ArticlePDF Available

Starting with rosuvastatin in primary hyperlipidemia--Is there more than lipid lowering?

Authors:

Abstract and Figures

The authors investigated the effects of rosuvastatin, beyond its lipid-lowering activity, on several nonlipid metabolic variables, along with its safety and tolerability, in patients treated for primary hyperlipidemia. Patients (n = 55) with primary hyperlipidemia were open-label assigned to the recommended starting dose of rosuvastatin 10 mg/day, and serum metabolic variables were measured at baseline and after 8 and 20 weeks. Treatment with rosuvastatin produced significant reductions in total cholesterol, low-density lipoprotein cholesterol (LDL-C), apolipoprotein B, nonhigh-density lipoprotein cholesterol (non HDL-C), and triglyceride concentrations, whereas HDL-C, apolipoprotein A-I, and lipoprotein(a) levels did not change significantly from baseline. The LDL-C treatment target was achieved in 71% of patients. No significant variations in renal function parameters (serum creatinine and creatinine clearance), insulin resistance estimates, and serum concentrations of uric acid, total homocysteine, vitamin B12, and folic acid were observed during the period of treatment. High-sensitivity C-reactive protein levels were significantly lowered by rosuvastatin therapy (median values, 3.1 vs 2.0 vs 1.9 mg/L, at 0, 8, and 20 weeks, respectively; p < 0.0001). In conclusion, rosuvastatin at 10 mg/day is a highly effective, safe, and well-tolerated monotherapy option for patients with primary hyperlipidemia, with a favorable antiinflammatory potential and nondeteriorating effects on renal function.
Content may be subject to copyright.
http://ang.sagepub.com/
Angiology
http://ang.sagepub.com/content/56/5/585
The online version of this article can be found at:
DOI: 10.1177/000331970505600510
2005 56: 585ANGIOLOGY
Konstantine Seferiadis and Moses S. Elisaf
Haralampos J. Milionis, Irene F. Gazi, Theodosios D. Filippatos, Vasilios Tzovaras, George Chasiotis, John Goudevenos,
: Is There More Than Lipid Lowering?−−Starting with Rosuvastatin in Primary Hyperlipidemia
Published by:
http://www.sagepublications.com
can be found at:AngiologyAdditional services and information for
http://ang.sagepub.com/cgi/alertsEmail Alerts:
http://ang.sagepub.com/subscriptionsSubscriptions:
http://www.sagepub.com/journalsReprints.navReprints:
http://www.sagepub.com/journalsPermissions.navPermissions:
http://ang.sagepub.com/content/56/5/585.refs.htmlCitations:
What is This?
- Sep 1, 2005Version of Record >>
at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from at Obafemi Awolowo University on October 11, 2013ang.sagepub.comDownloaded from
Starting with Rosuvastatin in Primary
Hyperlipidemia
Is There More Than Lipid Lowering?
Haralampos J. Milionis, MD,* Irene F. Gazi, MD,* Theodosios D. Filippatos, MD,*
Vasilios Tzovaras, MD,* George Chasiotis, PhD,
John Goudevenos, MD,*
Konstantine Seferiadis, PhD,
and Moses S. Elisaf, MD, FRSH, FASA,*
Ioannina, Greece
The authors investigated the effects of rosuvastatin, beyond its lipid-lowering activity, on
several nonlipid metabolic variables, along with its safety and tolerability, in patients treated
for primary hyperlipidemia. Patients (n = 55) with primary hyperlipidemia were open-label
assigned to the recommended starting dose of rosuvastatin 10 mg/day, and serum metabolic
variables were measured at baseline and after 8 and 20 weeks. Treatment with rosuvastatin
produced significant reductions in total cholesterol, low-density lipoprotein cholesterol (LDL-C),
apolipoprotein B, nonhigh-density lipoprotein cholesterol (non HDL-C), and triglyceride concen-
trations, whereas HDL-C, apolipoprotein A-I, and lipoprotein(a) levels did not change signifi-
cantly from baseline. The LDL-C treatment target was achieved in 71% of patients. No signif-
icant variations in renal function parameters (serum creatinine and creatinine clearance), insulin
resistance estimates, and serum concentrations of uric acid, total homocysteine, vitamin B12,
and folic acid were observed during the period of treatment. High-sensitivity C-reactive protein
levels were significantly lowered by rosuvastatin therapy (median values, 3.1 vs 2.0 vs 1.9
mg/L, at 0, 8, and 20 weeks, respectively; p <0.0001). In conclusion, rosuvastatin at 10 mg/day
is a highly effective, safe, and well-tolerated monotherapy option for patients with primary
hyperlipidemia, with a favorable antiinflammatory potential and nondeteriorating effects on
renal function.
Introduction
Rosuvastatin, a 3-hydroxy-3-methylglutaryl coen-
zyme A reductase inhibitor (statin), in its current-
ly recommended start dose of 10 mg/day, is con-
sidered highly effective in minimizing the need for
titrations to higher doses in order to achieve low-
density lipoprotein cholesterol (LDL-C) treatment
goals, thus reducing the frequency of medical con-
tacts.
1-3
There is less information with respect to
the actions of rosuvastatin on nonlipid variables,
which are associated with an increased risk of car-
Angiology 56:585–592, 2005
From the *Department of Internal Medicine, School of
Medicine, University of Ioannina,
Laboratory of Biochemistry,
University Hospital of Ioannina, Ioannina, Greece
Correspondence: Moses S. Elisaf, MD, Professor of Medicine,
Department of Internal Medicine, School of Medicine,
University of Ioannina, 451 10 Ioannina, Greece
Email: hmilioni@cc.uoi.gr
©2005 Westminster Publications, Inc, 708 Glen Cove Avenue,
Glen Head, NY 11545, USA
585
Angiology Volume 56, Number 5, 2005
diovascular disease, such as indices of renal func-
tion, glycemic status, serum uric acid concentra-
tions, and high-sensitivity CRP (hsCRP), as well as
serum total homocysteine (tHcy), folate, and vita-
min B12 concentrations. In this study, we assessed
the effects of rosuvastatin (at the recommended
start dose of 10 mg/day) on several serum meta-
bolic variables, beyond its lipid-lowering capaci-
ty, along with its safety and tolerability in patients
treated for primary hyperlipidemia.
Subjects and Methods
Patients attending the Outpatient Lipid Clinic of
the University Hospital of Ioannina, Greece, were
recruited for the study. Men and women with
dyslipidemia were screened for eligibility by med-
ical history, physical examination, and clinical
laboratory evaluation, including the lipid profile.
No participant had either symptomatic ischemic
heart disease or any other vascular disease. Any
lipid-lowering medications had to have been dis-
continued for at least 6 weeks before enrollment.
All participants gave their informed consent and
the study protocol was approved by the institu-
tional ethics committee.
Patients were assigned to the National
Cholesterol Education Program (NCEP) diet for 6
weeks and were advised to follow this diet
throughout the study.
4
Fasting serum lipid con-
centration entry criteria were total cholesterol
(TC) >240 mg/dL (>6.2 mmol/L) at weeks –4
and –2, and triglycerides (TG) <350 mg/dL (4.0
mmol/L).
Patients with (1) abnormal hepatic function
(aminotransferase levels >2 times the upper limit
of normal, and/or history of chronic liver disease,
such as cirrhosis or alcohol abuse); (2) impaired
renal function (serum creatinine concentrations
>1.8 mg/dL, 159 µmol/L); (3) diabetes mellitus
(fasting blood glucose >126 mg/dL, 7.0 mmol/L);
(4) raised TSH levels (greater than 5.0 µU/L);
and (5) any medical conditions that might pre-
clude successful completion of the study protocol
were excluded.
