You are on page 1of 16

Trends

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

HPLC methods for the


determination of simvastatin and
atorvastatin
Lucie Novakova, Dalibor Satnsky, Petr Solich
We review high-performance liquid chromatography (HPLC) methods for the determination of two major statins used in clinical
treatment simvastatin and atorvastatin in various fields of application, including bio-analytical assays, pharmaceutical assays
and environmental applications.
Statin molecules are known to be susceptible to interconversion of the lactone and acidic forms, so it is necessary to consider
this phenomenon during method development. We highlight liquid chromatography coupled to tandem mass spectrometry (LCMS/MS) methods, as they have become a method of choice in bio-analytical and environmental applications. We compare the
methods from the point of view of sensitivity. We discuss selection of the precursor ion for performing selected reaction
monitoring (SRM) in MS detection and sample preparation.
2008 Elsevier Ltd. All rights reserved.
Keywords: Atorvastatin; Bio-analytical method; Environmental analysis; High-performance liquid chromatography; HPLC; Interconversion;
Pharmaceutical analysis; Simvastatin; Tandem mass spectrometry
Abbreviations: [13CD3], Stable-isotope labeling using 13C and three atoms of deuterium; [d5], Stable isotope labeling using five atoms of deuterium;
2-OH-AT, 2-hydroxyatorvastatin; AcAc, Acetic acid; ACN, Acetonitrile, AmAc, Ammonium acetate; AT, Atorvastatin; AT-L, Atorvastatin lactone;
DFAT, Desfluoro-atorvastatin; DSAT, Diastereomer-atorvastatin; ESI, Electrospray ionization; Et-Ac, Ethylacetate; FAc, Formic acid; FD,
Fluorescence detection; GC, Gas chromatography; HLB, Hydrophilic lipophilic balance; HMG-CoA, 3-hydroxy-3-methylglutaryl coenzyme A;
HPLC, High-performance liquid chromatography; IS, Internal standard; LDL, Low-density lipoprotein; LLCE, Liquid-liquid cartridge extraction; LLE,
Liquid-liquid extraction; LOD, Limit of detection; LOQ, Limit of quantitation; LOV, Lovastatin; LOVA, Lovastatin acid; M, mol/l; MeOH, Methanol;
Me- SV, Methyl-simvastatin; MEV, Mevastatin; MP, Mobile phase; MS, Mass spectrometry; MS/MS, Tandem MS; MTBE, Methyl-tert-butyl-ether;
ODS, Octadecylsilica, C18; o-OH-AT, o-hydroxyatorvastatin; p-OH-AT, p-hydroxyatorvastatin; PRA, Pravastatin; QC, Quality control; ROS,
Rosuvastatin; SDS, Sodium dodecyl-sulphate; SIM, Selected ion monitoring; SPE, Solid-phase extraction; SRM, Selective reaction monitoring; SV,
Simvastatin; SVA, Simvastatin acid; THF, Tetrahydrofuran; TIS, Turbo ionspray; ulv, ultra-low volume; UV, Ultraviolet

1. Introduction
Lucie Novakova*,
Dalibor Satnsky, Petr Solich
Department of Analytical
Chemistry, Faculty of
Pharmacy, Charles University,
Heyrovskeho 1203, 500 05,
Hradec Kralove, Czech
Republic

Corresponding author.
Tel.: +420 495067345;
Fax: +420 495067164;
E-mail: novakoval@faf.cuni.cz

352

Statins include natural (lovastatin), semisynthetic (simvastatin, and pravastatin)


and synthetic compounds (fluvastatin,
atorvastatin, cerivastatin, rosuvastatin
and pitavastatin) and are potent, specific
and competitive inhibitors of 3-hydroxy-3methlyglutaryl coenzyme A (HMG-CoA)
reductase. They are highly effective in
reducing total cholesterol and low-density
lipoprotein (LDL) cholesterol levels in the
human body.
HMG-CoA reductase is the key enzyme
that catalyzes the conversion of HMG-CoA
to mevalonate, which is an early ratelimiting step in the biosynthetic pathway
of cholesterol (Fig. 1). High-plasma LDL
cholesterol is a risk factor of cardiovascu-

lar diseases, such as atherosclerosis, which


is characterized by deposition of cholesterol on the arterial wall [13]. Atherosclerosis of the coronary and peripheral
vasculature is the leading cause of death
worldwide. Statistics show that 3842% of
deaths are related to cardiovascular diseases in Western and other developed
countries [4]. Lowering cholesterol levels
can arrest or reverse atherosclerosis in all
vascular beds and it can significantly decrease the morbidity and the mortality
associated with atherosclerosis. The main
methods of treating hyperlipidemia (or
hypercholesterolemia) are dietary and
lifestyle changes and the administration of
hypolipidemic drugs [5].
Statins are commonly used to treat
several
forms
of
hypercholesterol-

0165-9936/$ - see front matter 2008 Elsevier Ltd. All rights reserved. doi:10.1016/j.trac.2008.01.013

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Trends

Figure 1. Biosynthesis of cholesterol. Cholesterol is synthesized from acetyl coenzyme A. The synthesis of mevalonate, mediated by HMG-CoA
reductase is the rate-limiting step, which regulates the cholesterol synthesis. It is the step where the statin molecule competitively effects the
synthesis of cholesterol.

emia.They have potent cholesterol-lowering effects and


they could reduce morbidity and mortality associated
with coronary heart disease significantly, as proved by
many clinical trials [2,68]. However, some statins
exhibit a number of adverse effects, such as myopathy or
rhabdomyolysis, so it is useful to monitor the levels of
statins in biological materials in order to establish an
appropriate dosage scheme, which would minimize
adverse effects and keep the cholesterol-lowering effect
[2].
Simvastatin and atorvastatin are the two most commonly occurring drugs in commercially available pharmaceutical formulations used for the clinical treatment of
hypercholesterolemia. Their structures can be seen in
Fig. 2.

2. Chemistry and pharmacokinetics


The first statin molecule mevastatin was discovered
by Endo et al. in 1976 as a fungal product extracted
from Penicillium citrinum [9]. Lovastatin, simvastatin and
pravastatin are also derivatives of fungal products.
Simvastatin and pravastatin are nowadays produced
semi-synthetically from lovastatin and mevastatin.
However, fluvastatin is a completely synthetic statin
with a very different structure from statins derived from
fungal products. It is a mevalonolactone derivative with
a fluorophenyl-substituted indole ring. Other synthetic
statins (cerivastatin, atorvastatin, rosuvastatin and
pitavastatin) have similar structures with fluorophenyl
groups. All of totally synthetic statins have open-ring

http://www.elsevier.com/locate/trac

353

Trends

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008


OH
O
CH3

H3C

H3C

CH3

H3C

Simvastatin (lactone form)

HO
COOH
OH

CH3

F
N

CH3
H
N
O

Atorvastatin (acid form)

Figure 2. Chemical structures of simvastatin and atorvastatin.

