Sei sulla pagina 1di 32

Review

pubs.acs.org/CR

New Advances in Separation Science for Metabolomics: Resolving


Chemical Diversity in a Post-Genomic Era
Naomi L. Kuehnbaum and Philip Britz-McKibbin*
Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Canada

*
S Supporting Information

drug development,5 nutrition,6 food safety/quality,7 and clinical


diagnostics8 to environmental chemistry and toxicology.9
Metabolomics (or metabonomics) is defined as the compre-
hensive analysis of all low molecular weight compounds (<1500
Da) in a sample, such as a cell, biofluid or organism. Although
several related “omics” subdisciplines have been recently coined
in the literature,10−14 they all share the common motif of
untargeted metabolite profiling using new advances in nuclear
magnetic resonance (NMR) and mass spectrometry (MS).
CONTENTS However, direct coupling of high resolution separation
1. Introduction: Separation Principles for Metabolite techniques to NMR15,16 and notably MS-based metabolo-
Profiling A mics17−19 greatly improves analytical performance when
1.1. The Origin of Metabolomics and Its Link to analyzing complex sample mixtures while providing comple-
Separation Science B mentary qualitative information for metabolite identification.
1.2. A Renaissance in Metabolism: The Advent of Indeed, known metabolites in human serum comprise greater
Metabolomics/Metabonomics C than 4000 chemically diverse compounds present at concen-
1.3. Chromatography: Solute Retention with tration levels that span over 9 orders of magnitude,20 which
Improved Column Technology D further expands to over 40 000 annotated compounds when
1.4. Electrophoresis: Ion Mobility with Tunable including exogenous metabolites derived from foods, micro-
Selectivity in Free Solution H biota, drugs, and synthetic chemicals.21 Similar chemical
2. New Advances in Separation Science for Metab- diversity also exists in plants as sessile organisms given their
olomics: More than Separation? K complex secondary metabolism.22 Thus, the total number of
2.1. The Volatile Metabolome by GC-MS M detectable yet identifiable compounds is extensive, which
2.2. The Nonvolatile Metabolome by LC-MS P necessitates the use of complementary separation platforms
2.3. The Ionic Metabolome by CE-MS S for unbiased screening of the metabolome.23
3. Sources of Bias and False Discovery in Metab- Since metabolites represent molecular end points of gene
olomics U expression and cell activity, metabolomics offers a holistic
3.1. Sample Pretreatment and Quality Assurance V approach for understanding the phenotype of an organism. In
3.2. Perspectives and Future Trends in Separa- this context, it plays a vital role in systems biology for
tion Science W elucidating the function of unknown genes24 and enzymes25
Associated Content X while revealing molecular mechanisms underlying drug
Supporting Information X action,26,27 dietary changes,28,29 exercise intervention30,31 and
Author Information X psychosocial impacts of environment.32 Unlike other “omics”
Corresponding Author X domains, the time scale of the metabolome as measured by
Notes X isotope tracer experiments is within milliseconds,33 where
Biographies X subtle changes in gene expression or protein activity lead to
Acknowledgments X large amplifications in metabolite concentrations in order to
References X maintain stability of highly interconnected metabolic net-
works.34 There is growing recognition that living systems
function as dynamic ecosystems or “superorganisms”36 which
1. INTRODUCTION: SEPARATION PRINCIPLES FOR underscores the complex interactions of host, diet and gut
microflora in human health.36−38 Thus, metabolomics provides
METABOLITE PROFILING
unique insight into the functional impact of gene expression39
Metabolomics offers a revolutionary framework for phenotyp-
ing individuals at a molecular level that is needed for new Special Issue: 2013 New Frontiers in Bioanalytical Chemistry
breakthroughs in cell biology and personalized medicine.1−3
Major interest in metabolomics is rapidly expanding in diverse Received: December 4, 2012
research fields ranging from biotechnology and agriculture,4

© XXXX American Chemical Society A dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

complementary to large-scale gene sequencing efforts40 and chromatography, rapid developments were also taking place
genome wide association studies (GWAS) that neglect the in gas chromatography (GC) that coincided with the
impact of environment in the etiology of human diseases.41 For development of sensitive postcolumn detectors for gas effluents
instance, most genetic mutations are not directly associated based on flame ionization detection (FID), 62 electron
with clinical phenotype, whereas aberrant metabolism is capture,63 and electron ionization (EI)-MS.64−66 Pauling et
strongly linked to disease severity and intoxication as in al.67 demonstrated that over 250 peaks from a headspace
classical phenylketonuria (PKU).42 Recent cytochrome P450 analysis of human urine vapor or breath condensates were
(CYP) genotyping initiatives for predicting drug toxicity among detected by GC-FID whose composition was reproducible
individuals have been largely unsuccessful since the expression when standardized methods were used for sample collection.
and functional activity of CYP isoforms is dependent on various The first commercial GC-MS instruments with temperature
confounding nongenetic variables, such as occupational programming further enhanced method performance for
exposure.43 Indeed, the majority of diagnostic tests, pharmaco- characterization of multicomponent mixtures in biological
logical agents and environmental pollutants are comprised of samples.68,69 Untargeted metabolite profiling soon became a
small molecules. However, there is currently no single approach reality with GC-MS instruments configured with quadrupole
amenable to comprehensive metabolite profiling because of the mass filters that allowed for repetitive mass scanning over the
chemical diversity and wide dynamic range of the metabolome full mass range during separation with improved resolution of
that remains largely uncharacterized.44 coeluting peaks.70−72 Also, the use of stable isotopes as internal
1.1. The Origin of Metabolomics and Its Link to Separation standards in GC-MS provided exceptional precision and
Science accuracy for quantitative analysis of metabolites when using
selective reaction monitoring,73 whereas retention index
The origin of metabolomics can be traced to developments in markers based on coinjection of a homologous series of n-
separation science and clinical medicine early last century, as alkane standards corrected for variation in retention times.74
well as to the hypothesis of “biochemical individuality” that The use of glass-based capillary columns to reduce axial
linked metabolite patterns present in biofluids with phenotypic diffusion with the subsequent availability of flexible open
traits associated with human health and disease susceptibil- tubular fused-silica capillaries soon transformed GC-MS into
ity.45,46 Williams and co-workers47 were early practitioners of the “gold standard” for metabolite profiling.75−78
metabolomics using paper chromatography to analyze individ- Although paper and gel electrophoresis have been important
ual metabolic patterns or “metabolic personality” in biological separation techniques for biomolecules since the seminal work
fluids derived from human subjects and animal models. His of Tiselius in 1930,79 it was not until narrow bore tubes80 and
breakthrough work countered genetic fatalism by demonstrat- subsequently fused-silica capillaries81,82 were introduced to
ing that dietary changes and nutritional supplementation can allow for fast separations of polar metabolites under a high
treat human behavioral disorders of obscure etiology, including voltage by capillary electrophoresis (CE). High separation
alcoholism and schizophrenia. The advent of paper and notably efficiency was realized due to the near flat profile of the
2D thin layer chromatography (TLC)48,49 further improved electroosmotic flow (EOF) relative to pressure-driven laminar
peak capacity for resolution of hundreds of “spots” from flow83 and the lack of mass transfer kinetics when performing
complex biofluids that were detected by their intrinsic UV electrophoresis in free solution. Selectivity in CE was readily
absorbance and/or reaction with specific colorimetric extended to include neutral solutes84 and their enantiomers85
reagents.50,51 However, the low separation efficiency, poor when using charged micelles and chiral selectors as additives in
automation, and lack of quantitative analyses limited the the background electrolyte (BGE), respectively. Despite earlier
applicability of planar chromatography for routine metabolite attempts at direct coupling of LC with MS via atmospheric
profiling applications.52 pressure ionization86−90 the first report of direct infusion
Higher resolution separation methods for metabolite electrospray ionization (ESI) of intact proteins by Fenn and co-
profiling awaited later advances in chromatography,53 including workers91 revolutionized metabolite profiling by providing a
column technology, benchtop instrumentation and computer “soft” ionization approach to desolvate ions prior to their
data processing and storage capabilities. For instance, extraction into the low pressure region of the mass analyzer.
Hamilton54 reported that up to 95 ninhydrin-positive The introduction of commercial benchtop tandem mass
compounds were resolved in human urine after separation on spectrometer instruments in the 1990s transformed metabolite
a synthetic anion-exchange resin with postcolumn chemical profiling by enabling multiplexed and high-throughput analysis
derivatization when using a commercial amino acid analyzer. by ESI-MS/MS that is rapidly replacing traditional biochemical
Similarly, Pitt et al.55 demonstrated that up to 150 UV- assays.92,93 The phenomenal explosion of LC-MS during the
absorbing metabolites were detected in human urine when past two decades stems from the ability to perform selective yet
using an ion-exchange resin with an optimized buffer gradient sensitive analyses of polar and thermally unstable metabolites
elution performed over 20 h. New advances in high not amenable by GC-MS without complex sample pretreat-
performance liquid chromatography (HPLC) awaited the ment.94 However, direct coupling of CE with ESI-MS has
development of uniformly packed columns prepared from proven to be more challenging relative to HPLC due to the
porous microparticles (i.e., type B silica <10 μm i.d.) modified high electric field and the low flow rate of the EOF that hinder
with octadecyl silica (i.e., reversed-phase, RP).56,57 This formation of a stable microspray.95,96 Although NMR remains
resulted in high separation efficiency and sample throughput the premier technique for structural elucidation of small
with over 100 metabolites resolved from an acidified urine molecules with early studies demonstrating its potential for
extract by RP-HPLC under 30 min.58 Separation selectivity was multicomponent analysis in biological fluids,97,98 direct
further expanded when using ion pairing reagents in the mobile coupling to HPLC99 and CE100 still remains nontrivial when
phase to increase the retention behavior of polar metabo- acquiring high quality spectra during sample elution. Because of
lites.59−61 Since the introduction of partition column fundamental limitations in the selectivity and sensitivity of
B dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 1. Literature survey of published research articles in metabolomics using a search query with keywords “metabolom* or metabonom*” in title
from 1998 to 2012 via Web of Knowledge 5.8 excluding reviews and abstract proceedings. The pie chart insert depicts the average fraction of articles
using NMR (46%), LC-MS (32%), GC-MS (17%), and CE-MS (5%) over the past five years (2008−2012). Overall, about 54% of all reports utilize
one or more separation methods that is critical for expanding the coverage of the metabolome. There is growing interest in using complementary
separation modes or multidimensional separations, as well as compiling data across multiple analytical platforms for expanded metabolite profiling.

single instrumental platforms, there is growing recognition of model animal organisms,119−121 where changes in urinary
the need for orthogonal separation techniques for expanded metabolic patterns represent a metabolic phenotype or
coverage of the metabolome,20,23,101−105 including integrated “metabotype” that is characteristic for different rodent
total analysis systems based on LC-solid-phase extraction species122 and genetic strains of mice.123 However, only a
(SPE)-NMR-MS.106 modest number of metabolites were quantified and identified in
1.2. A Renaissance in Metabolism: The Advent of urine, primarily abundant osmolytes, amino acids and organic
Metabolomics/Metabonomics acids. Subsequent work by Fiehn et al.124 introduced GC-MS-
based metabolomics for plant functional genomic studies based
Major international efforts at sequencing the whole genomes of on quantitative analysis of 326 distinct metabolites detected in
organisms from the mid-1990s and early 2000s107−109 sparked leaf extracts derived from four genotypes of Aradiposis thaliana.
renewed interest in metabolite profiling as a way to decipher Their work demonstrated that single loci mutants were
genes of unknown function or mutants that possess “silent” distinguished from wild type plants because of their character-
phenotypes.110,111 The initial euphoria derived from high- istic metabolic clustering after principal component analysis
throughput gene sequencing projects soon led to a realization (PCA). In this case, identification of about half of all detected
that gene regulation and interactions on a “systems-level” were molecular features was achieved by comparison of relative
critical for functional understanding of cell activity as a retention times in conjunction with EI-MS spectra from a
whole.112,113 Despite earlier developments in DNA microarray chemical library.124
and proteomic approaches in postgenomic research,114 the first The early adoptation of NMR in metabolomics stems from
published references to “metabolome” did not occur until 1998 its excellent robustness and reproducibility125,126 that is
by Tweeddale et al.115 and Oliver et al.116 that involved convenient for rapid screening of biofluids with minimal
comparative studies of wild-type and gene-deleted mutant sample handling127 as well as direct analysis of nanoliter tissue
strains of bacteria or yeast using Fourier-transformed infrared biopsies.128 Nevertheless, the limited sensitivity of 1H NMR
(FTIR) spectroscopy and 2D TLC separations, respectively. enables characterization of up to 50 metabolites with
The Ferenci group117 later reported the impact of oxidative concentrations above 10 μM in human serum.20 In addition,
stress in bacteria based on measured changes in 14C-glucose- stable isotope tracer experiments129 or chemical derivatiza-
labeled metabolites in Escherichia coli extracts when using 2D tion130 are needed for acquiring spectra for other lower
TLC with phosphorimaging in conjunction with an amino acid abundance nuclei, such as 13C. The use of 2D NMR pulse
analyzer. Although their work revealed increased intracellular sequence methods can increase spectral peak resolution
levels of valine and glutathione in paraquat-treated bacteria however at the cost of reduced sensitivity and longer
relative to controls, a major fraction of metabolites were acquisition times.131 In contrast, direct infusion-MS offers a
unresolved, whereas two spots that decreased upon paraquat sensitive yet high-throughput platform for metabolic phenotyp-
exposure remained unidentified.117 In 1999, Nicholson et al.118 ing132,133 that can measure hundreds of molecular features
introduced “metabonomics” as a way for assessing the without separation, where accurate mass and relative isotope
differential responses of rats exposed to various toxins based abundance measurements allow for determination of elemental
on time-dependent metabolite changes in urine when using 1H composition.134 However, it suffers from poor accuracy and
NMR together with multivariate statistical analysis. Several precision because of matrix-induced signal suppression that
follow-up studies were published in 2000 on the use of NMR- prevents reliable quantification when comparing different
based metabonomics for evaluating drug-induced toxicity in specimens without stable-isotope internal standards.135 More-
C dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

over, a search query for a putative elemental formula from of generation of variance determines overall peak width, which
public databases results in hundreds of potential chemical impacts separation efficiency (plate height, H), resolution and
structures and their isomers that is inadequate for structural peak capacity.162 The mass balance equation can be used to
elucidation of unknown metabolites.136 describe the movement of a solute as a boundary peak through
Thus, there is a clear need to expand metabolome coverage a tube, where both separative and nonseparative processes
with greater sensitivity and selectivity while ensuring robust influence its average linear velocity and rate of band
analytical performance to avoid false discoveries. This has led to broadening.163 Conditions that maximize separative transport
expanding interest in hyphenated separation techniques in while minimizing band dispersion enable higher separation
metabolomics first reported in 2002, such as hydrophilic efficiency that is required for resolving complex sample
interaction liquid chromatography (HILIC)-MS,137 RP-LC- mixtures prior to detection. The advent of comprehensive
MS,138 and CE-MS139 that are ideal for resolution of polar, two-dimensional separations further enhances separation
nonpolar, and ionic classes of metabolites, respectively. Despite performance via direct coupling of two columns in series that
earlier divergence in terminologies used in the literature, have complementary retention mechanisms, since the overall
metabolomics and metabonomics140 are now considered peak capacity of the system is the product of the peak capacity
interchangeable terms and in essence refer to the same for each dimension under ideal conditions.164 In chromatog-
paradigm of comprehensive identification and quantification raphy, separation is driven thermodynamically by the
of “all” metabolites in a given specimen;141 however, this is distribution equilibrium of a solute between a mobile phase
usually only reserved for biologically significant molecular and an immiscible stationary phase, whereas the extent of band
features identified across different samples/groups after broadening is dependent on diffusion and mass transfer
rigorous data preprocessing and multivariate analysis. This kinetics.165 Selectivity in chromatography is determined by
process is distinct from classical metabolite profiling involving differences in retention factor because of favorable changes in
quantitative analysis of targeted compound classes52 or the enthalpy or entropy of a solute upon its reversible transfer
metabolite fingerprinting, where rapid pattern recognition/ from mobile phase to stationary phase. The major modes of
classification of samples is desired without full resolution or separation in chromatography thus exploit specific types of
identification of specific metabolites.142 An “expanded” targeted noncovalent intermolecular interactions to retain solutes based
metabolomics approach143 has also been proposed for on adsorption, partitioning, or ion-exchange mechanisms;
quantitative analysis of hundreds of known standard metabo- nonretained solutes coelute with the mobile phase void volume
lites often across multiple analytical platforms.23,144,145 that defines the hold-up time of a chromatographic system.
However, a unique feature of metabolomics is related to Retention behavior is most readily optimized by changes in the
“hypothesis-free” testing for discovery of unknown or poorly solvent composition of the mobile phase in LC when using
characterized metabolites of biological or clinical significance; covalently modified silica particles,166 whereas temperature
indeed, this “untargeted” approach is critical for novel insights programming is used in GC to enhance solute volatility and
into complex mechanisms of drug/toxin action or disease desorption in wall-coated open tubular capillaries.167 Gradient
pathogenesis on a systemic level.26,146 To date, there have been elution programs are frequently used in order to achieve faster
over 2600 total publications in metabolomics (search query elution times relevant for optimal resolution of complex sample
with metabolom* or metabonom* in title from 1998 to 2012) mixtures; however, sufficient time for re-equilibration of the
that has increased rapidly over the past five years with about column is required for reproducible retention times.168,169 In
54% of all reports utilizing one or more separation techniques general, GC is the method a choice for volatile, nonpolar, and
based on LC (32%), GC (17%), or CE (5%) as depicted in thermally stable metabolites because of its high separation
Figure 1. Thus, NMR still remains a major platform for efficiency, reproducible retention times, and extensive EI-MS
metabolomics/metabonomics despite its lower sensitivity, database library.154 Selectivity in GC is determined primarily by
limited metabolome coverage and higher infrastructure costs. the polarity of the stationary phase since an inert gas (e.g., He)
Because of the lack of a truly universal method for serves to passively transport sample through the capillary,
metabolomics, recent large-scale investigations compile data where nonpolar analytes elute as a function of their volatility
across multiple instrumental platforms or multidimensional and molecular weight. The advent of cross-linked ionic liquid
separation systems.147 Despite several critical reviews dedicated stationary phases for GC-MS allows for unique dual retention
to NMR or MS-based metabolomics,19,148−150 there have been behavior for polar and nonpolar classes of metabolites with
few systematic reviews of separation science151−153 with most higher thermal stability (>350 °C) that has yet to be fully
reports restricted to specific separation platforms based on GC- explored in metabolomics.170,171 Further improvements to
MS,154 LC-MS,155−158 or CE-MS.159,160 Since William’s ultralow bleed stationary phases in conjunction with
pioneering work with 2D TLC for evaluating biochemical commercial two-dimensional gas chromatography-time-of-
individuality46 and Hornings’ classic work on metabolite flight-mass spectrometry (2D GC × GC-TOF-MS) instruments
profiling of human biological fluids by GC-MS,71 new advances with cryogenic modulation172 to minimize off-column peak
in separation science continue to play vital roles in recent broadening173 herald separations with unprecedented peak
metabolomics initiatives. capacity for metabolomics.
Recent developments in LC column technology over the past
1.3. Chromatography: Solute Retention with Improved decade has greatly improved separation efficiency while
Column Technology
expanding selectivity, which is now increasingly dominated by
Modern separation science has evolved rapidly since Mikhail columns packed with fused-core and sub-2 μm porous particles
Tswett’s seminal report on chromatography, which described for ultrafast separations.174 The ratio of the hold-up time to
the extraction, separation and recovery of chloroplasts from column efficiency (i.e., Poppe plot) can be used to assess the
plant extracts.161 A common principle underlying all separation resolving power of a separation system, which is favored with
techniques based on differential solute migration is that the rate columns that possess high porosity and low plate heights while
D dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 2. Comparison of base peak LC-MS chromatograms based on untargeted metabolite profiling of methanol extracts of human plasma
reconstituted in 0.1% formic acid (UPLC or UHPLC) and 10% methanol (HPLC) that highlights the increase in resolution power and sensitivity
along with 3-fold reduction in run time when analyzing the same sample under positive ion mode. LC conditions were (a) 2.1 mm × 100 mm BEH
C18 1.7 μm particle columns heated to 40 °C with a gradient-elution at 350 μL/min or (b) 2.1 mm × 100 mm Aquasil C18 3 μm column with
gradient-elution at 200 μL/min. The three major peaks circled represent common ions detected in both LC-MS conditions as a reference for
comparison of separation performance. Reprinted with permission from ref 187. Copyright 2009 American Chemical Society.

operating under high linear velocities.175 The introduction of greater than 700 bar or 10 000 psi when packed with 1.5 μm
monolithic columns based on a continuous porous silica rod porous particles that result in a 2-fold enhancement in peak
allows for fast separations because of its high porosity with plate efficiency relative to 3 μm particles with a concomitant
heights comparable to conventional 5 μm porous particles that reduction in analysis time under optimum linear flow rates.186
is achieved at much lower back pressures.176−178 However, For instance, UHPLC using a C18 stationary phase with
separation efficiency in monolithic columns is compromised by gradient elution provides higher resolution and improved
Eddy diffusion because of their radial heterogeneity in wide sensitivity relative to conventional HPLC along with a 3-fold
bore columns, which can be improved when using long and shorter run time as shown in Figure 2.187 The latter advantage
narrow capillary formats at low linear velocities.179,180 allows for the implementation of two LC-MS runs optimized
Alternatively, the reduction in particle size within packed for positive and negative ions in less time than a single HPLC
columns is an effective way to enhance separation efficiency181 separation that results in the identification of more than twice
by minimizing variance contributed by Eddy diffusion and mass as many plasma metabolites with greater precision.187 However,
transfer kinetics, which also increases total number of separation performance is ultimately limited by resistive heating
metabolites detected due to their elution as sharper peaks at very high pressures and extra-column band broadening
with higher signal/noise.182 Since pressure drop across packed notably when using micro- or nanoflow UHPLC systems.188
columns is inversely dependent on the square of particle size, Operating columns at elevated temperatures (>40 °C) offers a
specialized equipment is required for operating under optimum simple way for achieving faster separations at lower back
flow rates with particles having average diameter ranging from pressures because of the lower viscosity of the mobile phase
1.0 to 1.7 μm.183−185 For instance, commercial instruments that can enhance peak capacity up to 1000 with plate numbers
based on ultra high pressure or very high pressure liquid in excess of 100 000.189−191 Nevertheless, most siloxane-based
chromatography (UHPLC or vHPLC) generate back pressures materials have limited pH stability (pH ≈ 6−8) at high
E dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 3. Base peak chromatogram highlighting the chemical diversity of semipolar phytochemicals derived from methanol extracts of tomato
(Daniela) for comprehensive metabolite profiling of different tomato cultivars when using UHPLC-TOF-MS with an optimized solvent gradient
under negative ion mode detection. Over 135 different compounds, including 21 uniques phytochemicals reported for the first time in tomato fruit
were assigned based on their accurate mass, retention time, as well as UV absorbance and MS/MS spectral matching. Separations were performed on
a C18 analytical column (4.6 × 150 mm, 1.8 μm particle size) at 37 °C. Reprinted with permission from ref 206. Copyright 2010 Elsevier, Ltd.