Patients taking drugs such as angiotensin-
converting enzyme inhibitors, angiotensin II re-
ceptor antagonists, or calcium channel blockers
on a stabilized dose for at least 8 weeks before
entry to the study were considered eligible. Those
receiving drugs possibly affecting the laboratory
parameters tested were also excluded.
5,6
After a mean 6-week baseline period the pa-
tients were assigned to rosuvastatin 10 mg daily
for 20 weeks. A food record rating score was cal-
culated from 3-day diaries kept by the partici-
pants in order to assess compliance with the diet
throughout the study. Blood samples were taken
after a 14-hour overnight fast for the determina-
tion of serum lipid concentrations (including
apolipoproteins), urea, creatinine, glucose, in-
sulin, uric acid, hs-CRP, tHcy, vitamin B12, and
folic acid, as well as liver and muscle enzymes at
baseline and at 8 and 20 weeks following treat-
ment initiation.
Laboratory Determinations
The laboratory determinations were performed
by use of an Olympus AU 600 analyzer as previ-
ously described.
5,6
The creatinine clearance was
estimated by the Cockcroft-Gault formula:
Creatinine clearance (CrCl) = ([140-age] xbody
weight)/(72 xserum creatinine), where body
weight is in kg, age is in years, and serum creati-
nine is in mg/dL. For women, the result is multi-
plied by a factor of 0.85 to compensate for the
lower average muscle mass.
7
Glucose was measured by the hexokinase
method and serum insulin concentrations by the
AxSYM Insulin assay, which is based on the
Microparticle Enzyme Immunoassay technology
(Abbott Laboratories, Diagnostic Division, USA).
The HOmeostasis Model Assessment (HOMA)
was used to predict the homeostatic concentra-
tions of fasting insulin and glucose, which arise
from varying degrees of beta-cell deficiency and
insulin resistance.
8
The model is known to be
nonlinear but can be approximated by simple
mathematical approximations: insulin resistance
= (fasting insulin in µU/mL) x(fasting serum
glucose in mg/dL) x0.06/405.
9
Serum concentrations of hsCRP were mea-
sured by the N High-Sensitivity CRP method,
which is an in vitro diagnostic assay (provided
by Dade Behring Marburg GmbH, Marburg,
Germany) based on particle-enhanced immuno-
nephelometry. The reference range of this assay
is 0.175–55 mg/L.
5,6
The AxSYM Homocysteine assay, a fluores-
cence polarization immunoassay (Axis-Shield,
Oslo Norway, Abbott Laboratories), was used for
the quantitative measurement of total L-homo-
cysteine in human serum. Serum folate was mea-
sured by AxSYM Folate, an ion capture assay
(Abbott Laboratories, Illinois, USA). A chemilu-
minescent microparticle intrinsic factor assay was
Angiology Volume 56, Number 5, 2005
586
used for the quantitative determination of vita-
min B12 in human serum (The ARCHITECT B12
assay, Abbott Laboratories, Illinois, USA).
5,6
The concentrations of TC and TG were deter-
mined enzymatically on the Olympus AU 600
clinical chemistry analyzer (Olympus Diagnostica,
Hamburg, Germany). High-density lipoprotein
cholesterol (HDL-C) was determined in the su-
pernatant, after precipitation of the ApoB-con-
taining lipoproteins with dextran sulfate–Mg
2+
(Sigma Diagnostics, St. Louis, MO, USA). LDL-C
was calculated by using the Friedewald formula.
Apolipoproteins (Apo) A-I and B were measured
with a Behring Nephelometer BN100, and re-
agents (antibodies and calibrators) from Dade
Behring Holding GmbH (Liederbach, Germany).
The ApoA-I and ApoB assays were calibrated ac-
cording to the International Federation of Clinical
Chemistry (IFCC) standards. Lipoprotein(a)
[Lp(a)] levels were determined by means of par-
ticle-enhanced immunonephelometry using a
Behring Nephelometer BN100 and N Latex Lp(a)
Reagent from Dade Behring Marburg GmbH
(Liederbach, Germany). The lower limit of the
assay was 2.4 mg/dL (0.024 g/L). Liver and mus-
cle enzymes were measured by use of conven-
tional methods.
Statistical Analysis
All parameters were expressed as mean values ±
SD, except for Lp(a) and hsCRP, which were ex-
pressed as median values (range). Relationships
between variables were assessed by the Pearson’s
correlation coefficient, and Spearman correlation
coefficients for parametric and nonparametric
variables, respectively. Analysis of variance
(ANOVA) for repeated measurements was used
to assess post-rosuvastatin treatment changes in
parametric variables and Friedman’s ANOVA for
nonparametric variables as appropriate.
Significance was defined as p<0.05. Analyses
were performed using the SPSS 11.0 statistical
package for Windows (SPSS Inc, 1989–2001),
and Statistica 6.0 (Statsoft, Inc, 1984–2001).
Results
In total, 55 patients were included in the study
(29 men and 26 women) with a mean age of 52.5
±13.7 years and a body mass index of 25.8 ±4.6
kg/m
2
.
At baseline, serum tHcy concentrations corre-
lated positively with serum creatinine concentra-
tions (r=0.36, p=0.071), and inversely with
serum folic acid concentrations (r=–0.53,
p=0.010) and vitamin B12 levels (r=–0.49,
p=0.012).
Rosuvastatin therapy resulted in a significant
reduction in TC (by 37%), LDL-C (by 46.5%),
Apo B (by 35%), non HDL-C (ie, TC minus HDL-
C, by 44.2%), and TG (by 25%). HDL-C, Apo A-I,
and Lp(a) levels did not change significantly from
baseline following statin therapy (Table I). In
total, 39 of 55 patients (71%) achieved the LDL-
C treatment target of less than 130 mg/dL (3.4
mmol/L).
Glycemic parameters (fasting glucose, insulin,
HOMA index) and serum uric acid concentrations
were not significantly influenced by rosuvastatin
treatment (Table I).
Statin treatment produced no significant
changes in serum creatinine concentrations as
well as in the calculated creatinine clearance
(CrCl) measurements. (Figure 1, Table I). A sub-
group analysis of patients with baseline CrCl
equal to 70 mL/minute or less (n=15) revealed
no significant variations in serum creatinine (0.93
±0.15 vs 0.98 ±0.17 vs 0.92 ±0.16 mg/dL; 82
±13 vs 87 ±15 vs 81 ±14 µmol/L, p=NS) or
CrCl (61 ±8 vs 58 ±9 vs 59 ±7 mL/minute, p=
NS) following rosuvastatin therapy.
Rosuvastatin treatment resulted in a signifi-
cant reduction of serum hsCRP levels by 38.7%
(Table I).
Serum concentrations of tHcy were not mod-
ified in patients receiving rosuvastatin, and this
was also noted for serum folate and vitamin B12
values (Table I). There were no correlations be-
tween “posttreatment minus baseline” changes of
serum tHcy (DtHcy) concentrations and changes
in serum lipid concentrations, and between DtHcy
and changes in serum creatinine and/or creati-
nine clearance estimates. Finally, posttreatment
tHcy did not correlate with serum creatinine, fo-
late, or vitamin B12 concentrations after 20
weeks on rosuvastatin 10 mg/day.
