Risk Factors and Drug Interactions
Predisposing to Statin-Induced Myopathy
Implications for Risk Assessment, Prevention and Treatment
Yiannis S. Chatzizisis,
1,2
Konstantinos C. Koskinas,
2
Gesthimani Misirli,
1
Christos Vaklavas,
3
Apostolos Hatzitolios
4
and George D. Giannoglou
1
1 1st Cardiology Department, AHEPA University Hospital, Aristotle University Medical School,
Thessaloniki, Greece
2 Cardiovascular Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
3 Department of Internal Medicine, University of Texas Medical School at Houston, Houston, TX, USA
4 1st Propedeutic Department of Internal Medicine, AHEPA University Hospital, Aristotle University
Medical School, Thessaloniki, Greece
Contents
Abstract. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 171
1. Definition and Epidemiology of Statin-Induced Myopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173
2. Comparison between Statins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 173
3. Mechanisms of Statin-Induced Myopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174
4. Physicochemical and Pharmacokinetic Properties of Statins . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 174
5. Risk Factors that Precipitate Statin-Induced Myopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
5.1 Patient Characteristics. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
5.1.1 Demographic Characteristics . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 175
5.1.2 Genetic Factors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 176
5.1.3 Co-Morbidities . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
5.2 Statin Properties . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
5.2.1 Dose-Dependent Effects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 177
5.2.2 Physicochemical Properties of Statins: Lipophilicity versus Hydrophilicity. . . . . . . . . . . . . . . 178
5.3 Statin-Drug Interactions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 178
5.3.1 Interactions with Non-Hypolipidaemic Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 179
5.3.2 Interactions with Other Hypolipidaemic Agents . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 180
6. Recommendations for the Prevention and Management of Statin-Induced Myopathy . . . . . . . . . . 181
6.1 Prevention of Statin-Induced Myopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181
6.2 Management of Statin-Induced Myopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 181
7. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 182
Abstract HMG-CoA reductase inhibitors (‘statins’) represent the most effective and
widely prescribed drugs currently available for the reduction of low-density
lipoprotein cholesterol, a critical therapeutic target for primary and second-
ary prevention of cardiovascular atherosclerotic disease. In the face of the
established lipid lowering and the emerging pleiotropic properties of statins,
the patient population suitable for long-term statin treatment is expected to
further expand. An overall positive safety and tolerability profile of statins
REVIEW ARTICLE
Drug Saf 2010; 33 (3): 171-187
0114-5916/10/0003-0171/$49.95/0
ª 2010 Adis Data Information BV. All rights reserved.
has been established, although adverse events have been reported. Skeletal
muscle-related events are the most common adverse events of statin treat-
ment. Statin-induced myopathy can (rarely) manifest with severe and po-
tentially fatal cases of rhabdomyolysis, thus rendering the identification of
the underlying predisposing factors critical.
The purpose of this review is to summarize the factors that increase the
risk of statin-related myopathy. Data from published clinical trials, meta-
analyses, postmarketing studies, spontaneous report systems and case reports
for rare effects were reviewed. Briefly, the epidemiology, clinical spectrum
and molecular mechanisms of statin-associated myopathy are discussed. We
further analyse in detail the risk factors that precipitate or increase the like-
lihood of statin-related myopathy. Individual demographic features, genetic
factors and co-morbidities that may account for the significant inter-
individual variability in the myopathic risk are presented. Physicochemical
properties of statins have been implicated in the differential risk of currently
marketed statins. Pharmacokinetic interactions with concomitant medica-
tions that interfere with statin metabolism and alter their systemic bioavail-
ability are reviewed. Of particular clinical interest in cases of resistant
dyslipidaemia is the interaction of statins with other classes of lipid-lowering
agents; current data on the relative safety of available combinations are
summarized. Finally, we provide an update of current guidelines for the
prevention and management of statin myopathy.
The identification of patients with an increased proclivity to statin-
induced myopathy could allow more cost-effective approaches of monitoring
and screening, facilitate targeted prevention of potential complications, and
further improve the already overwhelmingly positive benefit-risk ratio of
statins.
Atherosclerotic cardiovascular disease is the
most frequent cause of morbidity and mortality
in developed countries. Low-density lipoprotein
cholesterol (LDL-C) reduction attenuates the pro-
gression of atherosclerosis and reduces the risk
of cardiovascular events. Among hypolipidae-
mic medications, HMG-CoA reductase inhibitors
(‘statins’) have unequivocally revolutionized both
primary and secondary prevention of cardiovas-
cular disease, due to their lipid-lowering potential
and pleiotropic effects that beneficially affect
atherosclerotic plaque stability.
[1]
Statins are com-
petitive inhibitors of HMG-CoA reductase, the
rate-limiting enzyme in cholesterol biosynthesis
that converts HMG-CoA into mevalonate. They
lower plasma LDL-C through intracellular cho-
lesterol depletion and upregulation of the expres-
sion of LDL receptors in hepatocytes.
[2]
The
number of patients receiving statins, often in
combination with other classes of lipid-lowering
agents, has expanded with the implementation of
more aggressive goals for LDL-C lowering,
[3]
while the additional benefit attributed to intensive
statin therapy has resulted in higher dose statin
regimens.
[4]
Accumulating evidence from controlled trials
and clinical experience demonstrates that statins
are well tolerated medicines with a good safety
profile.
[5-8]
The major and most common com-
plication to their use is a variety of skeletal
muscle-related events, which represent a clinically
important cause of statin intolerance and dis-
continuation. Statins confer a small but definite
risk of myopathy, a dose-dependent adverse ef-
fect associated with all statins (cla ss effect).
[9]
Muscular adverse effects are usuall y mild and
reversible; however, these adverse effects may be
a prelude to rhabdomyolysis, a very rare but po-
tentially serious and even life-threatening clinical
condition. The association of statins with cases of
172 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
severe myopathic events may have resulted in
excessive safety concerns for this revolutionary
class of medications.
[10]
Notwithstanding the
overall good safety profile, knowledge of the un-
derlying mechanisms and risk factors is required
for prompt identification and proper manage-
ment of adverse muscle events. The purpose of
this review is to investigate the risk factors that
precipitate statin-induced muscle adverse events,
and also to summarize current guidelines on the
administration of statins with regard to their
potential myotoxicity.
1. Definition and Epidemiology of
Statin-Induced Myopathy
In the present revie w the term ‘myopathy’ will
be used as a general term to describe all skeletal
muscle-related problems.
[11]
Several, often con-
troversial, terms have been used to describe the
clinical manifestations and laboratory findings of
statin-induced myopathy. The spectrum of myo-
pathy includes asymptomatic increase of creatine
kinase (CK), myalgia, myositis and rhabdomyo-
lysis, as summarized in table I. Rhabdomyolysis
represents the least frequent, though potentially
fatal, complication caused by skeletal muscl e
breakdown, which leads to the release of toxic
intracellular constituents into the blood circula-
tion and eventually causes acute renal failure.
The lack of consensus in the definition of
statin-induced muscle events hinders the precise
estimation of their true incidence. Because patients
with a considered high susceptibility to statin
toxicity are generally excluded from clinical trials
of statins, reported adverse event rates from con-
trolled trials may underestimate the true rate of
these adverse effects in an unselected patient po-
pulation. Complaints of muscle symptoms occur in
1.53.0% of clinical trial participants, while rates
widely range between 0.3% and 33% in routine
practice.
[12]
No conclusive evidence supports in-
creased myalgia associated with standard statin
doses,
[13]
although this has been reported in pa-
tients receiving higher doses.
[14]
The overall excess
risk of myopathy attributed to standard statin do-
ses is typically <0.01%.
[13]
Accordingtodatafrom
randomized clinical trials and cohort studies, the
incidence of myopathy is estimated at 5 patients
per 100 000 person-years and rhabdomyolysis at
1.6 patients per 100 000 person-years,
[15]
whereas
reporting rates in the US FDA Adverse Event
Reporting System datab ase (AERS) are 0.32.2
cases of myopathy and 0.313.5 cases of rhabdo-
myolysis per 1 000000 statin prescriptions.
[16]
A
recent, large-scale trial evaluating rosuvastatatin
20 mg daily reported comparable rates of myo-
pathy between recipients of drug and placebo
(0.1%).
[17]
Regarding higher dose statin regimens,
increased risk of myopathy has been reported for
simvastatin 80 mg daily
[18]
but not for higher doses
of atorvastatin (80 mg).
[4,19,20]
2. Comparison between Statins
Reports associating the use of all marketed
statins with the entire spectrum of myopathy
suggest that this is a class effect.
[21]
The safety
profiles of different statins at standard doses
seem comparable but not identical, as indicated
by the significantly greater risk and subsequent
withdrawal of cerivastatin,
[22,23]
while excess risk
seems to be clearly related to increased doses. On
the basis of current evidence, and in the absence
of randomized trials directly comparing the risk
of each available statin in comparable doses and
with standard definitions of myopathic events, no
definite conclusions on the relative myopathic
potential conferred by each of the currently
marketed statins can be drawn.
