Вы находитесь на странице: 1из 24

Anal Bioanal Chem (2010) 398:2952

DOI 10.1007/s00216-010-3741-5

REVIEW

Recent advances of capillary electrophoresis


in pharmaceutical analysis
Leena Suntornsuk

Received: 18 February 2010 / Revised: 8 April 2010 / Accepted: 9 April 2010 / Published online: 2 May 2010
# Springer-Verlag 2010

Abstract This review covers recent advances of capillary


electrophoresis (CE) in pharmaceutical analysis. The principle, instrumentation, and conventional modes of CE are
briefly discussed. Advances in the different CE techniques
(non-aqueous CE, microemulsion electrokinetic chromatography, capillary isotachophoresis, capillary electrochromatography, and immunoaffinity CE), detection techniques
(mass spectrometry, light-emitting diode, fluorescence,
chemiluminescence, and contactless conductivity), on-line
sample pretreatment (flow injection) and chiral separation
are described. Applications of CE to assay of active pharmaceutical ingredients (APIs), drug impurity testing, chiral
drug separation, and determination of APIs in biological
fluids published from 2008 to 2009 are tabulated.
Keywords Capillary electrophoresis .
Active pharmaceutical ingredient . Impurity .
Chiral separation . Biological fluid

Introduction
Chemical analysis is a discipline that is strongly associated
with drug discovery, and pharmaceutical control and
assurance, in order to provide the highest efficacy and
safety for patients. Unfortunately, counterfeit and substandard drugs are still available, with a value of up to

L. Suntornsuk (*)
Department of Pharmaceutical Chemistry, Faculty of Pharmacy,
Mahidol University,
447 Sri-Ayudhaya Rd, Rajathevee,
Bangkok 10400, Thailand
e-mail: pylll@mahidol.ac.th

40 billion US$ annually [1]. Among these, 32.5% may


not contain any active pharmaceutical ingredients (APIs),
20.4% may contain incorrect quantity of APIs, 21.7%
might contain the wrong APIs, 15.8% are in the wrong
packaging, 1.0% are copies of the original, and 8.6% contain impurities and contaminants. Quality control of pharmaceutical entities and products (qualitative and quantitative
analyses, purity testing, chiral separation, related substance
and stoichiometric determination) is, therefore, of paramount
importance clinically, pharmacologically, and to the pharmaceutical industry.
Capillary electrophoresis (CE) is a powerful analytical
method with significant importance in drug discovery and
manufacturing processes because of its unique separation
mechanism, speed, efficiency, and versatility. CE separation
depends on different migration of solutes in an electric field
and electrophoresis is performed in narrow-bore capillaries
filled with background electrolyte (BGE). Driving forces in
CE are electrophoretic migration and the electro-osmotic
flow (EOF). Compared with other techniques, the instrumentation of CE is simple consisting of electrodes, sampleintroduction systems, a capillary, a power supply, a detector,
and liquid-handling systems. Detection can be achieved
with on-line (UV/diode-array spectrophotometric, spectrofluorimetric and electrochemical detectors) or external
detectors (mass spectrometer, MS). Common CE modes are
capillary zone electrophoresis (CZE, based on analytes
charge-to-mass ratios), micellar electrokinetic chromatography (MEKC, based on chromatographic partition of analytes
between micelles and the background electrolyte), capillary
isoelectric focusing (CIEF, based on analytes isoelectric
points), capillary gel electrophoresis (CGE, based on
analytes size/molecular weight ratio) and capillary isotachophoresis (CITP, based on moving boundaries).

30

Advances of capillary electrophoresis in pharmaceutical


analysis
CE has grown from a simple to a sophisticated method
because of several innovations during recent decades.
This growth has expanded applications of CE to various
analytes including pharmaceuticals. Various techniques
have been established to improve CE selectivity, sensitivity,
and applicability. These include non-aqueous CE (NACE,
based on the use of pure or mixed organic solvents as
the BGE), microemulsion electrokinetic chromatography
(MEEKC, based on chromatographic partition of analytes
between microemulsion droplets and the aqueous BGE),
CITP, capillary electrochromatography (CEC, based on
chromatographic partition of analytes between stationary
phases and the BGE), and immunoaffinity capillary
electrophoresis (IACE). Different means of detection (MS,
light-emitting diode (LED), fluorescence, chemiluminescence (CL), and contactless conductivity (C4D) detectors),
and on-line sample pretreatment (flow injection, FI) have
also contributed to sensitivity and selectivity enhancement
in CE. Importantly, development of novel chiral selectors
(charged cyclodextrins (CD), dual CD systems, CDmediated MEKC and chiral surfactants) greatly facilitate
enantioseparation of chiral drugs.
Variation of capillary electrophoresis modes
A major advantage of CE is the availability of various
modes, which enable the separation and analysis of different compounds ranging from inorganic species to
biopolymers. It is well accepted that CZE and MEKC are
still mainly employed for drug analysis. Nevertheless, the
uses of other CE modes (NACE, MEEKC, CITP, CEC, and
IACE) are increasing and become valuable in this area
in order to improve the separation efficiency, selectivity,
sensitivity, or flexibility of CE.
Non-aqueous capillary electrophoresis (NACE)
NACE is a complimentary method to CZE. The use of
organic solvents, instead of water, in the BGE enhances
hydrophobic analyte solubility and method selectivity.
Analytes with similar charge-to-size ratios or analytes that
are degraded in aqueous media can be separated in nonaqueous media. CE selectivity can be easily manipulated
because of the different degrees of dissociation of the
electrolyte, ion-pairing effects, and ion conductivity in
organic solvents, because they usually have lower dielectric
constants than water [2]. Moreover, higher potentials can be
used to reduce analysis times, because a smaller electric
current is generated in organic solvent. NACE has attracted
analysts, because it provides better compatibility with

L. Suntornsuk

samples and different detectors, improves selectivity, and


generates a lower electric current. Geiser et al. recently
published two reviews on NACE with one focusing on
pharmaceutical analysis (drugs and related substances,
chiral substances, phytochemical extracts, and drugs in
biological fluids) [2, 3]. NACE has been applied to the
determination of fluoxetine, sertraline, citalopram, and
paroxetine in plasma [4], anesthetic drugs in plasma [5],
S-timolol and 1R,2S-ephedrine in raw material [6], eight
antidepressants in human serum [7], and five fluoroquinolones in urine [8].
Microemulsion electrokinetic chromatography (MEEKC)
MEEKC is a variant of CE in which microemulsion droplets are used as pseudo-stationary phases. Microemulsion
droplets, either the common oil-in-water or water-in-oil, are
thermodynamically stable transparent nanodroplets. These
droplets consist of oil and aqueous phase that are stabilized
by surfactants and co-surfactants. MEEKC differs from
MEKC in that microemulsion droplets are more flexible
and can swell better than micelles, providing a wider
separation window and higher resolution [9]. However,
preparation of the microemulsion can be complicated and
time-consuming. MEEKC is useful for analysis of neutral
and hydrophobic compounds and the determination of log
P values. Advances in MEEKC method development and
applications, including pharmaceutical analysis and chiral
separation, have been reviewed by Ryan et al. [9] and
McEvoy [10]. Applications of MEEKC in drug analysis
include the determination of coenzyme Q10 in human
plasma [11], dopa, phenylalanine, and tyrosine [12], paracetamol and its impurity in a suppository [13], and watersoluble and fat-soluble vitamins in formulations [14].
Capillary isotachophoresis (CITP)
CITP can be used as a preconcentration step to enhance
detection sensitivity prior to real separation. This is essential for trace and residue analysis such as biological and
environmental samples. The concept of CITP depends on
the analytes conductivity and the analyte zones are sandwiched between a leading electrolyte (containing highmobility ions) and a terminating electrolyte (containing
low-mobility ions). Analytes then migrate into discrete
zones according to their mobilities. Gebauer et al. and Mala
et al. have regularly published CITP reviews since 1997
[1521]. Other reviews, including one on miniaturized ITP,
have recently been published [2226]. CITP has applications
in analysis of amlodipine in urine [27, 28], neomycin
trisulfate in dosage forms [29], chiral separation of dimethindene and pheniramine enantiomers in dosage forms and
clinical samples [30, 31], peptide purity and counter ion

Recent advances of capillary electrophoresis in pharmaceutical analysis

contents in lecirelin (luteinizing hormone-releasing hormone


analog) [32], alendronate and pamidronate in aqueous
solution and urine [33], and 4-aminophenol (an impurity of
acetaminophen) in bulk and tablets [34].
Capillary electrochromatography (CEC)
CEC is a hybrid of CE and high performance liquid
chromatography (HPLC), which provides both selectivity
and efficiency. Stationary phase can be packed into columns or immobilized on capillary walls, when it is known
as open-tubular CEC. In addition to reversed-phase stationary phases (namely octadecylsilane, ODS), mixed-mode
stationary phases (a combination of hydrophobic and ionexchange interactions) have been developed, which increases applications of CEC in biochemical, omic, food,
phytochemical, pharmaceutical, forensic, and environmental research [35]. Monolithic columns prepared from
synthetic polymers have become a popular format of CEC
stationary phase, instead of packed capillaries [36]. Tanret
et al. reviewed monolithic stationary phases in CEC and
pressurized CEC (pCEC) [37]. Recently, the development
of molecular imprinted polymer (MIP) for CEC, including
organic polymer-based and silica-based MIP, has been
reviewed by Huang et al. [38]. CEC has been successfully
used for assays of steroid hormones [39], active compounds
in troxerutin tablets [40], and basic drugs in human serum
[41]. MIP in CEC has been used for chiral separation of
propranolol [42], naproxen [43], and ibuprofen, naproxen
and fenoprofen [44], ofloxacin [45], ephedrine and norephedrine isomers [46], illicit drugs [47, 48], oxytocin,
bovine serum albumin, bovine hemoglobin, ovalbumin,
and lysozyme [49], and tetrahydropalmatine and Trogers
base [50]. CEC with packed bead beds in microfluidic
devices has also been proposed for analysis of fluoresceinyl
isothiocyanate (FITC)-insulin, FITC-immunoglobulin G
(IgG), and their impurities using size-exclusion electrochromatography and strong cation-exchange electrochromatography [51]. Mesoporous silica nanoparticles immobilized
open-tubular capillary column with a coating of cellulose tris
(3,5-dimethylphenylcarbamate) for separation of enantiomers
in CEC was investigated for separation of the enantiomers of
several test racemates including indapamide, Trogers base,
praziquantel, tetrahydropalmatine, warfarin, and pindolol
[52]. CEC using a pentaerythritol diacrylate monostearatethylene dimethacrylate monolith has been used to separate
, , , and -tocopherols and -tocopherol acetate in
pharmaceutical preparations [53].
Immunoaffinity capillary electrophoresis (IACE)
IACE has emerged as an attractive alternative for CE
sensitivity and selectivity improvement because it combines

31

the advantages of immunoassay and separation techniques


[54]. The IACE setup consists of a concentrator (for sample
cleanup and enrichment) and a microreactor (for derivatization, chemical synthesis, or enzymatic reaction). Several
analytes can be simultaneously quantified and characterized
by serial application of samples to an array of immunoaffinity analyte concentrators housed within one instrument
[5560]. Nowadays, an analyte concentratormicroreactor
can be combined on-line with capillaries or microchip
channels, which makes IACE more powerful because of
automation, high-throughput, and parallel multiplex operation. IACE has been applied to the determination of
enantioselective binding of basic drugs to plasma [61],
analyses of neurotrophins in serum [62], total serum IgE
using magnetic bead-based IACE [63], recombinant human
erythropoietin and novel erythropoiesis stimulating protein
digest using IACEMS [64], androgenic anabolic steroids
[65], and hormones in biological fluids (blood, saliva and
urine) [66].
Detection techniques
Mass spectrometry (MS)
Coupling of separation techniques (HPLC, CE, and gas
chromatography (GC)) with MS has become a powerful
tool in chemical and biological assays. Although UV
detection (either direct or indirect measurements) is the
most widely used detector in CE, its sensitivity is low
because signals are directly related to an optical path length
afforded by an internal diameter of a capillary [67]. To
overcome this problem, an MS detector can be an interesting
choice because it provides higher sensitivity and capability to
differentiate overlapping peaks with distinct mass-to-charge
ratios (m/z). MS furnishes molecular weight and/or structural
information, which can be used for identification of unknowns or confirmation of identity.
Initial CEMS interfaces were complicated and sometimes irreproducible because a stable electrical contact at
the ion source-end of the capillary must be maintained.
In addition, large amounts of salts in the BGE can cause
ion suppression and contaminate the valuable ion
sources. Several developments have overcome these
problems, making CEMS a robust and sensitive method. CEMS interfaces mainly involve sheath-flow (coaxial sheath-flow and liquid-junction interfaces) and
sheathless interfaces. Three types of ion source are used
in CEMSatmospheric pressure ionization (electrospray
(ESI), atmospheric pressure chemical ionization (APCI) and
atmospheric pressure photoionization (APPI)), matrixassisted laser desorption ionization (MALDI), and inductively coupled plasma ionization. Mass analyzers in MS
include quadrupole, ion-trap (IT), time-of-flight (TOF), and

32

Fourier transform ion cyclotron resonance (FT-ICR).


Among these, ESI using a sheath flow interface is
commonly used for CEMS, whereas TOF provides high
data acquisition rate, high speed, high duty cycle, high
sensitivity, and no mass range limits [67]. Various aspects
of CEMS have been reviewed by several groups, for
example, CETOF/MS [68], trends in CEMS [68], NACE
MS [69], MEEKCMS [70], and CE and microchip-based
MS [71].
CEMS is gaining popularity for pharmaceutical
analysis because of the high sensitivity and structureinformative nature of the detector. This is extremely
valuable for drug discovery, pharmacokinetic studies,
drug impurity profiling, and quantification of APIs and
their metabolites in biological samples. Reviews on
applications of CEMS in clinical diagnosis [72], drug
analysis [73, 74], and small achiral and chiral solutes [75]
have been described. Studies on the ESI behavior of
selected drugs and their applications in CEMS have been
described by Smyth and Rodriguez [76]. CEMS has been
applied to analysis of illicit drugs in hair [77], drugs of
abuse and their metabolites in hair [78], short-chain
carnitine in human plasma [79], enantiomeric separation
of baclofen in tablets [80], and omeprazole in capsules
[81].
A well known benefit of CZE is the separation of
ionizable analytes, whereas MEKC and MEEKC are for
neutral species. MEEKC can be advantageous over
MEKC in terms of solubilizing power and large
migration window. Hyphenation of MEKC or MEEKC
with MS can further enhance method sensitivity and
selectivity. However, surfactants in MEKC or MEEKC
can cause several problems in MS, including ion source
contamination and baseline noise. Volatile surfactants,
low-molecular weight (unpolymerized) surfactants at low
concentrations [82], or partial-filling high-molecularweight surfactants (oligomers of monomeric surfactants
or polymeric sulfated amino acid surfactants) [8387], and
use of APCI or APPI have been proposed to overcome
these problems.
Schappler et al. compared CZEESI-MS, MEEKC
ESI-MS, and MEEKCAPPI-MS, in both positive and
negative modes, for separation of three -blockers, two
central stimulants, and nine diuretics [70]. the MEEKC
system consisted of 2.33% (w/w) SDS (equiv.
80 mmol L1), 7.34% (w/w) butan-1-ol (800 mmol L1),
and 0.82% (w/w) n-octane (50 mmol L1) in 10 mmol L1
ammonium acetate buffer (89.51% w/w). CZEESI-MS
either positive (using 15 mmol L1 ammonium formate
buffer, pH 2.5 as the BGE) or negative (using
10 mmol L1 ammonium acetate buffer, pH 9.2 as the
BGE) mode was not sufficient to separate the analyte
mixtures, which consisted of acidic, basic, and neutral

