaction between calcium intake
(2006). 51. Syed, F. A. et al. Skeletal effects of estrogen are and menarcheal age on bone mass gain: an eight-year
27. Xie, W., Ji, L., Zhao, T. & Gao, P. Identification of mediated by opposing actions of classical and follow‑up study from prepuberty to postmenarche.
transcriptional factors and key genes in primary nonclassical estrogen receptor pathways. J. Bone J. Clin. Endocrinol. Metab. 90, 44–51 (2005).
osteoporosis by DNA microarray. Med. Sci. Monit. 21, Miner. Res. 20, 1992–2001 (2005). 76. Winzenberg, T., Shaw, K., Fryer, J. & Jones, G. Effects
1333–1344 (2015). 52. Dallas, S. L., Prideaux, M. & Bonewald, L. F. of calcium supplementation on bone density in healthy
28. Keupp, K. et al. Mutations in WNT1 cause different The osteocyte: an endocrine cell … and more. children: meta-analysis of randomised controlled
forms of bone fragility. Am. J. Hum. Genet. 92, Endocr. Rev. 34, 658–690 (2013). trials. BMJ 333, 775–775 (2006).
565–574 (2013). 53. Rachner, T. D., Khosla, S. & Hofbauer, L. C. 77. Bischoff‑Ferrari, H. A. et al. Dietary calcium and serum
29. Zheng, H.‑F. et al. WNT16 influences bone mineral Osteoporosis: now and the future. Lancet 377, 25‑hydroxyvitamin D status in relation to BMD among
density, cortical bone thickness, bone strength, and 1276–1287 (2011). U. S. adults. J. Bone Miner. Res. 24, 935–942
osteoporotic fracture risk. PLoS Genet. 8, e1002745 54. Chow, J., Tobias, J. H., Colston, K. W. & Chambers, T. J. (2009).
(2012). Estrogen maintains trabecular bone volume in rats 78. Shea, B. et al. Meta-analyses of therapies for
30. Richards, J. B. et al. Bone mineral density, not only by suppression of bone resorption but also postmenopausal osteoporosis. VII. Meta-analysis
osteoporosis, and osteoporotic fractures: a genome- by stimulation of bone formation. J. Clin. Invest. 89, of calcium supplementation for the prevention of
wide association study. Lancet 371, 1505–1512 74–78 (1992). postmenopausal osteoporosis. Endocr. Rev. 23,
(2008). 55. Oursler, M. J. Estrogen regulation of gene expression 552–559 (2002).
31. Estrada, K. et al. Genome-wide meta-analysis in osteoblasts and osteoclasts. Crit. Rev. Eukaryot. 79. Elders, P. J. et al. Long-term effect of calcium
identifies 56 bone mineral density loci and reveals Gene Expr. 8, 125–140 (1998). supplementation on bone loss in perimenopausal
14 loci associated with risk of fracture. Nat. Genet. 56. Khastgir, G. et al. Anabolic effect of estrogen women. J. Bone Miner. Res. 9, 963–970 (1994).
44, 491–501 (2012). replacement on bone in postmenopausal women 80. Bischoff‑Ferrari, H. A. et al. Calcium intake and hip
32. Garnero, P., Sornay‑Rendu, E., Chapuy, M. C. with osteoporosis: histomorphometric evidence in fracture risk in men and women: a meta-analysis of
& Delmas, P. D. Increased bone turnover in late a longitudinal study. J. Clin. Endocrinol. Metab. 86, prospective cohort studies and randomized controlled
postmenopausal women is a major determinant 289–295 (2001). trials. Am. J. Clin. Nutr. 86, 1780–1790 (2007).
of osteoporosis. J. Bone Miner. Res. 11, 337–349 57. Hofbauer, L. C. & Schoppet, M. Clinical implications 81. Bolland, M. J. et al. Effect of calcium supplements
(1996). of the osteoprotegerin/RANKL/RANK system for bone on risk of myocardial infarction and cardiovascular
33. Sims, N. A. & Civitelli, R. Cell–cell signaling: and vascular diseases. JAMA 292, 490–495 (2004). events: meta-analysis. BMJ 341, c3691 (2010).
broadening our view of the basic multicellular unit. 58. Eghbali‑Fatourechi, G. et al. Role of RANK ligand 82. Wang, L., Manson, J. E., Song, Y. & Sesso, H. D.