temperatures, whereas residual silanol residues can adsorb basic silica-based octadecylsilane (ODS, C18) bonded-phase col-
solutes resulting in band broadening and peak asymmetry.192 umns in conjunction with an aqueous mobile phase with
However, bridged ethylene hybrid (BEH) modified silica methanol or acetonitrile as organic modifiers.200 Solvophobic
stationary phases reduce the number of residual silanol moieties theory201 and quantitative structure-retention relationships202
that contribute to undesirable ion-exchange processes while have been developed to accurately model the retention
also improving the pH/thermal stability of the column.193 The behavior of nonpolar and semipolar solutes based on their
recent development of “superficially porous” sub-3 μm fused- physicochemical properties when using type-B alkylsilica
core particles provide highly efficient columns without high stationary phases. Indeed, five different types of intermolecular
back pressures that achieve similar separation performance of interactions fully account for solute retention selectivity in RP-
sub-2 μm porous particles with standard HPLC instrumenta- LC, namely hydrophobic, steric hindrance, hydrogen bonding
tion.194 In this case, a 1.7 μm solid silica particle is used as acceptor or donor and Coulombic interactions (e.g., cation-
support for growing an outer thin porous shell (0.5 μm) exchange capacity).203,204 Recent computer simulations of
resulting in greater mass transfer kinetics even at high linear retention mechanisms in RP-LC suggest multiple sorption
flow velocities.195 sites involving both adsorption (at interface) and partition
Since the metabolome is composed of a diverse array of (within bonded phase) interactions at the molecular level that
compounds with different physicochemical properties, no single is solute dependent.205 Indeed, the extensive use of RP-LC
retention mechanism is adequate to resolve complex sample stems from its reproducible and predictable retention times,
mixtures that vary widely in their polarity, charge, and stability. wide applicability to various classes of metabolites/xenobiotics
In most cases, orthogonal separation modes are used in parallel and mobile phase compatibility for direct coupling to ESI-MS.
to analyze the same sample as a way to expand metabolome Figure 3 highlights the broad range of semipolar primary (e.g.,
coverage for both polar and nonpolar metabolites,196 such as organic acids) and secondary metabolites (e.g., flavonoids)
HILIC and RP-LC.102,197 Alternatively, there is growing from methanol extracts of tomato when using RP-LC that can
interest in developing “mixed-mode” retention mechanisms resolve over 135 different compounds when using an optimized
based on dual 198 or trimodal stationary phases within a single solvent gradient elution program.206 In this case, UHPLC-
column that exploit RP, cation-exchange or anion-exchange TOF-MS with negative ion mode enables differentiation of
retention mechanisms;199 however, method optimization is three major cultivars of tomatoes based on their characteristic
challenging given the large number of experimental variables profile of phytochemicals, notably biological active fractions of
that can impact separation performance. To date, RP-LC is by phenolic compounds. However, a major limitation of RP-LC is
far the most widely used mode of chromatography in the poor retention of hydrophilic/ionic metabolites due to their
metabolomic studies reported to date (>90%) when using coelution within the void volume. In this case, chemical
F dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 4. Comparison of the separation performance of HILIC-MS with two different types of columns using acidic (formic acid, FA) and basic
(ammonium acetate/carbonate, AA/AC) gradient elution conditions for model polar/ionic metabolites unretained by RP-LC. The two zwitter-ionic
HILIC columns were found to offer better separation performance for urinary metabolites and their isomers in comparison to ANP (type-C silica
hydride) and RP-LC (C18). The three LC-MS columns used for polar metabolite separations are (a) ZIC-HILIC, 150 × 4.6 mm 5 μm Merck; (b)
ZIC-pHILIC, 150 × 4.6 mm 5 μm Merck; and (c) CDH, cogent diamond hydride, 150 × 4.6 mm 4 μm MicroSolv. Because of their multivalent
charge status that is pH dependent, deleterious ionic interactions occur with both stationary phases that contribute to peak tailing and poor
separation efficiency notably for sugar phosphates, tricarboxylic acids, basic amino acids, and polyamines. Reprinted with permission from ref 222.
Copyright 2012 American Chemical Society.

derivatization can be implemented as a strategy to improve the HILIC represents a complementary normal-phase LC
retention of hydrophilic metabolites (e.g., amino acids, amines) separation mode for resolving polar/ionic metabolites and
when using RP-LC that also enhances their ionization efficiency their isomers that are poorly retained by RP-LC when using
with lower detection limits.207,208 A simple “label-free” high amounts of organic modifiers (e.g., acetonitrile) containing
approach to enhance the retention of polar/charged solutes 10−40% water.213 Under these conditions, retention of
in RP-LC is the use of ion pairing reagents61 in the mobile hydrophilic solutes occurs via partitioning onto a hydrated
phase, which also provides pH control while improving peak surface of the stationary phase relative to an organic-enriched
shape caused by unwanted silica interactions. However, ion mobile phase (i.e., biphasic system) with gradient elution
performed by increasing the water content. In contrast to
pairing RP-LC (and ion-exchange LC) is not readily compatible
conventional normal phase-LC, HILIC is more compatible for
with ESI-MS due to signal suppression and/or ion source
polar metabolite analysis when coupled to ESI-MS due to the
contamination by involatile electrolytes in the mobile phase, high content of organic modifier in the mobile phase that leads
such as trifluoroacetate under negative ion mode.209−211 For to improved ionization efficiency and solvent evaporation.214 A
this reason, the application of ion pair RP-LC24,211 for plethora of polar stationary phases have been developed for
separation of polar metabolites in biological samples is not HILIC ranging from unmodified silica to bonded-phase
widely used in metabolomics (<1% of total LC-MS papers) particles with nonionic (e.g., polyol), ionic (e.g., amine), or
since separations are constrained by the use of low zwitter-ionic (e.g., sulfoalkylbetaine) functional groups, which
concentrations of ion pair reagents together with nanoflow contribute to complex retention mechanisms distinct for each
ESI interfaces for adequate sensitivity.212 column type.215,216 Moreover, HILIC columns exhibit
G dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

increased retention time drifts and require longer re- separation optimization, such as Joule heating and electro-
equilibration times relative to RP-LC, while prone to poor kinetic dispersion.234 In the case of CEC, various approaches
separation performance for some classes of ionic metabolites, have been used to introduce RP partitioning during solute
such as estrogen conjugates217 and nucleotides.218 Alternatively, electromigration, including open tubular, monolithic, and
aqueous normal phase (ANP)-LC based on silica hydride-based porous particle column formats.235 For instance, ultrahigh
stationary phases (i.e., type-C silica columns) offer greater separation efficiency can be realized in CEC when using
stability since hydride groups replace the majority of surface- uniformly packed submicrometer silica colloidal crystals packed
active silanol moieties without end-capping, where hydrophilic in narrow bore capillaries without massive back-pressure or
partitioning occurs via a gradient elution program over a wide deleterious resistive heating since fluid transport is driven by
range of water content.219,220 Recent work has demonstrated the EOF.236 However, monolithic columns are generally
that ANP-LC-MS offers good retention time repeatability (CV favored in CEC since they do not require complicated packing
< 1%) with a similar total number of detectable features in procedures or the use of frits to restrict electromigration of
human urine as compared to RP-LC that would have otherwise porous particles since sol−gel formation occurs in situ with
eluted in the void volume.221 Recently, a systematic comparison covalent attachment of a porous silica rod onto the capillary
of the separation performance of RP (C18), ANP (silica surface.237 As separation efficiency in CE is directly dependent
hydride), and HILIC (zwitter-ionic) MS-based analysis of on the magnitude of the electric field, effective heat dissipation
human urine demonstrated that HILIC enabled detection of is required when using commercial instruments that permit the
the greatest fraction of significant features (70%) relative to application of 30 kV, whereas voltages up to 120 kV have been
ANP (53%) and RP (25%) with 12% of total features detected reported in CE systems with electrical shielding generating
across all three platforms; however, both HILIC and ANP plate numbers up to 6 million.238 Separation optimization in
required the application of two different elution conditions CE is dependent primarily on aqueous buffer conditions (e.g.,
under acidic (pH 2.8) and alkaline (pH 9.2) conditions to pH, electrolyte, ionic strength, organic modifier),239 which can
expand metabolome coverage as a result of the severe peak be performed in silico for rapid separation optimization based
skewing reported for several classes of ionic metabolites as on intrinsic properties of solutes and the buffer system. Unlike
shown in Figure 4.222 Despite these drawbacks, HILIC remains LC, separations in CE are typically performed isocratically
the most common separation mode used in LC-MS for polar using an aqueous buffer system, however nonaqueous solutions
metabolites, which is increasingly being used together with RP- (e.g., acetonitrile) can also be used as a way to enhance
LC for comprehensive metabolomic studies. solubility of nonpolar metabolites, as well as alter selectivity and
improve compatibility to ESI-MS.240 The latter detector
1.4. Electrophoresis: Ion Mobility with Tunable Selectivity
in Free Solution interface necessarily restricts the use of MEKC since involatile
micelles in the BGE contribute to ion suppression unless using
Separation in electrophoresis is driven electrokinetically under partial filling methods241 or alternative ion sources that better
an applied voltage based on differences in the electrophoretic tolerate matrix effects, such as atmospheric photoionization
mobility of an ion in free solution.223 The ideal state (limiting) (APPI).242 A unique way to enhance resolution and separation
mobility of an ion and its thermodynamic weak acid efficiency in CE is the use of “flow counterbalance” mode
dissociation constant (pKa) determine the effective charge operation via application of a pressure-induced243 or EOF
density of a weakly ionic species at a given buffer pH counterflow244 to balance or retard electrokinetic migration in
condition.224 Hybrid separation techniques that utilize both the capillary, which can be used to resolve minor differences in
electric field (i.e., mobility) and equilibria (i.e., thermodynamic) solute mobility, including enantiomers and isotopomers.
processes for separative transport, such as capillary electro- There is growing interest in CE-based separations in the
chromatography (CEC),83,225 MEKC,226 and dynamic com- pharmaceutical industry that demands robust analytical
plexation CE227 offer expanded selectivity for a wide range of performance,245 notably when performing chiral separations,
analytes, including neutral/nonpolar, and polar/ionic metabo- glycan analysis and quality control of biopharmaceuticals.
lites. For instance, resolution of neutral polyol enantiomers in Indeed, capillary gel electrophoresis was a key instrumental
CE when using charged arylboronic acids as selective sugar- platform in multiplexed gene sequencers that contributed to the
binding additives in the BGE can be achieved electrokinetically early completion of the Human Genome Project.246 However,
based on differences in their complex mobility even in cases there is a common misconception regarding the suitability of
when their binding constants are similar.228,229 When using CE as a robust platform in metabolomics notably in the context
soluble additives in CE, the retention factor of a solute is of its poor migration time reproducibility. Since bulk fluid
defined as the product of partition coefficient (or binding transport is distinct from pressure-driven flow in chromatog-
constant) and additive concentration,230 which allows for raphy, appropriate software tools are needed for data
independent control of a moving yet discrete “pseudo- processing to correct for migration time drifts in CE, which
stationary phase”231 Furthermore, multiple additives with is dependent on both the magnitude and direction of the
distinct binding mechanisms and/or charge states can be electrophoretic mobility of ion and the EOF.247 Indeed, the
used simultaneously for high resolution chiral separations, such major advantages of the EOF arise from its near flat-like profile
as anionic micelles and neutral cyclodextrins.232 Indeed, CE that minimizes solute axial diffusion during electromigration
offers a versatile microseparation format for analysis of volume- while serving as a natural electrokinetic pumping mechanism.
restricted samples with minimal sample pretreatment, including However, since the EOF is dependent on the surface properties
single cells.233 Because of the lack of pressure-driven laminar of the capillary and the electrolyte properties of the BGE,
flow, Eddy diffusion and mass transfer kinetics prevalent in absolute migration times are more variable in CE relative to
packed columns, separation efficiency in CE is typically an retention times in RP-LC or GC that are regulated by an
order of magnitude greater than conventional HPLC; however external peristaltic pump or pressure regulator;248 this issue can
other band broadening sources need to be minimized during be readily addressed by the use of a neutral EOF marker during
H dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 5. Overlay of extracted ion electropherograms depicting the separation of 20 different amino acids and acylcarnitines from dried blood spot
extracts that serve as diagnostic markers for in-born errors of metabolism in newborn screening programs. Note that metabolites of widely different
polarity can be analyzed simultaneously by CE-ESI-MS under a single elution condition while providing resolution of relevant isomeric and isobaric
ions (*), including cationic amino acids (Lys) and long-chain acylcarnitines (C16). Conditions used were 1 M formic acid, 15% v acetonitrile, pH 1.8
as BGE with a bare fused-silica capillary and a coaxial sheath liquid interface under positive ion mode. Reprinted with permission from ref 264.
Copyright 2009 American Chemical Society.

separation, which allows for precise determination of electro- Figure 5 highlights the selectivity of CE-MS for resolving a
phoretic mobility (CV < 1%) when using commercial wide range of polar metabolites and their isomers under a single
instruments with thermostatic control of the capillary. Indeed, elution condition, including polar amino acids and long-chain
mobility shift measurements are widely used in CE for reliable acylcarnitines that represent clinically relevant biomarkers for
determination of fundamental thermodynamic parameters in early detection of in-born errors of metabolism.264 In this case,
free solution, including pKa of weakly ionic drugs,249 the addition of acetonitrile as organic modifier in the BGE was
dissociation constants for affinity interactions250 and free needed to ensure adequate solubilization of surface-active
energy changes for protein unfolding.251 Alternatively, an acylcarnitines without compromising resolution of amino acid
internal standard can be used to determine relative migration stereoisomers, such as three Leu stereoisomers. However,
times,252 a migration time normalization can be performed caution is warranted when adopting CE-MS protocols based on
using two or more internals standards253 or dynamically coated coated capillaries under reversed polarity, where the sprayer
capillaries can be used to stabilize or suppress the EOF with serves as the anode to reverse the EOF.265 For instance, a
good interday precision (CV < 2%) for migration times.254 recent cross-platform comparison of GC-MS, LC-MS (i.e., ion
Recently, a temperature-corrected mobility scale was intro- pair-RP/HILIC), and CE-MS for analysis of polar metabolites
duced for data transformation to improve the long-term of central energy metabolism concluded that CE-MS lacked
reproducibility of CE-MS that accounts for multiple sources overall robustness.266 Regrettably, this result was based on a
of migration time variance, including EOF, Joule heating, flawed CE-MS method for anions that was subsequently
thermostatic control and suction effects by the nebulizer gas of discovered to cause premature column clogging at the capillary
the ion source.255 Although CE with laser-induced fluorescence outlet265 since its first introduction in 2002.139 Thus, similar to
detection offers ultrasensitive detection of trace levels of several major GC-MS,267 LC-MS,268 and NMR269 initiatives,
analytes with a dynamic range over 9 orders of magnitude,256 standardized CE-MS protocols require validation across
CE-MS suffers from poor concentration sensitivity relative to multiple laboratories to properly evaluate method robustness.
LC-MS because of the small sample volume injections (∼10 To date, metabolic profiling of cations by CE-ESI-MS under
nL) and postcapillary dilution effects when using a coaxial strongly acidic conditions with positive ion mode270 is a reliable
sheath liquid interface.257 Various CE-MS sheathless designs format consistently adopted by several research groups.271
have been developed to increase concentration sensitivity over However, further development is needed in CE-MS for anionic
the past twenty years;258 however, few configurations have metabolites under alkaline conditions that offers excellent
demonstrated sufficient robustness for routine analysis.259,260 In selectivity for polyprotic acids/strongly ionic metabolites that
general, CE-MS is ideal for the analysis of weakly/strongly ionic are not well served by LC separation modes based on HILIC or
metabolites that constitute the major fraction (>60%) of known ANP.262 Figure 6 demonstrates the excellent selectivity of CE-
metabolites or degradation products in primary metabolism,261 MS for a wide variety of anionic metabolites with negative ion
as well as secondary metabolites that are weakly retained or mode, including nucleotides, organic acids, sugar phosphates,
have poor separation efficiency in RP-LC or HILIC.262 In and redox-active cofactors.265 In this case, a platinum needle
comparison to GC-MS, metabolite profiling of cell extracts can was required to extend the capillary lifetime when operating CE
be performed by CE-MS with minimal sample pretreatment under reversed polarity conditions.272 Recent developments in
that increases sample throughput and overall reliability.263 low flow sprayer designs for CE-MS with improved
I dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 6. Anionic metabolite analysis by CE-MS under negative ion mode using a platinum needle with a COSMO(+) cationic polymer-coated
capillary and 50 mM ammonium acetate, pH 8.5 as BGE. Selected extracted electropherograms for a standard mixture of anionic metabolites (10
μM) relevant to glycolysis, pentose phosphate, and the TCA pathways in rat liver extracts, including sugar phosphates, organic acids, nucleotides, and
redox cofactors. The use of a platinum electrode instead of a conventional stainless steel sprayer improves sensitivity, migration time precision and
overall robustness, while avoiding capillary outlet clogging at the anode under reversed polarity conditions. Reprinted with permission from ref 265.
Copyright 2009 American Chemical Society.

Figure 7. Improved separation performance in 2D GC × GC-TOF-MS when using a polar/nonpolar column configuration to enhance peak capacity
(>9000) with greater loadability and separation efficiency, where a wider bore and thicker wall coated nonpolar stationary phase is used as the
column in the second dimension. Full scan chromatograms of methanol extracted fetal bovine serum samples with conventional setup A (2D 1m ×
0.1 mm, 0.1 μm, BPX5 column) and high loadability setup C (2D 2m × 0.32 mm, 0.25 μm, BPX5 column) where the first column is polar consisting
of 30 m × 0.25 mm, 0.25 μm, BPX50. Enlargements (100 s × 1.5 s) highlight better resolution of trimethylsilylated amino acids in the presence of
excess urea from plasma extracts when using the loadable column configuration, namely, serine (1), threonine (2), and urea (3). Reprinted with
permission from ref 287. Copyright 2008 Elsevier, Ltd.

sensitivity260,273,274 require further evaluation under negative the advent of two-dimensional gel electrophoresis as the
ion mode,272,275−277 which is prone to corona discharge with preeminent preparative tool in biochemistry and proteomics
higher background noise and poor spray stability.278 Despite research,114,279,280 there have been few precedents of online
J dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

two-dimensional CE × CE281,282 or LC × CE283 systems for eoisomers262,296 and enantiomers297,298 For instance, stereo-
metabolomics without off-line fractionation prior to ESI-MS.284 selective separations are critical for second-tier testing of
genetic diseases with improved sensitivity and selectivity, such
2. NEW ADVANCES IN SEPARATION SCIENCE FOR as plasma allo-Ile that is a pathognomonic marker of maple
METABOLOMICS: MORE THAN SEPARATION? syrup urine disease.299 Differential metabolic profiling of
Separation science plays several critical roles in metabolomics cellular responses to racemic drugs or single enantiomers also
for reliable quantification and improved identification of highlights the importance of drug stereochemistry on biological
metabolites in complex sample mixtures.285 Separation activity that can be resolved by appropriate separation
efficiency (plate height, H, or plate number, N) and peak techniques.300 In general, chiral resolution is realized by
capacity are two important parameters for determining the precolumn chemical derivatization to form diastereomeric
performance of a separation in terms of the total number of adducts that can be resolved by differences in their retention
unique compounds that are baseline resolved prior to detection. factor and/or mobility or the use of chiral-based stationary
Higher peak capacity within a wider separation window reduces phases or chiral additives in the BGE/mobile phase.301 Indeed,
solute coelution that can complicate relative quantification or there are few reported studies to date that have performed
spectral matching for unknown identification. For instance, the enantioselective metabolite profiling for resolution of optically
use of a shallow gradient elution program in RP-UHPLC active classes of metabolites. For example, chiral separations
expands peak capacity up to 1000 at the cost of longer analysis were used to improve discrimination of liver cirrhosis
times that exceed one hour when including column re- patients298 and bioequivalence of transgenic crops302 when
equilibration.190 Alternatively, the application of orthogonal measuring the enantiomeric excess ratio of D-amino acids.
RP and HILIC separation modes in parallel using optimized Another important function of separations notably when using
solvent elution programs (<15 min) enables a wider range of ESI-MS is improved detection of low abundance metabolites
compounds that can be detected by ESI-MS for global that is compromised by coelution of abundant co-ions and
metabolic profiling.286 Moreover, optimized 2D GC × GC/ involatile salts in a biological sample that lower sensitivity.209
TOF-MS using a polar and nonpolar column connected in This also impacts the quantitative performance of MS-based
series allows for a major boost in peak capacity to over 9000 metabolomics when measuring relative signal responses with
resolved features within a short total analysis time.287 Figure 7 acceptable accuracy that is independent of sample matrix. Given
highlights that the use of a wider bore/thicker nonpolar the wide disparity in solute ionization efficiency in ESI-MS,
stationary phase film as the second dimension column enhances reliable quantification of metabolites in complex biological
separation performance by increasing sample loadability that is samples is only feasible if separations are optimized to avoid
important when quantifying low abundance metabolites in coelution that is critical in metabolomics since purified
complex biological samples. However, because of the larger reference standards or stable-isotope internal standards are
retention time variability notably for HILIC, CE, and 2D GC × often unavailable.271 For instance, postcolumn infusions
GC separations,288 additional time resources are needed for experiments of a calibrator ion can be used in LC-MS to
data alignment, peak picking, and normalization that monitor potential ion suppression or enhancement effects
complicates data preprocessing and statistical analyses in during elution,303 where ESI is generally found to be more
metabolomics.289,290 Therefore, there is a fundamental trade- susceptible to signal suppression relative to atmospheric
off between comprehensive metabolite profiling when using pressure chemical ionization (APCI) or APPI even when
hyphenated separation platforms for deeper metabolome using UHPLC methods.304,305 One way to improve method
coverage and improved data quality in comparison to high robustness and sensitivity in metabolomics is to optimize
throughput screening approaches based on direct infusion sample workup in conjunction with separation conditions, such
(DI)-ESI-MS,291 desorption electrospray ionization (DESI)- as SPE-LC-MS.306 For instance, an optimized mixed-mode
MS,292 or 1H-NMR.293 For instance, a comparison of the solid-phase microextraction (SPME) procedure was used to
performance of DI-FT-ICR-MS relative to LC-ESI-TOF-MS enhance separation performance for analysis of both hydro-
for the discovery of putative serum markers of kidney cancer phobic and hydrophilic metabolites with reduced ionization
revealed that twice the number of significant molecular features suppression in LC-MS.307 However, since extraction is not
were detected by LC-MS; however, this required about a 20- exhaustive and depends on sorbent material, the number of
fold increase in total analysis time.294 Alternatively, a two-stage compounds detectable by LC-MS is reduced when compared to
strategy can be adapted based on rapid screening by DI-ESI-MS solvent-based extraction methods that denature protein. Also,
followed by confirmatory testing by hyphenated separation in most cases SPE/SPME extraction steps in metabolomics are
techniques that possess greater selectivity for confirmatory performed off-line prior to analysis that require long extraction
testing similar to expanded newborn screening initiatives.264 or desorption times that prevents absolute quantification since
However, this strategy has yet to be adapted in metabolomics recovery is highly solute dependent.308 Other sample cleanup
research as it risks false-positives due to a plethora of artifact methods offer better extraction efficiency but still are limited by
signals when using DI-ESI-MS as the primary screen, as well as complicated sample handling that contributes to higher process
false negatives due to matrix-induced ion suppression effects.295 variability, including single-drop microextraction309 and stir bar
For this reason, a compromise between sample throughput, exhaustive extraction.310 Alternatively, online sample precon-
metabolome coverage and peak capacity of a separation must centration together with desalting can be integrated during
be reached without sacrificing data quality when performing separation prior to ESI-MS when using a discontinuous
large-scale metabolomics investigations for biomarker discov- electrolyte system in CE-MS as highlighted in Figure 8.
ery.286 Overall, about a 50-fold enhancement in concentration
One major contribution of separations is the ability to resolve sensitivity can be realized in CE-MS for cationic metabolites
isobaric compounds since they are not readily distinguished by (e.g., Trp) under positive ion mode without ionization
high resolution MS or MS/MS, including diaster- suppression or extra-column band broadening.311 Although
K dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

feature, namely its accurate mass (m/z) and (corrected)


