You are on page 1of 16

Analytica Chimica Acta 870 (2015) 29–44

Contents lists available at ScienceDirect

Analytica Chimica Acta


journal homepage: www.elsevier.com/locate/aca

Tutorial

Tutorial review on validation of liquid chromatography–mass


spectrometry methods: Part I
Anneli Kruve a , Riin Rebane a , Karin Kipper a , Maarja-Liisa Oldekop a , Hanno Evard a ,
Koit Herodes a , Pekka Ravio b , Ivo Leito a, *
a
University of Tartu, Institute of Chemistry, Ravila 14a, 50411 Tartu, Estonia
b
Finnish Customs Laboratory, Tekniikantie 13, PL 53, FI-02151 Espoo, Finland

H I G H L I G H T S G R A P H I C A L A B S T R A C T

 The status of validation of LC–MS


methods is comprehensively
reviewed.
 Clarity is brought into validation-
related terminology.
 Recommendations on difficult vali-
dation-related issues in LC–MS are
given.

A R T I C L E I N F O A B S T R A C T

Article history: This is the part I of a tutorial review intending to give an overview of the state of the art of method
Received 4 June 2014 validation in liquid chromatography mass spectrometry (LC–MS) and discuss specific issues that arise
Received in revised form 31 January 2015 with MS (and MS/MS) detection in LC (as opposed to the “conventional” detectors). The Part I briefly
Accepted 9 February 2015
introduces the principles of operation of LC–MS (emphasizing the aspects important from the validation
Available online 13 February 2015
point of view, in particular the ionization process and ionization suppression/enhancement); reviews the
main validation guideline documents and discusses in detail the following performance parameters:
Keywords:
selectivity/specificity/identity, ruggedness/robustness, limit of detection, limit of quantification, decision
Liquid chromatography–mass spectrometry
limit and detection capability. With every method performance characteristic its essence and

Abbreviations: APCI, atmospheric pressure chemical ionization; API, atmospheric pressure ionization; APPI, atmospheric pressure photo ionization; ban, the slope of the
calibration function for the analyte; bint, the slope for the potential interferent; CCa, decision limit; CCb, detection capability; EMA/EMEA, European Medicines Agency; ESI,
electrospray ionization; Ex, effect of variation of parameter X; FDA, United States Food and Drug Administration; HRMS, high resolution mass spectrometer; ICH, International
Conference on Harmonization; IR, infrared spectroscopy; IUPAC, International Union of Pure and Applied Chemists; LC–MS, liquid chromatography–mass spectrometry; LLE,
liquid–liquid extraction; LoD, limit of detection; LoQ, limit of quantitation; LRMS, low resolution mass spectrometer; MEionization, matrix effect of ionization; MRM, multiple
reaction monitoring; MS/MS, tandem mass spectrometry; MSn, consequent reaction monitoring; NLS, neutral loss scan; NMR, nuclear magnetic resonance; Rs,
chromatographic resolution; RSD, relative standard deviation; s, standard deviation; SPE, solid phase extraction; SRM, selected reaction monitoring; TOF, time-of-flight;
UPLC/UHPLC, ultra-high performance liquid chromatography; UV–vis, ultraviolet–visible spectrophotometry; VIM, International Vocabulary of Metrology; a, probability of a
blank sample being considered as a positive sample; b, probability of falsely negative result.
The other part of this paper is published here http://dx.doi.org/10.1016/j.aca.2015.02.016
* Corresponding author. Tel.: +372 7 375 259; fax: +372 7 375 264.

http://dx.doi.org/10.1016/j.aca.2015.02.017
0003-2670/ ã 2015 Elsevier B.V. All rights reserved.
30 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

Validation terminology are addressed, the current status of treating it is reviewed and recommendations are given,
Electrospray how to determine it, specifically in the case of LC–MS methods.
Ionization suppression ã 2015 Elsevier B.V. All rights reserved.
Limit of detection
Robustness

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 31
2. MS as a detector for LC . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.1. Ionization sources . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.2. Ionization suppression or enhancement effect . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 32
2.3. Operation modes of MS as a detector . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 33
2.4. Practical aspects of LC–MS method development . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
3. Overview of the main validation guidelines and general situation with validation of LC–MS methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
4. Parameters of LC–MS methods . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
4.1. Selectivity, specificity, confirmation of identity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 34
4.1.1. Selectivity (specificity) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
4.1.2. Identity confirmation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 35
4.2. Ruggedness/robustness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
4.2.1. Definitions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 36
4.2.2. Specific aspects in LC–MS . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
4.2.3. Experimental design of ruggedness/robustness testing . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
4.2.4. Method parameters to be varied . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 37
4.2.5. Numerically expressing robustness/ruggedness . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 38
4.3. Limit of detection, limit of quantitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
4.3.1. Limit of detection . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 39
4.3.2. Limit of quantitation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 40
4.3.3. Decision limit (CCa) and detection capability (CCb) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
Acknowledgments . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 41

Anneli Kruve obtained her Ph.D. from University of Riin Rebane obtained her Ph.D. in analytical
Tartu (UT) in 2011. Since 2005 she has been involved chemistry from University of Tartu in 2012 with a
in HPLC and LC/MS method development and topic on optimization and validation of liquid
validation in various fields: bioanalysis, food analy- chromatographic methods with mass spectrometric
sis as well as industrial analysis. Starting from detection containing derivatization. For the past
2011 she works as a research fellow at UT Institute of eight years her main research area has been LC–MS
Chemistry. In 2008 and 2009 she has worked in analysis, including method development and vali-
University of Helsinki in the field on miniaturization dation for various analytes and development of
of MS ion sources. Her main research fields are novel derivatization reagents for LC–MS. She also
method development for pesticide analysis, funda- works as a quality assurance specialist in the
mental studies of ionization efficiency and design of Estonian Environmental Research Centre.
MS ionization sources.

Karin Kipper obtained her Ph.D. from University of Maarja-Liisa Oldekop obtained her M.Sc. from
Tartu (UT) in 2012. Since 2004 she has been involved University of Tarty (UT) in 2013 and is now a Ph.
in the bioanalytical method development and D. student in the chair of analytical chemistry at UT.
validation for HPLC-UV/vis and LC–MS analysis, Her main field of research is development of LC–MS
working at UT Institute of Pharmacology and methods using derivatization. The focus is on matrix
Institute of Chemistry. Starting from 2012 Karin influences on this type of analysis, stressing the
Kipper works as a research fellow at UT Institute of importance of trueness of the analysis results but
Chemistry. Her main research fields are pharma- also the sensitivity of the method. Since the
ceutical bioanalysis (pharmacokinetic/pharmacody- beginning of 2013 Maarja-Liisa Oldekop works as
namic studies), pharmaceuticals’ pathways in the quality manager of the UT Testing Centre, which
environment and development of novel eluent is an ISO/IEC 17025 accredited testing and calibra-
additives for LC–MS in order to improve separation tion laboratory.
and peak shape of basic compounds.
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 31

Hanno Evard graduated from the Applied Measure- Koit Herodes obtained his Ph.D. from University of
ment Science master’s programme at University of Tartu (UT) in 2002. Since 2008 he works as the head
Tartu in 2012. His master’s thesis was about paper of the UT Testing centre – a unit providing testing
spray ionization. He is now a Ph.D. student at and analysis services and accredited according to
University of Tartu. His research is focused on ISO 17025 by the Estonian Accreditation Centre.
fundamental studies and developing new applica- Since 2005 Koit Herodes works as associate profes-
tions for different mass spectrometry ion sources. sor of analytical chemistry at UT Institute of
Chemistry. He has been principal investigator of
numerous projects involving LC–MS analyses. Cur-
rently he is the principal investigator of the project
“Development of software for validation of chro-
matographic methods”, which aims at creating web-
based software for validation of chromatographic
methods.

Pekka Ravio obtained his M.Sc. from University of Ivo Leito obtained his Ph.D. from University of Tartu
Helsinki in 1981. Since 1984 he has been working in (UT) in 1998. During 2002–2005 he reorganized the
the Finnish Customs Laboratory being responsible UT Testing centre – a unit providing testing,
for various types of mass spectrometric analyses. His measurement, analysis and training services to
main area of expertise is pesticide residue analysis laboratories and industry – and was its director.
and quality assurance. Starting from 2005 Ivo Leito works as professor of
analytical chemistry at Institute of Chemistry,
University of Tartu. His main research directions
are on the borderline of analytical chemistry with
other disciplines: chemistry of superacids and
superbases; metrology in chemistry (MiC); liquid
chromatography and mass spectrometry; sensors
and their metrological characterization; applica-
tions of instrumental methods in analysis of
historical objects. He teaches analytical chemistry
and its metrological aspects at UT and has been
involved in setting up several international MiC-related educational activities.

1. Introduction Method validation is a key activity in chemical analysis,


indispensable for obtaining reliable results [6]. The higher the
During the recent 15 years liquid chromatography mass complexity of the method, the more important and voluminous, as
spectrometry (LC–MS) has evolved from a scientific curiosity into a rule, is validation [7,8]. Methods based on LC–MS are notorious
a routinely applied technique finding increasingly more use in for their complexity, on the one hand because of the instrument
routine field laboratories. This has become possible largely due to itself and on the other hand because LC–MS is often applied to the
the advent of the atmospheric pressure ionization (API) methods most complex samples [5,9,10]. Besides the intrinsic necessity of
[1]. The API sources are able to produce gas-phase ions with little validation, there are increasingly more regulations affecting
or no spontaneous decomposition from delicate and high laboratories that stipulate method validation as a requirement
molecular weight analytes. This, combined with the intrinsic [11–13] and scientific journals publishing analytical chemistry
sensitivity of mass spectrometers, has revolutionized large areas of research require validation data for the methods to be published. A
chemical analysis where traces of organic analytes are determined very large number of articles reporting new LC–MS methods are
in complex matrices. Among the ionization methods electrospray submitted for publication each year and method validation is
ionization (ESI) has proven especially versatile [1]. As a result, usually an intrinsic part of such articles. However, in spite of the
almost all fields of chemical analysis (bioanalytical and medical, number of articles submitted and published, miscalculation and
environmental, food, drug discovery [2], etc.) have experienced big misinterpretation of validation parameters is still common due to
changes. complexity of the methods and some ambiguity in the definitions
LC–MS offers additional selectivity and confirmation of identity of some of the validation parameters [14].
compared to “traditional” chromatography (i.e., chromatography The importance of validation has led to the emergence of a large
with unidimensional detector, such as UV absorbance) by number of validation guidance materials for laboratories, both of
determining the mass/charge ratio of the ion(s) or recording MS universal nature [15,16] and sector-specific [17–20]. Although
data (in the broadest sense of this word) for the whole there is a general agreement on the various validation parameters
chromatogram, often resulting in three-dimensional datasets. to be evaluated, diversity prevails about details and about the
This has proven immensely useful in the wide range of fields where methodology employed for validation and acceptance criteria
LC–MS is applied [1,3]. [21,22]: different recommendations and different sets of terminol-
The extensive additional possibilities, however, come at a cost: ogy are found in the different guidelines. As a conclusion, the
LC–MS systems are complex and a large number of parameters analytical community is still far from a consensus about exactly
have to be at or near optimal values in order to get the desired how validation should be carried out, both in general terms and as
performance [4,5]. This automatically means that whenever an applied to LC–MS [14,23,24–25].
analytical method based on LC–MS is developed, its performance This tutorial review tries to overview these issues and to
has to be carefully checked and monitored. Nevertheless, the address the validation of LC–MS methods. Most validation
instrument vendors are continuously making efforts to reduce the guidelines do not specifically address LC–MS at all [15,26]. To
complexity of the systems and their usage. In addition, the default some others, LC–MS-specific issues have just recently been added,
parameters are often sufficient for routine applications and the LC– especially to the ones targeted towards bioanalytical methods
MS system software is increasingly user-friendly. [18,19]. Recently a review has been published on the validation of
32 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