L. Suntornsuk

compounds, because of a lack of selectivity for neutral


molecules. As expected, separation of the investigated
compounds could not be achieved by MEEKCESI-MS,
because of the incompatibility of the surfactant, which
caused ion suppression. Generally, MEEKCAPPI-MS in
both positive and negative modes provided better separation than CZEESI-MS, with different migration orders
because of the partition and electrophoresis mechanisms.
Separation of the diuretics was achieved because of their
interactions with the microemulsions. Although limited
sensitivity was achieved, peak symmetry and efficiency
were greatly improved in MEEKCAPPI-MS. Last, but
not least, APPI is an attractive ion source for coupling
MEKC or MEEKC with MS, because it is less affected by
non-volatile BGE additives than ESI.
Wan et al. demonstrated the potential of MEEKC in
drug discovery by investigating the relationship between
brain tissue partitioning and microemulsion retention
factors of 42 central nervous system drugs [88]. Microemulsion compositions consisted of 80 mmol L1 lauric
acid, 37 mmol L1 octane (0.6% v/v), and 765 mmol L1
butanol (7% v/v) in 200 mmol L1 methyl ammonium
(apparent pH 11). The MEEKCIT-MS was compared
with LC and results revealed improved correlation
between log P values and the fraction unbound in brain
from MEEKC-IT-MS. The method was valuable for
prediction of braintissue binding and lipophilicity.
CEC can offer better selectivity and efficiency because
of chromatographic and electrophoretic mechanisms. But
CECMS is not yet fully established for day-to-day
analysis and improvements on interfaces, capillary
fabrication and modification, separating materials, and
packing methods are required. However, CECMS has
recently attracted interest in drug analysis and chiral
separation and these applications have been reported, for
example, chiral separation of -blockers (propranolol,
talinolol, nadolol, and labetalol) on vancomycin stationary phase [8991], impurity profiling of drugs with a total
of 22 analytes (six APIs and their related impurities) [92],
separation of seven -blockers (propranolol, esmolol,
metoprolol, bisoprolol, carteolol, celiprolol, and atenolol)
in urine samples by pCEC [93], and separation of acidic
drugs on avidin chemically immobilized on the fusedsilica surface in affinity CEC [94].
Light-emitting diode (LED) detectors
In addition to UV and MS detectors, LED detection can
be attractive in CE. The LED provides stable output, has
a wide range of emission wavelengths and consumes low
energy with long lifetime, low-cost, and small size [95].
Xiao et al. published a review on CE with LED-based
detection (LED-based absorption and LED-induced fluo-

Recent advances of capillary electrophoresis in pharmaceutical analysis

rescence detection) [96]. The use of the LED as a light


source in CE separation was introduced during 1994 [97]
and 1995 [98] and, since 1996, Haddad et al. have
continuously reported their work on CELED-based
absorption [99107]. The technique has been applied in
drug analysis, for instance, dopamine [108, 109], octopamine in human plasma [110], phenethylamine in artificial
urine samples [111], penicillamine in tablets and human
plasma [112], riboflavin in urine [113], and propranolol in
plasma [114].
Fluorescence detection
Fluorescence detection is a highly sensitive technique,
which can detect analytes at very low levels. Unfortunately, most analytes do not fluoresce very well and must
be derivatized with florescent reagents or excited by UV
irradiation, laser or LED to improve sensitivity. Schulze
et al. gave an intensive overview on native fluorescence
detection in CE and microchip CE using deep-UV light
excitation [115]. In drug analysis, this technique has been
employed for drug screening [116], separation of diuretics
in commercial formulations and urine on a microchip CE
[117], quantification of ofloxacin and its enantiomeric
metabolites in urine [118], analysis of tramadol and its
metabolites in urine [119], determination of salicylic acid
and gentisic acid in urine [120], detection of heroin
metabolites [121], quantification of psychotropic tryptamine derivatives from ecstasy tablets [122], aliphatic
amines in serum and carcinoma cells [123], amine
metabolites in plasma, urine, and saliva [124], aminothiols
in urine [125], anticancer drugs in plasma [126], and
vigabatrin in plasma [127].
Chemiluminescence (CL) detection
CL detection is based on measurement of electromagnetic radiation obtained from a chemical reaction that
produces electronically-excited intermediates or products
[128]. CL is classified as direct CL (luminescence) and
indirect CL (sensitized CL, donation of energy to other
molecules that luminescence). Reagents widely used in
CECL are ruthenium complexes, luminol and derivatives, and peroxyoxalates. CL emission can vary with time
and in different CL systems (pH, ionic strength, composition of solvent and solution, and temperature). Limitations of CL are lack of selectivity and requirement for
strict experimental control. However, CECL is a highly
sensitive method, which is powerful for clinical diagnosis,
and for pharmaceutical, food, and environmental analysis.
CECL has been used in studies of clindamycin interactions with hemoglobin [129], analyses of isoniazid and
p-aminosalicylic acid in tablets and serum [130], roxi-

33

thromycin in capsules and urine [131], puerarin in


pharmaceuticals, urine, and plasma [132], and quinolone
residues in pig urine [133].
Contactless conductivity (C4D) detection
UV has become the most widely used detector in CE,
however it might not be applicable for non-chromophore
species for example inorganic and organic ions. C4D can be
an alternative in these cases. This detector has been known
since the 1950s and a monograph on developments of C4D
was published by Pungor in 1965 [134]. Introduction of
C4D for CE quantification by Gas et al. [135] and axial
design of C4D in CE by Zemann et al. [136] have led to an
efficient method for detection of non-chromophore species.
A major consideration for C4D detection in CE is selection
of the appropriate BGE to maintain conductivity differences
between the BGE and the analytes. At the same time,
mobility differences between the analyte ions and co-ions
in the BGE should be minimal to obtain symmetric peaks.
Progress in C4D detection has been summarized in several
reviews [137145]. During the past 10 years, new configurations of C4D (axial capacitively coupled C4D for CZE
[146], portable CEC4D [147151] and CECC4D [152
154]) have been designed and broader applications (clinical
diagnosis, food, and enantiomer separation) have been
demonstrated [142, 155, 156]. Examples of CEC4D in
pharmaceutical analysis include determination of nonchromophore antibiotics (fosfomycin and tobramycin) in
human body fluid [157, 158], analysis of lecirelin and its
impurities [32], quantification of salicylate in urine [159],
detection of valproic acid in blood and urine [160],
separation of the enantiomers of tamsulosin [161], analyses
of basic drugs and amino acids [162], glucosamine and
potassium ion [163], and suxamethonium in formulations
[164].
On-line sample pretreatment
Flow injection (FI) is a flow-based technology that is
excellent for on-line sample pretreatment and preconcentration. FI is an interesting technique for coupling with
CE because it is simple, rapid and precise, and can be
automated and miniaturized [165, 166]. FICE hyphenation was first described in 1997 by Kub et al. [167] and
Fang et al. [168], and several reviews have subsequently
been published [165, 166, 169171]. Lu et al. focused on
the combination of FI with electrophoresis using capillaries and chips, including new interfaces, applications, and
microchip-based FICE systems [165]. Kub et al.
emphasized flow/sequential injection (SI) sample treatment coupled to CE (types of interfaces, types of injection
in an FI/SI-CE interface, FI/SI interfacing to sample

34

treatment, on-line monitoring with an FI/SI-CE interface,


coupling to microchip and applications) [166]. Trojanowicz presented recent developments in electrochemical
detectors (potentiometry, voltammetry, amperometry, and
arrays of electrochemical sensors) in flow analysis,
potentiometry, amperometry, and conductivity detection
in CE and in microfluidic systems and their applications
[171]. In 2008, Kub and Hauser published a chapter on
FICE focusing in on-line coupling of flow-injection
analysis and CE (electrokinetic, hydrodynamic and dual
opposite end injection, sample pretreatment, derivatization
for detection, FImicrochip CE and fast separations in
short capillaries) and electrokinetically pumped FI (electrophoretically mediated microanalysis in capillaries and
miniaturized systems) [172]. Importantly, future improvements in reduction of sample and reagents, preconcentration techniques to reduce reaction time, and novel
potential applications are still needed in FICE. In
pharmaceutical analysis, FICE has been used for the
separation of insulin, proinsulin, and c-peptide in rat
pancreatic islet excretions [173], determination of cold
medicines in preparations [174], and investigation of a
series of drughuman serum albumin interactions [175].
Chiral separation
The chirality of enantiomeric molecules is increasingly
important in biotechnology, chemistry, agriculture, and,
especially, in the pharmaceutical industry. It is estimated
that 50% of APIs are chiral [176]. Although the
pharmacological activity and toxicity of APIs greatly
depends on their chirality, only 25% are administered
as pure enantiomers. For example, enantiomers of ephedrine, amphetamine, NSAIDs, and -blockers have different efficacy, side effects, and toxicity. Chiral separation of
APIs is, therefore, essential to obtain the desirable
enantiomers. However, separation of chiral drugs may
not be straightforward because of their similar physicochemical properties. Separation techniques with high
resolving power and efficiency, for example HPLC, gas
chromatography (GC), and CE have been used for
separation of the enantiomers of chiral drugs. Chiral
separation by electrokinetic chromatography (EKC) is
now a major application of CE because of its high
resolution, flexibility, speed, and low cost. Moreover, the
availability of various and novel chiral selectors and the
simplicity of the method make it very attractive for
separation of the enantiomers of chiral drugs. The
mechanism of separation of enantiomers by EKC is based
on the formation of the corresponding diastereoisomers,
which have different physicochemical properties. Two
different procedures have been used for resolution of
enantiomers by EKC, by indirect and direct methods. The

L. Suntornsuk

indirect method involves coupling of the enantiomers with


an auxiliary chiral derivatizing reagent (CDR) to convert
them into diastereoisomers. The diastereoisomers can then
be separated by any achiral separation technique. The
indirect method is widely used in HPLC and GC, but it is
unpopular in CE because of limitations such as time
consumption, incomplete reaction, and requirements of
very pure CDRs. However, the indirect approach can be
beneficial either to increase the sensitivity by introducing
fluorescent groups to the structures of analytes or to
modify chemical structures of analytes for advantageous
interactions. Direct separation is based on the enantioselectivity of two enantiomers with chiral selectors added to
the BGE. It should be noted that separation of enantiomers
by CE is not based on charge-to-mass (size) ratio differences of enantiomers [177]. During the electrophoretic
run, the two enantiomers form labile diastereoisomer
complexes by intermolecular interactions (electrostatic
ionion, iondipole, and dipoledipole interactions,
hydrogen-bonds, interactions) [178]. The labile
complexes then move toward the detector with different
velocities. Cyclodextrins (neutral, charged CDs, dual-CD
systems, and CD-mediated MEKC), linear polysaccharides, proteins, macrocyclic antibiotics, crown ethers, chiral
metal complexes, chiral selectors in non-aqueous solvents,
and chiral surfactants can be used as chiral selectors in the
direct method. Among these, CDs and their derivatives are
the most common chiral selectors because they are
inexpensive, and result in rapid equilibrium of the CD
solute complexes and symmetric peaks. CEC also can be
used for chiral separation. Enantiomer separation by CEC
can be performed in open-tubular capillaries (OT-CEC) or
packed capillaries (P-CEC) [179]. In OT-CEC, chiral
selectors are covalently attached to or coated on the inner
surface of a capillary. OT-CEC columns are often prepared
by physical adsorption of proteins on the capillary wall or
covalent linking of different types of CDs to the capillary
wall for enantiomer separation. In P-CEC, a chiral
stationary phase (CSP) or an achiral stationary phase in
combination with chiral mobile phase is used. CSPs are
more frequently used and are available in various forms
(CD-CSP, brush-type CSP, macrocyclic antibiotics-CSP,
polysaccharide-CSP, protein-CSP, etc). The chiral recognition mechanisms are also based on molecular interactions, as previously mentioned.
There is no need to elaborate on chiral separation by
EKC and the chiral selectors used, because excellent
reviews on these topics have been extensively published
[177, 180185]. In addition, separation of the enantiomers
of specific class of drug (adrenergic drugs) by HPLC and
CE has been reviewed by Wang et al. [186]. Applications of
chiral drug separation by EKC are numerous and will be
discussed in the next section.

Recent advances of capillary electrophoresis in pharmaceutical analysis

Applications of capillary electrophoresis


in pharmaceutical analysis
CE and reversed-phase high-performance liquid chromatography (RP-HPLC) are complementary techniques which
are methods of choice for pharmaceutical analysis in the
21st century. CE is rapid, efficient, versatile, and low cost,
whereas HPLC is well established, accurate, sensitive,
reproducible, and robust. Despite some drawbacks (lower
sensitivity and reproducibility compared with HPLC),
numerous applications of CE in drug analysis are reported
daily and reviews on CE for drug analysis are regularly
published. In 2009, Gilpin et al. surveyed separation-based
(including CE), spectrometric-based, and other methods for
analysis of APIs and related drugs in bulk and formulations
between 2007 and 2008 [187]. In 2008, Separation Science
and Technology published a series of reviews on CE
including the role of CE in drug substance and drug product
development [188], the need for CE methods (in a
pharmacopeial monograph) [189], CE in impurity profiling
of drugs [190], the role of CE in biopharmaceutical
development and quality control [191], and CEC of
pharmaceuticals [192]. In 2007, Suntornsuk surveyed
recent applications of CE in pharmaceutical analysis
published during 20052007 [193]. In 2006, Altria et al.
reviewed use of CE for analysis of small-molecule
pharmaceuticals [194] and Morzunova reported CE in
pharmaceutical analysis [195]. In 2005, Ali et al. reviewed
pharmaceutical analysis by CE at nanolevel detection
sensitivity [196]. CE reviews on some classes of APIs have
also been published, for example metal-based drugs [197],
sulfonamides [198], antibiotics [199], pharmaceutical proteins [200], and glycoprotein pharmaceuticals [201].
This review surveys advances of CE in drug analysis
focusing on assay of APIs, determination of drug-related
impurities, chiral separation, and analysis of APIs in
biological samples. These recent examples do not cover
all published articles because of space limitations and are
selected mostly from publications during 2008 and 2009.
Assay of active pharmaceutical ingredients
Although CZEUV/diode-array detection (DAD) and
MEKCUV/DAD are commonly used for assays of APIs
in bulk drug and formulations, several advances of CE
(CECL, CEC4D, FICE and miniaturized CEamperometric detection (minCEAD)) have been increasingly
applied to this application (Table 1). Selected examples of
common CE (CZEUV, CZEindirect UV), and advanced
CE method in assay of APIs are described in detail.
Patel et al. reported the rapid separation and identification of various low-molecular-weight heparins (LMWHs)
and unfractionated heparin by CE using dalteparin sodium

35

as the test LMWHs [219]. LMWHs, which are clinically


used as anti-thrombotic agents, can be derived from native
heparin by enzymatic and chemical reactions. These
reactions result in various LMWHs with different pharmacodynamic and pharmacokinetic properties [231, 232]. CE
separation occurred in 10 min in 30 mmol L1 phosphate
buffer (pH 3.5) using an applied potential of 30 kV and
UV detection at 230 nm. Method linearity (r>0.997, for
2.550 mg mL1 dalteparin sodium), precision (RSD<
4.4%) and accuracy (<5.5%) were good. The method was
applied to the European Pharmacopeia LMWH standard,
dalteparin sodium, enoxaparin sodium, and heparin sodium.
In addition, it could be used for analysis of stressed
LMWHs under elevated temperature and oxidative conditions. Importantly, the method could detect batch-to-batch
variation of LMWHs.
Prutthiwanasan et al. developed an efficient and rapid
CZE method, using indirect UV detection, for assay of
three alkylphosphonate drugs (fosfomycin disodium
(FOS), clodronate disodium (CLO), and alendronate
sodium (ALN)) in pharmaceutical formulations [203].
The multiple probe BGE containing 30 mmol L1 benzoic
acid, 5 mmol L1 salicylic acid and 0.5 mmol L1
cetyltrimethylammonium bromide (CTAB) (pH 3.8), the
temperature (30 C), the applied potential (30 kV), and
detection at 220 were optimized. Addition of CTAB to the
two-probe BGE greatly improved peak symmetry and
EOF reversal, and the negative voltage polarity led to the
co-EOF flow and short analysis time. All analytes were
well resolved (resolution (R)>2.2) in 3.2 min. Good
linearity (r>0.999 in the ranges 201,000 g mL1 for
FOS, 1001,000 g mL 1 for CLO, and 100
750 g mL1 for ALN), repeatability (relative standard
deviation (RSD)<2.15%), recovery (99.3101.1%) and
sensitivity (limit of detection<50 g mL1) were obtained
by use of the proposed method. The method successfully
applied to analysis of FOS, CLO, and ALN in dosage
forms.
Wang et al. used CE with gold nanoparticles (10 nm)enhanced electrochemiluminescence (ECL) detection for
analysis of roxithromycin [131]. Tris(2,2-bipyridyl) ruthenium(II) (Ru(bpy)32+) was used as ECL reagent. CE
separation occurred in 10 mmol L1 phosphate buffer (pH
8.0) using a separating potential of 15 kV and injection at
10 kV for 10 s. The ECL emission was linear in a range of
24 nmol L1 to 0.24 mmol L1. The tris(2,2-bipyridyl)
ruthenium(II) (Ru(bpy)32+)-roxithromycin ECL emission
with gold nanoparticles (detection limit (DL)=8.4 nmol L1)
was ten times higher than that without the nanoparticles (DL
=84 nmol L1). The method could be used to determine
roxithromycin in pharmaceuticals and urine.
J et al. determined glucosamine and K+ in various
dosage forms (granules, capsules, and tablets) by CZEC4D

