ArticlePDF AvailableLiterature Review

Abstract

Alcohol is widely consumed across the world. It is consumed in both social and cultural settings. Until recently, two types of alcohol consumption were recognized: heavy chronic alcohol consumption or light consumption. Today, there is a new pattern of consumption among teenagers and young adults namely: binge drinking. Heavy alcohol consumption is detrimental to many organs and tissues, including bones, and is known to induce secondary osteoporosis. Some studies, however, have reported benefits from light alcohol consumption on bone parameters. To date, little is known regarding the effects of binge drinking on bone health. Here, we review the effects of three different means of alcohol consumption: light, heavy, and binge drinking. We also review the detailed literature on the different mechanisms by which alcohol intake may decrease bone mass and strength. The effects of alcohol on bone are thought to be both direct and indirect. The decrease in bone mass and strength following alcohol consumption is mainly due to a bone remodeling imbalance, with a predominant decrease in bone formation. Recent studies, however, have reported new mechanisms by which alcohol may act on bone remodeling, including osteocyte apoptosis, oxidative stress, and Wnt signalling pathway modulation. The roles of reduced total fat mass, increased lipid content in bone marrow, and a hypoleptinemia are also discussed.
CONSENSUS STATEMENT
Extraskeletal benefits and risks of calcium, vitamin D
and anti-osteoporosis medications
J.-J. Body &P. Bergmann &S. Boonen &J.-P. Devogelaer &
E. Gielen &S. Goemaere &J.-M. Kaufman &
S. Rozenberg &J.-Y. Reginster
Received: 9 November 2011 / Accepted: 19 December 2011
#The Author(s) 2012. This article is published with open access at Springerlink.com
Abstract
Summary Drugs used for the prevention and the treatment
of osteoporosis exert various favourable and unfavourable
extra-skeletal effects whose importance is increasingly
recognized notably for treatment selection.
Introduction The therapeutic armamentarium for the pre-
vention and the treatment of osteoporosis is increasingly
large, and possible extra-skeletal effects of available drugs
could influence the choice of a particular compound.
Methods The present document is the result of a national
consensus, based on a systematic and critical review of the
literature.
Results Observational research has suggested an inverse
relationship between calcium intake and cardiovascular dis-
eases, notably through an effect on blood pressure, but
recent data suggest a possible deleterious effect of calcium
supplements on cardiovascular risk. Many diverse studies
have implicated vitamin D in the pathogenesis of clinically
important non-skeletal functions or diseases, especially
muscle function, cardiovascular disease, autoimmune dis-
eases and common cancers. The possible effects of oral or
intravenous bisphosphonates are well-known. They have
been associated with an increased risk of oesophageal can-
cer or atrial fibrillation, but large-scale studies have not
J.-J. Body
Department of Medicine, CHU Brugmann,
Université Libre de Bruxelles,
Brussels, Belgium
P. Bergmann
Department of Radioisotopes, CHU Brugmann,
Université Libre de Bruxelles,
Brussels, Belgium
S. Boonen
Center for Metabolic Bone Diseases,
Katholieke University Leuven,
Leuven, Belgium
J.-P. Devogelaer
Department of Rheumatology, Saint Luc University Hospital,
Université Catholique de Louvain,
Brussels, Belgium
E. Gielen
Gerontology and Geriatrics Section,
Department of Experimental Medicine, K.U.Leuven,
Leuven, Belgium
S. Goemaere
Department of Rheumatology and Endocrinology,
State University of Gent,
Gent, Belgium
J.-M. Kaufman
Department of Endocrinology, State University of Gent,
Gent, Belgium
S. Rozenberg
Department of GynaecologyObstetrics,
Université Libre de Bruxelles,
Brussels, Belgium
J.-Y. Reginster
Department of Public Health, Epidemiology and Health
Economics, University of Liège,
Liège, Belgium
J.-Y. Reginster (*)
Bone and Cartilage Metabolism Research Unit,
CHU Centre-Ville, Policliniques L. BRULL,
Quai Godefroid Kurth 45 (9ème étage),
4020 Liege, Belgium
e-mail: jyreginster@ulg.ac.be
Osteoporos Int (2012) 23 (Suppl 1):S1S23
DOI 10.1007/s00198-011-1891-8
found any association with bisphosphonate use. Selective
oestrogen receptor modulators have demonstrated favour-
able or unfavourable extra-skeletal effects that vary between
compounds. Strontium ranelate has a limited number of
non-skeletal effects. A reported increase in the risk of ve-
nous thromboembolism is not found in observational stud-
ies, and very rare cases of cutaneous hypersensitivity
reactions have been reported. Denosumab has been intro-
duced recently, and its extra-skeletal effects still have to be
assessed.
Conclusion Several non-skeletal effects of bone drugs are
well demonstrated and influence treatment choices.
Keywords Bisphosphonate .Calcium .Denosumab .
Osteoporosis .SERM .Strontium ranelate .Vitamin D
Introduction
The pharmacological armamentarium for the management of
osteoporosis has considerably expanded. Indeed, ability to
substantially reduce fracture risk with a generally favourable
riskbenefit ratio is now documented in well-conducted large
clinical trials for a series of different molecules encompassing
different pharmacological classes and different modes of ac-
tion [1]. Osteoporosis is a highly prevalent problem in the
ageing population, and the absolute number of affected sub-
jects increases as a consequence of demographic evolutions.
Albeit at present only a fraction of these patients at risk are
treated, progress is being made and awareness increases of the
consequences of osteoporotic fractures in terms of personal
suffering and burden for the public health. Therefore, a large
and steadily increasing number of patients are likely to be
exposed for prolonged periods of treatment to osteoporosis
medication. Availability of several treatment alternatives con-
fronts the clinician with the difficulty to make the best choice
for the individual patient, whereas the large-scale and pro-
longed prescription of osteoporosis medication puts much
emphasis on safety issues.
To compare treatments, there is little evidence available
from direct comparative trials, and no direct comparisons
are available with fracture incidence as primary evaluation
criterion. To select the best choice treatmentfor their
individual patient, clinicians thus depend on indirect com-
parisons, with little possibility of reliable differentiation in
terms of efficacy, taking into account a variety of drug
characteristics in relation to the patients clinical profile
and preferences. In this context, consideration of the non-
skeletal actions of the osteoporosis medications will not
seldom intervene in the final choice, be it positively in terms
of perceived potential added valueor negatively because
of perceived potential risk for the patient. Aside from con-
troversies related to potential long-term osseous adverse
effects of osteoporosis treatments, a number of alleged
extra-skeletal safety issues have been raised in the recent
literature concerning as widely prescribed treatments as
calcium and bisphosphonates (BPs).
The present document is the result of a national consensus
based on a systematic review and a critical appraisal of the
literature. It aims at providing the clinicians with an overview
of what is the state of our knowledge on potentially deleterious
or beneficial non-skeletal actions of the main pharmacological
treatments of osteoporosis.
Methods
We included randomised controlled trials(RCTs), meta-
analyses as well as epidemiologic retrospective or prospective
studies and well documented case reports considering non-
skeletal actions of osteoporosis treatments. Relevant articles
related to treatment with calcium, vitamin D, bisphospho-
nates, selective oestrogen receptor modulators (SERMs),
strontium ranelate, teriparatide, parathyroid hormone (PTH)
and denosumab were identified through a systematic search,
from 1966 to 2011, in MEDLINE and databases such as
Cochrane Controlled Register. Following this extensive
search of the literature, a critical appraisal was obtained
through a consensus expert meeting.
Calcium
In the elderly, low calcium intake and vitamin D deficiency
result in a negative calcium balance. This stimulates the
secretion of PTH and induces age-associated secondary
hyperparathyroidism, which enhances bone turnover and
accelerates bone loss [2]. Adequate intake of calcium and
vitamin D, through diet and/or supplements, reverses this
secondary hyperparathyroidism and is recommended in the
prevention of osteoporotic fractures [1,3]. More specifically,
the National Institutes of Health (NIH) in the USA proposes a
recommended dietary allowance for calcium of 1,000 mg in
menaged50
70 years and 1,200 mg in men older than
70 years and women older than 50 years.
In combination with vitamin D substitution, calcium
supplements have proven anti-fracture efficacy when tar-
geted to persons at risk of calcium and/or vitamin D insuf-
ficiency, including elderly or institutionalized individuals,
osteoporosis patients on antiresorptive or anabolic medica-
tion and persons receiving glucocorticoids [48]. Benefits
are most apparent when a daily dose of 1,0001,200 mg
calcium is complemented with 800 IU vitamin D [6,8]. This
section reviews the evidence for the positive and negative
non-skeletal effects of calcium [9].
S2 Osteoporos Int (2012) 23 (Suppl 1):S1S23
Calcium as potentially protective against cardiovascular
events
Observational research has suggested an inverse relationship
between calcium intake and vascular diseases. In the Iowa
Womens Health Study in 34,486 postmenopausal women
aged 55 to 69 years, Bostick and colleagues found that the
highest quartile of total calcium intake (>1,425 mg/day),
when compared to the lowest quartile (<696 calcium/day),
was associated with a 33% reduction in ischaemic heart
disease mortality (risk ratio (RR) 0.67, 95% confidence
interval (CI) 0.47 to 0.94). According to the analysis, this
risk reduction was dependent of the high total intake of
calcium and could be attained by diet, supplements or both
[10]. Similarly, Knox found a strong negative correlation
between dietary calcium intake and mortality ratios for
ischemic heart disease [11]. In the NursesHealth Study
cohort of 85,764 women aged 39 to 59 years followed for
14 years, women in the highest quintile of total calcium
intake (median calcium 1,145 mg/day) had a lower risk of
stroke (RR 0.69, 95% CI 0.500.95) than those in the lowest
quintile (median calcium 395 mg/day) [12].
To explain this observed protection against vascular dis-
eases, potential beneficial effects of calcium on a number of
vascular risk factors have been postulated. In particular,
reductions in blood pressure, serum lipid concentration
and body weight might be involved, although the data, to
some extent, remain inconsistent [9].
An inverse relationship between calcium and blood pres-
sure has been observed in several studies. In a meta-analysis
of randomised controlled trials, both dietary calcium intake
and calcium supplements were associated with reduced blood
pressure, with a trend towards larger effects with dietary
intake. However, the effect size was relatively small, with a
mean reduction in systolic and diastolic blood pressure
of 1.44 mmHg (95% CI 2.20 to 0.68) and 0.84 mmHg
(95% CI 1.44 to 0.24), respectively [13]. In line with these
findings, a recent trial showed significantly lower rates of
hypertension amongst women aged over 45 years with a
dietary calcium intake of at least 679 mg/day. In women in
the highest quintile of dietary calcium intake (1,000 to
2,560 mg calcium/day), the relative risk reduction was 13%
(RR 0.87, 95% CI 0.81 to 0.93). However, in women taking
calcium supplements, even in the highest dosed quintile
(1,0002,100 mg), the risk of hypertension was unchanged
(RR 1.07, 95% CI 0.97 to 1.18) [14]. A recent Cochrane
review concluded that any association between calcium sup-
plements and reduction in blood pressure is uncertain and that
poor quality of individual trials and heterogeneity between
trials do not allow any firm conclusions [15]. Any antihyper-
tensive effect, if real, is at best small and transient [16].
Another potential cardioprotective mechanism might be a
reduction in serum lipid concentration, due to the binding of
calcium to fatty acids and bile acids in the gut, resulting in
malabsorption of fat, and a direct effect on adipocytes with
increased lipolysis [1719]. In a randomised controlled trial
in men, a diet fortified with calcium significantly reduced
total cholesterol, LDL cholesterol and apolipoprotein B
[18]. Similarly, in a randomised placebo-controlled trial in
postmenopausal women, a supplement of 1,000 mg calcium
during 12 months increased high-density lipoprotein (HDL)
cholesterol levels and HDL to low-density lipoprotein
(LDL) cholesterol ratio [20]. In another randomised study
in men and women, however, no significant effect of calcium
supplements (1,0002,000 mg) was seen on total cholesterol
or HDL cholesterol [21]. It is unclear, therefore, if and to what
extent calcium determines lipid profile.
Reduced body weight has been implicated as well. Several
large epidemiological studies have suggested that dietary cal-
cium intake and calcium supplements may be associated with
weight loss [22,23], an effect that might be mediated by the
same mechanisms affecting lipid profile [23]. However, sev-
eral systematic reviews of randomised controlled trials argued
against an inverse relationship between calcium (both dietary
intake and supplements) and body weight [2426], suggesting
that any conclusions are preliminary and that the implications
of calcium intake for body weight remain to be clarified.
Calcium supplements potentially associated with an increase
in cardiovascular risk
Whereas spontaneous calcium intake, up to 800 mg/day,
was not related to any cardiovascular deleterious effects,
the cardiovascular safety of calcium supplements has been
questioned. Rather than having a neutral or even beneficial
effect, increased exposure to calcium might actually increase
cardiovascular risk. In a meta-analysis published in 2010 by
Bolland and colleagues in the British Medical Journal, more
than 12,000 individuals from 15 double-blind placebo-
controlled randomised trials were enrolled, and an increase
in the incidence of myocardial infarction of about 30% was
seen in individuals on calcium supplements (500 mg daily)
compared to those on placebo [27]. More specifically, the
analysis of patient level data showed that the relative risk of
incident myocardial infarction in individuals allocated to cal-
cium increased by 31% (HR 1.31, 95% CI 1.02 to 1.67) and
trial level analysis showed a similar increase in risk by 27%
(HR 1.27, 95% CI 1.01 to 1.59). However, no significant
increase was observed in the incidence of a number of related
vascular endpoints, including the incidence of stroke (HR
1.20, 95% CI 0.96 to 1.50), death (HR 1.09, 95% CI 0.96 to
1.23) and the composite end point of myocardial infarction,
stroke and sudden death (HR 1.18, 95% CI 1.00 to 1.39).
The findings of this meta-analysis were partly driven by a
previous randomised placebo-controlled trial from the same
group that contributed 17% to the overall weight [28]. In
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S3
this trial, calcium supplements were associated with a sig-
nificant increase in HDL cholesterol levels but, neverthe-
less, also an increase in the risk of myocardial infarction [20,
28]. The authors postulated that calcium supplements may
acutely elevate serum calcium levels [29] and, as a result,
may enhance vascular calcification [28]. In fact, in a number
of observational studies, high serum calcium levels have
been associated with vascular calcification and an increased
risk of vascular events, including myocardial infarction,
stroke and death [30,31]. Further support for a potentially
deleterious effect of an acute increase in serum calcium
comes from the observation that, in the meta-analysis, die-
tary intake was not associated with myocardial infarction, in
line with observations that calcium from dairy products
hardly affects serum calcium levels [27].
Whilst the meta-analysis of Bolland and colleagues
should be interpreted as a strong signal that calcium supple-
ments (without vitamin D) may potentially increase the risk
of myocardial infarction, several limitations and even incon-
sistencies should be taken into account as well. First, the
statistical outcome was only borderline significant (HR
1.31, 95% CI 1.02 to 1.67; p00.035), with a broad 95%
confidence interval that approached 1 in the lower limit,
suggesting that the findings have to be interpreted with
caution. Also, the studies included in the analysis had been
designed to assess the effects of calcium on bone density
and fracture risk. None of the included trials had cardiovas-
cular outcomes as primary or even secondary endpoint. As a
result, cardiovascular events had not been adjudicated in a
standardized manner, which may have resulted in over- or
underreporting. Third, whilst the meta-analysis provided
evidence for an increased risk of myocardial infarction, no
increase was observed in the incidence of stroke, death or
the composite end point of myocardial infarction, stroke and
sudden death. In addition, trials that combined calcium and
vitamin D supplements, the recommend strategy to prevent
fractures in most elderly individuals, were excluded. In this
context, it should be noted that a number of large-scale
studies of calcium combined with vitamin D did not docu-
ment an increase in cardiovascular risk [32,33]. It is possi-
ble but not known if correction of vitamin D deficiency
might counteract any potential detrimental vascular effect
of calcium supplements [34,35]. Finally, with the exception
of the relatively small-sized trial from the same group [28],
individual trials with calcium supplements did not show a
significant increase in cardiovascular risk. In fact, a recent
randomised placebo-controlled trial by Lewis et al., not in-
cluded in the meta-analysis, did not find a higher risk of death
or first-time hospitalization from atherosclerotic vascular dis-
ease in patients on calcium supplements [36]. A subset anal-
ysis even suggested a cardioprotective effect of calcium
supplements in patients with pre-existing cardiovascular dis-
eases. Nevertheless, the meta-analysis by Bolland et al. should
be taken seriously, not as conclusive evidence but as a signif-
icant safety signal. Future studies with calcium should be
designed to include careful assessment of cardiovascular end-
points, preferably by independent and blinded adjudication.
Calcium and cancer risk
There is also much controversy about the effect of calcium
on the risk of cancer, with observational studies showing no
effect, a protective effect or even an increased cancer risk
[37]. Because the topic is diverse and the findings inconsis-
tent, this section will only briefly discuss the association
between calcium exposure and colorectal cancer, breast
cancer and prostate cancer, since these have received most
attention in recent years [9].
Whilst several observational studies concluded that cal-
cium intake does not affect the risk of colorectal cancer
[38], a number of cohort studies did find evidence for a
protective effect of high total calcium intake (dietary intake
plus supplements) [37,39,40]. In one of the main studies, a
NIH-funded 7-year prospective trial in 293,907 men and
198,903 women aged 50 to 71 years, the risk reduction for
colorectal cancer in the highest compared to the lowest
quintile of total calcium intake was 0.79 (95% CI 0.70 to
0.89) in men and 0.72 (95% CI 0.61 to 0.86) in women [37].
Moreover, in a meta-analysis of randomised controlled trials
in patients with previously removed colorectal adenomas
and randomly assigned to calcium (1,200, 1,600 or
2,000 mg) or placebo, calcium supplements were signifi-
cantly associated with a reduction in the risk of recurrent
adenomas, considered as the precursors of colorectal cancer
[41]. In line with these findings, the American College of
Gastroenterology recommends daily dietary supplementa-
tion with 3 g calcium carbonate (1,200 mg calcium) in the
prevention of recurrent colorectal adenomas [42].
Despite these data from observational studies and adenoma
prevention trials, it is still uncertain if calcium supplements
prevent colorectal cancer because large-scale long-term rand-
omised controlled trials are not available. The only major
randomised placebo-controlled study, the Womens Health
Initiative (WHI) trial in 36,282 postmenopausal women,
found no effect of daily supplementation with 1,000 mg cal-
cium and 400 IU of vitamin D for 7 years on colorectal cancer
risk [43]. A Cochrane review concluded that there is not
sufficient evidence to currently recommend the general use
of calcium supplements in the prevention of colorectal cancer
andthatmoreresearchisneeded[44].
The relationship between calcium exposure and breast
cancer is not clear either. Some observational studies in
premenopausal women found an inverse relationship be-
tween calcium intake and breast cancer [4547], but some
did not [37,48]. Similarly, in trials in postmenopausal
women, a protective effect has been reported [47], but most
S4 Osteoporos Int (2012) 23 (Suppl 1):S1S23
studies were negative [37,45,46,48]. If and to what extent
the source of calcium intake (dietary intake versus supple-
ments) plays any role is not known [48]. Overall, an inde-
pendent effect of calcium on the incidence of breast cancer
remains uncertain.