bioanalytical chromatographic methods [27], which is relevant ionization (APPI) sources. Literature references are by no means
also for LC–MS. exhaustive. Preference has been given to literature sources that
The specific aims of this tutorial review are threefold: either specifically focus on validation of LC–MS methods or where
some important aspect of LC–MS method validation is highlighted.
1. Give a critical overview of the state of the art of LC–MS method This review addresses LC–MS with analytes that are separated with
validation especially with respect to the relevant guidance the conventional LC columns, i.e., small to medium-size organic
materials; molecules (with molecular weight up to few thousands).
2. Draw attention to the aspects that are important specifically in
validating LC–MS methods (e.g., ionization suppression by 2. MS as a detector for LC
matrix effects);
3. Give recommendations on problematic issues in validation of The success of the LC–MS technique arises from its ability to
LC–MS methods. give three-dimensional data. First, the compounds are separated in
time by LC. Ions generated in the ionization source are then
This review is written with highly pragmatic approach and is separated according to their m/z ratios in the mass analyzer of MS.
expected to be useful besides scientific research laboratories also Finally, the MS detector measures the abundance of each ion.
for routine laboratories. Compared to the traditional LC detectors, such as ultraviolet–
Few words on general terminology are appropriate here. For visible spectrophotometry (UV–vis) or fluorescence, the MS
measurement-related terms the definitions given in the Interna- detector enables significantly more reliable identification of the
tional Vocabulary of Metrology (VIM) [28] are used in most cases. compounds eluting from LC. This is true for the conventional
The term method performance characteristic is used for e.g., limit nominal resolution MS (typical examples are triple quadrupole and
of detection (LoD), trueness, etc. The term method parameter is ion trap instruments), especially if MS/MS detection (see below) is
used for adjustable parameters, such as mobile phase pH, flow rate, used, and even more so for the high-resolution instruments (such,
column type, etc. The word technique is used in this tutorial for as time-of-flight (TOF) or Orbitrap). Therefore, among other
describing the generic (instrumental) platform for analysis, e.g., benefits, LC–MS reduces the risk of false positive identification.
LC–MS. The word method means an analytical methodology for Another advantage over other techniques is that difficult analytes
solving a specific analytical task (e.g., determination of a set of can be successfully determined in complicated matrices at low
drugs in a specific matrix, such as blood plasma, using LC–MS with levels.
ESI ionization). In some documents [28] procedure is recom-
mended in this meaning, but the term overwhelmingly used by the 2.1. Ionization sources
analytical community is method and we choose to use it. The term
approach is used in the context of determining method perfor- For a long time combining LC with MS was hindered by the
mance characteristics, e.g., there are different approaches for difficulties in transferring analyte molecules from the liquid phase
determining LoD, assessing linearity, etc. The term run refers to the to the gas phase as ions. These difficulties were solved by the
analysis of a single sample (i.e., a single chromatogram). The term invention of the API sources [30]. API sources enable formation of
(analytical) sequence refers to a set of samples analysed in gas-phase ions directly from liquid flow and thereby enable
sequence (e.g., with an autosampler) with the same method (i.e., a connecting LC to MS. The most common API sources, and in the
series of chromatograms). focus in this paper, are electrospray (ESI) [31], APCI [32] and APPI
With every method performance characteristic (e.g., trueness, [33,34]. API sources cover a wide range of analyte and solvent
limit of detection, . . . ) we explain its essence and terminology, polarity.
the current status of calculating and interpreting it and give our Both positive and negative ionization mode are available in
recommendations, how to treat it, specifically in the case of LC–MS most API sources, though positive mode is more extensively used.
methods. Validation is a complex and multifaceted activity, not Positive ions are usually formed as protonated analyte molecules
always easily separable from the process of analytical method [M + H]+ but also adducts with cations (mostly sodium, ammonium
development. Therefore, occasionally our suggestions address also or potassium), e.g., [M + Na]+ [35] or, in less common cases, as

method development. It is assumed that the system suitability has radical cations after loss of an electron M +.
already been verified, so system suitability checking is not For negative ionization mode, also different forms of ionization
addressed here. may occur. Similarly to positive ionization, the most significant is
This tutorial review is divided in two parts. The Part I briefly deprotonation [M  H]. Less common is adduct formation with
introduces the principles of operation of LC–MS (emphasizing the e.g., chloride [M + Cl], nitrate [M + NO3] or acetate [M +
aspects important from the validation point of view, in particular CH3COO]. Also radical anions via addition of electron may occur
the ionization process and ionization suppression/enhancement); [31].
reviews the main validation guideline documents and discusses in The ionization in API sources is “soft”, meaning that the formed
detail the following performance parameters: selectivity/specific- ions have little excess energy and therefore undergo little or no
ity/identity, ruggedness/robustness, limit of detection, limit of fragmentation. This is very different from the electron ionization
quantification, decision limit and detection capability. The Part II (EI), which is typically used in gas chromatography mass
[29] starts with briefly introducing the main quantitation methods spectrometry, where fragmentation (often extensive) is rather a
and then addresses the performance related to quantification: rule than an exception [1].
linearity of signal, sensitivity, precision, trueness, accuracy,
stability and measurement uncertainty. Its last section is devoted 2.2. Ionization suppression or enhancement effect
to practical considerations in validation.
Literature starting from year 2000 is mainly covered, but In API sources, especially in ESI, ionization efficiency of the
occasionally earlier works of lasting significance have been analytes may be strongly altered by the compounds co-eluting
included. ESI as the most popular ionization method is covered (matrix compounds either from the sample analysed or very late
more thoroughly (and is the default ionization method), but most eluting compounds from previous samples [36,37]) with the
of what is written also applies to the atmospheric pressure analyte [38,40]. The effect may either reduce (called ionization
chemical ionization (APCI) and atmospheric pressure photo suppression) or increase (ionization enhancement) the analyte
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 33

signal. Often both suppression and enhancement effects are some cases. Unfortunately, there are numerous analytes for
abbreviated in the term matrix effect. The term matrix effect which neither APCI nor negative mode ESI are suitable.
can also be understood in a broader sense – “the combined effect of (3) The two main LC-method-related matrix effect reduction
all components of the sample other than the analyte on the possibilities are improvement of chromatographic separation,
measurement of the quantity” [39]. It means any influence that e.g., with ultra-high performance liquid chromatography
sample matrix may have on an analytical result and is applicable to (UPLC/UHPLC), and sample dilution. Both have been used by
any analytical method. This paper focuses only on the narrow numerous authors [55–58]. Dilution has been shown to
meaning of the matrix effect – a factor affecting the ionization significantly reduce the ionization suppression [56–58].
process occurring in LC–MS ionization sources. Therefore, to avoid However, it is often impossible to dilute the sample sufficiently
confusion, we denote it as MEionization (as ME is often used in LC– so that ionization suppression will completely disappear,
MS literature) and preferably use terms ionization suppression and because the analyte concentration may fall below the limit
enhancement in text. of quantification. In such cases, the so-called extrapolative
The ionization suppression may result in more than 90% of the dilution approach has been found useful [45], which consists in
signal decrease but can also lead to the complete loss of MS signal diluting the sample as far as possible and if the suppression is
resulting in false negative results [38]. The mechanism of still present then extrapolating the analyte concentration
ionization suppression is very complex and several reviews have mathematically to infinite dilution.
been published [41,42]. Therefore, the aim of this chapter is not to
summarize the available literature, but to describe the most Sometimes it is too difficult (and therefore impractical) or
important aspects from the validation point of view. impossible to remove all of the matrix effect, therefore, approaches
Whenever possible and practical, ionization suppression should accounting for the matrix effect have also been developed. Most of
be eliminated or significantly reduced. If it is not possible to reduce them fall either into the category of internal standard usage or
ionization suppression to the level of being insignificant, it should matrix-matched calibration and are described in the Part II of this
be taken into account in calculation of the results. Several tutorial review [29]. Also some additional more complex
approaches [36,43–46] have been suggested and tested for approaches have been proposed [44,59,60] in the literature but
reducing the ionization suppression effect, mainly focusing on are rarely used in routine analysis. Therefore, these methods are
ESI ionization source. In broad terms the approaches can be not considered further in this paper.
categorized as (1) sample preparation based, (2) instrumental
modifications and (3) modifications in LC method: 2.3. Operation modes of MS as a detector

(1) Less than ideal sample preparation may be viewed as the main Intensive development of mass spectrometry instrumentation
reason of occurrence of ionization suppression [41]. In case of a has enabled an increasingly diverse range of possible operation
perfect sample preparation combined with the perfect modes [1]. This has led to the situation that nowadays the majority
chromatographic separation – leading to the chromatogram of analyses carried out with LC–MS use tandem mass spectromet-
where the analyte is completely separated from all matrix ric (often abbreviated as MS/MS) detection. In very broad terms,
components – ionization suppression/enhancement would not MS/MS means that the ions obtained from the sample are not
occur and would not have to be considered. Unfortunately, directly detected to obtain the signal but, depending on purpose,
perfect sample preparation methods are not available in most some of them (called precursor ions) are selected from the
cases. A number of literature sources address choosing the spectrum, manipulated (usually fragmented) and the resulting
most effective sample preparation method from the matrix ions (product ions) are eventually detected. MS/MS, as opposed to
effect point of view [47,48]. In LC–MS solid phase extraction conventional MS, is popular in routine analysis because it enables
(SPE), liquid–liquid extraction (LLE), precipitation/centrifuga- more reliable identification of the analyte (in the sense of lower
tion or combinations of these as well as other methods are used false positive rate) and higher signal-to-noise ratio.
for sample preparation. Bonfiglio et al. [47] compared different MS/MS is defined as “acquisition and study of the spectra of the
sample preparation techniques and found that for phenacetin product ions or precursor ions of m/z selected ions, or of precursor
and caffeine determination in endogenous plasma protein ions of a selected neutral mass loss” [61] and embraces several
precipitation is the least favorable technique for LC–ESI–MS operation modes. Different MS operation modes such as selected
analyses while LLE was the most favorable. Additionally, reaction monitoring (SRM), multiple reaction monitoring (MRM),
Souverain et al. [48] found LLE to be more effective sample consequent reaction monitoring (MSn) and neutral loss scan (NLS)
preparation technique than SPE for methadone determination, are frequently used. Even though mass spectrometer operation
because the latter tends to concentrate not only the analyte but mode does not change the general validation rules, some
also matrix compounds similar to the analyte (i.e., potentially modifications may be needed (mostly from selectivity point of
co-eluting from HPLC with the analyte). The reason probably is view). Therefore, a short overview of the most important operation
that for LLE a larger selection of extracting solvents is available modes is given below. For more details, please see Murray et al.
and therefore more freedom in varying selectivity is achievable. [61].
On the other hand, in the case of SPE, a solid phase similar to the In SRM the precursor ion with previously specified m/z
HPLC stationary phase is often used (often both are low polarity (corresponding to the ion formed from the analyte) is fragmented
C18 or C8 phases) and therefore little additional selectivity is and the signals of one or more specific product ions are measured
obtained. Additionally, it has been shown by Dams et al. [49] [61]. This operation mode increases the selectivity of MS by
that sample preconcentration may significantly increase decreasing the probability of false positive identification. Ions
ionization suppression. formed from matrix compounds with the same m/z as the analyte
(2) The main instrumental modification considered is using a non- ions generally have different molecular structures and thus yield
ESI ion source, such as APCI instead of ESI, since ionization in different product ions and will therefore not be detected. The
the APCI source has been demonstrated to be less affected by selectivity and reliability can be further improved if multiple
matrix effects [49–52]. Switching the ESI source from positive product ions from the same precursor ion are measured.
to negative ionization mode [53] or reducing the flow rate of In MRM the signals of product ions from multiple precursor ions
the effluent [54], have also been demonstrated to be efficient in are measured and used for quantitation of multiple analytes [61].
34 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

Consequent reaction monitoring (MSn) is similar to SRM, but and the International Union of Pure and Applied Chemists (IUPAC)
involves more fragmentation steps: the product ions of one [16] have developed guidelines for single laboratory method
fragmentation are used as precursor ions for a next fragmentation validation. The United States Food and Drug Administration (FDA)
and so on, until the ions are eventually detected. n denotes the has a guidance document for analytical method validation for
number of fragmentations plus one. In this context, SRM can be bioanalytical methods [18]. Similarly, the European Medicines
called MS2. NLS is monitoring the loss of an uncharged species from Agency (EMA, formerly EMEA) has a “Guideline on bioanalytical
precursor ions [61]. In this text, the general acronym MS (without a method validation” [19]. The general Eurachem guide “The Fitness
superscript) means mass spectrometry with or without fragmen- for Purpose of Analytical Methods” [15] and NordVal “Guide in
tation. Validation of Alternative Proprietary Chemical Methods” [26] are
not limited to any technique and can be used throughout the field
2.4. Practical aspects of LC–MS method development of analytical chemistry. There are also more specialized guidelines,
such as validation guideline for pesticide residue analysis in food
LC–MS offers a large variety of ionization methods, MS and feed by the SANCO [65] or European Commission Decision
operation modes and quantitation methods. This tremendously 2002/657/EC [66], that establish criteria and procedures for the
increases the opportunities but also the responsibility of the user. validation of analytical methods to ensure the quality and
When developing an LC–MS method some important aspects to comparability of analytical results generated by official control
consider are: laboratories.
Depending on the field of use, different guidelines may vary in
(1) There are a very large number of parameters in LC–MS methods depth and also in parameters described. In general, most guide-
that can be optimized. lines are targeted towards quantitative measurements. Even
(2) Some of the key performance characteristics – most notably the though qualitative procedures are part of some of the guidelines
efficiency of ionization of the analyte in the ion source – are [20], these are not exhaustively discussed in this review.
either difficult to control or are very sensitive to small changes In addition to previously mentioned official guidance docu-
in system parameters. ments, a number of articles, including review articles, have been
(3) MS detector in general displays inferior repeatability compared published on the topic of analytical method validation [6,21,64].
to most other detectors (in their respective working concen- Most of these are general but some address specific applications or
tration ranges), especially the UV–vis absorbance detector. The techniques. There have been number of articles specifically
repeatability standard deviation of MS signal, even if replicate focusing on the LC–MS method validation in toxicology
samples are analysed within a short time period, can be quite [25,64,67]. In addition, there is a large number of articles
high. The trueness of the quantitative MS results is decreased demonstrating validation of LC–ESI–MS methods for practical
first of all by different ion source related phenomena, such as analytical tasks with respect to certain validation guidelines such
ionization suppression/enhancement. This has important as FDA [68–72], ICH [73–75] Eurachem [76–83], EMA [84–86],
implications for determining trueness, precision and accuracy. IUPAC [87–89] or SANCO [90,91]. However, in most cases not all
(4) MS as a detector is mostly used for determination of very small parameters are investigated. There are also a number of cases
quantities of analyte. Therefore, a number of problems, such as when for newly developed methods thorough validation is carried
incomplete selectivity, non-ideal sample preparation, etc. can out but not according to any of the published guidelines [92–95]. In
be even further amplified. a number of cases only selected validation parameters are under
close interest and most commonly these are ionization suppres-
The benefits of MS as a detector (selectivity, identification sion/enhancement, limits of detection, precision parameters
ability) have led to very high expectations by LC–MS users which [85,96] and in many cases also recovery [97,98].
often do not become true. A most dangerous of them is the belief Many guidelines try to harmonize definitions required for
that the importance of chromatographic separation as well as validation characteristics and their basic requirements, especially
sample cleanup becomes less important because the MS detector the ICH guide, but they provide only a basis for general discussion
provides selectivity of its own [37,41] and consequently very short of the validation parameters, their calculation and interpretation.
LC columns can be used. Unfortunately, even though the right Therefore, a lot is put on the analysts’ shoulders. They have to (1)
choice of MS operation mode often guarantees sufficient detection identify parameters that are relevant to the performance of the
and identification ability, the accuracy of the obtained results may given analytical method, (2) design of a proper validation protocol
be strongly influenced by the ionization suppression/enhancement including acceptance criteria and (3) choose the methods for the
occurring in most API sources. Both ionization suppression [41] or appropriate data evaluation [99].
ion source contamination [62,63] result in variability of both MS
signal and the obtained results. 4. Parameters of LC–MS methods

3. Overview of the main validation guidelines and general In the following sections the main performance characteristics
situation with validation of LC–MS methods of analytical methods are examined with emphasis on LC–MS.