Safety and Tolerability of Rosuvastatin
Treatment
Rosuvastatin treatment (10 mg/day) was gener-
ally well tolerated. All participants completed the
study protocol. Clinically significant elevations in
alanine aminotransferase (ALT, values >3 times
the upper limit of normal on 2 consecutive occa-
sions) or in creatine kinase (CK) levels (values
Milionis Rosuvastatin in Primary Hyperlipidemia
587
Angiology Volume 56, Number 5, 2005
588
Table I. Serum parameters at baseline and after 8 and 20 weeks of treatment with
rosuvastatin 10 mg/day (n=55)
Variable Baseline Posttreatment p
8 weeks 20 weeks
Glucose, mg/dL 98 ±19 100 ±16098 ±18 NS
Insulin, µU/mL 9.6 ±4.7 9.6 ±4.2 9.5 ±3.1 NS
HOMA index 2.7 ±1.5 2.7 ±1.8 2.6 ±1.2 NS
TC, mg/dL 295 ±420186 ±260188 ±320<0.0001
LDL-C, mg/dL 213 ±420111 ±260114 ±300<0.0010
TG, mg/dL 148 ±690113 ±400111 ±370<0.0001
HDL-C, mg/dL 53 ±8053 ±8054 ±80NS
non-HDL-C, mg/dL 242 ±430133 ±230135 ±300<0.0001
Apo A-I, mg/dL 132 ±310132 ±330135 ±210NS
Apo B, mg/dL 122 ±36077 ±22 80 ±24 <0.0001
Lp(a), mg/dL 18 (2–131) 17 (2–119) 17 (2–116) *NS*
Creatinine, mg/dL 0.88 ±0.13 0.88 ±0.17 0.86 ±0.13 NS
CrCl, mL/minute 89 ±26 89 ±27 89 ±26 NS
Uric acid, mg/dL 5.0 ±1.3 5.0 ±1.4 4.9 ±1.2 NS
hsCRP, mg/L .3.1 (0.7–7.2) .2.0 (0.9–5.0) .1.9 (0.8–4.4) *<0.0001*
tHcy, µU/L 10.2 ±3.2010.1 ±3.0010.2 ±3.40NS
Vitamin B12, pg/mL 340 ±133 342 ±141 339 ±122 NS
Folate, ng/mL 6.3 ±2.5 6.4 ±2.1 6.3 ±2.0 NS
*Friedman’s ANOVA
To convert values for glucose to mmol/L, multiply by 0.05551. To convert values for insulin
to pmol/L, multiply by 6.945. To convert TC, LDL-C, non HDL-C, and HDL-C levels from
mg/dL to mmol/dL, multiply by 0.02586. To convert TG levels from mg/dL to mmol/L
multiply by 0.01129. To convert values for creatinine to mmol/L, multiply by 88.40. To
convert serum uric acid levels from mg/dL to µmol/L, multiply by 59.48.
HOMA: homeostasis model assessment; TC: total cholesterol; LDL-C: low-density lipoprotein
cholesterol; TG: triglycerides; HDL-C: high-density lipoprotein cholesterol; Apo: apolipopro-
tein; Lp(a): lipoprotein(a); CrCl: creatinine clearance; hsCRP: high-sensitivity C-reactive
protein; tHcy: total homocysteine; NS: nonsignificant.
>10 times the upper limit of normal) were not
observed during follow up.
One female patient reported muscle pain in
both upper limbs 3 days after rosuvastatin was
initiated. Physical examination was unremark-
able, and laboratory evaluation, including deter-
minations of aspartate aminotransferase (AST),
ALT, CK, lactic dehydrogenase (LDH), and al-
dolase levels, were within normal limits. Treat-
ment was not discontinued and the patient’s
symptoms resolved after 6 days.
Discussion
The study confirmed the efficacy of rosuvastatin
in improving the lipid profile (TC, LDL-C, Apo B,
non HDL-C, and TG reduction) in patients with
primary hyperlipidemia.
1-3
It is notable that a sig-
nificant proportion of patients (71%) achieved
the LDL-C treatment goal with rosuvastatin 10
mg/day. On the other hand, there was no signifi-
cant effect on serum HDL-C, Apo A-I, and Lp(a)
concentrations. This is in contrast to a previously
reported HDL-C-raising effect with rosuvastatin;
this difference could be attributed to the relative-
ly high pretreatment HDL-C levels.
1-3,10
Glycemic parameters and insulin resistance
estimates were not significantly altered by rosu-
vastatin. There are some reports suggesting that
statins can improve insulin resistance and de-
crease the long-term incidence of diabetes melli-
tus in dyslipidemic patients.
11,12
However, short-
term rosuvastatin treatment (10 mg/day for 20
weeks) did not seem to have any impact on in-
Milionis Rosuvastatin in Primary Hyperlipidemia
589
Figure 1.
Changes in serum creatinine
concentrations (A), and
creatinine clearance (B) at
baseline and after 8 and 20
weeks of treatment with
rosuvastatin 10 mg/day in the
study population (n=55).
A
B
sulin sensitivity in nondiabetic subjects with dys-
lipidemia. This finding is in agreement with re-
cently published data demonstrating that rosu-
vastatin does not enhance insulin sensitivity or
lower day-long glucose and insulin concentra-
tions in insulin-resistant, nondiabetic subjects
with combined hyperlipidemia.
13
No significant variations in renal function pa-
rameters (ie, serum creatinine and creatinine
clearance) were observed during the treatment
period. Dipstick-positive proteinuria has been de-
scribed in some patients who received 80 mg/day
of rosuvastatin in phase III trials.
14
However, fur-
ther assessment of the clinical program safety
database failed to show that this mild proteinuria
was predictive of any acute or progressive change
in renal function or that rosuvastatin therapy was
associated with deteriorating renal function.
14,15
The absence of change in renal function after a
5-month period of rosuvastatin therapy in our
study is in line with the findings of Vidt et al
15
in-
dicating an arrest in progression of renal disease
in dyslipidemic patients receiving rosuvastatin. In
terms of safety, it appears that the start dose (10
mg/day) of rosuvastatin does not have any im-
pact on kidney function in patients with primary
hyperlipidemia. Nevertheless, our results must be
interpreted within the context of certain limita-
tions, such as the short duration of our study (20
weeks) and lack of testing for hematuria or mi-
croalbuminuria.
There is evidence that statin therapy exerts
beneficial effects in patients with different types
of renal disease.
16
These effects include improve-
ment in renal function, decreased microalbumin-
uria, and a fall in blood pressure.
16-19
The under-
lying mechanism remains undefined and may in-
volve an increase in renal blood flow following
the lipid-lowering effect, which in turn may be
due to an increased production of nitric oxide
(NO).
20
A subgroup analysis in the Heart Protec-
tion Study showed that simvastatin significantly
reduced the fall in glomerular filtration rate in
high-risk patients with and without diabetes mel-
litus.
21
Atorvastatin shares this favorable effect as
shown in smaller as well as larger studies, in-
cluding the GREek Atorvastatin and Coronary
heart disease Evaluation (GREACE) Study.
22,23
The favorable effects on renal function with other
statins not seen with rosuvastatin 10 mg/day may
be ascribed to the administration of higher doses
of these agents and/or to patients with higher
levels of serum creatinine. Moreover, the “nonag-
gravating” effect of rosuvastatin on renal function
as shown in our study, favors cardiovascular dis-
ease protection, since there is evidence that ele-
vated serum creatinine concentrations, even with-
in normal range, are strong and independent pre-
dictors of coronary heart disease adverse out-
comes and stroke in high-risk patients.