acid forms [2,9]. All statins are absorbed rapidly following administration, reaching peak plasma concentration within 4 hours.
Statins exist in two forms, lactone and open-ring
hydroxy acid [10,11]. In vivo, the hydroxy acid forms
are the active drugs that lower plasma cholesterol while
the lactone forms are inactive (prodrug). The lactone
form of statin can be absorbed from the gastrointestinal
tract and transformed into the active drugs in liver and
non-hepatic tissues [11]. Depending upon chemical
structure, statins have different affinities for HMG-CoA
reductase, which determines their pharmacological
effects and different pharmacokinetic properties (e.g.,
tissue distribution, metabolic stability, enzymes and
transporters involved in their metabolism) [2]. We will
provide further information on simvastatin and atorvastatin, the most widely used statins in clinical treatment Fig. 2.
Simvastatin is a prodrug, which is administered as
an inactive lactone form. The lactone is absorbed from
gastrointestinal tract and hydrolyzed to the active
b-hydroxy acid form in the liver. Simvastatin and its
hydroxy acid are extensively (95%) bound to plasma
proteins. The substance undergoes extensive first-pass
metabolism in the liver and is mainly excreted in the bile.
About 85% of administered dose has been recovered
from the feces as metabolite and about 1015% from
urine, mainly as inactive forms [12,13].
Atorvastatin is administered in the open-ring hydroxy
acid form the active form. It is absorbed from the
354

http://www.elsevier.com/locate/trac

gastrointestinal tract and it undergoes extensive firstpass metabolism in the liver. Liver metabolism produces
two active hydroxy metabolites ortho-hydroxyatorvastatin and para-hydroxyatorvastatin and three
inactive metabolites corresponding to the lactone form.
More than 98% is bound to plasma proteins. It is
excreted mainly in feces via the bile, with a smaller
proportion excreted in the urine. About 70% of the total
plasma HMG-CoA activity is attributed to active metabolites of atorvastatin, even if their concentrations are
very low [1214].
As apparent from the information above, the levels
of statins in biological fluids are very low, probably
because only about 5% of dosed statin reaches the
systemic circulation. For atorvastatin and simvastatin,
the plasma concentrations are typically at ng/ml levels.
The active metabolites of atorvastatin are present at
plasma concentration corresponding to pg/ml levels
[13], the typical concentration range being 0.120 ng/
ml.

3. Interconversion of lactone and hydroxy acid


forms
A number of classes of drugs could potentially undergo
interconversion, which may occur during any of the
numerous steps of the bio-analytical method:
 in the biological matrix before collecting aliquots of
samples for the analysis;
 during extraction;
 during evaporation to dryness or reconstitution;
 in the solution in the injection vial; and,
 in the case of MS, in the ion source as well.
There are several categories of drugs that could undergo interconversion. HMG-CoA reductase inhibitors
are typical examples of such a class, where the interconversion occurs between lactone and open-ring hydroxy acid [10,11]. Another category could be
conversion between samples that contain a carboxylic
acid and its acylglucuronide [15]. Samples that contain
a thiol group and its disulfide may also interconvert to
each other [16]. Minimizing the interconversion will
depend on the conditions during the bio-analytical procedure, pH being one of the most important.
HMG-CoA reductase inhibitor simvastatin is administered in its lactone form, although the active form is
open-ring hydroxy acid. Both forms are found in postdose samples.
On the other hand, atorvastatin is administered as
the open-ring hydroxy acid, but the post-dose samples contain both acid and lactonized forms
[10,11,17,18].
For the samples of hydroxy acid chemical structure
and the corresponding lactone forms, it is important to
maintain pH between 4 and 5 in order to minimize

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

interconversion. Increasing the pH above 6 facilitates the


conversion of lactone to acid (in the ionized form); by
contrast, lowering pH facilitates the conversion of acid to
lactone or lactone to acid (in the non-ionized form). Most
assays utilize pH around 4.5 (see Tables 1 and 2).
It is important to optimize the method in order to
minimize such interconversion. Unfortunately, even
optimal conditions may not totally prevent interconversion. It is thus essential to design the composition of
the calibration standards and quality control (QC) samples in terms of the ratio of the concentration of one
analyte to that of the other so that the accuracy and
precision obtained for QC samples realistically reflect
accuracy and precision that will be obtained for the expected samples. Ideally, the composition of the QC samples should be identical to that of real samples. This is
difficult to establish at the early stage of assay, where the
composition of real samples is not known and will
probably change from sample to sample [10].
It is recommended that the example of methoddevelopment design should include QC samples prepared
in real biological material containing only the lactone
form, only the hydroxy acid form, and both in varying
ratios of lactone to hydroxy acid (1:1; 1:10; 10:1; 1:3;
and, 3:1) in order to get good method accuracy and
precision. Such QC samples should be prepared at various concentration levels inside the range of calibration
curve [10]. In this study, the influence of pH (analytical
conditions at pH 4.2, 7.3 and 1.8) on interconversion of
analytes was described in detail. Under the conditions
that showed practically no interconversion, the results
for all QC samples gave excellent values for accuracy (up
to 8.6% RSD) and precision (up to 8.7% RSD) with no
influence of the ratio of both analytes.
However, in another experimental design, the results
for accuracy and precision were strongly influenced by
this ratio, except for 1:1, which was the same ratio as in
the calibration standards. Thus, under the conditions
that allow interconversion between analytes, a method
validated for the quantitation of the two analytes using
calibration standards with the ratio of analyte concentration of 1:1 and QC samples with the same 1:1 ratio
can be used for accurate measurements of the analytes
in real samples only if such samples also contain the two
analytes in the ratio 1:1. Therefore, to obtain an accurate indication of the performance of the method for all
real samples, it is necessary to use QC samples that cover
the entire spectrum of composition of real samples.
To summarize, in developing a method for the quantitation of two analytes that can undergo interconversion, the first step is to select the conditions that will
eliminate or minimize interconversion. The second step
is judicious selection of the composition of the QC
samples and the composition of calibration standards,
which should cover the spectrum of the composition of
real samples [10].

Trends

4. Analytical methods
The determination of drugs is a multi-disciplinary task.
During the manufacturing process of a drug substance
and drug formulation, there is a need for QC analytical
methods to include all known and unknown impurities.
Bio-analytical methods are necessary for clinical trials,
therapeutic drug monitoring and individual dosagescheme adjustment. Recently, there has also been great
interest in monitoring pharmaceutical residues in the
environment, so environmentally-focused analytical
methods are also necessary.
Determination of drugs in biological materials is an
important step in drug discovery and drug development, as it provides pharmacokinetic information, and
is necessary when the treatment-dose schedule and
safety margins need to be established. High-performance liquid chromatography (HPLC) together with
various types of detection UV (ultraviolet), FD (fluorescence detection) and MS has become the method of
choice for bio-analytical method development. Gas
chromatography (GC) is somewhat suppressed because
of the need for a derivatization step prior to analysis in
order to obtain volatile derivatives of the drug molecule,
which is often not volatile in the case of pharmaceuticals.
As expected from the different structures of simvastatin and atorvastatin, analytical methods for their quantitative determination were developed individually.
Because of the structural properties, there are not many
analytical methods that determine these two compounds
together in one analytical run or even in combination
with other statin molecules. This is also probably
because statins are not used with other statins simultaneously during treatment of hyperlipidemic patients.
There are only two works that describe the determination of a number of statin structures together in one
analytical run:
 an environmental application where simvastatin,
atorvastatin, lovastatin and pravastatin (internal
standard (IS) = mevastatin) were determined in
aqueous samples [19]; and,
 a pharmaceutical application where atorvastatin,
lovastatin, pravastatin, rosuvastatin and simvastatin
were determined together for QC of pharmaceutical
formulations [20]. Theophylline was employed as IS
for quantitation. The analytical run took about 40
min.
There have been two reviews on analytical methods
for the determination of HMG-CoA reductase inhibitors.
The first, published in 2003, referred to lovastatin,
simvastatin, pravastatin, fluvastatin and atorvastatin
[12]. HPLC and GC methods were discussed. Generally,
fluorescence and UV detection were applied together
with HPLC. Only two methods for the determination of
atorvastatin were available at that time. The second
http://www.elsevier.com/locate/trac