Owing to the importance of method validation in the whole 4.1. Selectivity, specificity, confirmation of identity
field of analytical chemistry, various international organizations
and conferences have issued a number of guidance documents Selectivity and specificity refer to the ability of a method to
targeted toward single laboratory validation. Though different in measure the amount of the analyte that is claimed to be measured.
suggestions and requirements, all of these documents are IUPAC suggests using term “selectivity” to express the extent to
important and potentially helpful for validating any method [64]. which other substances interfere with the determination of an
Harmonized guidelines between European Union (EU), Japan analyte, while “specificity” is used to denote the ultimate
and the United States have been developed within the expert selectivity, meaning that no detectable interferences are supposed
working group of the International Conference on Harmonization to occur [100,101]. Analytical techniques are almost never specific
(ICH) of technical requirements for registration of pharmaceuticals by themselves, but validated analytical methods within their scope
for human use (ICH Q2(R1]) [17] AOAC International (AOAC) [20] of application can be specific. Sometimes the term “specificity” is
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 35

used to denote selectivity. For example, ICH and NordVal use the of combined “LC + detector” selectivity should be acceptable
term “specificity”, while AOAC, EMA, Eurachem, FDA and IUPAC use (considering of course the possible ionization suppression, see
“selectivity”. In this paper we use the IUPAC recommendation. above), but it would not be acceptable by the rules of ICH or AOAC
Selectivity of a method has to guarantee that the effects of as virtually all the organic compounds are detectable by infrared
interferents (degradation products, metabolites, etc.) in the spectroscopy (IR) and nuclear magnetic resonance (NMR). Out of
analyte signal are insignificant. The interferent can either act as the validation guidelines studied, only Eurachem foresees the
the analyte, and yield the signal indistinguishable from analyte’s possibility that interferents are not separated [15].
signal, or it can suppress/enhance the signal by altering sample In different situations different requirements apply for the
preparation, chromatographic separation or detector response. In selectivity achieved with chromatographic separation. For exam-
this chapter the former is dealt with, while the latter is discussed in ple, in the case of pharmaceutical preparations, all components of
the Part II of this article [29]. the sample must be revealed. So, all the components are
Samples almost always contain some compounds that have considered analytes and must be selectively detected. Situation
properties similar to the analyte. Therefore, the identity of a signal is different if for example, pesticide residues are determined in
presumably belonging to the analyte has to be confirmed. vegetables. Vegetable extract matrix is rich in compounds, which
Validation has to demonstrate that it is possible to confirm the may co-elute with analytes, and it would be advantageous if
identity of the analyte, but identity confirmation in the actual detector would be insensitive toward those endogenous com-
samples has to be done during routine use of the method. LC–MS is pounds, enabling selective detection of the pesticides.
often used for determining banned substances. In such analysis the For selectivity assessment, NordVal suggests analysis of a blank
large majority of samples are “negative” (analyte missing). In such sample and spiked blank samples [26]. Although it is suggested to
cases it is sometimes useful to have two separate methods – analyze a more concentrated extract of the blank to demonstrate
screening and confirmatory methods [20,65,66]. A screening absence of signal at analyte retention time, NordVal does not
method is simple, fast, can be qualitative and is deliberately specify any criteria for acceptable signal from the blank. Similarly
positively biased (i.e., the probability of false negatives is to NordVal EMA and FDA require analysis of blank matrices (at least
minimized). For example, for analysis of 120 pesticides and from six independent sources), but they also set a clear limit –
metabolites in infant food Anagnostopoulos et al. developed an signal from the blank at analyte retention time must be less than
MS2 screening method with one transition for each analyte and 20% of the limit of quantitation (LoQ) for the analyte [18,19]. The
separate confirmatory methods for each analyte containing two FDA guide adds that if LC–MS is used, analysis of six blank matrices
transitions [102]. For the analysis of emerging environmental may not be important, but matrix effects should be investigated
contaminants [103] (e.g., residues of personal care products and [18].
pharmaceuticals) qualitative screening methods based on liquid According to the European Commission decision 2002/657/EC,
chromatography coupled to high resolution mass spectrometer specificity is evaluated by analyzing at least 20 representative
(LC–HRMS) have proven to be especially suited [104,105]. Both blank matrices and additionally blank matrices fortified with
screening and confirmatory methods have to be validated. compounds likely to interfere with the analysis. It should be
Validation of screening methods is not within the scope of this demonstrated that analyte is not falsely identified, its identifica-
review and interested reader is directed to the following works: tion is not hindered and its quantification is not influenced [66].
[104–107]. Clearly, analyzing selected samples twice is disadvan- While the Eurachem guide just notes that interferents usually
tageous with respect to workload and methods, which are able to affect the slope of the calibration curve [15], IUPAC recommends
screen and confirm in one run are most welcome. The MS quantitation of selectivity by means of selectivity index, which is
operation modes described above, make LC–MS one of the most defined as ban/bint, where ban is the slope of the calibration function
selective techniques available, which also enables identity for the analyte and bint is the slope for the potential interferent
confirmation. [16]. bint can be determined by analyzing blank sample and blank
Different validation guidelines approach selectivity and identi- sample spiked with the interferent. Availability of the interfering
ty confirmation at different level of detail. The majority of the compound limits the use of selectivity index. In addition, no
guidelines [16–18,26] are very general and applicable to any acceptance criteria are given in the guideline for the selectivity
analytical technique. Such guidelines do not specifically address index. The present authors are not aware of any literature source
MS. In contrast the European Commission decision 2002/657/EC demonstrating the use of the selectivity index.
[66] and the SANCO guide [65] discuss LC–MS extensively. In conclusion, although MS is a highly selective detector,
interferences are not uncommon in LC–MS analysis [108,109].
4.1.1. Selectivity (specificity) During validation the analyst has to test as many possible
Selectivity evaluation in LC–MS usually starts from chro- interferents as reasonable, and continue monitoring selectivity
matographic resolution (Rs) between the analyte and the closest during routine use of the method. This can be done e.g., by
eluting peak. Rs at least 1.5 is required by AOAC [20] and 2 by FDA routinely analyzing a suitable control sample (either natural or
[18]. The Eurachem guide requires demonstration of separation self-prepared) containing the analyte (a known amount) and
from other components also on a column of different chemistry interferent(s).
[15]. In addition to this, AOAC, referring to FDA validation
guidelines, demands that no other compound should be detectable 4.1.2. Identity confirmation
when other selective methods are used – IR, NMR or MS [20]. For identity confirmation some of the validation guidelines
Similarly, ICH finds that if one is unable to demonstrate that a recognize the power of MS coupled to chromatography. AOAC
single analytical method is specific, two or more analytical recommends LC–MS (or gas chromatography–mass spectrometry
methods should be used [17]. (GC–MS)) full scan mass spectrum or identification of 3–4 frag-
These requirements of ICH and AOAC are really stringent, as ments or MSn [20]. Eurachem states that if the method under
they neglect detector-side selectivity. For example, if there is a co- evaluation is not specific, confirmation by independent methods
eluting compound at the analyte retention time, then it might be must be performed [15]. FDA [18] and ICH [17] are rather general,
possible to choose detector or detector setting (e.g., UV–vis the former stating that “Evidence should be provided that the
wavelength, MS mode or MRM transition) such as it responds only substance quantified is the intended analyte” and the latter
to the analyte. From the perspective of analyte detection, this kind requiring that positive results must be obtained by analyzing
36 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

samples containing the analyte and negative results from samples specific aspects of LC–MS. Therefore we recommend 2002/657/EC
not containing the analyte, and also negative results must be as guidance for LC–MS method selectivity and especially identity
obtained if structurally similar or closely related substances are confirmation.
analyzed.
Perhaps the most concrete guidance about assessing identity 4.2. Ruggedness/robustness
confirmation in LC–MS is given in the European Commission
decision 2002/657/EC [66] and the SANCO guide [65]. At least 4.2.1. Definitions
7 data points must be acquired over a chromatographic peak [66] The terms robustness and ruggedness refer to the ability of an
and the shape of the analyte’s peak in the sample must be similar to analytical method to remain unaffected by small variations in
that obtained from the calibration solution [65]. The relative method parameters (mobile phase composition, column age,
retention time (i.e., the ratio of the retention time of analyte to that column temperature, etc.) and influential environmental factors
of internal standard) should match that of the calibration solution (room temperature, air humidity, etc.) and characterize its
within 2.5% [65,66]. reliability during normal usage. The notion of remaining unaffect-
For identity confirmation 2002/657/EC [66] and the SANCO ed has two possible interpretations – it can be interpreted as (1) no
guide [65] also set criteria for mass spectra – relative abundances change of the detected amount of the analyte in a certain sample in
of analyte ions in sample compared to this in standard solution spite of the variation of the method parameter [66] or (2) no
should fall within specified ranges. For example, if the intensity of change of the critical performance characteristics (e.g., limit of
an ion is within 10–20% relative to base peak, its maximum quantitation) by the variation of the method parameter [65]. In
permitted tolerance is 30% (for full list of criteria, see [65,66]). experimental evaluation of robustness either one of these
Additionally it is suggested to compare the obtained mass spectra interpretations can be used.
to the reference spectra obtained with the same instrument. The definitions in the guidelines [15–17,20] as well as review
For identification at least four ions with specified tolerances articles [21,27,114,115] are very similar. Some guidelines use the
[66] must be registered. Due to soft nature of ionization methods term robustness and some use ruggedness. When used together
used in LC–MS fragmentation of quasi-molecular ion is seldom they are treated as synonyms in most cases [7,15,66,114–120]. The
observed in single stage (LC–)MS. In addition, in triple quadrupole only widespread guideline making difference between these terms
instruments full scan spectra lead to significantly higher limits of is the former USP [121], but the later versions of USP [122] use only
detection than monitoring specific ions. Therefore, fragmentation- the term robustness. Considering robustness as a method
based MS modes are recommended. The way of specifying the development parameter, EMA and FDA guidelines are missing
amount of data required for identity confirmation is different in the term. A recent review article emphasizes the importance of
2002/657/EC [66] and the SANCO guide [65]. European Commis- robustness testing and discusses different approaches thoroughly
sion [66] has introduced an elaborate system of identification [27].
points, where, for example, application of MS/MS for isolating one The above definitions imply changes made to the method
precursor ion (gives 1 point) and registering signals from two within the same laboratory. However, robustness can also be
fragments (1.5 points each) yields 4 identification points in total. described as the feasibility to reproduce the analytical method in
The required number of points for unambiguous identification is different laboratories or under different circumstances without the
3 or 4 depending on substance. The SANCO guide [65] simply states occurrence of unexpected differences in the obtained results [118].
that for identification using MS/MS experiment at least 2 product Along the similar lines it has been suggested that ruggedness
ions must be analyzed – thus coming to the conclusion similar to should be used as a parameter evaluating constancy of the results
that of European Commission [66]. when external factors such as analyst, laboratory, instrument,
The system of identification points is often used in case of liquid reagents and days are varied and robustness should be used as a
chromatography coupled to low resolution mass spectrometer parameter characterizing the stability of the method with respect
(LC–LRMS) analyses [52,110], but also in case of LC–HRMS methods to variations of the internal factors (parameters) of the method
[73,111]. For the best identity confirmation the mass spectrometer (e.g., parameters related to sample preparation, mobile phase
should be capable of precursor isolation-fragmentation and composition, mobile phase flow rate, injection volume, column
registration of the resulting full mass spectrum at high resolution. temperature etc.) [14,21,123].
This is acknowledged by the European Commission decision [66] The term robustness is in most cases understood in terms of
by assigning to HRMS double the identification points of LRMS influence of variations of method parameters on results. Our
(giving 2 points for a precursor ion or 2.5 for a transition). Utility of experience suggests, however, that an additional dimension –
HRMS for identification is also demonstrated by Pozo et al. [112]. robustness in terms of variability of sample matrix – is beneficial:
The SANCO guide [65] requires registration of at least two different matrices can lead to different matrix effects (either in the
diagnostic ions (at least one fragment and preferably quasi narrow or broad sense). On the example of blood plasma:
molecular ion). For the complete set of identification requirements depending on the personal variations in metabolism, diet, possible
see Refs. [65,66]. diseases, e.g., the composition (first of all but not limited to the
Also, directions for choosing proper diagnostic ions, which content of proteins, phospholipids and polyunsaturated fatty acids
enable achieving selectivity, are given in documents [65,66]. [124–126]) of blood plasma can vary significantly, even though
Quasi-molecular ion, product (fragment) ions with higher m/z and formally the matrix is the same – blood plasma [127]. The sample
fragments that originate from different parts of the molecular ion preparation procedure that is suitable for blood plasma of low
are recommended. Transitions associated with losses of common protein or phospholipid content may give different results for
fragments (for example loss of water), have been demonstrated to blood plasma with high protein or phospholipid content. This is
cause false positive as well as false negative results [112] and occasionally addressed in validation guidelines under selectivity
should therefore be avoided. [18,19]. However, the possible effects of this kind of variability are
Criteria, similar to those of SANCO and European Commission, not limited to the loss of selectivity, but can also influence recovery
have also been published by FDA [18] and Bethem et al. [113], but (and hence trueness), ionization suppression/enhancement as well
are rarely referenced. as limit of detection (LoD)/limit of quantitation (LoQ). It is thus
Compared to the validation guides (except SANCO), the useful to investigate the effect of sample matrix variability (in the
regulation 2002/657/EC has most thoroughly incorporated the case of formally identical matrices) more broadly than just for
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 37