24
Serum uric acid concentrations did not sig-
nificantly change from baseline after rosuvastatin
treatment. The pathophysiological mechanisms
underlying the association of serum uric acid with
vascular disease and all-cause mortality are not
fully clarified.
25
There is evidence that atorvas-
tatin may exert a hypouricemic effect, but this has
not been described with other statins.
6,26
Whether
higher doses of rosuvastatin affect serum urate
concentrations could be the subject of further in-
vestigation.
Rosuvastatin therapy had a neutral effect on
Hcy. Prospective and retrospective studies have
shown that elevated plasma concentrations of
tHcy are associated with a high prevalence of
cardiovascular disease, independently of con-
ventional risk factors.
5,27
Hcy is an intermediate
amino acid resulting from the metabolism of me-
thionine and is known to be toxic to vascular en-
dothelium.
5,27
tHcy concentrations are mainly de-
termined by folate and cobalamin status and by
renal function, which was also evident in our
study.
5,28
Pharmacologic agents, including lipid-
lowering drugs, may influence tHcy levels.
5,28,29
Fibrates have been shown to raise tHcy levels,
and this has been partly attributed to the impair-
ment of renal function,
5,30,31
whereas statin ther-
apy appears to exert a neutral effect on tHcy lev-
els, with the exception of high-dose simvastatin
(80 mg/day), which has been reported to lower
tHcy plasma concentrations.
5,32-36
In agreement
with our findings, in a placebo-controlled study
of 46 hypercholesterolemic patients, rosuvastatin
at 10 mg/day did not significantly affect tHcy
levels.
37
The lack of any influence on serum folic
acid and vitamin B12 levels together with the
nondeteriorating effect on renal function para-
meters may account for the “neutral” effect of ro-
suvastatin on tHcy.
A substantial decrease in hsCRP levels, which
are considered strong predictors of ischemic heart
disease and the severity of atherosclerosis, was
noted following statin treatment.
38
This CRP-low-
ering effect seems to be a shared property among
statins.
6,39
It has been shown that patients who
have low CRP levels after statin therapy have bet-
ter clinical outcomes than those with higher CRP
levels, regardless of the resultant level of LDL-C.
40
Moreover, in patients with coronary artery dis-
ease, intensive statin treatment leading to greater
Angiology Volume 56, Number 5, 2005
590
Homeostasis model assessment: Insulin resistance
and beta-cell function from fasting plasma glucose
and insulin concentrations in man. Diabetologia
28:412-419, 1985.
10. Schuster H, Barter PJ, Stender S, et al, and the
Effective Reductions in Cholesterol Using Rosuvastatin
Therapy I study group: Effects of switching statins on
achievement of lipid goals: Measuring Effective
Reductions in Cholesterol Using Rosuvastatin
Therapy (MERCURY I) study. Am Heart J 147:705-
713, 2004.
11. Paolisso G, Barbagallo M, Petrella G, et al: Effects of
simvastatin and atorvastatin administration on in-
sulin resistance and respiratory quotient in aged dys-
lipidemic non-insulin dependent diabetic patients.
Atherosclerosis 150:121–127, 2000.
12. Freeman DJ, Norrie J, Sattar N, et al: Pravastatin and
the development of diabetes mellitus. Evidence for a
protective treatment effect in the West of Scotland
Coronary Prevention Study. Circulation 103:357-362,
2001.
13. Lamendola C, Abbasi F, Chu JW, et al: Comparative
effects of rosuvastatin and gemfibrozil on glucose, in-
sulin, and lipid metabolism in insulin-resistant, non-
diabetic patients with combined hyperlipidemia. Am
J Cardiol 95:189-193, 2005.
14. Shepherd J, Hunninghake DB, Stein EA, et al: Safety
of rosuvastatin. Am J Cardiol 94:882-888, 2004.
15. Vidt DG, Cressman MD, Harris S, et al: Rosuvastatin-
induced arrest in progression of renal disease.
Cardiology 102:52-60, 2004.
16. Elisaf M, Mikhailidis DP: Statins and renal function.
Angiology 53:493-502, 2002.
17. Tsiara S, Elisaf M, Mikhailidis DP: Early vascular ben-
efits of statin therapy. Curr Med Res Opin 19:540-
556, 2003.
18. Kakafika A, Liamis G, Elisaf M, et al: Effect of ator-
vastatin on serum creatinine levels. Curr Med Res
Opin 17:230-231, 2001.
19. Youssef F, Gupta P, Seifalian AM, et al: The effect of
short-term treatment with simvastatin on renal func-
tion in patients with peripheral arterial disease.
Angiology 55:53-62, 2004.
20. Anderson TJ, Meredith IT, Yeung AC, et al: The ef-
fects of cholesterol-lowering and antioxidant therapy
on endothelium-dependent coronary vasodilation. N
Engl J Med 332:488-493, 1995.
21. Heart Protection Study Collaborative Group: MRC/
BHF Heart Protection Study of cholesterol-lowering
with simvastatin in 5,963 people with diabetes: A
randomised placebo-controlled trial. Lancet 361:
2005–2016, 2003.
22. Athyros VG, Papageorgiou AA, Elisaf M, et al, on be-
half of the GREACE Study Collaborative Group:
Statins and renal function in patients with diabetes
mellitus. Curr Med Res Opin 19:615-617, 2003.
23. Athyros VG, Mikhailidis DP, Papageorgiou AA, et al:
The effect of statins versus untreated dyslipidaemia
on renal function in patients with coronary heart dis-
ease. A subgroup analysis of the Greek atorvastatin
reductions in the levels of both the atherogenic
lipoproteins and CRP is associated with a greater
reduction in the rate of progression of athero-
sclerosis as compared with moderate statin treat-
ment.
41
Even at the starting dose of 10 mg/day,
rosuvastatin constitutes a potent lipid-lowering
and antiinflammatory therapeutic regimen.
In conclusion, rosuvastatin at the recom-
mended starting dose of 10 mg/day appears to be
a highly effective, safe, and well-tolerated mono-
therapy option for patients with primary hyper-
lipidemia. Its “neutral” effects on renal function
indices, glycemic control, and other “biomarkers,”
including serum uric acid and homocysteine, as
well as a CRP-related potential antiinflammatory
action offer considerable advantages.
REFERENCES
1. Scott LJ, Curran MP, Figgitt DP: Rosuvastatin: A re-
view of its use in the management of dyslipidemia.
Am J Cardiovasc Drugs 4:117-138, 2004.
2. Jones PH, Davidson MH, Stein EA, et al, and the
STELLAR Study Group: Comparison of the efficacy
and safety of rosuvastatin versus atorvastatin, sim-
vastatin, and pravastatin across doses (STELLAR
Trial). Am J Cardiol 92:152-160, 2003.
3. McKenney JM, Jones PH, Adamczyk MA, et al, for the
STELLAR Study Group: Comparison of the efficacy of
rosuvastatin versus atorvastatin, simvastatin, and
pravastatin in achieving lipid goals: Results from the
STELLAR trial. Curr Med Res Opin 19:689-698,
2003.
4. Executive summary of the third report of the Nation-
al Cholesterol Education Program (NCEP) Expert
Panel on Detection, Evaluation, and Treatment of
High Blood Cholesterol in Adults (Adult Treatment
Panel III). JAMA 285:2486-2497, 2001.
5. Milionis HJ, Papakostas J, Kakafika A, et al:
Comparative effects of atorvastatin, simvastatin and
fenofibrate on serum homocysteine levels in patients
with primary hyperlipidemia. J Clin Pharmacol
43:825-830, 2003.