CZEDAD
CZEDAD

100 mmol L1 borate buffer (pH 9.0)


50 mmol L1 phosphate buffer (pH 3.0)

Formulation
Tablet
Pharmaceuticals, plasma

Alginic acid

Atenolol, celiprolol, clorprenaline, fenoterol, metoprolol,


propranolol, terbutaline and clenbuterol
Captopril and indapamide

Pharmaceutical, nutraceutical
Nutraceutical formulations
pharmaceutical formulations
Tablet, serum
Infusion
Pharmaceutical formulation
Pharmaceutical formulations
Bulk drug, tablet, solution
Sceletium tablet
Standard
Ointment
Capsules

Glucosamine

Insulin and related synthetic analogues

Isoniazid, p-aminosalicylic acid

Linezolid, achiral impurity

Low molecular weight heparin

Low molecular weight, unfractionated heparin

Maprotiline, Desipramine, Moclobemide

Mesembrine and related alkaloids

Nadolol, labetalol

Neomycin

Oseltamivir

Standard

Glucosamine, K+

Tablet
Tablet

Etoricoxib

Ferulic acid, protocatechuic aldehyde, caffeic acid,


protocatechuic acid
FITC-IgC and FITC-insulin

Injection
Tablet

Ethambutol

[214]

CZEUV
CECMS
CZEUV
MEKCUV

40/60/1.6/0.4 or 0.1 mol L1 MeOHACNAcOHTEA


35 mmol L1 phosphate, 15 mmol L1 acetate buffer (pH 4.7)
10 mmol L1 boric acid (pH 10) with 40 mmol L1 SDS

[219]

CZEUV
50 mmol L1 phosphate buffer (pH 1.5)

[218]

CZEUV

[223]

[222]

[91]

[221]

[220]

[217]

MEKCUV

CZEUV

[130]

[216]

MEKC
DAD
CZECL

phosphate buffer (pH 2.35)

10 mmol L1 borate buffer (pH 9.2) with 100 mmol L1 SDS, 15% (v/v)
ACN
50 mmol L1 boric acid buffer (pH 10.0) with 0.5 mmol L1 luminal,
0.01 mmol L1 Cu (II)
125 mmol L1 Tris buffer (pH 2.0) with 150 mmol L1 SDS, 20% (v/v)
MeOH
50 mmol L1 phosphate buffer (pH 2.3), 100 mmol L1 ammonium formate
buffer (pH 3.5)
50 mmol L1 phosphate buffer (pH 3.5)

[163]
[215]

150 mmol L1 boric acid50 mmol L1 phosphate buffer (pH 7.0)

CZEUV

10 mmol L1 phosphate buffer (pH 6.7) with different amount ACN, SDS Microchip
and/or Tween
CECUV
30 mmol L1 acetate buffer (pH 5.2)
CZEC4D

[51]

[213]
CZEAD

20 mmol L1 borate buffer (pH 7.6)

[212]

[211]

[174]

[129]

60 mmol L1 acetic acidsodium acetate buffer (pH 4.6) with 5 mmol L1 CZEUV
CuSO4
CZEDAD
25 mmol L1 Trisphosphate buffer (pH 2.5)

LVSS-FI
CEUV
60 mmol L1 phosphate buffer and 20 mmol L1 boric acid buffer (pH 6.0) CZEUV

55 mmol L1 borate buffer (pH 9.3) with 15% (v/v) ACN

CEECL

50 mmol L1 phosphate buffer (pH 7.5)

pharmaceutical and clinical


sample
Pharmaceutical preparation

Clindamycin

Dextromethorphan, chlorphenamine maleate, pseudoephedrine


hydrochloride, paracetamol
Ertapenem

[209]
[210]

CZEDAD

10 mmol L1 borate buffer (pH 10.0)

Tablet, human urine

Clenbuterol, salbutamol, procaterol, fenoterol

[208]

10 mmol L1 acetate buffer (pH 3.0)methanol (20:80 and 15:85 (v/v) for CZEUV
CN/DOM and CN/NIC, respectively)
25 mmol L1 Trishydrochloride and 15 mmol L1 borate buffer (pH 8.87) CZEDAD

[207]

[206]

[205]

Raw material, tablet, capsule,


liquid formulation
Cinnarizinedomperidone (CN/DOM), cinnarizinenicergoline Combined tablet
(CN/NIC)
Ciprofloxacin, gatifloxacin, moxifloxacin, ofloxacin
Formulation

Chondroitin sulfates

CZEUV

75 mmol L1 Trisphosphate buffer (pH 2.4) with TiO2 nanoparticles

Tablet, capsule, injection

Alendronate, clodronate, fosfomycin

Ref.

Mini-CE
[202]
AD
30 mmol L1 benzoic acid, 5 mmol L1 salicylic acid, 0.5 mmol L1 CTAB CZEindirect [203]
(pH 3.8)
UV
MEKCUV [204]
10 mmol L1 sodium borate buffer (pH 9.0) with 50 mmol L1 SDS

12 mmol L1 sodium borate44 mmol L1 potassium phosphate (pH 7.2)

Tablet, capsule

Method

Acetaminophen and p-aminophenol

BGE

Matrix

Analyte

Table 1 Assay of active pharmaceutical ingredients

36
L. Suntornsuk

Vitamin supplement capsule

Pharmaceutical formulation

Pharmaceutical formulation

Tocopherols and tocopherol acetate

Water-soluble and fat-soluble vitamins

Ziprasidone

ACN, acetonitrile; AcOH, acetic acid; AD, amperometric detection; C4 D, contactless conductivity detection; CEC, capillary electrochromatography; CL, chemiluminescence; CTAB,
cetyltrimethylammonium bromide; CZE, capillary zone electrophoresis; DAD, diode array detector; ECL, electrochemiluminescence; ESI, electrospray ionization; FICE, flow injection capillary
electrophoresis; FITC, fluoresceinyl isothiocyanate; Ig G, immunoglobulin; LVSS, large volume sample stacking; MEEKC, microemulsion electrokinetic chromatography; MEKC, micellar electrokinetic
capillary chromatography; MeOH, methanol; mini-CE-AD, miniaturized capillary electrophoresis-amperometric detection; MS, mass spectrometry; SDS, sodium dodecyl sulfate; TEA, triethanolamine; TiO2,
titanium dioxide; Tris, tris(hydroxymethyl)aminomethane

[230]

[14]

MEEKC
DAD
CZEDAD

[53]

[164]

[229]

MEKC
DAD
CECDAD

10 mmol L1 phosphate buffer (pH 7.4) with 50 mmol L1 SDS, 25% (v/v)
2-propanol
100 mmol L1 Tris buffer (pH 9.3) with 10% (v/v) MeOH and 87% (v/v)
ACN
1.2% (w/w) SDS, 21% (v/v) 1-butanol, 18% (v/v) ACN, 0.8% (w/w) nhexane, 20 mmol L1 borax buffer (pH 8.7)
50 mmol L1 formate buffer (pH 3.0)
Tablet
Tris(8-quinolinolato) gallium(III), 8-quinolinol

CZEC4D

[228]
CZEDAD

Pharmaceutical formulation
Suxamethonium

100 mmol L1 Trisacetate buffer (pH 4.2) with 10% (v/v) ACN

Depot tablet
Salbutamol

100 mmol L1 TEAphosphoric acid (pH 2.6, 3.1) with 10% (v/v) ACN

[227]

[131]
CZEECL
Capsule, human urine
Roxithromycin

50 mmol L1 phosphate buffer (pH 8.0)

Pharmaceutical formulation
Recombinant human granulocyte colony-stimulating factor

50 mmol L1 borate buffer (pH 9.0)

[225]

[226]

MEKC
DAD
CZEDAD
Tablet
Pravastatin, degradation products

65 mmol L1 TEAphosphoric acid (pH 3.0)


Tablet
Piperaquine

25 mmol L1 borate buffer (pH 9.3) with 25 mmol L1 SDS

[224]

MEKC
DAD
CZEDAD
Tablet
Paracetamol, 4-aminophenol

50 mmol L1 phosphate buffer (pH 9.0) with 75 mmol L1 SDS

Matrix
Analyte

Table 1 (continued)

BGE

Method

Ref.

Recent advances of capillary electrophoresis in pharmaceutical analysis

37

[163]. C4D, based on conductivity measurement, was


selected as a detector because glucosamine is nonchromophore and can not be detected by UV detection.
Separation was achieved in <3 min in 30 mmol L1 acetate
buffer (pH 5.2) using an applied potential of 30 kV and
temperature of 25 C. When linearity was assessed, r was
0.997 for 100300 mg mL1 glucosamine and for 15
75 mg mL1 K+. LOD (S/N=3) was 9.3 and 2.9 mg mL1,
respectively. RSD for intermediate precision was 2.35%
and recovery was between 94.6 and 103.3%. The method
was successfully used for quantification of both analytes in
pharmaceuticals and neutraceuticals.
Liu et al. investigated FICE with on-line concentration by head-column field-amplification and largevolume sample stacking (LVSS) for analysis of
dextromethorphan hydrobromide, chlorpheniramine hydrogen maleate (Chl), pseudoephedrine hydrochloride
(Pse), and paracetamol (Par) [174]. The BGE was
55 mmol L1 borate buffer (pH 9.3) containing 15% (v/
v) ACN and injection was performed electrokinetically
between plugs of water. On-line concentration was
performed by injecting a water plug before sample
injection to clean the capillary tip, prevent sample
contamination with buffer, and avoid the samplebuffer
zone boundary disturbance. The flow injection provided
efficient continuous sample introduction resulting in
enhanced sampling frequencies and improved repeatability [233, 234]. Sensitivity enhancement of 30-fold was
observed, providing LOD of <2.84105 mg mL1. Good
repeatability was observed for peak area and height, with
RSD <3.98% for all analytes. This work has advantages in
terms of sensitivity improvement, simultaneous analysis of
APIs with large content differences (Cho and Par), and
improved resolution and peak symmetry.
Chu et al. proposed a novel miniaturized CE method
with carbon-disk electrode amperometric detection (miniCEAD) for analysis of acetaminophen and p-aminophenol in dosage forms [202]. The analytes could be
separated in 150 s in phosphateborate buffer (pH 7.2)
using a capillary 8.5 cm long, constructed on a plexiglass
plate, and a separation voltage of 2,000 V. The endcapillary 300-m carbon disc electrode amperometric
detector provided good S/N at low potential (+600 mV
vs. Ag/AgCl). The detection ranges for both analytes were
between 1.4106 and 5.9107 mol L1 with linearity up
to 1.0103 mol L1 and repeatability (as RSD) <2.3%.
The system was stable over 200 injections and could be
applied to analysis of acetaminophen and its impurity in
dosage forms both for single and combined formulations.
Other drugs (caffeine and chlorpheniramine) did not
interfere the analysis because they give no response with
amperometric detection under the CE conditions investigated.

38

Impurity testing
Impurities are defined as any extraneous materials present
in drug substances. They must be monitored, even if totally
inert or have superior pharmacological properties. They can
be organic or inorganic substances, for example residual
solvents (either volatile or non-volatile), and might originate from starting materials, by-products, intermediates,
degradation products, excipients, reagents, ligands, catalysts, or other materials (filter aids and carbon). Impurities
can affect efficacy and bioavailability, and lead to adverse
effects and toxicity; they must, therefore, be determined by
the manufacturer for regulatory and pharmacopeial reasons.
The International Conference on Harmonization (ICH) has
specified guidelines on impurities in new drug substances
[235] and new drug products [236], and for levels of
residual solvents [237]. Identification and quantification of
impurities are carried out by thin-layer chromatography
(TLC), high-performance TLC, HPLC, GC or CE, whereas
structure elucidation is performed by use of hyphenated
techniques (HPLCMS, CEMS or GCMS).
Three reviews on drug impurity analysis have recently
been published. Mallampati et al. illustrated CE capability and
applications on drug impurity profiling [190]. Bartos et al.
summarized advanced state-of-the-art drug impurity profiling
during 20032008 [238]. Jouyban et al. reviewed analysis of
pharmaceutical impurities by capillary electromigration
during 19802007 focusing on different classes of drugs,
for example chemotherapeutic agents, central nervous
system drugs, histamine receptor drugs, cardiovascular
drugs, anti-cancer drugs, anti-inflammatory drugs, and some
miscellaneous agents [239]. CE has become an essential tool
for drug impurity testing (Table 2). Selected examples on
CEMS, mixed MEKC, uses of sample stacking, and
chemometric techniques for drug impurity testing and
determination of heparin impurities by CE are described.
Hommerson et al. compared the suitability of various
ionization techniques (ESI, APCI, APPI, and thermospray ionization (TSI)) for drug impurity profiling of
lidocaine and proguanil by CEMS [248]. The BGE was
100 mmol L1 acetic acid (pH 4.5). Results showed that
ESI and TSI gave high ionization efficiency for charged
species with an LOD of 100 ng mL1 (in full-scan mode),
whereas APCI and APPI were appropriate for low and
high-polarity compounds. However, APCI and APPI
resulted in more pronounced fragmentation than ESI and
TSI, especially for proguanil and its impurities. Thus,
different ionization techniques provided complementary
information on drug impurity profiles; their ionization
fragmentation and efficiency were also different.
Furlanetto et al. optimized a mixed MEKC method for
analysis of budesonide (BD) and its four related substances
[242]. Experimental designs involving response surface

L. Suntornsuk

methodology were used to optimize the separating voltage


and the concentrations of buffer and surfactants. Baseline
separation of all compounds was obtained in 70 mmol L1
borate buffer (pH 8.8) containing 65 mmol L1 sodium
cholate (CHOL) and 10 mmol L1 3-(N,N-dimethylmyristylammonio)propanesulfonate (MAPS); the applied potential was 16 kV and the temperature 20 C. CHOL and
MAPS were used to form mixed micelles for selectivity
enhancement. Linearity (r>0.998) was observed for BD
and overall RSDs for migration times of impurities were
<8.4%. Recoveries were 99.0101.4% for BD and 98.1
104.8% with LOD <30 g mL1 for the impurities.
Acute hypersensitivity-like reactions in heparin therapy
attracted worldwide attention in 2008. Oversulfated chondroitin sulfate (OSCS) was identified as a toxic contaminant
that causes this adverse effect. Several groups have
proposed methods to separate and quantify OSCS. Somsen
et al. determined OSCS and dermatan sulfate (DS, a
harmless contaminant in heparin that can be used as an
indicator of the quality of heparin purification) in heparin
products by CEUV without sample pretreatment (derivatization, chemical degradation, or enzymatic digestion)
[251]. Types and concentrations of BGE, capillary temperature, sample concentrations and injection volume were
varied. Heparin, OSCS and DS were separated in 17 min in
850 mmol L1 Trisphosphate buffer (pH 3.0) the temperature was 25 C, the sample concentration 50 mg mL1,
injection was at 2.0 psi for 48 s, and the detection
wavelength was 200 nm. A small inner diameter (i.d.)
capillary (25 m) was used to prevent high current and
excessive Joule heating. Preliminary validation showed
linearity (r>0.99 in the ranges 0.510 mg mL1 for OSCS
and DS and 550 mg mL1 for heparin), repeatability (RSD
<4.5%), and detection limits (0.019, 1.1, and 0.26 mg mL1
for OSCS, heparin and DS, respectively) were acceptable.
The method could be applied to determine OSCS in
different batches of heparin down to at least the 0.1%
level. Although peak shapes and/or migration time of
OSCS and heparin from different sources were not
identical, because of the different composition of the
samples, the method was valuable for quality control of
heparin. Wielgos et al. attempted to determine heparin
impurities (OSCS and DS) by CE using high-molarity
phosphate buffers to achieve sample stacking and large
injection of heparin [252]. Detection of OSCS in porcine
intestinal heparin samples was achieved in approximately
20 min in 600 mmol L1 phosphate buffer (pH 3.5) using a
capillary length of 56 cm with an i.d. of 25 m. Resolution
enhancement was performed by counter-EOF (using negative voltage polarity at 30 kV) and injection at the cathode
(50 mbar for 40 s) to make the analytes move against the
EOF. The method was linear
(r=0.9994), precision was
good (RSD<5.5% for all analytes), and LOD was <0.1%

10 mmol L1 borate buffer (pH 8.0)

Standard, commercial product

Tablet

Thiamphenicol, florphenicol,
chloramphenicol, thiamphenicol
impurities
Tiapride hydrochloride, related
impurities

CZE
DAD

CZEUV
MEKC
DAD
CECMS,
NACE
MS
MEKC
UV

MEKC
UV
CEMS
MEKC
UV
CZEUV
CZEUV

CZEUV

CZE
DAD
MEKC
DAD
Mixed
MEKC
UV
MEKC
UV
CZEUV
MEKC
UV

Detector

[255]

[254]

[92]

[252]
[253]

[250]
[251]

[248]
[249]

[247]

[246]

[244]
[245]

[243]

[242]

[241]

[240]

Ref.