selectivity. See also the section of Trueness in part II of this review of variables plus one. However, none of the main effects are
[29]. confounded with each other. If the number of variables is large
There are studies where the terms robustness/ruggedness are (e.g., MS parameters are also included) and the number of factors
misinterpreted and actually decision threshold, detection capabil- examined exceeds the number of experiments, the supersaturated
ity (see below) [128] or measurement uncertainty [88] is designs are used [118]. The total variance of changes in results
evaluated. could be used as a measure of robustness. The significance of the
In this review we use the term robustness for expressing the effects of the parameters (factors) on the signals can be evaluated
stability of the method against small variations of the intrinsic using ANOVA or t-test (in case the relationship between the
method parameters. Since some changes in the method perfor- parameter change and signal is linear) [27,119].
mance can occur over longer period of time, the robustness is a The above described approaches are rigorous and powerful.
validation parameter that has to be monitored during the However, literature survey, as well as contacts with routine
validation and also after the validation procedure and during laboratories indicate, that these approaches are seldom used
the method lifetime. We use the term ruggedness for expressing [92,134–138]. The main reason is that these approaches require
the stability of the method against extraneous influencing factors. knowledge and experience with mathematical statistics. In most
We address changes of the method parameters (i.e., within- cases experiments with one-by-one variations (one variable at a
laboratory assessment of robustness) and variability of sample time approach) of the most important parameters are carried out
matrices. We do not explicitly address changes occurring when a [27,138–142].
method is transferred from one laboratory to another. Concluding, there is a difficult situation where on the one hand
analytical methods are very complex, have numerous parameters
4.2.2. Specific aspects in LC–MS and would, thus, benefit from validation with full rigor and, on the
As explained above, there are a very large number of adjustable other hand, available resources are always limited. Before starting
parameters in LC–MS methods and some of the key performance investigation of robustness it is crucial to find out, what are the
characteristics – most notably the ionization efficiency of the critical performance characteristics of the method. According to
analyte in the ion source – are either difficult to control or are these characteristics the method parameters to be varied are
sensitive to small changes in system parameters (or sample thereafter chosen. For example, if LoQ is very close to the LoQ
properties), resulting in poor instrument reproducibility between required by legislation, then changes in LoQ value have to be
the runs (between samples with formally identical matrix). monitored against small changes in method parameters. Most
Furthermore, LC–MS is very often used for the determination of influential method parameters for LoQ could be MS parameters,
very low levels of analytes and in highly complex matrices. mobile phase pH, extraction parameters (see Table 1). The main
Analysis of complex matrices often requires complex multi-step criteria for choosing parameters are (1) how much a given method
sample preparation procedures. parameter can influence the critical characteristic and (2) how
Put together, the above listed factors clearly indicate that in the likely it is that this parameter will change uncontrollably.
case of LC–MS method validation, investigation of ruggedness and Based on the common practice in literature and on our own
robustness is very important. experience we recommend the following:

4.2.3. Experimental design of ruggedness/robustness testing 1. Change parameters one by one (one variable at a time approach)
Because of the very large number of potentially variable in both directions from the nominal (optimal) value. Changes in
parameters it is reasonable to divide assessment of ruggedness into the parameters should be realistic in the context of normal use
separate parts. A very logical division would be to test ruggedness of the method.
separately for sample preparation and for the LC–MS analytical 2. Do not stop there! Often parameters may be mutually unrelated
part. (uncorrelated), but in some cases this does not hold. For
The experimental designs suggested for validation often involve example, change in mobile phase pH can decrease resolution
fractional factorial (saturated designs) [129] or the Plackett– between two adjacent peaks. Likewise, increase of mobile phase
Burmann design [119,130,131]. flow rate can also lead to decrease of resolution. While
In the Plackett–Burmann approach, N  1 parameters (varia- separately any of these two changes can still lead to no loss
bles) are studied in N experimental runs (with N being a multiple of of resolution, their occurrence together may lead to peak
4) [22,118,130]. The factors are investigated at two levels: low () overlap. Whether this is the case, can often be determined by
and high (+). From the obtained signals, factor effects are calculated educated inspection of the effects of the changes (without
according to: additional experiments) and noting possible problems.
P P 3. Effects from the change of parameters should be monitored. If
YðþÞ  YðÞ
Ex ¼ ; (1) necessary, graphical or statistical analysis of the effects should
N=2
be done.
where Ex is the effect of variation of parameter X, the sums indicate 4. Regarding the robustness tests results, if necessary, measures to
the signals where factor X is at (+) or () level. N is the number of improve the performance of the method should be taken.
design experiments. Both graphical and statistical interpretation
methods are described [22,129,132,133] for identifying significant
effects. 4.2.4. Method parameters to be varied
Parameters X indicate the different factors (operational factors The validation guidelines differ somewhat by their suggestions
and environmental factors) and their levels on robustness on which parameters should be varied and by how much.
estimation. The factors can be divided into quantitative (pH, ICH [17] states different conditions such as variations in mobile
temperature, concentration of the solutions), qualitative (column phase composition, usage of different columns (different ages,
age, recent changes of chromatographic column made by the different lots), column temperature and flow rate.
manufacturer) and mixture factors (fraction of the organic Eurachem [15] recommends using the AOAC guide and Youden
modifier on the eluent) [118]. ruggedness trial giving a short overview of ruggedness tests and
In the saturated designs, the number of experiments required, the influence on either precision or accuracy. Different parameters
without counting the central (initial) points, is equal to the number that could be varied and taken into account are for example,
38 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

Table 1
Recommended method parameters to be investigated during robustness studies.

Parameter Likelihood of uncontrollable Recommended extent of variation Comments


change
Liquid chromatography
pH Medium 0.5 units pH will have a strong effect on retention time (and
possibly resolution) if the analyte’s pKa value is within
1.5 units of the mobile phase pH
Concentration of additives in Medium 10% (relative) Salts, ion-pair reagents, modifiers can suppress/
eluent enhance analyte’s ionization in the ion source and
change its retention time and possibly resolution from
other compounds
Organic solvent content in the Low to Medium 2% Organic solvent content influences retention time (and
eluent possibly resolution) and analyte signal in LC–MS
Column temperature Low 5  C Column temperature influences the retention time
(and possibly resolution)
Eluent flow rate Low 20% Eluent flow rate influences the retention time (and
possibly resolution)
Column batch and age Medium – Changes in column can influence the retention time
(and possibly resolution)

Samples and sample preparation


Analyte extraction time; solvent High 20% Influences recovery and LoQ/LoD
amount and composition (in
liquid/liquid and solid phase
extraction, etc.)
Injection solvent composition Low/High 10% (relative) This is the solvent in which analyte is taken up during
the last stage of sample preparation. The
recommended extent of variation refers to the minority
component(s). This composition can influence
retention time and recovery and therefore also the
matrix effect (in broad sense), LoQ/LoD and stability.
The effect can be very serious in the case of UHPLC and
is usually not that critical in the conventional HPLC
Matrix effect in broad sense High 6 different Can be assessed under selectivity studies [19,18].
(sample matrix source) Influences trueness (recovery and ionization
suppression), LoQ/LoD

Mass spectrometry
Drying gas temp Low 10  C Drying gas temperature can influence analyte
ionization efficiency in the ion source
1
Nebulizer gas pressure/flow rate Low 5 psi/ 1 L min Nebulizer gas pressure/flow rate can influence analyte
ionization efficiency in the ion source
Ion source configuration High (if configurations can vary) According to the ion source design Ion source configuration can influence spray and
(nebulizer position) ionization efficiency in the ion source
Not applicable (if fixed source) Should be varied if source is used in
different configurations
Ion source condition (nebulizer High After analysis of samples versus Contamination can spontaneously accumulate when
aging, ion source contamination) cleaned system analyzing a series of samples

different analysts, instruments, reagents and variations in sample experience shows that in the case of LC–MS methods the MS
preparation or sample matrix. Evaluation of each parameter parameters are as important as the LC parameters (although
separately gives useful information about method robustness. several problems, such as matrix effects due to overlapping peaks,
From the results the effect of changes on method can be estimated analyte peaks falling outside the MS time window, etc are caused
and the factors can be determined for the parameters [143]. first of all by LC).
The NordVal guide [26] lists the most popular parameters to be Our recommendations on the choice of method parameters to
included in the robustness test: the composition of the samples, be varied during robustness study are collected in Table 1.
mobile phase pH, timing of individual (assay) steps, temperature Additionally, the possible changes of the critical performance
and presence of potentially interfering substances (e.g., tannins or characteristics (e.g., LoQ) and detected amount of analyte (e.g.,
other complexing agents, varying levels of lipids, endogenous decrease of trueness because of interference caused by incomplete
enzymes). resolution) are commented on. Differently from the LC–MS
Robustness can be studied by varying typical parameters in the method itself, sample preparation can have a large number of
chromatographic run that are capable of influencing analysis parameters that cannot be easily systematized. Therefore the
results [144] as well as the influence of the columns age or recommendations about sample preparation parameters are very
replacement. Our suggestions are presented in Table 1. It is well general.
known that even if two columns are formally of the same type their
performance can differ [145]. Additionally, it may be necessary to 4.2.5. Numerically expressing robustness/ruggedness
investigate robustness related to change of the instrument or some Robustness/ruggedness is usually expressed as relative stan-
of its components. This need will occur when either a component dard deviation (RSD)% of data obtained with the changed
(e.g., pump or detector) or the whole system is replaced. parameter inside the method with respect to the same data
In the literature limited attention has been paid to varying obtained using initially observed conditions. Expressing the
besides the LC parameters also the MS parameters [146,147]. Our robustness using the RSD [133] is a simple and well understood
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 39