[21]
An overall
Table I. The clinical spectrum of statin-induced myopathy
[11]
Condition Definition
Myopathy General term to describe all skeletal
muscle-related adverse effects
Asymptomatic CK elevation CK elevation without muscle
symptoms
Myalgia Muscle pain or weakness without
CK elevation
Myositis Muscle symptoms with CK
elevation typically <10 · ULN
Rhabdomyolysis Muscle symptoms with CK
elevation typically >10 · ULN, and
with creatinine elevation (usually
with brown urine and urinary
myoglobin)
CK = creatine kinase; ULN = upper limit of normal.
Statin-Induced Myopathy: Risk Factors and Drug Interactions 173
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
higher risk of rhabdomyolysis has been asso-
ciated with simvastatin 80 mg, whereas the lowest
incidence apparently occurs with fluvastatin and
pravastatin, presumably associated with their
weaker HMG-CoA reductase inhibitor capa-
city.
[12,15,24,25]
According to a meta-analysis
of trials comparing standard doses of all cur-
rently marketed statins except rosuvastatin,
atorvastatin was associated with the relatively
highest risk and fluva statin with the lowest risk of
adverse events in general, and muscular events in
particular.
[26]
A recently published meta-analysis
of randomized controlled trials comparing differ-
ent doses of atorvastatin (1080 mg) and rosu-
vastatin (540 mg) revealed no significant difference
in adverse muscul ar events between these two
statins at any dose ratio.
[27]
3. Mechanisms of Statin-Induced
Myopathy
The pathogenetic mechanisms of statin-induced
myopathy have been thoroughly reviewed by
Vaklavas et al.
[28]
and are presented briefly in this
review. The interruption of the HMG-CoA re-
ductase biosynthetic pathway and the consequent
intracellular depletion of downstream intermediate
metabolites (i.e. isopentenylated proteins geranyl
pyrophosphatase and farnesyl pyrophosphatase)
and end products (i.e. cholesterol, dolichols, ubi-
quinone) are considered the cornerstone of the
myotoxic effects of statins. Reduction of pre-
nylated proteins can result in dysprenylation of
proteins, including lamins and small guanosine
triphosphatases, thereby causing an imbalance in
the intracellular signalling cascades and enhancing
apoptosis. Sarcolemmal cholesterol deficiency, as a
result of the dynamic equilibrium between mem-
brane and plasma lipids, may adversely modify
membrane physical properties, integrity and fluid-
ity, thus resulting in membrane destabilization.
[29]
Inhibition of dolichol synthesis has been impli-
cated in defective N-linked glycosylation of plasma
membrane proteins and impaired response to
growth factors.
[28]
Ubiquinone or coenzyme Q10 (CoQ10) is a re-
cognized constituent of oxidative phosphorylation
and adenosine triphosphate production in mito-
chondria required to maintain cell integrity.
[30]
Consistently, the decreased CoQ10 biosynthesis
and thus energy depletion mediated by statins has
been postulated to account for the potential myo-
toxicity of statins. Statin-mediated reduction of
circulating, but not intramuscular,
[31]
CoQ10 levels
has been reported, while no direct association
between decreased intramuscular CoQ10 levels
and mitochondrial myopathy has been established.
Accordingly, CoQ10 deficiency may represent
a predisposing rather than etiopathogenic factor
of statin mediated myopathy, possibly in a syner-
gistic manner with coexisting CoQ10-depleting
conditions.
[32]
The equilibrium between intramuscular statin
transport and efflux may be a critical regulator
of intramuscular drug concentration and con se-
quently the risk of myopathy. Organic anion
transporting polypeptide (OATP) 2B1, a re-
cognized hepatic uptake transporter for statins,
has also been identified in skeletal myofibres,
[33]
and the OATP inhibitor estrone sulphate pro-
tected the skeletal myofibres against pravastatin-
and fluvastatin-induced toxicity. Furthermore,
isoforms -1, -4 and -5 of the multidrug resistance-
associated protein (MRP), a well characterized
statin efflux transporter, are highly expressed in
skeletal muscle, and the inhibition of MRP with
probenecid precipitates skeletal muscle toxicity in
rats treated with rosuvastatin,
[34]
implying that
MRP-1 may be involved in statin efflux at the
myocyte level.
4. Physicochemical and
Pharmacokinetic Properties of Statins
The physicochemical propert ies of statins,
which determine their bioavailability and thereby
affect the risk of myopathy, are summarized in
table II. Water solubility affects statin perme-
ability through cellular membranes of non-hepa-
tic (including muscular) cells and their ability to
cross the blood-brain barrier. Pravastatin, rosu-
vastatin and to some extent fluvastatin exhibit
hydrophilic properties, as opposed to the lipo-
philicity of the other statin molecules (i.e. ator-
vastatin, simvastatin and lovastatin).
[35]
174 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
The hepatic cytochrome P450 enzyme (CYP)
system is responsible for the metabolism of many
drugs, including statins to some extent with the
exception of pravastatin.
[36]
Lovastatin, simvas-
tatin and, to a lesser extent, atorvastatin are
metabolized by the CYP3A4 isozyme. Coadminis-
tration of the previously mentioned statins with
medications or food that either inhibit or are
substrates of CYP3A4 decreases the statins’ first-
pass metabolism, thereby resulting in increased
bioavailability.
[37]
Fluvastatin is mainly meta-
bolized by CYP2C9, and to a much lesser
extent by CYP3A4 and CYP2C8, and conse-
quently does not interact with CYP3A4 inhibi-
tors. Pravastatin is metabolized through several
pathways, including isomerization, sulfation,
glutathione conjugation and oxidation, and only
to a small extent (1%) by the CYP enzyme system;
it is the only statin with a significant renal excre-
tion (approximately 60% of the absorbed quan-
tity), in keeping with its hydrophilic nature.
[38]
This theoretically renders it safer as far as its
potential drug interactions are concerned.
[9]
Ro-
suvastatin undergoes minimal metabolism via the
CYP2C9 isoenzyme,
[39]
while 90% is eliminated
as the parent compound in the faeces.
The systematic bioavailability of statins is
quite low. All statins present a high affinity
with blood proteins (95%), except pravastatin
(approximately 50%). Atorvastatin and rosuvas-
tatin are the two statins with longer half-lives
(1316 hours) and this property is most probably
linked to their higher lipid-lowering efficacy.
5. Risk Factors that Precipitate
Statin-Induced Myopathy
When administering statins, physicians should
take into consideration a series of factors that
potentially increase the risk of myopathic events.
As summarized in figure 1, a constellation of
factors are associated with the risk of statin-
associated myopathy development, including
(i) patient characteristics (demographic character-
istics, co-morbidit ies, genetic factors); (ii) drug
properties (specific statin molecule, dose, phar-
macokinetic properties); and (iii) concomitant
interacting medications. Systemic exposure
is considered to play a pivotal role in statin-
associated myopathy, and risk factors that enhance
the respective risk may do so, at least partly, by
increasing either statin systemic bioavailability or
the sensitivity to increased statin blood levels.
5.1 Patient Characteristics
5.1.1 Demographic Characteristics
Certain demographic characteristics have
been associated with an increased risk of statin-
induced myopathy. It has been observed
epidemiologically that advanced age (particularly
>80 years), female sex, small body frame and
frailty increase the myopathic effect of
Table II. Physicochemical and pharmacokinetic properties of statins
Characteristic Lovastastin Simvastatin Pravastatin Fluvastatin Atorvastatin Rosuvastatin
Daily dosage (mg) 2080 1080 2080 4080 1080 1040
Origin Fungi Semisynthetic Fungi Synthetic Synthetic Synthetic
Prodrug Yes Yes No No No No
Solubility Lipophilic Lipophilic Hydrophilic Intermediate Lipophilic Hydrophilic
CNS permeation Yes Yes No No No No
Effect of food intake on
absorption
Increased
absorption
None Decreased
absorption
None None None
First-pass metabolism CYP3A4 CYP3A4 Multiple ways CYP2C9 CYP3A4 Limited CYP2C9
Protein binding (%)9595 50989090
Half-life (hours) 2323120.521316 19
Hepatic excretion (%)69 79 46 >68 Not available 63
Renal excretion (%)30 13 60 <6 <210
CYP = cytochrome P450 enzyme.
Statin-Induced Myopathy: Risk Factors and Drug Interactions 175
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
statins.
[11,13,40]
Myopathic symptoms may be
hard to differentiate from muscular complaints
commonly experienced in elderly patients. Poly-
pharmacy and age-related impairment of renal
function may in part account for the increased
risk of myopathy among elderly individuals. A
greater risk has been attributed to Chinese or
Japanese descent, although this concept is
inadequately supported by current evidence. Typi-
cally, Asians achieve similar benefits to Cauca-
sians at lower statin doses. Plasma levels of
rosuvastatin in particular have been shown to be
2-fold higher in Asian than in Caucasian in-
dividuals receiving similar rosuvastatin doses.
[41]
The smaller body mass index in Asians has been
postulated as the underlying cause of the differ-
ences in drug response in some
[42,43]
but not all
comparable studies.