ACN, acetonitrile; CEC, capillary electrochromatography; CHOL, sodium cholate; CZE, capillary zone electrophoresis; DAD, diode array detector; EDTA, ethylenediaminetetraacetic acid MAPS,
3-(N,N-dimethylmyristylammonio) propanesulfonate; MEKC, micellar electrokinetic capillary chromatography; MeOH, methanol; MS, mass spectrometry; NACE, non-aqueous capillary
electrophoresis; SDS, sodium dodecyl sulfate; S--CD, sulfated--cyclodextrin; TEA, triethanolamine; Tris, tris(hydroxymethyl)aminomethane; -CD, -cyclodextrin

OTCEC: 10 mmol L1 ammonium formate buffer (pH 3.5) or 10 mmol L1 ammonium acetate
(pH 7.0) NACE: 10 mmol L1 ammonium formate100 mmol L1 formic acid in 50:50
MeOHACN
100 mmol L1 borate buffer (pH 8.5) with 50 mmol L1 SDS

600 mmol L1 phosphate buffer (pH 2.6, 3.5)


15 mmol L1 borate buffer (pH 10) with 25 mmol L1 SDS

Raw material
Tablet
Standard

50 mmol L1 phosphate buffer-150 mmol L1 boric acid (pH 7.0)


850 mmol L1 Tris-phosphate buffer (pH 3.0)

Raw material
Raw material

Six pharmaceutical compounds,


process-related impurities

Oversulfated chondroitin sulfate


Oversulfated chondroitin sulfate,
dermatan sulfate
Oversulfated chondroitin sulfate
Rupatadine, degradation products

100 mmol L1 acetic (pH 4.5)


25 mmol L1 borate buffer (pH 9.5) with 30 mmol L1 SDS, 3% (v/v) ACN

Standard
Tablet

10 mmol L1 TEAphosphate buffer (pH 2.3), 5% (w/v) S--CD


100 mmol L1 borate buffer (pH 10.0) with 20 mmol L1 deoxycholic acid, 15 mmol L1 -CD

200 mmol L1 borate buffer (pH 8.0), 20 mmol L1 SDS, 2 mg mL1 EDTA

10 mmol L1 borate buffer (pH 9.8) with 100 mmol L1 SDS, 15% (v/v) 2-propanol

Suppository, cream

Ciclopiroxolamine, degradation
products
Clopidogrel bisulfate, its impurities
Gentamicin, degradation products

70 mmol L1 borate buffer (pH 8.8) with 65 mmol L1 CHOL, 10 mmol L1 MAPS

Tablet

Controlled-release capsule

Budesonide, related substances

10 mmol L1 borate buffer (pH 9.5) with 50 mmol L1 SDS, 20% (v/v) MeOH

25 mmol L1 citrate buffer (pH 7.0) with 20% (v/v) MeOH

25 mmol L1 phosphate buffer (pH 3.0)

Bulk drug, tablet

Atorvastatin, related substances

Indinavir sulfate, degradation


products
Lamivudine, stavudine, nevirapine,
thymine impurities
Lidocaine, proguanil
Nimesulide, related compounds

Tablet

Amlodipine besilate, impurity D

BGE

Standard, commercial products


Antibiotic carrier (e.g. PMMA
bead, mini-bead, bone
cement)
Bulk drug, capsules

Matrix

Analyte

Table 2 Impurity testing

Recent advances of capillary electrophoresis in pharmaceutical analysis


39

40

for OSCS, approximately 0.5% for DS, and 0.2% for


heparin sodium, calculated as percentage of total heparin.
Results from the CE method revealed a linear relationship
between peak area and bioactivity. A shorter capillary
(21.5 cm) and 600 mmol L1 lithium phosphate buffer (pH
2.5) were also used to provide high speed and highresolution analysis (5 min). Volpi et al. proposed an
alternative method for determination of OSCS in heparin
preparations by acid hydrolysis of heparin and OSCS
before CE analysis [250]. Heparin and OSCS are hydrolyzed to form glucosamine (2-amino-2-deoxy-D-glucose)
(GlcN) and galactosamine (2-amino-2-deoxy-D-galactose)
(GalN), respectively; they were then derivatized with
anthranilic acid (AA). Separation was carried out in a
capillary with an effective length of 65 cm (50 m i.d.) at
25 C, and 15 kV; 150 mmol L1 boric acid and
50 mmol L1 sodium dihydrogen phosphate buffer (pH
7.0) was used as the BGE. CE separation of GalN and GlcN
derivatized with AA was achieved in 10 min. Strong
absorbance at 214 nm enabled detection of OSCS at levels
as low as 1%. Response was a linear function of
concentration in the range 40400 g mL1 (r>0.980),
RSDs for migration time and peak area were <5.8%,
recovery was 92%, and LOD and LOQ were 200 pg and
500 pg, respectively. The method was robust within 10%
variation of experimental conditions (temperature, potential, and BGE composition). Application to real samples
revealed the presence of 38.9% OSCS (as GalN%) in
contaminated heparin raw material and 39.7% (as GalN%)
in the formulated contaminated heparin.
Although, OSCS is a toxic contaminant associated with
heparin therapy, chondroitin sulfate (CS) itself is used as a
dietary supplement for treatment of osteoarthritis. Its
efficacy can vary, however, because of to the quality of
the CS preparations. Malavaki et al. proposed a CZEDAD
method for quantification of CS, screening for other
glycosaminoglycan or DNA impurities, and determination
of hyaluronan impurities (hyaluronic acid, HA) in CS raw
materials, tablets, hard capsules, and liquid formulations
[207]. CS and HA separation was achieved in 12 min in
50 mmol L1 phosphate buffer (pH 3.0) using an applied
potential of 30 kV and a detection wavelength of 200 nm.
Excipients in CS formulations (gelatin, magnesium stearate,
titanium dioxide, ferric oxide, quinoline sulfate, sorbitol,
saccharin, and dyes) did not interfere with the analysis. The
linearity of the method was good (r>0.99 in the ranges 30
10,000 g mL1 for CS and 5500 g mL1 for HA), RSDs
were <6.6%, and relative error was 11%. LODs were 2 and
17 g mL1 for CS and HA, respectively.
Fayed et al. used CZE in an uncoated fused-silica
capillary for determination of clopidogrel bisulfate, a potent
anti-platelet and anti-thrombotic drug, and its impurities A,
B, and C [244]. Impurity A is a hydrolysis product,

L. Suntornsuk

impurity B is a racemic mixture of isomers, and impurity


C is the R enantiomer of clopidogrel. The CE conditions
were optimized and robustness testing was performed by
using a reduced central composite (RCC) experimental
design focusing on the effects of buffer composition and
pH, concentrations of the chiral selector, and the applied
potential. The predicted conditions were slightly modified
to avoid high current and baseline noise, and the final
conditions were 10 mmol L1 triethylaminephosphoric
acid buffer (pH 2.3) containing 5% (mass/volume, m/v)
sulfated -CD at 20 C and an applied potential of 12 kV.
Method robustness was tested using RCC by varying the
four factors by 1 to 2 units of the optimum values and
results revealed no significant changes on migration time
and resolution. The method was linear (r>0.996) in the
ranges 0.4300 g mL 1 for clopidogrel, 1.00
2.50 g mL1 for impurity A, 0.502.00 g mL1 for
impurity B, and 0.252.00 g mL1 for impurity C. LODs
and LOQs were less than 0.33 and 1.0 g mL1, respectively, for all analytes. Repeatability for clopidogrel, as
RSD, was <1.6% and recovery was 99.45%. The method
was successfully applied to analysis of clopidogrel and its
impurities in the bulk drug.
Michalska et al. investigated different sample-stacking
strategies (normal stacking mode (NSM) and stacking with
reverse migrating micelles (SRMM)) for MEKC analysis of
ertapenem (a Group 1 carbapenem antibiotic) and its
impurities in pharmaceutical formulations [211]. In NSM
mode, the best separation was obtained with 60 mmol L1
sodium dihydrogen phosphate and 20 mmol L1 boric acid
buffer (pH 6.0) containing 80 mmol L1 SDS, using an
uncoated fused-silica capillary and a potential of +18 kV.
All analytes migrated after the EOF within 11 min. For
SRMM, the separation was achieved in 10 min in
40 mmol L 1 sodium dihydrogen phosphate and
20 mmol L1 boric acid buffer (pH 6.0) containing
50 mmol L1 SDS using a neutral-coated capillary with a
reversed voltage polarity of 12 kV. Linearity (r>0.999, in
the range 0.00080.54 mg mL1 for ertapenem), RSD (<8%
for all analytes, calculated from corrected peak area), and
LOD (0.3 g mL1 for ertapenem) were comparable for
both NSM and SRMM. Recovery of ertapenem was not
statistically different (P=0.05) for the CE and HPLC
methods. The stacking enhancement factors were 183 to
4.75-fold for NSM and 1,289 to 4.29-fold for SRMM. The
method could be used for assay and stability studies of
ertapenem under various stress conditions (acid, base, and
heat).
Chiral drug separation
EKC is now a valuable technique for chiral drug
separation, and research in this area has increased

Recent advances of capillary electrophoresis in pharmaceutical analysis

exponentially, as reported in many reviews [177, 180


185]. EKC (mainly CZE mode) with neutral and/or
charged CD derivatives, are predominantly used for
separation of the enantiomers of chiral drugs (Table 3).
Other variations, for example CITP, MEEKC, NACE, CE
MS, CEC, and multiplexed CE have also been used to
resolve enantiomeric drugs. In addition to CD derivatives,
a few other chiral selectors have been used, including
eremomycin, -CD derivatized erythromycin, 1S,4R-(+)ketopinic acid ((+)-KPA) and 2R,3S,4R,5S-()-2,3:4,6-diO-isopropylidene-2-keto-l-gulonic acid (()-DIKGA),
dextran sulfate (DxS), and polygalacturonic acid. Examples of the use of different CDs, dual CD systems, ITPCZE, the sample stacking technique, and experimental
design for chiral drug separation are given below.
Suntornsuk et al. proposed rapid simultaneous separation
of baclofen enantiomers and impurity A by EKC in
100 mmol L1 sodium borate buffer (pH 9.9) containing
18 mmol L1 -CD and 1% (v/v) ACN; the temperature
was 45 C and the applied potential 27 kV [256].
Separation efficiency for the impurity was significantly
improved when electro-osmotic flow modification was
achieved by dynamic coating of the capillary with poly
(ethylene oxide), because impurity A was neutral under the
separating condition. Resolution of 2.7 (calculated for the
baclofen enantiomers) was achieved in 9.0 min with LODs
of 2 g mL1 for impurity A and 10 g mL1 for the
baclofen enantiomers. The linearity (r>0.999 in the range
550 g mL1 for impurity A and 50500 g mL1 for the
baclofen enantiomers), precision (RSD<3.37%), and recovery (100.3% for R-()-baclofen, 101.6% for S-(+)baclofen, and 96.1% for impurity A) of the method were
good. This was the first time an EKC method had been
developed for simultaneous separation of the baclofen
enantiomers and its impurity in bulk drug and tablets.
Desiderio et al. developed an EKC method for separation
of the enantiomers of baclofen using sulfobutylether--CD
(SBE--CD) as chiral selector and 0.25 mol L1 formic acid
containing 70% (v/v) methanol as the BGE [80]. Tandem MS
and on-line UV detection were used to detect the enantiomers. In this work the capillary was filled with the
(negatively charged) chiral selector during pre-run conditioning. When a positive voltage was applied, SBE--CD
migrated away from the MS detector at the capillary outlet,
in the opposite direction to baclofen generating a counter
current mode. This technique reduced analyte signal suppression during ionization, prevented ion source contamination, and required small amounts of chiral selectors. Tandem
MS (MS2 and MS3) improved method sensitivity (LOD=
0.1 g mL1 compared with 0.4 g mL1 in UV detection)
and selectivity. Linearity was good with r>0.99 for both
enantiomers with RSD<4.34% for method precision and
RSD<6.25% for method accuracy.

41

The chirality of -agonists and antagonists greatly


affects their bioactivity and side effects. Huang et al.
proposed FASI (field amplified sample injection)-CE for
simultaneous separation of the enantiomers of six blockers (carteolol, atenolol, sotalol, metoprolol, esmolol,
and propranolol) in human serum [259]. The optimized
BGE was 20 mmol L1 phosphate buffer (pH 5.5)
containing 1.5% (w/v) carboxymethyl--CD (CM--CD),
5% (v/v) methanol and 5% (v/v) ethanol. CM--CD was
chosen because it is negatively charged and has broad
enantioselectivity for the -blockers, which are basic
compounds. Resolution of the enantiomers was greatly
improved by formation of an inclusion complex, hydrogen
bonding, and pronounced electrostatic interaction of the
analytes with the chiral selector. A sensitivity enhancement
of 5 to 25-fold was achieved by FASI (injection at 10 kV
for 8 s), resulting in LODs between 0.01 and 0.5 g mL1.
Method linearity was good with r>0.994 (in the ranges
0.055 g mL1 for cartelol, 0.2525 g mL1 for atenolol,
sotalol, metoprolol and esmolol, and 2.525 g mL1 for
propranolol); RSDs of migration time and peak height were
<6.1% and recovery was between 82.1 and 98.2%.
Separation of the enantiomers of -blockers by CE using
CDs as chiral selectors has been performed by many
research groups [6, 257259, 268, 272, 279], although
with different optimized CE conditions. This is because of
the chemical structures of the -blockers investigated, the
experimental conditions, and the structures of the chiral
selectors. Gagyi et al. comparatively investigated the effects
of chemical structure on stereoselective recognition of
fourteen -blockers by using a variety of CDs, for example
hydroxypropyl--cyclodextrin (HP--CD), random methyl
-cyclodextrin (RAME), sulfated -cyclodextrin (S-CD), and sulfated -cyclodextrin (S--CD) [279]. Results
indicated that chemical structures (side chain substitution
and condensed rings) substantially affected stereoselective
recognition. For example, most -blockers were recognized
by HP--CD and RAME. Alprenolol and oxopenolol
enantiomers were resolved by S--CD, whereas pindolol
enantiomers were resolved by S--CD. Additionally, there
was no correlation between CD concentration or pH and
resolution of -blocker enantiomers. Generally, S enantiomers of -blockers had weaker interactions with CDs
and migrated before the R enantiomers. However, the S
enantiomer of carvediol migrated after the R enantiomer.
This phenomenon also was because of chemical structure
differences. Stereoselective recognition of -blockers containing more than one chiral center was even more
complicated and unpredictable, and requires intensive
future investigation.
Deeb et al. have demonstrated strategies in method
development for quantification of enantiomeric impurities
by use of CE [272]. First, initial CE conditions were

Drug
substance
Oral
suspension
Raw material

Dexamphetamine, 1R,2S-()-norephedrine, 1S,2S-(+)norpseudoephedrines, levoamphetamine


Linezolid

Aminoglutethimide

Cetirizine, levocetirizine

Amisulpride, impurities

Baclofen

Ibuprofen and metabolites

Alprenolol, pindolol, propanolol, isoxsuprine, ritodrine

Primaquine

Hexaconazole, penconazole, myclobutanil

Montelukast sodium, isomeric impurities

M--CD

S--CD

S--CD

SBE--CD

TM--CD

-CD

HP--CD

HP--CD

HP--CD

Ecstasy, methadone

Methadone and 2-ethylidene-1,5-dimethyl-3,3-diphenylpyrrolidine

Benzoxazolinonic aminoalcohol

HS--CD

HS--CD

S--CD

Atenolol, verapamil, isoprenaline, mandelic acid

R-flurbiprofen

IPA--CD

Propiconazole

Omeprazole

HS--CD

HS--CD

Amlodipine

HP--CD

HP--CD

Timolol

HDMS--CD

HDAS--CD

Standard

Brompheniramine, pheniramine, metoxyphenamine, doxylamine,


cyclopentolate, ketamine

-CD, S--CD, TM-CD, DM--CD, HP-CD


HDAS--CD

Oral fluid
specimen
Raw material

Raw material,
fresh grape
Plasma

Standard

Standard

Raw material

Tablet

Raw material

Plasma, urine

Tablet

Standard, raw
material
Pharmaceutical
formulation
Tablet, human
plasma
Tablet

Capsule

Urine

Human serum

Tablet

Tablet, human
urine

Betaxolol, sotalol, metoprolol, bisoprolol, bevantolol, timolol

Brompheniramine maleate, dexbrompheniramine maleate, pheniramine


maleate

CE--CDs

Raw material

Tablet

Carteolol, atenolol, sotalol, metoprolol, esmolol, propranolol

Salbutamol, cimaterol, clenbuterol, terbutaline, formoterol

-CD

CM--CD

Baclofen, impurity A

-CD

Matrix

CM--CD

Analyte

Chiral selector

Table 3 Chiral separation

CZEDAD

CZEDAD

18 mmol L1 Tris (pH 2.5, 7, and 10)