retention time, migration time, or retention index.314 Although
ultrahigh resolution MS instruments (e.g., FT-ICR-MS, Orbi-
trap etc.) may correctly assign a putative empirical formula, this
is subject to a high false-positive rate when adequate confidence
intervals are not properly used to ensure annotation
quality.315−317 Moreover, the confidence of annotation
decreases with lower signal/noise ratio (e.g., S/N < 20),318
such that low abundance ions typically remain uncharacterized
due to their poor mass accuracy (>10 ppm) and undetectable
isotope abundance patterns that are needed for discrimination
among numerous molecular formula.134,136,318 Even in the case
of a defined elemental formula, such as phenylalanine
(C9H11NO2), there are over 6 million putative chemical
structures as determined by Molgen Online187 with 2340
known structures with the same accurate mass archived in
PubChem (search query performed on 3/2013). However, a
greater chance for correct spectral assignment is achieved after
preselection of “authentic” molecular features with robust data
filtering319 or subtracting a process blank to all samples to
remove confounding background ions.187 As GC-EI-MS
provides reproducible mass spectra that are largely independent
of matrix-effects and vendor-specific MS instruments, this
allows for the development of universal database libraries for
mass spectral searches, such as NIST11.320 In most cases,
customized “in-house” mass spectral libraries are required for
improving the confidence of peak assignments in LC-MS
metabolomic studies because of the greater variability in ion
fragmentation processes when using ESI-MS/MS.187,321,322
Ultimately, structural elucidation from a molecular formula
requires additional data derived from collisional-induced
Figure 8. Time-resolved electropherograms and computer simulations dissociation (MS/MS), multistage MS (MSn) or EI-MS spectra
(as insets) highlighting the dynamics of electrokinetic focusing of a that are consistent with reference spectral libraries acquired
long/dilute sample plug of Trp in CE, where the capillary functions as under standardized conditions; however, the quality of mass
a preconcentrator, desalter and separator prior to ESI-MS. A long spectral matching is highly dependent on the specific database
sample plug (12.5 cm) of dilute Trp prepared in 200 mM ammonium that can result in different search query results (e.g., PubChem,
acetate, pH 7.0 with 15 mM NaCl is placed at increasing distances HMDB, Massbank, METLIN, etc.).21,323,324 Thus, one of the
from the capillary outlet (a) 7.5, (b) 25, and (c) 40 cm after it is main functions of separations is the resolution of multi-
injected within a discontinuous electrolyte system that is filled with 1
M formic acid, pH 1.8 as the BGE. About a 50-fold enhancement in
component mixtures prior to ionization in order facilitate mass
concentration sensitivity is realized without signal suppression. Analyte spectral matching with pure compound reference libraries.
peak numbering corresponds to concentration profiles of (1) Trp Indeed, it was long recognized that the inclusion of both mass
(experimental), (1*) Trp (predicted), (2) NH4+, (3) Na+, and (4) pH. spectral and retention time index criteria within customized
Reprinted with permission from ref 311. Copyright 2007 American GC-MS databases allows for greater confidence in identifying
Chemical Society. biologically significant molecular features.325 However, without
reference spectra or purified chemical standards, de novo
ion mobility-ESI-MS provides additional selectivity to resolve structural elucidation remains tenuous recent advances of in
isomeric metabolites because of the characteristic mobility drift silico mass fragmentation for comparing MS/MS spectra of
time of an ion, it does not offer preconcentration and/or unknown metabolites.326−330
desalting benefits similar to CE since electrophoretic The advent of computer-assisted design tools for separation
separations occur in the gas-phase after ionization.312 In optimization in GC/LC 202,331 and CE332,333 offers an
addition to resolving isobars/isomers, reducing matrix interfer- orthogonal approach for metabolite identification based on
ences that modulate signal response, boosting concentration accurate prediction of retention or migration times in silico. CE
sensitivity, as well as improving quantitative reliability, high is particularly amenable for predicting the mobility of polar
resolution separations also improve mass spectral matching to metabolites based on intrinsic physicochemical parameters of
reference libraries while providing qualitative information for an ion311 since separations are performed under defined
metabolite identification that is complementary to MS. isocratic conditions using a homogeneous aqueous buffer.
Separations can also be used in a preparative context to Indeed, fundamental electrokinetic, thermodynamic and
preconcentrate and fractionate eluting components from molecular properties of an ion not only permit modeling of
complex sample mixtures for subsequent off-line structural solute migration behavior in CE, but can also be used for virtual
elucidation by NMR.313 quantification (i.e., relative response factor) of polar metabo-
In most cases, the majority of detectable signals in MS-based lites by ESI-MS given their wide disparity in ionization
metabolomics studies are unknown, where a minimum of two efficiency.271 For instance, Sugimoto et al.253 demonstrated
independent parameters are necessary to describe a molecular that molecular descriptors can accurately predict migration
L dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

times of cationic metabolites in CE-TOF-MS when using it also enhances the performance of nominal mass resolution
support vector regression, which increases the assigned quadrupole mass filters, such that a larger fraction of
molecular features for urinary metabolites from 39% (i.e., metabolites can be identified relative to accurate mass alone
standard mass spectral library match alone) to over 52% when provided that retention time accuracy is within 0.25%.337,338 A
using a m/z and migration time tolerance of 15 ppm and 1.5 metabolomics workflow for identification of unknown metab-
min, respectively. Similar approaches have been reported for olites was recently introduced for GC-EI-TOF-MS based on
prediction of retention times when using RP-LC-MS,334,335 unbiased query searches of known chemical structures
HILIC-MS,336 and GC-MS136 based on in silico molecular deposited in the PubChem database,136 where in silico retention
descriptors to improve metabolite identification, which can index and mass spectra matching allowed for correct structure
reduce false negative assignments by about 40% as compared to assignment in 73% of cases among putative compounds within
accurate mass alone, including isomer discrimination.336 the top 5 hits. Moreover, the integration of both Kovats
However, model robustness is dependent on the selection of retention index estimation with EI-MS fragmentation pre-
an appropriate training/validation set of various classes of diction software filtered out 89% of all potential isomeric
metabolites while implementing measures to minimize structures not feasible by elemental formula alone. However,
retention time variance that can contribute to bias exceeding retention index predictions in GC using NIST MS software339
35% in HILIC.336 Another major challenge in predictive is not sufficiently robust for polar metabolites labeled with
modeling of retention times in chromatography is the nonlinear multiple trimethylsilyl groups since steric effects are not
change in isocratic retention factor that is solute dependent and included in retention modeling,340 which limits its applicability
not fully corrected by retention indexing or time warping for de novo structural elucidation of unknown molecular
algorithms, which assume similar retention behavior of features.136 Similarly, a dual in silico retention index and mass
neighboring eluting internal standards.337 This premise spectra model was reported for LC-MS to rank lead candidates
from PubChem search queries for subsequent screening using
multiple filters, including accurate mass, retention index,
precursor ion stability and CID fragmentation spectral
matching.341,342 New computational approaches for structural
elucidation of unknown metabolites are urgently needed for
ranking lead candidates based on a consensus scoring function
since a majority of annotated features remain largely
unidentified in metabolomics.343
2.1. The Volatile Metabolome by GC-MS
GC-MS represents an established separation platform in
metabolomics that is the method of choice for analysis of
volatile/nonpolar metabolites; however chemical derivatization
is required for polar metabolites to improve their volatility,
thermal stability, detectability, and retention while minimizing
Figure 9. Predicted RP-LC retention time and exact mass of 7307 deleterious column adsorption. Indeed, metabolome coverage
compounds from the KEGG metabolite database, which highlights by GC-MS is limited by the intrinsic thermal stability of the
their orthogonal relationship for metabolite identification. Simulated stationary phase and metabolite or their trimethylsilyl
compound identification revealed that a 2-fold increase in correct derivative344 that also excludes high molecular weight
structural assignment (70% of total compounds, P = 0.01) is realized
compounds with low vapor pressure unless using high
when accurate mass (1 ppm, 1 × 106 mass resolution) is supplemented
with accurate retention time (0.25% error). However, a major temperature GC-based columns.345 Plant sciences still domi-
improvement in compound identification is also realized when nate the use of GC-MS as a central platform in metabolomics
combining nominal mass resolution with predicted retention times since its first introduction in 2000124 with growing interest in
from only 1% to about 44% of total compounds, which is greater than food science, microbiology and biomedical applications.343 A
FT-ICR-MS alone. Reprinted with permission from ref 337. Copyright survey of GC-MS and 2D GC × GC-MS metabolomic studies
2011 Elsevier Ltd. recently published (Supporting Information Table S1) indicate
that overall methodologies are fairly standardized and differ
fundamentally prevents accurate modeling of analyte retention primarily in terms of sample pretreatment protocol as related to
behavior in LC-MS due to the wide range of solvent sample extraction and chemical derivatization for specimens
compositions or gradient elution programs used in separations ranging from biofluids,346,347 cells,348,349 tissues,350,351 and
for metabolomics. Recently, Boswell et al.337,338 introduced a whole organisms.352,353 In general, most methods use narrow
novel retention projection method for compound identification bore fused-silica capillaries with wall-coated 5%(diphenyl)-
in LC-MS based on their measured isocratic retention versus polydimethylsiloxane (PDMS) as a generic nonpolar stationary
eluent composition relationships that can tolerate different phase and He as an inert gas with elution gradient programs
solvent gradients, flow rates and instrument platforms. The performed with a maximum temperature under 320 °C within
inclusion of predicted retention times with good accuracy 50 min to reduce column bleed; however, some reports also
(<1%) substantially improves compound identification since utilize more polar stationary phases for improved selectivity in
retention times in RP-LC are highly orthogonal to accurate GC-MS and/or higher peak capacity in 2D GC × GC-MS, such
mass337 as depicted in Figure 9 for over 7,307 model as 50%(diphenyl)-PDMS354 and 14%(cyanopropylphenyl)-
compounds derived from the KEGG database. Interestingly, PDMS355 bonded phase columns. If sensitivity is an issue,
not only does retention time data with accurate mass double then splitless injection of the sample is performed, but it can
the total number of compounds identified by FT-ICR-MS, but result in column overloading, poor separation efficiency and
M dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

reduced column lifespan when exposed to excess residual and/or incomplete derivatives for polar/ionic metabolites that
silylating reagents. For these reasons, splitless injection is often vary significantly in their reactivity and stability, which
used in GC at a high temperature to induce flash evaporation, complicates absolute quantification with reduced sensitivity.364
where the initial temperature of oven is set low to recondense Excess silyl reagent and high temperatures are required to
and preconcentrate the sample zone at the column head; enhance reaction kinetics under anhydrous conditions that
however, this risks in situ thermal degradation of labile require stringent quality control measures during all stages of
metabolites in the injector, notably partially silylated deriva- sample handling, including stable-isotope internal standards,
tives. A similar type of analyte focusing mechanism is applied in blank controls and frequent injector liner replacements.154,354
2D GC × GC to reduce extra-column band broadening and Sample cleanup protocols to remove residual silylating reagents
enhance sensitivity during thermal modulation of eluting bands after derivatization using silica are ineffective due to
at the end of the first column with their subsequent transfer coextraction of most derivatized metabolites from sample.365
into a much shorter second column having complementary Alternatively, bulkier reagents can be used to impart greater
selectivity.173 Despite the advent of commercial 2D GC × GC- stability, such as tert-butyl(dimethylsilyl)trifluoroacetamide
MS instrumentation, time-consuming data processing and peak (MTBSTFA) derivatives that are 104 times more hydrolytically
picking procedures still hamper large-scale metabolomics stable than trimethylsilyl protecting groups.366 Reaction
studies.356 Clean injection port liners are critical in GC-MS conditions and GC-MS protocols can be systemically optimized
because of surface adsorption and carry-over contamination by when using a design of experiments to elucidate major factors
sample matrix, which can also potentially catalyze chemical impacting derivatization efficiency for rigorous method
reactions at elevated temperatures leading to sampling bias and validation.367 In addition, stable isotope labeling using
unanticipated artifacts.357 In most cases, EI-MS (70 eV) with MTBSTFA together with its heavier deuterated (d6) analog
positive ion mode is used as a universal ion source preferably offers a novel approach for differential metabolomics that
with TOF-MS, which allows for mass spectral matching with corrects for bias and the high variability associated with
known compounds within customized libraries or public silylation.368 In this case, relative quantification of metabolites
databases. Heuristic filtering of accurate mass measurements in human serum extracts is performed via ratiometric
(<3 ppm) with good isotope precision (<5%) of a molecular measurements of each encoded metabolite isotopomer pair.
ion (e.g., radical cation) in EI-MS allows for correct elemental Similarly, stable isotope labeling using a mixture of light and
formula assignment in most cases,315 whereas a tentative heavy (d9 ) isotopomers of MSTFA improves relative
chemical structure can be assigned based on its characteristic quantification of polar metabolites in human urine and plasma
fragmentation pattern and retention index with subsequent extracts by GC-MS.369 However, multiple silylated adducts with
verification using a reference standard.322 However, extensive polyprotic metabolites along with resolution of deuterated
fragmentation hampers reliable elemental composition in EI- isotopomers by GC complicates data processing; however, this
MS unless complemented by chemical ionization (CI) that is process does not impact quantitative performance since EI-MS
needed for generating more intense molecular ions and isotope is not subject to sample matrix effects unlike ESI-MS that
signatures.358 In addition, other more sensitive ionization requires coelution of isotopomers.368 Because of the long
modes are also available in GC-MS but rarely used in reaction times and elevated temperatures that are required for
metabolomic studies to date, including negative ion chemical oximation/silylation, microwave-assisted derivatization proto-
ionization (NICI) 359 or electron capture negative ion cols allow for rapid silylation with total reaction times under 5
(ECNI)360 detection that offer lower detection limits for min.370−372 Despite the continued use of classical trimethylsi-
metabolites as their perfluorobenzyl derivatives. lylation in GC-MS-based metabolomics in large-scale inter-
Trimethylsilylation remains the most widely used labeling laboratory studies267,354 ongoing controversies raise concerns
procedure in GC-MS-based metabolomics given its general regarding its suitability for metabolite profiling because of its
applicability to various classes of polar metabolites.361 After poor analytical performance for amino acids in comparison to
sample extraction, a two-step derivatization reaction is LC-fluorescence.373 Indeed, recent studies have demonstrated
performed involving methoximation in pyridine to prevent the advantages of precolumn derivatization based on alkylation
reversible cyclization of reducing sugars followed by silylation using methylchloroformate (MCF)355,374 or ethylchlorofor-
using bis(trimethylsilyl)trifluoroacetamide (BSTFA) or pref- mate375,376 that converts amino and nonamino organic acids
erably N-methyl(trimethylsilyl)-trifluoroacetamide (MSTFA) into volatile carbamate and ester derivatives, respectively.
catalyzed in the presence of 1% trimethylchlorosilane.154 Noteworthy, chemical derivatization occurs rapidly in biological
Although chemical derivatization is typically performed off- samples under aqueous alkaline conditions without heating,
line using in an autosampler vial, in-liner derivatization of prior extraction or stringent quality controls as needed for
volume-restricted biological samples or single cells can also be silylation. Similar to esterification derivatization of plasma fatty
achieved by GC-MS using a direct thermal desorption interface acids,377 alkylation reactions are performed in aqueous solution
inside the injector port.362 However, the optimum derivatiza- with high alcohol content for improved solubility and
tion temperature of the injector port must be kept sufficiently stabilization of long-chain carboxylic acids, where ethyl esters
low (70 °C) to prevent degradation of labile metabolites that of short-chain organic acids possess better retention in GC.376
restricts thermal focusing of sample at the column head. For instance, microwave-assisted oximation together with MCF
Despite its broad reactivity with various classes of functional results in stable derivatives that are less susceptible to sample
groups with good analytical performance for “class-1” matrix allowing for reliable GC-MS analyses while using a single
metabolites (e.g., sugars, fatty acids, organic acids), silylation internal standard without quartz liner replacement over several
is prone to numerous artifacts that leads to poor precision and months.378 Also, highly fluorinated alkyl chloroformates with
lower recoveries in the case of “class-2” (e.g., amines, encoded chlorine tags represent a novel class of label that
phosphates) and notably for “class-3” (e.g., amide, thiols) greatly enhances sensitivity when using GC-MS with ECNI
compounds.363 Moreover, silylation is associated with multiple detection.379,380 However, chloroformate reagents are more
N dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 10. Total ion chromatograms by GC-MS comparing metabolomic profiles of volatile urinary metabolites from cancer patients (leukemia,
colorectal, and lymphoma) contrasted with a healthy volunteer (control group) after sampling by HS-SPME for 60 min at 50 °C for pH-adjusted
urine samples with 17% w/v sodium chloride under agitation. A significant increase in the peak area of 2-methyl-3-phenyl-2-propenal, p-cymene,
anisole, 4-methyl-phenol, and 1,2-dihydro-1,1,6-trimethyl-naphthalene, along with lower levels of dimethyl disulfide were detected in cancer patients.
Reprinted with permission from ref 391. Copyright 2011 Nature.

susceptible to fragmentation in EI-MS or ECNI because of their ripe fruits among 94 different tomato genotypes using
weak carbamate bond that may lack a detectable radical cation unsupervised multivariate data analysis.388 Similarly, untargeted
without CI mass spectra validation.375 Thus, the development metabolite profiling of volatile organics from four apple
of improved extraction and derivatization protocols are still varieties was investigated by GC-TOF-MS, where an
needed for GC-MS to avoid the large sources of bias and experimental design was used to optimize HS-SPME conditions
process variability associated with classical trimethylsilylation381 to maximize total signal responses, including preheating time,
that ultimately limits the discrimination power of metabolomics extraction time, and temperature.389 A comparative analysis of
for molecular phenotyping biological samples.378 apple extracts after freeze-drying was found to reduce levels of
Head-space (HS) sampling of the volatile metabolome volatiles with greater variability relative to freshly processed
represents one of the most unique features of GC-MS that samples highlighting the impact of sample collection and
avoids many of the technical hurdles of sample processing of storage. In this case, differentiation of the four apples varieties
polar metabolites in biological samples. SPME is frequently was achieved by multivariate data analysis based on 10 key
used for direct sampling of the volatile fraction of organic volatile alcohols and ester derivatives, whereas residual levels of
compounds distributed above a solid or liquid specimen using a the pre/post-harvest scald inhibitor, N-phenylaniline was
fused-silica fiber coated with a specific sorbent that integrates measured in a specific apple variety prone to oxidation/
sampling, extraction, concentration and sample desorption in a discoloration during storage.389
solvent-free system.382−384 In addition, in-fiber chemical There is growing interest in using HS-SPME-GC-MS for
derivatization can also be integrated together with HS-SPME comprehensive analysis of volatile organics in biological
for single-step sample workup prior to GC-MS.385 Sampling samples since it reflects the metabolic condition and health
bias is inherent with SPME due to differences in sorption status of an individual.390 Recently, differential metabolite
kinetics and metabolite volatility that is also matrix dependent; profiling of volatile metabolites from skin was developed for
however, relative quantification with good precision is feasible early detection and diagnosis of melanoma relative to normal
when performed under standardized conditions. Because of the skin and benign naevus (mole) samples from a 3-mm punch
limited loading capacity of SPME, improved sample enrichment biopsy, as well as air blank samples.146 Three volatile
and lower detection limits can be achieved using headspace stir hydrocarbons were significantly elevated in both fresh and
bar sorptive extraction methods.386,387 Given the importance of frozen melanoma specimens as secondary metabolites of
odor and fragrant compounds in food quality, agriculture and membrane lipid peroxidation/oxidation stress, namely 4-
plant sciences, high throughput screening of volatiles from methyldecane, dodecane and undecane, whereas pyridine and
tomatoes was demonstrated by HS-SPME-GC-MS388 that 3-hexanol were expressed from naevi samples. Since this is a
detected 322 distinct plant-derived compounds after sub- noninvasive method that preserves skin morphology, the same
traction of SPME fiber blanks. Several major classes of volatile specimens used for volatile collection by HS-SPME were
organics were identified, including phenolics, lipid derivatives, subsequently analyzed by histopathological staining within 3 h
terpenoids and carotenoids that allows for the discrimination of of the original biopsy collection.146 The discovery of putative
O dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

markers for noninvasive diagnostics was recently reported by 2.2. The Nonvolatile Metabolome by LC-MS
Silva et al.,391 which detected up to 82 different metabolites in LC-MS has undergone the greatest expansion within
the headspace of urine samples for three different cancer types. metabolomics since the first publications in 2002137,138 because
A comparison of different sorbent materials and experimental of its wide selectivity for resolving various classes of involatile
conditions used in HS-SPME was performed to maximize or thermally unstable metabolites. Due to recent advances in
extraction efficiency. Figure 10 highlights differential volatile column technology, UHPLC now offers analogous separation
urinary metabolite signatures measured in three representative efficiency as GC based on solvent or temperature programming
urine samples from cancer patients relative to healthy controls for ultrafast separations with high peak capacity.190 For
by GC-MS. Optimal extraction was performed using a instance, a major fraction of known metabolites within the
carboxen-polydimethylsiloxane sorbent in HS-SPME, where human serum metabolome (>70%) comprise nonpolar lipid
benzene derivatives, terpenoids and phenols were significantly classes (e.g., phospholipids, glycerolipids, and steroids), which
elevated in the oncological group, including lower levels of are well-suited to RP-LC-MS.20 For this reason, thousands of
dimethyldisulfide;391 however, sample sizes were small and not molecular features are detected in metabolomic studies from
adequately age or gender matched for this pilot study since methanol extracts of plasma when using RP-LC-MS under both
positive and negative ion mode ESI.403 In addition, greater
volatile compounds from urine have been demonstrated to be
metabolome coverage can also be achieved by LC-MS when
gender-specific.392 Moreover, volatile organics derived from using complementary ion sources that have greater response
biological samples are known to be dependent on both genetic toward nonpolar metabolites, such as APCI.36,404 Hundreds of
and environmental factors, such as diet, lifestyle and microbiota different commercial stationary phases exist in LC, where
that contribute to significant interindividual variability. Indeed, proprietary column packings are characterized based on their
greater volatile metabolome complexity has been reported in hydrophobicity and silanophilic activity at pH 7 using model
human sweat in comparison to urine or saliva.386 Thus, nonpolar and polar solutes;405 indeed, similar C18 packed
standardized conditions are critical for ensuring reproducible columns made by various manufacturers often exhibit different
volatile extraction using HS-SPME without background hydrophobicity under analogous elution conditions. Thus,
contamination, including high purity chemical reagents because reproducible analytical performance by LC-MS in metabolo-
of the high amounts of salt added to sample for increasing mics can only be achieved when using batches of columns from
metabolite volatility.392 Because of the difficulty in collection of the same manufacturer while implementing preventative
breath samples for routine clinical diagnostics,393 metabolomic maintenance protocols (e.g., cleaning, reconditioning or
studies of volatile metabolites from urine were also conducted calibration) between blocks of runs.403 Moreover, quality
in a mouse model of lung cancer.394 To validate that volatile control samples that are intermittently injected throughout an
odorants represent authentic markers of lung cancer, sensor analysis block an effective approach for signal drift correction in
mice were trained to discriminate among tumor bearing and LC-MS that minimizes intralaboratory and interlaboratory
nontumor control mice based on their behavioral response variation with CV < 20%.268,406 Indeed, the temporal stability
of both measured signal and retention time in LC-MS are
similar to canine scent detection for lung and breast cancer.395
critical for relative quantification due to long-term changes in
To better correlate perceived odor quality/intensity in relation ion source, column properties or separation conditions. Similar
to its chemical composition, GC-MS together with online to GC-MS, rigorous optimization of preanalytical steps prior to
olfactometry396,397 offers a unique approach to understand food LC-MS, including sample collection/storage407 and sample
quality traits associated with human sensory responses (i.e., pretreatment/cleanup,408 as well as default software parameters
sensometabolome)398 using a trained panel of subjects who used in data alignment and peak picking are required to ensure
function as detectors for odor-active components. The high quality data as a way to reduce the false discovery rate.409
chemical-sensory characterization of offensive or unpalatable Also, since the eluent composition and intrinsic solute
odors can also be explored for monitoring livestock environ- properties determine ionization efficiency in ESI-MS,304 careful
ments that impact air quality on local and regional scales, optimization of separation conditions in LC-MS is needed
including human/animal health within confined feeding when stable isotope internal standards are unavailable. A survey
operations.399 For instance, untargeted metabolite profiling of of major LC-MS configurations in metabolomics indicates that
dairy manure odor was performed by HS-SPME-GC-MS with RP-HPLC using standard 3 μm and increasingly UHPLC with
simultaneous olfactometry (O),400 which revealed over 86 1.7 μm BEH particles typically use C18 bonded-phase columns
different types of volatile organics, including 17 compounds under elevated temperatures (>40 °C) for improved stability,
only detected by smell that were present below the detection performance and reproducibility (Supporting Information
limit of either FID or EI-MS. Dimethyltrisulfide was identified Table S2). However, most LC-MS methods differ widely in
above its odor detection threshold, which is a volatile terms of sample pretreatment and optimum solvent gradient
elution program reflecting a compromise between sample
pheromone produced by the fly attracting plant, dead-horse
throughput and peak capacity requirements. For instance, over
arum florets401 and a characteristic odorant emitted by 3500 features can be measured in human plasma when using a
fungating cancer wounds.402 Improved resolution and detection shallow gradient over 75 min in RP-LC for discovery of
of early eluting volatile organics was achieved by multidimen- putative biomarkers that better discriminate between methyl-
sional GC-MS with heart cutting thereby allowing for a shorter malonic and propionic academias.410 Although C18 remains by
extraction time of 30 min by HS-SPME.400 Thus, GC-MS-O far the most common RP stationary phase of choice in
allows for improved characterization of complex mixtures of UHPLC-MS, improved performance has been reported with
low abundance odor-active compounds, including sulfur- C8 columns when analyzing plasma extracts by reducing sample
containing compounds, fatty acids, and phenol/indole deriva- carry-over of lipids with shorter column wash times between
tives.400 runs.411 Unlike most LC-MS studies in metabolomics that use
P dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 11. Technical reproducibility of plasma metabolite profiling by ANP-LC-MS. (A) Overlay of chromatograms acquired for 56 repeated
analyses of a plasma specimen. (B) Profile plot overlay of normalized extracted ion intensities for 374 distinct metabolites, quantified as a function of
injection number for the 56 repeated plasma analyses depicted in A. Results depict flat run-to-run variation (CV = 6.5%) in the levels of the 374
metabolites with repeated analysis. (C) Peak overlay of 56 repeated assessments of extracted ion counts for various classes of polar metabolites in
plasma demonstrating long-term method reproducibility. Reprinted with permission from ref 26. Copyright 2012 PLoS.