approach. There is some similarity between RSD expressing than one day, the highest LoD value (i.e., the most conservative
robustness and RSD expressing intermediate precision (see Part LoD value) should be used so that the declared LoD would be
II of the review [29]). The main difference is that in the case of routinely achievable with high probability. It is important to
robustness determination the changes in the method are intro- stress that the word “conservative” here means that the
duced deliberately and their magnitude is controlled, while probability of false positives will become lower, while the
intermediate precision addresses unintentional changes that occur probability of false negatives will become higher.
under normal operation of the method in the laboratory. In LoD is matrix-sensitive and should be determined in a matrix
addition, robustness/ruggedness can be evaluated by the Student t- that matches the real sample matrix [15], i.e., using matrix-
test to assure the statistical significance of these obtained results matched samples. It can be difficult or even impossible to find
[144]. The review article [133] focuses on the variety of statistical blank matrix-matching samples and therefore a blank sample with
tests (e.g., the t- and F-tests) for the robustness evaluation. These a similar matrix can be used (see Chapter 3.2.2 in Part II of this
test the statistical significance of the changes obtained by the tutorial review [29]).
robustness evaluation. In addition, it has to be carefully evaluated if In the case of LC-MS/MS, multiple product ion intensities are
the changes are also significant from the fitness for purpose often monitored to confirm the analyte identity. The European
perspective, compared to other parameters influencing the Commission decision 2002/657/EC and SANCO set the requirement
method performance. that S/N ratios for all diagnostic ions should be 3. The same
guidelines also set the maximum allowed errors of relative ion
4.3. Limit of detection, limit of quantitation intensities and the number of diagnostic ions necessary for
different instrumentation and chemicals [65,66]. These require-
4.3.1. Limit of detection ments markedly influence LoD determination: LoD has to address
Limit of detection (LoD), also called detection limit, is loosely the diagnostic ion with the lowest intensity because as soon as the
defined as the lowest amount or lowest concentration of the presence of this ion cannot be stated with confidence, the identity
analyte in a sample which can be reliably detected and identified of the contaminant becomes uncertain. Different approaches for
with the method [15–17,26]. This does not imply the possibility to determining LoD are discussed below.
quantify the result [26]. Although this definition is used in many Signal-to-noise ratio (S/N) can be used to determine LoD if the
cases due to its simplicity it is not fully rigorous, because the method exhibits baseline noise. S/N is found by comparing signals
meaning of “reliably” is not clarified. Furthermore, when speaking from samples with known low analyte concentration and blank
about detecting an analyte near the detection limit, two types of samples. LoD is determined by establishing the minimum
false results are possible – false positive and false negative results – concentration at which S/N is over 3 or between 3 and 2 [17].
which are not fully accounted for in this definition. The common This approach cannot be always used with LC-MS/MS measure-
way of interpreting LoD data takes care of avoiding false positives, ments, because the baselines in the MS/MS mode do not always
but tolerates false negatives: If a peak that could belong to the exhibit noise. When the noise level cannot be reliably measured
analyte is there but the calculation gives a value below LoD then it (either there is no baseline noise or it is irregular) then the S/N
is not possible to reliably declare that the analyte has been values become highly variable or S/N cannot be calculated at all.
detected (and the result has to be reported as “below LoD”), However, if noise can be measured then the data analysis
because in reality the analyte may be present but at a very low programs usually enable calculation of S/N that can also be used.
concentration. Interestingly, explanation of this meaning of LoD is It is recommended to use the latter method to find S/N as it
not included in validation guidelines. provides values with lower variability. The S/N ratio can vary
An alternative approach of characterizing the detection significantly between days and even within a day, thus many
capability of a method has been proposed to account for these measurements should be made for reliable determination of LoD
shortcomings. This is achieved by specifying the lowest concen- [149].
tration levels related to analyte detection reliabilities in terms of Another approach to determine LoD is to measure a number of
probabilities of false positive and false negative results. These solutions with different concentrations close to the LoD, each with
lowest levels are termed as decision limit (CCa) and detection 10 separate samples. The lowest concentration where all 10 sam-
capability (CCb), respectively. This approach was first proposed by ples have positive results can be considered LoD [15,26]. This
Currie in 1968 [148], is briefly mentioned in the IUPAC validation approach was originally designed for qualitative analysis where
guideline [16] and more recently adopted into the EU food safety the result is expressed either as a positive (analyte detected) or a
legislation [66]. CCa and CCb are discussed further in a following negative (analyte not detected). Nevertheless, it can also be used
chapter. for quantitative analytical methods. This is so because, in fact, in
We discuss here determining the LoD of the “whole method”, the LoD region most quantitative methods become qualitative. The
expressed as the analyte concentration in the sample, e.g., mg of difference between positive and negative samples can be made for
analyte per kg of sample. This means that LoD should be example, based on the S/N: a concentration level where all
determined taking into account all steps in the method 10 samples have S/N over 3 can be used as LoD. Alternatively, a
(including sample preparation). We do not discuss here the visual assessment of the LoD can be used.
so-called instrument LoD (which is related to detecting analyte in If a blank sample can be measured, the following equations can
pure solvent). Since any alterations made to the method may be used to determine LoD:
change LoD, LoD should be determined for fully developed
LoD ¼ 3  sðblankÞ (2)
methods [16]. For analytical methods where LoD is not in the
validation range it is only necessary to estimate that it is low or
enough, but its numerical determination is not necessary [16].
LoD ¼ X ðblankÞ þ 3  sðblankÞ (3)
LoD is a highly variable parameter (i.e., it has poor day-to-day
reproducibility) and it should therefore be re-determined where X(blank) is mean value of the blank results and s(blank) is
regularly in order to reflect the actual operating performance the standard deviation of the blank values [15,26]. However,
[15]. Therefore, a conservative estimate of LoD is preferred, Eurachem requires the use of fortified samples for chromatograph-
especially for cases where LoD is calculated from the data ic techniques as measurement results can be obtained only when a
collected during a short time period. If LoD is estimated on more peak can be detected over noise [15]. The following equations can
40 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

be used for the fortified samples: compared to routine analysis and enables frequent LoD determi-
nation so that the determined values correspond to the actual
LoD ¼ 3  sðfortifiedÞ (4)
situation at the laboratory [149].
or In conclusion, we advise determining LoD at least 5 times over a
long period (e.g., 5 months) to acquire a sufficiently representative
LoD ¼ XðblankÞ þ 4:65  sðfortifiedÞ (5)
LoD result. The highest obtained LoD should be declared as the
where s(fortified) is the standard deviation of the fortified sample limit for the method, so that the laboratory would be with high
[15,26]. Samples should be fortified at the concentration level close probability routinely able to achieve this LoD. Although examples
to the LoD or at lowest acceptable level determined by the needs of can be found in the literature where LoD has been determined on
the method [15]. Eurachem also suggest an approach to take into different days [87] in most cases LoD is determined only once
account the number of repeated sample measurements made by [128,138,152]. It is also recommended to visually evaluate the
averaging the repeated results [15]. In case blank correlation is peaks in the LoD range to be sure that the calculations have not
used, Eurachem also suggests taking into account the repeated given unrealistic values. When stating LoD of the analytical
measurement made for blank and the sample [15]. If the method, the approach used to determine LoD should be specified
calibration graph intercept is statistically insignificant then there [17]. If the result of the sample on a specific day is visually above
is no difference between using concentration scale (after the LoD and a control sample is measured that shows lower LoD
calculating concentration values for all intensities) or intensity than previously reported then the analyte can be reported as
scale (after which LoD as concentration can be found from that present in the sample. Moreover, the performance of the method
value) in the above equations. However, if the intercept is should be monitored regularly by analyzing samples in the vicinity
statistically significant then only concentration scale can be used of LoD or a little above. Problems with the measurement method
[149]. The number of replicate determinations is important, are indicated by significant variation of results from the average or
because unless a large amount of data is collected LoD will be results drifting in one direction away from the average over a
subject to large random variations [16]. Eurachem and IUPAC longer period of time. In these cases the performance of the
recommend that at least 10 and 6, respectively, separate blank and method should be reevaluated and if necessary the source of
fortified samples should be measured [15,16]. If the intercept is not variance should be determined and fixed.
significantly different from zero it can be left out of the
calculations. This is necessary because in case of very large 4.3.2. Limit of quantitation
intercept values negative apparent values of LoD can be obtained, Limit of quantitation (LoQ) is defined as the lowest concentra-
which do not have physical meaning [149]. tion of analyte that can be determined with an acceptable
Another approach to determine LoD is from the equation repeatability and trueness [15]. LoQ is called lower limit of
s quantification [19], limit of quantitation [15], limit of quantifica-
LoD ¼ 3:3  (6) tion [16,18,26], quantification limit [153], quantitation limit [17] or
b
limit of determination [15,16,20] in different standards and
where b is calibration graph slope and s is the standard deviation of
guidelines. As with LoD, this definition is also not fully rigorous,
the signal. s can be determined in 3 different ways. First, s can be
but a more rigorous substitute, which would be widely accepted, is
determined as standard deviation of an appropriate number of
not available.
blank sample signals. Second, s can be determined as residual
Note that by definition quantitation is possible at either LoD or
standard deviation of the calibration line in the LoD region. Third, s
LoQ but only the associated uncertainty becomes comparable to
can be determined as the standard deviation of intercept [17].
the actual result when approaching LoD [15]. Therefore, LoQ is just
Visual evaluation can also be used to determine LoD. This is
an indicative value as results below it are not devoid of information
done by performing analysis of samples with known analyte
and can be fit for purpose. In case the result of a measurement is
concentrations and by visually establishing the minimum level at
between LoD and LoQ it can be reported that the analyte presence
which the analyte can be reliably detected [17]. Usually peak
has been detected in the sample but is below LoQ [154]. Moreover,
shapes and heights vary between samples with the same
it is suggested that LoQ should be found by expressing the
concentration. Therefore multiple measurements should be made.
uncertainty of the measurement as a function of concentration and
Other approaches have been suggested in articles on LoD
comparing the results to the uncertainty levels demanded of that
determination. These, for example, use information theory to take
method [15,16]. In this case at least 10 repeated measurements
into account different distributions of noise in different analytical
should be made in each calibration point [15].
systems [150] or suggest reporting LoD with confidence limits
Approaches to determine LoD and LoQ are similar. In the latter
[151]. However these approaches are not widespread or generally
only a greater multiplication coefficient in the equation is used or
accepted.
other higher demands are set on the same parameters. For
The multitude of possibilities of LoD determination can make it
example, S/N value of at least 10 is required at the LoQ
difficult to choose the most suitable approach. In a recent study
concentration level [17]. When using the approach with equation
[149] careful comparison of 10 LoD determination approaches was
carried out by some of the authors of this review for the LC-MS/MS LoQ ¼ XðblankÞ þ k  sðblankÞ (7)
analysis of meropenem, doripenem and cilastatine. The LoD values
the same variables can be used and the same amount of analyzed
obtained with different approaches differed by up to an order of
samples is required. However, the coefficient k is required to have
magnitude. It was shown that comparison of LoD values found by
values of 5, 6 or 10 [15,18,19,20,26]. In case of the ICH approach,
different approaches is not meaningful. Based on the study results
using the calibration line, the equation
the recommended LoD determination approach is the one using
Eq. (6) where s is the residual standard deviation of the regression s
LoQ ¼ 10  (8)
line. This approach is recommended as it gives conservative LoD b
estimates while using measurements made for calibration graph. It is used where again all the variables can be taken from the same
is important that if samples with analyte content close to LoD are datasets for both LoD and LoQ [17]. When using visual evaluation,
suspected then calibration points in the range of LoD are included. the LoQ is taken as the lowest concentration level where the
This approach of LoD determination requires only little extra work analyte can be quantified with acceptable level of precision and
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 41

trueness. [17]. It has also been suggested that LoQ can be found by
multiplying LoD by 2 [16].
Another approach to determine LoQ is to find precision,
trueness or recovery of the method at multiple concentration
levels. LoQ can then be taken as the lowest concentration where
these parameters are fit for purpose or meet the requirements of
the necessary legislation [15,18,65]. For example, FDA requires that
the intensities of the peaks at LoQ must have precision of 20% and
trueness of 80–120% [18], and SANCO requires that mean recovery
is in range of 70–120% and relative standard deviation of at least
20% [65].
LoQ is determined in most approaches from the same data as
LoD or is based on LoD and therefore in principle the same issues
occur. Nevertheless, as LoQ is higher than LoD and therefore the
results in the LoQ range have lower relative uncertainty, which
makes LoQ determination more reliable.
In our experience, the most appropriate approach to determine
LoQ is by using the Eq. (8) suggested by ICH where s is taken as
standard deviation of the calibration line residuals in the low
concentration range. Moreover, LoQ should be determined 5 times
over a longer period and the most conservative result should be Fig. 1. Relation between permitted limit, CCa and CCb [66].

stated as the methods’ performance level to increase its reliability.


CCa ¼ X þ k  sR (9)
The exact way of determining LoQ should be specified as with LoD
due to the differences of the results when different approaches are where X is intercept of the calibration graph, k is 1.64 or 2.33,
used. Moreover, the methods’ performance at the LoQ level can be depending on whether a is taken as 0.05 or 0.01 respectively, and
monitored with regular analysis of samples (either real contami- sR is the between-day reproducibility of blank samples or at the
nated samples or spiked blank samples) with concentrations close permitted limit set for that analyte [66]. From here CCb can be
to LoQ. found as
CCb ¼ CCa þ k  sR (10)
4.3.3. Decision limit (CCa) and detection capability (CCb)
In order to determine CCa and CCb a critical concentration where k and sR are the same as for CCa calculations. At least 20
needs to be defined above which the sample is said to contain the replicate samples should be measured to calculate sR [66]. These
analyte and below which no analyte is said to be present. When a calculations do not take into account the increase of standard
permitted limit is not established for the analyte, the critical value deviation with the increase of concentration.
is defined as the concentration at which the probability of a blank As more replicate measurements and more specific definitions
sample being considered as a positive sample (containing analyte) are used here compared to the LoD determination approaches, the
is a. If a is 0.05 (this is the commonly used probability level) then results are more reliable. Therefore CCa and CCb should be used
this means that the critical value is the concentration value above when the analysis method is operating at analyte concentrations
which the results of 5% or less of the blank samples lie and are close to LoD and reliable decisions, whether the analyte is present
falsely considered positive. This critical value is called CCa [66]. or not, are necessary. Although this approach increases the time
However, when the analyte level in the sample is indeed above CCa needed to determine LoD due to its superior reliability it is widely
then there is a danger of obtaining a result that is below CCa and is used in method validation [110,156,157].
therefore falsely counted as negative. This danger is not addressed
by CCa. Therefore, after finding CCa we can define an analyte Acknowledgments
concentration at which the probability of getting a falsely negative
result is b. This concentration level is often termed as CCb. If b is This work was supported by the institutional research funding
0.05 then CCb means a concentration where the analysis of the IUT20-14 and personal research funding PUT34 from the Estonian
sample would give results under the critical value only 5% of the Ministry of Education and Research, as well as the Estonian Science
times. CCb can be interpreted as LoD and some guidelines define Foundation grant No 8572 and project “Development of software
LoD this way [153,155] others term this concentration as CCb [66]. for validation of chromatographic methods” (with registration
In case a permitted limit for the analyte is established, the number 3.2.1201.13-0020) under the sub-measure “Supporting the
critical value is found by analyzing blank samples that are development of R&D of info and communication technology”
spiked at that permitted limit, not from the blank samples. funded by the EU Regional Development Fund.
Therefore, if a is taken as 0.05, CCa is the concentration above
which 5% of the highest results lie (and are considered falsely References
positive) that are obtained for samples with analyte concentra-
tion at (i.e., not exceeding) the permitted limit. CCb is then the [1] J.H. Gross, Mass Spectrometry, second ed., Springer, Heidelberg, 2011.
concentration at which the probability of obtaining results under [2] W.A. Korfmacher, Foundation Review: principles and applications of LC-MS
the respective CCa (i.e., obtaining false negatives) is 5% (if b is in new drug discovery, Drug Discovery Today 10 (2005) 1357–1367.
[3] B.W. Brooks, D.B. Huggett, Human Pharmaceuticals in the Environment.
taken as 0.05) [66]. The explanation of CCa and CCb can be found Current and Future Perspectives, Springer, New York, 2012.
in Fig. 1. [4] A. Kruve, K. Herodes, I. Leito, Optimization of electrospray interface and
By many standards and guidelines calculating CCa and CCb quadrupole ion trap mass spectrometer parameters in pesticide liquid
chromatography/electrospray ionization mass spectrometry analysis, Rapid
needs complex understanding of the statistics behind these Commun. Mass Spectrom. 25 (2010) 1159–1168.
definitions [155]. However simpler methods exist that are [5] M. Jemal, Y.Q. Xia, LC–MS development strategies for quantitative
presented here. CCa can be defined as bioanalysis, Curr. Drug Metab. 7 (2006) 491–502.
42 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