6. Milionis HJ, Kakafika AI, Tsouli SG, et al: Effects of
statin treatment on uric acid homeostasis in patients
with primary hyperlipidemia. Am Heart J 148:635-
640, 2004.
7. Cockroft DW, Gault MH: Prediction of creatinine
clearance from serum creatinine. Nephron 16:31-39,
1976.
8. Levy JC, Matthews DR, Hermans MP: Correct home-
ostasis model assessment (HOMA) evaluation uses
the computer program. Diabetes Care 21:2191-2192,
1998.
9. Matthews DR, Hosker JP, Rudenski AS, et al:
Milionis Rosuvastatin in Primary Hyperlipidemia
591
and coronary heart disease evaluation (GREACE)
study. J Clin Pathol 57:728-734, 2004.
24. Kaplan RC, Heckbert SR, Furberg CD, et al: Predic-
tors of subsequent coronary events, stroke, and death
among survivors of first hospitalized myocardial in-
farction. J Clin Epidemiol 55:654-664, 2002.
25. Ginsberg MH, Kozin F, O’Malley M, et al: Release of
platelet constituents by monosodium urate crystals. J
Clin Invest 60:999-1007, 1977.
26. Athyros VG, Elisaf M, Papageorgiou AA, et al, for the
GREACE Study Collaborative Group: Effect of statins
versus untreated dyslipidaemia on serum uric acid
levels in patients with coronary heart disease: A sub-
group analysis of the GREek Atorvastatin and
Coronary-heart-disease Evaluation (GREACE) Study.
Am J Kidney Dis 43:589-599, 2004.
27. Mangoni AA, Jackson SHD: Homocysteine and car-
diovascular disease: Current evidence and future
prospects. Am J Med 112:556-565, 2002.
28. Boushey CJ, Beresfors SAA, Omenn GS, et al: A quan-
titative assessment of plasma homocysteine as a risk
factor of vascular disease: Probable benefits of in-
creasing folic acid intakes. JAMA 274:1049-1057,
1995.
29. van Guldener C, Stehouwer CD: Homocysteine-low-
ering treatment: An overview. Expert Opin Pharma-
cother 2:1449-1460, 2001.
30. Westphal S, Dierkes J, Luley C: Effects of fenofibrate
and gemfibrozil on plasma homocysteine. Lancet
358:39-40, 2001.
31. Stulc T, Melenovsky V, Grauova B, et al: Folate sup-
plementation prevents plasma homocysteine increase
after fenofibrate therapy. Nutrition 17:721-723,
2001.
32. Luftjohann D, Sigit JI, Locatelli S, et al: High-dose
simvastatin (80 mg:day) decreases plasma concen-
trations of total homocyst(e)ine in patients with hy-
percholesterolemia. Atherosclerosis 155:265-266,
2001.
33. MacMahon M, Kirkpatrick C, Cummings CE, et al: A
pilot study with simvastatin and folic acid/vitamin
B12 in preparation for the Study of the Effectiveness
of Additional Reductions in Cholesterol and Homo-
cysteine (SEARCH). Nutr Metab Cardiovasc Dis 10:
195-203, 2000.
34. Giral P, Bruckert E, Jacob N, et al: Homocysteine and
lipid lowering agents. A comparison between ator-
vastatin and fenofibrate in patients with mixed hy-
perlipidemia. Atherosclerosis 154:421–427, 2001.
35.Malik J, Melenovsky V, Wichterle D, et al: Both
fenofibrate and atorvastatin improve vascular reac-
tivity in combined hyperlipidaemia (fenofibrate ver-
sus atorvastatin trial—FAT). Cardiovasc Res 52:290-
298, 2001.
36. Melenovsky V, Malik J, Wichterle D, et al: Compari-
son of the effects of atorvastatin or fenofibrate on
nonlipid biochemical risk factors and the LDL particle
size in subjects with combined hyperlipidemia. Am
Heart J 144:e6, 2002.
37. Lu TM, Ding YA, Leu HB, et al: Effect of rosuvastatin
on plasma levels of asymmetric dimethylarginine in
patients with hypercholesterolemia. Am J Cardiol
94:157-161, 2004.
38. Ridker PM, Cushman M, Stampfer MJ, et al: Inflam-
mation, aspirin and risk of cardiovascular disease in
apparently healthy men. N Engl J Med 336:973-979,
1997.
39. Chan KY, Boucher ES, Gandhi PJ, et al: HMG-CoA re-
ductase inhibitors for lowering elevated levels of C-
reactive protein. Am J Health Syst Pharm 61:1676-
1681, 2004.
40.Ridker PM, Cannon CP, Morrow D, et al, and the
Pravastatin or Atorvastatin Evaluation and Infection
Therapy-Thrombolysis in Myocardial Infarction 22
(PROVE IT-TIMI 22) Investigators: C-reactive protein
levels and outcomes after statin therapy. N Engl J
Med 352:20-28, 2005.
41. Nissen SE, Tuzcu EM, Schoenhagen P, et al, and the
Reversal of Atherosclerosis with Aggressive Lipid
Lowering (REVERSAL) Investigators: Statin therapy,
LDL cholesterol, C-reactive protein, and coronary
artery disease. N Engl J Med 352:29-38, 2005.
Angiology Volume 56, Number 5, 2005
592
... Chemical analysis of RSV is highly important in various stages of pharmaceutical development including assay of drug in dosage forms, stability, and bioequivalence samples. [2,3] However, detection of RSV in analytical methods is associated with many challenges with respect to its poor aqueous solubility and high lipophilicity, thus requires a suitable solvent system for its solubilization, retention, and elution from the stationary phase. [4] Based on these physiochemical characteristics, reversed-phase liquid chromatographic method is reported to be suitable. ...
Article
Full-text available
In the present work, we investigated the development of a bioanalytical HPLC method of rosuvastatin (RSV) calcium as per the Quality by Design (QbD)-based systematic chemometric tools. At first, the method objectives were framed and critical analytical attributes (CAAs) were chosen. Risk assessment and factor screening was performed using Hybrid Risk Matrix and Plackett–Burman design for identifying vital factors influencing the critical method parameters (CMPs). Monte-Carlo simulation analysis was conducted which confirmed excellent process robustness (Ppk >1.33) for the studied ranges of CMPs. Furthermore, systematic method development was carried out using custom experimental design, where mobile phase ratio, pH, and injection volume were taken as CMPs at three levels. The obtained trials were evaluated for peak area, retention time, theoretical plates, and peak tailing as CAAs. Mathematical response surface modeling was carried out and optimal chromatographic solution was identified using response optimizer plots. Method transfer was made to bioanalytical scale for estimation of the analyte in rat plasma samples. Extensive method validation was performed as per the ICH Q2 guideline, which indicated validation parameters within the acceptable limits. Overall, the studies construed successful development of QbD compliant HPLC method of rosuvastatin with potential utility bioanalytical testing.
... Rosuvastatin calcium (RSV) is an antilipidemic agent which competitively inhibits hydroxy-methylglutarylcoenzyme A and inhibits synthesis of cholesterol [1][2][3]. Among the several statins available for clinical management of hyperlipidemia, RSV is considered to be well tolerated in all age groups in the dose of 10-80 mg to reduce the total blood cholesterol and low-density lipids (LDL) [4,5]. ...
... If potent statin therapy does indeed decrease IL-1b production, and IL-b acts on the liver to increase homocysteine levels, it would be reasonable to expect that statin therapy that is sufficiently potent could decrease homocysteine. Indeed, 2 clinical trials and a rat study document this effect with rosuvastatin [67][68][69] -albeit other studies do not, possibly owing to suboptimal dose [70,71]. ...