standard narrow bore (2.1 mm i.d.) columns with relatively “off-line” fraction collection methods based on LC × LC-MS
high flow rates (300−500 μL/min), capillary LC columns (150 have only been reported in metabolomics417 because of
μm i.d.) packed with 1.7 μm trifunctional (T3) C18 particles technical hurdles associated with elution solvent compatibility,
provide better retention of polar metabolites with higher peak such as strong cation-exchange/HILIC,418 ion pair RP/RP,419
capacity and greater sensitivity for plasma lipids when coupled and RP/HILIC.420
to nanoflow ESI-MS.412 Alternatively, perfluorinated stationary LC-ESI-TOF-MS is widely used in metabolomics studies
phases offer extended selectivity for polar metabolites relative given its fast duty cycles for sharp eluting peaks in UHPLC,
to alkyl and phenyl-based RP columns since they exhibit mixed- whereas other mass analyzers have been used to reduce isobaric
mode retention mechanisms with greater solute retention at interferences, including ultrahigh mass resolution FT-ICR-
elevated temperatures and higher organic solvent content.413 MS421 and orbitrap instruments.422 Since ESI-MS rarely leads
For instance, pentafluorophenylpropyl (PFFP) columns offer to extensive in-source fragmentation, Q-TOF in conjunction
higher separation efficiency with better resolution for polar with CID experiments is required for structural elucidation of
metabolites and their stereoisomers, notably organic acids and unknown molecular features that may serve as putative markers
amino acids relative to amino-based HILIC or C18 RP of disease progression;423 however, unlike EI-MS, spectral
columns.218 Superheated water at elevated temperatures matching by MS/MS from archived database entries or via in
(>100 °C) lowers the viscosity of mobile phase when using silico modeling are nontrivial given complex fragmentation
UHPLC that also improves solvent eluotropic strength without reaction pathways that are dependent on both metabolite
organic modifier resulting in shorter analysis times, lower back structure and collisional energy selected.330 In most cases, only
pressure, and higher selectivity. Indeed, an optimized thermal a small fraction of metabolites in LC-MS (<20−30%) can be
and solvent gradient program in UHPLC-MS allows for fast yet unambiguously identified given limited access to authentic
high resolution separations when analyzing diluted urine commercial standards.424 Indeed, various salt adducts and
samples relative to a conventional isothermal gradient elution background artifacts are prevalent in ESI-MS that complicates
at elevated temperature.414 However, poor separation perform- data processing if not adequately filtered based on peaks that
ance was found when analyzing plasma extracts due to the satisfy minimum detection thresholds (S/N > 10) within a
higher content of lipids that are not adequately soluble in a defined retention window while comprising reproducible
primarily aqueous mobile phase (0−10% acetonitrile). High signals measured in pooled samples (CV < 15%).319 Given
temperature UHPLC is also a key strategy for comprehensive the large number of unknown molecular features that are not
2D LC × LC separations to minimize undersampling of eluting present in current databases, open access repositories for
first dimension peaks to the second column with ultrafast metabolomics studies and meta-data are required for
elution programs.415,416 Although 2D LC × LC separations dissemination, validation, and future identification of novel
using a switching valve has been developed in proteomics,283 metabolites, such as MetaboLights.425 However, the qualitative
Q dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 12. Extracted ion chromatograms of 1:5 of 12C-/13C-dansylated Trp, Ile, Leu, and Lys obtained by fast gradient RP-UHPLC with FT-ICR-
MS. Precolumn chemical derivatization of amines, amino acids and phenols was performed using light and heavy isotopes of dansyl chloride for 60
min at 60 °C with reaction quenched by methylamine prior to mixing of samples together prior to MS analysis. Dansylation labeling enhances solute
ionization efficiency and improves retention of polar metabolites while also allowing for relative quantification of coeluting isotope pairs without bias
due to matrix-induced signal suppression. Reprinted with permission from ref 438. Copyright 2009 American Chemical Society.

and quantitative performance of LC-MS is highly dependent on reproducibility (CV = 6.5%, n = 56) and separation efficiency of
optimization of separation conditions that generate high peak polar metabolites in murine plasma when using ANP-LC-MS,
capacity. To enhance resolution of polar/ionic metabolites that including various classes of cationic and anionic metabolites
are not well retained by conventional RP-LC, complementary that have better retention time precision relative to HILIC.26
retention mechanisms can be used in parallel for the same Recently, a novel strategy for comprehensive metabolite
sample, notably HILIC102,426 or ANP.220,427 Indeed, two profiling using aqueous and organic fractions of liver extracts
chromatographic systems also permit greater confidence in by UHPLC-MS was reported based on three different
peak assignment if the ion is detected under both separation stationary phases, namely, C8, C18, and HILIC (amide) for
and/or ionization modes while also allowing for detection of nonpolar lipids (organic), semipolar (aqueous), and polar
weakly retained compounds that coelute in the void front in (aqueous) metabolites, respectively.432 In this case, a differ-
one of the separation modes.424 For instance, a greater coverage ential metabolomics study was performed for identification of
of urinary metabolites can be analyzed when using HILIC-MS putative steatotic (fatty) markers of impaired liver function,
in positive/negative ESI that is largely complementary to RP- which revealed systemic dysregulation in lipid, bile acid and
LC-MS since urine is largely comprised of polar/ionic amino acid metabolism.
metabolites.222 Similarly, metabolomics studies performed on Although a mixed-mode SPME coating fiber offers a sample
several different specimens derived from the same organism cleanup method for biological fluids in LC-MS,307 the depth of
further enhances confidence of the biological significance of the metabolome is limited by the intrinsic physicochemical
putative disease markers when using complementary C18 and properties of a solute since small and hydrophilic metabolites
fluorinated-based stationary phases in LC-MS.428 However, in have poor ionization efficiency and high detection limits.271
many cases, unique metabolic fingerprints are characteristic to Although LC-MS was originally introduced as a “label-free”
specific organs or biofluids when using UHPLC-MS with a approach for metabolite profiling, various functional group-
PFPP column that optimally resolved up to 112 unique specific chemical labelling strategies have been introduced to
hydrophilic metabolites within 8 min unlike RP (C18) and improve analytical performance,208 including concentration
HILIC (amino) modes of separation.429 The combination of a sensitivity, separation selectivity, quantitative reliability and
zwitter-ionic HILIC and C18 RP-LC strategy for expanded qualitative identification.433 In most cases, a reagent that
profiling of the tear metabolome allowed for the identification possesses an ionizable charged group (i.e., quaternary
of 60 diverse classes of metabolites with a majority of ammonium) that is also bulky, hydrophobic, and contains an
compounds reported for the first time.430 Indeed, expanded isotopic signature (e.g., Br, Cl 13C, 2H, etc.) is preferred, where
coverage of the metabolome in human serum using both reactions can occur in free solution,207 on a solid support resin
HILIC and RP-LC data sets allows for better classification and for facile sample workup,434 or dynamically during elution by
staging of disease stage relative to healthy controls, such as formation of an ion pair complex.435 Although chemical
renal cell carcinoma.431 Alternatively, RP-LC in conjunction derivatization is often used to introduce a positive charge to
with ANP-LC can be used to reveal unanticipated perturbations weakly ionizable/neutral lipids,436 it can also be applied to
of xanthine oxidreductase inactivation in mice models induced reverse the charge of acidic metabolites upon adduct formation
by gene deletion or pharmacological inhibition, such as that enhances detection sensitivity over 2500-fold under
treatment with allopurinol.26 Figure 11 highlights the technical positive ion mode, such as fatty acids.437 Figure 12 highlights
R dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

the use of differential 12C/13C isotope dansylation labeling for online sample preconcentration together with a commercial low
relative quantification of amines, amino acids and phenols in flow/sheathless interface for CE-MS enhances concentration
human urine by LC-FT-ICR-MS under positive ion mode that sensitivity by over 2 orders of magnitude while maintaining
results in a signal enhancement ranging from 1 to 3 orders of high separation efficiency relative to a coaxial sheath liquid
magnitude while improving retention of polar amino acids by interface as shown in Figure 13.274 This approach allows for
RP-LC.438 Since identical sample aliquots are derivatized with deeper coverage of the urinary metabolome with nanomolar
light and heavy (2 × 13C) analogs of dansyl chloride and detection limits for most polar metabolites with over a 4-fold
subsequently mixed together prior to LC-MS, co-elution of a increase in detected molecular features (50−450 m/z),
pair of isotopomers offset by a m/z of 2 allows for reliable including low abundance ions above m/z 300, such as small
relative quantification without stable-isotope internal standards. peptides.274 Similarly, a sheathless CE-MS format based on a
Absolute quantification of dansyl chloride-reactive metabolites porous sprayer resulted in a 5-fold improvement in
is also feasible when isotopic encoding (13C) a pooled reference
sample relative to each individual sample, which allows for
detection of 1,058 pairs of putative metabolites in saliva
samples when using UHPLC-MS.421 However, de novo
identification of unknown metabolites remains a major obstacle
since comparison of retention times and MS/MS spectra
cannot be compared with archived public databases based on
underivatized metabolites. Moreover, an array of selective
chemical labels are ultimately needed for reacting with specific
functional groups in order to extend coverage of the
metabolome.434
2.3. The Ionic Metabolome by CE-MS
CE-MS represents a promising platform in metabolomics that
is applicable for resolution of polar/ionic and labile metabolites
that have poor retention in RP-LC or require complicated
sample handling prior to GC-MS.439 Moreover, high efficiency
separations of multivalent charged ions and their stereoisomers
can be realized by CE that are difficult to resolve by HILIC or
ANP without deleterious band broadening.222 For instance,
over 1692 molecular features were detected by CE-MS from
cell extracts of B. subtilus comprising charged and hydrophilic
metabolites that can be used for differentiation of bacterial Figure 13. (A) Base peak electropherogram (m/z 50−450) of human
sporulation.440 Direct analysis of strongly ionic and secondary urine obtained with sheathless CE-MS using a porous tip sprayer for
metabolites are also ideal for CE-MS without enzyme expanded metabolome coverage. Conditions: BGE, 10% acetic acid
deconjugation, including S-glutathione, N-glycine, O-glucur- (pH 2.2); sample injection, 2.0 psi for 30 s (∼1% of capillary volume).
onide, and O-sulfate conjugates, as well as their positional (B) Base peak electropherogram (m/z 50−450) of human urine
isomers.262,441 Recently, a comparison of CE-MS with RP- obtained with CE-MS using a coaxial sheath liquid interface.
UPLC-MS for comprehensive profiling of human urine Conditions: BGE, 10% acetic acid (pH 2.2); sample injection, 0.5
psi for 30 s (∼1% of capillary volume). Reprinted with permission
revealed that both techniques are largely complementary with from ref 274. Copyright 2012 American Chemical Society.
respect to metabolome coverage with CE-MS preferentially
detecting the majority of low molecular weight (m/z < 150)
and ionic metabolites coeluting in the void volume;442 however, concentration sensitivity notably for higher molecular weight
CE-MS detected 10-fold fewer significant molecular features in cations (m/z > 250) although response factor enhancement
urine for subject gender classification relative to UHPLC-MS among several small hydrophilic metabolites were not as
because of sensitivity limitations related to the much lower significant due to higher background noise in the low m/z
sample volumes injected (10 nL versus 30 μL). Indeed, CE as a range.273 Further studies are needed to evaluate the long-term
microseparation technique lends itself well to metabolomics robustness of the sheathless sprayer given the inherent fragility
studies of volume-restricted specimens and single cell analyses of the fluoride-etched porous tip,445 including batch-to-batch
needed for understanding accelerated aging processes in mice reproducibility and its performance under negative ion mode
models443 and cell heterogeneity in freshly isolated and conditions. Indeed, CE-MS methods continue to be reported
cultured neurons,444 respectively. Nevertheless, migration without adequate quality control practices or interlaboratory
time reproducibility, concentration sensitivity, and method trials needed for ensuring long-term reproducibility similar to
robustness remain key obstacles in CE-MS that has hindered its recent NMR,269 LC-MS,268 and GC-MS267 initiatives.
adaptation by the broader metabolomics community. This issue A survey of representative configurations used in CE-MS-
is partially explained by few commercial CE-MS vendors that based metabolomics to date (Supporting Information Table
offer full service support and the lack of customized software S3) highlight that separations of cations and anions are typically
tools developed for processing electrophoretic data, which is performed using bare or surface coated fused-silica capillaries
required for correction of migration times with good under strongly acidic (pH < 2) and alkaline (pH > 8)
precision.247,289 Indeed, several recent advances in CE-MS conditions, respectively. The main benefits of operating under
have largely addressed underlying concerns regarding method extreme pH conditions is that it ensures that weakly ionic
sensitivity and robustness. For instance, the integration of metabolites are adequately ionized while also generating a more
S dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

Figure 14. (a) Series of extracted ion electropherograms demonstrating comprehensive assessment of plasma thiol redox status by CE-MS in
conjunction with thiol-selective maleimide (NTAM) derivatization for simultaneous analysis of labile reduced thiols, as well as intact symmetric and
mixed oxidized disulfides over a wide dynamic range. (b) Total ion electropherogram for the same plasma specimen that highlights integration of
untargeted metabolite profiling for deeper insight into metabolic pathways modulated by thiol dysregulation. Reprinted with permission from ref
463. Copyright 2011 ACS.

stable EOF that is variable near neutral pH conditions due to Alternative capillary surfaces have also been explored in CE-
the weak acid ionization of surface silanol groups (pKa ≈ 6.5). MS, including polymer-based449 and sulfonic acid modified
Indeed, long-term variations in EOF in fused-silica capillaries fused-silica capillaries450 to better control the EOF, as well as
are highly dependent on electrolyte conditions and applied reduce ion adsorption. Soga and co-workers440 first introduced
voltage, as well as surface properties of the capillary wall that three different operating conditions in CE-MS-based metab-
alters with time due to cation adsorption446 if proper rinsing olomics for cations, anions and multivalent anions in order to
procedures are not implemented. Although dynamically coated achieve maximize metabolome coverage; the first method is
capillaries based on a triple layer of charged polymers improve widely adopted by several other research groups using strongly
long-term migration time reproducibility (CV < 1%) in high acidic formic acid (5% v) or acetic acid (10% v) as the BGE
salt urine samples,447 extra conditioning steps are needed for since it allows for excellent analytical performance for cationic
good performance, whereas surface adsorption of multivalent metabolites under suppressed EOF conditions.451−454 How-
anions can limit its applicability.440 In most cases, bare fused- ever, the two other methods for anion metabolite profiling that
silica capillaries with 50 μm i.d. are widely in CE-MS, however rely on a cationic coated polymer capillary (e.g., SMILE (+)) or
more narrow capillaries (e.g., 5−25 μm) have also been shown neutral polydimethylsiloxane-coated GC-capillary under re-
to improve separation efficiency by minimizing Joule heating versed polarity and negative ion mode440 have not gained wider
and siphoning effects for ultrafast separations (<1 min).448 acceptance due to inherent flaws impacting method robustness
T dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

and sample throughput.266 Both types of coated capillaries are intracellular oxidized glutathione was measured during
susceptible to hydrolysis with limited pH stability455 that can strenuous exercise that was regulated back to pre-exercise
lead to ion source contamination, whereas CE-MS under levels upon recovery. High dose oral N-acetylcysteine pretreat-
reversed polarity results in corrosion of the stainless steel ment was found to transiently downregulate resting metabolism
sprayer.265 Although more stable coated capillary formats with a while modulating metabolic responses to exercise-induced
platinum-based emitter are recently available,265 this is oxidative stress with faster recovery.31 Since oxidative stress is
ultimately unnecessary since anions (e.g., organic acids, sugar associated with aging and many chronic/acute diseases,462 a
phosphates, redox cofactors, nucleotides) can be readily new strategy for comprehensive plasma thiol redox status was
analyzed using bare fused-silica capillaries under normal introduced based on thiol-specific maleimide labeling for
polarity with pressure-assisted flow456 or methanol as an simultaneous determination of nanomolar levels of reduced
organic additive.457 Although ammonium acetate or bicarbon- thiols and free oxidized thiols as their intact symmetric and
ate are widely used volatile buffers for analysis of anionic mixed disulfides.463 Figure 14 highlights that artifact-free
metabolites using alkaline conditions in CE-MS, improved analysis of various plasma thiols can be achieved over three
sensitivity can be realized by using amine-based buffers as orders of dynamic range in CE-MS, where the use of a cationic
electrolytes in the BGE to reduce co-ion suppression, which maleimide label was found to stabilize reduced thiols as their
contributes to a 2-fold increase in the total number of thioether adducts while boosting sensitivity. Overall, maleimide
molecular features detected in human urine.272 Thus, caution labeling was not found to change the responses of other plasma
is warranted against the use of polymer-coated capillaries under metabolites, where a multivariate model was used to predict
reversed polarity for anionic metabolite profiling by CE-MS relative response factors for model thioether adducts with
given its many technical problems.458 various maleimide substituents.463 A study exploring the
Similar to recent developments in GC-MS and LC-MS-based correlation between sensory evaluation scores of Japanese
metabolomics, greater attention is required for optimization of sake to their metabolome using CE-MS was reported based on
sample pretreatment454 and separation/ionization conditions in four trained panelists who graded samples based on their
CE-MS459 to enhance analytical performance without bias. sweetness, sourness, bitterness and zatsumi, an unpleasant not
Although separation of neutral steroids is feasible by MEKC or clear flavor.464 A predictive “electric tongue” model for
CEC, direct coupling to ESI-MS limits selectivity by restricting estimating the taste and quality of different sake brands was
use of involatile surfactants or additives due to signal developed using support vector regression, which demonstrated
suppression. To date, CE-ESI-TOF-MS using a coaxial sheath good correlation notably with the undesirable zatsumi flavor
liquid interface with aqueous volatile buffers remains the most score. However, only 14% of detected peaks were identified in
common instrumental format for routine metabolomics studies. this study, whereas the overall significance of altered levels of
Leon et al.460 compared the metabolome profiles between three individual metabolites with respect to sensory responses was
isogenic wild-lines relative to three transgenic strains of maize not well understood.464 Recently, an extensive metabolomics
seed using CE-FT-ICR-MS after pressurized liquid extraction study was reported for the discovery of putative markers of liver
to evaluate their bioequivalence. In this case, genetically disease among 248 serum samples derived from seven
modified maize strains were differentiated from their parental subclasses of liver disorders and healthy controls.452 CE-
wild lines when using partial least-squares analysis, however TOF-MS-based metabolomics revealed that γ-glutamyl dipep-
several annotated features only expressed in genetically tides were found to be highly elevated in all liver diseases
modified strains were not identified based on their empirical relative to controls, which was associated with deleterious
formula with the exception of L-carnitine, apigeninidin and 5,6- oxidative stress due to depletion in reduced glutathione. In this
dihydroxyindole. One way to improve concentration sensitivity case, a specific set of γ-glutamyl dipeptides, transaminases, and
in CE-MS for anionic metabolite profiling is the use of chemical methionine sulfoxide were collectively used to discriminate
derivatization, such as N-butyl-4-aminomethylpyridium that among multiple liver disorders when using receiver-operating
contains a precharged quaternary amine, deuterium label (2H × characteristic curves as a way to provide a reliable diagnostic
9), and a reactive primary amine for labeling to carboxylic acids test without invasive liver biopsies.452 A similar approach was
after activation.461 In this case, CE separations can be reported for the discrimination of different types of cancer
performed under strongly acidic conditions and normal polarity using noninvasive saliva sampling with CE-MS from 211
when using positive ion mode ESI-MS since carboxylic acid different patients, including oral, pancreatic, and breast cancer
derivatives possess a net positive mobility. Unlike LC-MS, patients, as well as periodontal disease and healthy controls.465
deuterium-encoded isotopomers (d8) were found to coelute However, since salivary metabolite profiles are highly depend-
with light form derivatives in CE-MS thereby allowing for ent on various physiological and lifestyle factors466 standardized
relative quantification of urine samples without bias, which also sampling protocols and carefully designed cohort groups are
aided unknown structural identification by characteristic MS/ needed to minimize innate biological variation notably when
MS transitions.461 Reduced thiols are difficult to accurately collected from different hospital settings.
measure due to their susceptibility to oxidation, thiol-disulfide
exchange and poor ionization efficiency that requires careful 3. SOURCES OF BIAS AND FALSE DISCOVERY IN
optimization of all preanalytical and analytical steps, including METABOLOMICS
sample collection/storage, sample pretreatment and electro- The major aim of metabolomics is to measure statistically
phoretic separation conditions.441 Indeed, CE-MS allows for different metabolite responses between two or more groups of
reliable quantification of reduced thiols and their oxidized defined samples (e.g., placebo/treated, wild-type/mutant, etc.)
disulfides from red blood cell lysates that is needed for based on relative quantification of their average signal intensity
assessment of the synergistic interactions of nutrition and when using NMR and/or MS. However, the overall significance
exercise in human health.31 For instance, time-resolved of putative “marker” compounds will ultimately depend on the
metabolomic studies revealed that over a 50-fold increase in underlying stability of metabolite concentration levels in a given
U dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