Calcif. Tissue Int. 94, 2–3 (2014). in mediating increased bone resorption in early Systematic review: vitamin D and calcium
34. Khosla, S. New insights into androgen and estrogen postmenopausal women. J. Clin. Invest. 111, supplementation in prevention of cardiovascular
receptor regulation of the male skeleton. J. Bone 1221–1230 (2003). events. Ann. Intern. Med. 152, 315–323 (2010).
Miner. Res. 30, 1134–1137 (2015). 59. Nakashima, T. et al. Evidence for osteocyte regulation 83. Lewis, J. R. et al. The effects of calcium
This review describes the importance of oestrogen of bone homeostasis through RANKL expression. supplementation on verified coronary heart disease
action on the male skeleton and its role on bone Nat. Med. 17, 1231–1234 (2011). hospitalization and death in postmenopausal women:
homeostasis. 60. Mödder, U. I. et al. Regulation of circulating sclerostin a collaborative meta-analysis of randomized
35. Manolagas, S. C. From estrogen-centric to aging levels by sex steroids in women and in men. J. Bone controlled trials. J. Bone Miner. Res. 30, 165–175
and oxidative stress: a revised perspective of the Miner. Res. 26, 27–34 (2011). (2015).

14 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

84. Wallace, R. B. et al. Urinary tract stone occurrence 107. von Stengel, S., Kemmler, W., Engelke, K. & 132. Schilcher, J., Michaelsson, K. & Aspenberg, P.
in the Women’s Health Initiative (WHI) randomized Kalender, W. A. Effects of whole body vibration on bone Bisphosphonate use and atypical fractures of the
clinical trial of calcium and vitamin D supplements. mineral density and falls: results of the randomized femoral shaft. N. Engl. J. Med. 364, 1728–1737
Am. J. Clin. Nutr. 94, 270–277 (2011). controlled ELVIS study with postmenopausal women. (2011).
85. Serio, A. & Fraioli, A. Epidemiology of nephrolithiasis. Osteoporos Int. 22, 317–325 (2011). 133. Shane, E. et al. Atypical subtrochanteric and
Nephron 81 (Suppl. 1), 26–30 (1999). 108. Leung, K. S. et al. Effects of 18‑month low-magnitude diaphyseal femoral fractures: second report of a task
86. Bischoff‑Ferrari, H. A. et al. Prevention of nonvertebral high-frequency vibration on fall rate and fracture risks force of the American Society for Bone and Mineral
fractures with oral vitamin D and dose dependency: in 710 community elderly — a cluster-randomized Research. J. Bone Miner. Res. 29, 1–23 (2014).
a meta-analysis of randomized controlled trials. controlled trial. Osteoporos Int. 25, 1785–1795 134. Schilcher, J., Koeppen, V., Aspenberg, P.
Arch. Intern. Med. 169, 551–561 (2009). (2014). & Michaëlsson, K. Risk of atypical femoral fracture
87. Avenell, A., Mak, J. C. S. & O’Connell, D. Vitamin D 109. Kanis, J. A. et al. Smoking and fracture risk: during and after bisphosphonate use. N. Engl. J. Med.
and vitamin D analogues for preventing fractures in a meta-analysis. Osteoporos Int. 16, 155–162 (2005). 371, 974–976 (2014).
post-menopausal women and older men. 110. Krall, E. A. & Dawson-Hughes, B. Smoking increases 135. Dell, R. M. et al. Incidence of atypical nontraumatic
Cochrane Database Syst. Rev. 4, CD000227 (2014). bone loss and decreases intestinal calcium absorption. diaphyseal fractures of the femur. J. Bone Miner. Res.