In men, epidemiological studies have suggested that a
higher total intake of calcium might be associated with an
increased risk of developing prostate cancer. In these stud-
ies, total intake of calcium varied from more than 1,500 mg
to more than 2,000 mg/day [4951]. Calcium could poten-
tially suppress the active form of vitamin D (1,25-OH
2
-D
3
),
known to have an antiproliferative effect on prostate cancer
cells [50,52]. However, other studies could not confirm this
association and found no or only a weak relationship be-
tween calcium intake and prostate risk [37,5355], even at
very high intakes of calcium [37,54]. As with colon cancer
and breast cancer, conclusive evidence is lacking and more
studies are required.
Calcium and the risk of kidney stones
Since most kidney stones are composed of calcium oxalate,
an association with calcium intake is a theoretical concern.
In the prospective NursesHealth Study, women who took
supplemental calcium (1 to 500 mg/day) had a small but
significant increase in the risk of incident symptomatic
kidney stones (RR 1.20, 95% CI 1.021.41) compared to
those who did not take supplements [56]. Women in the
highest quintile of dietary calcium intake (median calcium
1,303 mg/day had, however, a lower risk (RR 0.65, 95% CI
0.500.83) compared to those in the lowest quintile (median
calcium 391 mg/day). Other trials also showed a slightly
increased risk of kidney stones in individuals on supplemental
calcium (1,000 mg/day) [32] and a lower risk in individuals on
a diet rich in calcium [57,58].
The lower incidence of kidney stones in individuals on
high dietary calcium intake is likely due to binding of
dietary calcium with dietary oxalate in the gut, with reduced
intestinal absorption and urinary excretion of oxalate. Cal-
cium supplements, on the other hand, do not bind dietary
oxalate when taken without meals. A combination of main-
tained oxalate excretion and increased calcium absorption
and excretion from supplements increases the risk of stone
formation [59].
In addition to beneficial musculoskeletal effects, espe-
cially when combined with vitamin D, calcium supple-
ments have been suggested to protect against colorectal
and breast cancer and to reduce some vascular risk
factors. At the same time, safety questions have been
raised about the role of calcium supplements in poten-
tially increasing cardiovascular events, prostate cancer
and kidney stones. Whilst these safety concerns have to
be taken seriously, currently available evidence is not
conclusive. In future research, priority should be given
to well-designed long-term studies to assess cardiovascular
and other safety endpoints.
Vitamin D
Rickets and osteomalacia are the diseases traditionally as-
sociated with severe vitamin D deficiency, defined as 25
(OH) vitamin D levels below 10 ng/ml (25 nmol/l). A
growing body of evidence has emerged indicating that less
severe degrees of vitamin D deficiency between 10 and
20 ng/ml (25 and 50 nmol/l) and even vitamin D insuffi-
ciency, defined as 25(OH) vitamin D levels between 20 and
30 ng/ml (50 and 75 nmol/l), impair gastrointestinal absorp-
tion of calcium and bone mineralization, contributing to the
pathogenesis of osteoporosis in older people [60]. Vitamin
D has an impact on bone density and bone quality. In
addition, by increasing muscle strength, adequate vitamin
D status reduces the risk of falling in older individuals (see
below). Therefore, vitamin D has a dual benefit for preven-
tion of fractures in the elderly, a benefit on bone density and
on muscle strength [61]. The importance of vitamin D for
the prevention and treatment of osteoporosis has notably
been reviewed in a previous Consensus of the Belgian Bone
Club [1].
Furthermore, many studies have implicated vitamin D
and its metabolites in the pathogenesis of a wide variety of
clinically important non-skeletal functions or diseases, es-
pecially muscle function, cardiovascular disease, autoim-
mune diseases and several common cancers. The principal
non-classical targets will be reviewed in this section. Whilst
the evidence on bone and muscle health is based on rando-
mised clinical trials, the evidence on other disease areas is
nevertheless of a lower level. Most trials are small to mod-
erate sized, and the outcomes of interest are only secondary
outcomes. Interestingly, a meta-analysis of 18 randomised
clinical trials including 57,311 individuals nevertheless con-
cluded that vitamin D supplementation was associated with
a decrease in total mortality (RR 0.93; 95% CI 0.770.96
compared to the control group) that could be due to effects
of vitamin D on the musculoskeletal system or, as summa-
rized below, on various non-skeletal diseases [35].
Vitamin D and muscular function
Vitamin D receptors have been shown to be present in
muscle tissue [62], and a direct effect of vitamin D on
muscle physiology is probable [63]. In muscle, vitamin D
activates protein kinase C, which promotes calcium release,
increasing the calcium pool that is essential for muscle
contraction [64]. The potential cell signalling pathways
affected by vitamin D in muscle have been recently
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S5
reviewed [65]. Vitamin D deficiency has long been clinical-
ly associated with impaired muscle strength [66] and is also
associated with loss of muscle mass [67]. With ageing, the
number of vitamin D receptors in muscle decreases and the
number of type II fibres, the first to be recruited to avoid
falls, also decreases [68]. Treatment of elderly stroke survi-
vors with 1,000 IU of vitamin D
2
daily increases mean type
II muscle fibre diameter by 2.5-fold over a 2-year period
[69]. Because muscle weakness is a major risk factor for
falls, it is not surprising that low vitamin D status is associ-
ated with an increased falls risk, as notably shown in a
longitudinal study [70]. A meta-analysis including seven
randomised, double-blind trials evaluating a daily dose of
7001,000 IU/day of vitamin D demonstrated that falling was
significantly reduced by 19% (RR 0.81; 95% CI 0.710.92) in
vitamin D supplemented individuals compared with those
receiving calcium or placebo [71]. This benefit may not de-
pend on additional calcium supplementation, was significant
within 25 months of treatment and extended beyond
12 months of treatment.
Vitamin D insufficiency and deficiency are associated
with an increase in muscle fat as demonstrated by a signif-
icant negative relationship between circulating 25(OH) vi-
tamin D levels and computed tomography measures of
percent muscle fat (p<0.001) [72]. Most studies have not
found a significant relationship between baseline 25(OH)
vitamin D levels and muscle strength [73]. However, cor-
rection of vitamin D deficiency has most often been associ-
ated with an improvement in muscle strength. Vitamin D
supplementation in vitamin D-deficient Asian Indians dur-
ing 6 months has thus shown an enhancement in skeletal
muscle strength and physical performance [74]. A recent
randomised, placebo-controlled, double-blind trial of
1,000 IU/day of vitamin D for 1 year showed a significant
increase in muscle strength and mobility in subjects in the
lowest tertile of baseline 25(OH) vitamin D values [75]. A
longer duration trial showed that vitamin D and calcium
supplementation during 20 months were superior to calcium
alone in reducing fall frequency and improving muscle
function in community-dwelling elderly subjects with 25
(OH) vitamin D levels below 31 ng/ml [76]. These studies
are in agreement with a recent systematic review and meta-
analysis where the authors confirmed a beneficial effect of
vitamin D supplementation on proximal muscle strength in
adults with vitamin D deficiency but no significant effect on
muscle strength in vitamin D replete adults [77].
Vitamin D and cardiovascular risk
A low level of 25(OH) vitamin D could be an independent
risk factor for cardiovascular events, although a causal rela-
tionship has yet to be supported by large interventional
trials. The evidence supporting a link between vitamin D
deficiency and myocardial diseases has recently been
reviewed [78]. In addition to possible direct effects due to
the presence of the vitamin D receptor and of the 1-alpha
hydroxylase enzyme in cardiac myocytes and other cells of
the cardiovascular system [79], vitamin D has significant
effects on several cardiovascular risk factors. Studies, rang-
ing from animal studies to clinical trials, have shown that
pharmacological doses of vitamin D notably reduce inflam-
mation [80], improve endothelial function [81], control the
secretion of insulin and improve insulin sensitivity [82].
Furthermore, as recently reviewed, vitamin D status has
been linked to arterial hypertension [83].
Several observational studies suggest that 25(OH) vita-
min D levels less than 15 ng/ml are associated with an
excess risk of cardiovascular events when compared to
levels >3040 ng/ml. A nested casecontrol study in
18,225 men in the Health Professionals Follow-up Study
(men aged 4075 years, free of cardiovascular disease at
baseline) showed that men with a 25(OH) vitamin D
level 15 ng/ml had an increased risk for myocardial
infarction relative to men with a level 30 ng/ml (RR
2.42; 95% CI 1.353.84) [84].Evenmenwitha25(OH)
vitamin D level 22.629.9 ng/ml had an increased risk
(RR 1.60; 95% CI 1.102.32) compared with those with
alevel30 ng/ml. In the Framingham offspring cohort
study, 25(OH) vitamin D was measured in 1,739 participants
without prior heart disease. At a mean follow-up of 5.4 years,
amongst those with hypertension, there was a 2-fold increase
in the risk of cardiovascular events for the participants with a
25(OH) vitamin D level <15 ng/ml compared to those with a
level 15 ng/ml [34]. The Ludwigshafen Risk and Cardiovas-
cular Health Study, a prospective cohort comprising 3,300
patients referred to coronary angiography and followed for
7.7 years, demonstrated a strong association between vitamin
D status and several cardiovascular outcomes, such as cardio-
vascular mortality [85], stroke [86], heart failure and sudden
cardiac death with the lowest risk amongst those with the
highest 25(OH) vitamin D levels [87]. However, such associ-
ations have not been found in other studies. In the Osteopo-
rotic Fractures in Men Study, vitamin D intake was evaluated
in 3,094 men and 25(OH) vitamin D was measured in 813
men. The authors found no association between vitamin D
intake or 25(OH) vitamin D levels and incidence of cardio-
vascular disease during a median follow-up of 4.4 years [88].
Similarly, serum levels of 25(OH) vitamin D levels were not
independently associated with cardiovascular mortality
in the prospective Rancho Bernardo study including
1,073 community-dwelling older adults followed up to
10.4 years [89]. On the other hand, in a cross-sectional
study of 2,722 subjects, the prevalence of hypertension
was found to be increased in subjects with 25(OH) vitamin D
levels <40 ng/ml; odds ratios were 2.7 (1.45.2), 2.0 (1.45.2)
and 1.3 (1.21.6) for 25(OH) vitamin D levels <15, 1529 and
S6 Osteoporos Int (2012) 23 (Suppl 1):S1S23
3039 ng/ml, respectively, compared with the >40-ng/ml
group [90]. This inverse relationship between 25(OH) vitamin
D levels and hypertension has been recently confirmed in a
meta-analysis of 18 studies [91]. These various sets of data
raise the question of whether vitamin D supplementation can
prevent hypertension and cardiovascular events.
The evidence of benefit of vitamin D supplementation
from randomised trials is, however, scarce. In a small trial,
8 weeks of supplementation with vitamin D3 (800 UI/day)
and calcium was reportedly more effective in reducing sys-
tolic blood pressure than calcium alone [92]. In the Wom-
ens Health Initiative trial, including 36,282 postmenopausal
women, vitamin D3 plus calcium supplementation did not
reduce blood pressure, nor the risk of developing hyperten-
sion over 7 years of follow-up; however, in this trial, sup-
plementation consisted only of 400 IU/day and adherence to
supplementation was only around 60% [93]. A recent meta-
analysis of eight randomised clinical trials in patients with a
mean baseline blood pressure above 140/90 mmHg con-
cluded that vitamin D reduces blood pressure modestly but
significantly [94]. In summary, results from different studies
are conflicting and trials specifically assessing effects of
vitamin D on cardiovascular diseases as a primary endpoint
are lacking. It is therefore premature to recommend supple-
mental vitamin D intake for the prevention of cardiovascular
diseases or hypertension [95].
Vitamin D and the immune system
Vitamin D receptors are present in almost all immune cells,
including activated T and B lymphocytes and antigen-
presenting cells. Immune cells also express vitamin D-
activating enzymes, allowing local conversion of inactive
vitamin D into calcitriol within the immune system [96].
Several autoimmune diseases such as type 1 diabetes melli-
tus or multiple sclerosis are more frequent in countries with
less sunshine, and vitamin D deficiency in early life
increases the risk of autoimmune diseases and infections
later on [96,97]. There are several epidemiological studies
that have reported an association between vitamin D defi-
ciency and susceptibility to respiratory infections, especially
tuberculosis and Gram-negative infections [98]. Studies using
animal models of autoimmune diseases have identified vita-
min D as a potential modulator of differentiation, proliferation
and secretion processes in autoimmune reaction [96]. Supple-
mentation in humans might thus be preventive in a number of
autoimmune disorders.
A Finnish birth-cohort study, including >10,000 children
born in 1966, showed that vitamin D supplementation dur-
ing the first year of life (2,000 IU/day) was associated with a
risk reduction of 78% for developing type 1 diabetes (fol-
lowed up until end 1997) compared to no supplementation
or use of lower doses [99]. A meta-analysis of data from
four casecontrol studies and one cohort study support the
beneficial effects of vitamin D in prevention of type 1
diabetes [100]. A more recent supplementation study, how-
ever, was negative [101]. Data indicate that treatment with
vitamin D could be beneficial in reducing the risk of devel-
oping multiple sclerosis and diminishing its exacerbations
[102]. Although contradictory data exist concerning supple-
mentation benefits in rheumatoid arthritis (RA) and system-
ic lupus erythematosus, an association between low levels of
25(OH) vitamin D levels and activity of both diseases has
been reported [103,104]. Furthermore, an inverse associa-
tion between higher intake of vitamin D and risk of rheu-
matoid arthritis was demonstrated in the Iowa Womens
Health Study [105]. However, we still lack non-biased large
cohort studies that can sustain the proposed benefits of
vitamin D supplementation for optimal immune function.
Large-scale intervention trials in humans that support the
findings in preclinical or observational studies are lacking
[96].
Vitamin D and cancer treatment and prevention
Many experimental data show that calcitriol stimulates ap-
optosis and differentiation and inhibits angiogenesis and
proliferation in tumour cells [106]. Numerous association
studies suggest that serum 25(OH) vitamin D levels are
inversely associated with the risk of many types of cancer.
Further, in some studies of patients with cancer, an associ-
ation between low 25(OH) vitamin D levels and poor prog-
nosis has been observed [107,108]. A meta-analysis of
available studies indicated that there is a trend for lower
incidence of colorectal carcinoma and adenoma with 25(OH)
vitamin D levels >20 ng/ml in a doseresponse association
[109]. For breast cancer, a pooled analysis of two studies with
880 cases and 880 controls demonstrated that individuals with
sufficient serum 25(OH) vitamin D levels had 50% lower risk
of breast cancer than those with levels <13 ng/ml [110]. In
addition, a large casecontrol study on 1,394 post-menopausal
breast cancer patients and 1,365 controls also showed that the
25(OH) vitamin D level was significantly associated with
lower breast cancer risk, particularly at levels above 20 ng/ml
[111]. Most evidence concerning the link between vitamin D
and cancer is derived from laboratory studies andobservational
investigations of 25(OH) vitamin D levels in association with
cancer incidence and outcome. There are, however, sev-
eral possible confounding factors and association cannot
prove causation. Moreover, results from prospective stud-
ies only are more heterogeneous and do not support a
significant association between vitamin D status and
breast cancer [112].
There have been no clinical trials with cancer incidence
or mortality as a primary outcome to support causality
between vitamin D status and cancer. One population-
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S7
based randomised clinical trial found that calcium plus
vitamin D supplementation decreased cancer incidence as
a secondary outcome. In that study including 1,179 healthy
postmenopausal women aged >55 years, the mean level of
25(OH) vitamin D at baseline was 29 ng/ml. Supplementa-
tion with 1,100 IU vitamin D/day increased serum 25(OH)
vitamin D to 38 ng/ml. After 4 years of treatment, the
supplemented group had a 60% lower risk of developing
cancer than the placebo group [113]. However, a recent re-
analysis has indicated that this inverse association between
vitamin D levels and cancer incidence disappeared after
adjustment for BMI and physical activity [9,112]. In anoth-
er randomised trial, the Womens Health Initiative, no effect
of calcium and 400 IU vitamin D/day was found on the
incidence of colorectal or breast cancer, which were second-
ary outcomes [114]. However, the dose of 400 IU used in
that trial may have been inadequate to raise 25(OH) vitamin
D blood levels significantly, particularly after factoring in
adherence levels. A recent review of randomised vitamin D
supplementation trials with cancer incidence as a secondary
endpoint concluded that the results were null [112]. More-
over, the recent large-scale Cohort Consortium Vitamin D
Pooling Project of Rarer Cancersshowed no evidence link-
ing higher serum 25(OH) vitamin D levels to reduced risks
of less common cancers, including endometrial, gastric,
kidney, pancreatic and ovarian cancers [115]. In summary,
the available evidence that vitamin D reduces cancer inci-
dence is inconsistent and inconclusive. Randomised con-
trolled trials assessing vitamin D supplementation for
cancer prevention are in progress. Their results are to be
awaited before promoting vitamin D supplementation to
reduce cancer risk.
As a general conclusion, the importance of vitamin D for
bone health and the prevention of osteomalacia and osteo-
porosis are well recognized. More recently, vitamin D defi-
ciency has been associated with other chronic conditions,
including cardiovascular disease, autoimmune diseases and
cancer. However, most evidence for the importance of vita-
min D in these conditions comes from laboratory studies
and observational investigations. Randomised controlled
trials are needed to determine whether long-term supple-
mentation with vitamin D has a favourable impact on the
development or clinical course of non-skeletal diseases
[116].
Bisphosphonates
BPs are the mainstay in the treatment of osteoporosis and
other metabolic bone diseases such as Pagets disease, as
well as in tumoural conditions such as multiple myeloma,
bone metastases and cancer-induced hypercalcaemia. Their
efficacy and safety have been thoroughly established on the
basis of multiple large pivotal trials dealing with their main
indications. Their daily use in clinical medicine since 1969
has confirmed the general conclusions of the trials. Their
strong affinity for the skeleton partially explains their excel-
lent safety profile for other systems of the body. Even at
high pharmacologic doses, their bone affinity grossly pre-
cludes tissue uptake outside the skeleton. First of all, intes-
tinal absorption after oral administration is weak, on the
order of less than 1%, even under ideal conditions (after a
prolonged fast, with a full glass of water, and remaining
fasting for at least 30 min in an upright position before any
other food or beverage intake), leading to very low peak
values in the plasma. After intravenous administration, how-
ever, if the plasma peak levels are higher, these levels are
transient and short-lived. Similarly to what is observed after
oral administration, serum levels rapidly decrease due to
their rapid adsorption on the surface of bone (±50%). The
rest is cleared by both glomerular filtration and proximal
tubular secretion (± the remaining 50%) [117]. The retention
time in the skeleton is extremely long and depends on the
individual bone affinity of the various BPs. Part of the
released BPs from the skeleton can be re-uptaken, and part
is eliminated in the urine. Even if their terminal half-life is
long, plasma levels remain very low. However, small
amounts have been detected in body fluids up to 8 years
after stopping the drug [118,119]. This justified some
warning regarding the use of BPs in premenopausal women
of child bearing age. Even if there has been no demonstrated
adverse foetal events in humans, large controlled studies are
lacking to confirm their widespread safe use [120]. Some
caution to restrict the use BPs to severe condition is still
justified.
Bisphosphonate and acute phase reaction
After the first intravenous administration of a nitrogen-
containing bisphosphonate (n-BP) (e.g. disodium pamidro-
nate, zoledronic acid, ibandronate), about 25% of patients
experienced flu-like symptoms, consisting of transient and
self-limited fever, myalgias and/or arthralgias for 2 to 3 days.