sample and the precision associated with the measurement. In collected from the same individual can distinguish subtle
many ways, metabolomic studies are confronted with the same presymptomatic phenotypes not evident by steady-state
problems that have plagued the failure of protein cancer experiments due to high biological variance.483 Since acceptable
biomarker discoveries to date.467 Sources of bias are attributed technical/process (RSD < 15%) and interlaboratory variance
to either fraud, false discoveries due to improper validation or (RSD < 20%)268 is often less than “normal” biological variance
true discoveries that ultimately lack adequate biological under standardized conditions (RSD < 60%), a minimum
significance or clinical value to patient care in the context of threshold change of about a 2-fold for pairwise comparison of
diagnosis, prognosis, subclass stratification or therapeutic relative responses in average metabolite signals is considered
monitoring.468 Bias is minimized with a carefully planned statistically significant when applying Bonferroni correction to
study design while implementing a fully validated metabolomics reduce the high false discovery rate with multiple hypothesis
workflow (i.e., preanalytical, analytical, postanalytical, and testing.484 False-discoveries in metabolomics often stem from
bioinformatic) that depends on knowledge of the chemical inadequate sample size or group class selection that contributes
properties and biological functions of a marker compound, to data overfitting when using multivariate statistics and
including its natural variation in sample and intrinsic chemical machine learning. In this case, stringent testing of predictive
stability. Indeed, there have been few reported metabolomic accuracy, such as cross-validation of a training set with
studies that have translated biomarker discovery into clinical independent testing of a hold-out set can be performed to
practice with the probable exception of sarcosine as a demonstrate biological significance.485
prognostic marker of prostate cancer469 that is still being 3.1. Sample Pretreatment and Quality Assurance
evaluated for its appropriate clinical utility.470
Early metabolomic studies have recognized the impact of Common sample preparation protocols used in metabolomics
diurnal cycle, diet, gender and estrus cycle on measured prior to separation486 include solvent extraction, ultrafiltration,
metabolite profiles in laboratory rats,471−473 which were long solid-phase extraction, and chemical derivatization, which can
known as “extrinsic” factors that impact metabolite profiling of lead to wide variations in recovery with distinct metabolic
biological fluids by GC-MS.67,474 For instance, recent studies profiles measured for the same sample.454 Thus, optimization of
have demonstrated that about 15% of identified metabolites in sample pretreatment is critical for ensuring stable yet
human plasma (e.g., fatty acids) and saliva (e.g., amino acids) representative metabolite concentration levels derived from
are under circadian control.475 The biological variance in the the original specimen while also being compatible to a
metabolome of yeast can vary from about 10% CV (n = 12) separation method and increasingly across multiple instrumen-
when cultured under defined conditions, but can exceed 40% tal platforms.487−489 A design of experiments can be used to
CV for more complex organisms depending on food source, optimize solvent extraction and chemical derivatization
such as C. elegans.476 Although different in-bred genetic strains conditions needed to understand significant variables and
of mice have distinct microflora populations reflected within their interactions that maximize recovery of metabolites.490 All
the host metabolome, this is found to be absent for genetically preanalytical steps during sample pretreatment need to be
distinct mice gestated by the same mother or isogenic mice investigated when using stable-isotope internal standards or
relocated to different laboratories highlighting the strong pooled samples as quality controls to properly evaluate absolute
influence of environment.477 Indeed, the innate biological metabolite recovery, process efficiency and residual enzyme
variability of metabolism places major constraints when activity with acceptable precision for various specimens.491−494
performing observational studies involving humans notably Seemingly minor details regarding sample handling can result in
when specimens are only collected at a single time frame as a significant changes in measured metabolite profiles, such as the
“metabolic snapshot”, such as a morning fasting blood impact of preservative agents in urine specimens,495 delayed
sample.31 For instance, a longitudinal study of biofluids storage of cerebrospinal fluid samples,496 slow deproteinization
collected from healthy postmenopausal female twins over of plasma,497 flash quenching of metabolism and extraction of
several months demonstrated that endogenous metabolite cell cultures,349 and storage temperature, prefractionation, and
concentration variation have a stable familial and individual- repeated freeze−thaw cycles on serum lipids.498 Indeed, the use
environmental component with an average variance of 47% in of different solvent extraction methods can contribute to
urine that increased to over 60% in plasma for major conflicting biological interpretations of intracellular metabolite
compounds detected by 1H NMR.478 Similarly, LC-MS-based profiles that raises questions on whether most published
metabolomics studies confirmed biological variability ranging metabolomic studies accurately reflect the metabolic status of a
from a median variance of 35% in human cerebrospinal fluid to sample without appropriate method validation.499 Therefore,
over 46% in the case of human plasma when using an optimized there is urgent need for the use of standardized protocols for
methanol extraction protocol for sample deproteinization prior sample collection, storage and processing500 that is critical for
to LC-MS.479 Thus, metabolomic studies based on drug or large-scale prospective studies in metabolomics.403,501 How-
lifestyle interventions suffer from significant intraindividual ever, certain labile classes of metabolites remain difficult to
(e.g., diurnal cycle, menstrual cycle, etc.) and interindividual quantify due to their inherent volatility, limited thermal or pH
variances (e.g., ethnicity, diet, etc.), including less well stability, photosensitivity or susceptibility to hydrolysis or
characterized effects, such as time of last meal, hydration oxidation, such as reduced thiols, unsaturated lipids, and redox-
status, and noncompliance with study protocols.480 In contrast active cofactors. Although the efficacy of liquid extraction
to large-scale randomized controlled trials, crossover inter- methods are often dependent on solvent polarity and pH
ventions offer a powerful approach in metabolomics481 for conditions,492,502 combining a two-step series of extractions503
measuring authentic treatment effects on an individual level or an optimized extraction using a single miscible solvent
instead of a group level since the same subject serves as their mixture488 enables greater sample throughput and broader
own control.482 Similarly, time-resolved metabolomics involv- metabolite coverage with acceptable reproducibility. Long-
ing stressor/challenge experiments with multiple specimens established protocols developed for targeted metabolite
V dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

profiling by GC-MS can also lead to unanticipated artifacts,

sub-2 μm or core-fused particles; elevated T/


2D GC × GC for higher peak capacity; high

thermal elution; chemical derivatization for


T ionic liquid columns; microwave-assisted

sheathless ion source; data transformations


such as urease treatment for sample cleanup of urine102 and

derivatization for improved performance


for precise migration times; chemical
methoximation/silylation derivatization of polar metabolites.504

alkylation in aqueous conditions


Given the recent expansion of metabolomics across various
research fields that employ different instrumental techniques, a

recent advances
consensus on a minimum set of reporting standards to enhance

improved performance
data quality is required while providing meta-data information
to promote transparency, best practices, and data ex-
change.314,505 For instance, quality assurance practices in
metabolomics typically incorporate the use of a series of
internal and/or recovery standards to evaluate variation in
sample processing that also improves data alignment,154 as well
as a comparison of different transformation methods on
biological information content,506 intermittent injection of a

migration time variability; poor concentration sensitivity;


HILIC/ANP; susceptible to ion suppression with ESI-

lack of interlab validation for ensuring robustness; few


complex sample pretreatment; stringent QC/validation;

variable retention times and complex mechanisms for


pooled sample as quality control for assessing long-term signal

not applicable to labile metabolites; artifact-prone

MS; limited MS/MS databases for identification


drift,507 and a blank subtraction or a dilution trend filter to
reject a majority of “spurious” peaks in ESI-MS.319 A variety of
proprietary, open-source and web-based software packages289

commercial CE-MS vendors/support


Table 1. Summary of Merits of Major Separation Platforms and Recent Advances for Comprehensive Metabolite Profiling
have been developed for data preprocessing of high resolution
MS-based metabolomics data sets, including file conversion,

limitations
mass filtering, retention time alignment, peak picking, data
normalization together with integrated bioinformatics tools for
statistical analysis and visualization;247,508−513 however system-
atic bias can occur since deconvolution algorithms are often not
optimized for specific instrumental platforms.102,514 Indeed,

silylation
there are ongoing efforts to enhance data preprocessing
strategies515 in order to improve the quality of peak selection
based on a “reliability index” of significant features derived from
a dilution series409 instead of maximizing the total number of
high separation efficiency; robust/mature platform with

high separation efficiency; online sample preconcentra-


EI/CI-MS; precise retention indices; extensive NIST

separation efficiency with UHPLC; robust perform-

tion/desalting; minimal sample handling; predictive


detectable features. This process is essential to downstream

ance of RP-LC with broad metabolome coverage


efforts dedicated to data analysis516 and structural identifica-
wide selectivity with RP, HILIC and ANP; high

tion330 that constitute two major bottlenecks in a metabolomics


workflow. Biological interpretation of complex metabolic
networks on a systems level also remains a major obstacle in
metabolomics517 that requires integration of metabolic flux
advantages

modeling of migration times

studies518 and “multi-omic” data approaches to unravel poorly


understood genotype-phenotype interactions.519,520
3.2. Perspectives and Future Trends in Separation Science
Separation science plays an important role in metabolomics by
enhancing the analytical performance of MS and NMR-based
database

methods needed for comprehensive profiling of metabolites in


complex biological samples. Given the chemical diversity and
dynamic range of the metabolome that remains largely
free approach for charged metabolites and peptides; ideal
derivatization required for polar metabolites; ideal with

uncharacterized, separation techniques based on chromatog-


approach for nonpolar/semipolar metabolites; ideal for
volatile, nonpolar, thermally stable metabolites; chemical

weakly ionic/strongly ionic and labile metabolites; label-


involatile and thermally unstable metabolites; label-free

raphy and electrophoresis offer distinct advantages for different


classes of metabolites as summarized in Table 1. Besides
involatile, nonpolar and semipolar metabolites.

resolving isobaric and isomeric compounds, high efficiency


separations also are needed for boosting concentration
metabolite compatibility

for multivalent ions and their isomers

sensitivity, reducing matrix-induced signal suppression effects,


while providing qualitative information that supports structural
headspace sampling with SPME

elucidation of unknown molecular features. The latter feature


requires theoretical models for accurate prediction of the
retention/migration behavior of diverse classes of metabolites
together with experimental conditions that provide reprodu-
cible yet robust separation performance. Indeed, deeper insight
into the biological significance of metabolites as putative
markers of health or disease status ultimately relies on correct
identification of a unique chemical structure from an empirical
formula. A compromise between sample throughput and
separation
platform

metabolomic coverage is required when selecting appropriate


GC-MS

CE-MS
LC-MS

separation mechanisms and elution conditions for generating


high quality data while minimizing various sources of bias that
W dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX
Chemical Reviews Review

contribute to false discoveries. Systematic optimization of all Notes


preanalytical steps in a metabolomics workflow (e.g., study The authors declare no competing financial interest.
design, sample collection/storage, sample pretreatment) is
essential to reduce biological variability needed for discrim- Biographies
ination of the metabolic phenotype among two or more groups
under standardized conditions. Moreover, postanalytical steps
related to data preprocessing, multivariate analysis and
biological interpretation require rigorous validation to avoid
data overfitting using appropriate statistical methods. GC-MS
and increasingly 2D GC × GC-MS offers high resolution
separation of volatile organics (i.e., volatile metabolome) when
using headspace sampling together with SPME, but requires
stringent quality control notably when using silylation.
UHPLC-MS is the method of choice for involatile metabolites
(i.e., nonvolatile metabolome) that include nonpolar and polar
compounds when using two or more separation modes based
on RP, HILIC, or ANP. The use of sub-2 μm and core−shell
particles in LC-MS with optimized solvent gradient elution
programs at elevated temperatures allow for fast separations
with high peak capacity. However, HILIC and ANP require Naomi L. Kuehnbaum is a Ph.D. student in bioanalytical chemistry in
further development to enhance separation performance the Department of Chemistry and Chemical Biology at McMaster
notably for highly charged/multivalent ions that are susceptible University in Hamilton (ON, Canada) after graduating with an H.B.Sc.
to adsorption. In this context, CE-MS is best suited for weakly in Chemistry and Forensic Science from the University of Toronto
and strongly ionic metabolites (i.e., ionic metabolome), as well Mississauga (ON, Canada). Her research is focused on the
as their stereoisomers, which allows for easy integration of development of novel methods based on capillary electrophoresis-
online sample preconcentration and desalting steps during mass spectrometry for metabolomics and the validation of synergistic
electromigration. Strategies to further improve the sensitivity nutritional and exercise interventions in human health.
and robustness in CE-MS notably for anionic metabolites are
needed for its wider acceptance, including quality assurance
protocols and interlaboratory testing of methods. Although LC-
MS and CE-MS are considered “label-free” methods for
metabolite profiling, chemical derivatization offers a promising
approach to enhance sensitivity, improve retention/mobility
and enable relative quantification without bias when using
isotope-encoded labels. There is growing interest in multiple
instrumental platforms for expanded coverage of the metab-
olome that takes advantage of the unique selectivity of GC, LC,
or CE separations in conjunction with optimized sample
extraction protocols and different ionization modes in MS.
However, multidimensional LC-MS and CE-MS separations are
still not adequately developed because of technical challenges
related to their direct coupling and solvent compatibility,
whereas hybrid separation modes based on supercritical fluid- Philip Britz-McKibbin is an Associate Professor of bioanalytical
LC-MS, CEC-MS, and MEKC-MS still remain largely unex- chemistry in the Department of Chemistry and Chemical Biology at
plored to date. Given the failure of massive research McMaster University in Hamilton (ON, Canada). His research
investments in the genomic sciences to translate biomarker program is focused on fundamental studies of capillary electrophoresis
discoveries into improved patient care,521,522 the onus is on and mass spectrometry for metabolomics, as well as characterization of
researchers using metabolomics to demonstrate that authentic biomolecular interactions in free solution. His research interests aim to
scientific discoveries lead to novel biological insights that develop improved assays for metabolite profiling, biomarker discovery,
positively impact human health and well-being. and drug screening for the prevention and treatment of genetic
disorders and chronic diseases.
ASSOCIATED CONTENT
ACKNOWLEDGMENTS
*
S Supporting Information
P.B.M. wishes to acknowledge funding support from National
Tables S1, S2, and S3. This information is available free of Science and Engineering Research Council of Canada and the
charge via the Internet at http://pubs.acs.org/. Ontario Genomics Institute. N.L.K acknowledges support in
the form of a Queen Elizabeth II Graduate Scholarship in
AUTHOR INFORMATION Science and Technology.
Corresponding Author REFERENCES
*Address: Department of Chemistry and Chemical Biology, (1) Patti, G. J.; Yanes, O.; Siuzdak, G. Nat. Rev. Mol. Cell Biol. 2012,
McMaster University, Hamilton, ON, L8S 4M1, Canada. Fax: 13, 263.
+1-905-522-2509. E-mail: britz@mcmaster.ca. (2) Veenstra, T. D. Genome Med. 2012, 4, 40.

X dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(3) Johnson, C. H.; Gonzalez, F. J. J. Cell. Physiol. 2012, 227, 2975. (36) Wikoff, W. R.; Anfora, A. T.; Liu, J.; Schultz, P. G.; Lesley, S. A.;
(4) Saito, K.; Matsuda, F. Ann. Rev. Plant Biol. 2010, 61, 463. Peters, E. C.; Siuzdak, G. Proc. Natl. Acad. Sci. U.S.A. 2009, 106, 3698.
(5) Zhao, L.; Nicholson, J. K.; Lu, A.; Wang, Z.; Tang, H.; Holmes, (37) Holmes, E.; Li, J.; Athanasiou, T.; Ashrafian, H.; Nicholson, J.
E.; Shen, J.; Zhang, X.; Li, J. V; Lindon, J. C. J. Proteome Res. 2012, 11, Trends Microbiol. 2011, 19, 349.
3509. (38) Wang, Z.; Klipfell, E.; Bennett, B. J.; Koeth, R.; Levison, B. S.;
(6) Jones, D. P.; Park, Y.; Ziegler, T. R. Annu. Rev. Nutr. 2012, 32, DuGar, B.; Feldstein, A. E.; Britt, E. B.; Fu, X.; Chung, Y.-M.; Wu, Y.;
183. Schauer, P.; Smith, J. D.; Allayee, H.; Tang, W. H. W.; DiDonato, J. A.;
(7) Antignac, J.-P.; Courant, F.; Pinel, G.; Bichon, E.; Monteau, F.; Lusis, A. J.; Hazen, S. L. Nature 2011, 472, 57.
Elliott, C.; Le Bizec, B. TrAC, Trends Anal. Chem 2011, 30, 292. (39) Lewis, N.; Nagarajan, H.; Palsson, B. Nat. Rev. Microbiol. 2012,
(8) D’Alessandro, A.; Giardina, B.; Gevi, F.; Timperio, A. M.; Zolla,
10, 291.
L. Blood Transfus. 2012, 10 (Suppl 2), s19.
(40) Levy, S.; Sutton, G.; Ng, P. C.; Feuk, L.; Halpern, A. L.; Walenz,
(9) Viant, M. R.; Sommer, U. Metabolomics 2013, 9, S144.
(10) Han, X.; Gross, R. W. J. Lipid Res. 2003, 44, 1071. B. P.; Axelrod, N.; Huang, J.; Kirkness, E. F.; Denisov, G.; Lin, Y.;
(11) Fricker, L. D.; Lim, J.; Pan, H.; Che, F.-Y. Mass Spectrom. Rev. MacDonald, J. R.; Pang, A. W. C.; Shago, M.; Stockwell, T. B.;
2006, 25, 327. Tsiamouri, A.; Bafna, V.; Bansal, V.; Kravitz, S. A.; Busam, D. A.;
(12) Mounicou, S.; Szpunar, J.; Lobinski, R. Chem. Soc. Rev. 2009, 38, Beeson, K. Y.; McIntosh, T. C.; Remington, K. A.; Abril, J. F.; Gill, J.;
1119. Borman, J.; Rogers, Y.-H. Y.-H.; Frazier, M. E.; Scherer, S. W.;
(13) Herrero, M.; Simo, C.; Garcia-Canas, V.; Ibanez, E.; Cifuentes, Strausberg, R. L.; Venter, J. C. PLoS Biol. 2007, 5, e254.
A. Mass Spectrom. Rev. 2012, 31, 49. (41) Adamski, J. Genome Med. 2012, 4, 34.
(14) Marshall, A. G.; Rodgers, R. P. Proc. Natl. Acad. Sci. U.S.A. 2008, (42) Chace, D.; Sherwin, J.; Hillman, S.; Lorey, F.; Cunningham, G.
105, 18090. Clin. Chem. 1998, 44, 2405.
(15) Jones, C. J.; Larive, C. K. Anal. Bioanal. Chem. 2012, 402, 61. (43) Fleeman, N.; Dundar, Y.; Dickson, R.; Jorgensen, A.;
(16) Agnolet, S.; Jaroszewski, J. W.; Verpoorte, R.; Staerk, D. Pushpakom, S.; McLeod, C.; Pirmohamed, M.; Walley, T.
Metabolomics 2010, 6, 292. Pharmacogenomics 2011, 11, 1.
(17) Lei, Z.; Huhman, D.; Sumner, L. J. Biol. Chem. 2011, 286, (44) Bowen, B.; Northen, T. J. Am. Soc. Mass. Spectrom. 2010, 21,
25435. 1471.
(18) Metz, T. O.; Zhang, Q.; Page, J. S.; Shen, Y.; Callister, S. J.; (45) Garrod, A. E. Lancet 1902, 2, 1616.
Jacobs, J. M.; Smith, R. D. Biomarkers Med. 2007, 1, 159. (46) Williams, R. J. Chem. Eng. News 1947, 25, 1112.
(19) Dettmer, K.; Aronov, P.; Hammock, B. Mass Spectrom. Rev. (47) Williams, R. J.; Berry, L. J.; Beerstecher, E. J. Proc. Natl. Acad.
2007, 26, 51. Sci. U.S.A. 1949, 35, 265.
(20) Psychogios, N.; Hau, D.; Peng, J.; Guo, A.; Mandal, R.; Bouatra, (48) Shostenkot, Y. V.; Georgievskii, V. P.; Levin, M. G. Anal. Chem.
S.; Sinelnikov, I.; Krishnamurthy, R.; Eisner, R.; Gautam, B.; Young, 2000, 55, 905.
N.; Xia, J.; Knox, C.; Dong, E.; Huang, P.; Hollander, Z.; Pedersen, T.; (49) Zakaria, M.; Gonnord, M.-F.; Guiochon, G. J. Chromatogr. 1983,
Smith, S.; Bamforth, F.; Greiner, R.; McManus, B.; Newman, J.;
27, 127.
Goodfriend, T.; Wishart, D. PLoS One 2011, 6, e16957.
(50) Williams, R. J.; Kirby, H. Science 1948, 107, 481.
(21) Wishart, D.; Jewison, T.; Guo, A.; Wilson, M.; Knox, C.; Liu, Y.;
(51) Armstrong, M. D.; Shaw, K. F.; Wall, P. E. J. Biol. Chem. 1956,
Djoumbou, Y.; Mandal, R.; Aziat, F.; Dong, E.; Bouatra, S.; Sinelnikov,
218, 293.
I.; Arndt, D.; Xia, J.; Liu, P.; Yallou, F.; Bjorndahl, T.; Perez-Pineiro,
(52) Robinson, A. B.; Robinson, N. E. Methods Mol. Biol. 2011, 708,
R.; Eisner, R.; Allen, F.; Neveu, V.; Greiner, R.; Scalbert, A. Nucleic
Acids Res. 2013, 41, D801. 1.
(22) Kind, T.; Scholtz, M.; Fiehn, O. PLoS One 2009, 4, e5440. (53) Gates, S. C.; Sweeley, C. C. Clin. Chem. 1978, 24, 1663.
(23) Mandal, R.; Guo, A. C.; Chaudary, K. K.; Liu, P.; Yallou, F. S.; (54) Hamilton, P. B. Anal. Chem. 1963, 35, 2055.
Dong, E.; Aziat, F.; Wishart, D. S. Genome Med. 2012, 4, 38. (55) Pitt, W. W. J.; Scott, S. D.; Johnson, W. F.; Jones, G. J. Clin.
(24) Lu, W.; Clasquin, M. F.; Melamud, E.; Amador-Noguez, D.; Chem. 1970, 16, 657.
Caudy, A.; Rabinowitz, J. D. Anal. Chem. 2010, 82, 3212. (56) Majors, R. E. Anal. Chem. 1973, 45, 755.
(25) Saito, N.; Robert, M.; Kitamura, S.; Baran, R.; Soga, T.; Mori, (57) Horváth, C. S.; Lipsky, S. R. J. Chromatogr. Sci. 1969, 7, 109.
H.; Nishioka, T.; Tomita, M. J. Proteome Res. 2006, 5, 1979. (58) Molnar, I.; Horvath, C. J. Chromatogr. 1977, 143, 391.
(26) Chen, Q.; Park, H.-C.; Goligorsky, M. S.; Fischer, S. M.; Gross, (59) Persson, B. A.; Karger, B. L. J. Chromatogr. Sci. 1974, 12, 521.
S. S. PLoS One 2012, 7, e37149. (60) Wittmer, D. P.; Nuessle, N. O.; Haney, W. G. J. Anal. Chem.
(27) Trupp, M.; Zhu, H.; Wikoff, W.; Baillie, R.; Zeng, Z.; Karp, P.; 1975, 47, 1422.
Fiehn, O.; Krauss, R.; Kaddurah-Daouk, R. PLoS One 2012, 7, e38386. (61) Horvath, C.; Melander, W.; Molnar, I.; Molnar, P. Anal. Chem.
(28) Van Duynhoven, J.; Vaughan, E.; Jacobs, D.; Kemperman, R.; 1977, 49, 2295.
Van Velzen, E.; Gross, G.; Roger, L.; Possemiers, S.; Smilde, A.; Doré, (62) McWilliam, I. G.; Dewar, R. A. Nature 1958, 181, 760.
J.; Westerhuis, J.; Van de Wiele, T. Proc. Natl. Acad. Sci. U.S.A. 2011, (63) Lovelock, J. E.; Lipsky, S. R. J. Am. Chem. Soc. 1960, 82, 431.
108 (Suppl.), 4531. (64) Gohlke, R. S. Anal. Chem. 1959, 31, 535.
(29) Van Velzen, E.; Westerhuis, J.; Van Duynhoven, J.; Van Dorsten, (65) Ryhage, R. Anal. Chem. 1964, 36, 759.
F.; Grün, C.; Jacobs, D.; Duchateau, G.; Vis, D.; Smilde, A. J. Proteome (66) Watson, J. T.; Bieman, K. Anal. Chem. 1964, 36, 1135.
Res. 2009, 8, 3317. (67) Pauling, L.; Robinson, A. R.; Teranishi, R.; Cary, P. Proc. Natl.
(30) Brugnara, L.; Vinaixa, M.; Murillo, S.; Samino, S.; Rodriguez, M. Acad. Sci. U.S.A. 1971, 68, 2374.
A.; Beltran, A.; Lerin, C.; Davison, G.; Correig, X.; Novials, A. PLoS (68) Dalgliesh, C. E.; Horning, E. C.; Horning, M. G.; Knox, K. L.;
One 2012, 7, e40600. Yarger, K. Biochem. J. 1966, 101, 792.
(31) Lee, R.; West, D.; Phillips, S.; Britz-McKibbin, P. Anal. Chem. (69) Stalling, D. L.; Gehrke, C. W.; Zumwalt, R. W. Biochem. Biophys.
2010, 82, 2959. Res. Commun. 1968, 31, 616.
(32) Capriotti, A. L.; Caruso, G.; Cavaliere, C.; Foglia, P.; Bizzarri, (70) Horning, E.; Horning, M. G. Methods Med. Res. 1970, 12, 369.
M.; Laganà, A. Chromatographia 2011, 73, 45. (71) Horning, E.; Horning, M. G. J. Chromatogr. Sci. 1971, 9, 129.
(33) Weckwerth, W.; Morgenthal, K. Drug Discovery Today 2005, 10, (72) Jellum, E.; Stokke, O.; Eldjarn, L. Clin. Chem. 1972, 18, 800.
1551. (73) Gaffney, T. E.; Hammar, G. C.; Holmstedt, B.; McMahon, R. E.
(34) Nielsen, J.; Oliver, S. Trends Biotechnol. 2005, 23, 544. Anal. Chem. 1971, 43, 307.
(35) Goodacre, R. J. Nutr. 2007, 137, 259S. (74) Kovats, E. Helv. Chim. Acta 1958, 41, 1915.