This paper stresses that, although vitamin D alone J. Bone Miner. Res. 14, 215–220 (1999). 27, 2544–2550 (2012).
may not prevent fracture, when given along with 111. Jensen, J., Christiansen, C. & Rødbro, P. Cigarette 136. Cummings, S. R. et al. Denosumab for prevention
calcium, it may prevent fracture. smoking, serum estrogens, and bone loss during of fractures in postmenopausal women with
88. Ceglia, L. Vitamin D and its role in skeletal muscle. hormone-replacement therapy early after menopause. osteoporosis. N. Engl. J. Med. 361, 756–765 (2009).
Curr. Opin. Clin. Nutr. Metab. Care 12, 628–633 N. Engl. J. Med. 313, 973–975 (1985). 137. Papapoulos, S. et al. The effect of 8 or 5 years of
(2009). 112. Oncken, C. et al. Impact of smoking cessation on bone denosumab treatment in postmenopausal women with
89. Cao, J. J. & Nielsen, F. H. Acid diet (high-meat protein) mineral density in postmenopausal women. osteoporosis: results from the FREEDOM Extension
effects on calcium metabolism and bone health. Curr. J. Womens Health (Larchmt) 15, 1141–1150 (2006). study. Osteoporos Int. 26, 2773–2783 (2015).
Opin. Clin. Nutr. Metab. Care 13, 698–702 (2010). 113. Schwartz, A. V., Nevitt, M. C., Brown, B. W. 138. Miller, P. D. et al. Effect of denosumab on bone density
90. Byberg, L., Bellavia, A., Orsini, N., Wolk, A. & Kelsey, J. L. Increased falling as a risk factor for and turnover in postmenopausal women with low bone
& Michaëlsson, K. Fruit and vegetable intake and risk fracture among older women: the study of osteoporotic mass after long-term continued, discontinued, and
of hip fracture: a cohort study of Swedish men and fractures. Am. J. Epidemiol. 161, 180–185 (2005). restarting of therapy: a randomized blinded phase 2
women. J. Bone Miner. Res. 30, 976–984 (2015). 114. Oliver, D. et al. Strategies to prevent falls and clinical trial. Bone 43, 222–229 (2008).
91. Fenton, T. R., Eliasziw, M., Lyon, A. W., Tough, S. C. fractures in hospitals and care homes and effect 139. Aubry‑Rozier, B., Gonzalez‑Rodriguez, E., Stoll, D.
& Hanley, D. A. Meta-analysis of the quantity of of cognitive impairment: systematic review and & Lamy, O. Severe spontaneous vertebral fractures
calcium excretion associated with the net acid meta‑analyses. BMJ 334, 82–82 (2007). after denosumab discontinuation: three case reports.
excretion of the modern diet under the acid-ash diet 115. Harwood, R. H. et al. Falls and health status in elderly Osteoporos Int. 27, 1923–1925 (2016).
hypothesis. Am. J. Clin. Nutr. 88, 1159–1166 (2008). women following first eye cataract surgery: 140. Leder, B. Z. et al. Denosumab and teriparatide
92. Dawson‑Hughes, B. & Harris, S. S. Calcium intake a randomised controlled trial. Br. J. Ophthalmol. 89, transitions in postmenopausal osteoporosis
influences the association of protein intake with rates 53–59 (2005). (the DATA Switch study): extension of a randomised
of bone loss in elderly men and women. Am. J. Clin. 116. Bischoff‑Ferrari, H. A. et al. Fall prevention with controlled trial. Lancet 386, 1147–1155 (2015).
Nutr. 75, 773–779 (2002). supplemental and active forms of vitamin D: 141. McClung, M. R. et al. Opposite bone remodeling effects
93. Sahni, S. et al. Protective effect of high protein a meta‑analysis of randomised controlled trials. of teriparatide and alendronate in increasing bone
and calcium intake on the risk of hip fracture in the BMJ 339, b3692 (2009). mass. Arch. Intern. Med. 165, 1762–1768 (2005).
Framingham offspring cohort. J. Bone Miner. Res. 25, 117. Panel on Prevention of Falls in Older Persons, 142. Neer, R. M. et al. Effect of parathyroid hormone
2770–2776 (2010). American Geriatrics Society and British Geriatrics (1–34) on fractures and bone mineral density in
94. Ensrud, K. E., Cauley, J., Lipschutz, R. & Society. Summary of the Updated American Geriatrics postmenopausal women with osteoporosis. N. Engl.