Acute phase reaction (APR) has been associated with the
release of serum inflammatory cytokines such as tumour
necrosis factor (TNFα) and IL-6, but not IL-1 [121]. The
origin of these pro-inflammatory agents was homed on
monocytes and/or macrophages [122] but also in human
peripheral blood γδ T cells, which could constitute the
trigger for activation of the former cells [123]. The APRs
were absent or at least strongly attenuated with subsequent
infusions with n-BPs. The APR has also been observed after
high-dose oral monthly ibandronate [124]. The post-
infusion syndrome can be reduced by acetaminophen
[125]. It has been suggested that the co-administration of
statins could prevent this reaction [123,126], but this
S8 Osteoporos Int (2012) 23 (Suppl 1):S1S23
preventative effect does not seem to be systematic [127]. On
the contrary, concomitant glucocorticoid (GC) therapy did
not alleviate it [128]. Depletion in 25(OH)D could constitute
a factor favouring the occurrence of APR after n-BPs infu-
sion in n-BP-naive patients, but this remains to be confirmed
[129].
Bisphosphonate and musculoskeletal pain
Some cases of prolonged musculoskeletal pain have been
reported [130] in up to 20% to 25% of patients on alendr-
onate and risedronate, as well as zoledronic acid [128,131].
The majority of patients experienced gradual relief of pain
after discontinuation of the drug. A few patients redevel-
oped pain following re-challenge of the drug. No plausible
explanation has been proposed for their occurrence, and the
association between BPs and musculoskeletal pain has
therefore been questioned [132].
Bisphosphonate and the risk of renal failure
In line with the renal elimination of BPs, it is not recom-
mended to prescribe BPs to patients with a creatinine clear-
ance less than 30 ml/min, and this is specified in the
Summary of Products Characteristics of BP who were
granted an European Marketing Authorisation. In all pivotal
studies of BPs, chronic kidney diseases (CKD) constituted
an exclusion criterion, based on the calculated estimated
glomerular filtration rate using the formula of Miller et al.
[133]. In these large studies, however, several patients with
CKD, but without other calcium metabolism abnormalities,
notably in serum calcium, phosphate, alkaline phosphatase,
vitamin D and PTH were included. Some exceptions to this
30-ml/min rule could therefore be theoretically possible
[133135]. Even if clinical trials and clear recommendations
in the population with CKD are lacking, many clinicians
suggested to halve the dose or reduce the frequency of
administration of BPs in CKD [135]. Potential indications
of BPs in CKD are the prevention of bone loss in kidney
after transplantation. However, in these cases, no antifrac-
ture efficacy has so far been demonstrated with BP use
[136138]. Moreover, some patients treated with IV pamidr-
onate developed low-bone turnover adynamic bone [137].
Calciphylaxis is a rare complication of CKD. Case reports
have suggested the potential usefulness of BPs in its treat-
ment [139,140]. Proteinuria and proximal tubular necrosis
has been described in mice and rats after parenteral doses of
pamidronate sodium and clodronate five to 20 times higher
than clinical doses used in humans [141]. However, acute
renal toxicity was also reported in humans after rapid infu-
sion of high doses of non-n-BPs [142]. Renal function
deterioration, defined by elevations in the serum creatinine
level, was observed in up to 15% of the patients receiving
4 mg of zoledronic acid over 15 min in trials of treatment for
bone metastases (compared with 6.7% to 11.5% in patients
on placebo) [143]. In the doses registered for the treatment
of postmenopausal osteoporosis, oral BPs did not adversely
affect the renal function. With intravenous zoledronic acid
infusions, with infusion times of 15 min, short-term
increases in serum creatinine have been observed for 9 to
11 days in a small subset of patients [144]. It seems there-
fore justified that patients be well hydrated and avoid simul-
taneous therapeutic agents at risk of impairing renal
function. Patients with a glomerular filtration rate less than
30 ml/min should ideally be excluded, the precise diagnosis
of bone loss in such patients being uncertain. Other kinds of
bone disease than osteoporosis could be present [144]. As
there exists no head-to-head comparative trial, it is not
possible to determine whether intravenous n-BPs such as
pamidronate disodium or ibandronate would have a different
renal safety profile than zoledronic acid [144].
Bisphosphonate and ocular risk
Cases of iritis, episcleritis and scleritis, but also conjuncti-
vitis, have been reported after therapy with n-BPs (mainly
alendronate, pamidronate disodium and zoledronic acid) in
up to 1% [145147]. This does not seem to constitute an
exclusive complication for n-BPs, but they were rarely
reported with first-generation BPs [148]. Eye inflammation
can resolve after local GC administration, but some patients
can recur after BP rechallenge. In severe cases of uveitis and
scleritis, it could be better to discontinue IV BP [149].
Bisphosphonate and the gastrointestinal tract
Digestive problems are at the origin of most drug with-
drawals with oral n-BPs, mainly due to oesophageal irrita-
tion and upper gastrointestinal side effects [150]. They are
poorly absorbed by the gastrointestinal tract, of the order of
about 1%. Moreover, their absorption is further reduced if
they are taken with food and beverage such as coffee, milk,
orange juice etc. Hence, the recommendation is to take them
in a fasting condition with a glass of water and to remain
fasting in an upright position for at least 30 min after
swallowing the drug until the first meal of the day. These
precautions help to prevent most upper gastrointestinal side
effects [151]. Moreover, the availability of weekly and
monthly BPs has further decreased the frequency of the
upper gastrointestinal tract symptoms [152157]. It has been
suggested that a lot of adverse events in upper gastrointes-
tinal tract might be already present prior to start BPs therapy
[158] and that clinicians and patients may sometimes inap-
propriately attribute gastrointestinal complaints to therapy
[159]. Irrespective of whether gastrointestinal symptoms in
individual patients are linked with oral BPs or not, it should
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S9
be remembered that such a link has not been reported with
intravenous therapy.
A study based on the General Practice Research Database
containing anonymised patient records of about six million
people in UK suggested a doubling of the incidence of
oesophageal cancer with 5 yearsuse of oral BPs [160],
but this was not confirmed in another analysis of the same
database [161]. No excess of gastric and colorectal cancer
was found. Moreover, in patients with Barretts oesophagus
on oral BPs, no increased risk of oesophageal adenocarci-
noma was observed [162]. Even if no definitive conclusion
can be drawn from these studies, upper gastrointestinal
investigation is recommended if a patient on BPs develops
dysphagia and pain.
Bisphosphonates and cardiovascular risk
In the pivotal study of zoledronic acid versus placebo in
postmenopausal osteoporotic women, atrial fibrillation
reported as serious adverse events (SAEs) was more fre-
quent in the actively treated patients (1.3% versus 0.5%;
p<0.001). This was not observed in the HORIZON recurrent
fracture trial, in which a similar frequency of seriousatrial
fibrillation was observed both in actively treated and placebo-
treated patients (1.1% versus 1.3%) [163]. Post hoc analyses
of previous main trials on alendronate, risedronate and ibandr-
onate having involved about 30,000 patients did not show any
clear-cut association with atrial fibrillation [164166]. It is
possible that a lot of BP-treated patients have increased risks
of cardiovascular events already before the start of therapy
[167,168]. Also, any potential cardiovascular risk should be
weighted against the benefits of BP therapy. These include the
well-documented antifracture efficacy, of course, but may also
include additional benefits like the mortality benefit after hip
fracture with zoledronic acid therapy, a 30% mortality reduc-
tion not simply attributable to anti-fracture efficacy [163,
169].
Bisphosphonate and hypocalcaemia
BPs and in particular n-BPs are potent inhibitors of osteo-
clastic bone resorption. They can therefore provoke hypo-
calcaemia, hypocalciuria and PTH reaction in some cases.
Etidronate, however, did not induce any fall in serum and
urine calcium because it acutely impaired the accretion of
calcium into bone, offsetting a hypocalcaemic response
[170]. Even with intravenous potent n-BPs, symptomatic
hypocalcaemia rarely occurs in the treatment of osteoporosis
under usual conditions, i.e. with supplemental calcium and
vitamin D, lack of pre-existing hypoparathyroidism and/or
renal failure.
Miscellaneous
Skin reactions like rash, pruritus and urticaria have been
rarely reported with BP use. Re-challenge was positive
in some cases [171]. Change of BP was not always
accompanied by resurgence of symptoms, suggesting
that BP-induced cutaneous reactions are probably not
attributable to a class effect [171].
Extremely rare case reports of damage to the oral
mucosa, apparently not related to osteonecrosis of the
jaw, have been reported with the incorrect administra-
tion of n-BPs. Discontinuation of the inappropriate use
allowed healing of the mucosa ulcers, even with main-
tained oral intake, but taken according to the prescription
instructions [172].
A few reports of transient hepatitis after months to years
of alendronate and/or risedronate, with liver biopsies
compatible with a drug-induced toxicity, have been
described [173,174]. Healing occurred soon or later
after stopping the drug.
Bisphosphonates and cancer
BPs constitute an efficacious therapy in order to prevent
skeletal complications in patients with bone metastases.
They might help to maintain functional independence and
quality of life [175]. Several BPs have shown some efficacy
in this regard, but owing to its easy mode of administration
and its potency, zoledronic acid became the most used drug.
Improved quality of life and prolonged disease-free survival
have been observed with adjuvant therapy with zoledronic
acid. In addition, zoledronic acid has shown a direct inhibi-
tion of tumorigenesis and cellular growth in preclinical
models. So far, clinical results remain controversial [160,
176183].
SAPHO syndrome
Synovitis, acne, pustulosis, hyperostosis and osteitis syn-
drome is a rare condition consisting of sterile inflammatory
osteoarticular disorders, frequently associated with skin
lesions resistant to conventional anti-inflammatory therapy
[184]. Several case reports have shown successful therapy
with infusions of pamidronate disodium and zoledronic acid
[185,186].
Multicentric reticulohistiocytosis
Multicentric reticulohistiocytosis is a rare systemic condi-
tion characterized by erosive polyarthritis frequently pro-
gressing to arthritis mutilans and papulonodular lesions on
the skin. Alleviation of the arthritis and concurrent reduction
S10 Osteoporos Int (2012) 23 (Suppl 1):S1S23
of the size and number of cutaneous nodules have been
observed in single case reports with therapy with alendronate,
pamidronate and zoledronic acid [187].
Hypertrophic osteoarthropathy
Hypertrophic osteoarthropathy can be disabling and resis-
tant to analgesic and anti-inflammatory drugs. Clubbing,
arthralgias, cutaneous and osseous (periosteal) proliferation
in the upper and lower extremities are frequently associated
with bronchogenic carcinoma and right-to-left cardiac
shunts. A few case reports have shown an effective
alleviation of symptoms after pamidronate disodium and
zoledronic acid in both benign and malignant conditions
[188].
There are potentially other indications for BPs such as
periodontitis leading to local bone loss. However, there is
not yet enough evidence to recommend a wide use of BPs in
the treatment of this condition. Moreover, the theoretical
albeit questioned risk of osteonecrosis of the jaw could deter
clinicians to use them thoughtlessly [189].
Selective oestrogen receptor modulators (SERMs)
SERMs and the risk of stroke
Several meta-analyses have reported an increased risk of
stroke with tamoxifen use. Braithwaite et al. [190] observed
a 49% increased stroke risk (RR 1.49; 95% CI 1.16 to 1.90).
Similarly, Bushnell and Goldstein [191] found an OR of
1.82 (95% CI 1.41 to 2.36) for ischemic stroke and 1.40
(1.14 to 1.72) for any stroke. During a mean follow-up
period of 4.9 years, the frequency of ischemic stroke was
0.71% with tamoxifen versus 0.39% for controls (absolute
increased risk, 0.32%; number needed to harm, 313).
In the Ruth study, the incidence of all strokes did not
differ between raloxifene (incidence rate per 100 woman-
years 00.95) and placebo (incidence rate 00.86) treatment
groups (p00.30). There was, however, in the group of
women assigned to raloxifene a higher incidence of fatal
strokes than amongst placebo users (incidence rates 00.22
and 0.15, respectively, p00.0499). No significant subgroup
interactions were found except that there was a higher
incidence of stroke associated with raloxifene use amongst
current smokers [192]. Lasofoxifene, contrary to other
SERMs, at a dose of 0.5 mg/day, as compared with placebo,
was associated with reduced stroke risk (2.5 versus 3.9 cases
per 1,000 person-years; hazard ratio 0.64; 95% CI 0.41 to
0.99) in a randomised osteoporosis trial (8,556 women)
[193].
SERMs and cardiovascular risk
In the meta-analysis conducted by Braithwaite et al. [190],
tamoxifen was associated with significantly decreased myo-
cardial infarction deaths (RR 0.62; 95% CI 0.41 to 0.93) but
not myocardial infarction incidence (RR 0.90; 95% CI 0.66
to 1.23). Five years of treatment with tamoxifen was asso-
ciated with reduced mortality from coronary heart disease
compared with that in the 2-year group (hazard ratio 00.67,
95% confidence interval 00.47 to 0.94. Ten years after
surgery, 2.1% of the patients in the 5-year group and 3.5%
of those in the 2-year group had died from coronary heart
disease.
Initial results from the breast prevention studies reported
that tamoxifen was associated with a doubling of the risk of
deep-vein thrombosis and pulmonary embolism. This was
reported for instance during the active treatment of the IBIS-
I trial (52 versus 23 cases, RR02.26, 95% CI 01.36 to 3.87),
but not after tamoxifen was stopped (16 versus 14 cases,
RR01.14, 95% CI00.52 to 2.53) [194]. Similarly,
Braithwaite et al., observed a 88% increased pulmonary
emboli risk (RR 1.88; 95% CI 1.77 to 3.01).
The Raloxifene Use for The Heart (RUTH) trial showed
that raloxifene had no overall effect on the incidence of
coronary events in women with established coronary heart
disease or coronary heart disease risk factors. In addition,
raloxifene had no effect on the incidence of coronary events
in any subgroup except in the case of a post hoc age subgroup
analysis using age categories defined in the WomensHealth
Initiative randomised trials. The effect of raloxifene on the
incidence of coronary events differed significantly by age
(interaction p00.0118). The incidence of coronary events in
women <60 years of age was significantly lower in those
assigned raloxifene (50 events) compared with placebo (84
events; hazard ratio 0.59; 95% confidence interval, 0.41 to
0.83; p00.003; absolute risk reduction, 36 per 1,000 women
treated for 1 year). No difference was found between
treatment groups in the incidence of coronary events in
women > or 060 and <70 or > or 070 years of age [195].
Adomaityte et al. [196] assessed the risk of raloxifene on
venous thromboembolism using a meta-analysis (nine trials,
24,523 postmenopausal women) and found a 62% increase
in odds of either DVT or PE (odds ratio 1.62; 95% CI 1.25
to 2.09). Similarly, raloxifene therapy was associated with
54% increase in odds of DVT (odds ratio 1.54; 95% CI 1.13
to 2.11) and 91% increase in odds of PE alone (odds ratio
1.91; 95% CI 1.05 to 3.47). The excess event rate, in the
More trial, was 1.8 per 1,000 woman-years (95% CI 0.54.1),
and the number needed to treat to cause one event was 170
(95% CI 100582) over 3.3 years [197]. Similarly to what is
observed with tamoxifen and with menopause hormone ther-
apy, the excess of risk is more pronounced during the first
2 years of use. Similar results were seen in the RUTH trial.
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S11
Overall, raloxifene use was associated with an increased VTE
risk (HR 1.44, 95% CI 1.061.95) versus placebo. Concomi-
tant use of aspirin or non-aspirin antiplatelet agents along with
raloxifene did not change VTE risk [198]. Still the risk with
raloxifene seems lower than with tamoxifen, since in the
updated report of the STAR trial (TAM versus RALOX),
Toxicity RRs (raloxifene/tamoxifen) were 0.75 (95% CI
0.600.93) for thromboembolic events.
Lasofoxifene was associated with reduced risks of coro-
nary heart disease events (5.1 versus 7.5 cases per 1,000
person-years; hazard ratio 0.68; 95% CI 0.50 to 0.93) [193].
There was a reduced risk of coronary revascularization
(hazard ratio 0.56; 95% CI 0.32 to 0.98), hospitalization
for unstable angina (hazard ratio 0.55; 95% CI 0.29 to
1.04) but no reduction of coronary death or nonfatal myo-
cardial infarction [199].
SERMs and global mortality and morbidity
In a post hoc analysis of the MORE osteoporosis treat-
ment trial (7,705 postmenopausal women), the global
index outcome (defined as described for the WHI trial;
i.e. occurrence of coronary heart disease, stroke, pulmo-
nary embolism, invasive breast cancer, endometrial can-
cer, colorectal cancer, hip fracture or death because of
other causes) resulted in annual rates of 1.39% and
1.83% in the raloxifene and placebo groups, respectively
(HR 0.75; 95% CI 0.620.92), which were compatible
with a favourable riskbenefit profile for raloxifene
[200]. A pooled analysis of mortality data was performed
from large clinical trials of raloxifene (60 mg/day) versus
placebo, including the MORE/CORE trials (7,705 post-
menopausal osteoporotic women followed for 4 years
and a subset of 4,011 participants followed for an additional
4 years; 110 deaths) and the RUTH trial (10,101 postmeno-
pausal women with coronary disease or multiple risk factors
for coronary disease followed for 5.6 years; 1,149 deaths).
All-cause mortality was 10% lower amongst women assigned
to raloxifene 60 mg/day versus placebo (relative hazard 0.90;
95% CI 0.801.00; p00.05). Lower overall mortality was
primarily due to lower rates of non-cardiovascular deaths,
especially a lower rate of non-cardiovascular, non-cancer
deaths [201]. The mechanism whereby raloxifene might
reduce the risk of non-cardiovascular death remains
unclear.
SERMs and cancer risk
It is well-known that tamoxifen is associated with signifi-
cantly increased risks of endometrial cancer (RR 2.70; 95%
CI 1.94 to 3.75) [190]. SERMS like tamoxifen and ralox-
ifene are approved in the USA, but not in Europe, for
reducing breast cancer risk in patients at risk of breast
cancer. It has been repeatedly shown that tamoxifen reduces
the risk of invasive ER-positive tumours [194].
On the hand, raloxifene did not increase risk for endo-
metrial hyperplasia (RR 1.3; 95% CI 0.45.1), or endome-
trial cancer (RR 0.9; 95% CI 0.32.7) [197]. In the updated
report of the STAR trial (TAM versus RALOX), Toxicity
RRs (raloxifene/ tamoxifen) were 0.55 (95% CI 0.360.83;
p00.003) for endometrial cancer (this difference was not
significant in the initial results) [202].