Y dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(75) Sauter, A. D.; Betowski, L. D.; Smith, T. R.; Strickler, V. A.; (110) Raamsdonk, L. M.; Teusink, B.; Broadhurst, D.; Zhang, N.;
Beimer, R. G.; Colby, B. N.; Wilkinson, J. E. J. High Resolut. Hayes, A.; Walsh, M. C.; Berden, J. A.; Brindle, K. M.; Kell, D. B.;
Chromatogr. Chromatogr. Commun. 1981, 4, 366. Rowland, J. J.; Westerhoff, H. V; Dam, K.; Oliver, S. G.; Van Dam, K.
(76) Settlage, J.; Jaeger, H. J. Chromatogr. Sci. 1984, 22, 192. Nat. Biotechnol. 2001, 19, 45.
(77) Chamberlin, B. A.; Sweeley, C. C. Clin. Chem. 1987, 33, 572. (111) Fiehn, O. Plant Mol. Biol. 2002, 48, 155.
(78) Kuhara, T.; Shinka, T.; Inoue, Y.; Ohse, M.; Zhen-wei, X.; (112) Dolinski, K.; Botstein, D. Genome Res. 2005, 15, 1611.
Yoshida, I.; Inokuchi, T.; Yamaguchi, S.; Takayanagi, M.; Matsumoto, (113) Sauer, U.; Heinemann, M.; Zamboni, N. Science 2007, 316,
I. J. Chromatogr. B. Biomed. Sci. Appl. 1999, 731, 141. 550.
(79) Tiselius, A. The moving boundary method of studying the (114) Pandey, A.; Mann, M. Nature 2000, 405, 837.
electrophoresis of proteins. Ph.D. Thesis, Almqvist & Wiksell, (115) Tweeddale, H.; Notley-McRobb, L.; Ferenci, T. J. J. Bacteriol.
University of Uppsala, Sweden, 1930. 1998, 180, 5109.
(80) Mikkers, F. E. P.; Everaerts, F. M.; Verheggen, T. P. E. M. J. (116) Oliver, S. G.; Winson, M. K.; Kell, D. B.; Baganz, F. Trends
Chromatogr. 1979, 169, 11. Biotechnol. 1998, 16, 373.
(81) Jorgenson, J. W.; Lukacs, K. D. Clin. Chem. 1981, 53, 1298. (117) Tweeddale, H.; Notley-McRobb, L.; Ferenci, T. Redox Rep.
(82) Jorgenson, J. W.; Lukacs, K. D. Clin. Chem. 1981, 27, 1551. 1999, 4, 237.
(83) Pretorius, V.; Hopkins, B. J.; Schieke, J. D. J. Chromatogr. A (118) Nicholson, J. K.; Lindon, J. C.; Holmes, E. Xenobiotica 1999,
1974, 99, 23. 29, 1181.
(84) Terabe, S.; Otsuka, K.; Ichikawa, K.; Tsuchiya, A.; Ando, T. (119) Warne, M. A.; Lenz, E. M.; Osborn, D.; Weeks, J. M.;
Anal. Chem. 1984, 56, 111. Nicholson, J. K. Biomarkers 2000, 5, 56.
(85) Terabe, S.; Ozaki, H.; Otsuka, K.; Ando, T. J. Chromatogr. A (120) Robertson, D. G.; Reily, M. D.; Sigler, R. E.; Wells, D. F.;
1985, 332, 211. Paterson, D. A.; Braden, T. K. Toxicol. Sci. 2000, 57, 326.
(86) Horning, E.; Carroll, D.; Dzidic, I.; Haegele, K.; Horning, M.; (121) Nicholls, A. W.; Nicholson, J. K.; Haselden, J. N.; Waterfield,
Stillwell, R. J. Chromatogr. Sci. 1974, 12, 725. C. J. Biomarkers 2000, 5, 410.
(87) Willoughby, R. C.; Browner, R. F. Anal. Chem. 1984, 56, 2626. (122) Griffin, J. L.; Walker, L. A.; Garrod, S.; Holmes, E.; Shore, R.
(88) Vestal, M. L.; Fergusson, G. J. Anal. Chem. 1985, 57, 2378. F.; Nicholson, J. K. Comp. Biochem. Physiol., Part B: Biochem. Mol. Biol.
(89) Caprioli, M.; Fan, T.; Cottrell, J. S. Anal. Chem. 1986, 58, 2949. 2000, 127, 357.
(90) Bruins, A. P.; Covey, T. R.; Henion, J. D. Anal. Chem. 1987, 59, (123) Gavaghan, C. L.; Holmes, E.; Lenz, E.; Wilson, I. D.;
2642. Nicholson, J. K. FEBS Lett. 2000, 484, 169.
(91) Fenn, J. B.; Mann, M.; Meng, C. K.; Wong, S. F.; Whitehouse, (124) Fiehn, O.; Kopka, J.; Dörmann, P.; Altmann, T.; Trethewey, R.
C. M. Science 1989, 246, 64. N.; Willmitzer, L. Nat. Biotechnol. 2000, 18, 1157.
(92) Rashed, M. S.; Bucknall, M. P.; Little, D.; Awad, A.; Jacob, M.; (125) Potts, B. C.; Deese, A. J.; Stevens, G. J.; Reily, M. D.;
Alamoudi, M.; Alwattar, M.; Ozand, P. T. Clin. Biochem. Rev. 1997, 43, Robertson, D. G.; Theiss, J. J. Pharm. Biomed. Anal. 2001, 26, 463.
1129. (126) Keun, H. C.; Ebbels, T. M. D.; Antti, H.; Bollard, M. E.;
(93) Chace, D. H.; Kalas, T. A.; Naylor, E. W. Clin. Chem. 2003, 49, Beckonert, O.; Schlotterbeck, G.; Senn, H.; Niederhauser, U.; Holmes,
1797. E.; Lindon, J. C.; Nicholson, J. K. Chem. Res. Toxicol. 2002, 15, 1380.
(94) Shushan, B. Mass Spectrom. Rev. 2010, 29, 930. (127) Brindle, J. T.; Antti, H.; Holmes, E.; Tranter, G.; Nicholson, J.
(95) Maxwell, E. J.; Chen, D. D. Y. Anal. Chim. Acta 2008, 627, 25. K.; Bethell, H. W.; Clarke, S.; Schofield, P. M.; McKilligin, E.;
(96) Smith, R. D.; Barinaga, C. J.; Udseth, H. R. Anal. Chem. 1988, Mosedale, D. E.; Grainger, D. J. Nat. Med. 2002, 8, 1439.
60, 1948. (128) Wong, A.; Jiménez, B.; Li, X.; Holmes, E.; Nicholson, J. K.;
(97) Nicholson, J. K.; Buckingham, M. J.; Sadler, P. J. Biochem. J. Lindon, J. C.; Sakellariou, D. Anal. Chem. 2012, 84, 3843.
1983, 211, 605. (129) Fan, T. W.; Lane, A. N.; Higashi, R. M. Russ. J. Plant Physiol.
(98) Bales, J. R.; Higham, D. P.; Howe, I.; Nicholson, J. K.; Sadler, P. 2003, 50, 879.
J. Clin. Chem. 1994, 30, 426. (130) Shaniah, N.; Desilva, M. A.; Gowda, G. A. N.; Raferty, M. A.;
(99) Cloarec, O.; Campbell, A.; Tseng, L. H.; Braumann, U.; Spraul, Hainline, B. E.; Raferty, D. Proc. Natl. Acad. Sci. U.S.A. 2007, 104,
M.; Scarfe, G.; Weaver, R.; Nicholson, J. K. Anal. Chem. 2007, 79, 11540.
3304. (131) Viant, M. R. Biochem. Biophys. Res. Commun. 2003, 310, 943.
(100) Jayawickrama, D.; Sweedler, J. J. Chromatogr. A. 2003, 1000, (132) Aharoni, A.; Ric de Vos, C.; Verhoeven, H.; Maliepaard, C.;
819. Kruppa, G.; Bino, R.; Goodenowe, D. Omics 2002, 6, 217.
(101) Williams, R.; Lenz, E.; Wilson, A.; Granger, J.; Wilson, I.; (133) Goodacre, R.; Vaidyanathan, S.; Bianchi, G.; Kell, D. B. Analyst
Major, H.; Stumpf, C.; Plumb, R. Mol. BioSyst. 2006, 2, 174. 2002, 127, 1457.
(102) Kind, T.; Tolstikov, V.; Fiehn, O.; Weiss, R. H. Anal. Biochem. (134) Kind, T.; Fiehn, O. BMC Bioinf. 2006, 7, 234.
2007, 363, 185. (135) Han, J.; Danell, R. M.; Patel, J. R.; Gumerov, D. R.; Scarlett, C.
(103) Ong, E. S.; Chor, C. F.; Zou, L.; Ong, C. N. Mol. BioSyst. 2009, O.; Speir, J. P.; Parker, C. E.; Rusyn, I.; Zeisel, S.; Borchers, C. H.
5, 288. Metabolomics 2008, 4, 128.
(104) Nevedomskaya, E.; Mayborada, O. A.; Deelder, A. M. Mol. (136) Kumari, S.; Stevens, D.; Kind, T.; Denkert, C.; Fiehn, O. Anal.
BioSyst. 2011, 7, 3214. Chem. 2011, 83, 5895.
(105) Kusano, M.; Redestig, H.; Hirai, T.; Oikawa, A.; Matsuda, F.; (137) Tolstikov, V.; Fiehn, O. Anal. Biochem. 2002, 301, 298.
Fukushima, A.; Arita, M.; Watanabe, S.; Yano, M.; Hiwasa-Tanase, K.; (138) Plumb, R. S.; Stumpf, C. L.; Gorenstein, M. V; Castro-Perez, J.
Ezura, H.; Saito, K. PLoS One 2011, 6, e16989. M.; Dear, G. J.; Anthony, M.; Sweatman, B. C.; Connor, S. C.;
(106) Schlotterbeck, G.; Ceccarelli, S. M. Bioanalysis 2009, 1, 549. Haselden, J. N. Rapid Commun. Mass Spectrom. 2002, 16, 1991.
(107) Fleischmann, R. D.; Adams, M. D.; White, O.; Clayton, R. A.; (139) Soga, T.; Ueno, Y.; Naraoka, H.; Ohashi, Y.; Tomita, M.;
Kirkness, E. F.; Kerlavage, A. R.; Bult, C. J.; Tomb, J. F.; Dougherty, B. Nishioka, T. Anal. Chem. 2002, 74, 2233.
A.; Merrick, J. M. Science 1995, 269, 496. (140) Nicholson, J. K.; Connelly, J.; Lindon, J. C.; Holmes, E. M.
(108) Goffeau, A. A.; Barrell, B. G.; Bussey, H.; Davis, R. W.; Dujon, Nat. Rev. Drug Discovery 2002, 1, 153.
B.; Feldmann, H.; Hoheisel, J. D.; Jacq, C.; Johnston, M.; Louis, E. J.; (141) Weckwerth, W.; Fiehn, O. Curr. Opin. Biotechnol. 2002, 13,
Mewes, H. W.; Murakami, Y.; Tettelin, H.; Oliver, S. G. Science 1996, 156.
274, 546. (142) Beckmann, M.; Parker, D.; Enot, D.; Duval, E.; Draper, J. Nat.
(109) Venter, J. C. Science 2001, 291, 1304. Protoc. 2008, 3, 486.

Z dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(143) Lu, W.; Bennett, B. D.; Rabinowitz, J. D. J. Chromatogr. B. (181) Unger, K. K.; Jilge, O.; Kinkel, J. N.; Hearn, M. T. W. J.
2008, 871, 236. Chromatogr. A 1986, 359, 61.
(144) He, Y.; Yu, Z.; Giegling, I.; Xie, L.; Hartmann, A. M.; Prehn, (182) Wilson, I. D.; Nicholson, J. K.; Castro-Perez, J.; Granger, J. H.;
C.; Adamski, J.; Kahn, R.; Li, Y.; Illig, T.; Wang-Sattler, R.; Rujescu, D. Johnson, K. A.; Smith, B. W.; Plumb, R. S. J. Proteome Res. 2005, 4,
Transl. Psychiatry 2012, 2, e149. 591.
(145) Suhre, K.; Meisinger, C.; Döring, A.; Altmaier, E.; Belcredi, P.; (183) MacNair, J. E.; Lewis, K. C.; Jorgenson, J. W. Anal. Chem.
Gieger, C.; Chang, D.; Milburn, M. V; Gall, W. E.; Weinberger, K. M.; 1997, 69, 983.
Mewes, H.-W.; Hrabé de Angelis, M.; Wichmann, H.-E.; Kronenberg, (184) MacNair, J. E.; Patel, K. D.; Jorgenson, J. W. Anal. Chem. 1999,
F.; Adamski, J.; Illig, T. PLoS One 2010, 5, No. e13953. 71, 700.
(146) Abaffy, T.; Duncan, R.; Riemer, D. D.; Tietje, O.; Elgart, G.; (185) Mellors, J. S.; Jorgenson, J. W. Anal. Chem. 2004, 76, 5441.
Milikowski, C.; DeFazio, R. A. PLoS One 2010, 5, No. e13813. (186) Anspach, J. A.; Maloney, T. D.; Colón, L. A. J. Sep. Sci. 2007,
(147) Mondello, L.; Tranchida, P. Q.; Dugo, P.; Dugo, G. Mass 31, 1207.
Spectrom. Rev. 2008, 27, 101. (187) Evans, A. M.; DeHaven, C. D.; Barrett, T.; Mitchell, M.;
(148) Zhang, A.; Sun, H.; Wang, P.; Han, Y.; Wang, X. Analyst 2012, Milgram, E. Anal. Chem. 2009, 81, 6656.
137, 293. (188) Uehara, T.; Yokoi, A.; Aoshima, K.; Tanaka, S.; Kadowaki, T.;
(149) Dunn, W. B.; Broadhurst, D. I.; Atherton, H. J.; Goodacre, R.; Tanaka, M.; Oda, Y. Anal. Chem. 2009, 81, 3836.
Griffin, J. L. Chem. Soc. Rev. 2011, 40, 387. (189) Greibrokk, T.; Andersen, T. J. Chromatogr. A 2003, 1000, 743.
(150) Bedair, M.; Sumner, L. W. TrAC, Trends Anal. Chem. 2008, 27, (190) Plumb, R. S.; Rainville, P.; Smith, B. W.; Johnson, K. A.;
238. Castro-Perez, J.; Wilson, I. D.; Nicholson, J. K. Anal. Chem. 2006, 78,
(151) Patti, G. J. J. Sep. Sci. 2011, 34, 3460. 7278.
(152) Issaq, H. J.; Abbott, E.; Veenstra, T. D. J. Sep. Sci. 2008, 31, (191) Plumb, R.; Mazzeo, J. R.; Grumbach, E. S.; Rainville, P.; Jones,
1936. M.; Wheat, T.; Neue, U. D.; Smith, B.; Johnson, K. A. J. Sep. Sci. 2007,
(153) Novotny, M. V; Soini, H. A.; Mechref, Y. J. Chromatogr. B 31, 1158.
2008, 866, 26. (192) Kirkland, J. J.; Henderson, J. W.; DeStefano, J. J.; Van Straten,
(154) Koek, M. M.; Jellema, R. H.; Van der Greef, J.; Tas, A. C.; M. A.; Claessens, H. A. J. Chromatogr. A 1997, 762, 97.
Hankemeier, T. Metabolomics 2011, 7, 30. (193) Hupp, A. M.; McGuffin, V. L. J. Chromatogr. A 2010, 1217,
(155) Theodoridis, G. A.; Gika, H. G.; Want, E. J.; Wilson, I. D. Anal. 6241.
Chim. Acta 2012, 711, 7. (194) Cunliffe, J. M.; Maloney, T. D. J. Sep. Sci. 2007, 30, 3104.
(156) Wang, X.; Sun, H.; Zhang, A.; Wang, P.; Han, Y. J. Sep. Sci. (195) Brice, R.; Zhang, X.; Colón, L. J. Sep. Sci. 2009, 32, 2723.
2011, 34, 3451. (196) Wilson, I.; Plumb, R.; Granger, J.; Major, H.; Williams, R.;
(157) Spagou, K.; Tsoukali, H.; Raikos, N.; Gika, H.; Wilson, I. D.; Lenz, E. J. Chromatogr. B 2005, 817, 67.
Theodoridis, G. J. Sep. Sci. 2010, 33, 716. (197) Gika, H. G.; Theodoridis, G. A.; Wilson, I. D. J. Sep. Sci. 2008,
(158) Pham-Tuan, H.; Kaskavelis, L.; Daykin, C.; Janssen, H. J. 31, 1598.
Chromatogr. B 2003, 789, 283. (198) Zhang, Y.; Carr, P. J. Chromatogr. A 2011, 1218, 763.
(159) Britz-McKibbin, P. Methods Mol. Biol. 2011, 708, 229. (199) Liu, X.; Pohl, C.; Woodruff, A.; Chen, J. J. Chromatogr. A 2011,
(160) Ramautar, R.; Mayboroda, O. A.; Somsen, G. W.; De Jong, G. 1218, 3407.
J. Electrophoresis 2011, 32, 52. (200) Dorsey, J. G.; Dill, K. A. Chem. Rev. 1989, 89, 331.
(161) Strain, H. H. J. Chem. Educ. 1955, 32, 601. (201) Vailaya, A.; Horváth, C. S. J. Phys. Chem. B 1997, 101, 5875.
(162) Giddings, J. C. Sep. Sci. 1969, 4, 181. (202) Héberger, K. J. Chromatogr. A 2007, 1158, 273.
(163) Bowser, M. T.; Bebault, G. M.; Peng, X.; Chen, D. D. Y. (203) Snyder, L.; Dolan, J.; Carr, P. J. Chromatogr. A 2004, 1060, 77.
Electrophoresis 1997, 18, 2928. (204) Zhang, Y.; Carr, P. W. J. Chromatogr. A 2009, 1216, 6685.
(164) Potts, L.; Stoll, D.; Li, X.; Carr, P. J. Chromatogr. A 2010, 1217, (205) Rafferty, J.; Zhang, L.; Siepmannm, J.; Schure, M. Anal. Chem.
5700. 2007, 79, 6551.
(165) Van Deemter, J. J.; Zuiderweg, F. J.; Klinkenberg, A. Chem. (206) Gómez-Romero, M.; Segura-Carretero, A.; Fernández-
Eng. Sci. 1956, 5, 271. Gutiérrez, A. Phytochemistry 2010, 71, 1848.
(166) Miyabe, K.; Okada, A. Analyst 2002, 127, 1420. (207) Shortreed, M.; Lamos, S.; Frey, B.; Phillips, M.; Patel, M.;
(167) Castello, G.; Moretti, P.; Vezzani, S. J. Chromatogr. A 2009, Belshaw, P.; Smith, L. Anal. Chem. 2006, 78, 6398.
1216, 1607. (208) Santa, T. Biomed. Chromatogr. 2011, 25, 1.
(168) Schellinger, A. P.; Stoll, D. R.; Carr, P. W. J. Chromatogr. A (209) Annesley, T. M. Clin. Chem. 2003, 49, 1041.
2005, 1064, 143. (210) Mallet, C. R.; Lu, Z.; Mazzeo, J. R. Rapid Commun. Mass
(169) Gika, H. G.; Macpherson, E.; Theodoridis, G. A.; Wilson, I. D. Spectrom. 2004, 18, 49.
J. Chromatogr. B 2008, 871, 299. (211) Coulier, L.; Bas, R.; Jespersen, S.; Verheij, E.; Van der Werf, M.
(170) Anderson, J. L.; Armstrong, D. W. Anal. Chem. 2005, 77, 6453. J.; Hankemeier, T. Anal. Chem. 2006, 78, 6573.
(171) Poole, C. F.; Poole, S. K. J. Sep. Sci. 2011, 34, 888. (212) Kiefer, P.; Delmotte, N.; Vorholt, J. A. Anal. Chem. 2011, 83,
(172) Rocha, S. M.; Caldeira, M.; Carrola, J.; Santos, M.; Cruz, N.; 850.
Duarte, I. F. J. Chromatogr. A 2012, 1252, 155. (213) Alpert, A. J. J. Chromatogr. A 1990, 499, 177.
(173) Wilson, R. B.; Siegler, W. C.; Hoggard, J. C.; Fitz, B. D.; (214) Cubbon, S.; Antonio, C.; Wilson, J.; Thomas-Oates, J. Mass
Nadeau, J. S.; Synovec, R. E. J. Chromatogr. A 2011, 1218, 3130. Spectrom. Rev. 2010, 29, 671.
(174) Fekete, S.; Oláh, E.; Fekete, J. J. Chromatogr. A 2012, 1228, 57. (215) Jandera, P. J. Sep. Sci. 2008, 31, 1421.
(175) Gritti, F.; Guiochon, G. J. Chromatogr. A 2012, 1228, 2. (216) Rappold, B. A.; Grant, R. P. J. Sep. Sci. 2011, 34, 3527.
(176) Minakuchi, H.; Nakanishi, K.; Soga, N.; Ishizuka, N.; Tanaka, (217) Nguyen, H. P.; Yang, S. H.; Wigginton, J. G.; Simpkins, J. W.;
N. Anal. Chem. 1996, 68, 3498. Schug, K. A. J. Sep. Sci. 2010, 33, 793.
(177) Tanaka, N.; Kobayashi, H.; Ishizuka, N.; Minakuchi, H.; (218) Yang, S.; Sadilek, M.; Lidstrom, M. E. J. Chromatogr. A 2010,
Nakanishi, K.; Hosoya, K.; Ikegami, T. J. Chromatogr. A 2002, 965, 35. 1217, 7401.
(178) Kobayashi, H.; Ikegami, T.; Kimura, H.; Hara, T.; Tokuda, D.; (219) Pesek, J.; Matyska, M. J. Sep. Sci. 2005, 28, 1845.
Tanaka, N. Anal. Sci. 2006, 22, 491. (220) Pesek, J.; Matyska, M.; Loo, J.; Fischer, S.; Sana, T. J. Sep. Sci.
(179) Ikegami, T.; Dicks, E.; Kobayashi, H.; Morisaka, H.; Tokuda, 2009, 32, 2200.
D.; Cabrera, K.; Hosoya, K.; Tanaka, N. J. Sep. Sci. 2004, 27, 1292. (221) Callahan, D.; De Souza, D.; Bacic, A.; Roessner, U. J. Sep. Sci.
(180) Guiochon, G. J. Chromatogr. A 2007, 1168, 101. 2009, 32, 2273.