[44,45]
Genetic differences in
statin metabolism involving the CYP450 enzymes
and the OATPs are more likely to account for the
heightened response to statins in Asians.
[46-48]
Although the increased systemic bioavailability
achieved with similar statin doses
[41]
has not been
clearly related to a higher myopathic risk in
Asians, rosuvastatin is labelled for lower doses
in Asians
[49]
and none of the statins are approved in
Japan at the highest doses approved in the US.
[50]
5.1.2 Genetic Factors
Genetic predisposition and interindividual
variability in susceptibility to statin-induced ad-
verse muscular events have been reported. A
number of candidate gene variants that encode
statin metabolizing enzymes and receptors,
[51,52]
OATPs
[53]
and CoQ10
[54]
have been implicated.
Genetic polymorphisms may result in variant
expression of the CYP isoenzymes both in the li-
ver and small intestine. Low hepatic or intestinal
expression of the CYP3A4 isoenzyme (‘poor
metabolizers’) results in decreased first-pass me-
tabolism for lovastatin, simvastatin and atorv-
astatin, which increases their bioavailability. In
a recently published study that performed a
genome-wide scan in patients with definite or in-
cipient myopathy receiving simvastatin 80 mg
daily, common variants in solute carrier organic
anion transporter (SLCO) 1B1 on chromosome
12, which encodes the OATP and thereby the
hepatic uptake of most statins, were linked to a
substantial excess risk of statin myopathy, with
60% of all myopathy cases attributable to one
specific common variant (rs4149056 C). Hetero-
zygotes for this variant displayed a 4-fold in-
crease in the incidence of myopathy, whereas the
homozygotes had a 17-fold increase.
[55]
Notably,
Dose
Pharmacokinetic properties
water solubility
first-pass metabolism
protein binding
Drug properties Patient characteristics Concomitant medications
Demographic characteristics
age
race
sex
Genetic predisposition
transporting polypeptides
CYP isoenzymes
Co-enzyme Q10 depletion
Co-morbidites
systemic diseases
infectious diseases
alcoholism/drug addiction
major surgical operations
myopathy
hereditary
acquired
Pharmacokinetic interactions
CYP metabolism
CYP3A4
CYP2C9
glucuronidation
Pharmacodynamic interactions
synergic myotoxicity
Increased systemic bioavailability
Enhanced susceptibility to elevated blood levels of statins
Myopathy
Fig. 1. Risk factors for statin-induced myopathy. CYP = cytochrome P450 enzyme.
176 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
these associations were demonstrated among
patients receiving high doses of simvastatin
(80 mg daily), indicating that genotypic control
could optimize tailored therapeutic adjustments
in patients under high-dose regimens and with
coexisting risk factors.
5.1.3 Co-Morbidities
The incidence of statin-induced rhabdomyo-
lysis may be higher in patients with existing
myopathies, either hereditary (e.g. carnitine pal-
mityl transferase II deficiency, McArdle’s diseas e
and myoadenylate deaminase deficiency) or ac-
quired (e,g. postpolyomyelitis syndrome).
[56,57]
Statins have also been implicated in the potential
aggravation of myasthenia gravis.
[58]
Further-
more, an underlying metabolic predisposition
consisting of biochemical abnormalities in mito-
chondrial or fatty acid metabolism in myocytes
may render some apparently healthy individuals
more susceptible to the development of statin-
induced myopathic outcomes than others.
[59]
Underlying chronic systemic diseases may
serve as non-modifiable risk factors that decrease
statin metabolism and excretion, and thereby in-
crease their syst emic bioavailability. These fac-
tors render some patients more suscept ible to
myopathy and increase the probability of adverse
muscle events, whi ch may ensue at any time
during the administration of a statin. Although
limited data suggest a beneficial cardiovascular
effect of statins in patients with moderate renal
impairment,
[60]
coexisting renal failure increases
the risk of statin-induced myopathic events.
[13]
Diabetes mellitus constitutes a further myopathic
risk factor in patients receiving statins, particu-
larly combined with advanced age and chronic
renal fail ure,
[11]
although there is no consensus of
opinion.
[61]
Enhanced risk of statin-induced
myopathy with excessive alcohol consumption
cannot be conclusively supported by data from
randomized trials as alcoholism is an exclusion
criterion in most trials. However, increased al-
cohol intake per se confers a myotoxic poten-
tial
[62]
and alcohol abuse could raise the blood
levels of statins.
[12]
Untreated hypothyroidism is
considered to increase the risk of statin myo-
pathy,
[11,25,63]
and statins may aggrava te the
muscle symptoms and CK elevation caused by
occult hypothyroidism. Liver dysfunction has
been considered a risk factor for statin myo-
pathy,
[11,64-66]
mainly due to the involvement of
the hepatobiliary system in the metabolism and
excretion of most statins. Although hepatic dys-
function has been associated with statin-induced
rhabdomyolysis in reports by regulatory autho-
rities,
[67]
the exclusion of patients with hepatic
failure from randomized controlled trials pre-
vents the establishment of a direct link between
impaired liver function and height ened risk of
myopathy.
[4,6,17]
Furthermore, acutely acting factors predis-
pose to myopathy independently, and may trig-
ger the development of severe myopathy, even
rhabdomyolysis, in statin-receiving individuals.
Such precipitating conditions include the use of
addictive drugs (e.g. amfetamines, cocaine,
heroin, LSD, ecstasy),
[62]
serious viral
[68]
or bac-
terial infection,
[69,70]
major trauma and intense
muscle activity. Statins can exacerbate exercise-
induced skeletal muscle injur y, as reported in an
observational study in patients receiving high-
dose statins
[25]
and as suggested by the greater
CK response to exercise in statin- compared with
placebo-treated patients.
[71]
Statin-related myo-
pathy has been reported in the setting of extensive
surgical operations;
[72,73]
therefore, a short-term
withdrawal of statins during hospitalization
for major surgery is recommended.
[11]
In the case
of vascular surgery in particular, including cor-
onary bypass procedures, statins should not be
discontinued
[74]
in light of their beneficial plaque-
stabilizing effect, wi th the exception of preopera-
tive muscular sympto ms, marked perioperative
tissue compression or prolonged postoperative
energy deprivation.
[75]
5.2 Statin Properties
5.2.1 Dose-Dependent Effects
While the therape utic benefit from statin
therapy is related to the achieved LDL-C reduc-
tion,
[76]
the risk of adverse muscular events ap-
pears to be a dose-dependent adverse effect
[21]
regardless of the degree of LDL-C decrease.
[15]
However, there does not ap pear to be a linear
Statin-Induced Myopathy: Risk Factors and Drug Interactions 177
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
relationship between plasma levels achieved by a
certain drug dose and the risk of adverse muscular
events. Increased myopathic risk has been de-
monstrated with higher than currently marketed
doses of simvastatin (160 mg)
[76]
and pravastatin
(160 mg).
[77]
An increased incidence of myopathy
has also been shown in patients with acute cor-
onary syndromes receiving simvastatin 80 mg daily
compared with placebo or simvastatin 20 mg.
[18]
A
higher incidence of statin-related myalgia was at-
tributed to atorvastatin 80 mg compared with
simvastatin 20 mg
[14]
but notably this did not occur
when atorvastatin 80 mg was compared with either
atorvastatin 10 mg
[19]
or placebo.
[20]
5.2.2 Physicochemical Properties of Statins:
Lipophilicity versus Hydrophilicity
In vitro research data indicate that pravastatin,
which is water soluble, is less myotoxic in relation
to lovastatin and simvastatin.
[78]
Although the in-
hibition of hepatocellular cholesterol synthesis was
comparable between these three statins, the effect
of pravastatin was 85 times weaker in rat myo-
cytes. Moreover, pravastatin was 100-200 times
(in an inversely dose-dependent mode) less myo-
toxic.
[78]
Overall, different statins seem to exert di-
verse dose-dependent effects on the HMG CoA
reductase activity of non-hepatic cells in vitro.The
decreased myotoxicity of pravastatin appears to be
related to its decreased penetration of the cell
membrane and thus uptake by extra-hepatic tis-
sues, presumably associated with the hydro-
philicity of the molecule. Pravastatin is taken up by
the hepatic cells via a sodium-independent bile
acid transporter, the OATP,
[79]
which, along with
sodium-dependent taurocholate cotransporting
polypeptide, also mediates the active hepatic up-
take of the hydrophilic rosuva statin molecule. The
lipid-rich membranes of non-hepatic cells, such as
muscle cells, lack OATP so that they function as a
barrier to hydrophilic statins while allowing pas-
sive diffusion to lipophilic statins. However, the
hydrophilicity of some statins per se has not been
proven to offer clinically significant muscular pro-
tection
[12]
and no clinical evidence supports a direct
association between the degree of lipophilicity
and the myotoxic potential
[9]
since cases of
rhabdomyolysis have also been attributed to hy-
drophilic statins.
5.3 Statin-Drug Interactions
The interaction of statins with other categories
of medications can enhance their myotoxic po-
tential (table III). Indeed, approximately 60% of
cases of statin-related rhabdomyolysis are related
to drug interactions.