The MORE trial found that 4 years of raloxifene therapy
also decreased the incidence of invasive breast cancer
amongst postmenopausal women with osteoporosis by
72% compared with placebo. The CORE (an extension trial)
examined the effect of four additional years of raloxifene
therapy. Incidences of invasive breast cancer and ER-
positive invasive breast cancer were reduced by 59%
(HR00.41; 95% CI00.24 to 0.71) and 66% (HR 00.34;
95% CI00.18 to 0.66), respectively, in the raloxifene group
compared with the placebo group. There was no difference
between the two groups in incidence of ER-negative inva-
sive breast cancer. Over the 8 years of both trials, the
incidences of invasive breast cancer and ER-positive inva-
sive breast cancer were reduced by 66% (HR00.34; 95%
CI00.22 to 0.50) and 76% (HR00.24; 95% CI 00.15 to
0.40), respectively, in the raloxifene group compared with
the placebo group [203]. It has further been suggested that
breast cancer risk reduction persists for some time in
patients who discontinue raloxifene although this conclu-
sion is limited by the post hoc analyses in unrandomised
patients and the small sample sizes [204]. Raloxifene re-
duced also the incidence of invasive breast cancer by 44%
(HR00.56; 95% CI00.38 to 0.83; absolute risk reduction 0
1.2 invasive breast cancers per 1,000 women treated for
1 year) in the RUTH trial [205]. The lower incidence of
invasive breast cancer reflected a 55% lower incidence of
invasive ER-positive tumours (HR 00.45; 95% CI 00.28 to
0.72). However, raloxifene treatment did not reduce the
incidence of non-invasive breast cancer or of invasive ER-
negative breast cancer. The reduced incidence of invasive
breast cancer was similar across subgroups, including those
defined by age, body mass index, family history of breast
cancer, prior use of postmenopausal hormones and 5-year
estimated risk of invasive breast cancer. An updated analysis
with an 81-month median follow-up of the STAR trial
(tamoxifen (20 mg/day) or raloxifene (60 mg/day) for
5 years in women at high-risk breast cancer) was published
in 2010 [202]. The RR (raloxifene/ tamoxifen) for invasive
breast cancer was 1.24 (95% CI 1.051.47) and for non-
invasive disease, 1.22 (95% CI 0.951.59). Compared with
initial results, the RRs widened for invasive and narrowed
for non-invasive breast cancer [202]. There were no signif-
icant mortality differences. Long-term raloxifene retained
76% of the effectiveness of tamoxifen in preventing invasive
S12 Osteoporos Int (2012) 23 (Suppl 1):S1S23
disease and grew closer over time to tamoxifen in preventing
non-invasive disease.
In the PEARL trial (n08,556), lasofoxifene 0.5 mg re-
duced the risk of total breast cancer by 79% (hazard ratio
0.21; 95% CI 0.08 to 0.55) and ER+ invasive breast cancer
by 83% (hazard ratio 0.17; 95% CI 0.05 to 0.57) compared
with placebo. This effect was similar regardless of Gail
score, whereas the effects were markedly stronger for women
with higher baseline estradiol levels [206].
SERMs and menopausal symptoms
In breast cancer patients, it has been well documented that
tamoxifen increases both severity and frequency of hot
flushes.
The situation is likely less severe when using raloxifene.
Some RCTs did not report an increased frequency or sever-
ity of vasomotor symptoms in women discontinuing oestro-
genprogestin as compared with placebo [207,208].
Nevertheless, other studies reported an increase in hot
flushes when using raloxifene [209], which led to the sug-
gestion of a gradual conversion to raloxifene from low-dose
oestrogen, with a progression from 60 mg every alternate
day to 60 mg/day.
It has been showed in short duration studies that it is
possible to avoid SERMs associated hot flushes and meno-
pausal symptoms, using a combination of a SEM (bazedox-
ifene) and estrogens (conjugated estrogens) [210].
Some non-skeletal side effects are favourable (breast
cancer protection); others on the other hand are unfavourable
(stroke risk, thromboembolism and endometrial cancer). The
presence and the magnitude of these side effects vary between
SERMs concluding that women with breast cancer treated
with tamoxifen have an 82% increased risk of ischemic stroke
and a 29% increased risk of any stroke, although the absolute
risk remains small.
Strontium ranelate
Strontium ranelate is a first-line treatment for the man-
agement of postmenopausal osteoporosis. Its dual mode
of action simultaneously reduces bone resorption and
increases bone formation [211]. Strontium ranelate has
a limited number of non-skeletal effects, for which most
of the evidence comes from post hoc analyses of these
two trials.
Strontium and cartilage
Osteoarthritis involves the degeneration of joint cartilage
and the adjacent bone, which leads to joint pain and
stiffness.
There is some preclinical evidence for an effect of stron-
tium ranelate on cartilage degradation. Strontium ranelate
has been demonstrated to stimulate the production of pro-
teoglycans in isolated human chondrocytes, leading to car-
tilage formation without affecting cartilage resorption [212].
There is also evidence for an impact on biomarkers of
cartilage degradation. Treatment with strontium ranelate
was associated with significantly lower levels of urinary
excretion of a marker of cartilage degradation (CTX-II)
(p<0.0001) [213,214].
The potential for a clinical effect of strontium ranelate in
osteoarthritis indicated that 3 yearstreatment with stron-
tium ranelate was associated with a 42% lower overall
osteoarthritis score (p00.0005 versus placebo) and a 33%
reduction in disc space narrowing score (p00.03 versus
placebo). These changes were concomitant to a 34% in-
crease in the number of patients free of back pain (p00.03
versus placebo) [215].
Strontium ranelate and cardiovascular risk
The possibility of a vascular effect was raised following
a pooled analysis of results in the SOTI and TROPOS
populations, which found a higher annual incidence of
venous thromboembolism over 5 years with strontium
ranelate than with placebo (0.9% versus 0.6%; relative
risk 1.4; 95% CI 1.02.0) [216]. Although these rates of
venous thromboembolism were similar to those in the
age-matched general population [217219], they merited
further investigation. The possibility of an impact was
therefore explored in a retrospective study in the Gen-
eral Practice Research Database (GPRD) [220]. The
GPRD was used to identify 11,546 women with osteo-
porosis but no treatment, 20,084 women with osteopo-
rosis treated with alendronate and 2,408 women with
osteoporosis treated with strontium ranelate; 115,009
women without osteoporosis were used as a comparator
group [220]. Women with osteoporosis but no treatment
were at greater risk for venous thromboembolism than
women without osteoporosis (hazard ratio 1.43; 95% CI
1.101.86; p00.007; age-adjusted model), possibly due
to the reduced mobility associated with bone disease.
On the other hand, there was no difference in the rates
of venous thromboembolism in the samples of women
with osteoporosis (no treatment, strontium ranelate or
alendronate). Similar findings have been reported from
other observational studies [221,222],whichallaystoa
great extent the concerns.
Strontium ranelate and cutaneous adverse reactions
The other non-skeletal effect of concern with strontium rane-
late is the occurrence of rare cases of cutaneous
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S13
hypersensitivity reactions, which are manifested as drug reac-
tion with eosinophilia and systemic symptoms (DRESS) or
toxic epidermal necrolysis [223226] (19-22). The pathogen-
esis of these hypersensitivity reactions remains unclear. Early
recognition and appropriate management, including drug
withdrawal, can improve the prognosis. The incidence of
these adverse reactions is extremely low, estimated at 1/
54,000 patient-years of treatment. This is most likely why
no cases were detected in the phase 3 clinical trials. Similarly,
no cases were reported in the observational study following
over 13,000 patients receiving strontium ranelate over 2 years
[222].
In conclusion, strontium ranelate has few non-skeletal
effects. A possible beneficial effect on cartilage degradation
and formation may translate into a new therapy for osteoarthri-
tis. Observational studies suggest no cause for concern over
possible vascular effects, whilst the rate of hypersensitivity
reactions with cutaneous effects remains very low.
Denosumab
Denosumab is a fully human monoclonal antibody that
inhibits the activity of the ligand for receptor activating
NFκB (RANKL), the main stimulator of osteoclastogenesis
and of osteoclast activity [227].
The potential extra skeletal effects of denosumab concern
its interaction with RANK function in non-skeletal tissues,
as RANK is largely expressed in several cell types, mainly
of the immunological and vascular systems [228].
Denosumab and the immune and inflammatory response
Besides its major role to regulate bone resorption, the
RANK/RANKL/OPG system is also an important regulator
of the immune system where it is produced by T cells and
enhances dendritic cells survival and antigen presentation
[229]. A theoretical concern is the possible effect of deno-
sumab on the susceptibility to infectious diseases and on the
risk of cancer. A deregulation of the immune system could
also lead to the appearance of atopic disease or autoimmune
diseases. Conversely, there could be a benefit in inflamma-
tory diseases. However, though RANK and RANK-L are
essential in mice for ontogeny of the lymphoid tissues [227],
patients with a mutation of the RANKL gene did not present
immunological defects [230]. Suppression of RANKL does
not interfere with inflammatory or immune response in
mature individuals, and RANKL inhibition did not prevent
inflammatory disease in several rat and mice models, except
in the IL-2-deficient mice whose lymphocytes over express
RANKL [229,231].
The only human model of inflammatory disease in which
denosumab has been used is RA. The authors followed at
MRI for 12 months 143 patients receiving 60 or 180 mg
injections of denosumab every 6 months. All patients were
treated with methotrexate. At 12 months, the MRI erosion
score was less increased from baseline in both denosumab
groups than in the patients receiving a placebo (p<0.012
and 0.007, respectively), but there was no evidence of an
effect of denosumab on joint space narrowing or on meas-
ures of RA disease activity [232]. Thus, denosumab cannot
substitute for DMARDs or anti-TNF in RA but could be an
interesting adjuvant in patients with progression of bone
erosions; beside, it could prevent osteoporosis associated
with RA, particularly in patients requiring glucocorticoid
treatment [233].
Concerning the problem of atopic disease and suscepti-
bility to infections, Stolina et al. have shown that mice
treated with OPG, the natural inhibitor of RANKL signal-
ling, did not differ from controls with regard to contact
hypersensitivity or infectious load induced by mycobacterial
infection [234]. There was no decrease of humoral or cellu-
lar immunity. Another study in mice showed that inhibition
of RANK signalling by a single dose of RANK-Fc 100 or
500 μg, which inhibits hypercalcaemia induced by 1,
25-dihydroxyvitamin D, did not decrease the immune
response to influenza infection [235].
In the first clinical study in postmenopausal women with
low bone density [236], the 1.9% of neoplasms in the
denosumab group versus none in the placebo or alendronate
groups was intriguing though not significant. However, in
the FREEDOM study, including nearly 4,000 patients trea-
ted for 3 years with denosumab, the incidence of neoplasia
did not differ significantly from the placebo group (3.7%
versus 3.2%) [237]. In this study, the authors found a sig-
nificant increase of eczema (3.0% versus 1.7%) and of
cellulitis (0.3% versus <0.1%) reported as SAEs in the
denosumab group but no difference in the overall proportion
of patients with skin infection. Other clinical trials did not
provide evidence for an increased risk of infectious compli-
cations either [238240]. Because denosumab is a relatively
recent treatment option, continued follow-up of any poten-
tial safety signals will be required, as with other agents in
osteoporosis.
Denosumab and cardiovascular risks
RANKL and OPG could also play a role in the regulation of
vascular calcification. Mice knocked out for OPG developed
extensive vascular calcifications [241]. OPG produced lo-
cally by endothelial cells could promote endothelial survival
and decrease atherotic plate mineralisation [228]. Several
clinical studies have shown that circulating OPG was higher
in patients with cardiovascular diseases, particularly in ter-
minal renal failure [242,243], an increase considered as a
reaction to the inflammatory signal [244]. One human study
S14 Osteoporos Int (2012) 23 (Suppl 1):S1S23
has shown conversely an inverse relationship between OPG
and echogenicity of carotid plaques, thus that individuals
with more fibrous and calcified plates had a lower serum
OPG concentration [245]. Inhibiting RANKL decreased
vascular calcifications in human RANKL knocked-in mice
with glucocorticoid induced osteoporosis [246]. Thus, one
could expect that besides protecting bone, denosumab could
decrease the risk of atherosclerosis. The clinical trials on
bone efficacy in osteoporosis and osteopenia did not show
differences in cardiovascular accidents in the denosumab-
treated patients. However, these studies were not designed
to study this end point, and the cardiovascular risk in the
patients included was not high (6.8% of the patients in the
placebo group of the FREEDOM study had a cardiovascular
event, stroke, coronary heart disease or peripheral vascular
disease). It would be interesting to look at high-risk subgroups
and to include cardiovascular events as an end point in osteo-
penia or osteoporosis studies conducted in patients at in-
creased risk of atheromatosis, like those with glucocorticoid
induced osteoporosis.
Teriparatide and parathyroid hormone(184)
The biological activity of the intact human PTH, i.e. PTH
(184), resides in its N-terminal sequence. Within the PTH
peptide family, teriparatide, the recombinant human PTH
(134) fragment has been most extensively developed
for clinical use in osteoporosis.
Miscellaneous effects
In clinical trials, commonly reported mild side effects have
been headaches (8%), nausea (8%), dizziness (9%) and leg
cramps (3%), with only for the latter two a significantly
higher incidence compared to placebo. These side effects tend
to occur within the first few hours following subcutaneous
injection [247,248].
Subcutaneous injection of 20 μg of teriparatide results in
a limited increase (around 0.8 mg/dl) of serum calcium,
peaking after 4 to 6 h, followed by a progressive return to
baseline before the next injection. These changes occur
usually within the physiologic range, with occasional, mild
hypercalcaemia having been observed in 11% of patients in
the pivotal clinical trial. Repeated or persistent hypercalcae-
mia necessitating reduction or cessation of concomitant
calcium supplementation and/or teriparatide dose reduction
occurred in about 3% of patients. In this trial, the 24-h urinary
calcium excretion showed a modest increase with a median of
30 mg/24 h. There were no clinical consequences, but patients
with history of hypercalciuria or of urinary calculi in the past
5 years were excluded from the trial. Significant increases of
serum uric acid have been observed in about 3% of patients.
Although these biochemical changes are generally mild, it has
been suggested that treatment with teriparatide should be
avoided in subjects with a history of nephrolithiasis or gout,
unless close monitoring is undertaken of serum and urinary
calcium excretion or serum uric acid [247,248].
The more limited data available on treatment with PTH
(184) suggests that at a proposed dose of 100 μg/day,
transient hypercalcaemia might be more frequent and mild
hypercalciuria observed in up to 10% of patients [249,250].
Mild local irritation with erythema at the injection site can
occur with teriparatide and PTH(184) [226,247].
Recently, teriparatide and PTH(184) have been proposed
as a possible therapeutic option for hypoparathyroidism [251,
252].
Conclusions
There is no doubt about the skeletal efficacy of bone drugs
as used in their registered indications: treatment of osteopo-
rosis in males and females, Pagets disease of bone, multiple
myeloma, bone metastases, cancer-induced hypercalcaemia,
prevention and treatment of glucocorticoid induced osteo-
porosis or bone loss after hormonal deprivation in hormone
sensitive cancers as, e.g. prostate or breast. Fractures can be
prevented and bone pain and progressive bone disease lim-
ited. In this manuscript, an extensive review of non-skeletal
effects of these drugs is presented. These can be either
beneficial or deleterious.
Beneficial non-skeletal effects are proven for vitamin D
and SERMs. Fall reduction, improved muscular function
and physical performance are observed for substitution with
adequate doses of vitamin D (800 IU/day) in deficient
populations. As the health impact of falls is broader than
for fractures only, fall reduction is a separate, valuable
clinical outcome. For SERMs, long-term (up to 8 years)
primary chemoprevention of oestrogen receptor positive
breast cancers in postmenopausal women is documented.
Viewing the lower level of evidence of non-vertebral fracture
reduction by SERMs compared to other anti-resorptive bone
drugs, breast cancer prevention contributes to the preferred
use of SERMs in a specific therapeutic niche determined by
younger age, axial osteoporosis and increased breast cancer
risk.
More recently, some studies illustrated a reduction in mor-
tality (with vitamin D, SERMs, IV bisphosphonate), which
was probably not related to the fracture reduction. This inter-
esting observation requires confirmation by additional large
scaled and long-term studies including specific endpoints on
cardiovascular risk factors and events and cancer.
Other promising beneficial effects are described for
strontium on cartilage and spinal osteoarthritis and for deno-
sumab on the prevention of bone erosions in rheumatoid
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S15
arthritis. More clinical trials are needed to validate a poten-
tial use in these therapeutic areas. Furthermore animal or
observational data support some speculation on potential
benefits of calcium on ischemic cardiac mortality and
stroke; of vitamin D on cardiovascular outcomes, autoim-
mune diseases and cancer prevention and of SERMs on
coronary events and of denosumab on the prevention of
vascular calcification.
The most frequent non-skeletal side effects of bone drugs
are the gastrointestinal intolerance of calcium supplements
and oral bisphosphonates, contributing in part to the
reported low adherence of these drugs, and the acute phase
reactions following intravenous amino-bisphosphonates
applications. More important side effects in terms of sever-
ity, but fortunately infrequent, are stroke and venous throm-
boembolic events for SERMs and endometrium cancer for
tamoxifen. A severe cutaneous hypersensitivity reaction,
described as DRESS syndrome, has been reported in ex-
tremely rare case (only 16 reported) in clinical practice with
strontium ranelate, although etiologic linkage remains
doubtful. Hypocalcaemia has rarely been observed in
bisphosphonate and denosumab trials (including calcium
and vitamin D repleted patients); moreover, it was mild,
transient and asymptomatic. Some studies, but not all, report
kidney stones and myocardial infarction as side effects of
calcium supplements and renal toxicity for iv pamidronate
and zoledronate. Speculative side effects are discussed: mus-
culoskeletal pain, uveitis, scleritis and oesophageal cancer for
oral bisphosphonates and atrial fibrillation for iv zoledronate,
coronary disease for SERMs, venous thromboembolism of
strontium ranelate and skin infections for denosumab.
In conclusion, some of the non-skeletal effects of bone
drugs, either beneficial or deleterious, may influence treatment
choices, whereas others still require more studies to reveal
additional insights into remaining questions concerning the
clinical management of patients with bone diseases.
Conflicts of interest Jean-Jacques Body has received speaker and
consultant fees from Amgen and Novartis and research support from
Amgen, Daiichi Sankyo, GlaxoSmithKline, Merck Sharp & Dohme,
Novartis, Nycomed, Servier and SMB.
Pierre Bergmann has received speaker fees from Servier and Roche.
Steven Boonen has received consulting fees and/or research support
from Amgen, Merck, Novartis and Servier
Jean-Pierre Devogelaer has no conflict of interest.
Evelien Gielen has no conflict of interest.
Stephan Goemaere has received speakers fees and/or research sup-
port from Amgen, Daiichi Sankyo, Eli Lilly, Glaxo Smith Kline, Merck
Sharp & Dohme, Novartis, Nycomed, Warner Chillcott, Sanofi-
Aventis, Servier and Roche.
Jean-Marc Kaufman has received consulting fees, paid advisory
boards, lecture fees and/or grant support from Amgen, Eli Lilly, Glaxo
Smith Kline, Merck, Novartis, Procter & Gamble, Roche, Sanofi
Aventis, Servier and Warner Chilcott.
Serge Rozenberg has received speakers or/and consultant fees from
Amgen, Merck Sharp & Dohme and Pfizer.
Jean-Yves Reginster on behalf of the Department of Public Health,
Epidemiology and Health Economics of the University of Liège,
Liège, Belgium has received consulting fees or paid advisory boards
from Servier, Novartis, Negma, Lilly, Wyeth, Amgen, GlaxoSmithKline,
Roche, Merckle, Nycomed, NPS, Theramex and UCB; lecture fees when
speaking at the invitation of a commercial sponsor from Merck Sharp and
Dohme, Lilly, Rottapharm, IBSA, Genevrier, Novartis, Servier, Roche,
GlaxoSmithKline, Teijin, Teva, Ebewee Pharma, Zodiac, Analis,
Theramex, Nycomed and Novo-Nordisk and grant support from indus-
tries Bristol Myers Squibb, Merck Sharp & Dohme, Rottapharm, Teva,
Lilly, Novartis, Roche, GlaxoSmithKline and Amgen, Servier.