[6] I. Taverniers, M. De Loose, E. Van Bockstaele, Trends in quality in the [33] A. Raffaell, A. Saba, Atmospheric pressure photoionization mass
analytical laboratory. II. Analytical method validation and quality assurance, spectrometry, Mass Spectrom. Rev. 22 (5) (2003) 318–331.
TrAC Trends Anal. Chem. 23 (2004) 535–552. [34] D.B. Robb, M.W. Blades, State-of-the-art in atmospheric pressure
[7] E. Trullols, I. Ruisánchez, F.X. Rius, Validation of qualitative analytical photoionization for LC/MS, Anal. Chem. 627 (2008) 34–49.
methods, TrAC Trends Anal. Chem. 23 (2004) 137–145. [35] A. Kruve, K. Kaupmees, J. Liigand, I. Leito, Sodium adduct formation efficiency
[8] I.R. Pizzutti, A. de Kok, R. Zanella, M.B. Adaime, M. Hiemstra, C. Wickert, O.D. in ESI source, J. Mass Spectrom. 48 (2013) 695–702.
Prestes, Method validation for the analysis of 169 pesticides in soya grain [36] B.K. Choi, A.I. Gusev, D.M. Hercules, Postcolumn introduction of an internal
without clean up, by liquid chromatography–tandem mass spectrometry standard for quantitative LC–MS analysis, Anal. Chem. 71 (1999)
using positive and negative electrospray ionization, J. Chromatogr. A 1142 4107–4110.
(2007) 123–136. [37] F.M. Lagerwerf, W.D. van Dongen, R.J.J.M. Steenvoorden, M. Honing, J.H.G.
[9] A. Van Eeckhaut, K. Lanckmans, S. Sarre, I. Smolders, Y. Michotte, Validation of Jonkman, Exploring the boundaries of bioanalytical quantitative LC–MS–MS,
bioanalytical LC-MS/MS assays: evaluation of matrix effects, J. Chromatogr. B Trends Anal. Chem. 19 (7) (2000) 418–427.
877 (2009) 2198–2207. [38] W.M.A. Niessen, P. Manini, R. Andreoli, Matrix effects in quantitative
[10] E. Rogatsky, D. Stein, Evaluation of matrix effect and chromatography pesticide analysis using liquid chromatography–mass spectrometry, Mass
efficiency: new parameters for validation of method development, J. Am. Soc. Spectrom. Rev. 25 (2006) 881–899.
Mass Spectrom. 16 (2005) 1757–1759. [39] G.G. Guilbault, M. Hjelm, Nomenclature for automated and mechanised
[11] ISO/IEC 17025,2005, General requirements for the competence of testing and analysis, Pure Appl. Chem. 61 (1989) 1657–1664.
calibration laboratories. [40] B.K. Matuszewski, M.L. Constanzer, C.M. Chavez-Eng, Strategies for the
[12] GLP–OECD Series on Principles of Good Laboratory Practice (GLP) and assessment of matrix effect in quantitative bioanalytical methods based on
Compliance Monitoring. HPLC-MS/MS, Anal. Chem. 75 (2003) 3019–3030.
[13] Commission directive 2009/90/EC: Laying down, pursuant to Directive 2000/ [41] P.J. Taylor, Matrix effects: the Achilles heel of quantitative high-performance
60/EC of the European Parliament and of the Council, technical specifications liquid chromatography–electrospray–tandem mass spectrometry, Clin.
for chemical analysis and monitoring of water status. Off. J. Eur. Union, 2009. Biochem. 38 (2005) 328–334.
[14] P. Araujo, Key aspects of analytical method validation and linearity [42] H. Trufelli, P. Palma, G. Famiglini, A. Capiello, An overview of matrix effects in
evaluation, J. Chromatogr. B 877 (2009) 2224–2234. liquid chromatography–mass spectrometry, Mass Spectrom. Rev. 30 (2011)
[15] (a) EurachemGuide: The Fitness for Purpose of Analytical Methods, 491–509.
Eurachem, Teddington, 1998. (b) B. Magnusson and U. Örnemark (Eds.) [43] H. Mei, Y. Hsieh, C. Nardo, X. Xu, S. Wang, K. Ng, W.A. Korfmacher,
Eurachem Guide: The Fitness for Purpose of Analytical Methods – A Investigation of matrix effects in bioanalytical high-performance liquid
Laboratory Guide to Method Validation and Related Topics, (second ed. 2014). chromatography/tandem mass spectrometric assays: application to drug
ISBN 978-91-87461-59-0. Available from www.eurachem.org. discovery, Rapid Commun. Mass Spectrom. 17 (2003) 97–103.
[16] Harmonized Guidelines for single-laboratory validation of method of [44] L. Alder, S. Lüderitz, K. Lindtner, S.-H. Stan, The ECHO technique–the more
analyses (IUPAC Technical Report), Pure Appl. Chem. 74(5) (2002) 835–855. effective way of data evaluation in liquid chromatography–tandem mass
[17] ICH harmonized tripartite guideline: validation of analytical procedures: text spectrometry analysis, J. Chromatogr. A 1058 (2004) 67–79.
and methodology Q2(R1), International Conference of harmonization of [45] A. Kruve, K. Herodes, I. Leito, Combating matrix effects in LC/ESI/MS: the
technical requirements for registration of pharmaceuticals for human use, extrapolative dilution approach, Anal. Chim. Acta 651 (2009) 75–80.
2005. [46] A. Kruve, I. Leito, Comparison of different methods aiming to account for/
[18] (a)Guidance for Industry: Bioanalytical method validation, U.S. Department overcome matrix effects in LC/ESI/MS on the example of pesticide analyses,
of Health and Human Services Food and Drug Administration, 2001. Anal. Methods 5 (2013) 3035–3044.
(b) Guidance for Industry: Bioanalytical method validation, Draft Guidance, U. [47] R. Bonfiglio, R.C. King, T.V. Olah, K. Merkle, The effects of sample preparation
S. Department of Health and Human Services Food and Drug methods on the variability of the electrospray ionization response for model
Administration http://www.fda.gov/downloads/Drugs/ drug compounds, Rapid Commun. Mass Spectrom. 13 (1999) 1175–1185.
GuidanceComplianceRegulatoryInformation/Guidances/UCM368107.pdf, [48] S. Souverain, S. Rudaz, J.-L. Veuthey, Matrix effect in LC–ESI–MS and LC–
2013. APCI–MS with off-line and on-line extraction procedure, J. Chromatogr. A
[19] Guidance on Bioanalytical Method Validation, European Medicines Agency, 1058 (2004) 61–66.
2011. [49] R. Dams, M.A. Huestis, W.E. Lambert, C.M. Murphy, Matrix effect in bio-
[20] AOAC Guidelines for Single-Laboratory Validation of Chemical Methods for analysis of illicit drugs with LC-MS/MS: influence of ionization type, sample
Dietary Supplements and Botanicals, Official Methods of Analysis, nineteenth preparation, and biofluid, J. Am. Soc. Mass Spectrom. 14 (2003) 1290–1294.
ed., Appendix K, AOAC, INTERNATIONAL, Gaithersburg, MD, 2012. [50] P. Keski-Rahkonen, K. Huhtinen, R. Desai, D.T. Harwood, D.J. Handelsman, M.
[21] S. Chandran, R.S.P. Singh, Comparison of various international guidelines for Poutanen, S. Auriola, LC–MS analysis of estradiol in human serum and
analytical method validation, Pharmazie 62 (2007) 4–14. endometrial tissue: comparison of electrospray ionization, atmospheric
[22] R. Bonfilio, E.C.L. Cazedey, M.B. de Araújo, H.R.N. Salgado, Analytical pressure chemical ionization and atmospheric pressure photoionization, J.
validation of quantitative high-performance liquid chromatographic Mass Spectrom. 48 (2013) 1050–1058.
methods in pharmaceutical analysis: a practical approach, Crit. Rev. Anal. [51] O.A. Ismaiel, M.S. Halquist, M.Y. Elmamly, A. Shalaby, H.T. Karnes, Monitoring
Chem. 42 (2012) 87–100. phospholipids for assessment of ion enhancement and ion suppression in ESI
[23] E. Rozet, R.D. Marini, E. Ziemons, B. Boulanger, P. Hubert, Advances in and APCI LC/MS/MS for chlorpheniramine in human plasma and the
validation, risk and uncertainty assessment of bioanalytical methods, J. importance of multiple source matrix effect evaluations, J. Chromatogr. B
Pharm. Biomed. 55 (2011) 848–858. 875 (2008) 333–343.
[24] G.A. Shabir, Validation of high-performance liquid chromatography methods [52] E. Beltrán, M. Ibánez, J.V. Sancho, F. Hernández, Determination of patulin in
for pharmaceutical analysis: understanding the differences and similarities apple and derived products by UHPLC-MS/MS. Study of matrix effects with
between validation requirements of the US Food and Drug Administration, atmospheric pressure ionisation sources, Food Chem. 142 (2014) 400–407.
the US Pharmacopeia and the International Conference on Harmonization, J. [53] E.M. Thurman, I. Ferrer, D. Barcelo, Choosing between atmospheric pressure
Chromatogr. A 987 (2003) 57–66. chemical ionization and electrospray ionization interfaces for the HPLC/MS
[25] F.T. Peters, Method validation using LC–MS, in: A. Polettini (Ed.), Applications analysis of pesticides, Anal. Chem. 73 (2001) 5441–5449.
of Liquid Chromatography–Mass Spectrometry in Toxicology, [54] A. Kloepfer, J.B. Quintana, T. Reemtsma, Operational options to reduce matrix
Pharmaceutical Press, London, 2006, pp. 71–96. effects in liquid chromatography–electrospray ionization-mass
[26] NordVal Protocol No. 2, Guide in validation of alternative propietary chemical spectrometry analysis of aqueous environmental samples, J. Chromatogr.
methods, 2010. A 1067 (2005) 153–160.
[27] O. González, M.E. Blanco, G. Iriarte, L. Bartolomé, M.I. Maguregui, R.M. Alonso, [55] E. Chambers, D.M. Wagrowski-Diehl, Z. Lu, J.R. Mazzeo, Systematic and
Bioanalytical chromatographic method validation according to current comprehensive strategy for reducing matrix effects in LC/MS/MS analyses, J.
regulations with a special focus on the non-well defined parameters limit Chromatogr. B 852 (2007) 22–34.
of quantification, robustness and matrix effect, J. Chromatogr. A 1353 (2014) [56] J.V. Sancho, O.J. Pozo, F.J. Lopez, F. Hernandez, Different quantitation
10–27. approaches for xenobiotics in human urine samples by liquid
[28] International Vocabulary of Metrology–Basic and General Concepts and chromatography/electrospray tandem mass spectrometry, Rapid Commun.
Associated Terms (VIM), JCGM 200:2012. Mass Spectrom. 16 (2002) 639–645.
[29] A. Kruve, R. Rebane, K. Kipper, M.-L. Oldekop, H. Evard, K. Herodes, P. Ravio, I. [57] A.I. Garcia-Valcarcel, J.L. Tadeo, A combination of ultrasonic assisted
Leito, Tutorial review on validation of liquid chromatography–mass extraction with LC-MS/MS for the determination of organophosphorus
spectrometry methods: Part II Anal. Chim. Acta, http://dx.doi.org/10.1016/ pesticides in sludge, Anal. Chim. Acta 641 (2009) 117–123.
j.aca.2015.02.016. [58] H. Stahnke, S. Kittlaus, G. Kempe, L. Alder, Reduction of matrix effects in liquid
[30] R.D. Smith, J.A. Loo, C.G. Endmonds, C.J. Barinaga, H.R. Udseth, New chromatography-electrospray ionization-mass spectrometry by dilution of
developments in biochemical mass spectrometry: electrospray ionization, the sample extracts: how much dilution is needed? Anal. Chem. 84 (2012)
Anal. Chem. 62 (9) (1990) 882–899. 1474–1484.
[31] N.B. Cech, C.G. Enke, Practical implications of some recent studies in [59] J. Zrostlikova, J. Hajšlova, J. Pouska, P. Begany, Alternative calibration
electrospray ionization fundamentals, Mass Spectrom. Rev. 20 (2001) 362–387. approaches to compensate the effect of co-extracted matrix components in
[32] R. Kostiainen, T. Kotiaho, T. Kuuranne, S. Auriola, Liquid chromatography/ liquid chromatography–electrospray ionisation tandem mass spectrometry
atmospheric pressure ionization-mass spectrometry in drug metabolism analysis of pesticide residues in plant materials, J. Chromatogr. A 973 (2002)
studies, J. Mass Spectrom. 38 (4) (2003) 357–372. 13–26.
A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44 43