[24]
The underlying mechan-
ism usually has a pharmacokinetic basis invol-
ving intestinal absorption, distribution, metabo-
lism, protein binding or excretion of statins. The
majority of reported cases pertain to competition
at the level of hepatic metabolism,
[80]
considering
that over half of currently available drugs are
metabolized by the CYP3A4 isoenzyme; inhibi-
tion of the CYP activity by co administered drugs
increases the risk of myopathic events. Simvas-
tatin and lovastatin appear to be more susceptible
to the inhibiting effect of other CYP3A4 sub-
strates than atorvastatin. Similarly, the inter-
action between fluvastatin and CYP2C9 inhibitors
or competitive substrates may be of clinical im-
portance, whereas CYP450 isoenzymes are mini-
mally involved in rosuvastatin clearance.
Table III. Substances that may precipitate statin-induced myopathy
Non-hypolipidaemic medicines
Ciclosporin
Macrolide antibacterials (erythromycin, clarithromycin)
Azole antifungals (itraconazole, ketoconazole, fluconazole)
Calcium channel antagonists (diltiazem, verapamil)
Nefazodone
HIV protease inhibitors (ritonavir, nelfinavir, indinavir)
Warfarin
Histamine H
2
receptor antagonists (cimetidine, ranitidine)
Omeprazole
Amiodarone
Hypolipidaemic medicines
Fibrates (gemfibrozil > bezafibrate, clofibrate, fenofibrate)
Niacin
Other substances
Grapefruit juice
Over-the-counter medications (Chinese red rice fungus)
178 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
Other sites of potential pharmacokinetic
interactions include inhibition of metabolism
by intestinal wall isoenzymes of the CYP system,
prevention of the OATP-mediated hepatocel-
lular uptake, blocking of biliary excretion and
inhibition of the renal elimination of hydrophilic
metabolites.
[80]
Pharmacodynamic interactions involving sta-
tins are less common because of the high selectivity
of statins as HMG-CoA reductase inhibitors.
The concomitant administration of agents with an
independent myotoxic effect, such as fibrates, can
synergistically increase the risk of statin-associated
myopathy. In that case, a pharmacokinetic com-
ponent may also come into play.
5.3.1 Interactions with Non-Hypolipidaemic Agents
Pharmacokinetic Interactions with Cytochrome
P450 Enzyme (CYP) 3A4 Inhibitors and Competing
Substrates
Inhibitors of CYP3A4 isoenzyme decrease
statin metabolism and thus increase their serum
levels and the likelihood of myopathy. Such en-
zymatic inhibitors include azole antifungals
(itraconazole, ketoconazole, fluconazole),
[37,81,82]
macrolide antibacterials (erythromycin, clari-
thromycin),
[83,84]
calcium channel antagonists
diltiazem
[85,86]
and verapamil, the antidepressant
nefazodone and the consumption of grapefruit
juice exceeding approximately 1 L daily. Grape-
fruit juice contain s 6
0
,7
0
-dihydroxybergamottin,
which acts as an inhibi tor of the intestinal
CYP3A4 isoenzyme resulting in decreased meta-
bolism and thereby enhanced bioavailability of
statins.
[80]
HIV protease inhibitors (ritonavir, nelfinavir,
indinavir) are recognized CYP3A4 inhibitors and
this property renders the myotoxic potential of
their combination with statins high risk,
[87]
in
particular those statins that rely to a large extent
on the CYP3A4 isoform for their metabolism. Of
clinical interest is the adverse effect of HIV pro-
tease inhibitors on the lipid profile,
[88]
which may
increase the risk of cardiovascular disease and
pancreatitis and often requires the administration
of lipid-lowering agents.
[89]
Statins are the most effective medicines for the
treatment of hypercholesterolaemia in patients
who have undergone transplantation,
[90]
and
their immunomodulatory properties appear to
provide general protection for the graft. How-
ever, ciclosporin (cyclosporine) inhibits both in-
testinal and hepatic CYP3A4 activity and can
therefore lead to increased bioavailability of sta-
tins metabolized by this cytochrome.
[91]
The more
lipophilic the stat in and the greater the systemic
exposure to unbound active statin compound, the
greater the potential for myopathy.
[73,92]
Pravas-
tatin and fluvastatin are less likely to interact
with ciclosporin on a pharmacokinetic basis.
However, ciclosporin has been reported to in-
crease serum levels of pravastatin.
[93]
Competi-
tion at the level of biliary clearance resulting in
reduced pravastatin removal through the bile
duct and prevention of the P-glycoprotein trans-
fer are considered the main pathomechanisms,
indicating that CY P3A4 is not the only site in-
volved in clinically relevant ciclosporin-statin
interactions.
The combination of statins with warfarin is
likely to increase the serum levels of warfarin,
thereby potentiating its anticoagulant effect.
[94]
Regular anticoagulation control and possibly
warfarin dose adjustment may thus be required.
However, the potentiating effect of warfarin on
statin levels has not been studied sufficiently.
[95]
A hypothesis has been articulated that as war-
farin constitutes the substrate of CYP2C9, and
partly of CYP3A4, it could compete with statins
in their enzymatic conversion.
Pharmacokinetic Interactions with CYP2C9
Inhibitors and Competing Substrates
Azole antifungal agents are recognized in-
hibitors of CYP2C9, as well as the previously
mentioned CYP3A4. This necessitates a higher
index of suspicion when they are administered in
patients receiving fluvastatin. For example, flu-
conazole has been reported to increase fluvastatin
bioavailability,
[96]
although no cases of rhab-
domyolysis attributable to such a combination
are known. Furtherm ore, histamine H
2
receptor
antagonists cimetidine and ranitidine, and the
proton pump inhibitor omeprazole, which are
Statin-Induced Myopathy: Risk Factors and Drug Interactions 179
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
also substrates of CYP2C9, enhance fluvastatin’s
systemic exposure, but without particular clinical
significance. Of note, omeprazole also appears to
possess a CYP3A4 induction capacity, poten-
tially increasing the biotransformation and thus
decreasing levels of statins that are substrates of
the CYP3A4 isoenzyme.
[37]
5.3.2 Interactions with Other Hypolipidaemic
Agents
Fibrates
In many cases of mixed dyslipidaemia, in dia-
betic patients or in patients with high triglycer-
ides despite the achieve ment of the desirable
LDL-C goal, the coadministration of statins with
fibrates is an attractive therape utic option. Ac-
cording to data from epidemiological studies and
clinical trials, the combination of any statin with
fibrates increases the risk of myopathy, which is
usually observed within the first 12 weeks by the
initiation of treatment. The incidence of myo-
toxicity with this combination is 0.12%.
[97]
Even
if most reports involve gemfibrozil,
[98,99]
other
fibrates (bezafibrate, clofibrate, fenofibrate) have
also been implicated in cases of rhabdomyolysis
when used alone and have an additive myotoxic
potential when combined with statins.
[99]
The
presence of gemfibrozil in most cases of rhabdo-
myolysis is partly explained by its wider clinical
use than other fibrates; however, the differential
safety profile of fibrates seems to remain even
after correction for the wider prescription of
gemfibrozil.
[100]
Since fibrates do not interfere with CYP-
mediated statin elimination, the additive adverse
effect when combined with statins appears to
have a predominantly pharmacodynamic basis
(synergy). The incidence of hospitalized patients
with rhabdomyolysis prescribed fibr ate mono-
therapy has been reported to be more than
5 times higher than with atorvastatin, pravastatin
or simvastatin monotherapy.
[101]
Pharmacoki-
netic parameters are also believed to account to
some degree for the observed interactions. Statin
glucoronidation is an intermediate step in the
conversion of active acid forms to lactones, which
in turn are metabolized by the hepatic P450 sys-
tem.
[102]
The inhibition of statin hydroxy acid
glucoronidation mediated by gemfibrozil
[103]
but
not fenofibrate and subsequent increased bio-
availability of statins is believed to partly account
for the increased myopathic risk associated with
the combination of statins with fibrates. Further-
more, statin-fibrate interactions may in part be
mediated through the activation of the peroxi-
some proliferator-activated family of nuclear re-
ceptors,
[104]
which have been shown to affect
CYP regulation. Moreover, gemfibrozi l has been
reported to reduce the renal clearance of pravas-
tatin by competitively acting at the transport
proteins
[105]
and it increases pravastatin and ro-
suvastatin concentrations by impeding their bili-
ary excretion.
[37]
Niacin
The addition of niacin in a statin-receiving
patient can yield complementary benefits in
achieving a comprehensive lipid control. The
concomitant administration of statins with high
doses of niacin has been associated with rhabdo-
myolysis in a limited number of anecdotal
reported c ases
[106]
through a mechanism that re-
mains unknown but appears to be unrelated to
statin serum levels. Niacin is not implicated as a
strong precipitating factor for statin-induced
myopathy, and the combination of statin and
niacin is considered to carry a lower risk than
statin-fibrate coadministration.
[21]
Based on cur-
rent evidence, no excessive risk of myopoat hy as a
result of a statin-niacin combination, compared
with that expected by adding one agent to the
other, can be supported.