355

Trends

356
http://www.elsevier.com/locate/trac

Table 1. HPLC analytical methods for the determination of simvastatin


Matrix sample
preparation

Stationary phase
analytical column

Mobile phase

Detection

Precursor ions notes

Time [min]

Validation data
LOQ/LOD

Ref.

SV, SVA
LOV, LOVA
PRA

Pu-Ehr tea SPE

Luna C18
(4.6 250 mm, 5 lm)

ACN, water, AcAc (70:30:0.5)

ESI+
MS-MS
SRM

Not stated

15

Not given stability


and interconversion study

[11]

SV, AT
LOV
PRA
IS = MEV

Aqueous samples
SPE

Genesis C18
(2.1 50 mm, 3 lm)

Gradient elution A:
ACN + 2 mM MeA + 0.1%
AcAc B: water + 2 mM
MeA + 0.1% AcAc

ESI+
MS-MS
SRM

[M + CH3NH3]+
[M + H]+
[M + CH3NH3]+
[M + CH3NH3]+

5.5

LOD = 0.11.0 ng/l


LOD = 0.11.2 ng/l
LOD = 0.11.2 ng/l
LOD = 1.015.4 ng/l

[19]

SV
AT, LOV
PRA, ROS
IS = theophylline

Pharmaceutical
formulation
metabolism study
in vitro
extraction MeOH

Interstil ODS 3V
(4.6 250 mm, 5 lm)

0.01 M AmAc pH 5.0


ACN
MeOH

UV 237 nm

40

LOQ = 0.1 lg/ml

[20]

SV, SVA
IS = LOV, LOVA

Human plasma
SPE

Capcell Pak C18


(4.6 150 mm, 5 lm)

ACN-water (80:20)

FD
360 nm
430 nm

Derivatization by
1-bromoacetylpyrene

35

LOQ = 0.1 ng/ml

[21]

SV, SVA
No IS

Human plasma
LLE

ODS Hypersil
(4.6 250 mm, 5 lm)

0.025 M sodium
dihydrogenphosphate pH4.5
ACN (35:65)

UV
238 nm

10

LOD = 15 ng/ml

[22]

SV
IS = LOV

Human plasma
LLE

Capcell Pak C18 UG


120U (1.5 250
mm, 5 lm)

ACN 20 mM potassium
phosphate
buffer pH 5.6 (65:35)

30

LOQ = 0.5 ng/l

[23]

SV and impurities
(SVA, LOV
Me-SV, dimmer
acetate ester
anhydro-SV
IS = propyphenazon

Tablets extraction MP

X-Terra (4.6
50 mm, 3.5 lm)

Microemulsion: 0.9%
diisopropylether
1.7% SDS 7.0%
co-surfactant 90% 25
mM di-sodium
phosphate pH 7.0

25

LOD = 5 ng/ml
LOQ = 10 ng/ml
(SV and SVA)

[24]

UV
238 nm

UV
238 nm

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Determined
substances

Table 1 (continued)
Matrix sample
preparation

Stationary phase
analytical column

Mobile phase

Detection

Precursor
ions notes

Time
[min]

Validation
data LOQ /LOD

Ref.

SV, LOV

Standard solutions

No LC

No LC

ESI+MSMS SRM

[M+H]+its
fragmentations
using MS

Not stated
fragmentation study

[25]

SV, SVA

Standard solutions

C18 (3.9 50 mm,


5 lm)

ACN, 3.0 mM FAc (70:25)

ESI+
MS-MS
SRM

[M + H]+
[M + NH4]+

2.5

Chromatographic and
mass resolution study

[26]

SV + impurities
No IS

SV substance tablets
Extraction ACN/FAc

Zorbax C8
(4.6 150 mm,
3.5 lm)

Gradient elution
A: 0.085 phosphoric acid
B:ACN
A: 0.001% FAc in water
B: 0.001% FAc in CAN

DAD
ESI+
MS-MS

45

Identification of
impurities

[27]

SV, SVA
IS = LOV, LOVA

Human plasma
SPE

Discovery C18
(4.6 50 mm, 5 lm)

ACN, MeOH, AmAc 0.1 M


(62:10:28)

ESI+/
MS-MS
SRM

[M + CH3CN + Na]+
[M H]

4.5

LOQ = 0.03 ng/ml SV


LOQ = 0.02 ng/ml SVA

[28]

SV, SVA
IS = LOV, LOVA

Human
plasma on-line
SPE

C18 (3.9 50 mm,


5 lm)

ACN, 3.0 mM FAc (70:25)

ESI+
MS-MS
SRM

[M + H]+

2.5

LOQ = 0.5 ng/ml SV

[29]

SV
IS = LOV

Human plasma LLE

Shim-pack ODS
(4.6 150 mm,
5 lm)

MeOHwater (9:1)

ESI+
MS
SV

[M + Na]+

LOD = 0.05 ng/ml

[30]

SV, SVA
IS = deuterium labeled

Eects of
mobile phase
on ionization/
fragmentation
plasma
LLCE

Kromasil C18
(2.0 50 mm, 4 lm)

ACN-buffer 2 mM pH 4.5
(70:30) buffers: AmAc,
hydrazine acetate, alkylammonium acetates

ESI+
MS-MS
SRM

[M H] SVA various SV: [M + H]+


[M + Na]+ [M + K]+
[M + NH4]+
[M + RNH3]+
R = alkyl

3.5

LOQ = 50 pg/ml

[31]

SV, SVA IS = stable


isotope labeled

Human plasma
LLCE

Kromasil C18
(2.0 50 mm, 5 lm)

ACN AmAc pH 4.5 by FAc


(75:25)

TIS
+
MS-MS
SRM

[M + H] SV
[M H] + SVA

3.5

LOQ = 50 pg/ml

[32]

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

http://www.elsevier.com/locate/trac

Determined
substances

(continued on next page)

Trends

357

358

[34]
LOQ = 0.05 ng/ml
Kromasil C18
(2.0 50 mm, 5 lm)
Human plasma LLE
SV, SVA
IS = stable isotope
labeled

ACN 1 mM methyl ammonium


acetate pH 4.5 (75:25)

TIS
+
MS-MS
SRM

[M + H] SV
[M H] + SVA

3.5

[33]
LOQ = 0.05 ng/ml
3.0
[M H] SVA
[M + CH3NH3]+ SV
TIS+
MS-MS SRM
Synergie Max RP
(2.0 50 mm, 4 lm)
Human plasma ulv-SPE

ACN 1 mM methyl ammonium


acetate pH 4.5 (80:20)

Precursor
ions notes

SV, SVA IS = deuterium


labeled

Table 1 (continued)

Stationary phase
analytical column
Matrix sample
preparation
Determined
substances

Mobile phase

Detection

Time
[min]

Ref.