100 mmol L1 phosphate buffer (pH 2.5)

CZEDAD

50 mmol L1 phosphate buffer (pH 2.5)

[276]

[274]
[275]

CZEDAD

50 mmol L1 phosphate buffer (pH 4.5) with 0.2% (w/v)

[273]

[272]

[271]

[270]

[269]

[268]

[267]

[80]

[266]

[265]

[264]

[263]

[81]

[28]

[6]

[262]

[261]

[260]

25 mmol L1 phosphate buffer (pH 7.0) with 50 mmol L1 SDS, MEKCDAD


MeOHACN (10:5%, v/v)
15 mmol L1 ammonium formate buffer (pH 2.5) with HS--CD CEESI-MS

25 mmol L1 phosphate buffer (pH 3.0) with 50 mmol L1 SDS CD modified-MEKC


DAD
20 mmol L1 borate buffer (pH 9.2) with, 10 mmol L1 SDS,
CD-MEKCDAD
10 mmol L1
50 mmol L1 phosphate buffer (pH 2.5)
CZEUV

CZEUV

CZEDAD

Chiral CEUV

20 mmol L1 TEAphosphate buffer (pH 5.0)


75 mmol L1 TEA phosphate buffer (pH 2.5) with different
alcohol
Tris phosphate buffer (pH 2.5)

CZEDAD
CZEMS

250 mmol L1 formic acid with 70% (v/v) MeOH

CZEUV

5 mmol L1 borate buffer (pH 8.7)


10 mmol L1 citrate buffer (pH 3.5)

CZEUV

NACEDAD

20 mmol L1 ammonium camphorsulfonate, 40 mmol L1


ammonium acetate in MeOH
50 mmol L1 Tris phosphate buffer (pH 3.0)

30 mmol L1 formic acid with potassium camphor sulfonate in NACEDAD


MeOH
ITP: potassium acetate (pH 4.75)glycine acetate (pH 3.2) CZE: ITP-CZEUV
glycine acetate (pH 3.2)
10 mmol L1 ammonium acetate buffer (pH 5.8)
CZEDAD, MS

50 mmol L

CZEUV

FASI-CZEUV

borate buffer (pH 9.0)

[258]

CZEDAD

20 mmol L1 phosphate buffer (pH 5.5)

[31]

ITP-CZEDAD

[259]

[257]

[256]

CZE UV

CZEUV

100 mmol L1 borate buffer (pH 9.9) with 1% (v/v) ACN

Ref.

50 mmol L1 phosphate buffer (pH 2.5) with


tetraalkylammonium-base ionic liquid
CE: 25 mmol L1 acetate buffer, (pH 3.1). ITP: 20 mmol L1
acetate buffer (pH 4.75) and 10 mmol L1 acetate buffer (pH
3.5)
50 mmol L1 acetate buffer (pH 4.75)

Method

BGE

42
L. Suntornsuk

CZEDAD
CZEUV
CZEUV
CZEUV

20 mmol L1 phosphate buffer (pH 2.5)


25 mmol L1 phosphate buffer (pH 8.0)
50 mmol L1 phosphate buffer (pH 6.5)
20 mmol L1 phosphate buffer (pH 3.01, 4.98)

Tablet
Raw material
Raw material

Escitalopram oxalate, citalopram hydrobromide, racemic citadiol

Pioglitazone, rosiglitazone

Flurbiprofen, fenoprofen, ibuprofen, indoprofen, ketoprofen,

[42]

()-DIKGA, 2R,3S,4R,5S-()-2,3:4,6-di-O-isopropylidene-2-keto-l-gulonic acid; (+)-KPA, 1S,4R-(+)-ketopinic acid; ACN, acetonitrile; AcOH, acetic acid; CEC, capillary electrochromatography;
CE--CD, carboxyethyl--cyclodextrin; CM--CD, carboxymethyl--cyclodextrin; CZE, capillary zone electrophoresis; DAD, diode array detector; DM--CD, heptakis(2,6-di-O-methyl)-cyclodextrin; DxS, dextran sulfate; EtOH, ethanol; FASI, field amplified sample injection; HDAS--CD, heptakis-(2,3-diacetyl-6-sulfato)--cyclodextrin; HDMS- CD, heptakis(2,3-di-Omethyl-6-O-sulfo)-cyclodextrin; HP--CD, hydroxypropyl--cyclodextrin; HP--CD, 2-hydroxypropyl--cyclodextrin; HS-CDs, high resolution power of sulfated cyclodextrins; HS--CD,
heptakis-(6-sulfo)--cyclodextrin; HS--CD, highly sulfated --cyclodextrin; IPA--CD, 6-monodeoxy-6-mono(2-hydroxy)propylamino--cyclodextrin; MEKC, micellar electrokinetic capillary
chromatography; MeOH, methanol; MIP, molecular imprinted polymer; MS, mass spectrometry; M--CD, methylated--cyclodextrin; NACE, non-aqueous capillary electrophoresis; RAMEB,
randomly methylated -cyclodextrin; SB--CD, sulfobutylether--cyclodextrin; SDS, sodium dodecyl sulfate; SI-S--CD, single isomer sulfate-substituted -cyclodextrin; S--CD, sulfated-cyclodextrin; S--CD, sulfated--cyclodextrin; S--CD, sulfated --cyclodextrin; TEA, triethanolamine; TM--CD, heptakis (2,3,6-tri-O-methyl) -cyclodextrin; -CD, -cyclodextrin; -CD,
-Cyclodextrin; -CD, -cyclodextrin

Standard

Nadolol, labetalol

MIPCECUV

[289]

[288]

[287]

[286]

[285]

[284]

[283]

[282]

[281]

[280]

[279]

[278]

[277]

Ref.

60 mmol L1 sodium acetate buffer (pH pH 7.0) with 85% (v/v) MIPCECUV
[45]
ACN
MeOHACN (40:60) with 0.8% (v/v) HOAc and 0.2% (v/v) TEA CECUV, vancomycin [91]
chiral stationary
phase

10 mmol L1 phosphate buffer (pH 7.0) with 20% (v/v) ACN

Standard
Standard

Ofloxacin

CZEUV

40 mmol L1 phosphate buffer (pH 4.0)

Drug
substance
Standard

CZEUV

40 mmol L1 KOH in MeOHEtOH (3:2 for (+)-KPA, 2:3 for


()-DIKGA)
10 mmol L1 phosphate buffer (pH 2.5)

NACEDAD

CZEUV

100 mmol L1 phosphate buffer (pH 2.5)

Raw material

Rotigotine, chiral impurities

Raw material

CZEDAD

80 mmol L1 phosphate buffer (pH 2.5)

Standard

9-Hydroxyrisperidone

S-timolol, and 1R,2S-ephedrine

CZEDAD

100 mmol L1 citrate buffer (pH 3.0)

Raw material

Five phenothiazines

Raw material

CZEUV

50 mmol L1 phosphate buffer (pH 2.5)

Chlorpheniramine, salsolinal

Multiplexed CZEUV

25 mmol L1 TEA phosphate (pH 2.5)

DxS (Mr: 1,000,000 or


Rotigotine, trihexyphenidyl
500,000)
Polygalacturonic acid (Mr: Nefopam hydrochloride, oxazepam, chlorpheniramine, chlorcyclizine,
2,000 to 100,000)
sulconazole, ketoconazole

Propanolol

-CD derivatized
erythromycin
(+)-KPA, ()-DIKGA

CZEDAD

25 mmol L1 phosphate buffer (pH 2.5)

Atropine sulfate, chloroquin maleate, citalopram HBr, metoprolol tartrate, Raw material
propanolol HCl, tramadol HCl, verapamil HCl, baclofen and ofloxacin
Mianserin, ketoprofen, alprenolol, aminoglutethimide, mepivacaine,
Raw material
fluoxetine, trihehyphenidyl, Trogers base
Twelve -blockers
Standard

HS--CD, HS--CD, HS-CD


HS--CD, HS--CD, HS-CD
S--CD, S--CD, HP-CD, RAMEB
Single and dual CD system
(SI-S--CD, MI-S-CD)
Dual CD system (HP-CD, S--CD)
Dual CD system (S--CD,
M--CD)
Dual CD system (-CD
and S--CD)
Dual CD system (DM-CD, SB--CD)
Eremomycin

Method

Matrix

BGE

Analyte

Chiral selector

Table 3 (continued)

Recent advances of capillary electrophoresis in pharmaceutical analysis


43

44

screened for the appropriate chiral selectors (from 22


neutral, anionic, or cationic CDs) and, second, the
optimum conditions (concentrations of chiral selectors
(010%), buffer pH (29) and amounts of organic solvents
(015% v/v)) were determined on the basis of resolution
and migration time. Fourteen chiral drugs were included in
the study and complete optimization was investigated for
atenolol, isoprenaline, verapamil, and mandelic acid. For
basic drugs (atenolol, isoprenaline, and verapamil), enantiomer separations (RS =3.3) were achieved in 50 mmol L1
phosphate buffer (pH 2.5) containing 2% HS--CD and 5%
methanol. For mandelic acid, 50 mmol L1 phosphate buffer
(pH 6.0) containing 1% 6-monodeoxy-6-monoamino--CD
and 5% methanol enabled the best separation (RS =1.6).
LODs were between 0.010 and 0.50 g mL1 and LOQs
(related to maximum concentrations, Cmax) were between
0.05 and 0.1%, where Cmax was 2.00 mg mL1 for atenolol
and 1.00 mg mL1 for isoprenaline, verapamil, and
mandelic acid. In impurity quantitation, relative LOQ or
percentage LOQ is more important than absolute LOQ
because the individual enantiomers are not available in their
completely pure forms. The authors also emphasized that
racemic mixtures should be used as test analytes in
optimization steps until a resolution of 3 is reached, because
they provide good S to R enantiomer ratios, enable peak
identification, and are less expensive and better quality than
the pure form of the enantiomers.
Although use of single CD derivatives for chiral drug
separation is common, this might be insufficient in
some cases and a dual CD system should be considered.
Chu et al. achieved simultaneous separation of the
enantiomers of rotigotine and related chiral impurities
(intermediates I and II) in 100 mmol L1 phosphate
buffer (pH 2.5) containing 2% (w/v) S--CD and 2% (w/v)
M--CD [282]. A single-CD system could not provide
satisfactory separation of the enantiomers of all analytes.
For instance, -CD gave tailing peaks for rotigotine and
its R form overlapped the enantiomers of intermediates I
and II, and M--CD resulted in partial enantioseparation
of intermediates I and II and overlapping with the R form
of rotigotine. S--CD enabled separation of the enantiomers of rotigotine, and separation of its R form from
intermediate II but the enantiomers of intermediate I
overlapped. The dual CD system containing neutral (M-CD) and charged (S--CD) CDs significantly improved
selectivity and enantiomer resolution (RS =1.3 to 7.3) of
the analytes because of inclusion complexation and
electrostatic interaction. Response was a linear function
of concentration in the range 0.005 to 0.25 mmol L1 (r
from 0.9978 to 0.9995). RSDs were <0.58% for migration
time and <3.78% for peak-area ratio. Recovery ranged
from 95.9% to 108.3% and LOD and LOQ for each
enantiomer were 0.003 and 0.01 mmol L1, respectively.

L. Suntornsuk

The method was applicable for chiral purity testing of


rotigotine raw material.
Sungthong et al. achieved separation of the enantiomers
of escitalopram and its related substances (R-citalopram, Rand S-citadiol) by CE [283]. Preliminary results indicated
separation was achieved in phosphate buffer (pH 2.5)
containing S--CD, and peak shapes were improved when
-CD was added into the BGE. In the final optimization,
central composite face-centered factorial design was used to
evaluate effects of S--CD and phosphate buffer concentrations, separation temperature, and applied potential.
Twenty-seven experiments were randomly run and data
were fitted by partial least-squares analysis. The optimum
CE conditions were 20 mmol L1 sodium phosphate buffer
(pH 2.5) containing dual CDs (22 mg mL1 S--CD and
0.5 mg mL1 -CD) at a capillary temperature of 28 C and
an applied potential of 20 kV. The predicted migration
time (8.7 vs. 8.4 min) and current (73 vs. 68 A) for the
optimized method were in good agreement with the true
values. The dual-CD system consisting of charged and
neutral CDs resulted in an efficient method for determination of related substances and analysis of the chiral purity
of escitalopram in a single run. Response was a linear
function of concentration in the ranges 2.5150 g mL1
and 2.550 g mL1 for R-citalopram and the citadiol
enantiomers, respectively (r>0.996). Intra-day and interday precision (as RSD) was <0.84% for migration time and
<9.28% for peak area ratio. Recovery of at least 88% was
estimated and the LODs and LOQs for all the analytes were
0.02% and 0.05%, respectively, relative to a concentration
of 5 mg mL1 escitalopram.
Miku et al. proposed the use of column-coupling
electrophoresis with a fiber-based DAD for enantioselective
analysis of traces of drugs (pheniramine and its analogs) in
pharmaceutical and clinical samples [31]. ITPCZE was
chosen to provide short analysis time, minimum generated
heat, reduced peak dispersion, and sufficient resolution with
good compatibility of the online coupled separation
systems. A charged CD (CE--CD) was selected as a
chiral selector for separation of the enantiomers of the
charged analytes because it was more efficient than neutral
CDs (-CD or hydroxyethyl HE--CD). The leading
electrolyte (pH 4.5) contained 10 mmol L1 sodium as
leading cation, 20 mmol L1 acetate as counter ion, and
hydroxyethylcellulose as EOF suppressor. The terminating
electrolyte contained 5 mmol L1 glycine or aminocaproic
acid as terminating cation and 10 mmol L1 acetate (pH 3.5
or 4.5) as counter ion. Good linearity (r>0.99 in the ranges
23.16231.6 g L1 for BP and 17.4174 g L1 for
PHM), precision (RSD<1.65%), recovery (>97.8% with
relative error <2.2%), and LOD and LOQ (6.8 and
12.7 g L1, respectively) were achieved by use of this
ITPCZE method.