[107]
Ezetimibe
Combined inhibition of intestinal cholesterol
absorption mediated by ezetimibe and hepatic
cholesterol synthesis via statins has emerged as a
challenging therapeutic option. Anecdotal reports
of myopathy attributed to the combination of sta-
tin plus ezetimibe
[108,109]
have not been confirmed
in the setting of randomized controlled trials. An
enhanced lipid-lowering effect and comparable
safety profile was shown when ezetimibe was ad-
ded to statins in patients with hypercholester-
olaemia.
[110]
The incidence of muscle-related events
was not higher in patients taking simvastatin
alone than the combination of simvastatin plus
180 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
ezetimibe according to pooled data from 17 re-
lative randomized clinical trials.
[111]
In recent stud-
ies, the addition of ezetimibe 10 mg resulted in
greater lipid-lowering efficacy than, and equal
safety and tolerability to, uptitration of atorvasta-
tin 2040mg in patients at moderately high risk
[112]
and to the doubling of atorvastatin 4080 mg in
patients at high risk for coronary heart disease.
[113]
Overall, current evidence cannot support enhanced
risk for statin-related myopathy by the coadmi-
nistration of ezetimibe.
[21]
6. Recommendations for the Prevention
and Management of Statin-Induced
Myopathy
6.1 Prevention of Statin-Induced Myopathy
Prevention and early recognition are the best
approaches to managing statin-related myopathy
and averting serious sequelae. Statin treatment
should begin with low doses that can be pro-
gressively increased; if necessary, the anticipated
clinical benefit should be weighed against the
increased myopathic risk. Coadministration of
interacting medications should be avoided
whenever possible. When a high-risk combina-
tion is necessary, small doses of the presumably
safest statin for any given combination should be
prescribed.
Patients should be counselled on the risk and
warning signs of myopathy, and on the possibility
of drug interactions. Any unexplained muscle
symptoms should be reported immediately to the
attending physician. Patients at high risk should
be followed up clinically, especially during the
first months of treatment. When acute clinical
conditions that can precipitate rhabdomyoly-
sis coexist, it is advisable to interrupt statins
temporarily.
Routine measurement of pretreatment CK
levels is not recommended because of the rarity of
myopathy when statins are prescribed at usual
doses in the general population. However, CK
should be measured in patients at high risk for
myopathy, at baseline and regularly during the
course of statin therapy.
[21]
If a statin-fibrate combination is required,
fenofibrate is the preferred option over gemfi-
brozil.
[80,100,114]
Thestatindosesshouldremain
below the maximal levels. Fluvastatin may be ap-
propriate for combination with gemfibrozil.
[115]
Particular attention is required for patients with
increased CK levels, and renal and hepatic
dysfunction.
On the basis of current evidence, routine CoQ10
supplementation cannot be recommended to pre-
vent statin-related myopathic events.
[21]
CoQ10
supplementation might be considered in the setting
of CoQ10-depleting conditions, such as advanced
age or multisystem diseases.
[30]
6.2 Management of Statin-Induced
Myopathy
Statin-induced myopathy is usually mild and
reversible upon statin discontinuation; however,
in very rare cases it may evolve towards severe
muscle damage, even rhabdomyolysis. If statin-
related myopathy is suspected, more common
causes of symptoms and/or CK elevation should
be ruled out by thorough history taking, physical
examination and laboratory tests. Other etiolo-
gies of muscle symptoms include unusual phy si-
cal activity, trauma, falls, accidents, seizures,
alcohol, drugs (corticosteroids, antipsychotics,
cocaine, amfetamines), occult hypothyroidism,
infections and autoimmune disorders (polymyo-
sitis, dermatomyositis, rheumatic polymyal-
gia).
[116]
Initial tests include CK levels, serum
thyroid-stimulating hormone levels, and renal
function with urinalysis if rhabdomyolysis is
suspected. In rare cases of persistent symptoma-
tology despite statin discontinuation, further in-
vestigations including electromyography and
muscle biopsy may be required, in consultation
with experts in muscle diseases.
[116]
The intensity of clinical symptoms along with
the magnitude of CK elevation should guide
clinical management, as summarized in figure 2.
In the case of CK levels <10 · the upper limit of
normal, without or with tolerable muscle symp-
toms, the statin may be continued at the same or
a smaller dose.
[116]
In the presence of tolerable
symptoms with CK serum elevation >10 · the
Statin-Induced Myopathy: Risk Factors and Drug Interactions 181
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
upper limit of normal, or if frank rhabd omyolysis
develops, statins should be discontinued. Rhab-
domyolysis prompts in-hospital supportive treat-
ment consisting of intravenous hydration and
monitoring for potential complications.
[117]
Intolerable muscle symptoms, regardless of CK
levels, require temporary statin interruption.
Once symptoms resolve, the same or lower dose
of the same or a different statin can be restarted.
Alternative statin regimens with an ostensibly
lower myopathic potential may include fluvasta-
tin extended release,
[118]
low-dose rosuvastatin,
[119]
and non-daily regimens with atorvastatin
[120-122]
or rosuvastatin.
[123-126]
If symptoms reoccur,
statin suspension should be permanent. Non-
statin lipid-lowering agents that have shown ef-
ficacy and safety in patien ts intolerant to statins
include ezetimibe,
[118,127]
alone or in combination
with bile acid-binding resin.
[128]
7. Conclusions
Statins represent the most effective class of
medications for reduction of LDL-C and there-
fore for the prevention of cardiovascula r athero-
sclerotic disease. Although myopathy is their
most common adverse effect, severe muscle-
related complications are very rare and should
not deter physicians from prescribing these gen-
erally safe and well tolerated agents. Several fac-
tors that may predispose to, or trigger, myopathic
events in statin-receiving patients have been
well characterized. Individual risk stratification,
taking into consideration patient characteristics,
Suspected statin-induced myopathy
Asymptomatic CK elevation
Exclude other causes
Measure CK levels
CK <10 × ULN CK >10 × ULN
CK <10 × ULN
CK >10 × ULN
CK >10 × ULN and renal function impairment
Normal
Myalgia
Myositis
TolerableIntolerable
RhabdomyolysisAssess symptom severity
Assess CK levels
Elevated
Yes
Yes
No
No
Continue statin
(same/reduced dose)
Treatment adjustment
based on CK monitoring
Continue statin
(same/reduced dose)
Treatment adjustment
based on symptoms
Restart statin once
symptoms resolve
(same/different statin,
same/reduced dose)
Symptoms recurrence?
Discontinue
statin
Discontinue statin Discontinue statin
Muscle-related symptoms ± CK elevation
Fig. 2. Assessment and management of statin-induced myopathy. CK = creatine kinase; ULN = upper limit of normal.
182 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
coadministered medications and statin pharma-
cological properties, should determine clinical
decision making. Considering the dose-dependen t
nature of statin-related myopathy, physicians
should start cautiously with lower doses in the
presence of predisposing conditions and weigh
the benefit of lipid lowering versus the potential
of excess risk when uptitrating doses. Combina-
tion therapy with other classes of hypolipidaemic
agents may be opted for when aggressive lipid-
lowering therapy is required. Since most patients
eligible for statins receive multiple concomitant
medications, often sharing the metabolic path-
way of the CYP system, recognition of potential
drug interactions is critical. Knowledge of the
pharmacokinetic properties of currently available
statins may allow the identification of the statin
at the presumably lowest risk for drug inter-
actions. In the clinical setting, counselling patients
on the risk and warning signs of myopathy will
increase awareness and allow prompt recogni-
tion and appropriate management of myopathic
events. In order to accurately estimate the true
incidence of statin-induced myopathy and en-
hance our understanding of potential risk factors,
a more complete and formal reporting of the
entire spectrum of muscle-related events attribu-
table to statins is required.
Acknowledgements
No sources of funding were used to assist in the prepara-
tion of this review. The authors have no conflicts of interest
to declare. Y.S. Chatzizisis and K.C. Koskinas contributed
equally to this work.
References
1. Chatzizisis YS, Jonas M, Beigel R, et al. Attenuation of
inflammation and expansive remodeling by valsartan
alone or in combination with simvastatin in high-risk
coronary atherosclerotic plaques. Atherosclerosis 2009;
203: 387-94
2. Istvan ES, Deisenhofer J. Structural mechanism for statin
inhibition of HMG-CoA reductase. Science 2001; 292
(5519): 1160-4
3. Grundy SM, Cleeman JI, Merz CN, et al., for the Co-
ordinating Committee of the National Cholesterol Edu-
cation Program and Endorsed by the National Heart,
Lung, and Blood Institute, American College of Cardiol-
ogy Foundation, and American Heart Association.