[60] A. Kaufmann, P. Butcher, Segmented post-column analyte addition; a concept analysis of melamine migration from melamine tableware, Talanta 10 (2012)
for continuous response control of liquid chromatography/mass 453–459.
spectrometry peaks affected by signal suppression/enhancement, Rapid [84] P. Montoro, M. Maldini, M. Russo, S. Postorino, S. Piacente, C. Pizza, Metabolic
Commun. Mass Spectrom. 19 (5) (2005) 611–617. profiling of roots of liquorice (Glycyrrhiza glabra) from different geographical
[61] K.K. Murray, R.K. Boyd, M.N. Eberlin, G.J. Langley, L. Li, Y. Naito, Definitions of areas by ESI/MS/MS and determination of major metabolites by LC–ESI/MS
terms relating to mass spectrometry (IUPAC recommendations 2013), Pure and LC–ESI/MS/MS, J. Pharm. Biomed. Anal. 54 (2011) 535–544.
Appl. Chem. 85 (7) (2013) 1515–1609. [85] M.J. Rumpler, Quantitative analysis of 11-nor-9-carboxy-
[62] A. Singleton, Recent advances in bioanalytical sample preparation for LC–MS tetrahydrocannbinol(THC-COOH) in urine by LC-MS/MS following a simple
analyses, Bioanalysis 4 (9) (2012) 1123–1140. filtration, J. Chromatogr. B 957 (2014) 77–83.
[63] A. Tan, S. Hussain, A. Musuku, R. Masse, Internal standard response variations [86] N. Venisse, C. Grignon, B. Brunet, S. Thévenot, A. Bacle, V. Migeot, A. Dupuis,
during incurred sample analysis by LC-MS/MS: case by case trouble- Reliable quantification of bisphenol A and its chlorinated derivatives in
shooting, J. Chromatogr. B 877 (27) (2009) 3201–3209. human urine using UPLC-MS/MS method, Talanta 125 (2014) 284–292.
[64] F.T. Peters, O.H. Drummer, F. Musshoff, Validation of new methods, Forensic [87] E.N. Ediage, J.D. Di Mavungu, S. Monbaliu, C. Van Peteghem, S. De Saeger, A
Sci. Int. 165 (2007) 216–224. validated multianalyte LC-MS/MS method for quantification of
[65] SANCO/12571/2013, Guidance document on analytical quality control and 25 mycotoxins in cassava flour, peanut cake and maize samples, J. Agric.
validation procedures for pesticide residues analysis in food and feed. 2013. Food Chem. 9 (2011) 5173–5180.
[66] European Commission Decision 2002/657/EC implementing Council [88] S.C. Utture, K. Banerjee, S. Dasgupta, S.H. Patil, M.R. Jadhav, S.S. Wagh, S.S.
Directive 96/23/EC concerning the performance of analytical methods and Kolekar, M.A. Anuse, P.G. Adsule, Dissipation and Distribution Behavior of
the interpretation of results, Off. J. Eur. Commun. L221 (2002) 8–36. Azoxystrobin, Carbendazim, and Difenoconazole in Pomegranate Fruits, J.
[67] E.I. Miller, F.M. Wylie, J.S. Oliver, Simultaneous detection and quantification Agric. Food Chem. 59 (2011) 7866–7873.
of amphetamines, diazepam and its metabolites cocaine and its metabolites, [89] M.M. De Zan, M.S. Cámara, J.C. Robles, S.V. Kergaravat, H.C. Goicoechea,
and opiates in hair by LC–ESI-MS–MS using a single extraction method, J. Development and validation of a simple stability-indicating high performance
Anal. Toxicol. 32 (2008) 457–469. liquid chromatographic method for the determination of miconazole nitrate in
[68] A.V. Ramani, P. Sengupta, R. Mullangi, Development and validation of a highly bulk and cream formulations, Talanta 79 (2009) 762–767.
sensitive and robust LC–ESI–MS/MS method for simultaneous quantitation of [90] C. Kmellár, P. Fodor, L. Pareja, C. Ferrer, M.A. Martínez-Uroz, A. Valverde, A.R.
simvastatin acid, amlodipine and valsartan in human plasma: application to a Fernandez-Alba, Validation and uncertainty study of a comprehensive list of
clinical pharmacokinetic study, Biomed. Chromatogr. 23 (2009) 615–622. 160 pesticide residues in multi-class vegetables by liquid chromatography–
[69] J. Feng, X.-W. Yang, R.-B. Huang, H.-Y. Zhang, M. He, Q.-C. Huang, tandem mass spectrometry, J. Chromatogr. A 1215 (2008) 37–50.
Development and validation of an LC–ESI–MS/MS method for the [91] K.P. Yánez, M.T. Martin, J.L. Bernal, M.J. Nozal, J. Bernal, Determination of
determination of nitidine chloride in rat plasma, J. Chromatogr. B 887-888 spinosad at trace levels in bee pollen and beeswax with solid–liquid
(2012) 43–47. extraction and LC–ESI-MS, J. Sep. Sci. 37 (2014) 204–210.
[70] J. Beyer, F.T. Peters, T. Kraemer, H. Maurer, Detection and validated [92] R.N. Rao, P.K. Maurya, M. Ramesh, R. Srinivas, S.B. Agwane, Development of a
quantification of nine herbal phenalkylamines and methcathinone in human validated high-throughput LC–ESI–MS method for determination of
blood plasma by LC-MS/MS with electrospray ionization, J. Mass Spectrom. sirolimus on dried blood spots, Biomed. Chromatogr. 24 (2010) 1356–1364.
42 (2007) 150–160. [93] M.J. Bogusz, H. Hassan, E. Al-Enazi, Z. Ibrahim, M. Al-Tufail, Application of LC–
[71] A. Gupta, P. Singhal, P.S. Shrivastav, M. Sanyal, Application of a validated ultra ESI–MS–MS for detection of synthetic adulterants in herbal remedies, J.
performance liquid chromatography–tandem mass spectrometry method for Pharm. Biomed. Anal. 41 (2006) 554–564.
the quantification of darunavir in human plasma for a bioequivalence study [94] H. Yamaguchi, A. Fujikawa, H. Ito, T. Tanaka, A. Furugen, K. Miyamori, N.
in Indian subjects, J. Chromatogr. B 879 (2011) 2443–2453. Takahashi, J. Ogura, M. Kobayashi, T. Yamada, N. Mano, K. Iseki, A rapid and
[72] H.N. Mistri, A.G. Jangid, A. Pudage, N. Gomes, M. Sanyal, P. Shrivastav, High sensitive LC/ESI–MS/MS method for quantitative analysis of docetaxel in
throughput LC-MS/MS method for simultaneous quantification of human plasma and its application to a pharmacokinetic study, J. Chromatogr.
lamivudine, stavudine and nevirapine in human plasma, J. Chromatogr. B B 893–894 (2012) 157–161.
879 (2007) 320–332. [95] A. Claus, G.M. Weisz, D.R. Kammerer, R. Carle, A. Schieber, A method for the
[73] H.F. Schröder, W. Gebhardt, M. Thevis, Anabolic doping, and lifestyle drugs, determination of acrylamide in bakery products using ion trap LC–ESI–MS/
and selected metabolites in wastewater-detection, quantification, and MS, Mol. Nutr. Food Res. 49 (2005) 918–925.
behaviour monitored by high-resolution MS and MSn before and after [96] M. Gros, M. Petrovi c, D. Barceló, Development of a multi-residue analytical
sewage treatment, Anal. Bioanal. Chem. 398 (2010) 1207–1229. methodology based on liquid chromatography–tandem mass spectrometry
[74] D. Yeniceli, E. Sener, O.T. Korkmaz, D. Dogrukol-Ak, N. Tuncel, A simple and (LC-MS/MS) for screening and trace level determination of pharmaceuticals
sensitive LC–ESI–MS (ion trap) method for the determination of bupropion in surface and wastewaters, Talanta 70 (2006) 678–690.
and its major metabolite, hydroxybupropion in rat plasma and brain [97] A. Sannino, L. Bolzoni, M. Bandini, Application of liquid chromatography with
microdialysates, Talanta 84 (2011) 19–26. electrospray tandem mass spectrometry to the determination of a new
[75] F.P. Gomes, P.L. García, J.M.P. Alves, A.K. Singh, E.R.M. Kedor-Hackmann, M.I.R. generation of pesticides in processed fruits and vegetables, J. Chromatogr. A
M. Santoro, Development and validation of stability-indicating HPLC 1036 (2004) 161–169.
methods for quantitative determination of pravastatin fluvastatin, [98] J. Wang, Determination of Five Macrolide Antibiotic Residues in Honey by LC–
atorvastatin, and rosuvastatin in pharmaceuticals, Anal. Lett. 42 (2009) ESI–MS and LC–ESI–MS/MS, J. Agric. Food Chem. 52 (2004) 171–181.
1784–1804. [99] G.A. Shabir, W.J. Lough, S.A. Arain, T.K. Bradshaw, Evaluation and application
[76] F. Calbiani, M. Careri, L. Elviri, A. Mangia, L. Pistara, I. Zagnoni, Development of best practice in analytical method validation, J. Liq. Chromatogr. Relat.
and in-house validation of a liquid chromatography–electrospray–tandem Technol. 20 (2007) 311–333.
mass spectrometry method for the simultaneous determination of Sudan I, [100] E. Rozet, A. Ceccato, C. Hubert, E. Ziemons, R. Oprean, S. Rudaz, B. Boulanger, P.
Sudan II Sudan III and Sudan IV in hot chilli products, J. Chromatogr. A 1042 Hubert, Analysis of recent pharmaceutical regulatory documents on
(2004) 123–130. analytical method validation, J. Chromatogr A. 1158 (2007) 111–125.
[77] A. Angioni, L. Porcu, F. Pirisi, LC/DAD/ESI/MS Method for the determination of [101] IUPAC, Compendium of Chemical Terminology, 2nd ed., (the Gold Book).
imidacloprid thiacloprid, and spinosad in olives and olive oil after field Compiled by A.D. McNaught, A. Wilkinson. Blackwell Scientific Publications,
treatment, J. Agric. Food Chem. 59 (2011) 11359–11366. Oxford (1997). XML on-line corrected version: http://goldbook.iupac.org
[78] J. Ascari, S. Dracz, F.A. Santos, J.A. Lima, M.H.G. Diniz, E.A. Vargas, Validation of (2006-) created by M. Nic, J. Jirat, B. Kosata, updates compiled by A. Jenkins.
an LC-MS/MS method for malachite green (MG) leucomalachite green (LMG), ISBN 0-9678550-9-8. doi:10.1351/goldbook, last update: 24-02-2014;
crystal violet (CV) and leucocrystal violet (LCV) residues in fish and shrimp, version: 2.3.3.
Food Addit. Contam. 29 (2012) 602–608. [102] C.J. Anagnostopoulos, P. Aplada Sarli, K. Liapis, S.A. Haroutounian, G.E.
[79] M. Careri, C. Corradini, L. Elviri, I. Nicoletti, I. Zagnoni, Liquid Miliadis, Validation of two variations of the QuEChERS method for the
chromatography-electrospray tandem mass spectrometry of cis-resveratrol determination of multiclass pesticide residues in cereal-based infant foods
and trans-resveratrol: development, validation, and application of the by LC-MS/MS, Food Anal. Method 5 (2012) 664–683.
method to red wine, grape, and winemaking byproducts, J. Agric. Food Chem. [103] S.D. Richardson, Environmental mass spectrometry: emerging contaminants
52 (2004) 6868–6874. and current issues, Anal. Chem. 82 (12) (2010) 4742–4774.
[80] H.G.F. Smienk, D. Calvo, P. Razquin, E. Domínguez, L. Mata, Single laboratory [104] R. Diaz, M. Ibáñez, J.V. Sancho, F. Hernández, Qualitative validation of a liquid
validation of a ready-to-use phosphatase inhibition assay for detection of chromatography–quadrupole-time of flight mass spectrometry screening
okadaic acid toxins, Toxins 4 (2012) 339–351. method for organic pollutants in waters, J. Chromatogr. A 1276 (2013) 47–57.
[81] B. Cordero, F. Canale, D. Del Rio, C. Bicchi, Identification quantitation,and [105] J. Nácher-Mestre, M. Ibáñez, R. Serrano, J. Pérez-Sánchez, F. Hernández,
method validation for flavan-3-ols in fermented ready-to-drink teas from the Qualitative screening of undesirable compounds from feeds to fish by liquid
Italian market using HPLC-UV/DAD and LC-MS/MS, J. Sep. Sci. 32 (2009) chromatography coupled to mass spectrometry, J. Agric. Food Chem. 61 (9)
3643–3651. (2013) 2077–2087.
[82] A.J. Giron, K. Deventer, P. Van Eenoo, Development and validation of an open [106] R. Macarthur, C. von Holst, A protocol for the validation of qualitative
screening method for diuretics, stimulants and selected compounds in methods of detection, Anal. Methods 4 (2012) 2744–2754.
human urine by UHPLC–HRMS for doping control, Anal. Chim. Acta 721 [107] H.G. Mol, P. Zomer, M. de Koning, Qualitative aspects and validation of a
(2012) 137–146. screening method for pesticides in vegetables and fruits based on liquid
[83] M. Mattarozzi, M. Milioli, C. Cavalieri, F. Bianchi, M. Careri, Rapid desorption chromatography coupled to full scan high resolution (orbitrap) mass
electrospray ionization-high resolution mass spectrometry method for the spectrometry, Anal. Bioanal. Chem. 403 (10) (2012) 2891–2908.
44 A. Kruve et al. / Analytica Chimica Acta 870 (2015) 29–44