Recent advances of capillary electrophoresis in pharmaceutical analysis

Determination of active pharmaceutical ingredients


in biological fluid
CE has been important in the analysis of APIs in biofluids
[72, 290292]. Recent examples of CE analysis of APIs in
biological fluid are listed in Table 4. Many techniques have
been developed for detection of drugs at trace levels, for
example CE (either CZE or MEKC)LIF, CELED induced
fluorescence, IACE, CEICP-optical emission spectrometry
(OES), MEEKC, NACE, CECL, CEMS, and CE-C4D.
Some of these techniques will be discussed in detail.
Coenzyme Q10 (CoQ10) is a lipid-soluble strong
antioxidant that protects circulating lipoproteins and cell
membranes from oxidative stress. Analysis of CoQ10 can
be cumbersome, because it is insoluble in water. Lucangioli
et al. established an ECK method using a novel microemulsion for analysis of CoQ10 in human plasma [11].
Separation was achieved in an uncoated silica capillary of
60 cm total length and 75 mm i.d. using an applied
potential of 20 kV, at room temperature, and detection was
at 214 nm. The microemulsion consisted of 1.4% (w/w)
sodium bis(2-ethylhexyl)sulfosuccinate (AOT), 4% (w/w)
cholic acid, 1% (w/w) octane, 8.5% (w/w) butanol, 0.1% (w/
w) et al. , and 85% (w/w) 10 mmol L1 Tris buffer at pH
9.0. Because of the extreme hydrophobicity of CoQ10 (log
P>10), separation could not be achieved by use of SDS
alone. AOT is a double-chain anionic hydrophobic surfactant that is valuable for highly hydrophobic compounds
such as tocopherol, CoQ10, and fat-soluble vitamins. Both
AOT and cholic acid as tensioactives were used to provide
better selectivity and efficiency. The selectivity of the
method was good, and there were no interferences from
endogenous substances (estradiol, cortisol, testosterone,
etc.). Linearity (r=0.9945 in the range 0.24 g mL1)
and accuracy (recovery=96.399.0%) were also good.
LODs and LOQs were 1 and 3 g mL1, respectively,
and intra-day and inter-day precision, as RSD, was <5.6%.
Results from plasma samples were in good agreement with
those from HPLC, with r between the two methods of
0.9938.
Schappler et al. determined propranolol hydrochloride
levels in plasma, without derivatization, by use of CELIF
with a diode solid-state laser at 266 nm [114]. Box
Behnken design was used to optimize the pH and ionic
strength of the BGE, temperature, and separation potential
in order to improve CE sensitivity (S/N) and efficiency (N).
The optimum S/N and N values estimated for use of
25 mmol L1 Trisphosphate (pH 2.8) at 30 C and 30 kV
were in good agreement with the experimental values.
Ordinary least-squares linear regression (OLS) after squareroot transformation was the most appropriate regression
model, with r=0.9983 (50850 ng mL1). The relative bias
was <7.9% and precision (as RSD) was <12.5%. The upper

45

and lower confidence limits of the mean bias were less than
30% and the lower limit of quantitation (LLOQ) was
50 ng mL1. The method was used to determine propranolol in a forensic case (post-mortem sample) and the
amount found was 156 ng mL1.
Belin et al. quantified valproic acid (VPA) in serum,
plasma, and urine using CEC4D [160]. The VPA molecule
does not contain a strong chromophore, so C4D was
selected because it is more sensitive than indirect UV
detection. The BGE system was predicted by use of Peak
Master 5.1, and 10 mmol L1 2-(N-morpholino)ethanesulfonic acid monohydrate/DL-histidine (MES/His) (pH 6.0)
was found to be optimum. High-speed analysis (within
3 min) was achieved by EOF reversal (dynamically coated
capillary with 50 mol L1 hexadecyltrimethyl ammonium
bromide (HTAB)) and a negative applied potential of
30 kV. A calibration curve determined from normalized
peak areas (ratios of migration time-corrected peak area of
VPA to caproic acid (internal standard)) was linear in the
range 1250 g mL1 (r=0.9993). Repeatability and
intermediate precision (as RSD) were <0.38% for migration
time and <3.5% for peak area. The LOD and LOQ were 24
and 80 ng mL1, respectively. Common drugs that may be
concurrently used by patients (acetaminophen, carbamazepine, digoxin, etc.) did not interfere with the analysis. The
method was validated by use of drug calibration standards
and results were comparable with manufactures values.

Future trends
Conventional and advanced CE techniques complement
one another in solving pharmaceutical analysis problems.
Nevertheless, further developments are desirable to
enhance method sensitivity, selectivity, and throughput.
Derivatization, sample stacking techniques, micro-solid
phase extraction, specific detectors (LIF, MS), and
extended light path capillaries have been developed to
improve CE sensitivity. Among these techniques, sample
stacking techniques (sample stacking, sweeping techniques, transient isotachophoresis, and dynamic pH junction) are simple, inexpensive, and do not require extra
instruments [20, 305, 306]. Sample stacking techniques
are valuable for determination of drugs at trace levels in
biological fluids [307, 308].
Microchip (miniaturized CE) and multiplexed CE
(arrays of capillaries in parallel formats) have been
developed to enable high-speed and/or high-throughput
analysis. Research in both areas is currently very active
but there is still room for improvements. Among microfluidic devices, microchip-CE is popular because of its
high speed, high throughput, low sample consumption,
and easy operation [309]. Integration of all analytical

CZEDAD
CZEDAD
CE-ICP-OES
CZEDAD

50 mmol L1 Tris-phosphate buffer (pH 2.3)


50 mmol L1 phosphate buffer (pH 2.5)
30 mmol L1 Tris-HCl buffer (pH 7.4)
100 mmol L1 borate (pH 9.0)

Plasma
Plasma
Plasma

Plasma
Cerebrospinal fluid
Plasma
Serum
Serum
Plasma
Plasma
Plasma, tablet
Plasma

Octopamine

Penicillamine

Propranolol

Plasma

Plasma
Plasma, tablet

Plasma
Urine
Plasma
Plasma

Plasma, pharmaceutical
formulation
Plasma

25 mmol L1 TRIS-phosphate buffer (pH 2.8)

10 mmol L1 borate buffer (pH 9.1)

25 mmol L1 Tris buffer (pH 4.0)


5 mmol L1 ammonium acetate (pH 3.6)
30 mmol L1 sodium-borate buffer (pH 10.0) with 10 mmol L1 SDS
Anti-neurotrophin FAb capillary, 0.1 mol L1 phosphate with 0.01% Brij 35 (pH 7.0)
50 mmol L1 ammonium acetate with 1 mol L1 acetic acid and 10 mmol L1 SDS
50 mmol L1 sodium phosphate buffer (pH 3.0) with 0.5 mmol L1 -CD (PDMA coated
capillary)
25 mmol L1 sodium borate buffer (pH 9.2)

20 mmol L1 borax (pH 10.5)


30 mmol L1 borate buffer (pH 9.4) with 0.01 mmol L1 luminal, 0.05 mmol L1 K3[Fe
(CN)6] in 0.6 mol L1 NaOH
2 mol L1 formic acid, 70 mmol L1 ammonium acetate in ACNMeOH (60:40)

10 mmol L1 Tris buffer (pH 9.0) with AOT, CA, octane, 1-butanol, PVA
(85:1.4:4:1:8.5:0.1)
60 mmol L1 Tris buffer (pH 4.0) with 40 mmol L1 SOS, 0.01% PVA
20 mmol L1 ammonium acetate (pH 4.0) in 50:50 ACNMeOH
80 mmol L1 phosphate, 60 mmol L1 TEA (pH 2.0) with 5% MeOH
1.25 mol L1 phosphoric acid in ACN

pCECMS
AEKC-DAD

20 mmol L1 ammonium acetate (pH 6.0) with 80% ACN


50 mmol L1 Tris solution with HSA

Urine
Plasma

[110]

CE-LED induced
fluorescence
CE-LED-induced
fluorescence
CE-LIF

[114]

[112]

[301]
[302]
[303]
[62]
[7]
[304]

[5]

NACEMS
CE-UV
CEMS
CE-UV
IACE
NACE-UV
MEKC-UV

[299]
[300]

[297]
[8]
[298]
[4]

FASS-CE-UV
NACEMS
CZEUV
NACE-DAD
CE-UV
CECL

[11]

MEEKC-DAD

[206]

[294]
[295]
[296]

[93]
[61]

[125]
[126]

MEKC-LIF
CZE-LIF

Urine
Plasma

Plasma, saliva, urine

Lidocaine, mepivacaine HCl and


bupivacaine HCl
Metformin
Methionine enkephalin
Mycophenolic acid
Neurotrophins
Eight tricyclic antidepressants (TCAs)
Nine TCAs

Donepezil
Five fluoroquinolone
Fluoxetine, norfluxoxetine
Fluoxetine, sertraline, citalopram,
paroxetine
Genistein
Levodopa

Coenzyme Q10

Aminothiols
Anthracyclines, daunorubicin,
doxorubicin and epirubicin
seven -blockers
Five antihistamines, phenothiazines,
anesthetics
Arginine and dimethylated arginines
Aripiprazole
Free calcium, calcium containing
species
Captopril, indapramide

[123]
[293]
[124]

MEKC-LIF
CZEUV

25 mmol L1 boric acid (pH 9.6) with 60 mmol L1 SDS


20 mmol L1 borax, 30 mmol L1 phosphate buffer (pH 8.6)

Ref

MEKC- LIF

Serum, carcinoma cell


Plasma, serum

Aliphatic amines
Allantoin, hypoxanthine, xanthine, and
uric acid
Amine metabolites

Method

BGE

20 mmol L1 borate buffer (pH 9.3) with 50 mmol L1 sodium cholate, 5 mmol L1 -CD,
20 mmol L1 Brij 35, 7% (v/v) MeOH
10 mmol L1 phosphate buffer (pH 12.0) with 10 mmol L1 SDS
105 mmol L1 borate buffer (pH 9.0) with 30% (v/v) MeOH,

Matrix

Analyte

Table 4 Determination of active pharmaceutical ingredients in biological fluid

46
L. Suntornsuk

ACN, acetonitrile; AEKC, affinity electrokinetic chromatography; AOT, sodium bis(2-ethylhexyl sulfosuccinate); Brij 35, polyoxyethylene glycol dodecyl ether; C4 D, contactless conductivity
detection; CA, cholic acid; CD, cyclodextrin; CL, chemiluminescence; CZE, capillary zone electrophoresis; DAD, diode array detector; ECL, electrogenerated chemiluminescence; ESI, electrospray ionization;
FASS, field amplified sample stacking; GLC, N-methylglucamine; HIS, histidine; HSA, human serum albumin; HTAB, hexadecyltrimethylammonium bromide; IACE, immunoaffinity electrokinetic
chromatography; ICP, inductively coupled plasma; IT, ion trap; LED, light-emitting diode; LIF, laser induced fluorescence; MEEKC, microemulsion electrokinetic chromatography; MEKC, micellar
electrokinetic chromatography; MeOH, methanol; MES, 2-(N-morpholino)ethane sulfonic acid; MS, mass spectrometry; NACE, non-aqueous capillary electrophoresis OES, optical emission spectrometry;
PDMA, polymethylacrylamide; PVA, poly(vinyl alcohol); SDS, sodium dodecyl sulfate; SOS, sodium octanesulfonate; TEA, triethanolamine; Tris, tris(hydroxymethyl)aminomethane

CECL
CE-ECL
CE-ESI-ITMSMS
CE-C4D
CE-LIF
8.0 mmol L1 borate buffer with 0.5 mmol L1 luminal, 0.1 mmol L1 K3[Fe(CN)6]
10 mmol L1 phosphate buffer (pH 9.0)
200 mmol L1 ammonium formate buffer, (pH 2.5)
10 mmol L1 MES with 10 mmol L1 DL-His, 50 mol L1 HTAB (pH 6.0)
50 mmol L1 borate buffer (pH 9.0) with 100 mmol L1 GLC
Plasma
Urine
Plasma
Plasma, serum, urine
Plasma
Puerarin
Quinolone residues
Short-chain carnitines
Valproic acid
Vigabatrin

[132]
[133]
[79]
[160]
[127]

Method
BGE
Matrix
Analyte

Table 4 (continued)

Ref

Recent advances of capillary electrophoresis in pharmaceutical analysis

47

channels can be achieved by electrokinetic pumping.


Glass is widely used as chip material, because the
fabrication techniques and its surface chemistry are well
understood, its EOF is high and reproducible and it is
commercially available. Polymers can be alternative
substrates because of their lower cost. Electrochemical
detection is standard for microchip format, because it is
sensitive, compatible with microfabrication techniques,
inexpensive, and can be miniaturized without loss of
performance. Advances in microchip CE include novel
materials [309, 310], applications (enantiomer separations
[311, 312] and protein-binding interactions [313]), and
hyphenated techniques (FImicrochip CE [165] and
microchip CEMS [71]). Advantages of nanomaterials
include low detection potentials, high sensitivity, stability,
resistance to passivation, improved LOD, and separation
performance. Carbon nanotubes (CNTs) have recently
been proposed as novel materials with unique geometric,
mechanical, electronic, and chemical properties [309].
CNT electrodes of small dimensions contain large active
surfaces which enhance electronic transfer and have strong
sorption capacity [314, 315]. CNTs have been used for
analysis of water-soluble vitamins (folic acid, pyridoxine
hydrochloride, and ascorbic acid) in pharmaceutical
preparations [309], neurotransmitters and metabolites in
brain homogenates [316], antioxidants [317], and catecholamine, herbicide, and phenolic pollutants [318].
Multiplexed CE also enables high-throughput analysis,
because several samples can be analyzed at the same time
in arrays of capillaries. Examples of multiplexed CE in
drug analysis include determination of log P [319322] and
pKa [323, 324], separation of enantiomers [278], and
enzyme activity screening [325]. Work on the development
of a low-flow multiplexed interface for CEESI-IT-MS
using a sequential spray has recently been described by
Chen et al. [326] and Li et al. [327].

Conclusions
During recent decades the high potential of CE in analytical
chemistry has been demonstrated, and has led to numerous
applications and advances. This article shows that CE is
now of equal importance to HPLC in drug analysis. The
methods complement each other in the highly competitive
pharmaceutical industry. On the basis of the advances
described in this review, CE can be used to separate and
analyze various classes of APIs in different matrices.
Variation of CE with new detectors enhances the flexibility
of the method and the capability of detection of new drug
entities with better sensitivity and selectivity. Evidently,
CE-based techniques will continue to grow further with
novel advances, and with improved robustness, sensitivity,

48

L. Suntornsuk

and speed; these make it a valuable tool for the pharmaceutical industry which will definitely facilitate drug
discovery, development, and quality control.

Acknowledgement The author thanks Athiporn Doomkaew, Brompoj Prutthiwanasan, Chusak Ardsoongnearn, Methinee Srisunakrua,
and Sumate Thiangthum for their technical assistance.

References
1.
2.
3.
4.
5.
6.

7.
8.
9.
10.
11.
12.
13.
14.
15.
16.
17.
18.
19.
20.
21.
22.
23.
24.
25.
26.
27.
28.
29.
30.
31.

http://www.who.int. Accessed 10 Jan 2010


Geiser L, Veuthey J-L (2009) Electrophoresis 30:3649
Geiser L, Veuthey J-L (2007) Electrophoresis 28:4557
Catai APF, Carrilho E, Lanas FM, Queiroz MEC (2009) J
Chromatogr A 1216:57795782
Morales-Cid G, Cardenas S, Simonet BM, Valcarcel M (2009)
Electrophoresis 30:16841691
Marini RD, Rozet E, Vander Heyden Y, Ziemons E, Boulanger
B, Bouklouze A, Servais A-C, Fillet M, Crommen J, Hubert Ph
(2007) J Pharm Biomed Anal 44:640651
Acedo-Valenzuela M-I, Galeano-Diaz T, Rodriguez AS, Martinez RC, Alvarez JD (2009) Electrophoresis 30:10521058
Morales-Cid G, Crdenas S, Simonet BM, Valcrcel M (2009)
Anal Chem 81:31883193
Ryan R, Donegan S, Power J, McEvoy E, Altria K (2009)
Electrophoresis 30:6582
McEvoy E, Marsh A, Altria K, Donegan S, Power J (2007)
Electrophoresis 28:193207
Lucangioli S, Sabrina F, Mario C, Valeria T (2009) Electrophoresis 30:18991905
Borst C, Holzgrabe U (2008) J Chromatogr A 1204:191196
McEvoy E, Donegan S, Power J, Altria K (2008) Chromatographia 68:4956
Yin C, Cao Y, Ding S, Wang Y (2008) J Chromatogr A
1193:172177
Gebauer P, Boek P (1997) Electrophoresis 18:21542161
Gebauer P, Boek P (2000) Electrophoresis 21:38973904
Gebauer P, Boek P (2002) Electrophoresis 23:38583864
Gebauer P, Mala Z, Boek P (2007) Electrophoresis 28:2632
Mala Z, Krivankova L, Gebauer P, Boek P (2007) Electrophoresis 28:243253
Mala Z, Slampova A, Gebauer P, Boek P (2009) Electrophoresis 30:215229
Gebauer P, Mala Z, Boek P (2009) Electrophoresis 30:2935
Monton MRN, Terabe S (2006) J Chromatogr B 841:8895
Nesmerak K, Nemcova I (2006) Anal Lett 39:10231040
Ivor CF (2007) Electrophoresis 28:1522
Petr J, Maier V, Horkov J, evck J, Strnsk Z (2006) J Sep
Sci 29:27052715
Chen L, Prest JE, Fielden PR, Goddard NJ, Manz A, Day PJR
(2006) Lab Chip 6:474487
Miku P, Markova K, Mark J, Nemec I, Valkov I, Havrnek
E (2009) Curr Pharm Anal 5:171178
Miku P, Markova K, Mark J, Nemec I, Valkov I, Havrnek
E (2008) J Chromatogr B 875:266272
Kurzawa M, Jastrzebska A, Szlyk E (2009) Chem Papers
63:255260
Miku P, Kubacak P, Valkov I, Havrnek E (2006) Meth Exp
Clin Pharmacol 28:595599
Miku P, Markova K, Mark J, Kaniansky D, Valkov I,
Havrnek E (2008) J Chromatogr A 1179:916