AA dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(222) Zhang, T.; Creek, D.; Barrett, M.; Blackburn, G.; Watson, D. (265) Soga, T.; Igarashi, K.; Itoh, C.; Mizobuchi, K.; Zimmermann,
Anal. Chem. 2012, 84, 1994. H.; Tomita, M. Anal. Chem. 2009, 81, 6165.
(223) Frost, N. W.; Jing, M.; Bowser, M. T. Anal. Chem. 2010, 82, (266) Büscher, J. M.; Czernik, D.; Ewald, J. C.; Sauer, U.; Zamboni,
4682. N. Anal. Chem. 2009, 81, 2135.
(224) Zusková, I.; Novotná, A.; Vceláková, K.; Gas, B. J. Chromatogr. (267) Allwood, J. W.; Erban, A.; De Koning, S.; Dunn, W. B.;
B 2006, 841, 129. Luedemann, A.; Lommen, A.; Kay, L.; Löscher, R.; Kopka, J.;
(225) Knox, J. H.; Grant, I. H. Chromatographia 1987, 24, 135. Goodacre, R. Metabolomics 2009, 5, 479.
(226) Terabe, S. Anal. Chem. 2004, 76, 241A. (268) Benton, H.; Want, E.; Keun, H.; Amberg, A.; Plumb, R.;
(227) Bowser, M.; Chen, D. Electrophoresis 1998, 19, 383. Goldfain-Blanc, F.; Walther, B.; Reily, M.; Lindon, J.; Holmes, E.;
(228) Kaiser, C.; Segui-Lines, G.; D’Amaral, J. C.; Ptolemy, A. S.; Nicholson, J.; Ebbels, T. Anal. Chem. 2012, 84, 2424.
Britz-McKibbin, P. Chem. Commun. 2008, 338. (269) Ward, J. L.; Baker, J. M.; Miller, S. J.; Deborde, C.; Maucourt,
(229) Fei, F.; Britz-McKibbin, P. Anal. Bioanal. Chem. 2010, 398, M.; Biais, B.; Rolin, D.; Moing, A.; Moco, S.; Vervoort, J.; Lommen,
1349. A.; Schäfer, H.; Humpfer, E.; Beale, M. H. Metabolomics 2010, 6, 263.
(230) Britz-McKibbin, P.; Chen, D. J. Chromatogr. A 1997, 781, 23. (270) Soga, T.; Heiger, D. N. Anal. Chem. 2000, 72, 1236.
(231) Terabe, S.; Otsuka, K.; Ando, T. Anal. Chem. 1985, 57, 834. (271) Chalcraft, K. R.; Lee, R.; Mills, C.; Britz-McKibbin, P. Anal.
(232) Nishi, H.; Fukuyama, T.; Terabe, S. J. Chromatogr. A 1991, Chem. 2009, 81, 2506.
553, 503. (272) Kok, M. G. M.; De Jong, G. J.; Somsen, G. W. Electrophoresis
(233) Nemes, P.; Knolhoff, A.; Rubakhin, S.; Sweedler, J. Anal. Chem. 2011, 32, 3016.
2011, 6810. (273) Hirayama, A.; Tomita, M.; Soga, T. Analyst 2012, 137, 5026.
(234) Gas, B.; Kenndler, E. Electrophoresis 2000, 21, 3888. (274) Ramautar, R.; Busnel, J.-M.; Deelder, A. M.; Mayboroda, O. A.
(235) Klampfl, C. W. J. Chromatogr. A 2004, 1044, 131. Anal. Chem. 2012, 84, 885.
(236) Malkin, D. S.; Wei, B.; Fogiel, A. J.; Staats, S. L.; Wirth, M. J. (275) Wu, Z.; Gao, W.; Phelps, M.; Wu, D.; Miller, D.; Dalton, J.
Anal. Chem. 2010, 82, 2175. Anal. Chem. 2004, 76, 839.
(237) Svec, F. Electrophoresis 2009, 30, S68. (276) Yanes, O.; Tautenhahn, R.; Patti, G.; Siuzdak, G. Anal. Chem.
(238) Hutterer, K. M.; Jorgenson, J. W. Anal. Chem. 1999, 71, 1293. 2011, 83, 2152.
(239) Beckers, J. L.; Bocek, P. Electrophoresis 2003, 24, 518. (277) Zhang, X.; Clausen, M.; Zhao, X.; Zheng, H.; Bertram, H. Anal.
(240) Geiser, L.; Veuthey, J. L. Electrophoresis 2009, 30, 36.
Chem. 2012, 84, 7785.
(241) Amundsen, L. K.; Kokkonen, J. T.; Sirén, H. J. Sep. Sci. 2008,
(278) Cech, N. B.; Enke, C. G. Mass Spectrom. Rev. 2002, 20, 362.
31, 803. (279) O’Farrell, P. H. J. Biol. Chem. 1975, 250, 4007.
(242) Hommerson, P.; Khan, A.; De Jong, G.; Somsen, G. J.
(280) Rabilloud, T.; Chevallet, M.; Luche, S.; Lelong, C. J. Proteomics
Chromatogr. A 2008, 1204, 197.
2010, 73, 2064.
(243) Henley, W.; Wilburn, R.; Crouch, A.; Jorgenson, J. Anal. Chem.
(281) Dickerson, J.; Ramsay, L.; Dada, O.; Cermak, N.; Dovichi, N.
2005, 77, 7024.
Electrophoresis 2010, 31, 2650.
(244) Yeung, K.; Lucy, C. Electrophoresis 1999, 20, 2554.
(282) Wang, T.; Ma, J.; Wu, S.; Sun, L.; Yuan, H.; Zhang, L.; Liang,
(245) Espada, A.; Molina-Martin, M. Drug Discovery Today 2012, 17,
Z.; Zhang, Y. J. Chromatogr. B 2011, 879, 804.
396.
(283) Evans, C.; Jorgenson, J. Anal. Bioanal. Chem. 2004, 378, 1952.
(246) Dovichi, N. J.; Zhang, J. Angew. Chem., Int. Ed. 2000, 39, 4463.
(284) Jia, L.; Tanaka, N.; Terabe, S. Electrophoresis 2005, 26, 3468.
(247) Sugimoto, M.; Hirayama, A.; Ishikawa, T.; Robert, M.; Baran,
(285) Metz, T. O.; Page, J. S.; Baker, E. S.; Tang, K.; Ding, J.; Shen,
R.; Uehara, K.; Kawai, K.; Soga, T.; Tomita, M. Metabolomics 2010, 6,
Y.; Smith, R. D. Trends Anal. Chem. 2008, 27, 205.
27.
(286) Want, E. J.; Wilson, I. D.; Gika, H.; Theodoridis, G.; Plumb, R.
(248) Jumppanen, J. H.; Riekkola, M.-L. Anal. Chem. 1995, 67, 1060.
(249) Shalaeva, M.; Kenseth, J.; Lombardo, F.; Bastin, A. J. Pharm. S.; Shockcor, J.; Holmes, E.; Nicholson, J. K. Nat. Protoc. 2010, 5,
Sci. 2008, 97, 2581. 1005.
(250) Colton, I.; Carbeck, J.; Rao, J.; Whitesides, G. M. Electro- (287) Koek, M. M.; Muilwijk, B.; Van Stee, L. L. P.; Hankemeier, T.
phoresis 1998, 19, 367. J. Chromatogr. A 2008, 1186, 420.
(251) Gavina, J.; Mazhab-Jafari, M.; Melacini, G.; Britz-McKibbin, P. (288) Koek, M.; Van der Kloet, F.; Kleemann, R.; Kooistra, T.;
Biochemistry 2009, 48, 223. Verheij, E.; Hankemeier, T. Metabolomics 2011, 7, 1.
(252) Yang, J.; Bose, S.; Hage, D. S. J. Chromatogr. A 1996, 735, 209. (289) Sugimoto, M.; Kawakami, M.; Robert, M.; Soga, T.; Tomita,
(253) Sugimoto, M.; Hirayama, A.; Robert, M.; Abe, S.; Soga, T.; M. Curr. Bioinf. 2012, 7, 96.
Tomita, M. Electrophoresis 2010, 31, 2311. (290) Aberg, K. M.; Alm, E.; Torgrip, R. J. Anal. Bioanal. Chem. 2009,
(254) Ramautar, R.; Mayboroda, O. A.; Derks, R. J.; Van Nieuwkoop, 394, 151.
C.; Van Dissel, J. T.; Somsen, G. W.; Deelder, A. M.; De Jong, G. J. (291) Junot, C.; Madalinski, G.; Tabet, J.; Ezan, E. Analyst 2010, 135,
Electrophoresis 2008, 29, 2714. 2203.
(255) Petersen, N. J.; Hansen, S. H. Electrophoresis 2012, 33, 1021. (292) Chen, H.; Pan, Z.; Talaty, N. N.; Cooks, R. G.; Raftery, D.
(256) Dada, O.; Essaka, D.; Hindsgaul, O.; Palcic, M.; Prendergast, J.; Rapid Commun. Mass Spectrom. 2006, 20, 1577.
Schnaar, R.; Dovichi, N. Anal. Chem. 2011, 83, 1748. (293) Dumas, M.-E.; Maibaum, E.; Teague, C.; Ueshima, H.; Zhou,
(257) Mokaddem, M.; Gareil, P.; Belgaied, J.-E.; Varenne, A. B.; Lindon, J.; Nicholson, J.; Stamler, J.; Elliott, P.; Chan, Q.; Holmes,
Electrophoresis 2009, 30, 1692. E. Anal. Chem. 2006, 78, 2199.
(258) Zamfir, A. D. J. Chromatogr. A 2007, 1159, 2. (294) Lin, L.; Yu, Q.; Yan, X.; Hang, W.; Zheng, J.; Xing, J.; Huang,
(259) Moini, M. Anal. Chem. 2001, 73, 3497. B. Analyst 2010, 135, 2970.
(260) Maxwell, E. J.; Zhong, X.; Chen, D. D. Anal. Chem. 2010, 82, (295) Stahnke, H.; Kittlaus, S.; Kempe, G.; Hemmerling, C.; Alder, L.
8377. J. Mass Spectrom. 2012, 47, 875.
(261) Ohashi, Y.; Hirayama, A.; Ishikawa, T.; Nakamura, S.; Shimizu, (296) Hinterwirth, H.; Lämmerhofer, M.; Preinerstorfer, B.; Gargano,
K.; Ueno, Y.; Tomita, M.; Soga, T. Mol. BioSyst. 2008, 4, 135. A.; Reischl, R.; Bicker, W.; Trapp, O.; Brecker, L.; Lindner, W. J. Sep.
(262) Kuehnbaum, N. L.; Britz-McKibbin, P. Anal. Chem. 2011, 83, Sci. 2010, 33, 3273.
8063. (297) Giuffrida, A.; León, C.; García-Cañas, V.; Cucinotta, V.;
(263) Williams, B.; Cameron, C.; Workman, R.; Broeckling, C.; Cifuentes, A. Electrophoresis 2009, 30, 1734.
Sumner, L.; Smith, J. Electrophoresis 2007, 28, 1371. (298) Waldhier, M. C.; Almstetter, M. F.; Nürnberger, N.; Gruber,
(264) Chalcraft, K. R.; Britz-McKibbin, P. Anal. Chem. 2009, 81, 307. M. A.; Dettmer, K.; Oefner, P. J. J. Chromatogr. A 2011, 1218, 4537.

AB dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(299) Oglesbee, D.; Sanders, K.; Lacey, J.; Magera, M.; Casetta, B.; (334) Hagiwara, T.; Saito, S.; Ujiie, Y.; Imai, K.; Kakuta, M.; Kadota,
Strauss, K.; Tortorelli, S.; Rinaldo, P.; Matern, D. Clin. Chem. 2008, 54, K.; Terada, T.; Sumikoshi, K.; Shimizu, K.; Nishi, T. Bioinformation
542. 2010, 5, 255.
(300) Wang, M.; Bai, J.; Chen, W.; Ching, C. PLoS One 2010, 5, (335) Albaugh, D.; Hall, L. L.; Hill, D.; Kertesz, T.; Parham, M.;
e15441. Grant, D. J. Chem. Inf. Model 2009, 49, 788.
(301) Stalcup, A. M. Annu. Rev. Anal. Chem. 2010, 3, 341. (336) Creek, D.; Jankevics, A.; Breitling, R.; Watson, D.; Barrett, M.;
(302) Herrero, M.; Ibáñez, E.; Martín-Alvarez, P.; Cifuentes, A. Anal. Burgess, K. Anal. Chem. 2011, 83, 8703.
Chem. 2007, 79, 5071. (337) Boswell, P. G.; Schellenberg, J. R.; Carr, P. W.; Cohen, J. D.;
(303) King, R.; Bonfiglio, R.; Fernandez-Metzler, C.; Miller-Stein, C.; Hegeman, A. D. J. Chromatogr. A 2011, 1218, 6732.
Olah, T. J. Am. Soc. Mass. Spectrom. 2000, 11, 942. (338) Boswell, P. G.; Schellenberg, J. R.; Carr, P. W.; Cohen, J. D.;
(304) Kostiainen, R.; Kauppila, T. J. J. Chromatogr. A 2009, 1216, Hegeman, A. D. J. Chromatogr. A 2011, 1218, 6742.
685. (339) Stein, S.; Babushok, V.; Brown, R.; Linstrom, P. J. Chem. Inf.
(305) Remane, D.; Meyer, M.; Wissenbach, D.; Maurer, H. Rapid Model. 2007, 47, 975.
Commun. Mass Spectrom. 2010, 24, 3103. (340) Schymanski, E. L.; Gallampois, C. M.; Krauss, M.; Meringer,
(306) León-González, Z.; Ferreiro-Vera, C.; Priego-Capote, F.; De M.; Neumann, S.; Schulze, T.; Wolf, S.; Brack, W. Anal. Chem. 2012,
Castro, M. D. L. J. Chromatogr. A 2011, 1218, 3013. 84, 3287.
(307) Vuckovic, D.; Pawliszyn, J. Anal. Chem. 2011, 83, 1944. (341) Hall, L. M. L. H.; Kertesz, T. M.; Hill, D. W.; Sharp, T. R.;
(308) Risticevic, S.; DeEll, J.; Pawliszyn, J. J. Chromatogr. A 2012, Oblak, E. Z.; Dong, Y. W.; Wishart, D. S.; Chen, M.-H.; Grant, D. F. J.
1251, 208. Chem. Inf. Model. 2012, 52, 1222.
(309) Jain, A.; Verma, K. K. Anal. Chim. Acta 2011, 706, 37. (342) Menikarachchi, L.; Cawley, S.; Hill, D.; Hall, L.; Lai, S.; Wilder,
(310) Lancas, F.; Queiroz, M.; Grossi, P.; Olivares, I. J. Sep. Sci. 2009, J.; Grant, D. Anal. Chem. 2012, 84, 9388.
32, 813. (343) Fiehn, O. TrAC, Trends Anal. Chem. 2008, 27, 261.
(311) Lee, R.; Ptolemy, A. S.; Niewczas, L.; Britz-McKibbin, P. Anal. (344) Kaal, E.; Janssen, H.-G. J. Chromatogr. A 2008, 1184, 43.
Chem. 2007, 79, 403. (345) Jung, H.-J. H.; Lee, W. W.-Y.; Yoo, Y. S. Y.; Chung, B. C. B.;
(312) Dwivedi, P.; Schultz, A. J.; Hill, H. H. Int. J. Mass Spectrom. Choi, M. H. M. Clin. Chim. Acta 2010, 411, 818.
2010, 298, 78. (346) Chan, E. C. Y.; Pasikanti, K. K.; Nicholson, J. K. Nat. Protoc.
(313) Bird, S. S.; Sheldon, D. P.; Gathungu, R. M.; Vouros, P.; Kautz, 2011, 6, 1483.
R.; Matson, W.; Kristal, B. S. Anal. Chem. 2012, 84, 9889. (347) Dettmer, K.; Almstetter, M. F.; Appel, I. J.; Nürnberger, N.;
(314) Sumner, L. W.; Amberg, A.; Barrett, D.; Beger, R.; Beale, M.
Schlamberger, G.; Gronwald, W.; Meyer, H. H. D.; Oefner, P. J.
H.; Daykin, C.; Fan, T. W.-M.; Fiehn, O.; Goodacre, R.; Griffin, J. L.;
Electrophoresis 2010, 31, 2365.
Hardy, N.; Higashi, R.; Kopka, J.; Lindon, J. C.; Lane, A. N.; Marriott,
(348) Mohler, R. E.; Tu, B. P.; Dombek, K. M.; Hoggard, J. C.;
P.; Nicholls, A. W.; Reily, M. D.; Viant, M. Metabolomics 2007, 3, 211.
Young, E. T.; Synovec, R. E. J. Chromatogr. A 2008, 1186, 401.
(315) Kind, T.; Fiehn, O. BMC Bioinf. 2007, 8, 105.
(349) Sellick, C. A.; Hansen, R.; Stephens, G. M.; Goodacre, R.;
(316) Matsuda, F.; Shinbo, Y.; Oikawa, A.; Hirai, M.; Fiehn, O.;
Dickson, A. J. Nat. Protoc. 2011, 6, 1241.
Kanaya, S.; Saito, K. PLoS One 2009, 7, e7490.
(350) Mal, M.; Koh, P. K.; Cheah, P. Y.; Chan, E. C. Y. Anal. Bioanal.
(317) Kilgour, D. P.; Mackay, C. L.; Langridge-Smith, P. R.;
O’Connor, P. B. Anal. Chem. 2012, 84, 7431. Chem. 2012, 403, 483.
(318) Weber, R. J. M.; Southam, A. D.; Sommer, U.; Viant, M. R. (351) Warren, C. R.; Aranda, I.; Cano, F. J. Metabolomics 2011, 8,
Anal. Chem. 2011, 83, 3737. 186.
(319) Jankevics, A.; Merlo, M.; De Vries, M.; Vonk, R.; Takano, E.; (352) Butler, J.; Mishur, R.; Bokov, A.; Hakala, K.; Weintraub, S.;
Breitling, R. Metabolomics 2012, 8, 29. Rea, S. PLoS One 2012, 7, e46140.
(320) Stein, S. Anal. Chem. 2012, 84, 7274. (353) McKelvie, J. R.; Yuk, J.; Xu, Y.; Simpson, A. J.; Simpson, M. J.
(321) Courant, F.; Royer, A. L.; Chéreau, S.; Morvan, M. L.; Metabolomics 2009, 5, 84.
Monteau, F.; Antignac, J. P.; Le Bizec, B. Analyst 2012, 137, 4958. (354) Begley, P.; Francis-McIntyre, S.; Dunn, W. B.; Broadhurst, D.
(322) Kind, T.; Wohlgemuth, G.; Lee, D. Y.; Lu, Y.; Palazoglu, M.; I.; Halsall, A.; Tseng, A.; Knowles, J.; Goodacre, R.; Kell, D. B. Anal.
Shahbaz, S.; Fiehn, O. Anal. Chem. 2009, 81, 10038. Chem. 2009, 81, 7038.
(323) Smith, C. A.; O’Maille, G.; Want, E. J.; Qin, C.; Trauger, S. A.; (355) Smart, K. F.; Aggio, R. B. M.; Van Houtte, J. R.; Villas-Bôas, S.
Brandon, T. R.; Custodio, D. E.; Abagyan, R.; Siuzdak, G. Ther. Drug G. Nat. Protoc. 2010, 5, 1709.
Monit. 2005, 27, 747. (356) Castillo, S.; Mattila, I.; Miettinen, J.; Orešič, M.; Hyötyläinen,
(324) Horai, H.; Arita, M.; Kanaya, S.; Nihei, Y.; Ikeda, T.; Suwa, K.; T. Anal. Chem. 2011, 83, 3058.
Ojima, Y.; Tanaka, K.; Tanaka, S.; Aoshima, K.; Oda, Y.; Kakazu, Y.; (357) Sverko, E.; Tomy, G. T.; Marvin, C. H.; Zaruk, D.; Reiner, E.;
Kusano, M.; Tohge, T.; Matsuda, F.; Sawada, Y.; Horai, Y. M.; Helm, P. A.; Hill, B.; McCarry, B. E. Environ. Sci. Technol. 2008, 42,
Nakanishi, H.; Ikeda, K.; Akimoto, N.; Maoka, T.; Takahashi, H.; Ara, 361.
T.; Sakurai, N.; Suzuki, H.; Shibata, D.; Neumann, S.; Iida, T.; Funatsu, (358) Abate, S.; Ahn, Y. G.; Kind, T.; Cataldi, T. R. I.; Fiehn, O.
K.; Matsuura, F.; Soga, T.; Taguchi, R.; Saito, K.; Nishioka, T. J. Mass Rapid Commun. Mass Spectrom. 2010, 24, 1172.
Spectrom. 2010, 45, 703. (359) Nakamura, S.; Sian, T. H.; Daishima, S. J. Chromatogr. A 2001,
(325) Wagner, C.; Sefkow, M.; Kopka, J. Phytochemistry 2003, 62, 19, 275.
887. (360) Rosenfelder, N.; Vetter, W. Mass Spectrom 2009, 23, 3807.
(326) Neumann, S.; Böcker, S. Anal. Bioanal. Chem. 2010, 398, 2779. (361) Fiehn, O.; Kopka, J.; Trethewey, R. N.; Willmitzer, L. Anal.
(327) Wolf, S.; Schmidt, S.; Müller-Hannemann, M.; Neumann, S. Chem. 2000, 72, 3573.
BMC Bioinf. 2010, 22, 148. (362) Koek, M. M.; Bakels, F.; Engel, W.; Van den Maagdenberg, A.;
(328) Wishart, D. S. Bioanalysis 2011, 3, 1769. Ferrari, M. D.; Coulier, L.; Hankemeier, T. Anal. Chem. 2010, 82, 156.
(329) Hill, D. W.; Kertesz, T. M.; Fontaine, D.; Friedman, R.; Grant, (363) Koek, M. M.; Muilwijk, B.; Van der Werf, M. J.; Hankemeier,
D. F. Anal. Chem. 2008, 80, 5574. T. Anal. Chem. 2006, 8, 1272.
(330) Kind, T.; Fiehn, O. Bioanal. Rev. 2010, 2, 23. (364) Little, J. L. J. Chromatogr. A 1999, 84, 1.
(331) Molnar, I. J. Chromatogr. A 2002, 965, 175. (365) Gu, Q.; David, F.; Lynen, F.; Rumpel, K.; Dugardeyn, J.; Van
(332) Hruska, V.; Jaros, M.; Gas, B. Electrophoresis 2006, 27, 984. Der Straeten, D.; Xu, G.; Sandra, P. J. Chromatogr. A 2011, 1218, 3247.
(333) Thormann, W.; Breadmore, M. C.; Caslavska, J.; Mosher, R. A. (366) Corey, E. J.; Venkateswarlu, A. J. Am. Chem. Soc. 1972, 94,
Electrophoresis 2010, 31, 726. 6190.