Implications of recent clinical trials for the National
Cholesterol Education Program Adult Treatment Panel
III Guidelines. Circulation 2004; 110: 227-39
4. Cannon CP, Braunwald E, McCabe CH, et al. Intensive
versus moderate lipid lowering with statins after acute
coronary syndromes. N Engl J Med 2004; 350: 1495-504
5. Schwarz GG, Olsson AG, Ezekowitz MD, et al. Effects of
atorvastatin on early recurrent ischemic events in acute
coronary syndromes. The MIRACLE study: a rando-
mized controlled trial. JAMA 2001; 285: 1711-8
6. Heart Protection Study Collaborative Group. MRC/BHF
Heart Protection Study of cholesterol lowering with sim-
vastatin in 20,536 high-risk individuals: a randomised
placebo-controlled trial. Lance t 2002; 360 (9326): 7-22
7. Shepherd J, Blauw GJ, Murphy MB, et al., PROSPER
study group. Pravastatin in elderly individuals at risk of
vascular disease (PROSPER): a randomised controlled
trial. PROspective Study of Pravastatin in the Elderly at
Risk. Lancet 2002; 360: 1623-30
8. Sever PS, Dahlof B, Poulter NR, et al., ASCOT in-
vestigators. Prevention of coronary and stroke events with
atorvastatin in hypertensive patients who have average or
lower-than-average cholesterol concentrations, in the
Anglo Scandinavian Cardiac Outcomes Trial Lipid
Lowering Arm (ASCOT-LLA): a multicentre randomised
controlled trial. Lancet 2003; 361: 1149-58
9. Evans M, Rees A. Effects of HMG-CoA reductase in-
hibitors on skeletal muscle: are all statins the same? Drug
Saf 2002; 25 (9): 649-63
10. Bolego C, Baetta R, Bellosta S, et al. Safety considerations
for statins. Curr Opin Lipidol 2002; 13 (6): 637-44
11. Pasternak RC, Smith SC Jr, Bairey-Merz CN, et al., and
Writing Committee Members. ACC /AHA/NHLBI clin-
ical advisory on the use and safety of statins. Circulation
2002; 106: 1024-8
12. Bays H. Statin safety: an overview and assessment of the
data: 2005. Am J Cardiol 2006; 97 Suppl. 8A: 6-26C
13. Armitage J. The safety of statins in clinical practice. Lancet
2007; 370: 1781-90
14. Pedersen TR, Faergeman O, Kastelein JP, et al. High-dose
atorvastatin vs usual-dose simvastatin for secondary pre-
vention after myocardial infarction. The IDEAL study: a
randomized controlled trial. JAMA 2005; 294: 2437-45
15. Law M, Rudnicka AR. Statin safety: evidence from the
published literature. Am J Cardiol 2006; 97 Suppl. 8A:
52-60C
16. Davidson MH, Clark JA, Glass LM, et al. Statin safety: an
appraisal from the Adverse Event Reporting System
(AERS). Am J Cardiol 2006; 97 Suppl. 8A: 32-43C
17. Ridker PM, Danielson E, Fonseca FA, et al., JUPITER
Study Group. Rosuvastatin to prevent vascular events in
men and women with elevated C-reactive protein. N Engl
J Med 2008; 359 (21): 2195-207
18. de Lemos JA, Blazing MA, Wiviott SD, et al. Early in-
tensive vs. a delayed conservative simvastatin strategy in
patients with acute coronary syndromes: phase Z of the A
to Z trial. JAMA 2004; 292: 1307-16
19. LaRosa JC, Grundy SM, Waters DD, et al. Intensive lipid
lowering with atorvastatin in patients with stable cor-
onary disease. N Engl J Med 2005; 352: 1425-35
Statin-Induced Myopathy: Risk Factors and Drug Interactions 183
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
20. The Stroke Prevention by Aggressive Reduction in
Cholesterol Levels (SPARCL) Investigators. High-dose
atorvastatin after stroke or transient ischemic attack.
N Engl J Med 2006; 355: 549-59
21. Thompson PD, Clarkson PM, Rosenson RS. An assess-
ment of statin safety by muscle experts. Am J Cardiol
2006; 97 Suppl.: 69-76C
22. US Food and Drug Administration. Office of Drug Safety
annual report 2001 [online]. Available from URL: http:
//www.fda.gov/AboutFDA/CentersOffices/CDER/ucm16
9925.htm. [Accessed 2009 Aug 5]
23. Staffa JA, Chang J, Green L. Cerivastatin and reports of
fatal rhabdomyolysis. N Engl J Med 2002; 346 (7): 539-40
24. Kashani A, Phillips CO, Foody JA, et al. Risks associated
with statin therapy: a systematic overview of randomized
clinical trials. Circulation 2006; 114: 2788-97
25. Bruckert E, Hayem G, Dejager S, et al. Mild to moderate
muscular symptoms with high-dosage statin therapy in
hyperlipidemic patients: the PRIMO study. Cardiovasc
Drugs Ther 2005; 19: 403-14
26. Silva MA, Swanson AC, Gandhi PG, et al. Statin-related
adverse events: a meta-analysis. Clin Ther 2006; 28: 26-33
27. Wlodarczyk J, Sullivan D, Smith M. Comparison of bene-
fits and risks of rosuvastatin versus atorvastatin from a
meta-analysis of head-to-head randomized controlled
trials. Am J Cardiol 2008; 102 (12): 1654-62
28. Vaklavas C, Chatzizisis YS, Ziakas A, et al. Molecular
basis of statin-associated myopathy. Atherosclerosis 2009
Jan; 202 (1): 18-28
29. Morita I, Sato I, Ma L, et al. Enhancement of membrane
fluidity in cholesterol-poor endothelial cells pre-treated
with simvastatin. Endothelium 1997; 5 (2): 107-13
30. Marcoff L, Thompson PD. The role of coenzyme Q10 in
statin-associated myopathy: a systematic review. J Am
Coll Cardiol 2007; 49: 2231-7
31. Laaksonen R, Jokelainen K, Sahi T, et al. Decreases in
serum ubiquinone concentrations do not result in reduced
levels in muscle tissue during short-term simvastatin
treatment in humans. Clin Pharmacol Ther 1995; 57: 62-6
32. Chatzizisis YS, Vaklavas C, Giannoglou GD. Coenzyme
Q10 depletion: etiopathogenic or predisposing factor in
statin associated myopathy [letter]? Am J Cardiol 2008
Apr 1; 101 (7): 1071
33. Sakamoto K, Mikami H, Kimura J. Involvement of or-
ganic anion transporting polypeptides in the toxicity of
hydrophilic pravastatin and lipophilic fluvas tatin in rat
skeletal myofibres. Br J Pharmacol 2008; 154: 1482-90
34. Dorajoo R, Pereira BP, Yu Z, et al. Role of multi-drug
resistance-associated protein-1 transporter in statin in-
duced myopathy. Life Sci 2008; 82: 823-30
35. Hamelin BA, Turgeon J. Hydrophilicity/lipophilicity: re-
levance for the pharmacology and clinical effects of
HMG-CoA reductase inhibitors. Trends Pharmacol Sci
1998; 19 (1): 26-37
36. Shitara Y, Sugiyama Y. Pharmacokinetic and pharmaco-
dynamic alterations of 3-hydroxy-3-methylglutaryl co-
enzyme A (HMG-CoA reductase inhibitors: drug-drug
interactions and interindividual differences in transporter
and metabolic enzyme functions. Pharmacol Ther 2006;
112: 71-105
37. Bellosta S, Paoletti R, Corsini A. Safety of statins: focus on
clinical pharmacokinetics and drug interactions. Circula-
tion 2004; 109 Suppl. III: III50-7
38. Haria M, McTavish D. Pravastatin: a reappraisal of its
pharmacological properties and clinical effectiveness in
the manag ement of coronary heart disease. Drugs 1997;
53 (2): 299-336
39. White CM. A review of the pharmacologic and pharma-
cokinetic aspects of rosuvastatin. J Clin Pharmacol 2002;
42: 963-70
40. Schech S, Graham D, Staffa J, et al. Risk factors for statin-
associated rhabdomyolysis. Pharmacoepidemiol Drug Saf
2007; 16 (3): 352-8
41. Lee E, Ryan S, Birmingham B, et al. Rosuvastatin phar-
macokinetics and pharmacogenetics in white and Asian
subjects residing in the same environment. Clin Pharma-
col Ther 2005; 78 (4): 330-41
42. Tan C-E, Ma S, Wai D, et al. Can we apply the National
Cholesterol Education Program Adult Treatment Panel
definition of the metabolic syndrome to Asians? Diabetes
Care 2004; 27: 1182-6
43. Matsuzawa Y, Kita T, Mabuchi H, et al., for the J-LIT
Study Group. Sustained redu ction of serum cholesterol in
low-dose 6-year simvastatin treatment with minimum side
effects in 51 321 Japanese hypercholesterolemic patients:
implication of the J-LIT study, a large scale nationwide
cohort study. Circ J 2003; 67: 287-94
44. Morales D, Chung N, Zhu J-R, et al. Efficacy and safety
of simvastatin in Asian and non-Asian coronary heart
disease patients: a comparison of the GOALLS and
STATT studies. Curr Med Res Opin 2004; 20: 1235-43
45. Tan CE, Loh LM, Tai ES. Do Singapore patients require
lower doses of statins? The SGH Lipid Clinic experience.