32. Sazelova P, Kasicka V, Solnova V, Koval D (2007) J


Chromatogr B 841:145151
33. Bexheti D, Anderson EI, Hutt AJ, Hanna-Brown M (2006) J
Chromatogr A 1130:137144
34. Eldridge SL, Almeida VK, Korir AK, Larive CK (2007) Anal
Chem 79:84468453
35. Huo Y, Kok WTh (2008) Electrophoresis 29:8093
36. Svec F (2009) Electrophoresis 30:S68S82
37. Tanret I, Mangelings D, Vander Heyden Y (2009) Chromatogr
Sci 47:407417
38. Huang Y-P, Liu Z-S, Zheng C, Gao R-Y (2009) Electrophoresis
30:155162
39. Wei GT, Hsu C-F, Liu F-K, Chou F-M (2005) J Chin Chem Soc
52:741751
40. Guo HZ, Wang LL, Bi KS, Sun YQ (2005) J Liq Chromatogr
28:647658
41. Dong J, Xie CH, Tian RJ, Wu RN, Hu JW, Zou H (2005)
Electrophoresis 26:34523459
42. Priego-Capote F, Ye L, Shakil S, Shamsi SA, Nilsson S (2008)
Anal Chem 80:28812887
43. Wang HF, Zhu YZ, Yan XP, Gao RY, Zheng JY (2006) Adv
Mater 18:32663270
44. Zaidi SA, Cheong WJ (2009) Electrophoresis 30:16031607
45. Zaidi SA, Han KM, Kim SS, Hwang DG, Cheong WJ (2009) J
Sep Sci 32:9961001
46. Li HF, Zheng H, Chen Z, Lin J-M (2009) Electrophoresis
30:10221029
47. Aturki Z, DOrazio G, Fanali S, Rocco A, Bortolotti F, Gottardo
R, Tagliaro F (2009) J Chromatogr A 1216:36523659
48. Choodum A, Thavarungkul P, Kanatharana P, Smith NW (2009)
Anal Sci 25:517522
49. Zheng C, Liu Z, Gao R, Zhang L, Zhang Y (2007) Anal Bioanal
Chem 388:11371145
50. Ou J, Li X, Feng S, Dong J, Dong X, Kong L, Ye M, Zou H
(2007) Anal Chem 79:639646
51. Jemere AB, Martinez D, Finot M, Harrison DJ (2009)
Electrophoresis 30:42374244
52. Dong X, Wu R, Dong J, Wu M, Zhu Y, Zou H (2008)
Electrophoresis 29:39333940
53. Chisuwan P, Nacapricha D, Wilairat P, Jiang Z, Smith NW
(2008) Electrophoresis 29:23012309
54. Guzman NA, Blanc T, Phillips TM (2008) Electrophoresis
29:32593278
55. Phillips TM, Wellner EF (2007) Electrophoresis 28:3041
3048
56. Amundsen LK, Siren H (2007) Electrophoresis 28:99113
57. Guzman NA, Stubbs RJ, Phillips TM (2006) Drug Discov
Today: Technol 3:2937
58. Guzman NA, Phillips TM (2005) Anal Chem 77:61A67A
59. Guzman NA (2004) Anal Bioanal Chem 378:3739
60. Guzman NA, Stubbs RJ (2001) Electrophoresis 22:3602
3628
61. Martnez-Gmez MA, Villanueva-Camaas RM, Sagrado S,
Medina-Hernndez MJ (2007) Electrophoresis 28:30563063
62. Kalish H, Phillips TM (2010) J Chromatogr B 878:194200
63. Chen H-X, Busnel J-M, Peltre G, Zhang X-X, Girault HH (2008)
Anal Chem 80:95839588
64. Gimenez E, Benavente F, de Bolos C, Nicolas E, Barbosa J,
Sanz-Nebot V (2009) J Chromatogr A 1216:25742582
65. Qi X-H, Zhang L-W, Zhang X-X (2008) Electrophoresis
29:33983405
66. Wellner EF, Kalish H (2008) Electrophoresis 29:34773483
67. Staub A, Schappler J, Rudaz S, Veuthey J-L (2009) Electrophoresis 30:16101623
68. Gasper A, Englmann M, Fekete A, Harir M, Schmitt-Kopplin P
(2008) Electrophoresis 29:6679

Recent advances of capillary electrophoresis in pharmaceutical analysis


69. Scriba GKE (2007) J Chromatogr A 1159:2841
70. Schappler J, Guillarme D, Rudaz S, Veuthey J-L (2008)
Electrophoresis 29:1119
71. Schappler J, Veuthey J-L, Rudaz S (2008) Sep Sci Tech 9:477521
72. Sniehotta M, Schiffer E, Zrbig P, Novak J, Mischak H (2007)
Electrophoresis 28:14071417
73. Smyth WF (2006) Electrophoresis 27:20512062
74. Smyth WF (2005) Electrophoresis 26:13341357
75. Shamsi SA, Miller BE (2004) Electrophoresis 25:39273961
76. Smyth WF, Rodriguez V (2007) J Chromatogr A 1159:159174
77. Gottardo R, Bortolotti F, De Paoli G, Pascali JP, Miksik I,
Tagliaro F (2007) J Chromatogr A 1159:185189
78. Gottardo R, Bortolotti F, De Paoli G, Pascali JP, Miksik I,
Tagliaro F (2007) J Chromatogr A 1159:190197
79. Desiderio C, Rossi AD, Inzitari R, Mancinelli A, Rossetti DV,
Castagnola M, Messana I (2008) Anal Bioanal Chem 390:1637
1644
80. Desiderio C, Rossetti DV, Perri F, Giardina B, Messana I,
Castagnola M (2008) J Chromatogr B 875:280287
81. Olsson J, Marlin ND, Blomberg LG (2007) Chromatographia
66:421425
82. Somsen GW, Mol R, De Jong GJ (2003) J Chromatogr A
1000:953961
83. Suomi J, Wiedmer SK, Jussila M, Riekkola ML (2002) J
Chromatogr A 970:287296
84. Frommberger M, Schmitt-Kopplin P, Menzinger F, Albrecht V,
Schmid M, Eberi L, Hartmann A, Kettrup A (2003) Electrophoresis 24:30673074
85. Amundsen LK, Kokkonen JT, Rovio S, Siren H (2004) J
Chromatogr A 1040:123131
86. Ozaki H, Terabe S (1998) J Chromatogr A 794:317325
87. Hou J, Zheng J, Shamsi SA (2007) J Chromatogr A 1159:208
216
88. Wan H, hman M, Holmn AG (2009) J Med Chem 52:16931700
89. Zheng J, Norton D, Shamsi SA (2006) Anal Chem 78:1323
1330
90. Zheng J, Shamsi SA (2006) Electrophoresis 27:21392151
91. Bragg W, Norton D, Shamsi SA (2008) J Chromatogr B
875:304316
92. Vassort A, Shaw PN, Ferguson PD, Szcs R, Barrett DA (2008)
Electrophoresis 29:35633574
93. Lu M, Zhang L, Qiu B, Feng Q, Xia S, Chen G (2008) J
Chromatogr A 1193:156163
94. Kitagawa F, Inoue K, Hasegawa T, Kamiya M, Okamoto Y,
Kawase M, Otsuka K (2006) J Chromatogr A 1130:219226
95. Jing X, Yan X, Shiheng C, Yafeng G (2009) Progress in
Chemistry 21:13251334
96. Xiao D, Yan L, Yuan H, Zhao S, Yang X, Choi MMF (2009)
Electrophoresis 30:189202
97. Bruno AE, Maystre F, Krattiger B, Nussbaum P, Gassmann E
(1994) Trends Anal Chem 13:190198
98. Tong W, Yeung ES (1995) J Chromatogr A 718:177185
99. Macka M, Andersson P, Haddad PR (1996) Electrophoresis
17:18981905
100. Macka M, Paull B, Andersson P, Haddad PR (1997) J
Chromatogr A 767:303310
101. Macka M, Nesterenko P, Andersson P, Haddad PR (1998) J
Chromatogr A 803:279290
102. Macka M, Nesterenko P, Haddad PR (1999) Microcolumn Sep
11:19
103. Vachirapatama N, Doble P, Yu Z, Macka M, Haddad PR (2001)
Anal Chim Acta 434:301307
104. Vachirapatama N, Macka M, Haddad PR (2002) Anal Bioanal
Chem 374:10821085
105. Johns C, Shaw MJ, Macka M, Haddad PR (2003) Electrophoresis 24:557566

49
106. King M, Paul B, Haddad PR, Macka M (2002) Analyst
127:15641567
107. Breadmore MC, Henderson RD, Fakhari AR, Macka M, Haddad
PR (2007) Electrophoresis 28:12521258
108. Shih C, Lin C (2005) Electrophoresis 26:962969
109. Huang H-M, Lin C-H (2005) J Chromatogr B 816:113119
110. Yu Q, Zhao S, Ye F, Li S (2007) Anal Biochem 369:187191
111. Tsai C, Liu JT, Shu YR, Chan PH, Lin CH (2006) J Chromatogr
A 1101:319323
112. Yang X, Yuan H, Wang C, Su X, Hu L, Xiao D (2007) J Pharm
Biomed Anal 45:362366
113. Su A-K, Lin C-H (2003) J Chromatogr B 785:3946
114. Schappler J, Staub A, Veuthey J-L, Rudaz S (2008) J
Chromatogr A 1204:183190
115. Schulze P, Belder D (2009) Anal Bioanal Chem 393:515525
116. Couderc F, Causse E, Bayle C (1998) Electrophoresis 19:2777
2790
117. Tolba K, Belder D (2007) Electrophoresis 28:29342941
118. Horstktter C, Blaschke G (2001) J Chromatogr B 754:169178
119. Soetebeer UB, Schierenberg MO, Schulz H, Grnefeld G,
Andresen P, Blaschke G (2000) J Chromatogr B 745:271278
120. Zaugg S, Zhang X, Sweedler J, Thormann W (2001) J
Chromatogr B 752:1731
121. Alnajjar A, Butcher JA, McCord B (2004) Electrophoresis
25:15921600
122. Huhn C, Ptz M, Martin N, Dahlenburg R, Pyell U (2005)
Electrophoresis 26:23912401
123. Deng Y-H, Wang H, Zhong L, Zhang H-S (2009) Talanta
77:13371342
124. Tseng H-M, Li Y, Barrett DA (2007) Anal Bioanal Chem
388:433439
125. Shen C-C, Tseng W-L, Hsieh M-M (2009) J Chromatogr A
1216:288293
126. Whitaker G, Lillquist A, Pasas SA, OConnor R, Regan F, Lunte
CE, Smyth MR (2008) J Sep Sci 31:18281833
127. Musenga A, Mandrioli R, Comin I, Kenndler E, Raggi M-A
(2007) Electrophoresis 28:35353541
128. Garcia-Campaa AM, Lara FJ, Gmiz-Gracia L, Huertas-Prez
JF (2009) Trends Anal Chem 28:973986
129. Wang J, Peng Z, Yang J, Wang X, Yang N (2008) Talanta
75:817823
130. Zhang X, Xuan Y, Sun A, Lv Y, Hou X (2009) Luminescence
24:243249
131. Wang J, Yang Z, Wang X, Yang N (2008) Talanta 76:8590
132. Han S (2009) J Chromatogr B 877:15911594
133. Fu Z, Liu Y, Wang L, Wang Y (2009) Chromatographia
69:11011105
134. Pungor E (1965) Oscillometry and conductiometry. Pergamon
Press, Oxford
135. Gas B, Demjanenko M, Vacik J (1980) J Chromatogr 192:253257
136. Zemann AJ, Schnell E, Volgger D, Bonn GK (1998) Anal Chem
70:563567
137. Zemann AJ (2001) Trends Anal Chem 20:346354
138. Tanyanyiwa J, Leuthardt S, Hauser PC (2002) Electrophoresis
23:36593666
139. Zemann AJ (2003) Electrophoresis 24:21252137
140. Guijt RM, Evenhuis CJ, Macka M, Haddad PR (2004)
Electrophoresis 25:40324057
141. Kub P, Hauser PC (2004) Electroanalysis 16:20092021
142. Solinova V, Kasicka V (2006) J Sep Sci 29:17431762
143. Pumera M (2007) Talanta 74:358364
144. Matysik FM (2008) Microchim Acta 160:114
145. Kub P, Hauser PC (2008) Anal Chim Acta 607:1529
146. Kub P, Hauser PC (2009) Electrophoresis 30:176188
147. Kub P, Nguyen HTA, Macka M, Haddad PR, Hauser PC
(2007) Electroanalysis 19:20592065

50
148. Kappes T, Galliker B, Schwarz MA, Hauser PC (2001) Trends
Anal Chem 20:133139
149. Kub P, Reinhardt M, Muller B, Hauser PC (2004) J Environ
Monit 6:169174
150. Xu Y, Wang WL, Li SFY (2007) Electrophoresis 28:15301539
151. Muhlberger H, Hwang W, Guber AE, Saile V, Hoffmann W
(2008) IEEE Sens J 8:572579
152. Hilder EF, Zemann AJ, Macka M, Haddad PR (2001) Electrophoresis 22:12731281
153. Kub P, Kub P, Kub V, Hauser PC, Boek P (2008) J
Chromatogr A 1190:377382
154. Oudhoff KA, Macka M, Haddad PR, Schoenmakers PJ, Kok WT
(2005) J Chromatogr A 1068:183187
155. Tanyanyiwa J, Hauser PC (2004) Electrophoresis 25:30103016
156. Felix FS, Quintino MSM, Carvalho AZ, Coelho LHG et al
(2006) J Pharm Biomed Anal 40:12881292
157. Petsch M, Mayer-Helm BX, Sauermann R, Joukhadar C,
Kenddler E (2004) Electrophoresis 25:22922298
158. Law WS, Kub P, Yuan LL, Zhao JH, Li SFY, Hauser PC
(2006) Electrophoresis 27:19321938
159. Caslavska J, Thormann W (2006) Electrophoresis 27:4618
4630
160. Belin GK, Krhenbhl S, Hauser PC (2007) J Chromatogr B
847:205209
161. Petr J, Maier V, Horakova J, Sevcik J (2006) Electrophoresis
27:47354745
162. Gong XY, Kub P, Tanyanyiwa J, Hauser PC (2005) J
Chromatogr A 1082:230234
163. J P, Los P, Spil Z, Pospiilov M, Polek M (2008)
Electrophoresis 29:35113518
164. Nussbaumer S, Fleury-Souverain S, Rudaz S, Bonnabry P,
Veuthey J-L (2009) J Pharm Biomed Anal 49:333337
165. Lu W-J, Chen Y-L, Zhu J-H, Chen X-G (2009) Electrophoresis
30:8391
166. Kub P, Karlberg B (2009) Anal Chim Acta 648:129145
167. Kub P, Engstrm A, Olsson JC, Thorsen G, Tryzell R,
Karlberg B (1997) Anal Chim Acta 337:117124
168. Fang ZL, Liu ZS, Shen Q (1997) Anal Chim Acta 346:135143
169. Chen XG, Fan LY, Hu ZD (2004) Electrophoresis 25:39623969
170. Chen YL, Lu WJ, Chen XG, Hu ZD (2007) Electrophoresis
38:3344
171. Trojanowicz M (2009) Anal Chim Acta 653:3658
172. Kub P, Hauser PC (2008) Comprehensive Anal Chem 54:287
307
173. Wu C-H, Scampavia L, Ruzicka J (2003) Analyst 128:11231130
174. Liu X, Zhang J, Chen X (2008) Anal Sci 24:10311037
175. Liu X, Li S, Zhang J, Chen X (2009) J Chromatogr B 877:3144
3150
176. Zhang Y, Wu D-R, Wang-Iverson DB, Tymiak AA (2005) Drug
Discov Today 10:571577
177. Chankvetadze B (2009) Electrophoresis 30:S211S221
178. Lammerhofer M (2005) J Chromatogr A 1068:330
179. Gbitz G, Schmid MG (2008) J Chromatogr A 1204:140156
180. Preinerstorfer B, Lmmerhofer M, Lindner W (2009) Electrophoresis 30:100132
181. Fanali S (2009) Electrophoresis 30:S203S210
182. Snchez-Hernndez L, Crego AL, Marina ML, Garcia-Ruiz C
(2008) Electrophoresis 29:237251
183. Chankvetadze B (2007) J Chromatogr A1168:4570
184. Van Eeckhaut A, Michotte Y (2006) Electrophoresis 27:2880
2895
185. Scriba GKE (2003) Electrophoresis 24:24092421
186. Wang Z, Ouyanga J, Baeyens WRG (2008) J Chromatogr B
862:114
187. Gilpin RK, Gilpin CS (2009) Anal Chem 81:46794694
188. Grosche O, Zeitz M (2008) Sep Sci Tech 9:95121