Funding This supplement was not sponsored by any outside com-
mercial interests. It was funded entirely by the Belgian Bone Club, a
non-profit scientific organisation.
Open Access This article is distributed under the terms of the Crea-
tive Commons Attribution Noncommercial License which permits any
noncommercial use, distribution, and reproduction in any medium,
provided the original author(s) and source are credited.
References
1. Body JJ, Bergmann P, Boonen S, Boutsen Y, Devogelaer JP,
Goemaere S, Kaufman JM, Rozenberg S, Reginster JY (2010)
Evidence-based guidelines for the pharmacological treatment of
postmenopausal osteoporosis: a consensus document by the Bel-
gian Bone Club. Osteoporos Int 21:16571680
2. Boonen S, Vanderschueren D, Geusens P, Bouillon R (1997)
Age-associated endocrine deficiencies as potential determinants
of femoral neck (type II) osteoporotic fracture occurrence in
elderly men. Int J Androl 20:134143
3. Boonen S, Bischoff-Ferrari HA, Cooper C, Lips P, Ljunggren O,
Meunier PJ, Reginster JY (2006) Addressing the musculoskeletal
components of fracture risk with calcium and vitamin D: a review
of the evidence. Calcif Tissue Int 78:257270
4. Boonen S, Vanderschueren D, Haentjens P, Lips P (2006) Calcium
and vitamin D in the prevention and treatment of osteoporosisa
clinical update. J Intern Med 259:539552
5. Group D (2010) Patient level pooled analysis of 68 500 patients
from seven major vitamin D fracture trials in US and Europe.
BMJ 340:b5463
6. Tang BM, Eslick GD, Nowson C, Smith C, Bensoussan A (2007)
Use of calcium or calcium in combination with vitamin D sup-
plementation to prevent fractures and bone loss in people aged 50
years and older: a meta-analysis. Lancet 370:657666
7. Avenell A, Gillespie WJ, Gillespie LD, OConnell D (2009)
Vitamin D and vitamin D analogues for preventing fractures
associated with involutional and post-menopausal osteoporosis.
Cochrane Database Syst Rev CD000227
8. Boonen S, Lips P, Bouillon R, Bischoff-Ferrari HA, Vanderschueren
D, Haentjens P (2007) Need for additional calcium to reduce the risk
of hip fracture with vitamin d supplementation: evidence from a
comparative metaanalysis of randomized controlled trials. J Clin
Endocrinol Metab 92:14151423
9. Chung M, Balk EM, Brendel M et al (2009) Vitamin D and
calcium: a systematic review of health outcomes. Evid Rep
Technol Assess (Full Rep) 1420
10. Bostick RM, Kushi LH, Wu Y, Meyer KA, Sellers TA, Folsom
AR (1999) Relation of calcium, vitamin D, and dairy food intake
to ischemic heart disease mortality among postmenopausal women.
Am J Epidemiol 149:151161
S16 Osteoporos Int (2012) 23 (Suppl 1):S1S23
11. Knox EG (1973) Ischaemic-heart-disease mortality and dietary
intake of calcium. Lancet 1:14651467
12. Iso H, Stampfer MJ, Manson JE, Rexrode K, Hennekens CH,
Colditz GA, Speizer FE, Willett WC (1999) Prospective study of
calcium, potassium, and magnesium intake and risk of stroke in
women. Stroke 30:17721779
13. Griffith LE, Guyatt GH, Cook RJ, Bucher HC, Cook DJ (1999)
The influence of dietary and nondietary calcium supplementation
on blood pressure: an updated metaanalysis of randomized con-
trolled trials. Am J Hypertens 12:8492
14. Wang L, Manson JE, Buring JE, Lee IM, Sesso HD (2008)
Dietary intake of dairy products, calcium, and vitamin D and
the risk of hypertension in middle-aged and older women. Hy-
pertension 51:10731079
15. Dickinson HO, Nicolson DJ, Cook JV, Campbell F, Beyer FR,
Ford GA, Mason J (2006) Calcium supplementation for the
management of primary hypertension in adults. Cochrane Data-
base Syst Rev CD004639
16. Reid IR, Horne A, Mason B, Ames R, Bava U, Gamble GD
(2005) Effects of calcium supplementation on body weight and
blood pressure in normal older women: a randomized controlled
trial. J Clin Endocrinol Metab 90:38243829
17. Govers MJ, Van der Meet R (1993) Effects of dietary calcium and
phosphate on the intestinal interactions between calcium, phos-
phate, fatty acids, and bile acids. Gut 34:365370
18. Denke MA, Fox MM, Schulte MC (1993) Short-term dietary
calcium fortification increases fecal saturated fat content and
reduces serum lipids in men. J Nutr 123:10471053
19. Zemel MB, Shi H, Greer B, Dirienzo D, Zemel PC (2000)
Regulation of adiposity by dietary calcium. FASEB J
14:11321138
20. Reid IR, Mason B, Horne A, Ames R, Clearwater J, Bava U, Orr-
Walker B, Wu F, Evans MC, Gamble GD (2002) Effects of
calcium supplementation on serum lipid concentrations in normal
older women: a randomized controlled trial. Am J Med 112:343
347
21. Bostick RM, Fosdick L, Grandits GA, Grambsch P, Gross M,
Louis TA (2000) Effect of calcium supplementation on serum
cholesterol and blood pressure. A randomized, double-blind,
placebo-controlled, clinical trial. Arch Fam Med 9:3138, dis-
cussion 39
22. Heaney RP (2003) Normalizing calcium intake: projected popu-
lation effects for body weight. J Nutr 133:268S270S
23. Parikh SJ, Yanovski JA (2003) Calcium intake and adiposity. Am
J Clin Nutr 77:281287
24. Barr SI (2003) Increased dairy product or calcium intake: is body
weight or composition affected in humans? The Journal of nutri-
tion 133:245S248S
25. Trowman R, Dumville JC, Hahn S, Torgerson DJ (2006) A
systematic review of the effects of calcium supplementation on
body weight. Br J Nutr 95:10331038
26. Lanou AJ, Barnard ND (2008) Dairy and weight loss hypothesis:
an evaluation of the clinical trials. Nutr Rev 66:272279
27. Bolland MJ, Avenell A, Baron JA, Grey A, MacLennan GS,
Gamble GD, Reid IR (2010) Effect of calcium supplements on
risk of myocardial infarction and cardiovascular events: meta-
analysis. BMJ 341:c3691
28. Bolland MJ, Barber PA, Doughty RN, Mason B, Horne A, Ames
R, Gamble GD, Grey A, Reid IR (2008) Vascular events in
healthy older women receiving calcium supplementation: rando-
mised controlled trial. BMJ 336:262266
29. Reid IR, Schooler BA, Hannan SF, Ibbertson HK (1986) The
acute biochemical effects of four proprietary calcium prepara-
tions. Aust N Z J Med 16:193197
30. Foley RN, Collins AJ, Ishani A, Kalra PA (2008) Calcium-
phosphate levels and cardiovascular disease in community-
dwelling adults: the Atherosclerosis Risk in Communities
(ARIC) Study. Am Heart J 156:556563
31. Vestergaard P, Mollerup CL, Frokjaer VG, Christiansen P,
Blichert-Toft M, Mosekilde L (2003) Cardiovascular events be-
fore and after surgery for primary hyperparathyroidism. World J
Surg 27:216222
32. Jackson RD, LaCroix AZ, Gass M et al (2006) Calcium plus
vitamin D supplementation and the risk of fractures. N Engl J
Med 354:669683
33. Hsia J, Heiss G, Ren H et al (2007) Calcium/vitamin D supple-
mentation and cardiovascular events. Circulation 115:846854
34. Wang TJ, Pencina MJ, Booth SL, Jacques PF, Ingelsson E, Lanier
K, Benjamin EJ, DAgostino RB, Wolf M, Vasan RS (2008)
Vitamin D deficiency and risk of cardiovascular disease. Circu-
lation 117:503511
35. Autier P, Gandini S (2007) Vitamin D supplementation and total
mortality: a meta-analysis of randomized controlled trials. Arch
Intern Med 167:17301737
36. Lewis JR, Calver J, Zhu K, Flicker L, Prince RL (2011) Calcium
supplementation and the risks of atherosclerotic vascular disease
in older women: results of a 5-year RCT and a 4.5-year follow-up.
J Bone Miner Res 26:3541
37. Park Y, Leitzmann MF, Subar AF, Hollenbeck A, Schatzkin A
(2009) Dairy food, calcium, and risk of cancer in the NIH-AARP
Diet and Health Study. Arch Intern Med 169:391401
38. Martinez ME, Willett WC (1998) Calcium, vitamin D, and colo-
rectal cancer: a review of the epidemiologic evidence. Cancer
Epidemiol Biomarkers Prev 7:163168
39. Wu K, Willett WC, Fuchs CS, Colditz GA, Giovannucci EL
(2002) Calcium intake and risk of colon cancer in women and
men. J Natl Cancer Inst 94:437446
40. Cho E, Smith-Warner SA, Spiegelman D et al (2004) Dairy
foods, calcium, and colorectal cancer: a pooled analysis of 10
cohort studies. J Natl Cancer Inst 96:10151022
41. Shaukat A, Scouras N, Schunemann HJ (2005) Role of supplemen-
tal calcium in the recurrence of colorectal adenomas: a metaanalysis
of randomized controlled trials. Am J Gastroenterol 100:390394
42. Bond JH (2000) Polyp guideline: diagnosis, treatment, and sur-
veillance for patients with colorectal polyps. Practice Parameters
Committee of the American College of Gastroenterology. Am J
Gastroenterol 95:30533063
43. Wactawski-Wende J, Kotchen JM, Anderson GL et al (2006)
Calcium plus vitamin D supplementation and the risk of colorec-
tal cancer. N Engl J Med 354:684696
44. Weingarten MA, Zalmanovici A, Yaphe J (2008) Dietary calcium
supplementation for preventing colorectal cancer and adenoma-
tous polyps. Cochrane Database Syst Rev CD003548
45. Shin MH, Holmes MD, Hankinson SE, Wu K, Colditz GA,
Willett WC (2002) Intake of dairy products, calcium, and vitamin
d and risk of breast cancer. J Natl Cancer Inst 94:13011311
46. Lin J, Manson JE, Lee IM, Cook NR, Buring JE, Zhang SM
(2007) Intakes of calcium and vitamin D and breast cancer risk in
women. Arch Intern Med 167:10501059
47. McCullough ML, Rodriguez C, Diver WR, Feigelson HS, Stevens
VL, Thun MJ, Calle EE (2005) Dairy, calcium, and vitamin D
intake and postmenopausal breast cancer risk in the Cancer Preven-
tion Study II Nutrition Cohort. Cancer Epidemiol Biomarkers Prev
14:28982904
48. Larsson SC, Bergkvist L, Wolk A (2009) Long-term dietary
calcium intake and breast cancer risk in a prospective cohort of
women. Am J Clin Nutr 89:277282
49. Rodriguez C, McCullough ML, Mondul AM, Jacobs EJ,
Fakhrabadi-Shokoohi D, Giovannucci EL, Thun MJ, Calle EE
(2003) Calcium, dairy products, and risk of prostate cancer in a
prospective cohort of United States men. Cancer Epidemiol Bio-
markers Prev 12:597603
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S17
50. Mitrou PN, Albanes D, Weinstein SJ, Pietinen P, Taylor PR,
Virtamo J, Leitzmann MF (2007) A prospective study of dietary
calcium, dairy products and prostate cancer risk (Finland). Int J
Cancer 120:24662473
51. Giovannucci E, Liu Y, Platz EA, Stampfer MJ, Willett WC
(2007) Risk factors for prostate cancer incidence and progression
in the health professionals follow-up study. Int J Cancer
121:15711578
52. Hedlund TE, Moffatt KA, Miller GJ (1996) Stable expression of
the nuclear vitamin D receptor in the human prostatic carcinoma
cell line JCA-1: evidence that the antiproliferative effects of 1
alpha, 25-dihydroxyvitamin D3 are mediated exclusively through
the genomic signaling pathway. Endocrinology 137:15541561
53. Koh KA, Sesso HD, Paffenbarger RS Jr, Lee IM (2006) Dairy
products, calcium and prostate cancer risk. Br J Cancer 95:1582
1585
54. Chan JM, Pietinen P, Virtanen M, Malila N, Tangrea J, Albanes
D, Virtamo J (2000) Diet and prostate cancer risk in a cohort of
smokers, with a specific focus on calcium and phosphorus (Fin-
land). Cancer Causes Control 11:859867
55. Rohrmann S, Platz EA, Kavanaugh CJ, Thuita L, Hoffman SC,
Helzlsouer KJ (2007) Meat and dairy consumption and subse-
quent risk of prostate cancer in a US cohort study. Cancer Causes
Control 18:4150
56. Curhan GC, Willett WC, Speizer FE, Spiegelman D, Stampfer
MJ (1997) Comparison of dietary calcium with supplemental
calcium and other nutrients as factors affecting the risk for kidney
stones in women. Ann Intern Med 126:497504
57. Curhan GC, Willett WC, Rimm EB, Stampfer MJ (1993) A
prospective study of dietary calcium and other nutrients and the
risk of symptomatic kidney stones. N Engl J Med 328:833838
58. Curhan GC, Willett WC, Knight EL, Stampfer MJ (2004) Dietary
factors and the risk of incident kidney stones in younger women:
NursesHealth Study II. Arch Intern Med 164:885891
59. Bihl G, Meyers A (2001) Recurrent renal stone disease-advances
in pathogenesis and clinical management. Lancet 358:651656
60. Holick MF (2007) Vitamin D deficiency. N Engl J Med 357:266281
61. Bischoff-Ferrari HA, Willett WC, Wong JB, Stuck AE, Staehelin
HB, Orav EJ, Thoma A, Kiel DP, Henschkowski J (2009) Pre-
vention of nonvertebral fractures with oral vitamin D and dose
dependency: a meta-analysis of randomized controlled trials.
Arch Intern Med 169:551561
62. Bischoff HA, Borchers M, Gudat F, Duermueller U, Theiler R,
Stahelin HB, Dick W (2001) In situ detection of 1,25-dihydrox-
yvitamin D3 receptor in human skeletal muscle tissue. Histochem
J 33:1924
63. Demay M (2003) Muscle: a nontraditional 1,25-dihydroxyvita-
min D target tissue exhibiting classic hormone-dependent vitamin
D receptor actions. Endocrinology 144:51355137
64. Capiati DA, Vazquez G, Boland RL (2001) Protein kinase C
alpha modulates the Ca2+ influx phase of the Ca2+ response to
1alpha,25-dihydroxy-vitamin-D3 in skeletal muscle cells. Horm
Metab Res 33:201206
65. Dirks-Naylor AJ, Lennon-Edwards S (2011) The effects of vita-
min D on skeletal muscle function and cellular signaling. J
Steroid Biochem Mol Biol
66. Venning G (2005) Recent developments in vitamin D defi-
ciency and muscle weakness among elderly people. BMJ
330:524526
67. Visser M, Deeg DJ, Lips P (2003) Low vitamin D and high
parathyroid hormone levels as determinants of loss of muscle
strength and muscle mass (sarcopenia): the Longitudinal Aging
Study Amsterdam. J Clin Endocrinol Metab 88:57665772
68. Bischoff-Ferrari HA, Borchers M, Gudat F, Durmuller U, Stahe-
lin HB, Dick W (2004) Vitamin D receptor expression in human
muscle tissue decreases with age. J Bone Miner Res 19:265269
69. Sato Y, Iwamoto J, Kanoko T, Satoh K (2005) Low-dose vitamin
D prevents muscular atrophy and reduces falls and hip fractures
in women after stroke: a randomized controlled trial. Cerebrovasc
Dis 20:187192
70. Snijder MB, van Schoor NM, Pluijm SM, van Dam RM, Visser
M, Lips P (2006) Vitamin D status in relation to one-year risk of
recurrent falling in older men and women. J Clin Endocrinol
Metab 91:29802985
71. Bischoff-Ferrari HA, Dawson-Hughes B, Staehelin HB, Orav JE,
Stuck AE, Theiler R, Wong JB, Egli A, Kiel DP, Henschkowski J
(2009) Fall prevention with supplemental and active forms of
vitamin D: a meta-analysis of randomised controlled trials. BMJ
339:b3692
72. Gilsanz V, Kremer A, Mo AO, Wren TA, Kremer R (2010)
Vitamin D status and its relation to muscle mass and muscle fat
in young women. J Clin Endocrinol Metab 95:15951601
73. Annweiler C, Beauchet O, Berrut G, Fantino B, Bonnefoy M,
Herrmann FR, Schott AM (2009) Is there an association between
serum 25-hydroxyvitamin D concentration and muscle strength
among older women? Results from baseline assessment of the
EPIDOS study. J Nutr Health Aging 13:9095
74. Gupta R, Sharma U, Gupta N, Kalaivani M, Singh U, Guleria R,
Jagannathan NR, Goswami R (2010) Effect of cholecalciferol and
calcium supplementation on muscle strength and energy metab-
olism in vitamin D-deficient Asian Indians: a randomized, con-
trolled trial. Clin Endocrinol (Oxf) 73:445451
75. Zhu K, Austin N, Devine A, Bruce D, Prince RL (2010) A
randomized controlled trial of the effects of vitamin D on muscle
strength and mobility in older women with vitamin D insufficien-
cy. J Am Geriatr Soc 58:20632068
76. Pfeifer M, Begerow B, Minne HW, Suppan K, Fahrleitner-
Pammer A, Dobnig H (2009) Effects of a long-term vitamin D
and calcium supplementation on falls and parameters of muscle
function in community-dwelling older individuals. Osteoporos
Int 20:315322
77. Stockton KA, Mengersen K, Paratz JD, Kandiah D, Bennell KL
(2011) Effect of vitamin D supplementation on muscle strength: a
systematic review and meta-analysis. Osteoporos Int 22:859871
78. Pilz S, Tomaschitz A, Drechsler C, Dekker JM, Marz W (2010)
Vitamin D deficiency and myocardial diseases. Mol Nutr Food
Res 54:11031113
79. Tishkoff DX, Nibbelink KA, Holmberg KH, Dandu L, Simpson
RU (2008) Functional vitamin D receptor (VDR) in the t-tubules
of cardiac myocytes: VDR knockout cardiomyocyte contractility.
Endocrinology 149:558564
80. Schleithoff SS, Zittermann A, Tenderich G, Berthold HK,
Stehle P, Koerfer R (2006) Vitamin D supplementation
improves cytokine profiles in patients with congestive heart
failure: a double-blind, randomized, placebo-controlled trial.