Singapore Med J 2003; 44: 635-8
46. Kim K, Johnson JA, Derendorf H. Differences in drug
pharmacokinetics between East Asians and Caucasians
and the role of genetic polymorphisms. J Clin Pharmacol
2004; 44: 1083-105
47. Wang A, Yu BN, Luo CH, et al. Ile 118Val genetic poly-
morphism of CYP3A4 and its effects on lipid-lowering
efficacy of simvastatin in Chinese hyperlipidemic patients.
Eur J Clin Pharmacol 2005; 60: 843-8
48. Liao JK. Safety and efficacy of statins in Asians. Am J
Cardiol 2007; 99 (3): 410-4
49. Food and Drug Administration Center for Drug Evaluation
and
Res
earch. FDA Public Health Advisory on Crestor
(rosuvastatin) [online]. Available from URL: http://www.
fda.gov/Drugs/DrugSafety/PublicHealthAdvisories/ucm051
756.htm [Accessed 2009 Aug 1]
50. Saito M, Hirata-Koizumi M, Urano T, et al. A literature
search on pharmacokinetic drug interactions of statins
and analysis of how such interactions are reflected in
package inserts in Japan. J Clin Pharm Ther 2005; 30:
21-37
51. Vermes A, Vermes I. Genetic polymorphisms in cyto-
chrome P450 enzymes: effect on efficacy and tolerability
of HMG-CoA reductase inhibitors. Am J Cardiovasc
Drugs 2004; 4: 247-55
52. Mulder AB, van Lijf HJ, Bon MA, et al. Association of
polymorphism in the cytochrome CYP2D6 and the effi-
184 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
cacy and tolerability of simvastatin. Clin Pharmaco l Ther
2001; 70: 546-51
53. Morimoto K, Ueda S, Seki N, et al. OATP-
C(OATP01B1)*15 is associated with statin-induced
myopathy in hypercholesterolemic patients. Clin Phar-
macol Ther 2005; 77: P21
54. Oh J, Ban MR, Miskie BA, et al. Genetic determinants of
statin intolerance. Lipids Health Dis 2007; 6: 7
55. The SEARCH Collaborative Group. SLCO1B1 variants
and statin-induced myopathy: a genomewide study.
N Engl J Med 2008; 359 (8): 789-99
56. Leung NM, Ooi TC, McQueen MJ. Use of statins and
fibrates in hyperlipidemic patients with neuromuscular
disorders. Arch Intern Med 2000; 132: 418-9
57. Franc S, Bruckert E, Giral P, et al. Rhabdomyolysis in
patients with preexisting myopathy, treated with anti-
lipemic agents. Presse Med 1997; 26: 1855-8
58. Oh SJ, Dhall R, Young A, et al. Statins may aggravate
myasthenia gravis. Muscle Nerve 2008; 38: 1101-7
59. Vladutiu R, Isackson P, Peltier W, et al. Genetic risk fac-
tors and metabolic abnormalities associated with lipid
lowering therapies. Muscle Nerve 2006; 34 (2): 153-62
60. Fried LF, Orchard TJ, Kasiske BL. Effect of lipid reduc-
tion on the progression of renal disease: a meta-analysis.
Kidney Int 2001; 59: 260-9
61. Nichols GA, Koro CE. Does statin therapy initiation in-
crease the risk for myopathy? An observational study of
32 225 diabetic and nondiabetic patients. Clin Ther 2007;
29 (8): 1761-70
62. Giannoglou GD, Chatzizisis YS, Misirli G. The syndrome
of rhabdomyolysis: pathophysiology and diagnosis. Eur J
Intern Med 2007; 18: 90-100
63. Bar SL, Holmes DT, Frohlich J. Asymptomatic hypo-
thyroidism and statin induced myopathy. Can Fam Phy-
sician 2007; 53 (3): 428-31
64. Sathasivam S, Lecky B. Statin induced myopathy. BMJ
2008; 337: a2286
65. Davidson MH, Robinson JG. Safety of aggressive lipid
management. J Am Coll Cardiol 2007; 49: 1753-62
66. Thompson PD, Clarkson P, Karas RH. Statin-associated
myopathy. JAMA 2003; 289 (13): 1681-90
67. Ronaldson KJ, O’Shea JM, Boyd IW. Risk factors for
rhabdomyolysis with simvastatin and atorvastatin. Drug
Saf 2006; 29 (11): 1061-7
68. Wong WM, Wai-Hung Shek T, Chan KH. Rhabdomyo-
lysis triggered by cytomegalovirus infection in a heart
transplant patient on concomitant cyclosporine and
atorvastatin therapy. Gastroenterol Hepatol 2004; 19 (8):
952-3
69. Finsterer J, Zuntner G. Rhabdomyolysis from simvastatin
triggered by infection and muscle exertion. South Med J
2005; 98 (8): 827-9
70. Betrosian A, Thireos E, Kofinas G, et al. Bacterial sepsis-
induced rhabdomyolysis. Intensive Care Med 1999; 25:
469-74
71. Thompson PD, Zmuda JM, Domalik LJ, et al. Lovastatin
increases exercise-induced skeletal muscle injury. Meta-
bolism 1997; 46: 1206-10
72. Wilhelmi M, Winterhalter M, Fisher S, et al. Massive post-
operative rhabdomyolysis following combined CABG/
abdominal aortic replacement: a possible association with
HMG-CoA reductase inhibitors. Cardiovasc Drugs Ther
2002; 16 (5): 471-5
73. East C, Alivizatos PA, Grundy SM, et al. Rhabdomyolysis
in patients receiving lovastatin after cardiac transplanta-
tion. N Engl J Med 1988; 318 (1): 47-8
74. Schouten O, Kertai MD, Bax JJ, et al. Safety of perio-
perative statin use in high-risk patients undergoing major
vascular surgery. Am J Cardiol 2005; 95 (5): 658-60
75. Antons KA, Williams CD, Baker SK, et al. Clinical per-
spectives of statin-induced rhabdomyolysis. Am J Med
2006; 119: 400-9
76. Davidson MH, Stein EA, Dujovne CA, et al. The efficacy
and six-week tolerability of sim vastatin 80 and
160 mg/day. Am J Cardiol 1997; 257 (79): 38-42
77. Rosenson RS, Bays HE. Results of two clinical trials on the
safety and efficacy of pravastatin 80 and 160 mg per day.
Am J Cardiol 2003; 91: 878-81
78. Masters BA, Palmoski MJ, Flint OP, et al. In vitro myotoxi-
city of the 3-hydroxy-3-methylglutaryl coenzyme A re-
ductase inhibitors, pravastatin, lovastatin and simvastatin,
using neonatal rat skeletal myocytes. Toxicol Appl Phar-
macol 1995; 131: 163-74
79. Ziegler K, Stunkel W. Tissue-selective action of pravastatin
due to hepatocellular uptake via sodium-independent bile
acid transporter. Biochem Biophys Acta 1992; 1139: 203-9
80. Bottorff MB. Statin safety and drug interactions: clinical
implications. Am J Cardiol 2006; 97 Suppl.: 27-31C
81. Lees RS, Lees AM. Rhabdomyolysis from the coadminis-
tration of lovastatin and the antifungal agent itracona-
zole. N Engl J Med 1995; 333: 664-5
82. Neuvonen PJ, Kantola T, Kivisto KT. Simvastatin but not
pravastatin is very susceptible to interaction with the
CYP3A4 inhibitor itraconazole. Clin Pharmacol Ther
1998; 63: 332-4
83. Kantola T, Kivisto KT, Neuvonen PJ. Erythromycin and
verapamil considerably increase serum simvastatin and
simvastatin acid concentrations. Clin Pharmacol Ther
1998; 64: 177-82
84. Lee AJ, Maddix DS. Rhabdomyolysis secondary to drug
interaction between simvastatin and clarithromycin. Ann
Pharmacother 2001; 35: 26-31
85. Mousa O, Brater DC, Sunblad KJ, et al. The interaction of
diltiazem with simvastatin. Clin Pharmacol Ther 2000; 67:
267-74
86. Lewin JJ, Nappi JM, Taylor MH. Rhabdomyolysis with
concurrent atorvastatin and diltiazem. Ann Pharmaco-
ther 2002; 36: 1546-8
87. Penzak SR, Chuck SK, Stajich GV. Safety and efficacy of
HMG-CoA reductase inhibitors for treatment of hyper-
lipidemia in patients with HIV infection. Pharmaco-
therapy 2000; 20: 1066-71
88. Lenhard JM, Croom DK, Weiel JE, et al. HIV protease
inhibitors stimulate hepatic triglyceride synthesis. Arter-
ioscler Thromb Vasc Biol 2000; 20: 2625-9
89. Chuk SK, Penzak SR. Risk-benefit of HMG-CoA re-
ductase inhibitors in the treatment of HIV protease
Statin-Induced Myopathy: Risk Factors and Drug Interactions 185
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
inhibitor-related hyperlipidaemia. Expert Opin Drug Saf
2002; 1 (1): 5-11
90. Gazi IF, Liberopoulos EN, Athyros VG, et al. Statins
and solid organ transplantation. Curr Pharm Des 2006;
12 (36): 4771-83
91. Pichard L, Domergue J, Fourtanier G, et al. Metabolism of
the new immumosuppresor cyclosporin G by human liver
cytochrome P450. Biochem Pharmacol 1996; 51 (5): 591-8
92. Norman DJ, Illingworth DR, Munson J, et al. Myolysis
and acute renal failure in a heart-transplant recipient re-
ceiving lovastatin. N Engl J Med 1988; 318: 46-7
93. Park JW, Siekmeier R, Merz M, et al. Pharmacokinetics
of pravastatin in heart-transplant patients taking
cyclosporin A. Int J Clin Pharmacol Therapeut 2002; 40:
439-50
94. Corsini A, Bellosta S, Baetta R, et al. New insights into the
pharmacodynamic and pharmacokinetic properties of
statins. Pharmacol Ther 1999; 84: 413-28
95. Moguorosi A, Bradley B, Showealter A, et al. Rhabdo-
myolysis and acute renal failure due to combina tion
therapy with simvastatin and warfarin. J Intern Med
1999; 246: 599-602
96. Kantola T, Backman JT, Niemi M, et al. Effect of fluco-
nazole on plasma fluvastatin and pravastatin concentra-
tions. Eur J Clin Pharmacol 2000; 56: 225-9
97. Shek A, Ferrill MJ. Statin-fibrate combination therapy.
Ann Phermacother 2001; 35: 908-17
98. Murdock DK, Murdock AK, Murdock RW, et al. Long-
term safety and efficacy of combination gemfibrozil and
HMG-CoA reductase inhibitors for the treatment of
mixed lipid disorders. Am Heart J 1999; 138: 151-5
99. Jones PH, Davidson MH. Reporting rate of rhabdomyolysis
with fenofibrate+statin versus gemfibrozil+any statin. Am J
Cardiol 2005; 95: 120-2
100. Franssen R, Vergeer M, Stroes ES, et al. Combination
statin-fibrate therapy: safety aspects. Diabetes Obes Me-
tab 2009 Feb; 11 (2): 89-94
101. Graham DJ, Staffa JA, Shatin D, et al. Incidence of hospi-
talized rhabdomyolysis in patients treated with lipid-
lowering drugs. JAMA 2004; 292: 2585-90
102. Prueksaritanont T, Subramanian R, Fang X, et al. Glu-
curonidation of statins in animals and humans: a novel
mechanism of statin lactonization. Drug Metab Dispos
2002; 30: 505-12
103. Prueksaritanont T, Zhao JJ, Ma B, et al. Mechani-
stic studies on metabolic interactions between gemfi
brozil and statins. J Pharmacol Exp Ther 2002; 301:
1042-51
104. Schoonjans K, Steals B, Auwerx J. Role of peroxisome
proliferator activated receptor in mediating effects of
fibrates and fatty acids on gene expression. J Lipid Res
1996; 37: 907-25
105. Kyrklund C, Backman JT, Neuvonen M, et al. Gemfibrozil
increases plasma pravastatin concentrations and reduces
pravastatin renal clearance. Clin Pharmacol Ther 2003;
73: 538-44
106. Reaven P, Witztum JL. Lovastatin, nicotinic acid, and
rhabdomyolysis. Ann Intern Med 1988; 109: 597-8
107. Bays H. Safety of niacin and simvastatin combination
therapy. Am J Cardiol 2008; 101 Suppl.: 3-8B
108. Fux R, Morike K, Gundel UF, et al. Ezetimibe and statin-
associated myopathy. Ann Intern Med 2004; 140: 671-2
109. Simard C, Poirier P. Ezetimibe-associated myopathy in
monotherapy and in combination with a 3-hydroxy-
3-methylglutaryl coenzyme A reductase inhibitor. Can J
Cardiol 2006; 22 (2): 141-4
110. Pearson TA, Denke MA, McBride PE, et al. A community-
based, randomized trial of ezetimibe added to statin
therapy to attain NCEP ATP III goals for LDL choles-
terol in hypercholesterolemic patients: the ezetimibe add-
on to statin for effectiveness (EASE) trial. Mayo Clin Proc
2005 May; 80 (5): 587-95
111. Davidson MH, Maccubbin D, Stepanavage M, et al. Stri-
ated muscle safety of ezetimibe/simvastatin (Vytorin). Am
J Cardiol 2006 Jan 15; 97 (2): 223-8
112. Conard SE, Bays HE, Leiter LA, et al. Efficacy and
safety of ezetimibe added on to atorvastatin (20 mg) ver-
sus uptitration of atorvastatin (to 40 mg) in hyper-
cholesterolemic patients at moderately high risk for
coronary heart disease. Am J Cardiol 2008 Dec 1; 102
(11): 1489-94
113. Leiter LA, Bays H, Conard S, et al. Efficacy and safety of
ezetimibe added on to atorvastatin (40 mg) compared with
uptitration of atorvastatin (to 80 mg) in hypercholester-
olemic patients at high risk of coronary heart disease. Am
J Cardiol 2008; 102: 1495-501
114. Davidson MH, Armani A, McKenney JM, et al. Safety
considerations with fibrate therapy. Am J Cardiol 2007;
99 Suppl.: 3-18C
115. Spence JD, Munoz CE, Hendricks L, et al. Pharmaco-
kinetics of the combination of fluvastatin and gemfibrozil.
Am J Cardiol 1995; 76 Suppl.: 80-3A
116. McKenney JM, Davidson MH, Jacobson TA, et al. Final
conclusions and recommendations of the National Lipid
Association Statin Safety Assessment Task Force. Am J
Cardiol 2006; 97 Suppl.: 89-94C
117. Chatzizisis YS, Misirli G, Hatzitolios AI, et al. The syn-
drome of rhabdomyolysis: complications and treatment.
Eur J Intern Med 2008; 19 (8): 568-74
118. Stein EA, Ballantyne CM, Windler E, et al. Efficacy
and tolerability of fluvastatin XL 80 mg alone, ezeti-
mibe alone, and the combination of fluvastatin XL 80 mg
with
ezetimibe
in patients with a history of muscle-related
side effects with other statins. Am J Cardiol 2008; 101:
490-6
119. Glueck CJ, Aregawi D, Agloria M, et al. Rosuvastatin
5 and 10 mg/d: a pilot study of the effects in hypercholes-
terolemic adults unable to tolerate other statins and reach
LDL cholesterol goals with nonstatin lipid-lowering
therapies. Clin Ther 2006; 28: 933-42
120. Matalka MS, Ravnan MC, Deedwania PC. Is alternate
daily dose of atorvastatin effective in treating patients
with hyperlipidemia? The Alternate Day Versus Daily
Dosing of Atorvastatin Study (ADDAS). Am Heart J
2002; 144: 674-7
121. Juszczyk MA, Seip RL, Thompson PD. Decreasing LDL
cholesterol and medication cost with every-other-day
statin therapy. Prev Cardiol 2005; 8: 197-9
122. Athyros VG, Tziomalos K, Kakafika AI, et al. Effective-
ness of ezetimibe alone or in combination with twice a
186 Chatzizisis et al.
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)
week atorvastatin (10 mg) for statin intolerant high-risk
patients. Am J Cardiol 2008; 101 (4): 483-5
123. Backes JM, Venero CV, Gibson CA, et al. Effectiveness
and tolerability of every-other-day rosuvastatin dosing in
patients with prior statin intolerance. Ann Pharmacother
2008; 42: 341-6
124. Backes JM, Moriarty PM, Ruisinger JF, et al. Effects of
once weekly rosuvastatin among patients with a prior
statin intolerance. Am J Cardiol 2007; 100: 554-5
125. Mackie BD, Satija S, Nell C, et al. Monday, Wednesday,
and Friday dosing of rosuvastatin in patients previously
intolerant to statin therapy [letter]. Am J Cardiol 2007;
99: 291
126. Gadarla M, Kearns A, Thompson PD. Efficacy of
rosuvastatin (5 mg and 10 mg) twice a week in patients
intolerant to daily statins. Am J Cardiol 2008; 10 (12):
1747-8
127. Gazi IF, Daskalopoulou SS, Nair DR, et al. Effect of
ezetimibe in patients who cannot tolerate statins or cannot
get to the low density lipoprotein cholesterol target
despite taking a statin. Curr Med Res Opin 2007; 23:
2183-92
128. Rivers SM, Kane MP, Busch RS, et al. Colesevelam
hydrochloride-ezetimibe combination lipid-lowering
therapy in patients with diabetes or metabolic syndrome
and a history of statin intolerance. Endocr Pract 2007; 13:
11-6
Correspondence: Professor George D. Giannoglou, 1st
Cardiology Department, AHEPA University Hospital,
Aristotle University Medical School, 1 St Kyriakidi Street,
54636 Thessaloniki, Greece.
Statin-Induced Myopathy: Risk Factors and Drug Interactions 187
ª 2010 Adis Data Information BV. All rights reserved. Drug Saf 2010; 33 (3)