Cummings, S. R. Weight change and fractures in older Society/British Geriatrics Society clinical practice J. Med. 344, 1434–1441 (2001).
women. Study of Osteoporotic Fractures Research guideline for prevention of falls in older persons. 143. Nakamura, T. et al. Randomized teriparatide human
Group. Arch. Intern. Med. 157, 857–863 (1997). J. Am. Geriatr. Soc. 59, 148–157 (2011). parathyroid hormone (PTH) 1–34 once-weekly efficacy
95. Cummings, S. R. et al. Risk factors for hip fracture 118. Institute of Medicine. Dietary Reference Ranges for research (TOWER) trial for examining the reduction
in white women. Study of Osteoporotic Fractures Calcium and Vitamin D (National Academies Press, in new vertebral fractures in subjects with primary
Research Group. N. Engl. J. Med. 332, 767–773 2011). osteoporosis and high fracture risk. J. Clin. Endocrinol.
(1995). 119. Michael, Y. L. et al. Primary care-relevant interventions Metab. 97, 3097–3106 (2012).
96. Crandall, C. J. et al. Postmenopausal weight change to prevent falling in older adults: a systematic evidence 144. Bang, U. C., Hyldstrup, L. & Jensen, J. E. B.
and incidence of fracture: post hoc findings from review for the U. S. Preventive Services Task Force. The impact of recombinant parathyroid hormone
Women’s Health Initiative Observational Study Ann. Intern. Med. 153, 815–825 (2010). on malignancies and mortality: 7 years of experience
and Clinical Trials. BMJ 350, h25 (2015). 120. Dawson-Hughes, B. et al. IOF position statement: based on nationwide Danish registers. Osteoporos Int.
97. Clynes, M. A. et al. Definitions of sarcopenia: vitamin D recommendations for older adults. 25, 639–644 (2014).
associations with previous falls and fracture in a Osteoporos Int. 21, 1151–1154 (2010). 145. Cosman, F. et al. Effects of intravenous zoledronic acid
population sample. Calcif. Tissue Int. 97, 445–452 121. Garvan Institute. Fracture Risk Calculator. plus subcutaneous teriparatide [rhPTH(1–34)] in
(2015). Garvan Institute http://www.garvan.org.au/ postmenopausal osteoporosis. J. Bone Miner. Res. 26,
98. Lang, T. et al. Cortical and trabecular bone mineral promotions/bone-fracture-risk/calculator/ (2016). 503–511 (2011).
loss from the spine and hip in long-duration 122. Billington, E. O., Gamble, G. D. & Reid, I. R. Reasons for 146. Cosman, F. et al. Effects of teriparatide in
spaceflight. J. Bone Miner. Res. 19, 1006–1012 discrepancies in hip fracture risk estimates using FRAX postmenopausal women with osteoporosis on prior
(2004). and Garvan calculators. Maturitas 85, 11–18 (2016). alendronate or raloxifene: differences between
99. Heinonen, A. et al. Bone mineral density in female 123. Dawson-Hughes, B. et al. Implications of absolute stopping and continuing the antiresorptive agent.
athletes representing sports with different loading fracture risk assessment for osteoporosis practice J. Clin. Endocrinol. Metab. 94, 3772–3780 (2009).
characteristics of the skeleton. Bone 17, 197–203 guidelines in the USA. Osteoporos Int. 19, 449–458 147. Recker, R. R. et al. Comparative effects of teriparatide
(1995). (2008). and strontium ranelate on bone biopsies and
100. Leichter, I. et al. Gain in mass density of bone following 124. Reid, I. R. Efficacy, effectiveness and side effects of biochemical markers of bone turnover in
strenuous physical activity. J. Orthop. Res. 7, 86–90 medications used to prevent fractures. J. Intern. Med. postmenopausal women with osteoporosis. J. Bone
(1989). 277, 690–706 (2015). Miner. Res. 24, 1358–1368 (2009).