... With respect to their impact on IS, previous reports of rosuvastatin showed no effect [15][16][17] or even worsening of IR [18]. Atorvastatin was neutral in our study which is consistent with most previous reports [8,19], although some demonstrated amelioration [20] or worsening of IR [21]. ...
Article
The impact of statins on glucose metabolism and adipokines remains controversial. We compared the effects of rosuvastatin and atorvastatin on glucose homeostasis, insulin sensitivity (IS), adiponectin and leptin levels as well as systemic inflammation in non-diabetic patients with dyslipidaemia. Thirty-six patients were randomly assigned to 10 mg/day of rosuvastatin (n = 18) or 20 mg/day of atorvastatin (n = 18) for 12 weeks. Total cholesterol (TC), low-density lipoprotein cholesterol (LDL-C), high-density lipoprotein cholesterol (HDL-C), non-HDL-C, triglycerides (TG), fasting plasma glucose, insulin, homeostasis model assessment-insulin resistance (HOMA-IR), quantitative IS check index (QUICKI), adiponectin, leptin and high-sensitivity C-reactive protein (hsCRP) were measured at baseline and after 4 and 12 weeks. Both statins significantly lowered TC, LDL-C, non-HDL-C and TG compared with baseline. Only rosuvastatin caused a significant reduction in insulin and HOMA-IR levels (-35%, p = 0.005 and -33%, p = 0.011 respectively) and a significant increase in QUICKI (+11%, p = 0.003) at 12 weeks. In terms of adipokines and hsCRP, no difference was observed after 4 and 12 weeks of treatment with either statin. Rosuvastatin compared with atorvastatin resulted in significant improvements in IS indices. No significant changes in adiponectin, leptin or hsCRP levels were observed at 4 and 12 weeks of treatment with either statin.
Article
Full-text available
Purpose: Limited data are available linking breakfast consumption to Heart Burn Syndrome (HBS). This study was done to investigate to find whether breakfast consumption is associated with HBS. This cross-sectional study was done to investigate the association between breakfast consumption and HBS among Iranian adults. Methods: This cross-sectional study was performed among 4763 general adults of Isfahan, Iran. Participants' patterns of breakfast eating were assessed by asking two questions from them. How often do you eat breakfast in a week?" Participants were able to respond as: "never or 1 day/wk", "2-4 days/wk", "5-6 days/wk", "every day". HBS was defined as the presence of HBS at sometimes, often or always using a Persian version of validated self-administered modified ROME III questionnaire. Results: Totally, 4763 patients with HBS completed this cross-sectional study, where about 32.4% of them intake breakfast less than one time per week. After controlling for potential confounders, participants who consumed breakfast every day had a 43% lower risk for having HBS as compared with those who had breakfast ≤ 1 times/wk (OR 0.57; 95% CI 0.41-0.80). A significant inverse relationship was found between breakfast consumption and frequent than scare HBS (OR 0.57; 95% CI 0.40-0.77) among the whole population, not in patients with HBS. No significant association was observed between breakfast intake and severity of HBS (OR 0.56; 95% CI 0.31-1.04). Conclusion: We found an inverse association between frequency of breakfast consumption and odds of HBS as well as the frequency of HBS among the adult population. Prospective studies are required to confirm these findings. Level of evidence: Level V, cross-sectional descriptive study.
Article
Rosuvastatin is one of the most potent statins and is currently widely prescribed. It is therefore important to know the dose-related magnitude of effect of rosuvastatin on blood lipids. Primary objectiveTo quantify the effects of various doses of rosuvastatin on serum total cholesterol, low-density lipoprotein (LDL)-cholesterol, high-density lipoprotein (HDL)-cholesterol, non-HDL-cholesterol and triglycerides in participants with and without evidence of cardiovascular disease. Secondary objectivesTo quantify the variability of the effect of various doses of rosuvastatin.To quantify withdrawals due to adverse effects (WDAEs) in the randomized placebo-controlled trials. We searched the Cochrane Central Register of Controlled Trials (CENTRAL) Issue 10 of 12, 2014 in The Cochrane Library, MEDLINE (1946 to October week 5 2014), EMBASE (1980 to 2014 week 44), Web of Science Core Collection (1970 to 5 November 2014) and BIOSIS Citation Index (1969 to 31 October 2014). No language restrictions were applied. Randomized controlled and uncontrolled before-and-after trials evaluating the dose response of different fixed doses of rosuvastatin on blood lipids over a duration of three to 12 weeks. Two review authors independently assessed eligibility criteria for studies to be included and extracted data. WDAEs information was collected from the placebo-controlled trials. One-hundred and eight trials (18 placebo-controlled and 90 before-and-after) evaluated the dose-related efficacy of rosuvastatin in 19,596 participants. Rosuvastatin 10 to 40 mg/day caused LDL-cholesterol decreases of 46% to 55%, when all the trials were combined using the generic inverse variance method. The quality of evidence for these effects is high. Log dose-response data over doses of 1 to 80 mg, revealed strong linear dose-related effects on blood total cholesterol, LDL-cholesterol and non-HDL-cholesterol. When compared to atorvastatin, rosuvastatin was about three-fold more potent at reducing LDL-cholesterol. There was no dose-related effect of rosuvastatin on blood HDL-cholesterol, but overall, rosuvastatin increased HDL by 7%. There is a high risk of bias for the trials in this review, which would affect WDAEs, but unlikely to affect the lipid measurements. WDAEs were not statistically different between rosuvastatin and placebo in 10 of 18 of these short-term trials (risk ratio 0.84; 95% confidence interval 0.48 to 1.47). The total blood total cholesterol, LDL-cholesterol and non-HDL-cholesterol-lowering effect of rosuvastatin was linearly dependent on dose. Rosuvastatin log dose-response data were linear over the commonly prescribed dose range. Based on an informal comparison with atorvastatin, this represents a three-fold greater potency. This review did not provide a good estimate of the incidence of harms associated with rosuvastatin because of the short duration of the trials and the lack of reporting of adverse effects in 44% of the placebo-controlled trials.
Article
Type 2 diabetes mellitus (T2DM) is associated with the development and progression of cardiovascular disease (CVD). Statins have an established efficacy in the management of dyslipidemia primarily by decreasing the levels of low-density lipoprotein cholesterol and thus decreasing CVD risk. They also have a favorable safety profile. Despite the statin-mediated benefit of CVD risk reduction a residual CVD risk remains, especially in T2DM patients with high triglyceride (TG) and low high-density lipoprotein cholesterol (HDL-C) values. Fibrates decrease TG levels, increase HDL-C concentrations, and improve many other atherosclerosis-related variables. Fibrate/statin co-administration improves the overall lipoprotein profile in patients with mixed dyslipidemia and may reduce the residual CVD risk during statin therapy. However, limited data exists regarding the effects of statin/fibrate combination on CVD outcomes in patients with T2DM. In the Action to Control Cardiovascular Risk in Diabetes (ACCORD) study the statin/fibrate combination did not significantly reduce the rate of CVD events compared with simvastatin/placebo in patients with T2DM. However, it did show a possible benefit in a pre-specified analysis in the subgroup of patients with high TG and low HDL-C levels. Furthermore, in the ACCORD study the simvastatin/fenofibrate combination significantly reduced the rate of progression of retinopathy compared with statin/placebo administration in patients with T2DM. The present review presents the available data regarding the effects of statin/fibrate combination in patients with T2DM and atherogenic mixed dyslipidemia.