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Validation
data LOQ /LOD

Trends

http://www.elsevier.com/locate/trac

work [13] referred to some MS bio-analytical methods


for the determination of statins.
Fig. 3 shows the trend in developing new methods for
the determination of simvastatin and atorvastatin. Simvastatin has been monitored since the early 1990s,
while, due to its later synthesis, atorvastatin has been
monitored since 1999. Since 2003, the number of newly
developed methods has been increasing for both analytes.
4.1. Simvastatin
LC-MS/MS (LC coupled to tandem MS) is nowadays the
method of choice for the determination of simvastatin
and its hydroxy acid (Table 1). While, in 2003, there
was the same number of GC methods (3), HPLC-UV or
FD (3) and LC-MS (3) [12], newly developed methods
employed MS/MS. Only simvastatin acid (open-acid
form, which arises from interconversion during analytical procedure or in the human body as an active form of
a prodrug) was monitored together with simvastatin; no
other metabolites were described.
Whereas the HPLC-FD method [21] using 1-bromoacetylpyrene for the derivatization could be very
convenient for bio-analytical purposes with its LOQ of
0.1 ng/ml, the HPLC-UV assay has borderline sensitivity
for the quantitation of simvastatin in biological materials
because of the poor UV-absorption properties of simvastatin molecule, giving an LOD of 15 ng/ml [22]. A more
sensitive HPLC-UV assay for the determination of simvastatin in human plasma was developed later with an
LOQ of 0.5 ng/ml [23]. However, lower sensitivity is no
problem for quantitation in pharmaceutical preparations. Such a method was developed by Malenovic et al.
[24] utilizing a microemulsion mobile phase for quantitation of simvastatin and its six impurities (simvastatin
acid, lovastatin, methyl-simvastatin, dimmer of simvastatin, acetate ester of simvastatin and anhydro-simvastatin) in tablets or the one previously cited [20].
HPLC-UV and FD analytical methods utilized C18
analytical columns for the retention/separation of simvastatin/simvastatin acid together with mobile phase
containing acetonitrile in combination with phosphate
buffer (to maintain pH around 45) or with water in an
older method [21]. X-terra hybrid stationary phase was
employed in one case together with application of a
microemulsion mobile phase [24]. Isocratic elution was
applied in most analyses. Detection was performed at
238 nm in all cases [2224]. A fluorescence method
utilized 360 nm and 430 nm [21]. Analytical run time
was usually very long, starting from 10 min [22] going
up to 35 min using FD [21]. Two bio-analytical methods
also determined simvastatin acid next to the lactone
form [21,22], as did a pharmaceutical QC method,
where simvastatin acid was determined among six
impurities using IS propyphenazon [24]. Lovastatin was
often employed as an IS because of its unique structural

http://www.elsevier.com/locate/trac

Determined substances

Matrix sample preparation

Stationary phase analytical column

Mobile phase

Detection

Precursor
ions (notes)

Time
[min]

Validation data
LOQ /LOD

Ref.

AT, SV
LOV, PRA
IS = MEV

Aqueous samples SPE

Genesis C18 (2.1


50 mm, 3 lm)

Gradient elution
A: ACN + 2 mM
MeA + 0.1% AcAc
B: water + 2 mM
MeA + 0.1% AcAc

ESI
+
MS-MS
SRM

[M + CH3NH3]+
[M + H]+
[M + CH3NH3]+
[M + CH3NH3]+

5.5

LOD = 0.11.0 ng/l


LOD = 0.11.2 ng/l
LOD = 0.11.2 ng/l
LOD = 1.015.4 ng/l

[19]

AT, LOV
PRA
ROS, SV
IS = theophylline

Pharmaceutical
formulation
metabolism study
in vitro extraction:
ACN:buffer

Interstil ODS 3V
(4.6 250 mm, 5 lm)

0.01 M AmAc pH 5.0


ACN
MeOH

UV
237 nm

40

LOQ = 0.1 lg/ml

[20]

AT impurities = DSAT,
DFAT, no IS

Bulk drug tablets


extraction MeOH

C18 Luna
(4.6 250 mm, 5 lm)

Gradient elution
ACN-ammonium acetate
buffer pH 4.0 THF

UV
248 nm

32

LOQ = 0.13 lg/ml


LOD = 0.013 lg/ml

[36]

AT, amolodipine
+ impurities
no IS

Tablets
MeOH extraction

Perfectsil Target ODS-3


(4.6 250 mm, 5 lm)

ACN 0.025 M NaH2HPO4


buffer pH 4.5 (55:45)

UV
237 nm

18

LOD = 0.35 lg/ml


LOQ = 2 lg/ml

[37]

AT amlodipine
No IS

Drug products
MeOH extraction

Lichrosphere 100 C18


(4.6 250 mm, 5 lm)

50 mM potassium dihydrogen
phosphate buffer pH 3.0
ACN (40:60)

UV
254 nm

LOD = 0.4 lg /ml

[38]

AT
IS = DCF

Human serum
LLE

Shim-pack CLC-ODS
(4.6 150 mm, 5 lm)

0.05 M sodium phosphate


buffer, pH 4.0 by
phosphoric acid
MeOH (33:67)

UV
247 nm

LOQ 4 ng/ml
LOD = 1 ng/ml

[39]

AT
IS = ibuprofen

Bulk drug tablets


human plasma
MeOH extraction

RP Supelcosil C18
(4.6 150 mm, 5 lm)

ACN:MeOH:water
(45:45:10)

UV
240 nm

LOD = 0.008 lg /ml


LOQ = 0. 18 lg/ml

[40]

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Table 2. HPLC analytical methods for the determination of atorvastatin

(continued on next page)

Trends

359

Trends

360
http://www.elsevier.com/locate/trac

Table 2 (continued)
Matrix sample preparation

Stationary phase analytical column

Mobile phase

Detection

Precursor
ions (notes)

Time
[min]

Validation data
LOQ /LOD

Ref.