101. McKay, H. A. et al. “Bounce at the Bell”: a novel 125. Ebetino, F. H. et al. The relationship between 148. Chavassieux, P. et al. Bone histomorphometry of
program of short bouts of exercise improves proximal the chemistry and biological activity of the transiliac paired bone biopsies after 6 or 12 months
femur bone mass in early pubertal children. bisphosphonates. Bone 49, 20–33 (2011). of treatment with oral strontium ranelate in 387
Br. J. Sports Med. 39, 521–526 (2005). 126. Murad, M. H. et al. Comparative effectiveness of drug osteoporotic women: randomized comparison to
102. Colberg, S. R. et al. Exercise and type 2 diabetes: treatments to prevent fragility fractures: a systematic alendronate. J. Bone Miner. Res. 29, 618–628 (2014).
the American College of Sports Medicine and the review and network meta-analysis. J. Clin. Endocrinol. 149. Meunier, P. J. et al. The effects of strontium ranelate
American Diabetes Association: joint position Metab. 97, 1871–1880 (2012). on the risk of vertebral fracture in women with
statement executive summary. Diabetes Care 33, 127. Black, D. M. et al. Once-yearly zoledronic acid for postmenopausal osteoporosis. N. Engl. J. Med. 350,
2692–2696 (2010). treatment of postmenopausal osteoporosis. N. Engl. 459–468 (2004).
103. Rubin, C. T., Bain, S. D. & McLeod, K. J. Suppression J. Med. 356, 1809–1822 (2007). 150. Reginster, J. Y. et al. Strontium ranelate reduces
of the osteogenic response in the aging skeleton. 128. Khosla, S. et al. Benefits and risks of bisphosphonate the risk of nonvertebral fractures in postmenopausal
Calcif. Tissue Int. 50, 306–313 (1992). therapy for osteoporosis. J. Clin. Endocrinol. Metab. women with osteoporosis: Treatment of Peripheral
104. Iwamoto, J., Takeda, T. & Ichimura, S. Effect of exercise 97, 2272–2282 (2012). Osteoporosis (TROPOS) study. J. Clin. Endocrinol.
training and detraining on bone mineral density in 129. Cartsos, V. M., Zhu, S. & Zavras, A. I. Bisphosphonate Metab. 90, 2816–2822 (2005).
postmenopausal women with osteoporosis. J. Orthop. use and the risk of adverse jaw outcomes. J. Am. 151. Tan, K. W., Wang, Y. S. & Tay, Y. K. Stevens–Johnson
Res. 6, 128–132 (2001). Dent. Assoc. 139, 23–30 (2008). syndrome due to strontium ranelate. Ann. Acad. Med.
105. Bolton, K. L. et al. Effects of exercise on bone density 130. Pazianas, M., Blumentals, W. A. & Miller, P. D. Singapore 40, 510–511 (2011).
and falls risk factors in post-menopausal women with Lack of association between oral bisphosphonates 152. Medicines and Healthcare Products Regulatory
osteopenia: a randomised controlled trial. J. Sci. Med. and osteonecrosis using jaw surgery as a surrogate Agency (MHRA). Strontium ranelate (Protelos): risk of
Sport 15, 102–109 (2012). marker. Osteoporos Int. 19, 773–779 (2008). serious cardiac disorders — restricted indications, new
106. Kemmler, W. et al. Exercise and fractures in 131. Lin, T. C., Yang, C. Y., Kao Yang, Y. H. & Lin, S. J. contraindications, and warnings. Drug Safety Update
postmenopausal women: 12‑year results of the Incidence and risk of osteonecrosis of the jaw among http://pad.res360.net/Content/Documents/
Erlangen Fitness and Osteoporosis Prevention Study the Taiwan osteoporosis population. Osteoporos Int. Strontium%20Safety%20Alert%20April%
(EFOPS). Osteoporos Int. 23, 1267–1276 (2012). 25, 1503–1511 (2014). 202013.pdf (2013).

NATURE REVIEWS | DISEASE PRIMERS VOLUME 2 | 2016 | 15


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.