L. Suntornsuk
189. Holzgrabe U (2008) Sep Sci Tech 9:245258
190. Mallampati S, Pauwels J, Hoogmartens J, Van Schepdael A
(2008) Sep Sci Tech 9:259315
191. Guo A, Camblin G, Han M, Meert C, Park S (2008) Sep Sci
Tech 9:357399
192. Adu JK, Euerby MR, Tettey JNA, Skellern GG (2008) Sep Sci
Tech 9:439476
193. Suntornsuk L (2007) J Chromatogr Sci 45:559577
194. Altria K, Marsh A, Snger-van de Griend C (2006) Electrophoresis 27:22632282
195. Morzunova TG (2006) Pharm Chem J 40:3952
196. Ali I, Aboul-Enein HY, Gupta VK, Li SFY (2005) J Cap Elect
Micro Tech 9:8599
197. Timerbaev AR, Keppler BK (2007) Anal Biochem 369:17
198. Hoff R, Kist TBL (2009) J Sep Sci 32:854866
199. Castro-Puyana M, Crego AL, Marina ML (2007) Electrophoresis
29:274293
200. Little MJ, Paquette DM, Roos PK (2006) Electrophoresis
27:24772485
201. Kamoda S, Kakehi K (2006) Electrophoresis 27:24952504
202. Chu Q, Jiang L, Tian X, Ye J (2008) Anal Chim Acta 606:246
251
203. Prutthiwanasan B, Suntornsuk L (2010) J Sep Sci 33:17
204. Volpi N (2008) Electrophoresis 29:35043510
205. Zhou S, Wang Y, De Beer T, Baeyens WRG, Fei GT, Dilinuer
M, Ouyang J (2008) Electrophoresis 29:23212329
206. Alnajjar AO (2008) Chromatographia 68:437442
207. Malavaki CJ, Asimakopoulou AP, Lamari FN, Theocharis AD,
Tzanakakis GN, Karamanos NK (2008) Anal Biochem 374:213
220
208. Abdelal AA, Kitagawa S, Ohtani H, El-Enany N, Belal F,
Walash MI (2008) J Pharm Biomed Anal 46:491497
209. Faria AF, De Souza MVN, De Oliveira MAL (2008) J Braz
Chem Soc 19:389396
210. Sirichai S, Khanatharana P (2008) Talanta 76:11941198
211. Michalska K, Pajchel G, Tyski S (2009) J Chromatogr A
1216:29342942
212. Faria AF, De Souza MVN, Bruns RE, De Oliveira MAL (2008) J
Chromatogr A 1202:224228
213. Dalmora SL, da Silva Sangoi M, da Silva LM, Macedo RO,
Barth T (2008) J Sep Sci 31:169176
214. Zhang L, Dong S, Yang Z, Wang Q, He P, Fang Y (2008) J
Pharm Biomed Anal 48:198200
215. Volpi N (2009) J Pharm Biomed Anal 49:868871
216. Ortner K, Buchberger W, Himmelsbach M (2009) J Chromatogr
A 1216:29532957
217. Michalska K, Pajchel G, Tyski S (2008) J Pharm Biomed Anal
48:321330
218. King JT, Desai UR (2008) Anal Biochem 380:229234
219. Patel RP, Narkowicz C, Hutchinson JP, Hilder EF, Jacobson GA
(2008) J Pharm Biomed Anal 46:3035
220. Vujic Z, Kuntic V, Ivkovic B (2008) Monatsh Chem 139:81
87
221. Patnala S, Kanfer I (2008) J Pharm Biomed Anal 48:440446
222. Huidobro AL, Garca A, Barbas C (2009) J Pharm Biomed Anal
49:13031307
223. Jabbaribar F, Mortazavi A, Jalali-Milani R, Jouyban A (2008)
Chem Pharm Bull 56:16391644
224. Nmeth T, Jankovics P, Nmeth-Palots J, Kszegi-Szalai H
(2008) J Pharm Biomed Anal 47:746749
225. Zhang Q, Li YF, Huang CZ (2008) Talanta 76:4448
226. Nigovi B, Vegar I (2008) Croatica Chem Acta 81:615622
227. Dalmora SL, DAvila FB, da Silva LM, Bergamo AC,
Zimmermann ES (2009) J Chromatogr B 877:24712476
228. Lodn H, Pettersson C, Arvidsson T, Amini A (2008) J
Chromatogr A 1207:181185

Recent advances of capillary electrophoresis in pharmaceutical analysis


229. Foteeva LS, Stolyarova NV, Timerbaev AR, Keppler BK (2008)
J Pharm Biomed Anal 48:218222
230. Farina C, Kremser L, Raggi MA, Kenndler E (2008) J Pharm
Biomed Anal 46:471476
231. Fareed J, Hoppensteadt D, Schultz C, Ma Q, Kujawski MF,
Neville B, Messmore H (2004) Curr Pharm Des 10:983999
232. Korir AK, Larive CK (2009) Anal Bioanal, Chem 393:155169
233. Wang S-L, Huang X-J, Fang Z-L (2001) Anal Chem 73:45454549
234. Fan LY, Cheng YQ, Chen HL, Chen XG, Liu LH, Hu ZD (2004)
Electrophoresis 25:31633167
235. ICH Harmonised Tripartite Guideline: Impurities in New Drug
Substances Q3A(R2) Current step 4 version dated 25 October
2006
236. ICH Harmonised Tripartite Guideline: Impurities in New Drug
Products Q3B(R2) Current step 4 version dated 2 June 2006
237. ICH Harmonised Tripartite Guideline: Impurities Guideline for
Residual Solvents Q3C(R3) Current step 4 version dated 17 July
1997
238. Bartos D, Gorog S (2008) Curr Pharm Anal 4:215230
239. Jouyban A, Kenndler E (2008) Electrophoresis 29:35313551
240. Jankovcs P, Nmeth T, Nmeth-Palots J, Kszegi-Szalai H
(2008) Chromatographia 38:S43S48
241. Nigovic B, Damic M, Injac R, Glavac NK, Strukelj B (2009)
Chromatographia 69:17
242. Furlanetto S, Orlandini S, Giannini I, Beretta G, Pinzauti S
(2009) Electrophoresis 30:633643
243. Li J, Jiang Y, Sun T, Ren S (2008) J Pharm Biomed Anal
47:929933
244. Fayed AS, Weshahy SA, Shehata MA, Hassan NY, Pauwels J,
Hoogmartens J, Schepdael AV (2009) J Pharm Biomed Anal
49:193200
245. Khn KD, Weber C, Kreis S, Holzgrabe U (2008) J Pharm
Biomed Anal 48:612618
246. Sekar R, Azhaguvel S (2009) Chromatographia 69:765769
247. Sekar R, Azhaguvel S (2008) Chromatographia 67:389398
248. Hommerson P, Khan AM, Bristow T, Harrison MW, de Jong GJ,
Somsen GW (2009) Rapid Commun Mass Spectrom 23:2878
2884
249. Zacharis CK, Tzanavaras PD, Notou M, Zotou A, Themelis DG
(2009) J Pharm Biomed Anal 49:201206
250. Volpi N, Maccari F, Linhardt RJ (2009) Anal Biochem 388:140
145
251. Somsen GW, Tak YH, Torano JS, Jongen PMJM, de Jong GJ
(2009) J Chromatogr A 1216:41074112
252. Wielgos T, Havel K, Ivanova N, Weinberger R (2009) J Pharm
Biomed Anal 49:319326
253. Nogueira DR, da Silva Sangoi M, da Silva LM, Todeschini V,
Dalmora SL (2008) J Sep Sci 31:30983105
254. Pajchel G, Michalska K, German R, Tyski S (2008) Chromatographia 68:587591
255. Wang Y, Zhou L, Hui Y, Chen X (2009) Chromatographia
70:293297
256. Suntornsuk L, Ployngam S (2010) J Pharm Biomed Anal
51:541548
257. Huang L, Lin J-M, Yu L, Xu L, Chen G (2009) Electrophoresis
30:10301036
258. Zhang H, Shao HAY, Zhang Z (2008) Chromatographia 68:5358
259. Huang L, Lin J-M, Yu L, Xu L, Chen G (2008) Electrophoresis
29:35883594
260. Kwaterczak A, Duszczyk K, Bielejewska A (2009) Anal Chim
Acta 645:98104
261. Kokiashvili NG, Wongwan S, Landgraf C, Michaelis K,
Hammitzsch-Wiedemann M, Scriba GKE (2009) J Pharm
Biomed Anal 50:10501053
262. Michalska K, Pajchel G, Tyski S (2008) J Chromatogr A
1180:179186

51
263. Rousseau A, Chiap P, Ivanyi R, Crommen J, Fillet M, Servais AC (2008) J Chromatogr A 1204:219225
264. Elbashir AA, Suliman FEO, Saad B, Aboul-Enein HY (2009)
Talanta 77:13881393
265. Chou Y-W, Huang W-S, Ko C-C, Chen S-H (2008) J Sep Sci
31:845852
266. Musenga A, Mandrioli R, Morganti E, Fanali S, Raggi MA
(2008) J Pharm Biomed Anal 46:966970
267. Glwka F, Karaniewicz M (2007) Electrophoresis 28:2726
2737
268. Denola NL, Quiming NS, Catabay AP, Saito Y, Jinno K (2008) J
Sep Sci 31:27012706
269. Elbashir AA, Saad B, Ali ASM, Saleh MI (2008) J AOAC Int
91:536541
270. Ibrahim WAW, Hermawan D, Sanagi MM, Aboul-Enein HY
(2009) J Sep Sci 32:466471
271. Shakalisava Y, Regan F (2008) J Sep Sci 31:11371143
272. Deeb SE, Hasemann P, Watzig H (2008) Electrophoresis
29:35523562
273. Ibrahim WAW, Hermawan D, Sanagi MM (2007) J Chromatogr
A 1170:107113
274. Schappler J, Guillarme D, Prat J, Veuthey J-L, Rudaz S (2008)
Electrophoresis 29:21932202
275. Martins LF, Yegles M, Wennig R (2008) J Chromatogr B 862:7985
276. Lipka E, Vaccher C, Bonte J-P (2009) Chromatographia 69:465
472
277. Zandkarimi M, Shafaati A, Foroutan SM (2008) Iranian J Pharm
Res 7:275281
278. Saavedra L, Nickerson B, Borjas RE, Lynen F, Sandra P (2008) J
Chromatogr B 875:248253
279. Gagyi L, Gyresi A, Kilr F (2008) J Biochem Biophys Methods
70:12681275
280. Liao W-S, Lin C-H, Chen C-Y, Kuo C-M, Liu Y-C, Wu J-C, Lin
C-E (2007) Electrophoresis 28:39223929
281. Danel C, Azaroual N, Brunel A, Lannoy D, Odou P, Dcaudin B,
Vermeersch G, Bonte J-P, Vaccher C (2009) Tetrahedron
Asymmetry 20:11251131
282. Chu B-L, Guo B, Zuo H, Wang Z, Lin J-M (2008) J Pharm
Biomed Anal 46:854859
283. Sungthong B, J P, Scriba GKE (2008) J Pharm Biomed Anal
46:959965
284. Jamali B, Bjrnsdottir I, Nordfang O, Hansen SH (2008) J
Pharm Biomed Anal 46:8287
285. Prokhorova AF, Shapovalova EN, Shpak AV, Staroverov SM,
Shpigun OA (2009) J Chromatogr A 1216:36743677
286. Dai R, Nie X, Li H, Saeed MK, Deng Y, Yao G (2007)
Electrophoresis 28:25662572
287. Hedeland Y, Lehtinen J, Pettersson C (2007) J Chromatogr A
1141:287294
288. Chu B-L, Feng Q, Wang Z, Lin J-M (2009) Chromatographia
70:817824
289. Zhu F, Du Y, Chen J, Chen B, Zhu Y, Zhai X, Xu S, Zhou W
(2009) Chromatographia 69:13151320
290. Lloyd DK (2008) J Chromatogr B 866:154166
291. Zhang J, Konecny J, Glatz Z, Hoogmartens J, Schepdael AV
(2007) Curr Anal Chem 3:197217
292. Carlucci F, Tabucchi A (2009) J Chromatogr B 877:33473357
293. Causs E, Pradelles A, Dirat B, Negre-Salvayre A, Salvayre R,
Couderc F (2007) Electrophoresis 28:381387
294. Zinellu A, Sotgia S, Zinellu E, Pinna A, Carta F, Gaspa L,
Deiana L, Carru C (2007) Electrophoresis 28:19421948
295. Musenga A, Saracino MA, Spinelli D, Rizzato E, Boncompagni G, Kenndler E, Raggi MA (2008) Anal Chim Acta
612:204211
296. Deng B, Zhu P, Wang Y, Feng J, Li X, Xu X, Lu H, Xu Q (2008)
Anal Chem 80:57215726

52
297. Yeh H-H, Yang Y-H, Ko J-Y, Chen S-H (2008) Electrophoresis
29:36493657
298. Lu C-C, Jong Y-J, Ferrance J, Ko W-K, Wu S-M (2007)
Electrophoresis 28:32903295
299. Feng H-Y, Hou S-R, Zheng N, Li X-J, Hu Z-B, Yuan Z-B (2008)
Chromatographia 68:431435
300. Zhao S, Bai W, Wang B, He M (2007) Talanta 73:142146
301. Wei S-Y, Yeh H-H, Liao F-F, Chen S-H (2009) J Sep Sci
32:413421
302. Ramautar R, Ratnayake CK, Somsen GW, de Jong GJ (2009)
Talanta 78:638642
303. Carlucci F, Anzini M, Rovini M, Cattaneo D, Merlini S,
Tabucchi A (2007) Electrophoresis 28:39083914
304. Lin S-C, Whang C-W (2008) J Sep Sci 31:39213929
305. Simpson SL, Quirino JP, Terabe S (2008) J Chromatogr A
1184:504541
306. Breadmore MC, Thabano JRE, Dawod M, Kazarian AA, Quirino
JP, Guijt RM (2009) Electrophoresis 30:230248
307. Musijowski J, Pobozy E, Trojanowicz M (2006) J Chromatogr A
1104:337345
308. Puig P, Borrull F, Calull M, Aguilar C (2005) Electrophoresis
26:954961
309. Crevilln AG, Pumera M, Gonzlez MC, Escarpa A (2008)
Electrophoresis 29:29973004
310. Pumera M, Escarpa A (2009) Electrophoresis 30:33153323

L. Suntornsuk
311. Sanchez-Hernndez L, Garca-Ruiz C, Marina ML, Crego AL
(2010) Electrophoresis 31:2843
312. Guihen E, Hogan A-M, Glennon JD (2009) Chirality 21:292298
313. Nie Z, Fung YS (2008) Electrophoresis 29:19241931
314. Ajayan PM (1999) Chem Rev 99:17871799
315. Trojanowicz M (2006) Trends Anal Chem 25:480489
316. Vlkov M, Schwarz MA (2007) J Chromatogr A 1142:214221
317. Crevillen AG, Pumera M, Gonzales MC, Escarpa A (2009) Lab
Chip 9:346353
318. Chicharro M, Arribas AS, Moreno M, Bermejo E, Zapardiel EA
(2007) Talanta 74:376386
319. Wehmeyr KR, Tu J, Yin Y (2003) LCGC North Am 21:1078
1088
320. Marsh A, Altria KD (2006) Chromatographia 64:327333
321. Wong KS, Kenseth J, Strasburg R (2004) J Pharm Sci 93:916931
322. Wan H, Holmen AG (2009) Comb Chem High Throughput
Screen 12:315329
323. Gong X, Figus M, Plewa J, Levorse DA, Zhou L, Welch CJ
(2008) Chromatographia 28:219225
324. Shalaeva M, Kenseth J, Lombardo F, Bastin A (2008) J Pharm
Sci 97:25812606
325. Dai J, Tu J, Anderson LN, Bao JJ, Liu C, Quay B, Wehmeyer
KR (2002) LCGC North America 20:606617
326. Chen C-J, Li F-A, Her G-R (2008) Electrophoresis 29:19972003
327. Li F-A, Wu M-C, Her G-R (2006) Anal Chem 78:53165321

Вам также может понравиться