AT novobicin
roxytromycin

Aqueous samples
SPE

YMC ODS-AQ (1.0 100 mm,


3 lm)

Gradient elution
A: ACN
B: 10 mM AmAc

ESI+
MS-MS
SRM

[M + H]+

5.0

ILOQ = 1 pg

[41]

AT, ATA
2-OH-AT/L
4-OH-AT/L
IS = deuterium labeled

Human serum LLE

YMC Basic (2.0 50 mm, 5 lm)

Gradient elution
A: water, MeOH, FAc 88%
B: ACN, MeOH, FAc 88%
(950 ml:50 ml:43 ll)

ESI+
MS-MS
SRM

[M + H]+

3.5

LOQ = 0.5 ng/ml

[42]

AT o-OH-AT, p-OH-AT
IS = deuterium labeled

Human, dog and rat


plasma LLE

YMC JSphere H 80 (C18)


(3.0 150 mm, 4 lm)

ACN, AcAc 0.1% (70:30)

ESI+
MS-MS
SRM

[M + H]+

LOQ = 0.250 ng/ml

[43]

AT, ATA
p-OH AT/L
o-OH AT/L
IS = metachalon

Human plasma SPE

Omnisphere C18
(2.0 30 mm, 3 lm)

Gradient elution
A: ACN, water, FAc 1mM
(30:70)
B: ACN, water, FAc 1mM
(60:40)

ESI+
MS-MS
SRM

[M + H]+

21

LOD = 0.06 ng/ml


LOD = 0.16 ng/ml o-AT

[18]

AT
p-OH AT
o-OH AT
IS = ROS

Human plasma LLE

Waters Symmetry C18


(4.6 100 mm, 5 lm)

0.03%
FAc ACN (30:70)

TSI+
MS-MS
SRM

[M + H]+

2.5

LOQ = 100 pg/ml

[44]

AT
2-OH-AT
IS = clindamycin

Human plasma LLE

Atlantis dC18 (4.0 100 mm,


3 lm)

ACN, AcAc (70:30)

ESI+
MS-MS
SRM

[M + H]+

4.0

LOD = 0.02 ng/ml


LOQ = 0.1 ng/ml

[45]

AT, AT-L
p-OH AT/L
o-OH AT/L
IS = deuterium labeled

Human plasma SPE

4 analytical columns in parallel


Genesis C18 (2.2 50 mm, 4 lm)

Gradient elution
A: 0.1% AcAc in water
B: ACN

ESI+
MS-MS
SRM

[M + H]+

1.65

[46]

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Determined Substances

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Trends

Analytical methods for simvastatin and atorvastatin

4
simvastatin
atorvastatin

0
1992

1997

1999

2000

2001

2002

2003

2004

2005

2006

2007

Figure 3. Analytical methods for the determination of simvastatin and atorvastatin first analytical methods for the determination of simvastatin
were published in the early 1990s, and the first for atorvastatin in 1999. The increasing trend for new methods to be developed for both statins
can be observed since 2003.

similarity with the molecule of simvastatin (Fig. 4). The


difference in the structure is only one methyl group, so
such a molecule could also be used as convenient IS in
MS detection, since it will have not just the same
retention and extraction properties, but also similar
ionization in the ion source.
Since 2000, there has been a growing number of
newly developed methods for the determination of simvastatin, where most of methods employed MS/MS
detection (Fig. 3). In the case of simvastatin, there were
not only methods for its quantitation also other methods
that studied detailed fragmentation [25], chromatographic and mass resolution necessary for analysis of
simvastatin and simvastatin acid [26], identification of
unknown impurities in simvastatin substance and tablets [27], and the influence of various media (used for
simvastatin standard solutions) for the interconversion
between simvastatin and simvastatin acid [11].
Quantitative LC-MS methods for simvastatin included,
above all, bio-analytical assays [2834] determining
simvastatin in human plasma, one pharmaceutical QC
application [27] and one environmental application
[19]. They employed MS/MS using a specific SRM (selected reaction monitoring) experiment except for one
work, where a single quadrupole was utilized for quantitation enabling only an SIM (selected ion monitoring)
experiment for quantitation [30].
Simvastatin in its lactone form was always determined
in electrospray positive ion mode (ESI+) or in positive
turbo ionspray (TIS+) respectively, while its hydroxy acid
form was determined in electrospray negative ion mode

(ESI ), negative turbo ionspray (TIS ) and ESI+ or TIS+.


The selection of precursor ion for the determination of
simvastatin lactone was not remotely unequivocal see
Tables 1 and 3. Simvastatin forms various adducts
influenced by mobile-phase composition, and, unfortunately, such adducts sometimes give higher intensity
than [M+H]+, which would be an ideal precursor ion for
SRM transition and was utilized in some studies
[26,29,32,34]. However, other authors found that simvastatin was not sensitive enough and they observed
[M+Na]+, [M+K]+, [M+NH4]+ in the spectra of simvastatin next to the [M+H]+, especially [M+Na]+ and
[M+NH4]+, which gave very high intensities [30,31].
They were therefore used as precursor ions for quantitation in some works [28,30].
Favorable experimental conditions could achieve high
sensitivity for [M+Na]+ over major molecular ions, as
published in earlier LC-MS/MS studies. Nevertheless, the
sodium content was found not to be totally controllable
during sample analysis. It caused variations in the
abundance of the sodium-adduct ion. Sources of sodium
(potassium respectively) are believed to originate from
the biological matrix or glass containers used during
sample preparation and analysis. Sodium acetate could
be added to the mobile phase as the way of maintaining
better consistency in the abundance of the sodium-adduct ion. Unfortunately, the non-volatile nature of the
sodium buffer makes it preferable not to use it during LCMS/MS assays. The factors affecting the relative abundance of [M+Na]+ and [M+H]+ include not only general
experimental conditions, such as mobile-phase-buffer
http://www.elsevier.com/locate/trac

361

Trends

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

OH
H3C
H3C

R=

Lovastatin

R
O

O
O
CH3

CH3
H3C

H3C

R=

H3C

Simvastatin

O
CH3
H3C
R=

D313C

[13CD3] Simvastatin

Figure 4. Similarity of simvastatin and lovastatin structures, and stable-isotope labeling of simvastatin.

content and pH, solvents and mass spectrometer


parameters, but also the materials of the sample and
solution containers. Generally, to reduce sodium content
to favor the formation of [M+H]+ ions, containers made
of glass and solutions or solvents containing an alcohol,
such as methanol, should be avoided throughout sample
preparation and analysis [31,35].
There were some attempts to favor the occurrence of
[M+NH4]+ by addition of ammonium ions into the
mobile phase [31]; however in this extensive study
methylammonium adduct [M+CH3NH3]+ was found to
be more convenient than other alkylammonium and
ammonium adducts tested. It was therefore chosen as a
precursor ion in this study and in one other work [33].
Monitored SRM transitions differed among the analytical
methods, and the details can be seen in Table 3.
The choice of precursor ion is important as well as
chromatographic and mass resolution [26]. In the case
of simvastatin, where mass unit 18 corresponds the
difference between lactone and hydroxy acid form and it
is, by coincidence, different by only one mass unit from
the 17 mass unit, which corresponds to the difference
between [M + H]+ and [M + NH4]+ ions of the lactone or
the hydroxy acid form arising in positive ionization
mode. The specificity of selected SRM must be verified or
chromatographic resolution of the acid and lactone form
is required [26].
Most LC-MS quantitative methods determined simvastatin hydroxy acid together with simvastatin, except for
one method, where only simvastatin was determined
[30]. Simvastatin and its hydroxy acid were separated on
C18 analytical column in all cases. Acetonitrile was
usually present in mobile phase at more than 70%.
Volatile additives including ammonium acetate, formic
362