[108] H. Kadar, B. Veyrand, J.P. Antignac, S. Durand, F. Monteau, B. Le Bizac, [133] B. Dejaegher, Y. Vander Heyden, Ruggedness and robustness testing, J.
Comparative study of low- versus high-resolution liquid chromatography- Chromatogr. A 1158 (2007) 138–157.
mass spectrometric strategies for measuring perfluorinated contaminants in [134] C. Benetti, R. Piro, G. Binato, R. Angeletti, G. Biancotto, Simultaneous
fish, Food Addit. Contam. A 28 (9) (2011) 1261–1273. determination of lincomycin and five macrolide antibiotic residues in honey
[109] R.J. Singh, R.L. Taylor, G. Satyanarayana Reddy, S.K.G. Grebe, C-3 Epimers can by liquid chromatography coupled to electrospray ionization mass
account for a Significant proportion of total circulating 25-hydroxyvitamin D spectrometry (LC-MS/MS), Food Addit. Contam. 23 (11) (2006) 1099–1108.
in infants, complicating accurate measurement and interpretation of vitamin [135] P. Jedziniak, T. Szprengier-Juszkiewicz, M. Olejnik, Determination of
D status, J. Clin. Endocr. Metab. 91 (8) (2006) 3055–3061. benzimidazoles and levamisole residues in milk by liquid chromatography–
[110] A. Gentili, F. Caretti, S. Bellante, L.M. Rocca, R. Curini, A. Venditti, mass spectrometry: screening method development and validation, J.
Development and validation of two multiresidue liquid chromatography Chromatogr. A 1216 (2009) 8165–8172.
tandem mass spectrometry methods based on a versatile extraction [136] R. Zeleny, S. Harbeck, H. Schimmel, Validation of a liquid chromatography–
procedure for isolating non-steroidal anti-inflammatory drugs from tandem mass spectrometry method for the identification and quantification
bovine milk and muscle tissue, Anal. Bioanal. Chem. 404 (2012) 1375–1388. of 5-nitroimidazole drugs and their corresponding hydroxy metabolites in
[111] A. Domenech, N. Cortes-Francisco, O. Palacios, J.M. Franco, P. Riobo, J.J. lyophilised pork meat, J. Chromatogr. A 1216 (2009) 249–256.
Lierena, S. Vichi, J. Caixach, Determination of lipophilic marine toxins in [137] M. Gasparini, M. Curatolo, W. Assini, E. Bozzoni, N. Tognoli, G. Dusi,
mussels. Quantification and confirmation criteria using high resolution mass Confirmatory method for the determination of nandrolone and trenbolone in
spectrometry, J. Chromatogr. A 1328 (2014) 16–25. urine samples using immunoaffinity cleanup and liquid chromatography–
[112] Ó.J. Pozo, J.V. Sancho, M. Ibáñez, F. Hernández, W.M.A. Niessen, Confirmation tandem mass spectrometry, J. Chromatogr. A 1216 (2009) 8059–8066.
of organic micropollutants detected in environmental samples by liquid [138] C.C.G.O. Lopes, H.R.N. Salgado, Development of a validated stability-
chromatography tandem mass spectrometry: achievements and pitfalls, indicating LC assay and stress degradation studies of linezolid in tablets,
TrAC Trends Anal Chem. 25 (10) (2006) 1030–1042. Chromatographia 69 (2009) S129–S135.
[113] R. Bethem, J. Boison, J. Gala, D. Heller, S. Lehotay, J. Loo, S. Musser, P. Price, S. [139] N. Kaul, H. Agrawal, A.R. Paradkar, K.R. Mahadik, The ICH guidance in
Stein, Establishing the fitness for purpose of mass spectrometric methods, J. practice: stress degradation studies on indinavir sulphate and development
Am. Soc. Mass Spectrom. 14 (2003) 528–541. of a validated specific stability-indicating HPTLC assay method, Il Farmaco 59
[114] J.A. Van Leeuwen, L.M.C. Buydens, B.G.M. Vandeginste, G. Kateman, P.J. (2004) 729–738.
Schoenmakers, M. Mulholland, RES, an expert system for the set-up and [140] L.B. Junior, F.T. Uchoa, S.S. Guterres, T.D. Costa, Development and validation of
interpretation of a ruggedness test in HPLC method validation. Part 1: the LC-MS/MS method for the simultaneous determination of quinine and
ruggedness test in HPLC method validation, Chemom. Intell. Lab. Syst. 10 doxycycline in pharmaceutical formulations, J. Liq. Chromatogr. Relat.
(1991) 337–347. Technol. 1 (2009) 2699–2711.
[115] E. Karageorgou, V. Samanidou, Youden test application in robustness assays [141] C. Yardimci, N. Özaltın, Method development and validation for the
during method validation, J. Chromatogr. A 65 (2014) 234–240. simultaneous determirnation of rosiglitazone and metformin in
[116] W.J. Youden, E.H. Steiner, Statistical manual of the association of official, Anal. pharmaceutical preparations by capillary zone electrophoresis, Anal.
Chem. (1975) 50–55. Chim. Acta 549 (2005) 88–95.
[117] R. Boque, A. Maroto, J. Riu, X. Rius, Validation of analytical methods, Grasas [142] S. Thomas, S. Shandilya, A. Bharati, S.K. Paul, A. Agarwal, C.S. Mathela,
Aceites 53 (1) (2002) 128–143. Identification, characterization and quantification of new impurities by
[118] V.V. Heyden, A. Nijhuis, J. Smeyers-Verbeke, B.G.M. Vandeginste, D.L. LC–ESI/MS/MS and LC–UV methods in rivastigmine tartrate active
Massart, Guidance for robustness/ruggedness tests in method validation, J. pharmaceutical ingredient, J. Pharm. Biomed. Anal. 57 (2012) 39–51.
Pharm. Biomed. Anal. 24 (2001) 723–753. [143] C. von Holst, A. Müller, E. Björklund, E. Anklam, In-house validation of a
[119] L. Cuadros-Rodriguez, R. Romero, J.M. Bosque-Sendra, Studies in research and simplified method for the determination of PCBs in food and feedingstuffs,
development of analytical processes, Crit. Rev. Anal. Chem. 35 (1) (2005) Eur. Food Res. Technol. 213 (2001) 154–160.
57–69. [144] N.A. Épshtein, Validation of HPLC techniques for pharmaceutical analysis,
[120] J. Caporal-Gautier, J.M. Nivet, P. Algranti, M. Guilloteau, M. Histe, M. Lallier, J.J. Pharm. Chem. J. 38 (2004) 212–228.
N’Guyen-Huu, R. Russotto, Guide de validation analytique, rapport d’une [145] T. Mirza, H.S.I. Tan, Determination of captopril in pharmaceutical tablets by
commission SFSTP, S. T. P. Pharma. Pratiques 2 (1992) 205–239. anion-exchange HPLC using indirect photometric detection; a study in
[121] The United States Pharmacopeia, 23rd ed., National Formulary 18, United systematic method development, J. Pharm. Biomed. Anal. 25 (1) (2001) 39–52.
States Pharmacopeial Convention, Rockville, USA, 1995. [146] G. Szekely, B. Henriques, M. Gil, C. Alvarez, Experimental design for the
[122] The United States Pharmacopeia, 35th ed., Rockville, The United States optimization and robustness testing of a liquid chromatography tandem
Pharmacopeial Convention, Inc., 2012, 3261–3279. mass spectrometry method for the trace analysis of the potentially genotoxic
[123] E. Aguilera, R. Lucena, S. Cardenas, M. Valcarcel, E. Trullols, I. Ruisanchez, 1,3-diisopropylurea, Drug Test. Anal. 6 (2014) 898–908.
Robustness in qualitative analyses: a practical approach, Trac-Trend Anal. [147] F. Calbiani, M. Careri, L. Elviri, A. Mangia, I. Zagnoni, Validation of an ion-pair
Chem. 25 (6) (2006) 621–627. liquid chromatography-electrospray-tandem mass spectrometry method for
[124] B. Sundström, G. Johansson, H. Kokkonen, T. Cederholm, S. Wållberg-Jonsson, the determination of heterocyclic aromatic amines in meat-based infant
Plasma phospholipid fatty acid content is related to disease activity in foods, Food Addit. Contam. 24 (8) (2007) 833–841.
ankylosing spondylitis, J. Rheumatol. 39 (2012) 327–333. [148] L.A. Currie, Limits for qualitative detection and quantitative determination,
[125] A.C. Thiébaut, M. Rotival, E. Gauthier, G.M. Lenoir, M.C. Boutron-Ruault, V. Anal. Chem. 40 (1968) 586–593.
Joulin, F. Clavel-Chapelon, V. Chajès, Correlation between serum [149] H. Evard, A. Kruve, I. Leito, LoD determination in LC/MS/MS: choosing the
phospholipid fatty acids and dietary intakes assessed a few years earlier, appropriate methodology and comparability of the results, J. Chromatogr. A
Nutr. Cancer 61 (2009) 500–509. (2015) submitted for publication.
[126] C.E. Childs, M. Romeu-Nadal, G.C. Burdge, P.C. Calder, The polyunsaturated [150] J. Fonollosa, A. Vergara, R. Huerta, S. Marco, Estimation of the limit of detection
fatty acid composition of hepatic and plasma lipids differ by both sex and using information theory measures, Anal. Chim. Acta 810 (2014) 1–9.
dietary fat intake in rats, J. Nutr. 140 (2010) 245–250. [151] W. Huber, Basic calculations about the limit of detection and its optimal
[127] P. Bastos-Amador, F. Royo, E. Gonzalez, J. Conde-Vancells, L. Palomo-Diez, F.E. determination, Accred. Qual. Assur. 8 (2003) 213–217.
Borras, J.M. Falcon-Perez, Proteomic analysis of microvesicles from plasma of [152] N. Lindegardh, W. Hanpithakpong, Y. Wattanagoon, P. Singhasivanon, N.J.
healthy donors reveals high individual variability, J. Proteomics 75 (2012) White, N.P.J. Day, Development and validation of a liquid chromatographic–
3574–3584. tandem mass spectrometric method for determination of oseltamivir and its
[128] M.E. Dasenaki, N.S. Thomaidis, Multi-residue determination of seventeen metabolite oseltamivir carboxylate in plasma, saliva and urine, J. Chromatogr.
sulfonamides and five tetracyclines in fish tissue using a multi-stage LC–ESI– B 859 (2007) 74–78.
MS/MS approach based on advanced mass spectrometric techniques, Anal. [153] L.A. Currie, Nomenclature in evaluation of analytical methods including
Chim. Acta 672 (2010) 93–102. detection and quantification capabilities, Pure Appl. Chem. 67 (1995)
[129] B. Dejaegher, X. Capron, J. Smeyers-Verbeke, Y. Vander Heyden, 1699–1723.
Randomization tests to identify significant effects in experimental design for [154] Directive 2000/60/EC of the European Parliament and of the Council
robustness testing, Anal. Chim. Acta 564 (2006) 184–200. establishing a framework for Community action in the field of water policy,
[130] R.L. Plackett, J.P. Burman, The design of optimum multifactorial experiments, CELEX-EUR. Off. J. L 327 (2000) 1–72.
Biometrica 33 (4) (1946) 305–325. [155] ISO 11843–6:2013 Capability of detection – Part 6: Methodology for the
[131] Y. Vander Heyden, K. Luypaert, C. Hartmann, D.L. Massart, J. Hoogmartens, J. determination of the critical value and the minimum detectable value in
De Beer, Ruggedness tests on the high-performance liquid chromatography Poisson distributed measurements by normal approximations.
assay of the United States Pharmacopeia XXII for tetracycline hydrochloride. [156] S. Szilagyi, B. de la Calle, Development and validation of an analytical method
A comparison of experimental designs and statistical interpretations, Anal. for the determination of semicarbazide in fresh egg and in egg powder based
Chim. Acta 312 (1995) 245–262. on the use of liquid chromatography tandem mass spectrometry, Anal. Chim.
[132] Y. Vander Heyden, C. Hartmann, D.L. Massart, A.M.J. Hollands, P. Nuyten, P. Acta 572 (2006) 113–120.
Schoenmakers, Ruggedness testing of a size-exclusion chromatographic [157] M. Rokka, K. Peltonen, Simultaneous determination of four coccidiostats in
assay for low-molecular-mass polymers, J. Chromatogr. A 756 (1) (1996) eggs and broiler meat: validation of an LC-MS/MS method, Food Addit.
89–106. Contam. 23 (2006) 470–478.

You might also like