Article
Full-text available
Objective: Cardiovascular complications represent a major cause of morbidity and mortality in patients undergoing vascular surgery. This was a prospective randomized, open-label study to investigate the effect of lipid-lowering treatment by statin monotherapy or intensified by combining statin with ezetimibe on a 12-month prognosis after vascular surgery. Methods: Patients were randomly assigned to receive rosuvastatin (RSV) 10 mg/d or rosuvastatin 10 mg/d plus ezetemibe (RSV/EZT) 10 mg/d, starting prior to scheduled surgical procedure. The primary end point was the first major cardiovascular event, including death from cardiac causes, nonfatal myocardial infarction, ischemic stroke, and unstable angina. Results: A total of 136 patients assigned to RSV and 126 to RSV/EZT completed the study protocol. As many as 6.6% of patients in the RSV group experience a major cardiovascular event within 30 days after surgery versus 5.6% in the RSV/EZT group (P = .72). From month 1 to 12 of the follow-up period, primary end point was observed (9 taking RSV vs 2 in the RSV/EZT group [P = .04]). Intensified lipid-lowering therapy with RSV/EZT was associated with a greater decrease in low-density lipoprotein cholesterol levels compared with RSV (75.87 ± 31.64 vs 87.19 ± 31.7, P = .004), while no differential effect on triglyceride, high-density lipoprotein cholesterol or high-sensitivity C-reactive protein levels was noted between groups. Conclusion: Our findings indicate that statin therapy intensified by ezetimibe may reduce the incidence of cardiovascular events within the first 12 months after vascular surgery. Nonetheless, whether the use of ezetimibe as an add-on therapy to reduce cardiovascular risk in these patients needs to be tested in larger future studies.
Article
The focus of this review is on the role of apolipoprotein C-II (apoC-II) in lipoprotein metabolism and the potential effects on the risk of cardiovascular disease (CVD). We searched PubMed/Scopus for articles regarding apoC-II and its role in lipoprotein metabolism and the risk of CVD. Apolipoprotein C-II is a constituent of chylomicrons, very low-density lipoprotein, low-density lipoprotein, and high-density lipoprotein (HDL). Apolipoprotein C-II contains 3 amphipathic α-helices. The lipid-binding domain of apoC-II is located in the N-terminal, whereas the C-terminal helix of apoC-II is responsible for the interaction with lipoprotein lipase (LPL). At intermediate concentrations (approximately 4 mg/dL) and in normolipidemic subjects, apoC-II activates LPL. In contrast, both an excess and a deficiency of apoC-II are associated with reduced LPL activity and hypertriglyceridemia. Furthermore, excess apoC-II has been associated with increased triglyceride-rich particles and alterations in HDL particle distribution, factors that may increase the risk of CVD. However, there is not enough current evidence to clarify whether increased apoC-II causes hypertriglyceridemia or is an epiphenomenon reflecting hypertriglyceridemia. A number of pharmaceutical interventions, including statins, fibrates, ezetimibe, nicotinic acid, and orlistat, have been shown to reduce the increased apoC-II concentrations. An excess of apoC-II is associated with increased triglyceride-rich particles and alterations in HDL particle distribution. However, prospective trials are needed to assess if apoC-II is a CVD marker or a risk factor in high-risk patients.
Article
Metabolic syndrome (MetS) is associated with increased cardiovascular mortality and its management incorporates hypolipidemic, antidiabetic and antihypertensive drugs. However, several classes of these drugs, such as biguanides, fibrates and hydrochlorothiazide have been reported to raise circulating total homocysteine (tHcy) levels. During the last decades, numerous large-scale epidemiological studies have identified Hcy as a moderate independent cardiovascular risk factor. Therefore, drug-induced hyperhomocysteinemia in MetS patients may add one cardiovascular risk factor in these high-risk patients. The present systematic review summarizes data from studies which investigated the effects of the above-mentioned drugs on tHcy, and calculates the treatment effect of each drug class on tHcy levels. We also discuss the underlying pathophysiology and the issues that should be addressed in the future.
Article
Full-text available
Clinical trials evaluating the effectiveness of therapy with hydroxymethylglutaryl-coenzyme A (HMG-CoA) reductase inhibitors for reducing elevated C-reactive protein (CRP) levels and associated coronary events are reviewed. Atherosclerotic plaque growth may be attenuated with therapy aimed at minimizing inflammation. Because increased levels of CRP have been associated with arterial-wall inflammation, statins can prevent ischemia by both inhibiting deposition of lipids and decreasing inflammation. Evaluation of recent clinical trials, including WOSCOPS, PRINCE, AFCAPS/ TexCAPS, MIRACL, CURVES, REVERSAL, and JUPITER, demonstrated the correlation of statin therapy with decreased levels of CRP. WOSCOPS found that patients with CRP values of > 4.59 mg/L at baseline were at the highest risk of coronary events. The PRINCE trial evaluated the antiinflammatory effects of pravastatin and found a mean 16.9% reduction in CRP levels after 24 weeks of therapy. AFCAPS/TexCAPS researchers found that lovastatin provded a 14.8% reduction in the median levels of CRP (p < 0.001). The MIRACL study showed that atorvastatin reduced CRP levels by 83% (p < 0.001). Researchers in the CURVES study found a significant reduction in CRP levels with pravastatin, simvastatin, and atorvastatin compared with baseline (p < 0.025). Results of the REVERSAL study linked atorvastatin with a 36.4% decrease in CRP levels, while pravastatin was associated with a 5.2% decrease (p < 0.0001). JUPITER is ongoing and will determine whether long-term use of rosuvastatin can reduce the rate of coronary events. The lowering of elevated CRP levels by statins may reduce the risk of coronary events independently of the effect of statins on lipid levels.
Article
Full-text available
The release of human platelet constituents by the etiologic agent of gout, the monosodium urate crystal, is described here. In suspensions of washed platelets, response to urate crystals proceeded in two phases: A secretory phase involved the rapid active release of serotonin, ATP, and ADP with little loss of lactic dehydrogenase or beta-glucuronidase. A lytic phase involved the slower loss of all platelet constituents. Both phases were inhibited by iodoacetate plus dinitrophenol, suggesting an energy requirement. In ultrastructural studies, lysis of washed platelets which appeared to contain crystals was seen. Urate crystals were also shown to induce serotonin release and platelet lysis in citrated platelet-rich plasma. Since urate crystals are deposited at a variety of sites, urate crystal-platelet interaction in vivo is a possibility. Such interactions, leading to release of platelet constituents, might contribute to gouty inflammation or to enhanced atherogenesis.
Article
Full-text available
We examined the development of new diabetes mellitus in men aged 45 to 64 years during the West of Scotland Coronary Prevention Study. Our definition of diabetes mellitus was based on the American Diabetic Association threshold of a blood glucose level of >/=7.0 mmol/L. Subjects who self-reported diabetes at baseline or had a baseline glucose level of >/=7.0 mmol/L were excluded from the analyses. A total of 5974 of the 6595 randomized subjects were included in the analysis, and 139 subjects became diabetic during the study. The baseline predictors of the transition from normal glucose control to diabetes were studied. In the univariate model, body mass index, log triglyceride, log white blood cell count, systolic blood pressure, total and HDL cholesterol, glucose, and randomized treatment assignment to pravastatin were significant predictors. In a multivariate model, body mass index, log triglyceride, glucose, and pravastatin therapy were retained as predictors of diabetes in this cohort. We concluded that the assignment to pravastatin therapy resulted in a 30% reduction (P:=0.042) in the hazard of becoming diabetic. By lowering plasma triglyceride levels, pravastatin therapy may favorably influence the development of diabetes, but other explanations, such as the anti-inflammatory properties of this drug in combination with its endothelial effects, cannot be excluded with these analyses.