http://www.elsevier.com/locate/trac

acid and methyl-ammonium acetate were added in order


to enhance ionization and to get better sensitivity of the
method. Ammonium acetate buffer often had pH of 4.5
in order to minimize the interconversion between simvastatin and simvastatin hydroxy acid, because it is
much stronger than in the case of atorvastatin. Analytical runs were very short, ranging from 2.5 min up to
5 min. Isocratic elution was applied in most cases.
The best ISs for precise and accurate quantitation in
MS are stable-isotope-labeled standards. In the case of
simvastatin labeling usually occurs at one of the methyl
group, [13CD3]-simvastatin is obtained [31,32], see
Fig. 4. Four works employed deuterium-labeled ISs [31
34]. All other works utilized lovastatin and lovastatin
acid ISs, as its high stated above, because of its high
similarity with the structure of simvastatin Fig. 4.
Method sensitivity expressed as LOQ was typically about
0.05 ng/ml in almost all methods.
4.2. Atorvastatin
Analytical methods, even those newly developed, for
the determination of atorvastatin and its five possible
metabolites, including the lactone form of atorvastatin,
p-hydroxyatorvastatin and o-hydroxyatorvastatin and
their lactone forms (overview in Table 2), still employ
HPLC-UV detection in spite of its lower sensitivity.
However, such methods could be successfully applied
for the analysis of atorvastatin in drug substances or
drug formulations together with impurities, as reported by Ertuk et al. [36], Mohammadi et al. [37]
(where amlodipine was also determined in combined
tablet formulation), as it was in the work of Chaudhari et al. [38] but without the determination of
impurities.

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Trends

Table 3. Specific SRM transition for the determination of simvastatin, its various precursor ions, simvastatin hydroxy acid and their stable-isotope
labeled forms
Compound determined
Simvastatin

Molecular weight

Precursor ion
+

418.27

[M + H]

[M + Na]+
[M + CH3NH3]+
[M + CH3CN + Na]+
[13CD3]-simvastatin

[M + H]+

422.27

[M + CH3NH3]+
Simvastatin acid

436.27

[M H]

[13CD3]-simvastatin acid

440.27

[M + H]+
[M H]

Two HPLC-UV assays for the determination of atorvastatin in biological materials have been published.
Determination of atorvastatin in human serum using
diclofenac as the IS was published by Bahrami et al. [39]
with an LOQ of 4 ng/ml. Altuntas et al. developed a
method for the determination of atorvastatin in human
serum, bulk drug and tablets using ibuprofen as the IS
with an LOQ of 18 ng/ml [40].
HPLC-UV methods generally utilize the C18 stationary
phase. In most cases, a combination of acetonitrile and
buffer (ammonium acetate or phosphate) with the aim of
keeping pH between 4 and 5. Both isocratic and gradient
elution has been utilized. Detection has usually been
performed at 237 nm [20,37], 247 nm [36,39] or 254
nm [38]. Analytical run times have been very variable,
the shortest about 34 min [39,41], the longest about
30 min [36]. None of HPLC-UV methods also determined
the active metabolites of atorvastatin or its lactone form.

Specific SRM transition (SIM)

Ref.

419.1 285.1
419.1 199.1
441 (SIM)
450.3 285.1
481.2 440.9

[29,32]
[34]
[30]
[31,33]
[28]

423.2 285.1
423.2 199.1
454.3 285.1

[32]
[34]
[31,33]

435.3 114.0
435.3 319.1
437.3 303
439.2 319.1

[28]
[3134]
[29]
[3134]

Fluorescence detection has not been employed in the


determination of atorvastatin, probably due to the difficulties with the derivatization step and the repeatability
of the procedure.
Methods about 10100 times more sensitive have
been developed using MS/MS detection, most of which
were for bio-analytical applications, determining atorvastatin in human plasma. One method referred to the
determination of atorvastatin together with other drugs
(novobiocin and roxytromycin in aqueous samples [41]
for the environmental applications).
Bio-analytical methods [18,4246] have, in all cases,
employed MS/MS using specific SRM conditions with
electrospray ionization in positive-ion mode (ESI+). This
may be surprising, taking into account the carboxylic
acid group present in the structure of atorvastatin.
However, several works confirmed that no higher
sensitivity was obtained with electrospray ionization in

Table 4. Specific SRM transition for the determination of atorvastatin, its metabolites and their stable-isotope-labeled forms
Compound determined

Molecular weight

Precursor ion
+

Specific SRM transition

Ref.

559.2 440.2
564.2 440.2
564.2 445.2
541.2 448.2
546.2 448.2
546.2 453.2

[18,4246]
[42]
[43,46]
[18,42,46]
[42]
[46]

Atorvastatin
[d5]-atorvastatin

558.25
563.25

[M + H]
[M + H]+

Atorvastatin lactone
[d5]-atorvastatin lactone

540.25
545.25

[M + H]+
[M + H]+

p-hydroxy atorvastatin
[d5]-p-hydroxyatorvastatin
p-hydroxyatorvastatin lactone
[d5]-p-hydroxyatorvastatin lactone

574.25
579.25
556.25
561.25

[M + H]+
[M + H]+
[M + H]+
[M + H]+

575.2 440.2
580.2 445.2
557.2 448.2
562.2 453.2

[18,4246]
[42,43,46]
[18,42,46]
[42,46]

o-hydroxyatorvastatin

574.25

[M + H]+

[d5]-o-hydroxyatorvastatin
o-hydroxyatorvastatin lactone
[d5]-o-hydroxyatorvastatin lactone

579.25
556.25
561.25

[M + H]+
[M + H]+
[M + H]+

575.2 440.2
575.2 466.2
580.2 445.2
557.2 448.2
562.2 453.2

[4246]
[18,44]
[42,43,46]
[18,42,46]
[42,46]

http://www.elsevier.com/locate/trac

363

Trends

364
http://www.elsevier.com/locate/trac

Table 5. Extraction procedures utilized for sample preparation during simvastatin analysis
Matrix

Extraction
procedure

Stationary phase
LLCE cartridge

SPE eluent/LLE
extraction agent

Stabilization/
pre-extraction
treatment

Interconversion
data

Validation data:
recovery

Ref.

SV, SVA
LOV, LOVA, PRA

Pu-Ehr tea

SPE

1. Extraction in water at 100C


2. Supelco DSC C18

90% MeOH

None

Given at various
conditions

recSV = 95.098.9%

[11]

SV, AT
LOV, PRA

Aqueous samples

SPE

1. Filtration
2. Oasis HLB

MeOH

pH 4.5 by 3.0 M H2SO4

1%

recSV = 6984%

[19]

SV, SVA

Human plasma

SPE

C8

1. MeOH, water
6:4 2. ACN

None

Not significant

recSV = 40%
recSVA = 40%

[21]

SV, SVA

Human plasma

LLE

ACN:water
(60:40) ACN

None

Not given

recSV = 95.296.3%
recSVA = 92.895.1%

[22]

SV

Human plasma

LLE

diethylether

10 M HCl

Not given

recSV = 86.990.7%

[23]

SV, SVA

Human plasma

SPE

Oasis HLB

ACN 0.1 M AmAc


pH 4.5 (75:25)

None

Not given

recSV = 88.8%
recSVA = 85.6%

[28]