PRIMER

153. European Medicines Agency. Protelos/Osseor to 172. Silverman, S. L. Quality-of‑life issues in osteoporosis. 190. Cosman, F. et al. Clinician’s guide to prevention
remain available but with further restrictions. EMA Curr. Rheumatol. Rep. 7, 39–45 (2005). and treatment of osteoporosis. Osteoporos Int. 25,
http://www.ema.europa.eu/docs/en_GB/document_ 173. Pike, C. et al. Direct and indirect costs of non-vertebral 2359–2381 (2014).
library/Referrals_document/Protelos_and_Osseor/ fracture patients with osteoporosis in the US. This is a recent guide for clinicians who manage
European_Commission_final_decision/ PharmacoEconomics 28, 395–409 (2010). osteoporosis.
WC500173034.pdf (2014). 174. Eekman, D. A. et al. Indirect costs account for half 191. Compston, J. et al. Diagnosis and management of
154. Lindsay, R., Hart, D. M., Forrest, C. & Baird, C. of the total costs of an osteoporotic fracture: osteoporosis in postmenopausal women and older
Prevention of spinal osteoporosis in oophorectomised a prospective evaluation. Osteoporos Int. 25, men in the UK: National Osteoporosis Guideline
women. Lancet 2, 1151–1153 (1980). 195–204 (2014). Group (NOGG) update 2013. Maturitas 75, 392–396
155. Rossouw, J. E. et al. Risks and benefits of estrogen 175. Gold, D. T. & Silverman, S. L. The Downward Spiral of (2013).
plus progestin in healthy postmenopausal women Vertebral Osteoporosis: Consequences (Cedars-Sinai 192. Kanis, J. A. et al. Goal-directed treatment of
— principal results from the Women’s Health Initiative Medical Center, 2003). osteoporosis in Europe. Osteoporos Int. 25,
randomized controlled trial. JAMA 288, 321–333 176. Ballane, G., Cauley, J. A., Luckey, M. M. 2533–2543 (2014).
(2002). & Fuleihan Gel, H. Secular trends in hip fractures 193. International Osteoporosis Foundation. Guideline
156. Anderson, G. L. et al. Effects of conjugated, equine worldwide: opposing trends East versus West. J. Bone references. IOF https://www.iofbonehealth.org/
estrogen in postmenopausal women with Miner. Res. 29, 1745–1755 (2014). guideline-references (accessed 12 Sep 2016).
hysterectomy — the Women’s Health Initiative 177. Brauer, C. A., Coca‑Perraillon, M., Cutler, D. M. 194. Cummings, S. R. et al. Goal-directed treatment of
randomized controlled trial. JAMA 291, 1701–1712 & Rosen, A. B. Incidence and mortality of hip fractures osteoporosis. J. Bone Miner. Res. 28, 433–438 (2013).
(2004). in the United States. JAMA 302, 1573–1579 195. Austin, M. et al. Relationship between bone mineral
157. Rossouw, J. E. et al. Postmenopausal hormone (2009). density changes with denosumab treatment and risk
therapy and risk of cardiovascular disease by age 178. Hiligsmann, M. et al. Trends in hip fracture incidence reduction for vertebral and nonvertebral fractures.
and years since menopause. JAMA 297, 1465–1477 and in the prescription of antiosteoporosis J. Bone Miner. Res. 27, 687–693 (2012).
(2007). medications during the same time period in Belgium 196. Jacques, R. M. et al. Relationship of changes in total
158. Reid, I. R. et al. A comparison of the effects of (2000–2007). Arthritis Care Res. (Hoboken) 64, hip bone mineral density to vertebral and nonvertebral
raloxifene and conjugated equine estrogen on bone 744–750 (2012). fracture risk in women with postmenopausal
and lipids in healthy postmenopausal women. 179. Wright, N. C. et al. The recent prevalence of osteoporosis treated with once-yearly zoledronic
Arch. Intern. Med. 164, 871–879 (2004). osteoporosis and low bone mass in the United States acid 5 mg: the HORIZON-Pivotal Fracture Trial (PFT).