AC dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(367) Danielsson, A. P. H.; Moritz, T.; Mulder, H.; Spégel, P. (403) Zelena, E.; Dunn, W. B. W. B.; Broadhurst, D.; Francis-
Metabolomics 2012, 8, 50. McIntyre, S.; Carroll, K. K. M.; Begley, P.; O’Hagan, S.; Knowles, J. J.
(368) Huang, X.; Regnier, F. E. Anal. Chem. 2008, 80, 107. D.; Halsall, A. A.; HUSERMET Consortium; Wilson, I.; Kell, D. B. D.
(369) Lien, S. K.; Kvitvang, H. F.; Bruheim, P. J. Chromatogr. A 2012, B.; Wilson, I. D. Anal. Chem. 2009, 81, 1357.
1274, 118. (404) Nordstrom, A.; Want, E.; Northen, T.; Lehtio, J.; Siuzdak, G.
(370) Hong, Z.; Lin, Z.; Liu, Y.; Tan, G.; Lou, Z.; Zhu, Z.; Chai, Y.; Anal. Chem. 2008, 80, 421.
Fan, G.; Zhang, J.; Zhang, L. J. Chromatogr. A 2012, 1254, 14. (405) Neue, U. D.; Tran, K. V.; Iraneta, P. C.; Alden, B. A. J. Sep. Sci.
(371) Liebeke, M.; Wunder, A.; Lalk, M. Anal. Biochem. 2010, 401, 2003, 26, 174.
312. (406) Kamleh, M. A.; Ebbels, T. M.; Spagou, K.; Masson, P.; Want,
(372) Kouremenos, K. A.; Harynuk, J. J.; Winniford, W. L.; Morrison, E. J. Anal. Chem. 2012, 84, 2670.
P. D.; Marriott, P. J. J. Chromatogr. B 2010, 878, 1761. (407) Gika, H. G.; Theodoridis, G. A.; Wilson, I. D. J. Chromatogr. A
(373) Noctor, G.; Bergot, G.; Mauve, C.; Thominet, D.; Lelarge- 2008, 1189, 314.
Trouverie, C.; Priou, J. Metabolomics 2005, 3, 161. (408) t’Kindt, R.; De Veylder, L.; Storme, M.; Deforce, D.; Van
(374) Villas-Bôas, S. G.; Delicado, D. G.; Akesson, M.; Nielsen, J. Bocxlaer, J. J. Chromatogr. B 2008, 871, 37.
Anal. Biochem. 2003, 322, 134. (409) Eliasson, M.; Rännar, S.; Madsen, R.; Donten, M.; Marsden-
(375) Gao, X.; Pujos-Guillot, E.; Martin, J.-F.; Galan, P.; Juste, C.; Jia, Edwards, E.; Moritz, T.; Shockcor, J.; Johansson, E.; Trygg, J. Anal.
W.; Sebedio, J.-L. Anal. Biochem. 2009, 393, 163. Chem. 2012, 84, 6869.
(376) Tao, X.; Liu, Y.; Wang, Y.; Qiu, Y.; Lin, J.; Zhao, A.; Su, M.; Jia, (410) Wikoff, W. R.; Gangoiti, J. A.; Barshop, B. A.; Siuzdak, G. Clin.
W. Anal. Bioanal. Chem. 2008, 391, 2881. Chem. 2007, 53, 2169.
(377) Han, L.-D.; Xia, J.-F.; Liang, Q.-L.; Wang, Y.-M. Y.; Hu, P.; Li, (411) Bruce, S. J.; Jonsson, P.; Antti, H.; Cloarec, O.; Trygg, J.;
P.; Luo, G.-A. Anal. Chim. Acta 2011, 689, 85. Marklund, S. L.; Moritz, T. Anal. Biochem. 2008, 372, 237.
(378) Villas-Bôas, S. G.; Smart, K. F.; Sivakumaran, S.; Lane, G. A. (412) Gao, X.; Zhang, Q.; Meng, D.; Isaac, G.; Zhao, R.; Fillmore, T.
Metabolites 2011, 1, 3. L.; Chu, R. K.; Zhou, J.; Tang, K.; Hu, Z.; Moore, R. J.; Smith, R. D.;
(379) Vincenti, M.; Fasano, F.; Valsania, M. C.; Guarda, P.; Katze, M. G.; Metz, T. O. Anal. Bioanal. Chem. 2012, 402, 2923.
Richardson, S. D. Anal. Bioanal. Chem. 2010, 397, 43. (413) Bell, D. S.; Cramer, H. M.; Jones, A. D. J. Chromatogr. A 2005,
(380) Hušek, P.; Šimek, P.; Hartvich, P.; Zahradníčková, H. J. 1095, 113.
Chromatogr. A 2008, 1186, 391. (414) Gika, H. G.; Theodoridis, G.; Extance, J.; Edge, A. M.; Wilson,
(381) Kanani, H.; Chrysanthopoulos, P. K.; Klapa, M. I. J. I. D. J. Chromatogr. B 2008, 871, 279.
Chromatogr. B 2008, 871, 191. (415) Stoll, D.; Wang, X.; Carr, P. Anal. Chem. 2008, 80, 268.
(382) Zhang, Z.; Pawliszyn, J. Anal. Chem. 1993, 65, 1843. (416) D’Attoma, A.; Grivel, C.; Heinisch, S. J. Chromatogr. A 2012,
(383) Musteata, F. M.; Pawliszyn, J. TrAC Trends Anal. Chem. 2007, 1262, 148.
26, 36. (417) Stoll, D. R. Anal. Bioanal. Chem. 2010, 397, 979.
(384) Vas, G.; Vekey, K. J. Mass Spectrom. 2004, 39, 233. (418) Fairchild, J. N.; Horvath, K.; Gooding, J. R.; Campagna, S. R.;
(385) Llop, A.; Pocurull, E.; Borrull, F. J. Sep. Sci. 2011, 34, 1531. Guiochon, G. J. Chromatogr. A 2010, 1217, 8161.
(386) Penn, D. J.; Oberzaucher, E.; Grammer, K.; Fischer, G.; Soini, (419) Guo, K.; Peng, J.; Zhou, R.; Li, L. J. Chromatogr. A 2011, 1218,
H. A.; Wiesler, D.; Novotny, M. V.; Dixon, S. J.; Xu, Y.; Brereton, R. G. 3689.
J. R. Soc., Interface 2007, 4, 331. (420) Liu, Y.; Xue, X.; Guo, Z.; Xu, Q.; Zhang, F.; Liang, X. J.
(387) Kaur, N.; Cabral, J. L.; Morin, A.; Waldron, K. C. J. Chromatogr. A 2008, 1208, 133.
Chromatogr. A 2011, 1218, 324. (421) Zheng, J.; Dixon, R. A.; Li, L. Anal. Chem. 2012, 84, 10802.
(388) Tikunov, Y.; Lommen, A.; De Vos, C. H.; Verhoeven, H. A.; (422) Dunn, W. B.; Broadhurst, D.; Brown, M.; Baker, P. N.;
Bino, R. J.; Hall, R. D.; Bovy, A. G. Plant Physiol. 2005, 139, 1125. Redman, C. W. G.; Kenny, L. C.; Kell, D. B. J. Chromatogr. B 2008,
(389) Aprea, E.; Gika, H.; Carlin, S.; Theodoridis, G.; Vrhovsek, U.; 871, 288.
Mattivi, F. J. Chromatogr. 2011, 1218, 4517. (423) Ciborowski, M.; Teul, J.; Martin-Ventura, J. L.; Egido, J.;
(390) Shirasu, M.; Touhara, K. J. Biochem. 2011, 150, 257. Barbas, C. PLoS One 2012, 7, e31982.
(391) Silva, C. L.; Passos, M.; Câmara, J. S. Br. J. Cancer 2011, 105, (424) Roux, A.; Xu, Y.; Heilier, J.; Olivier, M.; Ezan, E.; Tabet, J.;
1894. Junot, C. Anal. Chem. 2012, 84, 6429.
(392) Zhang, S.; Liu, L.; Steffen, D.; Ye, T.; Raftery, D. Metabolomics (425) Haug, K.; Salek, R.; Conesa, P.; Hastings, J.; De Matos, P.;
2011, 8, 323. Rijnbeek, M.; Mahendraker, T.; Williams, M.; Neumann, S.; Rocca-
(393) Phillips, M.; Cataneo, R. N.; Ditkoff, B. A.; Fisher, P.; Serra, P.; Maguire, E.; González-Beltrán, A.; Sansone, S.; Griffin, J.;
Greenberg, J.; Gunawardena, R.; Kwon, C. S.; Tietje, O.; Wong, C. Steinbeck, C. Nucleic Acids Res. 2012, 47, D781.
Breast Cancer Res. 2006, 99, 19. (426) Cubbon, S.; Bradbury, T.; Wilson, J.; Thomas-Oates, J. Anal.
(394) Matsumura, K.; Opiekun, M.; Oka, H.; Vachani, A.; Albelda, S.; Chem. 2007, 79, 8911.
Yamazaki, K.; Beauchamp, G. PLoS One 2010, 5, e8819. (427) De Carvalho, L.; Zhao, H.; Dickinson, C.; Arango, N.; Lima,
(395) McCulloch, M.; Jezierski, T.; Broffman, M.; Hubbard, A.; C.; Fischer, S.; Ouerfelli, O.; Nathan, C.; Rhee, K. Chem. Biol. 2010,
Turner, K. Integr. Cancer Ther. 2006, 5, 30. 17, 323.
(396) Delahunty, C.; Eyres, G.; Dufour, J. J. Chromatogr. A 2008, (428) Tsutsui, H.; Maeda, T.; Min, J. Z.; Inagaki, S.; Higashi, T.;
1186, 123. Kagawa, Y.; Toyo’oka, T. Clin. Chim. Acta 2011, 412, 861.
(397) d’Acampora Zellner, B.; Dugo, P.; Dugo, G.; Mondello, L. J. (429) Lv, H.; Palacios, G.; Hartil, K.; Kurland, I. J. J. Proteome Res.
Chromatogr. A 2008, 1186, 123. 2011, 10, 2104.
(398) Hufnagel, J. C.; Hofmann, T. J. Agric. Food Chem. 2008, 56, (430) Chen, L.; Zhou, L.; Chan, E. C. Y.; Neo, J.; Beuerman, R. W. J.
9190. Proteome Res. 2011, 10, 4876.
(399) Lo, Y.; Koziel, J.; Cai, L.; Hoff, S.; Jenks, W.; Xin, H. J. Environ. (431) Lin, L.; Huang, Z.; Gao, Y.; Yan, X.; Xing, J.; Hang, W. J.
Qual. 2008, 37, 521. Proteome Res. 2011, 10, 1396.
(400) Laor, Y.; Koziel, J.; Cai, L.; Ravid, U. J. Air Waste Manage. (432) García-Cañaveras, J.; Donato, M.; Castell, J.; Lahoz, A. J.
Assoc. 2008, 58, 1187. Proteome Res. 2011, 10, 4825.
(401) Stensmyr, M. C.; Urru, I.; Collu, I.; Celander, M.; Hansson, B. (433) O’Maille, G.; Go, E. P.; Hoang, L.; Want, E. J.; Smith, C.;
S.; Angioy, A.-M. Nature 2002, 420, 625. O’Maille, P.; Nordstrom, A.; Morita, H.; Qin, C.; Uritboonthai, W.;
(402) Shirasu, M.; Nagai, S.; Hayashi, R.; Ochiai, A.; Touhara, K. Apon, J.; Moore, R.; Garrett, J.; Siuzdak, G. Spectroscopy 2008, 22, 327.
Biosci., Biotechnol., Biochem. 2009, 73, 2117. (434) Carlson, E.; Cravatt, B. Nat. Methods 2007, 4, 429.

AD dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

(435) Soukup-Hein, R. J.; Remsburg, J.; Dasgupta, P.; Armstrong, D. (468) Diamandis, E. P. BMC Med. 2012, 10, 87.
Anal. Chem. 2007, 79, 7346. (469) Sreekumar, A.; Poisson, L.; Rajendiran, T.; Khan, A.; Cao, Q.;
(436) Woo, H. K.; Go, E. P.; Hoang, L.; Trauger, S. A.; Bowen, B.; Yu, J.; Laxman, B.; Mehra, R.; Lonigro, R.; Li, Y.; Nyati, M.; Ahsan, A.;
Siuzdak, G.; Northen, T. R. Rapid Commun. Mass Spectrom. 2009, 23, Kalyana-Sundaram, S.; Han, B.; Cao, X.; Byun, J.; Omenn, G.; Ghosh,
1849. D.; Pennathur, S.; Alexander, D.; Berger, A.; Shuster, J.; Wei, J.;
(437) Yang, W.; Adamec, J.; Regnier, F. Anal. Chem. 2007, 79, 5150. Varambally, S.; Beecher, C.; Chinnaiyan, A. Nature 2009, 457, 910.
(438) Guo, K.; Li, L. Anal. Chem. 2009, 81, 3919. (470) Stephan, C.; Jentzmik, F.; Jung, K. Eur. Urol. 2010, 58, 19.
(439) Barbas, C.; Moraes, E. P.; Villaseñor, A. J. Pharm. BIomed. Anal. (471) Bollard, M.; Holmes, E.; Lindon, J.; Mitchell, S.; Branstetter,
2011, 55, 823. D.; Zhang, W.; Nicholson, J. K. Anal. Biochem. 2001, 295, 194.
(440) Soga, T.; Ohashi, Y.; Ueno, Y.; Naraoka, H.; Tomita, M.; (472) Plumb, R.; Granger, J.; Stumpf, C.; Wilson, I.; Evans, J.; Lenz,
Nishioka, T. J. Proteome Res. 2003, 2, 488. E. Analyst 2003, 128, 819.
(441) Lee, R.; Britz-McKibbin, P. Anal. Chem. 2009, 81, 7047. (473) Shi, H.; Vigneau-Callahan, K. E.; Shestopalov, A. I.; Milbury, P.
(442) Ramautar, R.; Nevedomskaya, E.; Mayboroda, O.; Deelder, A.; E.; Matson, W. R.; Kristal, B. S. J. Nutr. 2002, 132, 1039.
Wilson, I.; Gika, H.; Theodoridis, G.; Somsen, G.; De Jong, G. Mol. (474) Hutterer, F.; Roboz, J.; Sarkozi, L.; Ruhig, A.; Bacchin, P. Clin.
BioSyst. 2011, 7, 194. Chem. 1971, 17, 789.
(443) Nevedomskaya, E.; Ramautar, R.; Derks, R.; Westbroek, I.; (475) Dallmann, R.; Viola, A. U.; Tarokh, L.; Cajochen, C.; Brown, S.
Zondag, G.; Pluijm, I.; Deelder, A. M.; Mayboroda, O. A.; Van der A. Proc. Natl. Acad. Sci. U.S.A. 2012, 108, 2625.
Pluijm, I. J. Proteome Res. 2010, 9, 4869. (476) Szeto, S.; Reinke, S.; Lemire, B. J. Biomol. NMR 2011, 49, 245.
(444) Nemes, P.; Knolhoff, A.; Rubakhin, S.; Sweedler, J. ACS Chem. (477) Friswell, M. K.; Gika, H.; Stratford, I. J.; Theodoridis, G.;
Neurosci. 2012, 3, 782. Telfer, B.; Wilson, I. D.; McBain, A. J. PLoS One 2010, 5, e8584.
(445) Busnel, J.-M.; Schoenmaker, B.; Ramautar, R.; Carrasco- (478) Nicholson, G.; Rantalainen, M.; Maher, A.; Li, J.; Malmodin,
Pancorbo, A.; Ratnayake, C.; Feitelson, J. S.; Chapman, J. D.; Deelder, D.; Ahmadi, K.; Faber, J.; Hallgrímsdóttir, I.; Barrett, A.; Toft, H.;
A. M.; Mayboroda, O. A. Anal. Chem. 2010, 82, 9476. Krestyaninova, M.; Viksna, J.; Neogi, S.; Dumas, M.; Sarkans, U.;
(446) Bello, M.; Capelli, L.; Righetti, P. J. Chromatogr. A 1994, 684, Silverman, B.; Donnelly, P.; Nicholson, J.; Allen, M.; Zondervan, K.;
311. Lindon, J.; Spector, T.; McCarthy, M.; Holmes, E.; Baunsgaard, D.;
(447) Ramautar, R.; Toraño, J. S.; Somsen, G. W.; De Jong, G. J. Holmes, C. Mol. Syst. Biol. 2011, 7, 525.
Electrophoresis 2010, 31, 2319. (479) Crews, B.; Wikoff, W.; Patti, G.; Woo, H.; Kalisiak, E.;
(448) Grundmann, M.; Matysik, F. Anal. Bioanal. Chem. 2011, 400, Heideker, J.; Siuzdak, G. Anal. Chem. 2009, 81, 8538.
269. (480) Favé, G.; Beckmann, M.; Lloyd, A. J.; Zhou, S.; Harold, G.; Lin,
(449) Tanaka, Y.; Higashi, T.; Rakwal, R.; Wakida, S.; Iwahashi, H. W.; Tailliart, K.; Xie, L.; Draper, J.; Mathers, J. C. Metabolomics 2011,
Electrophoresis 2008, 29, 2016.
7, 469.
(450) Harada, K.; Ohyama, Y.; Tabushi, T.; Kobayashi, A.; Fukusaki,
(481) Van Velzen, E.; Westerhuis, J.; Van Duynhoven, J.; Van
E. J. Biosci. Bioeng. 2008, 105, 249.
Dorsten, F.; Hoefsloot, H.; Jacobs, D.; Smit, S.; Draijer, R.; Kroner, C.;
(451) Lee, R.; Britz-McKibbin, P. Electrophoresis 2010, 31, 2328.
Smilde, A. J. Proteome Res. 2008, 7, 4483.
(452) Soga, T.; Sugimoto, M.; Honma, M.; Mori, M.; Igarashi, K.;
(482) Van der Greef, J.; Hankemeier, T.; McBurney, R. N.
Kashikura, K.; Ikeda, S.; Hirayama, A.; Yamamoto, T.; Yoshida, H.;
Pharmacogenomics 2006, 7, 1087.
Otsuka, M.; Tsuji, S.; Yatomi, Y.; Sakuragawa, T.; Watanabe, H.; Nihei,
(483) Krug, S.; Kastenmüller, G.; Stückler, F.; Rist, M. J. M.; Skurk,
K.; Saito, T.; Kawata, S.; Suzuki, H.; Tomita, M.; Suematsu, M. J.
Hepatol. 2011, 55, 896. T.; Sailer, M.; Raffler, J.; Römisch-Margl, W.; Adamski, J.; Prehn, C.;
(453) Ramautar, R.; Shyti, R.; Schoenmaker, B.; De Groote, L.; Frank, T.; Engel, K. K.-H.; Hofmann, T.; Luy, B.; Zimmermann, R.;
Derks, R. J.; Ferrari, M. D.; Van den Maagdenberg, A. M.; Deelder, A. Moritz, F.; Schmitt-Kopplin, P.; Krumsiek, J.; Kremer, W.; Huber, F.;
M.; Mayboroda, O. A. Anal. Bioanal. Chem. 2012, 404, 2895. Oeh, U.; Theis, F. F. J.; Szymczak, W.; Hauner, H.; Suhre, K.; Daniel,
(454) Simó, C.; Ibáñez, C.; Gómez-Martínez, A.; Ferragut, J. A.; H. FASEB J. 2012, 26, 2607.
Cifuentes, A. Electrophoresis 2011, 32, 1765. (484) Patti, G. J.; Tautenhahn, R.; Siuzdak, G. Nat. Protoc. 2012, 7,
(455) Soga, T.; Ishikawa, T.; Igarashi, S.; Sugawara, K.; Kakazu, Y.; 508.
Tomita, M. J. Chromatogr. A 2007, 1159, 125. (485) Broadhurst, D. I.; Kell, D. B. Metabolomics 2007, 2, 171.
(456) Harada, K.; Fukusaki, E.; Kobayashi, A. J. Biosci. Bioeng. 2006, (486) Vuckovic, D. Anal. Bioanal. Chem. 2012, 403, 1523.
101, 403. (487) Geier, F. M.; Want, E. J.; Leroi, A. M.; Bundy, J. G. Anal. Chem.
(457) Sato, S.; Yanagisawa, S. Metabolomics 2010, 6, 529. 2011, 83, 3730.
(458) Timischl, B.; Dettmer, K.; Kaspar, H.; Thieme, M.; Oefner, P. (488) Saric, J.; Want, E.; Duthaler, U.; Lewis, M.; Keiser, J.;
J. Electrophoresis 2008, 29, 2203. Shockcor, J.; Ross, G.; Nicholson, J.; Holmes, E.; Tavares, M. Anal.
(459) Nilsson, S.; Bylund, D.; Jörntén-Karlsson, M.; Petersson, P.; Chem. 2012, 84, 6963.
Markides, K. Electrophoresis 2004, 25, 2100. (489) Beltran, A.; Suarez, M.; Rodríguez, M. A.; Vinaixa, M.; Samino,
(460) Leon, C.; Rodriguez-Meizoso, I.; Lucio, M.; Garcia-Cañas, V.; S.; Arola, L.; Correig, X.; Yanes, O. Anal. Chem. 2012, 84, 5838.
Ibañez, E.; Schmitt-Kopplin, P.; Cifuentes, A. J. Chromatogr. A 2009, (490) Gullberg, J.; Jonsson, P.; Nordström, A.; Sjöström, M.; Moritz,
1216, 7314. T. Anal. Biochem. 2004, 331, 283.
(461) Yang, W.-C.; Regnier, F. E.; Adamec, J. Electrophoresis 2008, (491) T’Kindt, R.; Morreel, K.; Deforce, D.; Boerjan, W.; Van
29, 4549. Bocxlaer, J. J. Chromatogr. B 2009, 877, 3572.
(462) Jones, D. P. Rejuvenation Res. 2006, 9, 169. (492) Canelas, A.; Ten Pierick, A.; Ras, C.; Seifar, R.; Van Dam, J.;
(463) D’Agostino, L. A.; Lam, K. P.; Lee, R.; Britz-McKibbin, P. J. Van Gulik, W.; Heijnen, J. Anal. Chem. 2009, 81, 7379.
Proteome Res. 2011, 592. (493) Shin, M. H.; Lee do, Y.; Liu, K. H.; Fiehn, O.; Kim, K. H. Anal.
(464) Sugimoto, M.; Koseki, T.; Hirayama, A.; Abe, S.; Sano, T.; Chem. 2010, 82, 6660.
Tomita, M.; Soga, T. J. Agric. Food Chem. 2010, 58, 374. (494) El Rammouz, R.; Létisse, F.; Durand, S.; Portais, J.-C.; Moussa,
(465) Sugimoto, M.; Wong, D. T.; Hirayama, A.; Soga, T.; Tomita, Z. W.; Fernandez, X. Anal. Biochem. 2010, 398, 169.
M. Metabolomics 2010, 6, 78. (495) Smith, L.; Maher, A.; Want, E.; Elliott, P.; Stamler, J.; Hawkes,
(466) Sugimoto, M.; Saruta, J.; Matsuki, C.; To, M.; Onuma, H.; G.; Holmes, E.; Lindon, J.; Nicholson, J. K. Anal. Chem. 2009, 81,
Kaneko, M.; Soga, T.; Tomita, M.; Tsukinoki, K. Metabolomics 2013, 4847.
DOI: 10.1007/s11306-012-0464-y. (496) Rosenling, T.; Stoop, M.; Smolinska, A.; Muilwijk, B.; Coulier,
(467) Ranshoff, D. F. Nat. Rev. 2005, 5, 142. L.; Shi, S.; Dane, A.; Christin, C.; Suits, F.; Horvatovich, P.; Wijmenga,

AE dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX


Chemical Reviews Review

S.; Buydens, L.; Vreeken, R.; Hankemeier, T.; Van Gool, A.; Luider,
T.; Bischoff, R. Clin. Chem. 2011, 57, 1703.
(497) Boernsen, K. O.; Gatzek, S.; Imbert, G. Anal. Chem. 2005, 77,
7255.
(498) Zivkovic, A. M.; Wiest, M. M.; Nguyen, U. T.; Davis, R.;
Watkins, S. M.; German, J. B. Metabolomics 2009, 5, 507.
(499) Duportet, X.; Bastos, R.; Aggio, M.; Carneiro, S.; Villas-Bôas, S.
G. Metabolomics 2012, 8, 41.
(500) Elliott, P.; Peakman, T. C. Int. J. Epidemiol. 2008, 37, 234.
(501) Dunn, W. B.; Broadhurst, D.; Ellis, D. I.; Brown, M.; Halsall,
A.; O’Hagan, S.; Spasic, I.; Tseng, A.; Kell, D. B. Int. J. Epidemiol. 2008,
37, i23.
(502) Want, E. J.; O’Maille, G.; Smith, C. A.; Brandon, T. R.;
Uritboonthai, W.; Qin, C.; Trauger, S. A.; Siuzdak, G.; Uritboonthai,
W. Anal. Chem. 2006, 78, 743.
(503) Masson, P.; Alves, A.; Ebbels, T.; Nicholson, J.; Want, E. Anal.
Chem. 2010, 82, 7779.
(504) Kanani, H. H.; Klapa, M. I. Metab. Eng. 2007, 9, 39.
(505) Fernie, A.; Aharoni, A.; Willmitzer, L.; Stitt, M.; Tohge, T.;
Kopka, J.; Carroll, A.; Saito, K.; Fraser, P.; DeLuca, V. Plant Cell. 2011,
23, 2477.
(506) Van den Berg, R.; Hoefsloot, H.; Westerhuis, J.; Smilde, A.;
Van der Werf, M. BMC Genomics 2006, 7, 142.
(507) Sangster, T.; Major, H.; Plumb, R.; Wilson, A. J.; Wilson, I. D.
Analyst 2006, 131, 1075.
(508) Xia, J.; Mandal, R.; Sinelnikov, I. V.; Broadhurst, D.; Wishart,
D. S. Nucleic Acids Res. 2012, 40, W127.
(509) Tautenhahn, R.; Patti, G. J.; Rinehart, D.; Siuzdak, G. Anal.
Chem. 2012, 84, 5035.
(510) Wei, X.; Shi, X.; Kim, S.; Zhang, L.; Patrick, J.; Binkley, J.;
McClain, C.; Zhang, X. Anal. Chem. 2012, 84, 7963.
(511) Creek, D.; Jankevics, A.; Burgess, K.; Breitling, R.; Barrett, M.
Bioinformatics 2012, 28, 1048.
(512) Pluskal, T.; Castillo, S.; Villar-Briones, A.; Orešič, M. BMC
Bioinf. 2010, 11, 395.
(513) Melamud, E.; Vastag, L.; Rabinowitz, J. D. Anal. Chem. 2010,
82, 9818.
(514) Dragsted, L. O. Metabolites 2012, 2, 77.
(515) Hendriks, M. M. W. B.; Van Eeuwijk, F. A.; Jellema, R. H.;
Westerhuis, J. A.; Reijmers, T. H.; Hoefsloot, H. C. J.; Smilde, A. K.
TrAC, Trends Anal. Chem. 2011, 30, 1685.
(516) Hrydziuszko, O.; Viant, M. R. Metabolomics 2012, 8, 161.
(517) Duarte, N. C.; Becker, S. A.; Jamshidi, N.; Thiele, I.; Mo, M. L.;
Vo, T. D.; Srivas, R.; Palsson, B. Ø. Proc. Natl. Acad. Sci. U.S.A. 2007,
104, 1777.
(518) Zhang, G.-F.; Sadhukhan, S.; Tochtrop, G. P.; Brunengraber,
H. J. Biol. Chem. 2011, 286, 23631.
(519) Bordbar, A.; Mo, M. L.; Nakayasu, E. S.; Schrimpe-Rutledge, A.
C.; Kim, Y.-M.; Metz, T. O.; Jones, M. B.; Frank, B. C.; Smith, R. D.;
Peterson, S. N.; Hyduke, D. R.; Adkins, J. N.; Palsson, B. Ø. Mol. Syst.
Biol. 2012, 8, 558.
(520) Ishii, N.; Nakahigashi, K.; Baba, T.; Robert, M.; Soga, T.;
Kanai, A.; Hirasawa, T.; Naba, M.; Hirai, K.; Hoque, A.; Ho, P.;
Kakazu, Y.; Sugawara, K.; Igarashi, S.; Harada, S.; Masuda, T.;
Sugiyama, N.; Togashi, T.; Hasegawa, M.; Takai, Y.; Yugi, K.; Arakawa,
K.; Iwata, N.; Toya, Y.; Nakayama, Y.; Nishioka, T.; Shimizu, K.; Mori,
H.; Tomita, M. Science 2007, 316, 593.
(521) Ptolemy, A. S.; Rifai, N. Scand. J. Clin. Lab. Invest., Suppl. 2010,
242, 6.
(522) Anderson, N. L.; Ptolemy, A. S.; Rifai, N. Clin. Chem. 2013, 59,
194.

AF dx.doi.org/10.1021/cr300484s | Chem. Rev. XXXX, XXX, XXX−XXX

Potrebbero piacerti anche