SV, SVA

Human plasma

on-line SPE

Oasis HLB

ACN 3.0 mM
FAc (10:90)

0.1 M sodium acetate pH 4.2

<1%

recSV P 75%
recSVA P 38%

[29]

SV

Human plasma

LLE

Et-Ac

None

Not given

rec = 101.4%

[30]

SV, SVA

Plasma

LLCE

ChemElut cartridge

MTBE

100 mM AmAc pH 4.5

<0.07%

recSV = 78%
recSVA = 87%

[31]

SV, SVA

Human plasma

LLCE

ChemElut cartridge

MTBE

100 mM AmAc pH 4.5

<0.07%

recSV = 66%
recSVA = 73%

[32]

SV, SVA

Human plasma

ulv-SPE

Oasis HLB l-elution


96-well SPE plate

ACN:water (95:5)

100 mM AmAc pH 4.5

<0.08%

recSV = 66%
recSVA = 73%

[33]

SV, SVA

Human plasma

LLE

96-well LLE plate

MTBE

100 mM AmAc pH 5.0

<0.06%
lactonization
<0.07%
hydrolysis

recSV = 87.1%
recSVA = 72.7%

[34]

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Substances
isolated

http://www.elsevier.com/locate/trac

Substances isolated

Matrix

Extraction
procedure

Stationary phase

SPE eluent LLE extraction agent

Stabilization /preextraction treatment

Interconversion
data

Validation data:
recovery

Ref.

AT, SV
LOV, PRA

Aqueous samples

SPE

1. Filtration - Soxhlet
2. Oasis HLB

MeOH

pH 4.5 by 3.0 M H2SO4

10%

recAT = 6486%

[19]

AT

Human serum

LLE

Et-Ac

0.1 M phosphate
buffer pH 7.0

Not given

recAT = 95%

[39]

AT novobiocin
roxytromycin

Aqueous samples

SPE

1. Filtration - Soxhlet
2. HLB

MeOH

pH 4.0 by
3.5 M H2SO4

Not given

recAT = 70.572.2%

[41]

AT, ATA
2-OH-AT/L
4-OH-AT/L

Human serum

LLE

MTBE

0.1 M sodium
acetate buffer pH 5.0

Given at various
conditions

rec = 60100%
all analytes

[42]

AT o-OH-AT,
p-OH-AT

Human, dog
and rat plasma

LLE

Diethyl ether

0.1 M NaOH

Given at various
conditions

recAT = 100107%

[43]

AT, ATA
p-OH AT/L
o-OH AT/L

Human plasma

SPE

Varian C18

ACN 0.1 M AmAc (95:5)

1 M sodium
formate pH 3

<4%

rec = 5378%
all analytes

[18]

AT
p-OH AT
o-OH AT

Human plasma

LLE

Diethyl ether
dichloromethane (7:3)

10% o-phosphoric acid

Not given

recAT = 54.2%

[44]

AT
2-OH-AT

Human plasma

LLE

Diethyl ether
dichloromethane (60:40)

0.01 M sodium
acetate pH 6.0

Not given

recAT = 89.692.5%

[45]

AT, AT-L
p-OH AT/L
o-OH AT/L

Human plasma

SPE

Isolute C-18

95% MeOH

100 mM AmAc
pH 4.6

Not given

Not given

[46]

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

Table 6. Extraction procedures utilized for sample preparation during atorvastatin analysis

Trends

365

Trends

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008

negative-ion mode (ESI ), probably because this compound contained two nitrogen groups [42,43]. The
precursor ion chosen for quantitation was [M+H]+ in all
methods developed (see Table 2). The monitored SRM
transition for atorvastatin therefore utilized 559 440
(which was also the most intensive transition in all
methods).
Compared to HPLC-UV methods, all LC-MS/MS
methods determined atorvastatin together with its phydroxy and o-hydroxy metabolites, except [45] where
only the o-hydroxy metabolite was determined. All
metabolites including hydroxy acid and lactone forms
were determined by Jemal et al. [42], Hermann et al.
[18] and Van Pelt et al. [46]. Atorvastatin metabolites
were all also determined in ESI+ using [M + H]+ as a
precursor ion; the specific transition for each metabolite
and their stable isotope-labeled forms can be seen in
Table 4.
Atorvastatin and its metabolites have been separated
on the C18 analytical column using acetonitrile usually about 70% (to shorten retention times of analytes;
containing methanol only they would be eluted much
later) as part of the mobile phase, together with acetic
or formic acid as additives to enhance ionization. Both
isocratic and gradient elution have been utilized. Analytical run times were, in most cases, about 6 min or less,
except for one method [18].
The best ISs for precise and accurate quantitation in
MS are stable-isotope-labeled standards. In the case of
atorvastatin and its metabolites, [d5] labeling usually
occurs on the phenyl ring, which does not contain
fluorine [42,43]. Only three works employed deuteriumlabeled ISs [42,43,46] because they were very expensive
and sometimes they were not easily available. Other
works utilized ISs of various structures, including
metachalon [18], rosuvastatin [44] or clindamycin [46].
Method sensitivity expressed as LOQ was quite similar for
all methods, typically in the range 0.5 ng/ml [42] to 0.1
ng/ml [45]. [Please check this range?]

5. Sample preparation
A convenient sample-preparation procedure should isolate the analytes from the complex matrix while
removing endogenous interfering substances. This is
often the most time-consuming, critical step of analysis.
The analytes should be pre-concentrated in order to increase the sensitivity and the selectivity of the method.
The procedures for sample preparation of simvastatin
and atorvastatin have in most cases included LLE (liquidliquid extraction), and SPE (solid-phase extraction).
However, simple extraction into the organic solvent has
also been used in pharmaceutical applications
[20,24,27,3638,40] see Tables 1 and 2. There have
also been specific approaches to sample preparation (e.g.,
366

http://www.elsevier.com/locate/trac

LLCE (liquid-liquid cartridge extraction) [31,32] or online SPE utilizing a column-switching technique).
Tables 5 and 6 give overviews of LLE and SPE samplepreparation techniques utilized for the isolation of simvastatin and atorvastatin, focusing on data recovery,
interconversion during sample preparation (details given
in the work of Herman et al. [18], Zhao et al. [32] and
Zhang et al. [34]) and stabilization data.
Statins have mostly been isolated using SPE on the
Oasis HLB cartridge, employing methanol for elution, or
using LLE, where MTBE, ethyl acetate or diethyl ether
have generally been applied as extraction agents.

6. Conclusions
Analysis of statins is a current problem, because these
drugs are widely used for the treatment of hypercholesterolemia.
This review has presented HPLC methods for the
determination of simvastatin and atorvastatin in various
fields of application (pharmacology, clinical medicine
and environmental). We divided the methods according
to the type of detection. We discussed interconversion
between acid and lactone form of statins.
LC-MS/MS methods have undoubtedly become the
method of choice. In order to get high selectivity and
sensitivity, MS/MS techniques employ specific SRM
conditions, which are convenient, especially in bioanalytical applications. There is no problem in determining the metabolites, even if they are not also sufficiently separated chromatographically.
Chromatographic conditions must be carefully arranged, as must selection of a precursor ion for a specific
SRM transition in order not retain the sensitivity and the
selectivity of the method.