Am J Clin Nutr 83:754759
81. Sugden JA, Davies JI, Witham MD, Morris AD, Struthers AD
(2008) Vitamin D improves endothelial function in patients with
type 2 diabetes mellitus and low vitamin D levels. Diabet Med
25:320325
82. Pittas AG, Lau J, Hu FB, Dawson-Hughes B (2007) The role of
vitamin D and calcium in type 2 diabetes. A systematic review
and meta-analysis. J Clin Endocrinol Metab 92:20172029
83. Pilz S, Tomaschitz A, Ritz E, Pieber TR (2009) Vitamin D status
and arterial hypertension: a systematic review. Nat Rev Cardiol
6:621630
84. Giovannucci E, Liu Y, Hollis BW, Rimm EB (2008) 25-
Hydroxyvitamin D and risk of myocardial infarction in men: a
prospective study. Arch Intern Med 168:11741180
85. Dobnig H, Pilz S, Scharnagl H, Renner W, Seelhorst U, Wellnitz
B, Kinkeldei J, Boehm BO, Weihrauch G, Maerz W (2008)
Independent association of low serum 25-hydroxyvitamin d and
S18 Osteoporos Int (2012) 23 (Suppl 1):S1S23
1,25-dihydroxyvitamin d levels with all-cause and cardiovascular
mortality. Arch Intern Med 168:13401349
86. Pilz S, Dobnig H, Fischer JE, Wellnitz B, Seelhorst U, Boehm
BO, Marz W (2008) Low vitamin d levels predict stroke in
patients referred to coronary angiography. Stroke 39:26112613
87. Pilz S, Marz W, Wellnitz B, Seelhorst U, Fahrleitner-Pammer A,
Dimai HP, Boehm BO, Dobnig H (2008) Association of vitamin
D deficiency with heart failure and sudden cardiac death in a large
cross-sectional study of patients referred for coronary angiography.
J Clin Endocrinol Metab 93:39273935
88. Messenger W, Nielson CM, Li H, Beer T, Barrett-Connor E, Stone
K, Shannon J (2012) Serum and dietary vitamin D and cardiovas-
cular disease risk in elderly men: a prospective cohort study. Nutr
Metab Cardiovasc Dis. doi:10.1016/j.numecd.2011.10.019
89. Jassal SK, Chonchol M, von Muhlen D, Smits G, Barrett-Connor
E (2010) Vitamin d, parathyroid hormone, and cardiovascular
mortality in older adults: the Rancho Bernardo study. Am J
Med 123:11141120
90. Bhandari SK, Pashayan S, Liu IL, Rasgon SA, Kujubu DA, Tom
TY, Sim JJ (2011) 25-Hydroxyvitamin D levels and hypertension
rates. J Clin Hypertens (Greenwich) 13:170177
91. Burgaz A, Orsini N, Larsson SC, Wolk A (2011) Blood 25-
hydroxyvitamin D concentration and hypertension: a meta-
analysis. J Hypertens 29:636645
92. Pfeifer M, Begerow B, Minne HW, Nachtigall D, Hansen C
(2001) Effects of a short-term vitamin D(3) and calcium supple-
mentation on blood pressure and parathyroid hormone levels in
elderly women. J Clin Endocrinol Metab 86:16331637
93. Margolis KL, Ray RM, Van Horn L et al (2008) Effect of calcium
and vitamin D supplementation on blood pressure: the Womens
Health Initiative Randomized Trial. Hypertension 52:847855
94. Witham MD, Nadir MA, Struthers AD (2009) Effect of vitamin D
on blood pressure: a systematic review and meta-analysis. J
Hypertens 27:19481954
95. Geleijnse JM (2011) Vitamin D and the prevention of hyperten-
sion and cardiovascular diseases: a review of the current evi-
dence. Am J Hypertens 24:253262
96. Mathieu C (2011) Vitamin D and immune system: getting it right.
IBMS BoneKEY 8:178186
97. Shoenfeld N, Amital H, Shoenfeld Y (2009) The effect of mela-
nism and vitamin D synthesis on the incidence of autoimmune
disease. Nat Clin Pract Rheumatol 5:99105
98. Hughes DA, Norton R (2009) Vitamin D and respiratory health.
Clin Exp Immunol 158:2025
99. Hypponen E, Laara E, Reunanen A, Jarvelin MR, Virtanen SM
(2001) Intake of vitamin D and risk of type 1 diabetes: a birth-
cohort study. Lancet 358:15001503
100. Zipitis CS, Akobeng AK (2008) Vitamin D supplementation in
early childhood and risk of type 1 diabetes: a systematic review
and meta-analysis. Arch Dis Child 93:512517
101. Brekke HK, Ludvigsson J (2007) Vitamin D supplementation and
diabetes-related autoimmunity in the ABIS study. Pediatr Diabetes
8:1114
102. Pierrot-Deseilligny C (2009) Clinical implications of a possible
role of vitamin D in multiple sclerosis. J Neurol 256:14681479
103. Amital H, Szekanecz Z, Szucs G et al (2010) Serum concentra-
tions of 25-OH vitamin D in patients with systemic lupus eryth-
ematosus (SLE) are inversely related to disease activity: is it time
to routinely supplement patients with SLE with vitamin D? Ann
Rheum Dis 69:11551157
104. Cutolo M, Otsa K, Uprus M, Paolino S, Seriolo B (2007) Vitamin
D in rheumatoid arthritis. Autoimmun Rev 7:5964
105. Merlino LA, Curtis J, Mikuls TR, Cerhan JR, Criswell LA, Saag
KG (2004) Vitamin D intake is inversely associated with rheu-
matoid arthritis: results from the Iowa Womens Health Study.
Arthritis Rheum 50:7277
106. Fleet JC (2008) Molecular actions of vitamin D contributing to
cancer prevention. Mol Aspects Med 29:388396
107. Tretli S, Hernes E, Berg JP, Hestvik UE, Robsahm TE (2009)
Association between serum 25(OH)D and death from prostate
cancer. Br J Cancer 100:450454
108. Goodwin PJ, Ennis M, Pritchard KI, Koo J, Hood N (2009)
Prognostic effects of 25-hydroxyvitamin D levels in early breast
cancer. J Clin Oncol 27:37573763
109. International Agency for Research on Cancer (IARC) (2008)
Vitamin D and Cancer. In Cancer IAfro (ed) IARC Working
Group reports. World Health Organisation, Lyon, pp 1221
110. Garland CF, Gorham ED, Mohr SB, Grant WB, Giovannucci EL,
Lipkin M, Newmark H, Holick MF, Garland FC (2007) Vitamin
D and prevention of breast cancer: pooled analysis. J Steroid
Biochem Mol Biol 103:708711
111. Abbas S, Linseisen J, Slanger T, Kropp S, Mutschelknauss EJ,
Flesch-Janys D, Chang-Claude J (2008) Serum 25-hydroxyvitamin
D and risk of post-menopausal breast cancerresults of a large case-
control study. Carcinogenesis 29:9399
112. Manson JE, Mayne ST, Clinton SK (2011) Vitamin D and pre-
vention of cancerready for prime time? N Engl J Med
364:13851387
113. Lappe JM, Travers-Gustafson D, Davies KM, Recker RR, Heaney
RP (2007) Vitamin D and calcium supplementation reduces cancer
risk: results of a randomized trial. Am J Clin Nutr 85:15861591
114. Chlebowski RT, Johnson KC, Kooperberg C et al (2008) Calcium
plus vitamin D supplementation and the risk of breast cancer. J
Natl Cancer Inst 100:15811591
115. Helzlsouer KJ (2010) Overview of the Cohort Consortium Vita-
min D Pooling Project of rarer cancers. Am J Epidemiol 172:49
116. Souberbielle JC, Body JJ, Lappe JM et al (2010) Vitamin D and
musculoskeletal health, cardiovascular disease, autoimmunity
and cancer: recommendations for clinical practice. Autoimmun
Rev 9:709715
117. Pazianas M, Cooper C, Ebetino FH, Russell RG (2010) Long-
term treatment with bisphosphonates and their safety in postmen-
opausal osteoporosis. Ther Clin Risk Manag 6:325343
118. Green JR, Rogers M (2002) Pharmacological profile of zole-
dronic acid: a highly potent inhibitor of bone resorption. Drug
Dev Res 55:210224
119. Papapoulos SE, Cremers SC (2007) Prolonged bisphosphonate
release after treatment in children. N Engl J Med 356:10751076
120. McNicholl DM, Heaney LG (2010) The safety of bisphosphonate
use in pre-menopausal women on corticosteroids. Curr Drug Saf
5:182187
121. Thiebaud D, Sauty A, Burckhardt P, Leuenberger P, Sitzler
L, Green JR, Kandra A, Zieschang J, Ibarra de Palacios P
(1997) An in vitro and in vivo study of cytokines in the
acute-phase response associated with bisphosphonates. Calcif
Tissue Int 61:386392
122. Sauty A, Pecherstorfer M, Zimmer-Roth I, Fioroni P, Juillerat L,
Markert M, Ludwig H, Leuenberger P, Burckhardt P, Thiebaud D
(1996) Interleukin-6 and tumor necrosis factor alpha levels after
bisphosphonates treatment in vitro and in patients with malignan-
cy. Bone 18:133139
123. Hewitt RE, Lissina A, Green AE, Slay ES, Price DA, Sewell AK
(2005) The bisphosphonate acute phase response: rapid and co-
pious production of proinflammatory cytokines by peripheral
blood gd T cells in response to aminobisphosphonates is inhibited
by statins. Clin Exp Immunol 139:101111
124. Miller PD, McClung MR, Macovei L et al (2005) Monthly oral
ibandronate therapy in postmenopausal osteoporosis: 1-year results
from the MOBILE study. J Bone Miner Res 20:13151322
125. Recker RR, Lewiecki EM, Miller PD, Reiffel J (2009) Safety of
bisphosphonates in the treatment of osteoporosis. Am J Med 122:
S2232
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S19
126. Thompson K, Rogers MJ (2004) Statins prevent bisphosphonate-
induced gamma, delta-T-cell proliferation and activation in vitro.
J Bone Miner Res 19:278288
127. Srivastava T, Haney CJ, Alon US (2009) Atorvastatin may have
no effect on acute phase reaction in children after intravenous
bisphosphonate infusion. J Bone Miner Res 24:334337
128. Reid DM, Devogelaer JP, Saag K et al (2009) Zoledronic acid and
risedronate in the prevention and treatment of glucocorticoid-
induced osteoporosis (HORIZON): a multicentre, double-blind,
double-dummy, randomised controlled trial. Lancet 373:1253
1263
129. Bertoldo F, Pancheri S, Zenari S, Boldini S, Giovanazzi B,
Zanatta M, Valenti MT, Dalle Carbonare L, Lo Cascio V (2010)
Serum 25-hydroxyvitamin D levels modulate the acute-phase
response associated with the first nitrogen-containing bisphosph-
onate infusion. J Bone Miner Res 25:447454
130. Bock O, Boerst H, Thomasius FE, Degner C, Stephan-Oelkers M,
Valentine SM, Felsenberg D (2007) Common musculoskeletal
adverse effects of oral treatment with once weekly alendronate
and risedronate in patients with osteoporosis and ways for their
prevention. J Musculoskelet Neuronal Interact 7:144148
131. Black DM, Delmas PD, Eastell R et al (2007) Once-yearly
zoledronic acid for treatment of postmenopausal osteoporosis.
N Engl J Med 356:18091822
132. Caplan L, Pittman CB, Zeringue AL, Scherrer JF, Wehmeier KR,
Cunningham FE, Eisen SA, McDonald JR (2010) An observa-
tional study of musculoskeletal pain among patients receiving
bisphosphonate therapy. Mayo Clin Proc 85:341348
133. Miller PD, Roux C, Boonen S, Barton IP, Dunlap LE, Burgio DE
(2005) Safety and efficacy of risedronate in patients with age-
related reduced renal function as estimated by the Cockcroft and
Gault method: a pooled analysis of nine clinical trials. J Bone
Miner Res 20:21052115
134. Jamal SA, Bauer DC, Ensrud KE, Cauley JA, Hochberg M,
Ishani A, Cummings SR (2007) Alendronate treatment in women
with normal to severely impaired renal function: an analysis of
the fracture intervention trial. J Bone Miner Res 22:503508
135. Toussaint ND, Elder GJ, Kerr PG (2009) Bisphosphonates in
chronic kidney disease; balancing potential benefits and adverse
effects on bone and soft tissue. Clin J Am Soc Nephrol 4:221233
136. Fan SL, Almond MK, Ball E, Evans K, Cunningham J (2000)
Pamidronate therapy as prevention of bone loss following renal
transplantation. Kidney Int 57:684690
137. Coco M, Glicklich D, Faugere MC et al (2003) Prevention of
bone loss in renal transplant recipients: a prospective, randomized
trial of intravenous pamidronate. J Am Soc Nephrol 14:2669
2676
138. Palmer SC, McGregor DO, Strippoli GF (2007) Interventions for
preventing bone disease in kidney transplant recipients. Cochrane
Database Syst Rev CD005015
139. Shiraishi N, Kitamura K, Miyoshi T et al (2006) Successful
treatment of a patient with severe calcific uremic arteriolopathy
(calciphylaxis) by etidronate disodium. Am J Kidney Dis
48:151154
140. Monney P, Nguyen QV, Perroud H, Descombes E (2004) Rapid
improvement of calciphylaxis after intravenous pamidronate ther-
apy in a patient with chronic renal failure. Nephrol Dial Trans-
plant 19:21302132
141. Body JJ (2006) The risk of cumulative renal effects of intrave-
nous bisphosphonates. Support Cancer Ther 3:7783
142. Bounameaux HM, Schifferli J, Montani JP, Jung A, Chatelanat F
(1983) Renal failure associated with intravenous diphosphonates.
Lancet 1:471
143. Ibrahim A, Scher N, Williams G et al (2003) Approval summary
for zoledronic acid for treatment of multiple myeloma and cancer
bone metastases. Clin Cancer Res 9:23942399
144. Miller PD (2011) The kidney and bisphosphonates. Bone 49:77
81
145. Rey J, Daumen-Legre V, Pham T, Bernard P, Dahan L, Acquaviva
PC, Lafforgue P (2000) Uveitis, an under-recognized adverse
effect of pamidronate. Case report and literature review. Joint
Bone Spine 67:337340
146. Malik AR, Campbell SH, Toma NM (2002) Bilateral acute ante-
rior uveitis after alendronate. Br J Ophthalmol 86:1443
147. Durnian JM, Olujohungbe A, Kyle G (2005) Bilateral acute
uveitis and conjunctivitis after zoledronic acid therapy. Eye
(Lond) 19:221222
148. Fietta P, Manganelli P, Lodigiani L (2003) Clodronate induced
uveitis. Ann Rheum Dis 62:378
149. Fraunfelder FW, Fraunfelder FT, Jensvold B (2003) Scleritis and
other ocular side effects associated with pamidronate disodium.
Am J Ophthalmol 135:219222
150. Lufkin EG, Argueta R, Whitaker MD, Cameron AL, Wong VH,
Egan KS, OFallon WM, Riggs BL (1994) Pamidronate: an
unrecognized problem in gastrointestinal tolerability. Osteoporos
Int 4:320322
151. de Groen PC, Lubbe DF, Hirsch LJ, Daifotis A, Stephenson W,
Freedholm D, Pryor-Tillotson S, Seleznick MJ, Pinkas H, Wang
KK (1996) Esophagitis associated with the use of alendronate. N
Engl J Med 335:10161021
152. Cryer B, Miller P, Petruschke RA, Chen E, Geba GP, Papp AE
(2005) Upper gastrointestinal tolerability of once weekly alendr-
onate 70 mg with concomitant non-steroidal anti-inflammatory
drug use. Aliment Pharmacol Ther 21:599607
153. Greenspan S, Field-Munves E, Tonino R, Smith M, Petruschke R,
Wang L, Yates J, de Papp AE, Palmisano J (2002) Tolerability of
once-weekly alendronate in patients with osteoporosis: a random-
ized, double-blind, placebo-controlled study. Mayo Clin Proc
77:10441052
154. Eisman JA, Rizzoli R, Roman-Ivorra J, Lipschitz S, Verbruggen
N, Gaines KA, Melton ME (2004) Upper gastrointestinal and
overall tolerability of alendronate once weekly in patients with
osteoporosis: results of a randomized, double-blind, placebo-
controlled study. Curr Med Res Opin 20:699705
155. Bobba RS, Beattie K, Parkinson B, Kumbhare D, Adachi JD
(2006) Tolerability of different dosing regimens of bisphospho-
nates for the treatment of osteoporosis and malignant bone dis-
ease. Drug Saf 29:11331152
156. Cadarette SM, Katz JN, Brookhart MA, Sturmer T, Stedman
MR, Levin R, Solomon DH (2009) Comparative gastrointes-
tinal safety of weekly oral bisphosphonates. Osteoporos Int
20:17351747
157. Rosen CJ, Hochberg MC, Bonnick SL et al (2005) Treatment
with once-weekly alendronate 70 mg compared with once-
weekly risedronate 35 mg in women with postmenopausal oste-
oporosis: a randomized double-blind study. J Bone Miner Res
20:141151
158. Vestergaard P, Schwartz K, Pinholt EM, Rejnmark L, Mosekilde
L (2010) Gastric and esophagus events before and during treat-
ment of osteoporosis. Calcif Tissue Int 86:110115
159. Cryer B, Bauer DC (2002) Oral bisphosphonates and upper
gastrointestinal tract problems: what is the evidence? Mayo Clin
Proc 77:10311043
160. Green J, Czanner G, Reeves G, Watson J, Wise L, Beral V (2010)
Oral bisphosphonates and risk of cancer of oesophagus, stomach,
and colorectum: casecontrol analysis within a UK primary care
cohort. BMJ 341:c4444
161. Cardwell CR, Abnet CC, Cantwell MM, Murray LJ (2010) Ex-
posure to oral bisphosphonates and risk of esophageal cancer.
JAMA 304:657663
162. Nguyen DM, Schwartz J, Richardson P, El-Serag HB (2010)
Oral bisphosphonate prescriptions and the risk of esophageal
S20 Osteoporos Int (2012) 23 (Suppl 1):S1S23
adenocarcinoma in patients with Barretts esophagus. Dig Dis Sci
55:34043407
163. Lyles KW, Colon-Emeric CS, Magaziner JS et al (2007) Zole-
dronic acid and clinical fractures and mortality after hip fracture.
N Engl J Med 357:17991809
164. Cummings SR, Schwartz AV, Black DM (2007) Alendronate and
atrial fibrillation. N Engl J Med 356:18951896
165. Karam R, Camm J, McClung M (2007) Yearly zoledronic acid in
postmenopausal osteoporosis. N Engl J Med 357:712713, au-
thor reply 714-715
166. Lewiecki EM, Cooper C, Thompson E, Hartl F, Mehta D, Papapoulos
SE (2010) Ibandronate does not increase risk of atrial fibril-
lation in analysis of pivotal clinical trials. Int J Clin Pract 64:821
826
167. Varma R, Aronow WS, Basis Y, Singh T, Kalapatapu K, Weiss
MB, Pucillo AL, Monsen CE (2008) Relation of bone mineral
density to frequency of coronary heart disease. Am J Cardiol
101:11031104
168. Choi SH, An JH, Lim S et al (2009) Lower bone mineral density
is associated with higher coronary calcification and coronary
plaque burdens by multidetector row coronary computed tomog-
raphy in pre- and postmenopausal women. Clin Endocrinol (Oxf)
71:644651
169. Eriksen EF, Lyles KW, Colon-Emeric CS et al (2009) Antifrac-
ture efficacy and reduction of mortality in relation to timing of the
first dose of zoledronic acid after hip fracture. J Bone Miner Res
24:13081313
170. McCloskey EV, Yates AJ, Beneton MN, Galloway J, Harris S,
Kanis JA (1987) Comparative effects of intravenous diphospho-
nates on calcium and skeletal metabolism in man. Bone 8(Suppl
1):S3541
171. Brinkmeier T, Kugler K, Lepoittevin JP, Frosch PJ (2007) Ad-
verse cutaneous drug reaction to alendronate. Contact Dermatitis
57:123125
172. Krasagakis K, Kruger-Krasagakis S, Ioannidou D, Tosca A
(2004) Chronic erosive and ulcerative oral lesions caused by
incorrect administration of alendronate. J Am Acad Dermatol
50:651652
173. Yanik B, Turkay C, Atalar H (2007) Hepatotoxicity induced by
alendronate therapy. Osteoporos Int 18:829831
174. Phillips MB (2007) Risedronate-induced hepatitis. Am J Med
120:e12
175. Coleman R, Cook R, Hirsh V, Major P, Lipton A (2011) Zole-
dronic acid use in cancer patients: more than just supportive care?