159. Ettinger, B. et al. Reduction of vertebral fracture risk based on bone mineral density at the femoral neck or J. Bone Miner. Res. 27, 1627–1634 (2012).
in postmenopausal women with osteoporosis treated lumbar spine. J. Bone Miner. Res. 29, 2520–2526 197. Felsenberg, D. et al. Incidence of vertebral fracture
with raloxifene — results from a 3‑year randomized (2014). in Europe: results from the European Prospective
clinical trial. JAMA 282, 637–645 (1999). 180. Looker, A. C., Melton, L. J. 3rd, Borrud, L. G. Osteoporosis Study (EPOS). J. Bone Miner. Res. 17,
160. Barrett‑Connor, E. et al. Effects of raloxifene on & Shepherd, J. A. Changes in femur neck bone 716–724 (2002).
cardiovascular events and breast cancer in density in US adults between 1988–1994 198. Curtis, E. M. et al. Epidemiology of fractures in the
postmenopausal women. N. Engl. J. Med. 355, and 2005–2008: demographic patterns and United Kingdom 1988–2012: variation with age, sex,
125–137 (2006). possible determinants. Osteoporos Int. 23, 771–780 geography, ethnicity and socioeconomic status. Bone
161. Sharifi, M. & Lewiecki, E. M. Conjugated estrogens (2012). 87, 19–26 (2016).
combined with bazedoxifene: the first approved tissue 181. Jha, S., Wang, Z., Laucis, N. & Bhattacharyya, T. 199. McClung, M. et al. Odanacatib anti-fracture efficacy and
selective estrogen complex therapy. Expert Rev. Clin. Trends in media reports, oral bisphosphonate safety in postmenopausal women with osteoporosis:
Pharmacol. 7, 281–291 (2014). prescriptions, and hip fractures 1996–2012: results from the phase III long-term odanacatib fracture
162. Mirkin, S., Ryan, K. A., Chandran, A. B. & Komm, B. S. an ecological analysis. J. Bone Miner. Res. 30, trial. ACR http://acrabstracts.org/abstract/odanacatib-
Bazedoxifene/conjugated estrogens for managing the 2179–2187 (2015). anti-fracture-efficacy-and-safety-in-postmenopausal-
burden of estrogen deficiency symptoms. Maturitas This paper shows the recent trend for fewer women-with-osteoporosis-results-from-the-phase-iii-
77, 24–31 (2014). prescriptions of oral bisphosphonates and relates long-term-odanacatib-fracture-trial/ (2014).
163. Lindsay, R., Gallagher, J. C., Kagan, R., Pickar, J. H. this to media announcements. 200. Greenspan, S. L. et al. Effect of recombinant human
& Constantine, G. Efficacy of tissue-selective estrogen 182. Eisman, J. A. et al. Making the first fracture the last parathyroid hormone (1–84) on vertebral fracture and
complex of bazedoxifene/conjugated estrogens for fracture: ASBMR Task Force report on secondary bone mineral density in postmenopausal women with
osteoporosis prevention in at‑risk postmenopausal fracture prevention. J. Bone Miner. Res. 27, osteoporosis — a randomized trial. Ann. Intern. Med.
women. Fertil. Steril. 92, 1045–1052 (2009). 2039–2046 (2012). 146, 326–339 (2007).
164. Silverman, S. L. et al. Efficacy of bazedoxifene 183. McClung, M. R. et al. Romosozumab in 201. Reid, I. R. Short-term and long-term effects of
in reducing new vertebral fracture risk in postmenopausal women with low bone mineral osteoporosis therapies. Nat. Rev. Endocrinol. 11,
postmenopausal women with osteoporosis: results density. N. Engl. J. Med. 370, 412–420 (2014). 418–428 (2015).
from a 3‑year, randomized, placebo-, and active- 184. Fierce Biotech. Amgen and UCB announce positive
controlled clinical trial. J. Bone Miner. Res. 23, top-line results from the phase 3 study of Acknowledgements
1923–1934 (2008). Romosozumab in postmenopausal women R.E. was supported by a Senior Investigator Award from
165. Cummings, S. R. et al. Lasofoxifene in postmenopausal with osteoporosis. Fierce Biotech http:// the National Institute of Health Research. L.C.H. was
women with osteoporosis. N. Engl. J. Med. 362, www.fiercebiotech.com/press-releases/ funded by Deutsche Forschungsgemeinschaft, SFB‑655 and
686–696 (2010). amgen-and-ucb-announce-positive-top-line-results- Transregio‑67.