Acknowledgement
The authors gratefully acknowledge the financial support of IGA MZ CR No. 1A/8689-4.
References
[1] L.Y. Ye, P.S. Firby, M.J. Moore, Ther. Drug. Monit. 22 (2000) 737.
[2] Y. Shitara, Y. Sugiyama, Pharmacol. Therapeut. 112 (2006)
71.
[3] K.J. Williams, I. Tabas, Atherosclerosis Thrombosis Vascular Biol.
15 (1995) 551.
[4] A.A. White, Stats. Med. 14 (1995) 697.
[5] D.J. Betteridge, P.M. Dodson, P.N. Durrington, E.A. Hughes, M.F.
Laker, D.P. Nicholls, J.A. Rees, C.A. Seymour, G.R. Thompson,
A.F. Winder, Postgrad. Med. J. 69 (1993) 359.
[6] World Health Organization, World Health Report, Report of the
Director-General, WHO, Geneva, Switzerland, 1998.
[7] F.M. Sacks, Am. J. Cardiol. 88 (suppl.) (2001) 14N.

Trends in Analytical Chemistry, Vol. 27, No. 4, 2008


[8] S. Bertolini, G.B. Bon, L.M. Campbell, M. Farnier, J. Lagan,
G. Mahla, Atherosclerosis 130 (1997) 191.
[9] A. Endo, M. Kuroda, Y. Tsujita, J. Antibiot. 29 (1976) 1346.
[10] M. Jemal, Y.Q. Xia, J. Pharm. Biomed. Anal. 22 (2000) 813.
[11] D.J. Yang, L.S. Hwang, J. Chromatogr., A 1119 (2006) 277.
[12] S. Erturk, A. Onal, S.M. Cetin, J. Chromatogr, B 793 (2003) 193.
[13] R. Nirogi, K. Mudigonda, V. Kandikere, J. Pharm. Biomed. Anal.
44 (2007) 379.
[14] W. Jacobsen, B. Kuhn, A. Soldner, G. Kirchner, K.F. Sewing,
P.A. Kollman, L.Z. Benet, U. Christians, Drug. Metab. Dispos.
28 (2000) 1369.
[15] S. Khan, D.S. Teitz, M. Jemal, Anal. Chem. 70 (1998) 1622.
[16] H.F. Gilbert, Methods Enzymol. 251 (1995) 8.
[17] M.J. Morris, J.D. Gilbert, J.Y.K. Hsieh, B.K. Matuszewski,
H.G. Ramjit, W.F. Bayne, Biol. Mass. Spectrom. 22 (1993) 1.
[18] M. Hermann, H. Christensen, J.L.E. Reubsaet, Anal. Bioanal.
Chem. 382 (2005) 1242.
[19] X.S. Miao, C.D. Metcalfe, J. Chromatogr., A 998 (2003) 133.
[20] M.K. Pasha, S. Muzeeb, S.J.S. Basha, D. Shashikumar, R. Mullangi,
N.R. Srinivas, Biomed. Chromatogr. 20 (2006) 282.
[21] H. Ochiai, N. Uchiyama, K. Imagaki, S. Hata, T. Kamei, J.
Chromatogr., B 694 (1997) 211.
[22] G. Carlucci, P. Mazzeo, L. Biordi, M. Bologna, J. Pharm. Biomed.
Anal. 10 (1992) 693.
[23] B.C. Kim, E. Ban, J.S. Park, Y.K. Song, C.K. Kim, J. Liq.
Chromatogr. Relat. Technol. 27 (2004) 3089.
[24] A. Malenovic, M. Medenica, D. Ivanovic, B. Jancic, Chromatographia Suppl. 63 (2006) S95.
[25] H. Wang, Y. Wu, Z. Zhao, J. Mass Spectrom. 36 (2001) 58.
[26] M. Jemal, Z. Ouyang, Rapid Commun. Mass Spectrom. 14 (2000)
1757.
[27] M. Vuletic, M. Cindric, J.D. Koruznjak, J. Pharm. Biomed. Anal. 37
(2005) 715.
[28] B. Barrett, J. Huclova, V. Borek-Dohalsky, B. Nemec, I. Jelnek,
J. Pharm. Biomed. Anal. 41 (2006) 517.

Trends
[29] M. Jemal, Z. Ouyang, M.L. Powell, J. Pharm. Biomed. Anal. 23
(2000) 323.
[30] H. Yang, Y. Feng, Y. Luan, J. Chromatogr., B 785 (2003) 369.
[31] J.J. Zhao, A.Y. Yang, J.D. Rogers, J. Mass Spectrom. 37 (2002)
421.
[32] J.J. Zhao, I.H. Xie, A.Y. Yang, B.A. Roadcap, J.D. Rogers, J. Mass
Spectrom. 35 (2000) 1133.
[33] A.Y. Yang, L. Sun, D.G. Musson, J.J. Zhao, J. Pharm. Biomed.
Anal. 38 (2005) 521.
[34] N. Zhang, A. Yang, J.D. Rogers, J.J. Zhao, J. Pharm. Biomed. Anal.
34 (2004) 175.
[35] Y. Wu, J. Zhao, J. Henion, W.A. Korfmacher, C.C. Lin, J. Mass
Spectrom. 32 (1997) 379.
[36] S. Erturk, E.S. Aktas, L. Ersoy, S. Ficicioglu, J. Pharm. Biomed.
Anal. 33 (2003) 1017.
[37] A. Mohammadi, N. Rezanour, M.A. Dogahen, F.G. Bidkorbeh,
M. Hashem, R.B. Walker, J. Chromatogr., B 846 (2007) 215.
[38] B.G. Chaudhari, N.M. Patel, P.B. Shah, Chem. Pharm. Bull. 55
(2007) 241.
[39] G. Bahrami, B. Mohammadi, A. Mirzaeei, A. Kiani, J. Chromatogr., B 826 (2005) 41.
[40] T.G. Altuntas, N. Erk, J. Liq. Chromatogr. Relat. Technol. 27
(2004) 83.
[41] X.S. Miao, C.D. Metcalfe, J. Mass Spectrom. 38 (2003) 27.
[42] M. Jemal, Z. Ouyang, B.C. Chen, D. Teitz, Rapid Commun. Mass.
Spectrom. 13 (1999) 1003.
[43] W.W. Bullen, R.A. Miller, R.N. Hayes, J. Am. Soc. Mass. Spectrom.
10 (1999) 55.
[44] R.V.S. Nigori, V.N. Kandikere, M. Shukla, K. Mudigonda,
S. Maurya, R. Boosi, Y. Anjaneyulu, Biomed. Chromatogr.
20 (2006) 924.
[45] V. Borek-Dohalsky, J. Huclova, B. Barret, B. Nemec, I. Ulc,
I. Jelnek, Anal. Bioanal. Chem. 386 (2006) 275.
[46] C.K. Van Pelt, T.N. Corso, G.A. Schultz, S. Lowes, J. Henion, Anal.
Chem. 73 (2001) 582.

http://www.elsevier.com/locate/trac

367

You might also like