Cancer 117:1123
176. Gnant M, Clezardin P (2012) Direct and indirect anticancer
activity of bisphosphonates: a brief review of published literature.
Cancer Treat Rev (in press)
177. Normanno N, De Luca A, Gallo M, Lamura L, Perrone F (2011)
Zoledronic acid in early-stage breast cancer. Lancet Oncol
12:991
178. Coleman RE, Marshall H, Cameron D et al (2011) Breast-cancer
adjuvant therapy with zoledronic acid. N Engl J Med 365:1396
1405
179. Gnant M (2011) Zoledronic acid in breast cancer: latest findings
and interpretations. Ther Adv Med Oncol 3:293301
180. Zhao X, Xu X, Zhang Q, Jia Z, Sun S, Zhang J, Wang B,
Wang Z, Hu X (2011) Prognostic and predictive value of
clinical and biochemical factors in breast cancer patients with
bone metastases receiving metronomiczoledronic acid.
BMC Cancer 11:403
181. Chlebowski RT, Col N (2011) Bisphosphonates and breast cancer
prevention. Anticancer Agents Med Chem
182. Rennert G, Pinchev M, Rennert HS, Gruber SB (2011) Use of
bisphosphonates and reduced risk of colorectal cancer. J Clin
Oncol 29:11461150
183. Sassa S, Okabe H, Nemoto N, Kikuchi H, Kudo H, Sakamoto S
(2009) Ibandronate may prevent colorectal carcinogenesis in
mice with ulcerative colitis. Anticancer Res 29:46154619
184. Kahn MF, Chamot AM (1992) SAPHO syndrome. Rheum Dis
Clin North Am 18:225246
185. Kopterides P, Pikazis D, Koufos C (2004) Successful treatment of
SAPHO syndrome with zoledronic acid. Arthritis Rheum 50:2970
2973
186. Siau K, Laversuch CJ (2010) SAPHO syndrome in an adult
with ulcerative colitis responsive to intravenous pamidronate:
a case report and review of the literature. Rheumatol Int
30:10851088
187. Codriansky KA, Runger TM, Bhawan J, Kantarci A, Kissin EY
(2008) Multicentric reticulohistiocytosis: a systemic osteoclastic
disease? Arthritis Rheum 59:444448
188. King MM, Nelson DA (2008) Hypertrophic osteoarthropathy
effectively treated with zoledronic acid. Clin Lung Cancer
9:179182
189. Schonfeld SE (2010) Strategies for managing periodontal inflam-
mation. J Calif Dent Assoc 38:272283
190. Braithwaite RS, Chlebowski RT, Lau J, George S, Hess R, Col
NF (2003) Meta-analysis of vascular and neoplastic events asso-
ciated with tamoxifen. J Gen Intern Med 18:937947
191. Bushnell CD, Goldstein LB (2004) Risk of ischemic stroke with
tamoxifen treatment for breast cancer: a meta-analysis. Neurolo-
gy 63:12301233
192. Mosca L, Grady D, Barrett-Connor E et al (2009) Effect of
raloxifene on stroke and venous thromboembolism according to
subgroups in postmenopausal women at increased risk of coro-
nary heart disease. Stroke 40:147155
193. Cummings SR, Ensrud K, Delmas PD et al (2010) Lasofoxifene
in postmenopausal women with osteoporosis. N Engl J Med
362:686696
194. Cuzick J, Forbes JF, Sestak I, Cawthorn S, Hamed H, Holli K,
Howell A (2007) Long-term results of tamoxifen prophylaxis for
breast cancer96-month follow-up of the randomized IBIS-I
trial. J Natl Cancer Inst 99:272282
195. Collins P, Mosca L, Geiger MJ, Grady D, Kornitzer M, Amewou-
Atisso MG, Effron MB, Dowsett SA, Barrett-Connor E, Wenger
NK (2009) Effects of the selective estrogen receptor modulator
raloxifene on coronary outcomes in the Raloxifene Use for The
Heart trial: results of subgroup analyses by age and other factors.
Circulation 119:922930
196. Adomaityte J, Farooq M, Qayyum R (2008) Effect of raloxifene
therapy on venous thromboembolism in postmenopausal women.
A meta-analysis Thromb Haemost 99:338342
197. Grady D, Ettinger B, Moscarelli E, Plouffe L Jr, Sarkar S, Ciaccia
A, Cummings S (2004) Safety and adverse effects associated with
raloxifene: multiple outcomes of raloxifene evaluation. Obstet
Gynecol 104:837844
198. Duvernoy CS, Yeo AA, Wong M, Cox DA, Kim HM (2010)
Antiplatelet therapy use and the risk of venous thromboembolic
events in the Raloxifene Use for the Heart (RUTH) trial. J
Womens Health 19:14591465
199. Ensrud K, LaCroix A, Thompson JR et al (2010) Lasofoxifene
and cardiovascular events in postmenopausal women with osteo-
porosis: five-year results from the Postmenopausal Evaluation
and Risk Reduction with Lasofoxifene (PEARL) trial. Circulation
122:17161724
200. Barrett-Connor E, Cauley JA, Kulkarni PM, Sashegyi A, Cox
DA, Geiger MJ (2004) Riskbenefit profile for raloxifene: 4-year
data From the Multiple Outcomes of Raloxifene Evaluation
(MORE) randomized trial. J Bone Miner Res 19:12701275
201. Grady D, Cauley JA, Stock JL, Cox DA, Mitlak BH, Song J,
Cummings SR (2010) Effect of raloxifene on all-cause mortality.
Am J Med 123(469):e461467
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S21
202. Vogel VG, Costantino JP, Wickerham DL et al (2010) Update of
the National Surgical Adjuvant Breast and Bowel Project Study
of Tamoxifen and Raloxifene (STAR) P-2 Trial: preventing breast
cancer. Cancer Prev Res (Phila) 3:696706
203. Martino S, Cauley JA, Barrett-Connor E, Powles TJ, Mershon J,
Disch D, Secrest RJ, Cummings SR (2004) Continuing outcomes
relevant to Evista: breast cancer incidence in postmenopausal
osteoporotic women in a randomized trial of raloxifene. J Natl
Cancer Inst 96:17511761
204. Vogel VG, Qu Y, Wong M, Mitchell B, Mershon JL (2009)
Incidence of invasive breast cancer in postmenopausal women
after discontinuation of long-term raloxifene administration. Clin
Breast Cancer 9:4550
205. Grady D, Cauley JA, Geiger MJ, Kornitzer M, Mosca L, Collins
P, Wenger NK, Song J, Mershon J, Barrett-Connor E (2008)
Reduced incidence of invasive breast cancer with raloxifene
among women at increased coronary risk. J Natl Cancer Inst
100:854861
206. LaCroix AZ, Powles T, Osborne CK et al (2010) Breast cancer
incidence in the randomized PEARL trial of lasofoxifene in
postmenopausal osteoporotic women. J Natl Cancer Inst 102:1706
1715
207. Palacios S, Farias ML, Luebbert H et al (2004) Raloxifene is not
associated with biologically relevant changes in hot flushes in
postmenopausal women for whom therapy is appropriate. Am J
Obstet Gynecol 191:121131
208. Gordon S, Walsh BW, Ciaccia AV, Siddhanti S, Rosen AS,
Plouffe L Jr (2004) Transition from estrogenprogestin to ralox-
ifene in postmenopausal women: effect on vasomotor symptoms.
Obstet Gynecol 103:267273
209. Davis SR, ONeill SM, Eden J, Baber R, Ekangaki A, Stocks JM,
Thiebaud D (2004) Transition from estrogen therapy to raloxifene
in postmenopausal women: effects on treatment satisfaction and
the endometriuma pilot study. Menopause 11:167175
210. Utian W, Yu H, Bobula J, Mirkin S, Olivier S, Pickar JH (2009)
Bazedoxifene/conjugated estrogens and quality of life in post-
menopausal women. Maturitas 63:329335
211. Marie PJ, Felsenberg D, Brandi ML (2010) How strontium rane-
late, via opposite effects on bone resorption and formation, pre-
vents osteoporosis. Osteoporos Int 22:16591667
212. Henrotin Y, Labasse A, Zheng SX, Galais P, Tsouderos Y, Crielaard
JM, Reginster JY (2001) Strontium ranelate increases cartilage
matrix formation. J Bone Miner Res 16:299308
213. Alexandersen P, Karsdal MA, Qvist P, Reginster JY, Christiansen
C (2007) Strontium ranelate reduces the urinary level of cartilage
degradation biomarker CTX-II in postmenopausal women. Bone
40:218222
214. Alexandersen P, Karsdal MA, Byrjalsen I, Christiansen C (2011)
Strontium ranelate effect in postmenopausal women with differ-
ent clinical levels of osteoarthritis. Climacteric 14:236243
215. Bruyere O, Delferriere D, Roux C et al (2008) Effects of stron-
tium ranelate on spinal osteoarthritis progression. Ann Rheum
Dis 67:335339
216. European Medicines Agency (2009) Strontium ranelate. Summa-
ry of product characteristics, 3 June 2010. European Medicines
Agency, London
217. Naess IA, Christiansen SC, Romundstad P, Cannegieter SC,
Rosendaal FR, Hammerstrom J (2007) Incidence and mortality
of venous thrombosis: a population-based study. J Thromb Hae-
most 5:692699
218. Oger E (2000) Incidence of venous thromboembolism: a
community-based study in Western France. EPI-GETBP Study
Group Groupe dEtude de la Thrombose de Bretagne Occidentale
Thromb Haemost 83:657660
219. Silverstein MD, Heit JA, Mohr DN, Petterson TM, OFallon
WM, Melton LJ 3rd (1998) Trends in the incidence of deep vein
thrombosis and pulmonary embolism: a 25-year population-based
study. Arch Intern Med 158:585593
220. Breart G, Cooper C, Meyer O, Speirs C, Deltour N, Reginster JY
(2010) Osteoporosis and venous thromboembolism: a retrospec-
tive cohort study in the UK General Practice Research Database.
Osteoporos Int 21:11811187
221. Osborne V, Layton D, Perrio M, Wilton L, Shakir SA (2010)
Incidence of venous thromboembolism in users of strontium
ranelate: an analysis of data from a prescription-event monitoring
study in England. Drug Saf 33:579591
222. Breart G, Jakob FJ, Palacios S et al (2010) New interim analysis
of a prospective observational cohort study of patients treated
with strontium ranelate. Osteoporos Int S 1:S166
223. Jonville-Bera AP, Crickx B, Aaron L, Hartingh I, Autret-Leca E
(2009) Strontium ranelate-induced DRESS syndrome: first two
case reports. Allergy 64:658659
224. Lee HY, Lie D, Lim KS, Thirumoorthy T, Pang SM (2009)
Strontium ranelate-induced toxic epidermal necrolysis in a patient
with post-menopausal osteoporosis. Osteoporos Int 20:161162
225. Pernicova I, Middleton ET, Aye M (2008) Rash, strontium rane-
late and DRESS syndrome put into perspective. European Med-
icine Agency on the alert Osteoporos Int 19:18111812
226. Musette P, Brandi ML, Cacoub P, Kaufman JM, Rizzoli R,
Reginster JY (2010) Treatment of osteoporosis: recognizing and
managing cutaneous adverse reactions and drug-induced hyper-
sensitivity. Osteoporos Int 21:723732
227. Kong YY, Yoshida H, Sarosi I et al (1999) OPGL is a key
regulator of osteoclastogenesis, lymphocyte development and
lymph-node organogenesis. Nature 397:315323
228. Baudhuin M, Lamoureux F, Duplomb L, Redini F, Heymann D
(2007) RANKL, RANK, osteoprotegerin: key partners of
osteoimmunology and vascular diseases. Cell Mol Life Sci
64:23342350
229. Ferrari-Lacraz S, Ferrari S (2011) Do RANKL inhibitors (deno-
sumab) affect inflammation and immunity? Osteoporos Int
22:435446
230. Sobacchi C, Frattini A, Guerrini MM et al (2007) Osteoclast-poor
human osteopetrosis due to mutations in the gene encoding
RANKL. Nat Genet 39:960962
231. Ashcroft AJ, Cruickshank SM, Croucher PI et al (2003) Colonic
dendritic cells, intestinal inflammation, and T cell-mediated bone
destruction are modulated by recombinant osteoprotegerin. Im-
munity 19:849861
232. Cohen SB, Dore RK, Lane NE, Ory PA, Peterfy CG, Sharp JT,
van der Heijde D, Zhou L, Tsuji W, Newmark R (2008) Denosu-
mab treatment effects on structural damage, bone mineral density,
and bone turnover in rheumatoid arthritis: a twelve-month, mul-
ticenter, randomized, double-blind, placebo-controlled, phase II
clinical trial. Arthritis Rheum 58:12991309
233. Andrews NA (2008) Denosumab and the treatment of rheumatoid
arthritis: in an occupied field, where will a RANKL inhibitor fit
in? Bone Key 5:351356
234. Stolina M, Guo J, Faggioni R, Brown H, Senaldi G (2003)
Regulatory effects of osteoprotegerin on cellular and humoral
immune responses. Clin Immunol 109:347354
235. Miller RE, Branstetter D, Armstrong A, Kennedy B, Jones J,
Cowan L, Bussiere J, Dougall WC (2007) Receptor activator of
NF-kappa B ligand inhibition suppresses bone resorption and
hypercalcemia but does not affect host immune responses to
influenza infection. J Immunol 179:266274
236. McClung MR, Lewiecki EM, Cohen SB et al (2006) Denosumab
in postmenopausal women with low bone mineral density. N Engl
J Med 354:821831
237. Cummings SR, San Martin J, McClung MR et al (2009) Deno-
sumab for prevention of fractures in postmenopausal women with
osteoporosis. N Engl J Med 361:756765
S22 Osteoporos Int (2012) 23 (Suppl 1):S1S23
238. Kendler DL, Roux C, Benhamou CL, Brown JP, Lillestol M,
Siddhanti S, Man HS, San Martin J, Bone HG (2010) Effects of
denosumab on bone mineral density and bone turnover in post-
menopausal women transitioning from alendronate therapy. J
Bone Miner Res 25:7281
239. Brown JP, Prince RL, Deal C et al (2009) Comparison of the
effect of denosumab and alendronate on BMD and biochemical
markers of bone turnover in postmenopausal women with low
bone mass: a randomized, blinded, phase 3 trial. J Bone Miner
Res 24:153161
240. Miller PD, Wagman RB, Peacock M, Lewiecki EM, Bolognese
MA, Weinstein RL, Ding B, San Martin J, McClung MR (2011)
Effect of denosumab on bone mineral density and biochemical
markers of bone turnover: six-year results of a phase 2 clinical
trial. J Clin Endocrinol Metab 96:394402
241. Bucay N, Sarosi I, Dunstan CR et al (1998) Osteoprotegerin-
deficient mice develop early onset osteoporosis and arterial cal-
cification. Genes Dev 12:12601268
242. Ziegler S, Kudlacek S, Luger A, Minar E (2005) Osteoprotegerin
plasma concentrations correlate with severity of peripheral artery
disease. Atherosclerosis 182:175180
243. Mesquita M, Demulder A, Damry N, Melot C, Wittersheim E,
Willems D, Dratwa M, Bergmann P (2009) Plasma osteoprote-
gerin is an independent risk factor for mortality and an early
biomarker of coronary vascular calcification in chronic kidney
disease. Clin Chem Lab Med 47:339346
244. Kobayashi-Sakamoto M, Hirose K, Isogai E, Chiba I (2004) NF-
kappaB-dependent induction of osteoprotegerin by Porphyromo-
nas gingivalis in endothelial cells. Biochem Biophys Res Com-
mun 315:107112
245. Vik A, Mathiesen EB, Noto AT, Sveinbjornsson B, Brox J,
Hansen JB (2007) Serum osteoprotegerin is inversely associated
with carotid plaque echogenicity in humans. Atherosclerosis
191:128134
246. Helas S, Goettsch C, Schoppet M, Zeitz U, Hempel U, Morawietz
H, Kostenuik PJ, Erben RG, Hofbauer LC (2009) Inhibition of
receptor activator of NF-kappaB ligand by denosumab attenuates
vascular calcium deposition in mice. Am J Pathol 175:473478
247. Hodsman AB, Bauer DC, Dempster DW et al (2005) Parathyroid
hormone and teriparatide for the treatment of osteoporosis: a
review of the evidence and suggested guidelines for its use.
Endocr Rev 26:688703
248. Neer RM, Arnaud CD, Zanchetta JR et al (2001) Effect of
parathyroid hormone (134) on fractures and bone mineral den-
sity in postmenopausal women with osteoporosis. N Engl J Med
344:14341441
249. Hodsman AB, Hanley DA, Ettinger MP, Bolognese MA, Fox J,
Metcalfe AJ, Lindsay R (2003) Efficacy and safety of human
parathyroid hormone-(1-84) in increasing bone mineral density in
postmenopausal osteoporosis. J Clin Endocrinol Metab 88:5212
5220
250. Antoniucci DM, Sellmeyer DE, Bilezikian JP, Palermo L, Ensrud
KE, Greenspan SL, Black DM (2007) Elevations in serum and
urinary calcium with parathyroid hormone (1-84) with and without
alendronate for osteoporosis. J Clin Endocrinol Metab 92:942947
251. Winer KK, Sinaii N, Reynolds J, Peterson D, Dowdy K, Cutler
GB Jr (2010) Long-term treatment of 12 children with chronic
hypoparathyroidism: a randomized trial comparing synthetic hu-
man parathyroid hormone 134 versus calcitriol and calcium. J
Clin Endocrinol Metab 95:26802688
252. Winer KK, Yanovski JA, Sarani B, Cutler GB Jr (1998) A
randomized, cross-over trial of once-daily versus twice-daily
parathyroid hormone 134 in treatment of hypoparathyroidism.
J Clin Endocrinol Metab 83:34803486
Osteoporos Int (2012) 23 (Suppl 1):S1S23 S23
... While evidence to date supports decreased matrix IGF-1 as a strong candidate for alcohol-impaired bone healing, we acknowledge that additional undefined factors in bone matrix or in circulation are likely to be important mediators of alcohol's bioactivity. Because multiple classes of systemic factors influence bone, including hormones, chemokines and cytokines (Johnson et al., 2022;Gonzalez-Reimers et al., 2014;Maurel et al., 2012;Mercer et al., 2015), understanding the precise mechanisms will require additional research. ...