166. Lips, P. & van Schoor, N. M. Quality of life in patients phase-3-study-romosozumab (2016).
with osteoporosis. Osteoporos Int. 16, 447–455 185. Leder, B. Z. et al. Effects of abaloparatide, a human Author contributions
(2005). parathyroid hormone-related peptide analog, on Introduction (R.E.); Epidemiology (T.W.O.); Mechanisms/
167. Gold, D. T. The clinical impact of vertebral fractures: bone mineral density in postmenopausal women pathophysiology (L.C.H.); Diagnosis, screening and prevention
quality of life in women with osteoporosis. Bone 18, with osteoporosis. J. Clin. Endocrinol. Metab. 100, (B.L.); Management (I.R.R.); Quality of life (D.T.G.); Outlook
185S–189S (1996). 697–706 (2015). (S.R.C.); Overview of Primer (R.E.).
168. Cauley, J. A., Thompson, D. E., Ensrud, K. C., 186. Tucker, M. E. Novel agent abaloparatide reduces
Scott, J. C. & Black, D. Risk of mortality following fractures, including wrist. Medscape http:// Competing interests
clinical fractures. Osteoporos Int. 11, 556–561 www.medscape.com/viewarticle/841015 (2015). R.E. has received consulting fees from Amgen, AstraZeneca,
(2000). 187. Rizzoli, R. et al. Continuous treatment with GlaxoSmithKline (GSK), Immunodiagnostic Systems, Ono
This study, using data from the Fracture odanacatib for up to 8 years in postmenopausal Pharma, Lilly and Roche Diagnostics, and grant support from
Intervention Trial, was one of the first to examine women with low bone mineral density: a phase 2 Amgen, Immunodiagnostic Systems, Lilly and AstraZeneca.
mortality resulting from major osteoporotic study. Osteoporos Int. 27, 2099–2107 (2016). L.C.H. has received honoraria for serving on the advisory
fractures and the first to identify excess mortality 188. [No authors listed.] Merck announces data from board and for giving lectures from Amgen, Eli Lilly, Merck,
associated with clinical vertebral fractures. pivotal phase 3 fracture outcomes study for Novartis and UCB. B.L. has received consulting fees from
169. Randell, A. G. et al. Deterioration in quality of life odanacatib, an investigational oral, once-weekly Amgen, Merck, Eli Lilly and UCB, and grant support from
following hip fracture: a prospective study. treatment for osteoporosis. Business Wire Novo Nordisk, Eli Lilly and Orkla Health. I.R.R. has received
Osteoporos Int. 11, 460–466 (2000). http://www.businesswire.com/news/home/ research funding and honoraria from Merck, Amgen and
170. Papaioannou, A. et al. Determinants of health-related 20140915006286/en/Merck-Announces-Data- Novartis. S.R.C. serves as a consultant to Amgen, Radius,
quality of life in women with vertebral fractures. Pivotal-Phase-3-Fracture (2014). Merck and Eli Lilly about the design of studies and
Osteoporos Int. 17, 355–363 (2006). 189. [No authors listed.] Merck provides update on interpretation of results, and has received grant support from
171. Bhattacharya, R., Shen, C. & Sambamoorthi, U. odanacatib development program. Business Wire Amgen for systematic review of medical risk factors for hip
Excess risk of chronic physical conditions associated http://www.businesswire.com/news/ fracture. He has not received funds for lectures or other
with depression and anxiety. BMC Psychiatry 14, 10 home/20160902005107/en/Merck-Update- promotional activities. D.T.G. and T.W.O. have no conflicts of
(2014). Odanacatib-Development-Program (2016). interest or competing interests to report.

16 | 2016 | VOLUME 2 www.nature.com/nrdp


©
2
0
1
6
M
a
c
m
i
l
l
a
n
P
u
b
l
i
s
h
e
r
s
L
i
m
i
t
e
d
,
p
a
r
t
o
f
S
p
r
i
n
g
e
r
N
a
t
u
r
e
.
A
l
l
r
i
g
h
t
s
r
e
s
e
r
v
e
d
.

You might also like