Article
Background: Little is known about the effect of dyslipidemia on serum uric acid (SUA) levels, and less is known about the effect of statin treatment on them. The GREek Atorvastatin and Coronary-heart-disease Evaluation study suggested that a mean atorvastatin dose of 24 mg/d achieves the National Cholesterol Educational Program treatment goals and significantly reduces morbidity and mortality in patients with coronary heart disease (CHD) in comparison to the usual care. Here, we report the time course of SUA levels in usual-care patients undertreated for their dyslipidemia (12% were administered statins) in comparison to structured-care patients treated with atorvastatin in the vast majority (98%). Methods: Mean on-study SUA levels (up to 48 months) were compared with those at baseline by using analyses of variance to assess differences over time within and between treatment groups. Cox multivariate analysis was used to investigate whether changes in SUA levels during the study were clinically relevant. Results: All patients had normal renal function at baseline; serum creatinine (SCr) levels less than 1.3 mg/dL (< 115 mu mol/L) and moderately elevated SUA levels (mean, 7.1 +/- 0.9 [SD] mg/dL [425 52 mu mol/L]; upper normal limit, 7.0 mg/dL [415 mu mol/L]). Usual-care patients (n = 800) showed an increase in SUA levels by 3.3% (P < 0.0001). Structured-care patients (n = 800) had an 8.2% reduction in SUA levels (P < 0.0001). In all patients not administered diuretics (n = 1,407), SUA level changes showed a positive correlation with changes in SCr levels (r = 0.82; P < 0.0001) and an inverse correlation with estimated glomerular filtration rate (r = -0.77; P < 0.0001). After adjustment for 19 predictors of all CHD-related events, Cox multivariate analysis involving backward stepwise logistic regression showed a hazard ratio (HR) of 0.89 (95% confidence interval [CI], 0.78 to 0.96; P = 0.03) with every 0.5-mg (30-mu mol/L) reduction in SUA level, an HR of 0.76 (95% Cl, 0.62 to 0.89; P = 0.001) with every 1-mg (60-mu mol/L) reduction, an HR of 1.14 (95% Cl, 1.03 to 1.27; P = 0.02) with every 0.5-mg increase, and an HR of 1.29 (95% Cl, 1.17 to 1.43; P = 0.001) with every 1-mg increase in SUA levels. Conclusion., Data suggest that SUA level is an independent predictor of CHD recurrent events. Atorvastatin treatment significantly reduces SUA levels in patients with CHD, thus offsetting an additional factor associated with CHD risk.
Article
Patients with coronary artery disease and abnormalities of serum lipids often have endothelial vasodilator dysfunction, which may contribute to ischemic cardiac events. Whether cholesterol-lowering or antioxidant therapy can restore endothelium-dependent coronary vasodilation is unknown. We randomly assigned 49 patients (mean serum cholesterol level, 209 +/- 33 mg per deciliter [5.40 +/- 0.85 mmol per liter]) to receive one of three treatments: an American Heart Association Step 1 diet (the diet group, 11 patients); lovastatin and cholestyramine (the low-density lipoprotein [LDL]-lowering group, 21 patients); or lovastatin and probucol (the LDL-lowering-antioxidant group, 17 patients). Endothelium-dependent coronary-artery vasomotion in response to an intracoronary infusion of acetylcholine (10(-8) to 10(-6) M) was assessed at base line and after one year of therapy. Vasoconstrictor responses to these doses of acetylcholine are considered to be abnormal. Treatment resulted in significant reductions in LDL cholesterol levels of 41 +/- 22 percent in the LDL-lowering-antioxidant group and 38 +/- 20 percent in the LDL-lowering group (P < 0.001 vs. the diet group). The maximal changes in coronary-artery diameter with acetylcholine at base line and at follow-up were -19 and -2 percent, respectively, in the LDL-lowering-antioxidant group, -15 and -6 percent in the LDL-lowering group, and -14 and -19 percent in the diet group (P < 0.01 for the LDL-lowering-antioxidant group vs. the diet group; P = 0.08 for the LDL-lowering group vs. the diet group). (The negative numbers indicate vasoconstriction). Thus, the greatest improvement in the vasoconstrictor response was seen in the LDL-lowering-antioxidant group. The improvement in endothelium-dependent vasomotion with cholesterol-lowering and antioxidant therapy may have important implications for the activity of myocardial ischemia and may explain in part the reduced incidence of adverse coronary events that is known to result from cholesterol-lowering therapy.
Article
This study was conducted in preparation for the Study Evaluating Additional Reduction in Cholesterol and Homocysteine (SEARCH). SEARCH is a 12,000 patient 2X2 factorial study in post-myocardial infarction patients that will compare simvastatin 20 mg with simvastatin 80 mg to evaluate whether greater LDL-C reductions with simvastatin provide greater coronary event reductions. SEARCH will also test the hypothesis that lowering plasma homocysteine with folic acid and vitamin B12 will reduce coronary events. This pilot study was performed to determine whether any clinically meaningful interaction between simvastatin and folic acid/vitamin B12 exists. Following a 2-week diet/placebo run-in period, 141 patients with primary hypercholesterolaemia were randomised to one of three treatments for 6 weeks: 80 mg/day simvastatin and 2 mg folic acid/0.8 mg vitamin B12 daily (combination group); or 80 mg/day simvastatin and placebo vitamins (simvastatin alone group); or 2 mg folic acid/0.8 mg vitamin B12 daily and placebo simvastatin (vitamins alone group). The combination group and simvastatin alone group experienced similar serum lipid changes with reductions in LDL-cholesterol of 55.2% and 51.5% respectively. The combination group and vitamins alone group experienced similar homocysteine lowering with reductions in homocysteine of 25.3% and 23.1% respectively. All therapies were well tolerated. There was no detectable antagonistic effect when simvastatin and folic acid/vitamin B12 were administered concomitantly.
Article
Hyperhomocysteinemia is a risk factor for cardiovascular disease. Elevation in homocysteine levels has recently been demonstrated during lipid lowering treatment with fibrates. We compared the effect of a statin and a fibrate (atorvastatin and fenofibrate) on plasma levels of homocysteine and other thiol compounds in hyperlipidemic patients. The study was of open randomized, parallel design with a preliminary screening phase, and a 6 week placebo period. After the placebo period, patients were allocated randomly to atorvastatin or fenofibrate for a 6 month period. Plasma thiols were assayed by high pressure liquid chromatography with fluorescence detection. There were 29 patients in the fenofibrate group and 24 in the atorvastatin group. Fenofibrate induced a significant increase in both homocysteine and cysteine plasma levels (+35.8 and +18%, respectively, P<0.0001); by contrast, cysteinylglycine remained stable. There were no significant changes in any thiol compounds in the atorvastatin group. Both treatments induced a significant decrease in uric acid, although fenofibrate was noticeably more effective than atorvastatin (-22.8 and -6.4%, respectively). Fenofibrate induced a non-significant increase in creatinine (12%) while atorvastatin reduced it (4.7%, NS). Our study confirms that the induction of elevations in plasma homocysteine and cysteine levels are a distinct feature of the pleiotropic effects of fibrates. Further studies are needed not only to investigate the potential deleterious effects of this modification, but also to define the specific mechanism which underlies such fibrate-mediated action.