Article
Full-text available
Allografts play an important role in treatment of complex bone fractures and deformities. The purpose of this study was to test the hypothesis that alcohol consumption impairs graft incorporation and bone healing by two mechanisms: (1) by lowering osteoinductive capacity and (2) by suppressing bone formation. We performed experiments using a demineralized allogeneic bone matrix (DBM) model in which DBM harvested from donor rats fed control or ethanol diet was implanted subcutaneously into recipient rats fed control or ethanol diet. We also evaluated the efficacy of intermittent parathyroid hormone (PTH) on bone graft incorporation (DBM from donor rats fed alcohol or control diet) using a critical size defect model. Bone formed during osteoinduction was measured by micro-computed tomography. Experiment 1: Bone volume was lower in DBM harvested from ethanol-consuming donors 6 weeks following implantation into recipients fed control diet, indicating that exposure of the donor rats to ethanol lowered osteoinductive capacity. Experiment 2: Bone volume was lower in DBM harvested 3 weeks following implantation from ethanol-consuming donors into ethanol-consuming recipients compared to DBM harvested from control donors implanted into control recipients or DBM harvested from control donors implanted into ethanol-consuming recipients. Experiment 3: Ethanol consumption by donors resulted in a tendency for lower DBM bone volume (p = 0.085) whereas PTH treatment resulted in higher DBM bone volume in the critical size defect model. Our results suggest that chronic heavy alcohol consumption by allograft donors may impair osteoinduction and this negative outcome may be worsened by alcohol intake during bone healing. Additionally, PTH has the potential to increase osteoinduction in DBM harvested from both abstinent and alcohol-consuming donors.
... 1,2 One of the causes of chronic hypophosphatemia in adults is the excess action of fibroblast growth factor 23 (FGF23), which is the key regulator of phosphate homeostasis. FGF23 promotes excess phosphate excretion into the urine by downregulating the expression of sodium-phosphate cotransporters (NaPi2a/NaPi2c) in the proximal tubules and reduces serum 1,25-dihydroxyvitamin D. 3,4 Although chronic alcoholism is known to cause hypophosphatemia and bone fragility through mechanisms that are mainly related to vitamin D deficiency and damage to the renal tubules, 5,6 we recently reported 2 alcoholic patients with hypophosphatemic osteomalacia, which was obviously induced by FGF23 oversecretion, and the condition was remedied by abstinence. 7 Although FGF23 (in a physiological state) is specifically secreted from mature osteocytes or osteoblasts in bone, the source of excess FGF23 in these alcoholic patients is uncertain. ...
Article
Full-text available
Excess fibroblast growth factor 23 (FGF23), a mature osteocyte-derived phosphaturic hormone, causes chronic hypophosphatemic osteomalacia in adults. This rare condition was recently reported in 2 alcoholic patients, with marked improvement upon cessation of alcohol consumption, suggesting a link between alcohol and FGF23-related hypophosphatemia within the highly limited cases. This study aimed to investigate whether the source of excess FGF23 in alcohol-induced FGF23-related hypophosphatemic osteomalacia is the bone or the other organs. To achieve this goal, an immunohistochemical approach for the bone obtained from a patient was employed. Initial attempts at quantifying FGF23 in the bone using conventional immunohistochemistry (IHC) faced issues in quantifiability and sensitivity for low FGF23 expression levels. Therefore, next-generation IHC with phosphor-integrated dots (PIDs) was applied, which enabled the quantification of FGF23 expression in the bone across a broad range. Preliminary analyses using IHC with PIDs on normal bone samples (n = 12) provided a reference level (154.5 PID particles per cell). IHC with PIDs quantified suppressed physiological FGF23 expression in the bone samples from 3 patients with tumor-induced osteomalacia, where FGF23 is oversecreted from a tumor (13.6 PID particles per cell). Subsequently, bone samples obtained from a 70-year-old male with alcohol-induced FGF23-related hypophosphatemic osteomalacia were analyzed, showing a higher number of PID particles per cell (199.4 PID particles per cell) than the reference level. This study suggests that orthotopic, bone-derived FGF23 is implicated in alcohol-induced FGF23-related hypophosphatemic osteomalacia. Furthermore, the study also demonstrated that highly sensitive IHC with PIDs could aid in the differential diagnosis of FGF23-related hypophosphatemia of unknown origin. Specifically, a bone sample with a low number of PID particles per cell indicates an excess ectopic secretion of FGF23; a bone sample with a normal to high number of PID particles per cell indicates an excess orthotopic secretion of FGF23.
... A potential explanation was that people with light alcohol consumption were included as having drinking history along with heavy drinkers. Although heavy alcohol consumption (>50 g/day) is toxic to bones, light alcohol consumption (0-12.5 g/day) might actually help increase BMD in the hip and other weight-bearing locations, promote calcitonin release, and decrease bone resorption [58][59][60]. These effects are beneficial for maintaining physical skills and activities, and hence might result in a lower risk of falls and HF. ...
Article
Full-text available
Background Hip fracture (HF) is one of the most prevalent orthopedic conditions among the elderly, with falls being the primary risk factor for HF. With the surge of aged population, China is facing great challenges from HF and falls. However, a comprehensive long-term observation of risk factors affecting HF and falls and their association are little reported at a national level. Methods The longitudinal cohort was established using the China Health and Retirement Longitudinal Study (CHARLS) data from 2011 to 2018. The incidence density and multi-risk-stratified lifetime risk (up to 90 years of age) of falls and HF were studied at index ages of 50, 60, and 70, as well as the lifetime risk stratified by six regions in China, based on the modified Kaplan–Meier method with Statistical Analysis System (SAS). Results This study identified 17 705 subjects aged 50–89. The incidence density of falls was 65.07 and 47.53 per 1000 person-years in women and men, respectively. The incidence density of HF was also higher in women at 5.58 per 1000 person-years than in men at 4.88. By age 50, the lifetime risk of experiencing a HF was 18.58 % for women and 13.72 % for men. Vision and hearing abilities were significantly related to the lifetime risk of both falls and HF. Obesity-related factors presented age-relevant relationships with lifelong risks. Lack of naps, poor lower limb strength, and physical capabilities were indicative of HF risk. The north-western region of China had the lowest lifetime risk of falls but highest risk of HF, while other regions showed a consistent trend between falls and HF. Conclusion The aging population worldwide faces a considerable risk of falls and HF. Several risk factors were identified in this study using a Chinese population, relating to disease history, lifestyle habits, health status and physical function, and the risks differed among six regions in China. Future precautionary management programs, as well as patient self-awareness are necessary for improving the prevention of falls and HF to reduce their incidence in the aging population. The translational potential of this article With the greatest aged population worldwide, China faces the unparalleled challenge on public health. The study poses the lifetime risk of hip fracture and falls stratified by multiple risk factors in people from 45 to 90 in a national scale, which would shed a light on the early and continuous prevention of such injury.
... Despite extensive research, the precise pathological mechanisms underlying alcohol-induced osteoporosis remain inadequately understood. Prior studies have developed an animal model of alcohol-induced osteoporosis by administering ethanol intraperitoneally at a dosage of 3 g/kg [40,41] which enable researchers to closely study the effects of alcohol on bone health and potential therapeutic interventions in a controlled setting. Therefore, we based our study on the animal model mentioned. ...
Article
Full-text available
Background: Alcohol-induced osteoporosis is a significant health concern, impairing bone formation and enhancing resorption, thereby weakening skeletal integrity. This study examines the effects of palm vitamin E on bone histomorphometry in a male rat model of alcohol-induced osteoporosis. Methods: Three-month-old Sprague–Dawley rats were randomized into seven groups, with one baseline control group (BC) and six experimental groups undergoing a two-phase treatment. In the first month, the control group received normal saline, while experimental groups received intraperitoneal alcohol (3 g/kg) three times weekly. For the subsequent two months, alcohol treatment continued in one group (A), while others received olive oil (C), saline (AN), alpha-tocopherol (AA), or palm vitamin E (AE) orally. Results: Femur histomorphometric analysis post-sacrifice showed that alcohol exposure significantly decreased osteoblastic activity and impaired bone microarchitecture, evidenced by reduced Ob.S/BS, OS/BS, OV/BV, Tb.Th, BV/TV, and Tb.N, alongside increased Oc.S/BS, ES/BS, and Tb.Sp. Both alpha-tocopherol and palm vitamin E improved bone parameters, with palm vitamin E showing superior efficacy except in OV/BV. Conclusions: These findings suggest that palm vitamin E may offer a therapeutic benefit for mitigating alcohol-induced bone damage.
... Tobacco reduces bone density, causing a loss in bone quality and increasing the number of fracture cases [170]. Regarding alcohol, it has been shown that continued consumption of large amounts of alcohol negatively affects bone mass since there is an overactivation of osteoclasts with the consequent loss of mineralized matrix [171]. As previously mentioned, the mechanical load that allows for better bone regulation, and that can be achieved through movement or external load, positions exercise as a powerful preventive and management tool during the treatment of osteoporosis [172]. ...
Article
Full-text available
Osteoporosis is a chronic disease that is characterized by a loss of bone density, which mainly affects the microstructure of the bones due to a decrease in bone mass, thereby making them more fragile and susceptible to fractures. Osteoporosis is currently considered one of the pandemics of the 21st century, affecting around 200 million people. Its most serious consequence is an increased risk of bone fractures, thus making osteoporosis a major cause of disability and even premature death in the elderly. In this review, we discuss its causes, the biochemical mechanisms of bone regeneration, risk factors, pharmacological treatments, prevention and the effects of diet, focusing in this case on compounds present in a diet that could have palliative and preventive effects and could be used as concomitant treatments to drugs, which are and should always be the first option. It should be noted as a concluding remark that non-pharmacological treatments such as diet and exercise have, or should have, a relevant role in supporting pharmacology, which is the recommended prescription today, but we cannot ignore that they can have a great relevance in the treatment of this disease.
Article
Osteoporosis, a metabolic disorder distinguished by decreased bone density and degradation of bone tissue microarchitecture, is a silent disease that evolves without any clinical symptoms or signs. An individual may not be aware of osteoporosis until a fracture occurs. The lifetime risk of osteoporosis is estimated to be between 10 and 20%. The disease can have intrinsic causes, like genetic predisposition, aging, and lack of sex hormones. However, it can also occur secondary to calcium and vitamin D deficiencies due to the influence of various factors. With a global increase in osteoporotic fractures, there is a need for macro-level and micro-level interventions to prevent and treat osteoporosis and its complications. This review highlights the crucial role of various biomolecules in diagnosing and managing osteoporosis and emphasizes the importance of further research in the field.
Preprint
Full-text available
Chronic alcohol consumption (CAC) is associated with an enhanced risk of bone fracture, reduced bone density, and osteoporosis. We have previously shown using a rhesus macaque model of voluntary ethanol consumption that CAC induces functional, transcriptomic, and epigenomic changes in hematopoietic stem and progenitor cells (HSPCs) and their resultant monocytes/macrophages, skewing them towards a hyper-inflammatory response. Here, we extended those studies and investigated alterations in osteoclasts, which, in postnatal life, are differentiated from HSPCs and play a critical role in maintaining bone homeostasis. Analysis using spectral flow cytometry revealed a skewing of HSPCs towards granulocyte-monocyte progenitors (GMPs) with the CAC group that was in concordance with an increased number of colony-forming unit-granulocyte/macrophage (CFU-GM). Additionally, HSPCs from animals in the CAC group incubated with M-CSF and RANKL were more likely to differentiate into osteoclasts, as evidenced by increased Tartrate-Resistant Acid Phosphatase (TRAP) staining and bone resorption activity. Moreover, single-cell RNA sequencing of differentiated HSPCs identified three clusters of osteoclast precursors in the CAC group with enhanced gene expression in pathways associated with cellular response to stimuli, membrane trafficking, and vesicle-mediated transport. Collectively, these data show that CAC-derived hematopoietic progenitor cells exhibit a higher capacity to differentiate into osteoclast precursors. These findings provide critical insights for future research on the mechanisms by which CAC disrupts monopoiesis homeostasis and enhances osteoclast precursors, thereby contributing to reduced bone density.
Article
The differentiation of adipocytic and osteogenic cells has been investigated in cultures of adult rat marrow stromal cells. Adipocytic differentiation was assessed using morphological criteria, changes in expression of procollagen mRNAs, consistent with a switch from the synthesis of predominantly fibrillar (types I and III) to basement membrane (type IV) collagen, and the induction of expression of aP2, a specific marker for differentiation of adipocytes. Osteogenic differentiation was assessed on the basis of changes in the abundance of the mRNAs for the bone/liver/kidney isozyme of alkaline phosphatase and the induction of mRNAs for bone sialoprotein and osteocalcin. In the presence of foetal calf serum and dexamethasone (10(−8) M) there was always differentiation of both adipocytic and osteogenic cells. When the steroid was present throughout primary and secondary culture the differentiation of osteogenic cells predominated. Conversely, when dexamethasone was present in secondary culture only, the differentiation of adipocytes predominated. When marrow stromal cells were cultured in the presence of dexamethasone in primary culture and dexamethasone and 1,25-dihydroxyvitamin D3 (1,25(OH)2D3; 10(−8) M) in secondary culture, the differentiation of adipocytes was inhibited whereas the differentiation of osteogenic cells was enhanced, as assessed by an increase in expression of osteocalcin mRNA. The results, therefore, demonstrate an inverse relationship between the differentiation of adipocytic and osteogenic cells in this culture system and are consistent with the possibility that the regulation of adipogenesis and osteogenesis can occur at the level of a common precursor in vivo.
Article
Background: Published reports on the effect of alcohol consumption on bone mineral density (BMD) are inconsistent. Objective: The objective of this study was to examine the relation between alcohol intake and BMD, calcitropic hormones, calcium absorption, and other biochemical indexes of bone and mineral metabolism in elderly women. Design: The results presented are derived from baseline observations of 489 elderly women (aged 65–77 y) recruited for an osteoporosis study. The nondrinking group comprised 297 women and the drinking group comprised 148 women. Furthermore, the effect of different alcohol intakes (≤28.6, >28.6 to ≤57.2, >57.2 to ≤142.9, and >142.9 g/wk) was studied. Results: Women who consumed alcohol had significantly higher spine (10%), total body (4.5%), and midradius (6%) BMD than did nondrinkers. An alcohol intake >28.6 g/wk was associated with higher BMD; maximum effect was seen with an intake of >28.6 to ≤57.2 g/wk (16%, 12%, and 14% increase in spine, total body, and midradius BMD, respectively). There was a marked reduction in bone remodeling markers, serum osteocalcin, and the ratio of urinary cross-linked N-telopeptides of type 1 collagen to creatinine with alcohol consumption, suggesting that increased BMD with alcohol consumption could be due to reduced bone remodeling. Further, serum parathyroid hormone concentrations were significantly lower in alcohol drinkers than in nondrinkers and could be one of the causes of decreased bone resorption. Conclusions: Moderate alcohol intake was associated with higher BMD in postmenopausal elderly women. The protective effect of alcohol may have been a result of lower bone remodeling due to reduced parathyroid hormone concentrations or factors such as increased estrogen concentrations.
Article
371 Background: Bisphosphonates are commonly used for the treatment of osteoporosis and of bone metastases due to breast cancer and were recently reported to be associated with reduced risk of breast cancer, but their association with risk of other cancers is unknown. Methods: The Molecular Epidemiology of Colorectal Cancer (MECC) study is a population-based case-control study in northern Israel of colorectal cancer cases and age/sex/clinic/ethnic-group matched controls. Use of bisphosphonates prior to diagnosis was assessed in a subset of 933 pairs of post-menopausal female cases and controls, enrolled in Clalit Health Services (CHS), using computerized pharmacy records. Results: The use of bisphosphonates for more than one year prior to diagnosis, but not for less than a year, was associated with a significantly reduced relative risk of colorectal cancer (odds ratio=0.50, 95% CI: 0.35-0.71). This association remained statistically significant after adjustment for, vegetable consumption, sports activity, family history of colorectal cancer, , BMI, use of low-dose aspirin, statins, vitamin D and post-menopausal hormones (OR=0.40, 0.24-0.64). Concomitant use of bisphosphonates and statins did not further reduce the risk. Conclusions: The use of oral bisphosphonates for more than 1 year was associated with a 60% relative reduction in the risk of colorectal cancer similar to the recently reported association of this drug class with reduction in breast cancer risk. No significant financial relationships to disclose.
Article
Tolerability of different dosing regimens of bisphosphonates for the treatment of osteoporosis and malignant bone disease
Article
Tolerability of different dosing regimens of bisphosphonates for the treatment of osteoporosis and malignant bone disease
Article
Previous studies have demonstrated that lasofoxifene, a nonsteroidal selective estrogen-receptor modulator, decreases bone resorption, bone loss, and low-density lipoprotein cholesterol in postmenopausal women. Its effects of the risk of fractures, breast cancer, and cardiovascular disease are unclear. The postmenopausal evaluation and risk-reduction with lasofoxifene trial was an international, randomized, placebo-controlled trial that investigated the effects of lasofoxifene on the risk of fractures, estrogen receptor (ER)-positive breast cancer, and cardiovascular disease in a population of postmenopausal women with osteoporosis. The study subjects were a population of women between the ages of 59 and 80 years who had a bone mineral density T score of -2.5 or less at the femoral neck or spine. Participants were randomized to receive once-daily lasofoxifene at a dose of either 0.25 mg (low-dose, n = 2852) or 0.5 mg (high-dose, n = 2852) or placebo (n = 2852, control group) for 5 years. The trial was conducted at 113 sites in 32 countries. Vertebral and nonvertebral fractures and ER-positive breast cancer were the primary study end points. Major coronary heart disease events and stroke were the secondary endpoints. Compared with placebo, treatment with the high-dose lasofoxifene was associated with a reduction in the risk of vertebral fractures (13.1 cases vs. 22.4 cases per 1000 person-years; hazard ratio [HR], 0.58; 95% confidence interval [CI], 0.47-0.70), nonvertebral fractures (18.7 vs. 24.5 cases per 1000 person-years; HR, 0.76; 95% CI, 0.64-0.91), ER-positive breast cancer (0.3 vs. 1.7 cases per 1000 person-years; HR, 0.19; 95% CI, 0.07-0.56), major coronary heart disease events (5.1 vs. 7.5 cases per 1000 person-years; HR, 0.68; 95% CI, 0.50―0.93), and stroke (2.5 vs. 3.9 cases per 1000 person-years; HR, 0.64; 95% CI, 0.41-0.99). At 5 years compared to the placebo group, postmenopausal women receiving low-dose lasofoxifene showed reduced rates of vertebral fractures (16.0 vs. 22.4 per 1000 person-years; HR, 0.69; 95% CI, 0.57-0.83) and stroke (2.4 vs. 3.9 cases per 1000 person-years; HR, 0.61; 95% CI, 0.39-0.96). Both drug doses were associated with increased risk of a venous thromboembolic event: Compared to the placebo which had 1.4 venous thromboembolic events per 1000 person-years, the low-dose group had 3.8 events per 1000 person-years (HR, 2.67; 95% CI, 1.55-4.58) and the high-dose group had 2.9 events per 1000 person-years (HR, 2.06; 95% CI, 1.17- 3.61). No increased risk of endometrial hyperplasia or endometrial cancer was observed at 5 years with either dose. Deaths per 1000 person-years were 7.0 and 5.7 for low dose and high dose, respectively, compared to 5.1 for the placebo (P = ns for both comparisons). These findings show that treatment of postmenopausal women with osteoporosis using a daily dose of lasofoxifene of 0.5 mg is associated with reduced risks of vertebral and nonvertebral fractures, ER-positive breast cancer, major coronary heart disease, and stroke, and an increased risk of thromboembolic events.